Next Article in Journal
MutaPT: A Multi-Task Pre-Trained Transformer for Classifying State of Disorders of Consciousness Using EEG Signal
Previous Article in Journal
Age-Based Developmental Biomarkers in Eye Movements: A Retrospective Analysis Using Machine Learning
Previous Article in Special Issue
The Diverse Roles of Reactive Astrocytes in the Pathogenesis of Amyotrophic Lateral Sclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neuroinflammation in Glioblastoma: Progress and Perspectives

Department of Pathology, School of Medicine, South China University of Technology, Guangzhou 510006, China
*
Author to whom correspondence should be addressed.
Brain Sci. 2024, 14(7), 687; https://doi.org/10.3390/brainsci14070687
Submission received: 10 May 2024 / Revised: 25 June 2024 / Accepted: 7 July 2024 / Published: 9 July 2024
(This article belongs to the Special Issue Advances in Neuroinflammation)

Abstract

:
Glioblastoma is the most common and malignant primary brain tumor, with high morbidity and mortality. Despite an aggressive, multimodal treatment regimen, including surgical resection followed by chemotherapy and radiotherapy, the prognosis of glioblastoma patients remains poor. One formidable challenge to advancing glioblastoma therapy is the complexity of the tumor microenvironment. The tumor microenvironment of glioblastoma is a highly dynamic and heterogeneous system that consists of not only cancerous cells but also various resident or infiltrating inflammatory cells. These inflammatory cells not only provide a unique tumor environment for glioblastoma cells to develop and grow but also play important roles in regulating tumor aggressiveness and treatment resistance. Targeting the tumor microenvironment, especially neuroinflammation, has increasingly been recognized as a novel therapeutic approach in glioblastoma. In this review, we discuss the components of the tumor microenvironment in glioblastoma, focusing on neuroinflammation. We discuss the interactions between different tumor microenvironment components as well as their functions in regulating glioblastoma pathogenesis and progression. We will also discuss the anti-tumor microenvironment interventions that can be employed as potential therapeutic targets.

1. Introduction

Glioblastoma, also known as glioblastoma multiforme (GBM), is the most common and lethal primary brain tumor in adults, with an aggressive nature and poor treatment response [1,2]. Despite a multimodal treatment regimen including surgical resection, radiation, and chemotherapy, the prognosis of glioblastoma patients remains poor, with a median survival of only 12–15 months [3,4]. One formidable challenge in advancing glioblastoma therapy is the complexity of the tumor microenvironment (TME) [3,4,5,6]. The TME of glioblastoma is a highly dynamic and heterogeneous system that not only consists of cancerous cells but also various types of non-cancerous cells, the predominant part of which are resident or infiltrating inflammatory cells [7,8,9,10]. Over the past decades, the heterogeneous nature of glioblastoma has been extensively studied and regarded as a key factor in the poor treatment efficacy of the disease. However, most of these studies are cancer cell-centric, which may underestimate the role of the tumor microenvironment, especially neuroinflammation, in glioblastoma pathogenesis and progression.
Neuroinflammation is the inflammatory response of the brain characterized by the infiltration of various immune cells and the release of inflammation-related cytokines, chemokines, and growth factors. In glioblastoma, neuroinflammation also has the typical features of enhanced vascularization, hypoxic tumor microenvironment, and immune-suppressive milieu. All these characteristics, together with the presence of the blood–brain barrier, make the neuroinflammatory microenvironment in glioblastoma a unique pathologic process. Accumulating evidence has suggested that the neuroinflammatory microenvironment plays an important role in glioblastoma progression, invasion, and treatment response [11,12,13]. It can affect the biological behavior of tumor cells directly through inflammation–tumor cell interactions or indirectly via the release of related cytokines, chemokines, and growth factors. Neuroinflammation has increasingly been recognized as a key player and potential therapeutic target in glioblastoma [11,14,15]. A deeper understanding of the inflammatory microenvironment in glioblastoma and its interactions with the cancer cells could provide the basis for more efficient therapies.
In this review, we discuss the known and emerging concepts related to the role of the tumor microenvironment in glioblastoma carcinogenesis and progression, focusing on neuroinflammation. We discuss the components of the tumor microenvironment in glioblastoma, especially neuroinflammation-related components, and their impacts on tumor invasion and progression. We also review the anti-tumor microenvironment interventions that can potentially be employed as therapeutic targets in glioblastoma.

2. The Blood–Brain Barrier and Vasculature in Glioblastoma

Anatomically, the human brain is protected by a natural membrane called the blood–brain barrier (BBB), which is composed of endothelial cells connected by tight junctions and surrounded by pericytes, astrocytes, and the basement membrane [16,17]. Under normal conditions, the protective BBB has a major role in maintaining normal brain function by preventing toxins and pathogens from entering the brain through circulation [16,17]. The traditional belief even holds that the human brain is immunologically privileged since the BBB is impermeable to immune cells. This perceived dogma has recently been changed with the breakthrough findings that the human brain actually possesses a conventional and functional lymphatic system like other organs [18,19]. Moreover, it has been increasingly accepted that the integrity of BBB is actually compromised under pathological conditions, e.g., tumor-associated inflammation in glioblastoma, which leads to increased permeability of BBB and the infiltration of inflammatory cells into the brain [20,21].
These changes, together with other factors, e.g., the rapid growth of glioblastoma cells, contribute greatly to the proliferation of microvasculature, one of the most characterized pathologic features of glioblastoma [22]. A variety of angiogenic factors and chemokines have been described as being involved in the formation of vasculature in glioblastoma; the elevated level of vascular endothelial growth factor (VEGF) is regarded as the predominant one [22]. Additionally, hypoxia-inducible factor (HIF), which can actually enhance the expression of VEGF, is also reported to be a key player in vessel formation [22]. However, it is important to note that the vasculature in glioblastoma is poorly organized, hyper-dilated, and has leaky vessels. This abnormal vasculature leads to the leaking of blood components into tumor tissues and also attracts inflammatory cells, which release proangiogenic factors, thereby further enhancing the endothelial cell proliferation in glioblastoma TME.

3. Composition of Tumor Microenvironment in Glioblastoma

It is now widely accepted that the tumor microenvironment (TME) of glioblastoma is a complex and dynamic system consisting of both non-cellular and cellular components [5,7,8,9,10,11,23] (Figure 1). The non-cellular components include an extracellular matrix (ECM) in which cells are embedded and various soluble factors (e.g., growth factors, cytokines, and chemokines). The cellular components include stromal cells and inflammatory cells, and inflammatory cells are predominant. It was reported that inflammatory cells in glioblastoma TME can constitute up to 30–50% of the tumor mass, which consists of resident microglia, infiltrated macrophages, and less abundant lymphoid T cells, NK cells, dendritic cells, and neutrophils. The inflammatory cells co-exist and interact with cancer cells, stromal cells, and non-cellular components, which shape the glioblastoma TME in both direct and indirect ways. The TME, especially neuroinflammation, has been regarded as the new therapeutic target for glioblastoma treatment [5,7,8,9,10,11,12,13,23,24,25].

3.1. Non-Cellular Components of TME in Glioblastoma

Extracellular matrix (ECM). The ECM is the non-cellular component that provides an important physical scaffold for all the cellular components embedded in it. However, the ECM in the brain is unique and different from the ECM normally found in many other tissues. The ECM of the brain is largely composed of hyaluronic acid (HA), proteoglycans, and glycoproteins and demonstrates a mesh-like appearance as compared with the fibrous ECM in other tissues [26]. In the case of glioblastoma, the components of the ECM change, which can be physically reflected in the increased stiffness of TME in glioblastoma [27]. Recent studies have shown that some ECM components (e.g., HA) increase in glioblastoma tissue as compared to non-tumor tissue and contribute to the increased mobility and invasiveness of glioblastoma cells [28].
Soluble molecular chemicals. For the soluble chemical components, a variety of inflammation mediators, including cytokines, chemokines, and growth factors, have been identified in the TME of glioblastoma and are involved in various signaling pathways [29]. Cytokines are signaling proteins secreted from specific immune cells, and the action modes of the cytokines include pro-inflammatory functions (e.g., IL-6, IL-8, and TNF-α) and anti-inflammatory functions (e.g., IL-4, IL-10, and TGF-β). Chemokines are small proteins that serve to mediate the migration of different cell types throughout the body. The chemokines that are highly expressed in the TME of glioblastoma have been characterized by CXCL2, IL-8, and CCL2, which can promote tumor invasion via facilitating cell proliferation, tumor growth, and angiogenesis. Additionally, tumor acidosis and low oxygen concentration are also important hallmarks of glioblastoma TME [30].

