1
|
Liao Y, Du L, Qiu E, Zeng Y. Characterization of growth arrest-specific transcript 5 and growth arrest-specific transcript 5-related m6A gene signature in glioma: An observational study. Medicine (Baltimore) 2024; 103:e39414. [PMID: 39331894 PMCID: PMC11441912 DOI: 10.1097/md.0000000000039414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 08/02/2024] [Indexed: 09/29/2024] Open
Abstract
Glioma remains a significant clinical challenge and poses a dismal patient prognosis. This study focused on the long noncoding ribonucleic acid growth arrest-specific transcript 5 (GAS5) and explored the role of GAS5 and GAS5-related m6A genes in glioma. We explored the mechanisms of GAS5 expression in glioma using bioinformatic analysis based on glioma data from the Cancer Genome Atlas, GSE1142, and Chinese Glioma Genome Atlas databases. Kaplan-Meier curve analysis, nomogram construction, immune cell infiltration, drug sensitivity, mutations, and pathway analyses were performed to determine the GAS5 mechanism in glioma. Spearman correlation and weighted gene co-expression analyses were used to identify the GAS5-related m6A gene. Furthermore, we explored the correlation between GAS5, GAS5-related m6A gene, and clinical traits using analysis of variance. The Kaplan-Meier curve analysis suggested that patients with high expressions of GAS5 had better survival. The nomogram constructed indicated that GAS5 was an independent prognostic factor. Furthermore, GAS5 significantly correlated with plasma cells. GAS5 expression was significantly associated with biological processes, including oxidative phosphorylation, proteasome, and ribosome mitotic spindle. GAS5 expression was associated with sensitivity to erlotinib and gemcitabine. Differentially expressed GAS5 was significant in histology (P = 2.8e-09), grade (P = 3.7e-05), isocitrate dehydrogenase (IDH) mutation (P = 3.4e-17), 1p/19q co-deletion (Codel) status (P = 1.7e-08), and IDH mutation status and 1p/19q Codel status (P = 2.9e-18). Heterogeneous nuclear ribonucleoproteins C1/C2 (HNRNPC) gene was significant in IDH mutation (P = .008) and IDH mutation status and 1p/19q Codel status (P = 2.1e-05). GAS5 and HNRNPC expressions reflected the malignant grade of glioma and are associated with prognosis. The abnormal expression of GAS5 could be an important biomarker for guiding erlotinib and gemcitabine use in glioma treatment. GAS5 and heterogeneous nuclear ribonucleoproteins C1/C2 are potential diagnostic and prognostic markers for glioma.
Collapse
Affiliation(s)
- Yutian Liao
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China
| | - Li Du
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China
| | - Eryue Qiu
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China
| | - Yiqian Zeng
- Department of Trauma Intensive Care Unit, Zhuzhou Central Hospital, Zhuzhou, China
| |
Collapse
|
2
|
Lad M, Beniwal AS, Jain S, Shukla P, Kalistratova V, Jung J, Shah SS, Yagnik G, Saha A, Sati A, Babikir H, Nguyen AT, Gill S, Rios J, Young JS, Lui A, Salha D, Diaz A, Aghi MK. Glioblastoma induces the recruitment and differentiation of dendritic-like "hybrid" neutrophils from skull bone marrow. Cancer Cell 2024; 42:1549-1569.e16. [PMID: 39255776 PMCID: PMC11446475 DOI: 10.1016/j.ccell.2024.08.008] [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: 03/08/2023] [Revised: 05/28/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024]
Abstract
Tumor-associated neutrophil (TAN) effects on glioblastoma (GBM) biology remain under-characterized. We show here that neutrophils with dendritic features-including morphological complexity, expression of antigen presentation genes, and the ability to process exogenous peptide and stimulate major histocompatibility complex (MHC)II-dependent T cell activation-accumulate intratumorally and suppress tumor growth in vivo. Trajectory analysis of patient TAN scRNA-seq identifies this "hybrid" dendritic-neutrophil phenotype as a polarization state that is distinct from canonical cytotoxic TANs, and which differentiates from local precursors. These hybrid-inducible immature neutrophils-which we identified in patient and murine glioblastomas-arise not from circulation, but from local skull marrow. Through labeled skull flap transplantation and targeted ablation, we characterize calvarial marrow as a contributor of antitumoral myeloid antigen-presenting cells (APCs), including TANs, which elicit T cell cytotoxicity and memory. As such, agents augmenting neutrophil egress from skull marrow-such as intracalvarial AMD3100, whose survival-prolonging effect in GBM we report-present therapeutic potential.
Collapse
Affiliation(s)
- Meeki Lad
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Angad S Beniwal
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Saket Jain
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Poojan Shukla
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Venina Kalistratova
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Jangham Jung
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Sumedh S Shah
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Garima Yagnik
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Atul Saha
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Ankita Sati
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Husam Babikir
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Alan T Nguyen
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Sabraj Gill
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Jennifer Rios
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Jacob S Young
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Austin Lui
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Diana Salha
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Aaron Diaz
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Manish K Aghi
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA.
| |
Collapse
|
3
|
Lu T, Yee PP, Chih SY, Tang M, Chen H, Aregawi DG, Glantz MJ, Zacharia BE, Wang HG, Li W. LC3-associated phagocytosis of neutrophils triggers tumor ferroptotic cell death in glioblastoma. EMBO J 2024; 43:2582-2605. [PMID: 38806658 PMCID: PMC11217441 DOI: 10.1038/s44318-024-00130-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 04/24/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
Necrosis in solid tumors is commonly associated with poor prognostic but how these lesions expand remains unclear. Studies have found that neutrophils associate with and contribute to necrosis development in glioblastoma by inducing tumor cell ferroptosis through transferring myeloperoxidase-containing granules. However, the mechanism of neutrophilic granule transfer remains elusive. We performed an unbiased small molecule screen and found that statins inhibit neutrophil-induced tumor cell death by blocking the neutrophilic granule transfer. Further, we identified a novel process wherein neutrophils are engulfed by tumor cells before releasing myeloperoxidase-containing contents into tumor cells. This neutrophil engulfment is initiated by integrin-mediated adhesion, and further mediated by LC3-associated phagocytosis (LAP), which can be blocked by inhibiting the Vps34-UVRAG-RUBCN-containing PI3K complex. Myeloperoxidase inhibition or Vps34 depletion resulted in reduced necrosis formation and prolonged mouse survival in an orthotopic glioblastoma mouse model. Thus, our study unveils a critical role for LAP-mediated neutrophil internalization in facilitating the transfer of neutrophilic granules, which in turn triggers tumor cell death and necrosis expansion. Targeting this process holds promise for improving glioblastoma prognosis.
