Immune Microenvironment of Gliomas

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Immunology".

Deadline for manuscript submissions: closed (31 July 2021) | Viewed by 25099

Special Issue Editors


E-Mail Website
Guest Editor
Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02-093 Warsaw, Poland

E-Mail Website
Guest Editor
1. Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
2. IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Italy

Special Issue Information

Dear Colleagues,

Malignant gliomas are rapidly progressing, incurable tumors of the central nervous system (CNS). Numerous studies of human and experimental rodent gliomas have revealed considerable heterogeneity in the tumor microenvironment, which is composed of reactive astrocytes, endothelial cells, and numerous immune cells. Infiltrating immune cells mostly consist of glioma-associated microglia and macrophages (GAMs), myeloid-derived suppressor cells (MDSCs), granulocytes, and T lymphocytes. Myeloid cells have a few intact innate immune functions, but instead of initiating antitumor immune responses, they are reprogrammed and secrete proinvasive and immunosuppressive molecules, supporting tumor growth and enabling effective antitumor T cell responses. Findings from linage tracing and single cell RNA sequencing studies have revealed cell type heterogeneity among immune infiltrates, a significant contribution of peripheral myeloid cells to GAMs, and specific functional responses of discrete subpopulations. Accumulating MDSCs inhibit cytotoxic responses mediated by natural killer cells and block the activation of tumor-reactive CD4+ T helper cells and cytotoxic CD8+ T cells. Malignant gliomas induce various states of T-cell dysfunction, resulting in decreased proliferative capacity and effector function. The presence of regulatory T cells contributes to the lack of effective immune activation against malignant gliomas. The complexity of cell-to-cell interactions in the glioma microenvironment is far from well understood. Identification of those mediators may facilitate the development of immunotherapies for gliomas, as immunomodulatory and immune evasion mechanisms employed by malignant gliomas are listed as the main obstacles to glioma immunotherapy. Tumor antigenic heterogeneity, an immunosuppressive microenvironment, unique properties of the CNS that limit T cell entry, and potential immune‐based toxicities impede the development of effective immunotherapies for gliomas. The low antigenicity of gliomas and the low abundance and perivascular location of CD8 T cells may further contribute to the limited clinical efficacy of checkpoint inhibitors in glioma patients. In this Special Issue, we will summarize and present new findings related to the heterogeneity and complexity of interactions between tumor and immune components in the glioma microenvironment.

Prof. Bozena Kaminska
Prof. Cristina Limatola
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Published Papers (5 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

18 pages, 3898 KiB  
Article
The Presence and Potential Role of ALDH1A2 in the Glioblastoma Microenvironment
by Stephanie Sanders, Denise M. Herpai, Analiz Rodriguez, Yue Huang, Jeff Chou, Fang-Chi Hsu, Darren Seals, Ryan Mott, Lance D. Miller and Waldemar Debinski
Cells 2021, 10(9), 2485; https://doi.org/10.3390/cells10092485 - 20 Sep 2021
Cited by 6 | Viewed by 3163
Abstract
Glioblastoma (GBM) is the most aggressive malignant glioma. Therapeutic targeting of GBM is made more difficult due to its heterogeneity, resistance to treatment, and diffuse infiltration into the brain parenchyma. Better understanding of the tumor microenvironment should aid in finding more effective management [...] Read more.
Glioblastoma (GBM) is the most aggressive malignant glioma. Therapeutic targeting of GBM is made more difficult due to its heterogeneity, resistance to treatment, and diffuse infiltration into the brain parenchyma. Better understanding of the tumor microenvironment should aid in finding more effective management of GBM. GBM-associated macrophages (GAM) comprise up to 30% of the GBM microenvironment. Therefore, exploration of GAM activity/function and their specific markers are important for developing new therapeutic agents. In this study, we identified and evaluated the expression of ALDH1A2 in the GBM microenvironment, and especially in M2 GAM, though it is also expressed in reactive astrocytes and multinucleated tumor cells. We demonstrated that M2 GAM highly express ALDH1A2 when compared to other ALDH1 family proteins. Additionally, GBM samples showed higher expression of ALDH1A2 when compared to low-grade gliomas (LGG), and this expression was increased upon tumor recurrence both at the gene and protein levels. We demonstrated that the enzymatic product of ALDH1A2, retinoic acid (RA), modulated the expression and activity of MMP-2 and MMP-9 in macrophages, but not in GBM tumor cells. Thus, the expression of ALDH1A2 may promote the progressive phenotype of GBM. Full article
(This article belongs to the Special Issue Immune Microenvironment of Gliomas)
Show Figures