3.2. Cellular Components of TME in Glioblastoma

3.2.1. Stromal Cells

In glioblastoma TME, the stromal cells consist of astrocytes, neurons, and vascular endothelial cells. There is growing interest in the study of glioblastomas to communicate with astrocytes and neurons [31]. Among them, astrocytes have been found to undergo reactive astrogliosis during the growth of the tumor, which could further contribute to tumor cell infiltration. These tumor-associated reactive astrocytes have also been characterized as regulating the immune environment in glioblastoma [32,33]. The neurons have also been found in the pathologic process of glioblastoma, and they may interact with glioblastoma cells via paracrine stimulation, synaptic transmission, and some other indirect means [34]. For the endothelial cells, as mentioned above, glioblastoma is one of the most vascularized malignancies with extensive endothelial cell proliferation and even the formation of glomerular structures [22,35]. Various angiogenic factors have been reported to contribute to this hallmark, including the hypoxic tumor environment, which leads to highly elevated expressions of vascular endothelial growth factor (VEGF) and, therefore, proliferation of the endothelial cells. However, these newly formed microvasculature are abnormal blood vessels and cannot provide enough blood flow and oxygen to the tumor tissue, which will accelerate the necrosis of tumor tissues, another hallmark of glioblastoma.

3.2.2. Inflammatory Cells

Microglia/Macrophages. Microglia and macrophages are collectively referred to as tumor-associated macrophages (TAMs), which account for 30% of the total tumor volume and are the predominant inflammatory cell populations in glioblastoma [32]. Microglial cells are the resident myeloid cells in the brain, while macrophages are infiltrating monocytes derived from the peripheral blood due to the breakdown of the BBB under pathological conditions, e.g., tumors. Microglia and macrophages have similar functions and are difficult to differentiate in most cases. However, some studies reported that they have different localization sites within the tumor tissue: resident microglia are typically found at the tumor periphery, while infiltrating macrophages tend to be more enriched in the tumor core. Recent studies reported the employment of single-cell sequencing to precisely differentiate these two cell subpopulations [36].
According to their phenotype and function, TAMs are further classified into two subtypes: the pro-inflammatory subtype (M1 macrophages) and the anti-inflammatory subtype (M2 macrophages). M1 macrophages exhibit immune-supportive and anti-tumoral functions, while M2 macrophages have immune-suppressive and pro-tumoral functions [37]. The dual function of TAMs in glioblastoma pathogenesis and progression has been a robustly debated topic in the neuroinflammation field [37,38]. The acquisition of the M1 or M2 phenotype depends on the cytokines expressed in TME. It has been found that the M1 phenotype is acquired after being stimulated with pro-inflammatory factors, such as Toll-like receptor 4 (TLR-4) ligands and interferon-gamma (IFN-γ), and then eliminates tumor cells by producing inflammatory factors (e.g., TNF-a) [39,40]. On the other hand, the M2 phenotype is triggered after receiving stimulation with anti-inflammatory factors, for example, IL-4 and IL-10. M2-phenotype TAMs show less cytotoxicity in tumor cells by producing anti-inflammatory factors (e.g., TGF-β) and are associated with promoting tumor growth. However, it should be noted that the phenotypes of TAMs are dynamic, and as the tumor progresses, the M1 and M2 phenotypes can switch to each other [30,31]. Due to their dominant number, TAMs have been regarded as promising therapeutic targets for glioblastoma treatment. The cytokines that can contribute to TAM infiltration have been characterized, including colony-stimulating factor-1 (CSF-1), CCL2, and CCL5. Moreover, multiple immunotherapy approaches that could target TAMs have been tried [37], which is detailed in the Section 4 below.
T cells. T cells are also an important population of inflammatory cells in TME of glioblastoma, in spite of the fact that they constitute only a small proportion of the total cell numbers in TME (~0.25%) [41]. Of particular importance are the regulatory T cells (Tregs). Tregs are a unique population of CD4+ T cells that can regulate the overall immune homeostasis in an immunosuppressive manner [42,43]. In glioblastoma, Tregs inhibit the anti-tumor response and promote tumor-killing tolerance by secreting immunomodulatory cytokines (e.g., TGF-β and IL-10). This will, in turn, inhibit the production of anti-tumor cytokines, such as IL-2 and IFN-γ, leading to a decrease in effector cells necessary to control tumor growth. Tregs are reported to be recruited to the TME of glioblastoma by specific cytokines such as CXCR3 and CCR5, which can be secreted by glioblastoma cells and innate immune cells within the brain. In addition to Tregs, CD8+ cytotoxic T cells are another important subpopulation of T cells in glioblastoma, which can induce a tumor-killing effect and mediate tumor regression like natural killer cells. Various immunotherapy efforts have been tried to boost the cytotoxic CD8+ T cell function to treat glioblastoma, such as immune checkpoint inhibitors and CAR-T cell therapy [44,45,46]. However, these therapies are generally less efficacious in glioblastoma as compared to other malignancies due to the relatively low number of tumor-infiltrating T cells in the TME of glioblastoma.
Natural killer cells. Natural killer (NK) cells have also been characterized as an important part of inflammatory cells in glioblastoma TME. Although they account for a relatively small proportion (~2% of total infiltrating inflammatory cells) like T cells, NK cells are critical for the anti-tumor immune response in glioblastoma [47]. NK cells can not only provoke tumor cell apoptosis through their direct natural cytotoxicity (e.g., granzyme B and perforin); they can also control tumor growth via secreting cytokines or regulating the activity of other inflammatory cells. For example, NK cells have been demonstrated to be able to regulate T cell-mediated immune responses by maintaining the function of dendritic cells and promoting tumor antigen presentation. On the other hand, NK cells can also be regulated by the TME. For example, glioblastoma cells express transforming growth factor (TGF-β), which can inhibit the activation of NK cell function. Glioblastoma cells can also express unique MHC-I molecules to inhibit the function of NK cells by acting as inhibitory receptor ligands [48]. Therefore, although glioblastoma is often infiltrated by NK cells, these NK cells are functionally inhibited by glioblastoma cells and TME.
Dendritic cells. Dendritic cells (DCs) are a class of professional antigen-processing and presenting cells that play key roles in cancer immunity [49]. Similar to NK cells, DCs are recruited to glioblastoma via specific chemokines such as CXCL1 and CCL5. It has been shown that DCs can produce anti-tumor cytokines (e.g., IL-12), which in turn recruit more CD8+ T cells [50]. Preclinical studies have shown that the activation of DCs can improve long-term tumor survival in the mouse model of glioblastoma [50]. Clinical studies of DC vaccines in glioblastoma patients have also shown some efficacy in improving the median overall survival [50]. However, it remains to be elucidated for the standardization of DC vaccine therapy, e.g., the antigens used and the injection sites [48]. Therefore, future work on improving the efficacy of DC-based therapy in more clinical trials is needed [51].
Neutrophil cells. Neutrophil cells are the most abundant population of granulocytes in the human body, which account for approximately 70% of the total number of white blood cells. In glioblastoma, neutrophils are observed to be negatively correlated with the prognosis of glioblastoma patients [52,53,54]. Neutrophils are commonly found in the center area of the glioblastoma tumor bulk and aid in tumor progression and angiogenesis. Neutrophils are attracted to the TME core by specific chemokines, e.g., CXCL8 and IL-8. They can also promote tumor proliferation and angiogenesis by secreting elastase. Recently, it was found that neutrophils are involved in the proliferation and invasion of glioblastoma cells by activating the NF-κB signaling. Additionally, there was a positive feedback loop between IL-8 expression and neutrophil infiltration into tumor sites [55]. In glioma, it was also found that the neutrophil-to-lymphocyte ratio (NLR) in the peripheral blood was positively associated with tumor grading, in which an increase in NLR may indicate a higher tumor grade and poorer patient outcomes. Moreover, compared with traditional molecular prognostic markers, e.g., IDH1 mutations, NLR can better evaluate the prognosis of glioblastoma patients and guide the treatment regimen.