Collapse
Affiliation(s)
- Tong Lu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Patricia P Yee
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
- Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Stephen Y Chih
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
- Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Miaolu Tang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Han Chen
- Transmission Electron Microscopy (TEM) Core, Penn State College of Medicine, Hershey, PA, USA
| | - Dawit G Aregawi
- Division of Neurooncology and Skull Base Surgery, Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
- Department of Neurology, Penn State College of Medicine, Hershey, PA, USA
| | - Michael J Glantz
- Division of Neurooncology and Skull Base Surgery, Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Brad E Zacharia
- Division of Neurooncology and Skull Base Surgery, Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
- Department of Otolaryngology-Head and Neck Surgery, Penn State College of Medicine, Hershey, PA, USA
| | - Hong-Gang Wang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
4
|
Lin H, Liu C, Hu A, Zhang D, Yang H, Mao Y. Understanding the immunosuppressive microenvironment of glioma: mechanistic insights and clinical perspectives. J Hematol Oncol 2024; 17:31. [PMID: 38720342 PMCID: PMC11077829 DOI: 10.1186/s13045-024-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Glioblastoma (GBM), the predominant and primary malignant intracranial tumor, poses a formidable challenge due to its immunosuppressive microenvironment, thereby confounding conventional therapeutic interventions. Despite the established treatment regimen comprising surgical intervention, radiotherapy, temozolomide administration, and the exploration of emerging modalities such as immunotherapy and integration of medicine and engineering technology therapy, the efficacy of these approaches remains constrained, resulting in suboptimal prognostic outcomes. In recent years, intensive scrutiny of the inhibitory and immunosuppressive milieu within GBM has underscored the significance of cellular constituents of the GBM microenvironment and their interactions with malignant cells and neurons. Novel immune and targeted therapy strategies have emerged, offering promising avenues for advancing GBM treatment. One pivotal mechanism orchestrating immunosuppression in GBM involves the aggregation of myeloid-derived suppressor cells (MDSCs), glioma-associated macrophage/microglia (GAM), and regulatory T cells (Tregs). Among these, MDSCs, though constituting a minority (4-8%) of CD45+ cells in GBM, play a central component in fostering immune evasion and propelling tumor progression, angiogenesis, invasion, and metastasis. MDSCs deploy intricate immunosuppressive mechanisms that adapt to the dynamic tumor microenvironment (TME). Understanding the interplay between GBM and MDSCs provides a compelling basis for therapeutic interventions. This review seeks to elucidate the immune regulatory mechanisms inherent in the GBM microenvironment, explore existing therapeutic targets, and consolidate recent insights into MDSC induction and their contribution to GBM immunosuppression. Additionally, the review comprehensively surveys ongoing clinical trials and potential treatment strategies, envisioning a future where targeting MDSCs could reshape the immune landscape of GBM. Through the synergistic integration of immunotherapy with other therapeutic modalities, this approach can establish a multidisciplinary, multi-target paradigm, ultimately improving the prognosis and quality of life in patients with GBM.
Collapse
Affiliation(s)
- Hao Lin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Duanwu Zhang
- Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
Sun C, Wang S, Ma Z, Zhou J, Ding Z, Yuan G, Pan Y. Neutrophils in glioma microenvironment: from immune function to immunotherapy. Front Immunol 2024; 15:1393173. [PMID: 38779679 PMCID: PMC11109384 DOI: 10.3389/fimmu.2024.1393173] [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: 02/28/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Glioma is a malignant tumor of the central nervous system (CNS). Currently, effective treatment options for gliomas are still lacking. Neutrophils, as an important member of the tumor microenvironment (TME), are widely distributed in circulation. Recently, the discovery of cranial-meningeal channels and intracranial lymphatic vessels has provided new insights into the origins of neutrophils in the CNS. Neutrophils in the brain may originate more from the skull and adjacent vertebral bone marrow. They cross the blood-brain barrier (BBB) under the action of chemokines and enter the brain parenchyma, subsequently migrating to the glioma TME and undergoing phenotypic changes upon contact with tumor cells. Under glycolytic metabolism model, neutrophils show complex and dual functions in different stages of cancer progression, including participation in the malignant progression, immune suppression, and anti-tumor effects of gliomas. Additionally, neutrophils in the TME interact with other immune cells, playing a crucial role in cancer immunotherapy. Targeting neutrophils may be a novel generation of immunotherapy and improve the efficacy of cancer treatments. This article reviews the molecular mechanisms of neutrophils infiltrating the central nervous system from the external environment, detailing the origin, functions, classifications, and targeted therapies of neutrophils in the context of glioma.
Collapse
Affiliation(s)
- Chao Sun
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Siwen Wang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Zhen Ma
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Jinghuan Zhou
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Zilin Ding
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Guoqiang Yuan
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yawen Pan
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
6
|
Kaminska P, Ovesen PL, Jakiel M, Obrebski T, Schmidt V, Draminski M, Bilska AG, Bieniek M, Anink J, Paterczyk B, Jensen AMG, Piatek S, Andersen OM, Aronica E, Willnow TE, Kaminska B, Dabrowski MJ, Malik AR. SorLA restricts TNFα release from microglia to shape a glioma-supportive brain microenvironment. EMBO Rep 2024; 25:2278-2305. [PMID: 38499808 PMCID: PMC11094098 DOI: 10.1038/s44319-024-00117-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 03/20/2024] Open
Abstract
SorLA, encoded by the gene SORL1, is an intracellular sorting receptor of the VPS10P domain receptor gene family. Although SorLA is best recognized for its ability to shuttle target proteins between intracellular compartments in neurons, recent data suggest that also its microglial expression can be of high relevance for the pathogenesis of brain diseases, including glioblastoma (GBM). Here, we interrogated the impact of SorLA on the functional properties of glioma-associated microglia and macrophages (GAMs). In the GBM microenvironment, GAMs are re-programmed and lose the ability to elicit anti-tumor responses. Instead, they acquire a glioma-supporting phenotype, which is a key mechanism promoting glioma progression. Our re-analysis of published scRNA-seq data from GBM patients revealed that functional phenotypes of GAMs are linked to the level of SORL1 expression, which was further confirmed using in vitro models. Moreover, we demonstrate that SorLA restrains secretion of TNFα from microglia to restrict the inflammatory potential of these cells. Finally, we show that loss of SorLA exacerbates the pro-inflammatory response of microglia in the murine model of glioma and suppresses tumor growth.
Collapse
Affiliation(s)
- Paulina Kaminska
- Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Peter L Ovesen
- Max-Delbrueck Center for Molecular Medicine, 13125, Berlin, Germany
| | - Mateusz Jakiel
- Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
- Institute of Computer Science, 01-248, Warsaw, Poland
| | - Tomasz Obrebski
- Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
| | - Vanessa Schmidt
- Max-Delbrueck Center for Molecular Medicine, 13125, Berlin, Germany
| | | | - Aleksandra G Bilska
- Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
- Museum and Institute of Zoology, Polish Academy of Sciences, 00-679, Warsaw, Poland
| | | | - Jasper Anink
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, 1105AZ, Amsterdam, The Netherlands
| | - Bohdan Paterczyk
- Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
| | | | - Sylwia Piatek
- Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland
| | - Olav M Andersen
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, 1105AZ, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland, 2103 SW, Heemstede, The Netherlands
| | - Thomas E Willnow
- Max-Delbrueck Center for Molecular Medicine, 13125, Berlin, Germany
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark
| | - Bozena Kaminska
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | | | - Anna R Malik
- Faculty of Biology, University of Warsaw, 02-096, Warsaw, Poland.
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| |
Collapse
|
7
|
Kienzler JC, Becher B. Immunity in malignant brain tumors: Tumor entities, role of immunotherapy, and specific contribution of myeloid cells to the brain tumor microenvironment. Eur J Immunol 2024; 54:e2250257. [PMID: 37940552 DOI: 10.1002/eji.202250257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/10/2023]
Abstract
Malignant brain tumors lack effective treatment, that can improve their poor overall survival achieved with standard of care. Advancement in different cancer treatments has shifted the focus in brain tumor research and clinical trials toward immunotherapy-based approaches. The investigation of the immune cell landscape revealed a dominance of myeloid cells in the tumor microenvironment. Their exact roles and functions are the subject of ongoing research. Current evidence suggests a complex interplay of tumor cells and myeloid cells with competing functions toward support vs. control of tumor growth. Here, we provide a brief overview of the three most abundant brain tumor entities: meningioma, glioma, and brain metastases. We also describe the field of ongoing immunotherapy trials and their results, including immune checkpoint inhibitors, vaccination studies, oncolytic viral therapy, and CAR-T cells. Finally, we summarize the phenotypes of microglia, monocyte-derived macrophages, border-associated macrophages, neutrophils, and potential novel therapy targets.
