A large body of literature provides evidence for the promising potential of immunotherapy in the treatment of GB. Being notorious for its extensive local invasion into deeper areas of the CNS, GB is always difficult to resect. Due to this obstacle, immunotherapy is taking the forefront as a promising treatment option for those diagnosed with GB. Various immune cell types are reported to specifically attack the tumor cells using myriad mechanisms of recognition and destruction. Many of those cells will be discussed separately below.
1.1.1. Lymphocytes/CAR-T
When considering immune cells, it is always important to discuss the role of T and B lymphocytes. Both cell types work together to formulate the immune responses our bodies have towards invasive antigens and substances. They can easily cross the BBB under certain physiological and pathological conditions
[1], and they perform well when combating tumors. Nair and colleagues induced certain T cells in vitro to become CMV pp65-specific, thus acquiring the capability to recognize and kill tumor cells at an increased rate
[2]. T cells can elicit a powerful mechanism to eliminate internal and external pathogens. Therefore, they are being used therapeutically to manage different malignancies with promising outcomes
[1].
In a recent study, Lee-Chang et al.
[3] developed a vaccine utilizing B cells activated with CD40 agonism and IFNy stimulation. This vaccine aims to travel to secondary lymphoid organs and increase antigen cross-presentation. As a result, this vaccine promotes the survival and functionality of CD8+ T cells
[3]. It was found that when this vaccine was combined with other treatments such as radiation and PD-L1 blockade, this combination was able to elicit immunological memory that prevented the growth of new tumor cells.
The chimeric antigen receptors (CAR-) T cells are a product of genetic engineering in an effort to achieve a long-term outcome in cases of malignancies such as GB. CAR-T cells are genetically programmed to attack tumor cells by recognizing the surface proteins expressed
[4]. They are developed to target tumor-associated antigens (TAA), such as interleukin 13 receptor α 2 (IL13Rα2), epidermal growth factor receptor variant III (EGFRvIII), human epidermal growth factor receptor 2 (HER2), and erythropoietin-producing hepatocellular carcinoma A2 (EphA2)
[5][6]. These artificial proteins are composed of an extracellular antigen-binding domain, a transmembrane domain, and an intracellular T-Cell signaling domain like Cd3 ζ (with or without costimulatory components)
[7]. Different generations of CAR-T cells have been studied with outstanding outcomes in redirecting the cytotoxic nature of T lymphocytes to become independent of the major histocompatibility complex (MHC) restrictions and without requiring antigen priming
[8]. First generation CAR-T cells have an antigen recognition domain (scFv) and the activating signal CD3 ζ, a costimulatory molecule, can be added to form a second and third generation CAR with two costimulatory molecules
[9]. Recently, Bielamowicz et al. Bielamowicz, Fousek
[10] stated that GB cells overexpress different and targetable surface antigens, such as HER2, IL13Rα2, EphA2 and EGFRvIII which have been targeted using CAR-T cells with promising outcomes.
EGFRvIII is a variant of EGFR present in 31–64% of patients with GB that promote tumor cell proliferation, invasion, and angiogenesis in the tumor environment
[11]. EFRON vIII CAR-T cells localized to intracerebral tumors reduce the expression of EGFRvIII in cancer cells
[12]. Chen et al.
[11] generated an EGFRvIII-targeting CAR (806-28Z CAR) using the epitope of 806 antibody, which is only fully exposed on EGFRvIII or activated EGFR, for in vitro and in vivo testing with an SQ xenograft established by injecting 1 × 10
7 GL261/EGFRvIII cells mixed with Matrigel (4:1) into the right limb of C57BL/6 mice. They reported dose-dependent cytotoxicity against mouse GL261/EGFRvIII cells, effective inhibition of tumor growth, effective lysis of mixed heterogenous GL261 cells accompanied with high concentrations of granzyme B which can be used as a predictive marker to determine the effectiveness of CAR-T cells as a therapeutic approach
[11]. Likewise, Ravanpay et al.