4. Anti-TME Intervention for the Therapy of Glioblastoma

The functional role of TME, especially neuroinflammation, in the pathogenesis and tumor progression of glioblastoma makes anti-TME intervention a major novel therapy strategy for glioblastoma treatment [12,56,57,58,59]. Currently, there are two main types of TME-based therapy for glioblastoma: anti-vasculature therapy and neuroinflammation-based therapy. The latter could be further classified into four strategies: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell (CAR-T) therapies, vaccines, and oncolytic viruses (OVs) [57,58,59,60,61,62,63,64,65,66]. Additionally, a combined multimodal therapy of these different strategies is also extensively studied. In the following, we will provide an overview of each of these therapeutic approaches in the clinical setting for glioblastoma treatment (Table 1, Table 2, Table 3 and Table 4).

4.1. Anti-Vasculature Therapy

Due to the hallmark of microvascular proliferation in glioblastoma, anti-vasculature has become one of the most studied therapy approaches. A series of clinical trials have been performed to test the effectiveness of anti-vasculature therapy in glioblastoma [22,35]. The majority of these studies focus on blocking the VEGF/VEGFR signaling pathway, either through a monoclonal antibody against VEGF or with small-molecule inhibitors against VEGFR. For example, VEGF inhibition with bevacizumab, a humanized monoclonal antibody targeting VEGF, has shown effects in improving glioblastoma patients’ survival [67,68,69]. Moreover, it was found that anti-VEGF therapy can decrease vasogenic brain edema and improve blood perfusion and subsequent oxygenation, which creates conditions for better drug delivery and the efficacy of other treatments. It can also decrease the immune suppression in glioblastoma TME. Therefore, there are some strategies to combine anti-VEGF therapy with other treatment regimens, such as combining anti-vasculature therapy with immune-based approaches.
Overall, however, anti-VEGF therapy has benefitted only a subset of glioblastoma patients; the outcome in most anti-VEGF studies failed to demonstrate the benefit in patient survival [70,71,72]. There are several underlying reasons for this treatment failure. One of the major problems is the inefficient drug delivery to the tumor, which is frequent in almost all types of therapeutics for brain disease because of the BBB. Other reasons include the existence of VEGF-independent angiogenesis, such as neoangiogenesis through the CXCR4/CXCL12 axis [73]. Therefore, efforts that target angiogenesis through different action mechanisms will be helpful to increase treatment efficacy. Further analysis also revealed that the effects of anti-VEGF therapy may be dependent on the glioblastoma genetic subtypes, e.g., IDH1 mutation status, suggesting the necessity of patient subtype stratification before clinical trials [74]. Table 1 lists some of the completed phase II or III clinical trials of anti-vasculature therapy for the treatment of glioblastoma.

4.2. Neuroinflammation-Based Therapy

4.2.1. Immune Checkpoint Inhibitors

Immune checkpoint inhibitors (ICIs) primarily refer to the monoclonal antibodies that can target the cell immune checkpoints and allow for a more robust anti-tumor effect [75]. The majority of studies on ICIs have focused on programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte antigen 4 (CTLA-4), which are all important proteins of immune checkpoint pathways [76]. PD-1 is a cell surface protein of T cells and normally acts as a T cell checkpoint that keeps T cells from attacking tumor cells by binding with PD-L1. The use of PD-1 inhibitors has led to increased survival in patients with various tumor types, including glioblastoma [75,76,77]. For example, in neoadjuvant anti-PD-1 immunotherapy (pembrolizumab) in recurrent glioblastoma, satisfactory results were observed in the overall survival of patients receiving neoadjuvant pembrolizumab compared to the control group [78]. Moreover, functional activation of tumor-infiltrating T lymphocytes was observed, and interferon responses were induced in TME.
In the majority of clinical trials applying the anti-PD-1 antibody to recurrent glioblastoma patients, however, very limited efficacy in improving patients’ survival was observed. Generally, the anti-tumor effects of ICI treatment are less efficacious in glioblastoma than in other malignancies (e.g., melanoma). For example, in a phase 3 trial for a PD-1 inhibitor, although the safety of the treatment was found to be consistent with that in other tumor types, no clinical benefit was observed [79]. The scarcity of T cells in glioblastoma TME is a potential reason because the existence of infiltrating T cells in TME is fundamental to the success of ICI treatment [80]. With continuous efforts in this field, e.g., the characterization of new checkpoints and the combination of ICI treatment with other treatment regimens, the treatment efficacy of ICIs may be improved. Table 2 lists some of the completed phase II or III clinical trials of ICI therapy for the treatment of glioblastoma.

4.2.2. Vaccine Therapy

Vaccine therapy utilizes one or multiple tumor-associated antigens to stimulate anti-tumor effects and has been extensively studied in multiple malignancies, including glioblastoma [81,82,83]. There are several types of tumor vaccines that are being used in cancer treatment, and the peptide-based vaccine and the dendritic cell (DC)-based vaccine are two main strategies for glioblastoma [84,85]. Peptide-based vaccines are vaccines developed based on short peptides that have epitopes with cancer cells and can act as antigenic targets to induce effective anti-tumor responses. Peptide-based vaccines have simple structures and are relatively easy to manipulate. Moreover, they have relatively lower variability as compared to other vaccines. One of the most frequently used tumor-associated antigens for peptide vaccines in glioblastoma is EGFRvIII, a deletion mutation found in approximately 20% of glioblastoma patients [84]. Dendritic cell (DC)-based vaccines employ DCs primed with whole tumor cell lysates or tumor-associated antigens to stimulate the adaptive immune system and, therefore, to control the growth of tumors. Currently, both peptide- and DC-based vaccines have been investigated in clinical trials in glioblastoma patients and represent attractive approaches for the immunotherapy of glioblastoma [82,83,84,85,86]. For example, in a phase II clinical trial of the EGFRvIII peptide vaccine study, a substantial increase in patient survival was observed [87]. For the DC-based vaccines, preclinical studies have demonstrated promising results for vaccine treatment in combination with PD-1 inhibitors. Moreover, a phase I clinical study revealed that the combination of DC-based vaccine therapy with TMZ is safe and tolerable in glioblastoma patients. Additionally, in phase III clinical trials in newly diagnosed and recurrent glioblastoma patients, it was found that autologous tumor lysate-loaded DCs could extend the patients’ survival [88].
While vaccine therapy presents an attractive method for glioblastoma therapy, disappointing results remain. In a phase III clinical trial that used the EGFRvIII peptide vaccine (rindopepimut), the results did not show a significant improvement in patient survival [89]. The potential reason is the heterogeneous and unstable expression of EGFRvIII in glioblastoma cells, which leads to the outgrowth of tumor cells that lack this antigen. Future studies are needed to reveal the effectiveness of this treatment and its impact on the overall survival of glioblastoma patients. Table 3 lists some of the completed phase II or III clinical trials of vaccine therapy for the treatment of glioblastoma.

4.2.3. CAR-T Cell Therapy

CAR-T cell therapy is a novel type of immunotherapy in which T cells are modified to bind chimeric antigen receptors (CARs) to increase their ability to recognize and target tumor cells [90,91,92]. Currently, clinical trials employing CAR-T cell therapy in cancer treatment have shown safety and encouraging results, especially in hematologic malignancies. The effectiveness of CAR-T cell therapy in treating solid malignancies, e.g., glioblastoma, has also been identified [90,92,93]. There are three commonly used antigens for CAR-T cell therapy in glioblastoma: EGFRvIII, HER2, and IL-13 receptor (IL-13R) [90,91,92]. For example, a phase I clinical trial targeting EGFRvIII CAR-T cell therapy demonstrated that the intravenous administration could transfer the CAR-T cells to the brain tumor site and that the EGFRvIII level on glioblastoma tumor cells was reduced by the CAR-T cell treatment [94]. Moreover, this study also demonstrated the effects of CAR-T cell therapy on improving the immunosuppressive tumor environment, indicating the promising perspective of combinational therapy of CAR-T cell therapy with other treatment approaches. In another phase I clinical trial employing IL13R-targeted CAR-T cell therapy, it was found that the intracranial injection of CAR-T cells can achieve an anti-tumor effect in glioblastoma treatment [95]. In a phase I study employing HER2-specific CAR-T cell therapy, of the 17 recruited glioblastoma patients, only 8 demonstrated clinical benefits in overall survival [96].
As for the confounding and limited effects of CAR-T cell therapy in glioblastoma, one major problem is the heterogeneity of target-antigen expressions in tumor cells, which finally leads to heterogeneous treatment effects. Another substantial issue is how to maximize and maintain the activity of the injected CAR-T cells. It has been reported that CAR-T cell administration can induce immunosuppressive responses in the brain [94]. Therefore, successful treatment needs the development of engineered CAR-T cells that are resistant to immunosuppression. Table 4 lists some of the completed phase II or III clinical trials of CAR-T cell therapy for the treatment of glioblastoma.