Collapse
Affiliation(s)
- Jenny C Kienzler
- Institute of Experimental Immunology, Inflammation Research Lab, University of Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, Inflammation Research Lab, University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Elguindy M, Young JS, Mondal I, Lu RO, Ho WS. Glioma-Immune Cell Crosstalk in Tumor Progression. Cancers (Basel) 2024; 16:308. [PMID: 38254796 PMCID: PMC10813573 DOI: 10.3390/cancers16020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/21/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Glioma progression is a complex process controlled by molecular factors that coordinate the crosstalk between tumor cells and components of the tumor microenvironment (TME). Among these, immune cells play a critical role in cancer survival and progression. The complex interplay between cancer cells and the immune TME influences the outcome of immunotherapy and other anti-cancer therapies. Here, we present an updated view of the pro- and anti-tumor activities of the main myeloid and lymphocyte cell populations in the glioma TME. We review the underlying mechanisms involved in crosstalk between cancer cells and immune cells that enable gliomas to evade the immune system and co-opt these cells for tumor growth. Lastly, we discuss the current and experimental therapeutic options being developed to revert the immunosuppressive activity of the glioma TME. Knowledge of the complex interplay that elapses between tumor and immune cells may help develop new combination treatments able to overcome tumor immune evasion mechanisms and enhance response to immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | - Winson S. Ho
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
9
|
Brosque A, Friedmann-Morvinski D. Drivers of heterogeneity in the glioblastoma immune microenvironment. Curr Opin Cell Biol 2023; 85:102279. [PMID: 37984008 DOI: 10.1016/j.ceb.2023.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/22/2023]
Abstract
Glioblastoma is the most common and aggressive primary brain tumor, characterized by a highly complex and heterogeneous tumor immune microenvironment (TIME). In this review, we discuss the impact of tumor-intrinsic and tumor-extrinsic drivers that contribute to heterogeneity in the adult glioblastoma TIME, focusing on four main factors: genetic drivers, sex, age, and standard of care therapy. We describe recent insights into how each of these factors affects key aspects ranging from TIME composition to therapy response, with an emphasis on the cross-talk between tumor and immune cells. Deciphering these local interactions is fundamental to understanding therapy resistance and identifying novel immunomodulatory strategies.
Collapse
Affiliation(s)
- Alina Brosque
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel. https://twitter.com/alibrosque
| | - Dinorah Friedmann-Morvinski
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
10
|
Ross JL, Hambardzumyan D. Et tu neutrophils? The brain reprograms neutrophils to facilitate tumor progression. Immunity 2023; 56:2469-2471. [PMID: 37967529 DOI: 10.1016/j.immuni.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 11/17/2023]
Abstract
Neutrophils have remained understudied in malignant brain tumors. In a recent issue of Cell, Maas et al. analyze brain tumor-patient samples and demonstrate that the brain microenvironment reprograms infiltrating neutrophils to enhance their longevity and increase their immune-suppressive and pro-angiogenic properties.
Collapse
Affiliation(s)
- James L Ross
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Dolores Hambardzumyan
- Departments of Oncological Sciences and Neurosurgery, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Maas RR, Soukup K, Fournier N, Massara M, Galland S, Kornete M, Wischnewski V, Lourenco J, Croci D, Álvarez-Prado ÁF, Marie DN, Lilja J, Marcone R, Calvo GF, Santalla Mendez R, Aubel P, Bejarano L, Wirapati P, Ballesteros I, Hidalgo A, Hottinger AF, Brouland JP, Daniel RT, Hegi ME, Joyce JA. The local microenvironment drives activation of neutrophils in human brain tumors. Cell 2023; 186:4546-4566.e27. [PMID: 37769657 DOI: 10.1016/j.cell.2023.08.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 07/11/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023]
Abstract
Neutrophils are abundant immune cells in the circulation and frequently infiltrate tumors in substantial numbers. However, their precise functions in different cancer types remain incompletely understood, including in the brain microenvironment. We therefore investigated neutrophils in tumor tissue of glioma and brain metastasis patients, with matched peripheral blood, and herein describe the first in-depth analysis of neutrophil phenotypes and functions in these tissues. Orthogonal profiling strategies in humans and mice revealed that brain tumor-associated neutrophils (TANs) differ significantly from blood neutrophils and have a prolonged lifespan and immune-suppressive and pro-angiogenic capacity. TANs exhibit a distinct inflammatory signature, driven by a combination of soluble inflammatory mediators including tumor necrosis factor alpha (TNF-ɑ) and Ceruloplasmin, which is more pronounced in TANs from brain metastasis versus glioma. Myeloid cells, including tumor-associated macrophages, emerge at the core of this network of pro-inflammatory mediators, supporting the concept of a critical myeloid niche regulating overall immune suppression in human brain tumors.
Collapse
Affiliation(s)
- Roeltje R Maas
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland; Neuroscience Research Center, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Klara Soukup
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland
| | - Nadine Fournier
- Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; Translational Data Science Group, Swiss Institute of Bioinformatics, Lausanne 1011, Switzerland; Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne 1011, Switzerland
| | - Matteo Massara
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Sabine Galland
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Mara Kornete
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland
| | - Vladimir Wischnewski
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Joao Lourenco
- Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; Translational Data Science Group, Swiss Institute of Bioinformatics, Lausanne 1011, Switzerland
| | - Davide Croci
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland
| | - Ángel F Álvarez-Prado
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Damien N Marie
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland
| | - Johanna Lilja
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland
| | - Rachel Marcone
- Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; Translational Data Science Group, Swiss Institute of Bioinformatics, Lausanne 1011, Switzerland
| | - Gabriel F Calvo
- Department of Mathematics & MOLAB-Mathematical Oncology Laboratory, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| | - Rui Santalla Mendez
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Pauline Aubel
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Leire Bejarano
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Pratyaksha Wirapati
- Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne 1011, Switzerland
| | - Iván Ballesteros
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Andrés Hidalgo
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain; Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Andreas F Hottinger
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Jean-Philippe Brouland
- Department of Pathology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne 1011, Switzerland
| | - Roy T Daniel
- L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Monika E Hegi
- L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland; Neuroscience Research Center, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne 1011, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne 1011, Switzerland.
| |
Collapse
|
12
|
Tsai HC, Tong ZJ, Hwang TL, Wei KC, Chen PY, Huang CY, Chen KT, Lin YJ, Cheng HW, Wang HT. Acrolein produced by glioma cells under hypoxia inhibits neutrophil AKT activity and suppresses anti-tumoral activities. Free Radic Biol Med 2023; 207:17-28. [PMID: 37414347 DOI: 10.1016/j.freeradbiomed.2023.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/23/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023]
Abstract
Acrolein, which is the most reactive aldehyde, is a byproduct of lipid peroxidation in a hypoxic environment. Acrolein has been shown to form acrolein-cysteine bonds, resulting in functional changes in proteins and immune effector cell suppression. Neutrophils are the most abundant immune effector cells in circulation in humans. In the tumor microenvironment, proinflammatory tumor-associated neutrophils (TANs), which are termed N1 neutrophils, exert antitumor effects via the secretion of cytokines, while anti-inflammatory neutrophils (N2 neutrophils) support tumor growth. Glioma is characterized by significant tissue hypoxia, immune cell infiltration, and a highly immunosuppressive microenvironment. In glioma, neutrophils exert antitumor effects early in tumor development but gradually shift to a tumor-supporting role as the tumor develops. However, the mechanism of this anti-to protumoral switch in TANs remains unclear. In this study, we found that the production of acrolein in glioma cells under hypoxic conditions inhibited neutrophil activation and induced an anti-inflammatory phenotype by directly reacting with Cys310 of AKT and inhibiting AKT activity. A higher percentage of cells expressing acrolein adducts in tumor tissue are associated with poorer prognosis in glioblastoma patients. Furthermore, high-grade glioma patients have increased serum acrolein levels and impaired neutrophil functions. These results suggest that acrolein suppresses neutrophil function and contributes to the switch in the neutrophil phenotype in glioma.