[13] used EFGR806-CAR through intracranial administration to treat xenograft GB mouse models. Placing the CAR-T cells near the target site lowered the risk of side effects outside the CNS and ensured consistent regression of orthotopic glioma.
HER2 antigen is over expressed in about 80% of GB cases and was incorporated in the design of a third generation anti-HER2 CAR (anti-HER2 scFv-CD28-CD137-CD3ζ) combined with a PD1 blockade and anti-HER2 scFv from the 4D5 antibody for CAR construction to avoid a decreased binding of CAR to antigen
[9]. The costimulatory molecule, CD28, induced and increased production of IL-2, enhancing the clonal expansion and endurance of CAR T cells that, when combined with 4-1BB/CD137, were more efficient in INF-γ production and lysis of tumor cells
[9]. TanCAR combined two antigen recognition domains for HER2 and IL13Rα2 previously proved to provide a “near-complete tumor elimination” in previous work by Hedge et al.
[14]. Higher density of TanCAR-mediated HER2-IL13Rα2 heterodimers was observed on STED super resolution microscopes and confirmed by PLA in addition to increased IFN-γ and IL-2 secretion; all supporting the antitumor characteristic of this therapy
[15]. Bielamowicz et al.
[10] demonstrated that nearly 100% of tumor cells were killed, in vitro and in vivo, with UCAR-T, a trivalent transgene combining IL13Rα2 binding IL-13 mutein, HER2-specific single chain variable fragment 9scFv), FRP5, and Epha2-specific scFv 4H5 with CD28 as a costimulatory molecule and ζ-signaling domain of the T cell receptor (TCR). The UCAR T cell showed better signaling, increased engagement of a larger domain of GB cells and an almost entire activation and proliferation of the 3 CARs as demonstrated in surface staining
[10].
In efforts to evidence long term immunity after therapy with CAR T cells, Pituch et al.
[6] proposed the utilization of IL13Rα2-CARCD28ζ CAR-T cells and observed an increased number of CD8α+DC cells known to efficiently cross-present both cell-bound and soluble antigens in the MHC class I, therefore inducing a CD8+ T cell response. Krenciute et al.
[5] designed the IL13Rα2-CAR.IL15T cell by modifying T cells with a retroviral vector that encoded an IL13Rα2-specific scFv with CD28.ζ endodomain and a retroviral vector that encoded inducible caspase-9, NGFR with a shortened cytoplasmic domain and IL15 separated by 2A sequence demonstrating that the action of IL15 in IL13Rα2-CAR T cells enhanced their effector functions. Although IL15 did not show any significant improvement in the proliferation of IL13Rα2-CAR T cells or cytokine production after the first antigen-specific stimulation, it showed significant proliferation after the third stimulation
[5]. In light of the fact that steroids (e.g., dexamethasone) form part of the standard protocol of treatment for patients with different malignancies for alleviation of symptoms, it was demonstrated that the antitumor response and presence of intracranial IL13BBζ in T cell-treated mice were not significantly impaired after dexamethasone was given, when compared to the control group, by Brown, Aguilar
[16].
Another molecule believed to be effective if used with CAR T cells is B7-H3, a type I transmembrane protein encoded by chromosome 15, which has costimulatory and co-inhibitory functions on T cell subsets
[17]. B7-H3.CAR with CD28 costimulation showed a faster antitumor effect in comparison with 4-1BB co-stimulation with no markable difference in antiproliferative activity in general
[18]. Although both costimulations showed cross-reactivity to murine B7-H3 without toxicity when infused systematically, and antitumor activity, using in vitro and xenograft GB murine models, allowed the elimination of both differentiated tumor cells and cancer stem cells (CSCs). The low expression of B7-H3 in one third of GB cells demonstrated effective killing by B7-H3.CAR-T cells
[18].
In an effort to understand the dynamics of the cytotoxic effects of T-cells in GB to establish better delivery of therapies, Murty, Haile
[19] used intravital microscopy to evidence the use of CAR-T cells along with focal radiation, achieving complete tumor regression in vivo. In another study, the IV administration of IL13BBζ was shown to be ineffective, possibly due to deficient cell trafficking to the intracranial tumors, pointing out intracranial therapy with CAR-T cells as a better option for long-term survival
[16].