4.2.4. Oncolytic Virus Therapy

Oncolytic virus (OV) is able to infect cancer cells to present tumor-associated antigens and then lyse the tumor cells. Moreover, it was found that the cellular proteins released from the OV-lysed tumor cells can activate the anti-tumor immune response in multiple ways. For example, viruses can activate macrophages, and activated macrophages can enhance the infiltration of T cells into TME and, therefore, improve the immunosuppressive characteristic of glioblastoma. Therefore, OV therapy is becoming a very promising approach for the treatment of malignancies [97,98]. In glioblastoma, the effects of OV on tumor-killing have also been widely studied [98,99,100,101,102]. Multiple types of viruses are being tested for OV therapy, including retrovirus, adenovirus, herpes simplex virus, poliovirus, and measles virus [97,98]. In 2018, the recombinant oncolytic poliovirus PVSRIPO was tested in recurrent glioblastoma patients [103]. The study confirmed the potential of intratumor infusions of PVSRIPO for improving patients’ clinical outcomes. It was observed that the survival rate among patients who received PVSRIPO therapy was higher than that of historical controls.
While OV therapy has become an important focus of anti-tumor therapy, its safety and efficacy need to be tested in future research. The initial studies usually used replication-incompetent viruses to avoid complications (e.g., encephalitis), and now, an increasing number of types of viruses have been utilized as aforementioned. However, no full safety or preliminary efficacy data are currently available in the public domain. While the main goal of the current work is not to discuss each of these studies in detail, a comprehensive discussion of oncolytic virus therapy for glioblastoma can be found in several other excellent review papers [94,95,96,97]. Table 5 lists some of the completed phase II/III clinical trials of OV therapy for glioblastoma treatment.

5. Challenges and Perspectives

Despite encouraging results achieved so far, however, the anti-TME treatment for glioblastoma is still facing many challenges [5,58]. One of the primary impediments is the BBB. Although the BBB is compromised and more permeable during the tumor state, the anti-tumor drugs cannot cross the BBB inadequately to achieve sufficient drug accumulation in the tumor. For this reason, multiple efforts have been made to deliver pharmaceutical agents to the brain efficiently. One such important advance is focused ultrasound (FUS), which can increase the permeability of the BBB in a temporary way and enhance the delivery of drugs to the brain [104]. Currently, FUS-mediated BBB disruption has demonstrated robustness in non-invasive drug delivery to the brain and provides encouraging perspectives for the treatment of brain diseases, including glioblastoma. Another important impediment to achieving effective treatment responses, especially for immunotherapy-based approaches, is the immunosuppressive nature of glioblastoma TME. Therefore, strategies that could boost the immune response in glioblastoma TME will be helpful—for example, recruiting cytotoxic or tumor-killing inflammation cells, improving immunosuppressive properties through drugs, and transforming a ‘cold’ tumor into a ‘hot’ tumor. The third important impediment to developing effective treatment responses is the aforementioned complexity of the glioblastoma microenvironment. The enormous inter-tumor and intra-tumor heterogeneity of glioblastoma has made it one of the most difficult-to-treat malignancies in the world. Therefore, continued efforts are needed to fully understand the complex cellular and molecular components as well as their interactions involved in the TME of glioblastoma. At the same time, a synergic combination of different treatment strategies may lead to a promising curing regimen. Actually, there have been efforts to employ combinatorial therapies between immunotherapy and the current standard of care [105].

6. Conclusions

In summary, this review highlights the functions of neuroinflammation in glioblastoma at the cellular, molecular, and therapeutic levels. While increasing and promising results have been achieved in the anti-neuroinflammation therapy of glioblastoma, there are still many challenges. The immunosuppressive and heterogeneous characteristics of the glioblastoma microenvironment ultimately lead to resistance to anti-inflammatory therapies. Continued efforts into the tumor microenvironment will help our understanding of how these components interact with one another and contribute to the therapeutic response. This will lead to the development of more efficient and targeted therapy strategies for the treatment of glioblastoma in the future.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81702945).