Figure 1

24 pages, 2739 KiB  
Article
Microglia-Derived Small Extracellular Vesicles Reduce Glioma Growth by Modifying Tumor Cell Metabolism and Enhancing Glutamate Clearance through miR-124
by Carmela Serpe, Lucia Monaco, Michela Relucenti, Ludovica Iovino, Pietro Familiari, Ferdinando Scavizzi, Marcello Raspa, Giuseppe Familiari, Laura Civiero, Igea D’Agnano, Cristina Limatola and Myriam Catalano
Cells 2021, 10(8), 2066; https://doi.org/10.3390/cells10082066 - 12 Aug 2021
Cited by 28 | Viewed by 3113
Abstract
Brain homeostasis needs continuous exchange of intercellular information among neurons, glial cells, and immune cells, namely microglial cells. Extracellular vesicles (EVs) are active players of this process. All the cells of the body, including the brain, release at least two subtypes of EVs, [...] Read more.
Brain homeostasis needs continuous exchange of intercellular information among neurons, glial cells, and immune cells, namely microglial cells. Extracellular vesicles (EVs) are active players of this process. All the cells of the body, including the brain, release at least two subtypes of EVs, the medium/large EVs (m/lEVs) and small EVs (sEVs). sEVs released by microglia play an important role in brain patrolling in physio-pathological processes. One of the most common and malignant forms of brain cancer is glioblastoma. Altered intercellular communications constitute a base for the onset and the development of the disease. In this work, we used microglia-derived sEVs to assay their effects in vitro on murine glioma cells and in vivo in a glioma model on C57BL6/N mice. Our findings indicated that sEVs carry messages to cancer cells that modify glioma cell metabolism, reducing lactate, nitric oxide (NO), and glutamate (Glu) release. sEVs affect Glu homeostasis, increasing the expression of Glu transporter Glt-1 on astrocytes. We demonstrated that these effects are mediated by miR-124 contained in microglia-released sEVs. The in vivo benefit of microglia-derived sEVs results in a significantly reduced tumor mass and an increased survival of glioma-bearing mice, depending on miR-124. Full article
(This article belongs to the Special Issue Immune Microenvironment of Gliomas)
Show Figures

Graphical abstract

Review

Jump to: Research

21 pages, 1242 KiB  
Review
Single-Cell Omics in Dissecting Immune Microenvironment of Malignant Gliomas—Challenges and Perspectives
by Bozena Kaminska, Natalia Ochocka and Pawel Segit
Cells 2021, 10(9), 2264; https://doi.org/10.3390/cells10092264 - 31 Aug 2021
Cited by 22 | Viewed by 5697
Abstract
Single-cell technologies allow precise identification of tumor composition at the single-cell level, providing high-resolution insights into the intratumoral heterogeneity and transcriptional activity of cells in the tumor microenvironment (TME) that previous approaches failed to capture. Malignant gliomas, the most common primary brain tumors [...] Read more.
Single-cell technologies allow precise identification of tumor composition at the single-cell level, providing high-resolution insights into the intratumoral heterogeneity and transcriptional activity of cells in the tumor microenvironment (TME) that previous approaches failed to capture. Malignant gliomas, the most common primary brain tumors in adults, are genetically heterogeneous and their TME consists of various stromal and immune cells playing an important role in tumor progression and responses to therapies. Previous gene expression or immunocytochemical studies of immune cells infiltrating TME of malignant gliomas failed to dissect their functional phenotypes. Single-cell RNA sequencing (scRNA-seq) and cytometry by time-of-flight (CyTOF) are powerful techniques allowing quantification of whole transcriptomes or >30 protein targets in individual cells. Both methods provide unprecedented resolution of TME. We summarize the findings from these studies and the current state of knowledge of a functional diversity of immune infiltrates in malignant gliomas with different genetic alterations. A precise definition of functional phenotypes of myeloid and lymphoid cells might be essential for designing effective immunotherapies. Single-cell omics studies have identified crucial cell subpopulations and signaling pathways that promote tumor progression, influence patient survival or make tumors vulnerable to immunotherapy. We anticipate that the widespread usage of single-cell omics would allow rational design of oncoimmunotherapeutics. Full article
(This article belongs to the Special Issue Immune Microenvironment of Gliomas)
Show Figures