Collapse
Affiliation(s)
- Hong-Chieh Tsai
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan; School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Zhen-Jie Tong
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, 333, Taiwan; Department of Anaesthesiology, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan; Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 333, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan; School of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan; Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei Municipal, 236, Taiwan
| | - Pin-Yuan Chen
- School of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan; Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan; School of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Ko-Ting Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan; School of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Ya-Jui Lin
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan; School of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Hsiao-Wei Cheng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Hsiang-Tsui Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan; Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan; Doctor Degree Program in Toxicology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
13
|
Yuile A, Wei JQ, Mohan AA, Hotchkiss KM, Khasraw M. Interdependencies of the Neuronal, Immune and Tumor Microenvironment in Gliomas. Cancers (Basel) 2023; 15:2856. [PMID: 37345193 PMCID: PMC10216320 DOI: 10.3390/cancers15102856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
Gliomas are the most common primary brain malignancy and are universally fatal. Despite significant breakthrough in understanding tumor biology, treatment breakthroughs have been limited. There is a growing appreciation that major limitations on effective treatment are related to the unique and highly complex glioma tumor microenvironment (TME). The TME consists of multiple different cell types, broadly categorized into tumoral, immune and non-tumoral, non-immune cells. Each group provides significant influence on the others, generating a pro-tumor dynamic with significant immunosuppression. In addition, glioma cells are highly heterogenous with various molecular distinctions on the cellular level. These variations, in turn, lead to their own unique influence on the TME. To develop future treatments, an understanding of this complex TME interplay is needed. To this end, we describe the TME in adult gliomas through interactions between its various components and through various glioma molecular phenotypes.
Collapse
Affiliation(s)
- Alexander Yuile
- Department of Medical Oncology, Royal North Shore Hospital, Reserve Road, St Leonards, NSW 2065, Australia
- The Brain Cancer Group, North Shore Private Hospital, 3 Westbourne Street, St Leonards, NSW 2065, Australia
- Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Joe Q. Wei
- Department of Medical Oncology, Royal North Shore Hospital, Reserve Road, St Leonards, NSW 2065, Australia
- Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Aditya A. Mohan
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| | - Kelly M. Hotchkiss
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| | - Mustafa Khasraw
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA
| |
Collapse
|
14
|
Mu J, Gong J, Shi M, Zhang Y. Analysis and validation of aging-related genes in prognosis and immune function of glioblastoma. BMC Med Genomics 2023; 16:109. [PMID: 37208656 DOI: 10.1186/s12920-023-01538-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 05/09/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a common malignant brain tumor with poor prognosis and high mortality. Numerous reports have identified the correlation between aging and the prognosis of patients with GBM. The purpose of this study was to establish a prognostic model for GBM patients based on aging-related gene (ARG) to help determine the prognosis of GBM patients. METHODS 143 patients with GBM from The Cancer Genomic Atlas (TCGA), 218 patients with GBM from the Chinese Glioma Genomic Atlas (CGGA) of China and 50 patients from Gene Expression Omnibus (GEO) were included in the study. R software (V4.2.1) and bioinformatics statistical methods were used to develop prognostic models and study immune infiltration and mutation characteristics. RESULTS Thirteen genes were screened out and used to establish the prognostic model finally, and the risk scores of the prognostic model was an independent factor (P < 0.001), which indicated a good prediction ability. In addition, there are significant differences in immune infiltration and mutation characteristics between the two groups with high and low risk scores. CONCLUSION The prognostic model of GBM patients based on ARGs can predict the prognosis of GBM patients. However, this signature requires further investigation and validation in larger cohort studies.
Collapse
Affiliation(s)
- Jianhua Mu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jianan Gong
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Miao Shi
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Yinian Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
Yeo AT, Shah R, Aliazis K, Pal R, Xu T, Zhang P, Rawal S, Rose CM, Varn FS, Appleman VA, Yoon J, Varma H, Gygi SP, Verhaak RG, Boussiotis VA, Charest A. Driver Mutations Dictate the Immunologic Landscape and Response to Checkpoint Immunotherapy of Glioblastoma. Cancer Immunol Res 2023; 11:629-645. [PMID: 36881002 PMCID: PMC10155040 DOI: 10.1158/2326-6066.cir-22-0655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/20/2022] [Accepted: 03/03/2023] [Indexed: 03/08/2023]
Abstract
The composition of the tumor immune microenvironment (TIME) is considered a key determinant of patients' response to immunotherapy. The mechanisms underlying TIME formation and development over time are poorly understood. Glioblastoma (GBM) is a lethal primary brain cancer for which there are no curative treatments. GBMs are immunologically heterogeneous and impervious to checkpoint blockade immunotherapies. Utilizing clinically relevant genetic mouse models of GBM, we identified distinct immune landscapes associated with expression of EGFR wild-type and mutant EGFRvIII cancer driver mutations. Over time, accumulation of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) was more pronounced in EGFRvIII-driven GBMs and was correlated with resistance to PD-1 and CTLA-4 combination checkpoint blockade immunotherapy. We determined that GBM-secreted CXCL1/2/3 and PMN-MDSC-expressed CXCR2 formed an axis regulating output of PMN-MDSCs from the bone marrow leading to systemic increase in these cells in the spleen and GBM tumor-draining lymph nodes. Pharmacologic targeting of this axis induced a systemic decrease in the numbers of PMN-MDSC, facilitated responses to PD-1 and CTLA-4 combination checkpoint blocking immunotherapy, and prolonged survival in mice bearing EGFRvIII-driven GBM. Our results uncover a relationship between cancer driver mutations, TIME composition, and sensitivity to checkpoint blockade in GBM and support the stratification of patients with GBM for checkpoint blockade therapy based on integrated genotypic and immunologic profiles.