Acknowledgments

We thank Zhong Lan for helpful discussions and insightful comments.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wen, P.Y.; Weller, M.; Lee, E.Q.; Alexander, B.M.; Barnholtz-Sloan, J.S.; Barthel, F.P.; Batchelor, T.T.; Bindra, R.S.; Chang, S.M.; Chiocca, E.A.; et al. Glioblastoma in adults: A Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro-Oncology 2020, 22, 1073–1113. [Google Scholar] [CrossRef] [PubMed]
  2. Schaff, L.R.; Mellinghoff, I.K. Glioblastoma and Other Primary Brain Malignancies in Adults: A Review. J. Am. Med. Assoc. 2023, 329, 574–587. [Google Scholar] [CrossRef] [PubMed]
  3. Tan, A.C.; Ashley, D.M.; López, G.Y.; Malinzak, M.; Friedman, H.S.; Khasraw, M. Management of glioblastoma: State of the art and future directions. CA—Cancer J. Clin. 2020, 70, 299–312. [Google Scholar] [CrossRef] [PubMed]
  4. Aldape, K.; Brindle, K.M.; Chesler, L.; Chopra, R.; Gajjar, A.; Gilbert, M.R.; Gottardo, N.; Gutmann, D.H.; Hargrave, D.; Holland, E.C.; et al. Challenges to curing primary brain tumours. Nat. Rev. Clin. Oncol. 2019, 16, 509–520. [Google Scholar] [CrossRef] [PubMed]
  5. Bikfalvi, A.; da Costa, C.A.; Avril, T.; Barnier, J.V.; Bauchet, L.; Brisson, L.; Cartron, P.F.; Castel, H.; Chevet, E.; Chneiweiss, H.; et al. Challenges in glioblastoma research: Focus on the tumor microenvironment. Trends Cancer 2023, 9, 9–27. [Google Scholar] [CrossRef] [PubMed]
  6. Becker, A.P.; Sells, B.E.; Haque, S.J.; Chakravarti, A. Tumor Heterogeneity in Glioblastomas: From Light Microscopy to Molecular Pathology. Cancers 2021, 13, 761. [Google Scholar] [CrossRef] [PubMed]
  7. Gieryng, A.; Pszczolkowska, D.; Walentynowicz, K.A.; Rajan, W.D.; Kaminska, B. Immune microenvironment of gliomas. Lab. Investig. 2017, 97, 498–518. [Google Scholar] [CrossRef] [PubMed]
  8. Virtuoso, A.; Giovannoni, R.; De Luca, C.; Gargano, F.; Cerasuolo, M.; Maggio, N.; Lavitrano, M.; Papa, M. The Glioblastoma Microenvironment: Morphology, Metabolism, and Molecular Signature of Glial Dynamics to Discover Metabolic Rewiring Sequence. Int. J. Mol. Sci. 2021, 22, 3301. [Google Scholar] [CrossRef]
  9. Di Nunno, V.; Franceschi, E.; Tosoni, A.; Gatto, L.; Bartolini, S.; Brandes, A.A. Tumor-Associated Microenvironment of Adult Gliomas: A Review. Front. Oncol. 2022, 12, 891543. [Google Scholar] [CrossRef]
  10. Sharma, P.; Aaroe, A.; Liang, J.Y.; Puduvalli, V.K. Tumor microenvironment in glioblastoma: Current and emerging concepts. Neuro-Oncol. Adv. 2023, 5, vdad009. [Google Scholar] [CrossRef]
  11. Nóbrega, A.H.L.; Pimentel, R.S.; Prado, A.P.; Garcia, J.; Frozza, R.L.; Bernardi, A. Neuroinflammation in Glioblastoma: The Role of the Microenvironment in Tumour Progression. Curr. Cancer Drug Targets 2024, 24, 579–594. [Google Scholar] [CrossRef]
  12. Alghamri, M.S.; McClellan, B.L.; Hartlage, M.S.; Haase, S.; Faisal, S.M.; Thalla, R.; Dabaja, A.; Banerjee, K.; Carney, S.V.; Mujeeb, A.A.; et al. Targeting Neuroinflammation in Brain Cancer: Uncovering Mechanisms, Pharmacological Targets, and Neuropharmaceutical Developments. Front. Pharmacol. 2021, 12, 680021. [Google Scholar] [CrossRef] [PubMed]
  13. Catalano, M. Editorial: Brain Tumors and Neuroinflammation. Front. Cell. Neurosci. 2022, 16, 941263. [Google Scholar] [CrossRef]
  14. Bianconi, A.; Aruta, G.; Rizzo, F.; Salvati, L.F.; Zeppa, P.; Garbossa, D.; Cofano, F. Systematic Review on Tumor Microenvironment in Glial Neoplasm: From Understanding Pathogenesis to Future Therapeutic Perspectives. Int. J. Mol. Sci. 2022, 23, 4166. [Google Scholar] [CrossRef]
  15. Bianconi, A.; Palmieri, G.; Aruta, G.; Monticelli, M.; Zeppa, P.; Tartara, F.; Melcarne, A.; Garbossa, D.; Cofano, F. Updates in Glioblastoma Immunotherapy: An Overview of the Current Clinical and Translational Scenario. Biomedicines 2023, 11, 1520. [Google Scholar] [CrossRef]
  16. Obermeier, B.; Daneman, R.; Ransohoff, R.M. Development, maintenance and disruption of the blood-brain barrier. Nat. Med. 2013, 19, 1584–1596. [Google Scholar] [CrossRef] [PubMed]
  17. Alahmari, A. Blood-Brain Barrier Overview: Structural and Functional Correlation. Neural Plast. 2021, 2021, 6564585. [Google Scholar] [CrossRef] [PubMed]
  18. Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.S.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef]
  19. Cugurra, A.; Mamuladze, T.; Rustenhoven, J.; Dykstra, T.; Beroshvili, G.; Greenberg, Z.J.; Baker, W.; Papadopoulos, Z.; Drieu, A.; Blackburn, S.; et al. Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and CNS parenchyma. Science 2021, 373, eabf7844. [Google Scholar] [CrossRef] [PubMed]
  20. Steeg, P.S. The blood-tumour barrier in cancer biology and therapy. Nat. Rev. Clin. Oncol. 2021, 18, 696–714. [Google Scholar] [CrossRef]
  21. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef]
  22. Mosteiro, A.; Pedrosa, L.; Ferrés, A.; Diao, D.; Sierra, A.; González, J.J. The Vascular Microenvironment in Glioblastoma: A Comprehensive Review. Biomedicines 2022, 10, 1285. [Google Scholar] [CrossRef]
  23. Baghba, R.; Roshangar, L.; Jahanban-Esfahlan, R.; Seidi, K.; Ebrahimi-Kalan, A.; Jaymand, M.; Kolahian, S.; Javaheri, T.; Zare, P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun. Signal. 2020, 18, 59. [Google Scholar] [CrossRef]
  24. Dapash, M.; Hou, D.; Castro, B.; Lee-Chang, C.; Lesniak, M.S. The Interplay between Glioblastoma and Its Microenvironment. Cells 2021, 10, 2257. [Google Scholar] [CrossRef] [PubMed]
  25. Crivii, C.B.; Bosca, A.B.; Melincovici, C.S.; Constantin, A.M.; Marginean, M.; Dronca, E.; Sufletel, R.; Gonciar, D.; Bungardean, M.; Sovrea, A. Glioblastoma Microenvironment and Cellular Interactions. Cancers 2022, 14, 1092. [Google Scholar] [CrossRef]
  26. Lam, D.; Enright, H.A.; Cadena, J.; Peters, S.K.G.; Sales, A.P.; Osburn, J.J.; Soscia, D.A.; Kulp, K.S.; Wheeler, E.K.; Fischer, N.O. Tissue-specific extracellular matrix accelerates the formation of neural networks and communities in a neuron-glia co-culture on a multi-electrode array. Sci. Rep. 2019, 9, 4159. [Google Scholar] [CrossRef]
  27. Belousov, A.; Titov, S.; Shved, N.; Garbuz, M.; Malykin, G.; Gulaia, V.; Kagansky, A.; Kumeiko, V. The Extracellular Matrix and Biocompatible Materials in Glioblastoma Treatment. Front. Bioeng. Biotech. 2019, 7, 341. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, C.; Sinha, S.; Jiang, X.Y.; Murphy, L.; Fitch, S.; Wilson, C.; Grant, G.; Yang, F. Matrix Stiffness Modulates Patient-Derived Glioblastoma Cell Fates in Three-Dimensional Hydrogels. Tissue Eng. Part A 2021, 27, 390–401. [Google Scholar] [CrossRef]
  29. Yeo, E.C.F.; Brown, M.P.; Gargett, T.; Ebert, L.M. The Role of Cytokines and Chemokines in Shaping the Immune Microenvironment of Glioblastoma: Implications for Immunotherapy. Cells 2021, 10, 607. [Google Scholar] [CrossRef]
  30. Corbet, C.; Feron, O. Tumour acidosis: From the passenger to the driver’s seat. Nat. Rev. Cancer 2017, 17, 577–593. [Google Scholar] [CrossRef] [PubMed]
  31. Uyar, R. Glioblastoma microenvironment: The stromal interactions. Pathol. Res. Pract. 2022, 232, 153813. [Google Scholar] [CrossRef]
  32. Lin, C.C.J.; Yu, K.; Hatcher, A.; Huang, T.W.; Lee, H.K.; Carlson, J.; Weston, M.C.; Chen, F.J.; Zhang, Y.Q.; Zhu, W.Y.; et al. Identification of diverse astrocyte populations and their malignant analogs. Nat. Neurosci. 2017, 20, 396–405. [Google Scholar] [CrossRef]