Graphical abstract

14 pages, 1105 KiB  
Review
The Interplay between Glioblastoma and Its Microenvironment
by Mark Dapash, David Hou, Brandyn Castro, Catalina Lee-Chang and Maciej S. Lesniak
Cells 2021, 10(9), 2257; https://doi.org/10.3390/cells10092257 - 31 Aug 2021
Cited by 52 | Viewed by 7865
Abstract
GBM is the most common primary brain tumor in adults, and the aggressive nature of this tumor contributes to its extremely poor prognosis. Over the years, the heterogeneous and adaptive nature of GBM has been highlighted as a major contributor to the poor [...] Read more.
GBM is the most common primary brain tumor in adults, and the aggressive nature of this tumor contributes to its extremely poor prognosis. Over the years, the heterogeneous and adaptive nature of GBM has been highlighted as a major contributor to the poor efficacy of many treatments including various immunotherapies. The major challenge lies in understanding and manipulating the complex interplay among the different components within the tumor microenvironment (TME). This interplay varies not only by the type of cells interacting but also by their spatial distribution with the TME. This review highlights the various immune and non-immune components of the tumor microenvironment and their consequences f the efficacy of immunotherapies. Understanding the independent and interdependent aspects of the various sub-populations encapsulated by the immune and non-immune components will allow for more targeted therapies. Meanwhile, understanding how the TME creates and responds to different environmental pressures such as hypoxia may allow for other multimodal approaches in the treatment of GBM. Ultimately, a better understanding of the GBM TME will aid in the development and advancement of more effective treatments and in improving patient outcomes. Full article
(This article belongs to the Special Issue Immune Microenvironment of Gliomas)
Show Figures

Figure 1

17 pages, 1342 KiB  
Review
Immune Microenvironment Landscape in CNS Tumors and Role in Responses to Immunotherapy
by Hinda Najem, Mustafa Khasraw and Amy B. Heimberger
Cells 2021, 10(8), 2032; https://doi.org/10.3390/cells10082032 - 09 Aug 2021
Cited by 11 | Viewed by 4333
Abstract
Despite the important evolution of immunotherapeutic agents, brain tumors remain, in general, refractory to immune therapeutics. Recent discoveries have revealed that the glioma microenvironment includes a wide variety of immune cells in various states that play an important role in the process of [...] Read more.
Despite the important evolution of immunotherapeutic agents, brain tumors remain, in general, refractory to immune therapeutics. Recent discoveries have revealed that the glioma microenvironment includes a wide variety of immune cells in various states that play an important role in the process of tumorigenesis. Anti-tumor immune activity may be occurring or induced in immunogenic hot spots or at the invasive edge of central nervous system (CNS) tumors. Understanding the complex heterogeneity of the immune microenvironment in gliomas will likely be the key to unlocking the full potential of immunotherapeutic strategies. An essential consideration will be the induction of immunological effector responses in the setting of the numerous aspects of immunosuppression and evasion. As such, immune therapeutic combinations are a fundamental objective for clinical studies in gliomas. Through immune profiling conducted on immune competent murine models of glioma and ex vivo human glioma tissue, we will discuss how the frequency, distribution of immune cells within the microenvironment, and immune modulatory processes, may be therapeutically modulated to lead to clinical benefits. Full article
(This article belongs to the Special Issue Immune Microenvironment of Gliomas)
Show Figures

Figure 1

Back to TopTop