Nevertheless, tumor recurrence is the top burden in the development of effective therapies for GB. Patients treated with IL13Rα2-targeted CAR-T cells showed recurrence with loss and/reduced expression of IL13Rα2 reducing the efficacy of the therapy and making it even more difficult after treatments
[8]. Moreover, to achieve complete eradication of GB, there are some barriers yet to concur, such as the suboptimal penetration of CAR-T cells within the tumor stroma, the poor effector function of T cells which inhibits a continuous antigen-driven stimulation and the minimal antigen specificity as a consequence of the heterogeneity of the GB tumor that could cause off-site toxicities
[19]. There is also a need for further investigation on how to enhance the endurance of CAR-T cells within the tumor environment to eradicate large tumors or even better early detection and eradication of tumor recurrence
[18].
1.1.2. Natural Killer (NK) Cells
The use of NK cells is the most preferred immunotherapy approach discussed in the literature regarding the GB treatment. The NK cells can be used for the targeted killing of glioma cells. Further, they can be used in combination with other immunotherapies including inhibitors for immune checkpoints, drugs targeting immune-related genes, or specific antibodies that block the action of proteins protecting NK cells from immunosuppression
[20][21][22][23]. Although a large body of evidence suggests a positive effect of NK cells as immunotherapy in GB treatment, the major hurdle is to mitigate the suppression of the cytotoxic effects of NK cells.
A study by Lee et al. reported the potential use of NK cells in the inhibition of systemic metastasis of GB cells in the mice model. This was attributed to the cytotoxic effects of NK cells against GB cells. Therefore, adequate supplementation of NK cells to the brain can be considered as a promising immunotherapy to treat GB
[21]. The killer Ig-like receptor (KIR) genotypes in NK cells are correlated with various tumor types. The presence of KIR2DS2 immuno-genotype NK cells was shown to be associated with more potent cytotoxic activity against GB
[20].
Another approach discussed in the literature includes the adoptive transfer of CAR-modified immune cells for the treatment of GB. A Han et al. study elucidated the use of CAR-engineered NK cells in the treatment of GB via targeting wild-type EGFR as well as mutant form EGFRvIII. These EGFR-CAR engineered NK cells demonstrated increased tumor cell lysis capacity, stimulated production of IFN-γ, and further suppressed the tumor growth and subsequently improved survival outcome for a long period
[24]. The observations of another study published in 2016 were concordant with the above-mentioned evidence
[25].
A study by Tanaka et al. reported another approach using a combination of ex vivo-expanded highly purified natural killer cells (genuine induced NK cells (GiNK)) and the chemotherapeutic agent temozolomide for the treatment of GB. This therapy has been shown to help to stimulate anticancer effects including the stimulation of tumor cell death in human GB cells in vitro
[26]. In addition, another study demonstrated that the use of the pretreatment approach of GB with another anticancer agent, bortezomib, helped to stimulate the cytotoxicity of NK cells by inducing TRAIL-R2 expression and enhanced GB lysis due to increased IFN-γ release
[27]. Recent research involving the treatment of NK cells with IL-2/HSP70 stimulated BBB crossing and the subsequent antitumor effects of NK cells and resulted in a substantial tumor growth inhibition and prolonged survival in an in vivo study
[28].
In a study published in 2005, tumor-derived RNA transfected dendritic cells (DCs) were shown to increase the cytotoxic ability of NK-like T cells by recognizing and killing the tumor cells using adaptive as well as innate immune systems, thereby enhancing antitumor effects against the tumor from which RNA was originated
[29].
Furthermore, NK cells were used as a vehicle for oncolytic enterovirus delivery in several recently published studies
[30][31]. Recently published evidence by Shaim et al. reported an innovative mechanism of NK cell immune evasion by GB stem cells by targeting the integrin-TGF-β axis, leading to its inhibition and consequently improving the antitumor effects of NK cells against GB
[32].