  33. Heiland, D.H.; Ravi, V.M.; Behringer, S.P.; Frenking, J.H.; Wurm, J.; Joseph, K.; Garrelfs, N.W.C.; Strähle, J.; Heynckes, S.; Grauvogel, J.; et al. Tumor-associated reactive astrocytes aid the evolution of immunosuppressive environment in glioblastoma. Nat. Commun. 2019, 10, 2541. [Google Scholar] [CrossRef] [PubMed]
  34. Quail, D.F.; Joyce, J.A. The Microenvironmental Landscape of Brain Tumors. Cancer Cell 2017, 31, 326–341. [Google Scholar] [CrossRef] [PubMed]
  35. Ahir, B.K.; Engelhard, H.H.; Lakka, S.S. Tumor Development and Angiogenesis in Adult Brain Tumor: Glioblastoma. Mol. Neurobiol. 2020, 57, 2461–2478. [Google Scholar] [CrossRef] [PubMed]
  36. Kaminska, B.; Ochocka, N.; Segit, P. Single-Cell Omics in Dissecting Immune Microenvironment of Malignant Gliomas-Challenges and Perspectives. Cells 2021, 10, 2264. [Google Scholar] [CrossRef] [PubMed]
  37. Khan, F.; Pang, L.Z.; Dunterman, M.; Lesniak, M.S.; Heimberger, A.B.; Chen, P.W. Macrophages and microglia in glioblastoma: Heterogeneity, plasticity, and therapy. J. Clin. Investig. 2023, 133, e163446. [Google Scholar] [CrossRef] [PubMed]
  38. Hu, X.M. Microglia/macrophage polarization: Fantasy or evidence of functional diversity? J. Cereb. Blood Flow Metab. 2020, 40, S134–S136. [Google Scholar] [CrossRef]
  39. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
  40. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
  41. Woroniecka, K.I.; Rhodin, K.E.; Chongsathidkiet, P.; Keith, K.A.; Fecci, P.E. T-cell Dysfunction in Glioblastoma: Applying a New Framework. Clin. Cancer Res. 2018, 24, 3792–3802. [Google Scholar] [CrossRef]
  42. Humphries, W.; Wei, J.; Sampson, J.H.; Heimberger, A.B. The Role of Tregs in Glioma-Mediated Immunosuppression: Potential Target for Intervention. Neurosurg. Clin. N. Am. 2010, 21, 125–137. [Google Scholar] [CrossRef] [PubMed]
  43. Li, C.X.; Jiang, P.; Wei, S.H.; Xu, X.F.; Wang, J.J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 2020, 19, 116. [Google Scholar] [CrossRef] [PubMed]
  44. Kelly, W.J.; Giles, A.J.; Gilbert, M. T lymphocyte-targeted immune checkpoint modulation in glioma. J. Immunother. Cancer 2020, 8, e000379. [Google Scholar] [CrossRef] [PubMed]
  45. Chongsathidkiet, P.; Jackson, C.; Koyama, S.; Loebel, F.; Cui, X.Y.; Farber, S.H.; Woroniecka, K.; Elsamadicy, A.A.; Dechant, C.A.; Kemeny, H.R.; et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat. Med. 2018, 24, 1459–1468. [Google Scholar] [CrossRef]
  46. Jackson, C.M.; Choi, J.; Lim, M. Mechanisms of immunotherapy resistance: Lessons from glioblastoma. Nat. Immunol. 2019, 20, 1100–1109. [Google Scholar] [CrossRef]
  47. Sedgwick, A.J.; Ghazanfari, N.; Constantinescu, P.; Mantamadiotis, T.; Barrow, A.D. The Role of NK Cells and Innate Lymphoid Cells in Brain Cancer. Front. Immunol. 2020, 11, 1549. [Google Scholar] [CrossRef]
  48. Burster, T.; Gärtner, F.; Bulach, C.; Zhanapiya, A.; Gihring, A.; Knippschild, U. Regulation of MHC I Molecules in Glioblastoma Cells and the Sensitizing of NK Cells. Pharmaceuticals 2021, 14, 236. [Google Scholar] [CrossRef] [PubMed]
  49. Böttcher, J.P.; Sousa, C.R.E. The Role of Type 1 Conventional Dendritic Cells in Cancer Immunity. Trends Cancer 2018, 4, 784–792. [Google Scholar] [CrossRef] [PubMed]
  50. Srivastava, S.; Jackson, C.; Kim, T.; Choi, J.; Lim, M. A Characterization of Dendritic Cells and Their Role in Immunotherapy in Glioblastoma: From Preclinical Studies to Clinical Trials. Cancers 2019, 11, 537. [Google Scholar] [CrossRef]
  51. Peng, Q.; Qiu, X.Y.; Zhang, Z.H.; Zhang, S.L.; Zhang, Y.Y.; Liang, Y.; Guo, J.Y.; Peng, H.; Chen, M.Y.; Fu, Y.X.; et al. PD-L1 on dendritic cells attenuates T cell activation and regulates response to immune checkpoint blockade. Nat. Commun. 2020, 11, 4835. [Google Scholar] [CrossRef]
  52. Massara, M.; Persico, P.; Bonavita, O.; Poeta, V.M.; Locati, M.; Simonelli, M.; Bonecchi, R. Neutrophils in Gliomas. Front. Immunol. 2017, 8, 1349. [Google Scholar] [CrossRef] [PubMed]
  53. Masucci, M.T.; Minopoli, M.; Carriero, M.V. Tumor Associated Neutrophils. Their Role in Tumorigenesis, Metastasis, Prognosis and Therapy. Front. Oncol. 2019, 9, 1146. [Google Scholar] [CrossRef] [PubMed]
  54. Wu, L.; Zhang, X.H.F. Tumor-Associated Neutrophils and Macrophages-Heterogenous but Not Chaotic. Front. Immunol. 2020, 11, 553967. [Google Scholar] [CrossRef] [PubMed]
  55. Khan, S.; Mittal, S.; McGee, K.; Alfaro-Munoz, K.D.; Majd, N.; Balasubramaniyan, V.; de Groot, J.F. Role of Neutrophils and Myeloid-Derived Suppressor Cells in Glioma Progression and Treatment Resistance. Int. J. Mol. Sci. 2020, 21, 1954. [Google Scholar] [CrossRef] [PubMed]
  56. Di Nunno, V.; Franceschi, E.; Tosoni, A.; Gatto, L.; Bartolini, S.; Brandes, A.A. Glioblastoma Microenvironment: From an Inviolable Defense to a Therapeutic Chance. Front. Oncol. 2022, 12, 852950. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, X.H.; Zhao, L.L.; Zhang, H.; Zhang, Y.R.; Ju, H.Y.; Wang, X.Y.; Ren, H.; Zhu, X.; Dong, Y.C. The immunosuppressive microenvironment and immunotherapy in human glioblastoma. Front. Immunol. 2022, 13, 1003651. [Google Scholar] [CrossRef] [PubMed]
  58. Yasinjan, F.; Xing, Y.; Geng, H.Y.; Guo, R.; Yang, L.; Liu, Z.L.; Wang, H. Immunotherapy: A promising approach for glioma treatment. Front. Immunol. 2023, 14, 1255611. [Google Scholar] [CrossRef] [PubMed]
  59. Sampson, J.H.; Maus, M.V.; June, C.H. Immunotherapy for Brain Tumors. J. Clin. Oncol. 2017, 35, 2450–2456. [Google Scholar] [CrossRef] [PubMed]
  60. Lim, M.; Xia, Y.X.; Bettegowda, C.; Weller, M. Current state of immunotherapy for glioblastoma. Nat. Rev. Clin. Oncol. 2018, 15, 422–442. [Google Scholar] [CrossRef]
  61. Sampson, J.H.; Gunn, M.D.; Fecci, P.E.; Ashley, D.M. Brain immunology and immunotherapy in brain tumours. Nat. Rev. Cancer 2020, 20, 12–25. [Google Scholar] [CrossRef]
  62. Xu, S.C.; Tang, L.; Li, X.Z.; Fan, F.; Liu, Z.X. Immunotherapy for glioma: Current management and future application. Cancer Lett. 2020, 476, 1–12. [Google Scholar] [CrossRef] [PubMed]
  63. Chokshi, C.R.; Brakel, B.A.; Tatari, N.; Savage, N.; Salim, S.K.; Venugopal, C.; Singh, S.K. Advances in Immunotherapy for Adult Glioblastoma. Cancers 2021, 13, 3400. [Google Scholar] [CrossRef] [PubMed]
  64. Ma, Y.F.; Yang, Z.G.; Huntoon, K.; Jiang, W.; Kim, B.Y.S. Advanced Immunotherapy Approaches for Glioblastoma. Adv. Ther. 2021, 4, 2100046. [Google Scholar] [CrossRef]
  65. Mende, A.L.; Schulte, J.D.; Okada, H.; Clarke, J.L. Current Advances in Immunotherapy for Glioblastoma. Curr. Oncol. Rep. 2021, 23, 21. [Google Scholar] [CrossRef] [PubMed]
  66. Mahmoud, A.B.; Ajina, R.; Aref, S.; Darwish, M.; Alsayb, M.; Taher, M.; AlSharif, S.A.; Hashem, A.M.; Alkayyal, A.A. Advances in immunotherapy for glioblastoma multiforme. Front. Immunol. 2022, 13, 944452. [Google Scholar] [CrossRef] [PubMed]
  67. Gilbert, M.R.; Dignam, J.J.; Armstrong, T.S.; Wefel, J.S.; Blumenthal, D.T.; Vogelbaum, M.A.; Colman, H.; Chakravarti, A.; Pugh, S.; Won, M.; et al. A Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma. N. Engl. J. Med. 2014, 370, 699–708. [Google Scholar] [CrossRef] [PubMed]
  68. Ferrara, N.; Hillan, K.J.; Gerber, H.P.; Novotny, W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat. Rev. Drug Discov. 2004, 3, 391–400. [Google Scholar] [CrossRef] [PubMed]
  69. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.A.; Paleologos, N.; Nicholas, M.K.; Jensen, R.; et al. Bevacizumab Alone and in Combination with Irinotecan in Recurrent Glioblastoma. J. Clin. Oncol. 2009, 27, 4733–4740. [Google Scholar] [CrossRef] [PubMed]