Collapse
Affiliation(s)
- Alan T. Yeo
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Sackler School of Graduate Studies, Tufts University School of Medicine, Boston, Massachusetts
| | - Rushil Shah
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Konstantinos Aliazis
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Rinku Pal
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Tuoye Xu
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Piyan Zhang
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Shruti Rawal
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Frederick S. Varn
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Vicky A. Appleman
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Joon Yoon
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Hemant Varma
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Roel G.W. Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Vassiliki A. Boussiotis
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Al Charest
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Chen Z, Soni N, Pinero G, Giotti B, Eddins DJ, Lindblad KE, Ross JL, Puigdelloses Vallcorba M, Joshi T, Angione A, Thomason W, Keane A, Tsankova NM, Gutmann DH, Lira SA, Lujambio A, Ghosn EEB, Tsankov AM, Hambardzumyan D. Monocyte depletion enhances neutrophil influx and proneural to mesenchymal transition in glioblastoma. Nat Commun 2023; 14:1839. [PMID: 37012245 PMCID: PMC10070461 DOI: 10.1038/s41467-023-37361-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
Myeloid cells comprise the majority of immune cells in tumors, contributing to tumor growth and therapeutic resistance. Incomplete understanding of myeloid cells response to tumor driver mutation and therapeutic intervention impedes effective therapeutic design. Here, by leveraging CRISPR/Cas9-based genome editing, we generate a mouse model that is deficient of all monocyte chemoattractant proteins. Using this strain, we effectively abolish monocyte infiltration in genetically engineered murine models of de novo glioblastoma (GBM) and hepatocellular carcinoma (HCC), which show differential enrichment patterns for monocytes and neutrophils. Eliminating monocyte chemoattraction in monocyte enriched PDGFB-driven GBM invokes a compensatory neutrophil influx, while having no effect on Nf1-silenced GBM model. Single-cell RNA sequencing reveals that intratumoral neutrophils promote proneural-to-mesenchymal transition and increase hypoxia in PDGFB-driven GBM. We further demonstrate neutrophil-derived TNF-a directly drives mesenchymal transition in PDGFB-driven primary GBM cells. Genetic or pharmacological inhibiting neutrophils in HCC or monocyte-deficient PDGFB-driven and Nf1-silenced GBM models extend the survival of tumor-bearing mice. Our findings demonstrate tumor-type and genotype dependent infiltration and function of monocytes and neutrophils and highlight the importance of targeting them simultaneously for cancer treatments.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Nishant Soni
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gonzalo Pinero
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Devon J Eddins
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Medicine, Lowance Center for Human Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Katherine E Lindblad
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - James L Ross
- Emory University Department of Microbiology and Immunology, Emory Vaccine Center, Atlanta, GA, 30322, USA
| | | | - Tanvi Joshi
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Angelo Angione
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wes Thomason
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Aislinn Keane
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nadejda M Tsankova
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sergio A Lira
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Amaia Lujambio
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eliver E B Ghosn
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Medicine, Lowance Center for Human Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Alexander M Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
17
|
Lad BM, Beniwal AS, Jain S, Shukla P, Jung J, Shah SS, Yagnik G, Babikir H, Nguyen AT, Gill S, Young JS, Lui A, Salha D, Diaz A, Aghi MK. Glioblastoma induces the recruitment and differentiation of hybrid neutrophils from skull bone marrow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.24.534105. [PMID: 36993266 PMCID: PMC10055347 DOI: 10.1101/2023.03.24.534105] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Tumor-associated neutrophil (TAN) effects on glioblastoma biology remain under-characterized. We show here that 'hybrid' neutrophils with dendritic features - including morphological complexity, expression of antigen presentation genes, and the ability to process exogenous peptide and stimulate MHCII-dependent T cell activation - accumulate intratumorally and suppress tumor growth in vivo . Trajectory analysis of patient TAN scRNA-seq identifies this phenotype as a polarization state which is distinct from canonical cytotoxic TANs and differentiates intratumorally from immature precursors absent in circulation. Rather, these hybrid-inducible immature neutrophils - which we identified in patient and murine glioblastomas - arise from local skull marrow. Through labeled skull flap transplantation and targeted ablation, we characterize calvarial marrow as a potent contributor of antitumoral myeloid APCs, including hybrid TANs and dendritic cells, which elicit T cell cytotoxicity and memory. As such, agents augmenting neutrophil egress from skull marrow - such as intracalvarial AMD3100 whose survival prolonging-effect in GBM we demonstrate - present therapeutic potential.
Collapse
|
18
|
Wu S, Li X, Hong F, Chen Q, Yu Y, Guo S, Xie Y, Xiao N, Kong X, Mo W, Wang Z, Chen S, Zeng F. Integrative analysis of single-cell transcriptomics reveals age-associated immune landscape of glioblastoma. Front Immunol 2023; 14:1028775. [PMID: 36761752 PMCID: PMC9903136 DOI: 10.3389/fimmu.2023.1028775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
Glioblastoma (GBM) is the most malignant tumor in center nervous system. Clinical statistics revealed that senior GBM patients had a worse overall survival (OS) comparing with that of patients in other ages, which is mainly related with tumor microenvironment including tumor-associated immune cells in particular. However, the immune heterogeneity and age-related prognosis in GBM are under studied. Here we developed a machine learning-based method to integrate public large-scale single-cell RNA sequencing (scRNA-seq) datasets to establish a comprehensive atlas of immune cells infiltrating in cross-age GBM. We found that the compositions of the immune cells are remarkably different across ages. Brain-resident microglia constitute the majority of glioblastoma-associated macrophages (GAMs) in patients, whereas dramatic elevation of extracranial monocyte-derived macrophages (MDMs) is observed in GAMs of senior patients, which contributes to the worse prognosis of aged patients. Further analysis suggests that the increased MDMs arisen from excessive recruitment and proliferation of peripheral monocytes not only lead to the T cell function inhibition in GBM, but also stimulate tumor cells proliferation via VEGFA secretion. In summary, our work provides new cues for the correlational relationship between the immune microenvironment of GBM and aging, which might be insightful for precise and effective therapeutic interventions for senior GBM patients.
Collapse
Affiliation(s)
- Songang Wu
- Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, College of Chemistry and Chemical Engineering, Xiamen University, Fujian, China
| | - Xuewen Li
- Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,National Institute for Data Science in Health and Medicine, School of Life Sciences, Xiamen University, Fujian, China
| | - Fan Hong
- Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,National Institute for Data Science in Health and Medicine, School of Life Sciences, Xiamen University, Fujian, China
| | - Qiang Chen
- Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,National Institute for Data Science in Health and Medicine, School of Life Sciences, Xiamen University, Fujian, China
| | - Yingying Yu
- Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,National Institute for Data Science in Health and Medicine, School of Life Sciences, Xiamen University, Fujian, China
| | - Shuanghui Guo
- Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,National Institute for Data Science in Health and Medicine, School of Life Sciences, Xiamen University, Fujian, China
| | - Yuanyuan Xie
- Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, College of Chemistry and Chemical Engineering, Xiamen University, Fujian, China,Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China
| | - Naian Xiao
- Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, College of Chemistry and Chemical Engineering, Xiamen University, Fujian, China,Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China
| | - Xuwen Kong
- Department of Automation, School of Aerospace Engineering, Xiamen University, Fujian, China
| | - Wei Mo
- Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,National Institute for Data Science in Health and Medicine, School of Life Sciences, Xiamen University, Fujian, China
| | - Zhanxiang Wang
- Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, College of Chemistry and Chemical Engineering, Xiamen University, Fujian, China,Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,*Correspondence: Feng Zeng, ; Shaoxuan Chen, ; Zhanxiang Wang,
| | - Shaoxuan Chen
- Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,National Institute for Data Science in Health and Medicine, School of Life Sciences, Xiamen University, Fujian, China,*Correspondence: Feng Zeng, ; Shaoxuan Chen, ; Zhanxiang Wang,
| | - Feng Zeng
- Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, College of Chemistry and Chemical Engineering, Xiamen University, Fujian, China,Department of Neuroscience, Fujian Key Laboratory of Brain Tumors Diagnosis and Precision Treatment, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, China,Department of Automation, School of Aerospace Engineering, Xiamen University, Fujian, China,*Correspondence: Feng Zeng, ; Shaoxuan Chen, ; Zhanxiang Wang,
| |
Collapse
|
19
|
The Tumor Immune Microenvironment in Primary CNS Neoplasms: A Review of Current Knowledge and Therapeutic Approaches. Int J Mol Sci 2023; 24:ijms24032020. [PMID: 36768342 PMCID: PMC9917056 DOI: 10.3390/ijms24032020] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Primary CNS neoplasms are responsible for considerable mortality and morbidity, and many therapies directed at primary brain tumors have proven unsuccessful despite their success in preclinical studies. Recently, the tumor immune microenvironment has emerged as a critical aspect of primary CNS neoplasms that may affect their malignancy, prognosis, and response to therapy across patients and tumor grades. This review covers the tumor microenvironment of various primary CNS neoplasms, with a focus on glioblastoma and meningioma. Additionally, current therapeutic strategies based on elements of the tumor microenvironment, including checkpoint inhibitor therapy and immunotherapeutic vaccines, are discussed.
Collapse
|
20
|
Friedmann-Morvinski D, Hambardzumyan D. Monocyte-neutrophil entanglement in glioblastoma. J Clin Invest 2023; 133:163451. [PMID: 36594465 PMCID: PMC9797336 DOI: 10.1172/jci163451] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Glioblastoma (GBM) is the most belligerent and frequent brain tumor in adults. Research over the past two decades has provided increased knowledge of the genomic and molecular landscape of GBM and highlighted the presence of a high degree of inter- and intratumor heterogeneity within the neoplastic compartment. It is now appreciated that GBMs are composed of multiple distinct and impressionable neoplastic and non-neoplastic cell types that form the unique brain tumor microenvironment (TME). Non-neoplastic cells in the TME form reciprocal interactions with neoplastic cells to promote tumor growth and invasion, and together they influence the tumor response to standard-of-care therapies as well as emerging immunotherapies. One of the most prevalent non-neoplastic cell types in the GBM TME are myeloid cells, the most abundant of which are of hematopoietic origin, including monocytes/monocyte-derived macrophages. Less abundant, although still a notable presence, are neutrophils of hematopoietic origin and intrinsic brain-resident microglia. In this Review we focus on neutrophils and monocytes that infiltrate tumors from the blood circulation, their heterogeneity, and their interactions with neoplastic cells and other non-neoplastic cells in the TME. We conclude with an overview of challenges in targeting these cells and discuss avenues for therapeutic exploitation to improve the dismal outcomes of patients with GBM.
Collapse
Affiliation(s)
- Dinorah Friedmann-Morvinski
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, and,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, Tisch Cancer Institute, and,Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
21
|
Picca A, Finocchiaro G. Deciphering diffuse glioma immune microenvironment as a key to improving immunotherapy results. Curr Opin Oncol 2022; 34:653-660. [PMID: 36000367 DOI: 10.1097/cco.0000000000000895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Immunotherapeutic approaches have yet to demonstrate their clinical efficacy in diffuse gliomas. Evidence is mounting that the central nervous system is subject to immune surveillance, but brain tumours manage to escape due to factors intrinsic to their tumoral immune microenvironment (TME). This review aims to discuss the recently characterized molecular bases of the glioma TME and the potentially actionable targets to improve immunotherapeutic results in these hard-to-treat cancers. RECENT FINDINGS Single-cell studies defined the composition of the glioma immune TME and its peculiarities compared with other solid cancers. In isocitrate dehydrogenase (IDH) wildtype gliomas, the TME is enriched in myeloid cells (monocyte-derived macrophages and resident microglia) with mainly immunosuppressive functions. Lymphocytes can infiltrate the glioma TME, but are exposed to multiple immunomodulating signals that render them in a state of deep exhaustion. IDH mutant gliomas produce the oncometabolite D-2-hydroxyglutarate with negative effects on leukocyte recruitment and function, resulting in the induction of an 'immune-desert' TME. SUMMARY Several molecular pathways have been recently identified in the induction of an 'immune-hostile' microenvironment in diffuse gliomas, unravelling potential vulnerabilities to targeted immunotherapies.
Collapse
Affiliation(s)
- Alberto Picca
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, F-75013 Paris, France. Equipe labellisée LNCC
| | - Gaetano Finocchiaro
- Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Croci D, Santalla Méndez R, Temme S, Soukup K, Fournier N, Zomer A, Colotti R, Wischnewski V, Flögel U, van Heeswijk RB, Joyce JA. Multispectral fluorine-19 MRI enables longitudinal and noninvasive monitoring of tumor-associated macrophages. Sci Transl Med 2022; 14:eabo2952. [PMID: 36260692 DOI: 10.1126/scitranslmed.abo2952] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
High-grade gliomas, the most common and aggressive primary brain tumors, are characterized by a complex tumor microenvironment (TME). Among the immune cells infiltrating the glioma TME, tumor-associated microglia and macrophages (TAMs) constitute the major compartment. In patients with gliomas, increased TAM abundance is associated with more aggressive disease. Alterations in TAM phenotypes and functions have been reported in preclinical models of multiple cancers during tumor development and after therapeutic interventions, including radiotherapy and molecular targeted therapies. These findings indicate that it is crucial to evaluate TAM abundance and dynamics over time. Current techniques to quantify TAMs in patients rely mainly on histological staining of tumor biopsies. Although informative, these techniques require an invasive procedure to harvest the tissue sample and typically only result in a snapshot of a small region at a single point in time. Fluorine isotope 19 MRI (19F MRI) represents a powerful means to noninvasively and longitudinally monitor myeloid cells in pathological conditions by intravenously injecting perfluorocarbon-containing nanoparticles (PFC-NP). In this study, we demonstrated the feasibility and power of 19F MRI in preclinical models of gliomagenesis, breast-to-brain metastasis, and breast cancer and showed that the major cellular source of 19F signal consists of TAMs. Moreover, multispectral 19F MRI with two different PFC-NP allowed us to identify spatially and temporally distinct TAM niches in radiotherapy-recurrent murine gliomas. Together, we have imaged TAMs noninvasively and longitudinally with integrated cellular, spatial, and temporal resolution, thus revealing important biological insights into the critical functions of TAMs, including in disease recurrence.
Collapse
Affiliation(s)
- Davide Croci
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland.,Agora Cancer Research Center, Lausanne 1011, Switzerland
| | - Rui Santalla Méndez
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland.,Agora Cancer Research Center, Lausanne 1011, Switzerland
| | - Sebastian Temme
- Department of Anesthesiology, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf 40225, Germany.,Experimental Cardiovascular Imaging, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf 40225, Germany
| | - Klara Soukup
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland.,Agora Cancer Research Center, Lausanne 1011, Switzerland
| | - Nadine Fournier
- Agora Cancer Research Center, Lausanne 1011, Switzerland.,Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne 1011, Switzerland
| | - Anoek Zomer
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland.,Agora Cancer Research Center, Lausanne 1011, Switzerland
| | - Roberto Colotti
- In Vivo Imaging Facility (IVIF), Department of Research and Training, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Vladimir Wischnewski
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland.,Agora Cancer Research Center, Lausanne 1011, Switzerland
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf 40225, Germany.,Institute for Molecular Cardiology, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf 40225, Germany
| | - Ruud B van Heeswijk
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Lausanne 1011, Switzerland.,Agora Cancer Research Center, Lausanne 1011, Switzerland
| |
Collapse
|
23
|
Chryplewicz A, Scotton J, Tichet M, Zomer A, Shchors K, Joyce JA, Homicsko K, Hanahan D. Cancer cell autophagy, reprogrammed macrophages, and remodeled vasculature in glioblastoma triggers tumor immunity. Cancer Cell 2022; 40:1111-1127.e9. [PMID: 36113478 PMCID: PMC9580613 DOI: 10.1016/j.ccell.2022.08.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/29/2022] [Accepted: 08/15/2022] [Indexed: 01/10/2023]
Abstract
Glioblastoma (GBM) is poorly responsive to therapy and invariably lethal. One conceivable strategy to circumvent this intractability is to co-target distinctive mechanistic components of the disease, aiming to concomitantly disrupt multiple capabilities required for tumor progression and therapeutic resistance. We assessed this concept by combining vascular endothelial growth factor (VEGF) pathway inhibitors that remodel the tumor vasculature with the tricyclic antidepressant imipramine, which enhances autophagy in GBM cancer cells and unexpectedly reprograms immunosuppressive tumor-associated macrophages via inhibition of histamine receptor signaling to become immunostimulatory. While neither drug is efficacious as monotherapy, the combination of imipramine with VEGF pathway inhibitors orchestrates the infiltration and activation of CD8 and CD4 T cells, producing significant therapeutic benefit in several GBM mouse models. Inclusion up front of immune-checkpoint blockade with anti-programmed death-ligand 1 (PD-L1) in eventually relapsing tumors markedly extends survival benefit. The results illustrate the potential of mechanism-guided therapeutic co-targeting of disparate biological vulnerabilities in the tumor microenvironment.
Collapse
Affiliation(s)
- Agnieszka Chryplewicz
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland; Agora Translational Cancer Research Center, Lausanne, Switzerland
| | - Julie Scotton
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Mélanie Tichet
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland; Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Anoek Zomer
- Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Ksenya Shchors
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Johanna A Joyce
- Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland; Department of Oncology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman (SCCL), Lausanne/Geneva, Switzerland
| | - Krisztian Homicsko
- Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland; Department of Oncology, University of Lausanne, Lausanne, Switzerland; Swiss Cancer Center Leman (SCCL), Lausanne/Geneva, Switzerland
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland; Agora Translational Cancer Research Center, Lausanne, Switzerland; Lausanne Branch, Ludwig Institute for Cancer Research, Lausanne, Switzerland; Swiss Cancer Center Leman (SCCL), Lausanne/Geneva, Switzerland.
| |
Collapse
|
24
|
Pang L, Khan F, Heimberger AB, Chen P. Mechanism and therapeutic potential of tumor-immune symbiosis in glioblastoma. Trends Cancer 2022; 8:839-854. [PMID: 35624002 PMCID: PMC9492629 DOI: 10.1016/j.trecan.2022.04.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/20/2022]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal form of brain tumor in human adults. Myeloid-lineage cells, including macrophages, microglia, myeloid-derived suppressor cells (MDSCs), and neutrophils, are the most frequent types of cell in the GBM tumor microenvironment (TME) that contribute to tumor progression. Emerging experimental evidence indicates that symbiotic interactions between cancer cells and myeloid cells are critical for tumor growth and immunotherapy resistance in GBM. In this review, we discuss the molecular mechanisms whereby cancer cells shape a myeloid cell-mediated immunosuppressive TME and, reciprocally, how such myeloid cells affect tumor progression and immunotherapy efficiency in GBM. Moreover, we highlight tumor-T cell symbiosis and summarize immunotherapeutic strategies intercepting this co-dependency in GBM.
Collapse
Affiliation(s)
- Lizhi Pang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Fatima Khan
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Amy B Heimberger
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peiwen Chen
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
25
|
Multimodal imaging of the dynamic brain tumor microenvironment during glioblastoma progression and in response to treatment. iScience 2022; 25:104570. [PMID: 35769877 PMCID: PMC9234718 DOI: 10.1016/j.isci.2022.104570] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/02/2022] [Accepted: 06/06/2022] [Indexed: 12/13/2022] Open
Abstract
Tumors evolve in a dynamic communication with their native tissue environment and recruited immune cells. The diverse components of the tumor microenvironment (TME) can critically regulate tumor progression and therapeutic response. In turn, anticancer treatments may alter the composition and functions of the TME. To investigate this continuous dialog in the context of primary brain cancers, we developed a multimodal longitudinal imaging strategy. We combined macroscopical magnetic resonance imaging with subcellular resolution two-photon intravital microscopy, and leveraged the power of single-cell analysis tools to gain insights into the ongoing interactions between different components of the TME and cancer cells. Our experiments revealed that the migratory behavior of tumor-associated macrophages is different in genetically distinct glioblastomas, and in response to macrophage-targeted therapy. These results underscore the importance of studying cancer longitudinally in an in vivo setting, to reveal complex and dynamic alterations in the TME during disease progression and therapeutic intervention.
Collapse
|
26
|
Regev O, Kizner M, Roncato F, Dadiani M, Saini M, Castro-Giner F, Yajuk O, Kozlovski S, Levi N, Addadi Y, Golani O, Ben-Dor S, Granot Z, Aceto N, Alon R. ICAM-1 on Breast Cancer Cells Suppresses Lung Metastasis but Is Dispensable for Tumor Growth and Killing by Cytotoxic T Cells. Front Immunol 2022; 13:849701. [PMID: 35911772 PMCID: PMC9328178 DOI: 10.3389/fimmu.2022.849701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Breast tumors and their derived circulating cancer cells express the leukocyte β2 integrin ligand Intercellular adhesion molecule 1 (ICAM-1). We found that elevated ICAM-1 expression in breast cancer cells results in a favorable outcome and prolonged survival of breast cancer patients. We therefore assessed the direct in vivo contribution of ICAM-1 expressed by breast cancer cells to breast tumorigenesis and lung metastasis in syngeneic immunocompetent mice hosts using spontaneous and experimental models of the lung metastasis of the C57BL/6-derived E0771 cell line, a luminal B breast cancer subtype. Notably, the presence of ICAM-1 on E0771 did not alter tumor growth or the leukocyte composition in the tumor microenvironment. Interestingly, the elimination of Tregs led to the rapid killing of primary tumor cells independently of tumor ICAM-1 expression. The in vivo elimination of a primary E0771 tumor expressing the ovalbumin (OVA) model neoantigen by the OVA-specific OVA-tcr-I mice (OT-I) transgenic cytotoxic T lymphocytes (CTLs) also took place normally in the absence of ICAM-1 expression by E0771 breast cancer target cells. The whole lung imaging of these cells by light sheet microscopy (LSM) revealed that both Wild type (WT)- and ICAM-1-deficient E0771 cells were equally disseminated from resected tumors and accumulated inside the lung vasculature at similar magnitudes. ICAM-1-deficient breast cancer cells developed, however, much larger metastatic lesions than their control counterparts. Strikingly, the vast majority of these cells gave rise to intravascular tumor colonies both in spontaneous and experimental metastasis models. In the latter model, ICAM-1 expressing E0771- but not their ICAM-1-deficient counterparts were highly susceptible to elimination by neutrophils adoptively transferred from E0771 tumor-bearing donor mice. Ex vivo, neutrophils derived from tumor-bearing mice also killed cultured E0771 cells via ICAM-1-dependent interactions. Collectively, our results are a first indication that ICAM-1 expressed by metastatic breast cancer cells that expand inside the lung vasculature is involved in innate rather than in adaptive cancer cell killing. This is also a first indication that the breast tumor expression of ICAM-1 is not required for CTL-mediated killing but can function as a suppressor of intravascular breast cancer metastasis to lungs.
Collapse
Affiliation(s)
- Ofer Regev
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Marina Kizner
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Francesco Roncato
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Maya Dadiani
- Cancer Research Center, Sheba Medical Center, Ramat-Gan, Israel
| | - Massimo Saini
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Francesc Castro-Giner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Olga Yajuk
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem, Israel
| | - Stav Kozlovski
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Nehora Levi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Shifra Ben-Dor
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem, Israel
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
27
|
Wang G, Wang J, Niu C, Zhao Y, Wu P. Neutrophils: New Critical Regulators of Glioma. Front Immunol 2022; 13:927233. [PMID: 35860278 PMCID: PMC9289230 DOI: 10.3389/fimmu.2022.927233] [Citation(s) in RCA: 11] [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: 04/24/2022] [Accepted: 06/06/2022] [Indexed: 11/22/2022] Open
Abstract
In cancer, neutrophils are an important part of the tumour microenvironment (TME). Previous studies have shown that circulating and infiltrating neutrophils are associated with malignant progression and immunosuppression in gliomas. However, recent studies have shown that neutrophils have an antitumour effect. In this review, we focus on the functional roles of neutrophils in the circulation and tumour sites in patients with glioma. The mechanisms of neutrophil recruitment, immunosuppression and the differentiation of neutrophils are discussed. Finally, the potential of neutrophils as clinical biomarkers and therapeutic targets is highlighted. This review can help us gain a deeper and systematic understanding of the role of neutrophils, and provide new insights for treatment in gliomas.
Collapse
Affiliation(s)
- Guanyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinpeng Wang
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chaoshi Niu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Stereotactic Neurosurgical Institute, Hefei, China
- Anhui Province Key Laboratory of Brain Function and Brain Disease, Hefei, China
- Anhui Provincial Clinical Research Center for Neurosurgical Disease, Hefei, China
| | - Yan Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengfei Wu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Stereotactic Neurosurgical Institute, Hefei, China
- Anhui Province Key Laboratory of Brain Function and Brain Disease, Hefei, China
- Anhui Provincial Clinical Research Center for Neurosurgical Disease, Hefei, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
| |
Collapse
|
28
|
Ali M, Fulci G, Grigalavicius M, Pulli B, Li A, Wojtkiewicz GR, Wang C, Hsieh KLC, Linnoila JJ, Theodossiou TA, Chen JW. Myeloperoxidase exerts anti-tumor activity in glioma after radiotherapy. Neoplasia 2022; 26:100779. [PMID: 35247801 PMCID: PMC8894277 DOI: 10.1016/j.neo.2022.100779] [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: 10/20/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 11/27/2022]
Abstract
Background Host immune response is a critical component in tumorigenesis and immune escape. Radiation is widely used for glioblastoma (GBM) and can induce marked tissue inflammation and substantially alter host immune response. However, the role of myeloperoxidase (MPO), a key enzyme in inflammation and host immune response, in tumorigenesis after radiotherapy is unclear. In this study, we aimed to determine how post-radiation MPO activity influences GBM and outcome. Methods We injected C57BL/6J or MPO-knockout mice with 005 mouse GBM stem cells intracranially. To observe MPO's effects on post-radiation tumor progression, we then irradiated the head with 10 Gy unfractionated and treated the mice with a specific MPO inhibitor, 4-aminobenzoic acid hydrazide (ABAH), or vehicle as control. We performed semi-quantitative longitudinal molecular MRI, enzymatic assays and flow cytometry to assess changes in inflammatory response and tumor size, and tracked survival. We also performed cell culture experiments in murine and human GBM cells to determine the effect of MPO on these cells. Results Brain irradiation increased the number of monocytes/macrophages and neutrophils, and boosted MPO activity by ten-fold in the glioma microenvironment. However, MPO inhibition dampened radiation-induced inflammation, demonstrating decreased MPO-specific signal on molecular MRI and attenuated neutrophil and inflammatory monocyte/macrophage recruitment to the glioma. Compared to saline-treated mice, both ABAH-treated and MPO-knockout mice had accelerated tumor growth and reduced survival. We further confirmed that MPO decreased tumor cell viability and proliferation in cell cultures. Conclusion Local radiation to the brain initiated an acute systemic inflammatory response with increased MPO-carrying cells both in the periphery and the GBM, resulting in increased MPO activity in the tumor microenvironment. Inhibition or absence of MPO activity increased tumor growth and decreased host survival, revealing that elevated MPO activity after radiation has an anti-tumor role.
Collapse
Affiliation(s)
- Muhammad Ali
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; K.G. Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Norway
| | - Giulia Fulci
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mantas Grigalavicius
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Benjamin Pulli
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anning Li
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory R Wojtkiewicz
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Cuihua Wang
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kevin Li-Chun Hsieh
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jenny J Linnoila
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Theodossis A Theodossiou
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - John W Chen
- Institute for Innovation in Imaging and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Mastandrea I, Sher D, Magod P, Friedmann-Morvinski D. Isolation and characterization of the immune cell fraction from murine brain tumor microenvironment. STAR Protoc 2022; 3:101106. [PMID: 35098162 PMCID: PMC8783153 DOI: 10.1016/j.xpro.2021.101106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Ignacio Mastandrea
- The School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Corresponding author
| | - Divsha Sher
- The School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Prerna Magod
- The School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dinorah Friedmann-Morvinski
- The School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Corresponding author
| |
Collapse
|
30
|
Glioblastoma Microenvironment and Cellular Interactions. Cancers (Basel) 2022; 14:cancers14041092. [PMID: 35205842 PMCID: PMC8870579 DOI: 10.3390/cancers14041092] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/31/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary This paper summarizes the crosstalk between tumor/non-tumor cells and other elements of the glioblastoma (GB) microenvironment. In tumor pathology, glial cells result in the highest number of cancers, and GB is considered the most lethal tumor of the central nervous system (CNS). The tumor microenvironment (TME) is a complex peritumoral hallo composed of tumor cells and several non-tumor cells (e.g., nervous cells, stem cells, fibroblasts, vascular and immune cells), which might be a key factor for the ineffective treatment since the microenvironment modulates the biologic status of the tumor with the increase in its evasion capacity. A deeper understanding of cell–cell interactions in the TME and with the tumor cells could be the basis for a more efficient therapy. Abstract The central nervous system (CNS) represents a complex network of different cells, such as neurons, glial cells, and blood vessels. In tumor pathology, glial cells result in the highest number of cancers, and glioblastoma (GB) is considered the most lethal tumor in this region. The development of GB leads to the infiltration of healthy tissue through the interaction between all the elements of the brain network. This results in a GB microenvironment, a complex peritumoral hallo composed of tumor cells and several non-tumor cells (e.g., nervous cells, stem cells, fibroblasts, vascular and immune cells), which might be the principal factor for the ineffective treatment due to the fact that the microenvironment modulates the biologic status of the tumor with the increase in its evasion capacity. Crosstalk between glioma cells and the brain microenvironment finally inhibits the beneficial action of molecular pathways, favoring the development and invasion of the tumor and its increasing resistance to treatment. A deeper understanding of cell–cell interactions in the tumor microenvironment (TME) and with the tumor cells could be the basis for a more efficient therapy.
Collapse
|
31
|
Anisman H, Kusnecov AW. Cancer biology and pathology. Cancer 2022. [DOI: 10.1016/b978-0-323-91904-3.00004-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|