  70. Clavreul, A.; Pourbaghi-Masouleh, M.; Roger, E.; Menei, P. Nanocarriers and nonviral methods for delivering antiangiogenic factors for glioblastoma therapy: The story so far. Int. J. Nanomed. 2019, 14, 2497–2513. [Google Scholar] [CrossRef]
  71. Martin, J.D.; Seano, G.; Jain, R.K. Normalizing Function of Tumor Vessels: Progress, Opportunities, and Challenges. Annu. Rev. Physiol. 2019, 81, 505–534. [Google Scholar] [CrossRef]
  72. Lombardi, G.; De Salvo, G.L.; Brandes, A.A.; Eoli, M.; Rudà, R.; Faedi, M.; Lolli, I.; Pace, A.; Daniele, B.; Pasqualetti, F.; et al. Regorafenib compared with lomustine in patients with relapsed glioblastoma (REGOMA): A multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2019, 20, 110–119. [Google Scholar] [CrossRef] [PubMed]
  73. Belotti, D.; Pinessi, D.; Taraboletti, G. Alternative Vascularization Mechanisms in Tumor Resistance to Therapy. Cancers 2021, 13, 1912. [Google Scholar] [CrossRef]
  74. Thomas Sandmann, R.B.; Garcia, J.; Li, C.; Cloughesy, T.; Chinot, O.L.; Wick, W.; Nishikawa, R.; Mason, W.; Henriksson, R.; Saran, F.; et al. Patients with Proneural Glioblastoma May Derive Overall Survival Benefit from the Addition of Bevacizumab to First-Line Radiotherapy and Temozolomide: Retrospective Analysis of the AVAglio Trial. J. Clin. Oncol. 2015, 33, 10. [Google Scholar] [CrossRef] [PubMed]
  75. Korman, A.J.; Garrett-Thomson, S.C.; Lonberg, N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat. Rev. Drug Discov. 2022, 21, 509–528. [Google Scholar] [CrossRef]
  76. Berger, K.N.; Pu, J.J. PD-1 pathway and its clinical application: A 20 year journey after discovery of the complete human-gene. Gene 2018, 638, 20–25. [Google Scholar] [CrossRef] [PubMed]
  77. Han, Y.Y.; Liu, D.D.; Li, L.H. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar] [PubMed]
  78. Cloughesy, T.F.; Mochizuki, A.Y.; Orpilla, J.R.; Hugo, W.; Lee, A.H.; Davidson, T.B.; Wang, A.C.; Ellingson, B.M.; Rytlewski, J.A.; Sanders, C.M.; et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 2019, 25, 477–486. [Google Scholar] [CrossRef] [PubMed]
  79. Lim, M.; Weller, M.; Idbaih, A.; Steinbach, J.; Finocchiaro, G.; Raval, R.R.; Ansstas, G.; Baehring, J.; Taylor, J.W.; Honnorat, J.; et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated promoter. Neuro-Oncology 2022, 24, 1935–1949. [Google Scholar] [CrossRef] [PubMed]
  80. Han, S.; Ma, E.L.; Wang, X.N.; Yu, C.Y.; Dong, T.; Zhan, W.; Wei, X.Z.; Liang, G.B.; Feng, S.Z. Rescuing defective tumor-infiltrating T-cell proliferation in glioblastoma patients. Oncol. Lett. 2016, 12, 2924–2929. [Google Scholar] [CrossRef]
  81. Sayegh, E.T.; Oh, T.; Fakurnejad, S.; Bloch, O.; Parsa, A.T. Vaccine therapies for patients with glioblastoma. J. Neuro-Oncol. 2014, 119, 531–546. [Google Scholar] [CrossRef]
  82. Zhao, B.H.; Wu, J.M.; Li, H.Z.; Wang, Y.K.; Wang, Y.N.; Xing, H.; Wang, Y.; Ma, W.B. Recent advances and future challenges of tumor vaccination therapy for recurrent glioblastoma. Cell Commun. Signal 2023, 21, 74. [Google Scholar] [CrossRef] [PubMed]
  83. Platten, M.; Bunse, L.; Riehl, D.; Bunse, T.; Ochs, K.; Wick, W. Vaccine Strategies in Gliomas. Curr. Treat. Options Neurol. 2018, 20, 11. [Google Scholar] [CrossRef] [PubMed]
  84. Swartz, A.M.; Batich, K.A.; Fecci, P.E.; Sampson, J.H. Peptide vaccines for the treatment of glioblastoma. J. Neuro-Oncol. 2015, 123, 433–440. [Google Scholar] [CrossRef] [PubMed]
  85. Li, L.H.; Zhou, J.; Dong, X.T.; Liao, Q.J.; Zhou, D.B.; Zhou, Y.H. Dendritic cell vaccines for glioblastoma fail to complete clinical translation: Bottlenecks and potential countermeasures. Int. Immunopharmacol. 2022, 109, 108929. [Google Scholar] [CrossRef] [PubMed]
  86. Zhang, M.; Choi, J.; Lim, M. Advances in Immunotherapies for Gliomas. Curr. Neurol. Neurosci. 2022, 22, 1–10. [Google Scholar] [CrossRef] [PubMed]
  87. Schuster, J.; Lai, R.K.; Recht, L.D.; Reardon, D.A.; Paleologos, N.A.; Groves, M.D.; Mrugala, M.M.; Jensen, R.; Baehring, J.M.; Sloan, A.; et al. A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: The ACT III study. Neuro-Oncology 2015, 17, 854–861. [Google Scholar] [CrossRef] [PubMed]
  88. Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F.; et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef] [PubMed]
  89. Liau, L.M.; Ashkan, K.; Brem, S.; Campian, J.L.; Trusheim, J.E.; Iwamoto, F.M.; Tran, D.D.; Ansstas, G.; Cobbs, C.S.; Heth, J.A.; et al. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination with Extension of Survival Among Patients with Newly Diagnosed and Recurrent Glioblastoma A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol. 2023, 9, 112–121. [Google Scholar] [CrossRef] [PubMed]
  90. Chen, D.; Yang, J. Development of novel antigen receptors for CAR T-cell therapy directed toward solid malignancies. Transl. Res. 2017, 187, 11–21. [Google Scholar] [CrossRef]
  91. Bagley, S.J.; Desai, A.S.; Linette, G.P.; June, C.H.; O’Rourke, D.M. CAR T-cell therapy for glioblastoma: Recent clinical advances and future challenges. Neuro-Oncology 2018, 20, 1429–1438. [Google Scholar] [CrossRef]
  92. Li, L.; Zhu, X.Q.; Qian, Y.; Yuan, X.L.; Ding, Y.; Hu, D.S.; He, X.; Wu, Y. Chimeric Antigen Receptor T-Cell Therapy in Glioblastoma: Current and Future. Front. Immunol. 2020, 11, 594271. [Google Scholar] [CrossRef] [PubMed]
  93. Jenkins, M.R.; Drummond, K.J. CAR T-Cell Therapy for Glioblastoma. N. Engl. J. Med. 2024, 390, 1329–1332. [Google Scholar] [CrossRef] [PubMed]
  94. O’Rourke, D.M.; Nasrallah, M.P.; Desai, A.; Melenhorst, J.J.; Mansfield, K.; Morrissette, J.J.D.; Martinez-Lage, M.; Brem, S.; Maloney, E.; Shen, A.; et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 2017, 9, aaa0984. [Google Scholar] [CrossRef]
  95. Brown, C.E.; Alizadeh, D.; Starr, R.; Weng, L.H.; Wagner, J.R.; Naranjo, A.; Ostberg, J.R.; Blanchard, M.S.; Kilpatrick, J.; Simpson, J.; et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N. Engl. J. Med. 2016, 375, 2561–2569. [Google Scholar] [CrossRef] [PubMed]
  96. Ahmed, N.; Brawley, V.; Hegde, M.; Bielamowicz, K.; Kalra, M.; Landi, D.; Robertson, C.; Gray, T.L.; Diouf, O.; Wakefield, A.; et al. HER2-Specific Chimeric Antigen Receptor-Modified Virus-Specific T Cells for Progressive Glioblastoma A Phase 1 Dose-Escalation Trial. JAMA Oncol. 2017, 3, 1094–1101. [Google Scholar] [CrossRef] [PubMed]
  97. Aurelian, L. Oncolytic viruses as immunotherapy: Progress and remaining challenges. Oncotargets Ther. 2016, 9, 2627–2637. [Google Scholar] [CrossRef] [PubMed]
  98. Lawler, S.E.; Speranza, M.C.; Cho, C.F.; Chiocca, E.A. Oncolytic Viruses in Cancer Treatment A Review. JAMA Oncol. 2017, 3, 841–849. [Google Scholar] [CrossRef] [PubMed]
  99. Wollmann, G.; Ozduman, K.; van den Pol, A.N. Oncolytic Virus Therapy for Glioblastoma Multiforme. Cancer J. 2012, 18, 69–81. [Google Scholar] [CrossRef] [PubMed]
  100. Martikainen, M.; Essand, M. Virus-Based Immunotherapy of Glioblastoma. Cancers 2019, 11, 186. [Google Scholar] [CrossRef]
  101. Liu, P.H.; Wang, Y.N.; Wang, Y.K.; Kong, Z.R.; Chen, W.Q.; Li, J.T.; Chen, W.L.; Tong, Y.R.; Ma, W.B.; Wang, Y. Effects of oncolytic viruses and viral vectors on immunity in glioblastoma. Gene Ther. 2022, 29, 115–126. [Google Scholar] [CrossRef]
  102. Qi, Z.B.; Long, X.Y.; Liu, J.Y.; Cheng, P. Glioblastoma microenvironment and its reprogramming by oncolytic virotherapy. Front. Cell. Neurosci. 2022, 16, 819363. [Google Scholar] [CrossRef] [PubMed]
  103. Desjardins, A.; Gromeier, M.; Herndon, J.E.; Beaubier, N.; Bolognesi, D.P.; Friedman, A.H.; Friedman, H.S.; McSherry, F.; Muscat, A.M.; Nair, S.; et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N. Engl. J. Med. 2018, 379, 150–161. [Google Scholar] [CrossRef] [PubMed]
  104. Durham, P.G.; Butnariu, A.; Alghorazi, R.; Pinton, G.; Krishna, V.; Dayton, P.A. Current clinical investigations of focused ultrasound blood-brain barrier disruption: A review. Neurotherapeutics 2024, 21, e00352. [Google Scholar] [CrossRef] [PubMed]
  105. Niedbala, M.; Malarz, K.; Sharma, G.; Kramer-Marek, G.; Kaspera, W. Glioblastoma: Pitfalls and Opportunities of Immunotherapeutic Combinations. Oncotargets Ther. 2022, 15, 437–468. [Google Scholar] [CrossRef]
Figure 1. Components of the tumor microenvironment (TME) in glioblastoma.
Figure 1. Components of the tumor microenvironment (TME) in glioblastoma.
Brainsci 14 00687 g001
Table 1. Clinical trials of anti-vasculature therapy for glioblastoma treatment.
Table 1. Clinical trials of anti-vasculature therapy for glioblastoma treatment.
ClinicalTrials.gov IDTargetBrief Description of the TrialPhaseYear of Start to Completion
1NCT01753713VEGFDovitinib in treating patients with recurrent or progressive glioblastoma22012–2017
2NCT01609790VEGFBevacizumab with or without trebananib in treating patients with recurrent brain tumors22012–2022
3NCT02330562VEGFMarizomib alone or in combination with bevacizumab
in patients with recurrent glioblastoma
1 and 22015–2021
4NCT02511405VEGFA phase 3, pivotal trial of VB-111 plus bevacizumab vs. bevacizumab in patients with recurrent glioblastoma32015–2018
5NCT02342379VEGFTH-302 in combination with bevacizumab for glioblastoma22015–2019
Note: The data are retrieved from clinicaltrials.gov.
Table 2. Clinical trials of ICIs for glioblastoma treatment.
Table 2. Clinical trials of ICIs for glioblastoma treatment.
ClinicalTrials.gov IDTargetBrief Description of the TrialPhaseYear of Start to Completion
1NCT02550249PD-1Neoadjuvant nivolumab in glioblastoma22015–2017
2NCT02336165PD-1Phase 2 study of durvalumab (MEDI4736) in patients with glioblastoma22015–2021
3NCT02337491PD-1Pembrolizumab +/− bevacizumab for recurrent glioblastoma22015–2020
4NCT02617589PD-1An investigational immuno-therapy study of nivolumab compared to temozolomide, each given with radiation therapy, for newly diagnosed patients with glioblastoma32016–2022
5NCT02798406PD-1Combination adenovirus + pembrolizumab to trigger immune virus effects22016–2021
6NCT03018288PD-1Radiation therapy plus temozolomide and pembrolizumab with and without HSPPC-96 in newly diagnosed glioblastoma22017–2022
7NCT02794883CTLA-4Tremelimumab and durvalumab in combination or alone in treating patients with recurrent malignant glioma22016–2020
8NCT03367715CTLA-4Nivolumab, ipilimumab, and short-course radiotherapy in adults with newly diagnosed MGMT unmethylated glioblastoma22018–2022
Note: The data are retrieved from clinicaltrials.gov.
Table 3. Clinical trials of vaccine therapy for glioblastoma treatment.
Table 3. Clinical trials of vaccine therapy for glioblastoma treatment.
ClinicalTrials.gov IDVaccine TypeTitlePhaseYear of Start to Completion
1NCT00293423Peptide vaccineGP96 heat shock protein-peptide complex vaccine in treating patients with recurrent or progressive glioma1 and 22005–2013
2NCT00458601Peptide vaccinePhase II study of rindopepimut (CDX-110) in patients with glioblastoma multiforme22007–2016
3NCT00643097Peptide vaccineVaccine therapy in treating patients with newly diagnosed glioblastoma22007–2016
4NCT00905060Peptide vaccineHSPPC-96 vaccine with temozolomide in patients with newly diagnosed glioblastoma22009–2014
5NCT01480479Peptide vaccinePhase III study of rindopepimut/GM-CSF in patients with newly diagnosed glioblastoma32011–2016
6NCT01920191Peptide vaccinePhase I/II trial of IMA950 multi-peptide vaccine plus poly-ICLC in glioblastoma1 and 22013–2016
7NCT00639639DC vaccineVaccine therapy in treating patients with newly diagnosed glioblastoma12006–2022
8NCT00323115DC vaccinePhase II feasibility study of dendritic cell vaccination for newly diagnosed glioblastoma22006–2013
9NCT00846456DC vaccineSafe study of dendritic cell (DC) based therapy targeting tumor stem cells in glioblastoma1 and 22009–2013
10NCT01006044DC vaccineEfficacy and safety of autologous dendritic cell vaccination in glioblastoma after complete surgical resection22009–2014
11NCT01213407DC vaccineDendritic cell cancer vaccine for high-grade glioma22010–2015
12NCT02465268DC vaccineVaccine therapy for the treatment of newly diagnosed glioblastoma22016–2023
Note: The data are retrieved from clinicaltrials.gov.
Table 4. Clinical trials of CAR-T cell therapy for glioblastoma treatment.
Table 4. Clinical trials of CAR-T cell therapy for glioblastoma treatment.
ClinicalTrials.gov IDTargetBrief Description of the TrialPhaseYear of Start to Completion
1NCT01082926GRm13Z40-2Phase I study of cellular immunotherapy for recurrent/refractory malignant glioma using intratumoral infusions of GRm13Z40-2, an allogeneic CD8+ Cytolitic T-Cell line genetically modified to express the IL 13-Zetakine and HyTK and to be resistant to glucocorticoids, in combination with interleukin-212010–2013
2NCT01109095HER2CMV-specific cytotoxic T lymphocytes expressing CAR targeting HER2 in patients with glioblastoma12010–2018
3NCT01454596EGFRvIIICAR T cell receptor immunotherapy targeting EGFRvIII for patients with malignant gliomas expressing EGFRvIII1 and 22012–2019
4NCT03726515EGFRvIIICART-EGFRvIII + pembrolizumab in glioblastoma12019–2021
Note: The data are retrieved from clinicaltrials.gov.
Table 5. Clinical trials of OV therapy for glioblastoma treatment.
Table 5. Clinical trials of OV therapy for glioblastoma treatment.
ClinicalTrials.gov IDVirus TypeBrief Description of the TrialPhaseYear of Start to Completion
1NCT00028158Herpes Simplex VirusSafety and effectiveness study of G207, a tumor-killing virus, in patients with recurrent brain cancer1 and 22002–2003
2NCT00528684ReovirusSafety and efficacy study of REOLYSIN® in the treatment of recurrent malignant gliomas12006–2010
3NCT01301430ParvovirusParvovirus H-1 (ParvOryx) in patients with progressive primary or recurrent glioblastoma multiforme.1 and 22011–2015
4NCT01956734AdenovirusVirus DNX2401 and temozolomide in recurrent glioblastoma12013–2015
5NCT02197169AdenovirusDNX-2401 with interferon-gamma (IFN-γ) for recurrent glioblastoma or gliosarcoma brain tumors12014–2018
6NCT02798406AdenovirusCombination adenovirus + pembrolizumab to trigger immune virus effects22016–2021
7NCT03072134AdenovirusNeural stem cell-based virotherapy of newly diagnosed malignant glioma12017–2021
Note: The data are retrieved from clinicaltrials.gov.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, X.; Gou, W.; Zhang, X. Neuroinflammation in Glioblastoma: Progress and Perspectives. Brain Sci. 2024, 14, 687. https://doi.org/10.3390/brainsci14070687

AMA Style

Li X, Gou W, Zhang X. Neuroinflammation in Glioblastoma: Progress and Perspectives. Brain Sciences. 2024; 14(7):687. https://doi.org/10.3390/brainsci14070687

Chicago/Turabian Style

Li, Xin, Wenting Gou, and Xiaoqin Zhang. 2024. "Neuroinflammation in Glioblastoma: Progress and Perspectives" Brain Sciences 14, no. 7: 687. https://doi.org/10.3390/brainsci14070687

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop