Next Article in Journal
Advanced Magnetic Resonance Imaging (MRI) Techniques: Technical Principles and Applications in Nanomedicine
Next Article in Special Issue
Biomarkers of Response and Resistance to Immunotherapy in Microsatellite Stable Colorectal Cancer: Toward a New Personalized Medicine
Previous Article in Journal
Regulation of Let-7a-5p and miR-199a-5p Expression by Akt1 Modulates Prostate Cancer Epithelial-to-Mesenchymal Transition via the Transforming Growth Factor-β Pathway
Previous Article in Special Issue
Serum sCD25 Protein as a Predictor of Lack of Long-Term Benefits from Immunotherapy in Non-Small Cell Lung Cancer: A Pilot Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Immunotherapy for the Treatment of Adult Glioblastoma: Overcoming Chemical and Physical Barriers

1
Foundation Medicine, Inc., Cambridge, MA 02141, USA
2
Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
3
Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
4
Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
5
Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(7), 1627; https://doi.org/10.3390/cancers14071627
Submission received: 28 February 2022 / Revised: 18 March 2022 / Accepted: 22 March 2022 / Published: 23 March 2022

Abstract

:

Simple Summary

The poor prognosis for glioblastoma (GBM) despite the existence of a standard-of-care treatment of resection, radiotherapy, and adjuvant chemotherapy has necessitated the exploration of other therapeutic avenues. One particularly promising avenue is an immunotherapeutic approach in which the body′s immune system is artificially stimulated to directly identify and attack the tumor cells. A variety of methods including immune checkpoint inhibition, T-cell transfer, vaccination, and a viral approach are being developed for GBM. Barriers such as tumor heterogeneity, the physical blood–brain barrier, the immunosuppressive nature of GBM, and the limited number of identifiable GBM-specific targets have reduced the efficacy of the aforementioned approaches. In the following review, we document the advances in immunotherapy, the barriers to implementation, and the development of a new technology (microbubble-enhanced focused ultrasound) to overcome the physical barriers to immunotherapy.

Abstract

Glioblastoma, or glioblastoma multiforme (GBM, WHO Grade IV), is a highly aggressive adult glioma. Despite extensive efforts to improve treatment, the current standard-of-care (SOC) regimen, which consists of maximal resection, radiotherapy, and temozolomide (TMZ), achieves only a 12–15 month survival. The clinical improvements achieved through immunotherapy in several extracranial solid tumors, including non-small-cell lung cancer, melanoma, and non-Hodgkin lymphoma, inspired investigations to pursue various immunotherapeutic interventions in adult glioblastoma patients. Despite some encouraging reports from preclinical and early-stage clinical trials, none of the tested agents have been convincing in Phase III clinical trials. One, but not the only, factor that is accountable for the slow progress is the blood–brain barrier, which prevents most antitumor drugs from reaching the target in appreciable amounts. Herein, we review the current state of immunotherapy in glioblastoma and discuss the significant challenges that prevent advancement. We also provide thoughts on steps that may be taken to remediate these challenges, including the application of ultrasound technologies.

1. Introduction

Glioblastoma is one of the most common primary malignant adult brain tumors, typified by its aggressiveness. The current standard-of-care treatment includes maximal resection and radiotherapy, followed by adjuvant chemotherapy with the DNA alkylator temozolomide [1]. The median overall survival (MOS) following GBM diagnosis is 12–15 months [1]. A multitude of factors complicates the treatment of GBM including (1) the heterogeneous nature of the tumors, both within a patient and between patients; and (2) the highly impermeable blood–brain barrier (BBB), which limits the effective delivery of many standard therapeutics.
A recent promising advancement has been an immunotherapeutic approach, which may involve either antagonizing the tumor′s inherent immune-suppressive properties or, conversely, inducing a glioma-specific immune response using either exogenous or endogenous agents. Immunotherapy has recently been popularized by the impressive outcomes in hematogenous malignancies [2]. However, for immunotherapy to be successful in solid tumors such as GBM, it must overcome tumor heterogeneity and the physical barriers imposed by the BBB and the tumor microenvironment (TME).
To overcome heterogeneity, key mutations which underlie GBM pathogenesis are continuously being elucidated so that targeted immunotherapeutics can be more effectively developed to combat the complexity of GBM. To overcome the BBB, ultrasound is being developed as a modality for transiently and noninvasively disrupting the BBB for the passage of therapeutics [3,4,5,6]. In the following review, we detail the advances in immunotherapies and how their efficacy can be enhanced by ultrasound technologies.

2. Current Immunotherapy Options and Developments

The human immune system is a complex regulatory environment that must constantly be able to distinguish between “self” and foreign matter. The immune system can be split into “innate” and “adaptive” immunity. Innate immunity does not improve with repeated encounters and consists of phagocytic cells (neutrophils, monocytes) and pro-inflammatory cells (eosinophils, basophils, and mast cells) [7]. Adaptive immunity learns and improves upon repeated exposure to pathogens. The main players in adaptive immunity are B and T lymphocytes, which produce antigen-specific immunoglobulins and induce foreign cell lysis [7]. Following activation, part of the immune system′s natural response is to return the hyperactive immune response to basal levels. Cells such as regulatory T-cells (Tregs) release anti-inflammatory cytokines leading to a diminished immune response [8]. Similarly, cell–cell signaling via inhibitory immunoreceptors such as PD-1, CTLA-4, LAG3, TIM3, TIGIT, and BTLA can attenuate an upregulated immune response [9]. The most promising immunotherapy approaches to treating glioblastoma are immune checkpoint inhibition [10,11], T-cell transfer therapy [12], vaccination [13], and oncolytic virus therapy (OVT). These methods harness the immune system to recognize and focally target tumor cells.

2.1. Immune Checkpoint Inhibitors

Immune checkpoint inhibitors (ICIs) avert the inactivation of CD8+ T-cells by preventing checkpoint receptors from binding with their ligands (Figure 1A). The critical immune checkpoint targets for ICIs in glioma include programmed cell death protein-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4). In the canonical pathway, when the PD-L1 ligand on the target cells interacts with the PD-1 receptor on the T-cells, intracellular tyrosine residues on the PD-1 cytoplasmic region lead to recruitment of Src homology 2 domain-containing protein tyrosine phosphatase-2 (SHP-2) [14]. This causes spleen tyrosine kinase (Syk) and phospholipid inositol-3-kinase (PI3K) to be phosphorylated, resulting in T-cell exhaustion and a suppressed immune response [14,15]. Glioblastoma cells can co-opt this machinery by overexpressing PD-L1, thereby evading the immune response [16]. Through a similar mechanism, glioblastoma cells also can upregulate CTLA-4, which promotes T-cell anergy through blockade of the B7/CD28 co-stimulatory signal [17].
A Phase III trial (CheckMate 143) compared the efficacy of the PD-1 inhibitor nivolumab, either alone or in combination with the CTLA-4 inhibitor ipilimumab, versus the vascular endothelial growth factor (VEGF) inhibitor bevacizumab in a subset of patients with recurrent glioblastoma (Table 1) [18]. Nivolumab was not superior to bevacizumab; the MOS was 9.8 and 10.0 months under nivolumab and bevacizumab, respectively. In another Phase III trial (CheckMate 498), nivolumab combined with radiotherapy also failed to prolong survival in patients with newly diagnosed MGMT-unmethylated glioblastoma compared with the SOC [19]. However, in a small randomized Phase II trial with 35 recurrent glioblastoma patients, the administration of the PD-1 inhibitor pembrolizumab before and after surgery significantly prolonged overall survival compared with adjuvant administration alone: 13.7 vs. 7.5 months, respectively [20].
In a single-arm Phase II study, a regimen consisting of avelumab (a monoclonal PD-L1 antibody) and axitinib (a tyrosine kinase inhibitor targeted against multiple VEGF receptors) was prescribed to patients with recurrent glioblastoma. It was well tolerated but failed to meet the study threshold for activity [21]. In another Phase II study, the addition of the PD-L1 inhibitor durvalumab to standard therapy moderately prolonged survival in patients with newly diagnosed MGMT-unmethylated glioblastoma compared with historical controls: 15.1 vs. 12.7 months, respectively [22]. The ambiguity of these clinical trial results involving CTLA-4 or PD-L1 inhibitors suggests the need for further research into a combinatorial approach, which may be feasible, given that each pathway leads to unique alterations in cytokine release [34].

2.2. T-Cell Transfer Therapies

T-cell transfer therapy or adoptive T-cell therapy is a type of immunotherapy that encompasses two main approaches: tumor-infiltrating lymphocyte (TIL) therapy and chimeric antigen receptor (CAR) T-cell therapy (Figure 1B).
In TIL therapy, T-lymphocytes invading the TME are collected via routine biopsy or surgery, isolated using fluorescence-activated cell sorting (FACS), and then selectively expanded using IL-2 stimulation [35,36,37]. The logic behind this approach is that T-cells found in or near the tumor already have a “proven track record” for identifying cancerous cells, but there are too few of them to overcome immunosuppression. Moreover, TIL therapy significantly reduces off-target effects due to their inherent specificity to the tumor [37]. Mathewson et al. performed single-cell transcriptome sequencing in a group of patients with isocitrate dehydrogenase (IDH)-mutant and IDH-wildtype glioblastoma [38]. They described the potential effectors of anti-tumor immunity in a population of cytotoxic TILs expressing several natural killer (NK) cell genes, including the CD161-encoding gene KLRB1. The inactivation of KLRB1 or antibody-mediated CD161 blockade resulted in increased T-cell cytotoxicity against tumor cells in vitro and an enhanced response in vivo.
CAR T-cell therapy introduces synthetic T-cell receptors into T-cells, which confer the ability to recognize tumor-specific surface antigens and initiate an MHC-independent immune response [39,40,41]. CAR T-cell therapy has had great efficacy in hematogenous malignancies but has been difficult to implement in solid tumors due to the immunosuppressive environment of the TME [2,40]. Moreover, solid tumors lack highly specific surface antigens, which can lead to numerous off-target effects when using CAR T-cell therapy. Two small Phase I trials tested CAR T-cell therapy in EGFRvIII-positive recurrent glioblastoma. Although EGFRvIII-targeted CAR T-cells found their way from peripheral blood to the tumor, no meaningful response was detected [23,42]. This lack of response to anti-EGFRvIII CAR T-cells may be attributable to the significant intra- and inter-tumoral heterogeneity of EGFRvIII expression in glioblastoma as well as to adaptive changes in the local TME, which include changes in antigen expression over time. For instance, following treatment, EGFRvIII was lost in a group of patients.

2.3. Vaccination

Tumor vaccines elicit an immune response against one or several tumor antigens (Figure 1C). Vaccines usually consist of peptides or proteins, but may also constitute antigen-laden dendritic cells. Immunostimulants such as poly ICLC are often co-administered with tumor vaccines to enhance adaptive immunity.
In a single-arm, multicenter, open-label Phase I trial performed in patients with newly diagnosed Grade 3 and 4 IDH1-mutant astrocytoma, an IDH1-specific peptide vaccine induced an immune response in 30 out of 32 (93.3%) patients [24]. The 3-year progression-free and overall survival rates were 63% and 84%, respectively. The 2-year progression-free rate among patients with an immune response was 82%, while the two patients without an immune response had tumor progression within 2 years of diagnosis.
Another vaccine approach involves a vaccination against survivin, an antiapoptotic protein expressed by many tumor types [25,43,44]. Survivin expression in GBM has been associated with increased recurrence, chemotherapy resistance, and poor overall prognosis [25,43,44,45,46]. The SurVaxM vaccine contains a synthetic long peptide mimic that spans the human survivin protein sequence; it expresses MHC Class I epitopes and stimulates the MHC Class II-restricted T-cell responses required for cytotoxic CD8+ T-cell activity against tumors [25]. A Phase I trial of SurVaxM against recurrent GBM demonstrated no serious adverse events and prolonged overall survival following vaccination (86.6 weeks) compared with historical overall survival (30 weeks) [25]. A subsequent study identified that glioma patients routinely expressed elevated serum levels of CD9+/GFAP+/SVN+ exosomes, associated with tumor progression, compared with healthy controls [47]. Patients treated with antisurvivin therapy showed decreased levels of these exosomes. Monitoring of CD9+/GFAP+/SVN+ exosomes may be a promising adjunct to the use of MRI in disease surveillance. Current trials are underway to evaluate SurVaxM’s efficacy in newly diagnosed GBM [26]. However, identifying plausible new vaccine targets for GBM remains difficult due to the heterogeneity of GBM tumors.

2.4. Oncolytic Virus Therapy

In recent years, the use of OVT has shown promise in the treatment of GBMs. OVT utilizes intratumoral delivery of viral vectors to either deliver oncolytic gene therapy into the TME or to cause direct cytotoxicity through viral infection and replication [48,49]. OVT also has pro-immunogenic effects due to the induction of immunogenic cell death (ICD) in infected tumor cells. In ICD, the destruction of tumor cells by OVT leads to the release of antigenic molecules into the TME which both recruits and activates local dendritic cells, with the subsequent stimulation of specific T-cells [49].
The earliest trials of oncolytic therapy in GBM used murine fibroblasts to deliver the replication-defective herpes simplex virus 1 (HSV1) thymidine kinase (tk) gene to GBMs, which conferred increased chemosensitivity to antiviral agents such as acyclovir, ganciclovir, and valganciclovir [48,50]. However, this trial failed to show prolonged survival in the OVT group, which was hypothesized to be the result of low gene transduction rates due to the nonmigratory nature of murine fibroblasts [50]. More recently, a genetically engineered replication selective HSV1 virus, G207, has shown safety and efficacy in clinical trials. G207 contains a deletion of the diploid γ134.5 neurovirulence gene and has viral ribonucleotide reductase (UL 39) disabled by the insertion of Escherichia coli lacZ. This allows for conditional replication in tumor cells while preventing the infection of normal cells [51]. A Phase I trial showed a median survival of 15.9 months in 13 GBM patients treated with intratumoral G207, with no evidence of HSV encephalitis [27,52]. A separate Phase I trial demonstrated the safety of G207 administration in conjunction with radiotherapy [27], while a more recent trial showed its safety in the treatment of pediatric high-grade gliomas [28]. HSV-vector mediated delivery of gene therapy offers significant promise in the treatment of GBM, and a current Phase I trial is investigating the use of a new drug, rQnestin34.5v.2, after a preclinical study suggested its low toxicity to humans [53,54].
Another development in OVT was the use of intratumoral injection of aglatimagene besadenovec (GliatakTM), a replication-defective adenovirus vector-mediated delivery of HSV1-tk (AdV-tk), in conjunction with subsequent valaciclovir therapy. Phase I trials of Gliatak conducted by Chiocca and colleagues demonstrated the safety of the therapy and an impressive radiographic response [55], while the Phase II trial showed a statistically significant improvement in the MOS of GBM patients treated with Gliatak after gross total resection (GTR) compared with patients treated with the standard of care after gross total resection (25.1 months vs. 16.3 months, respectively) [29]. Importantly, the survival benefit was even further improved at 2 and 3 years compared with the standard of care treatment, but no difference was noted if the resection was subtotal [29]. However, another Phase III clinical trial named the Aspect trial, which utilized AdV-tk, showed no significant improvement in overall survival when patients were treated with intratumoral injections of AdV-tk compared with the standard of care treatment group [30]. It should be noted that the ASPECT trial had uneven use of temozolomide, and radiotherapy was not administered concomitantly with the gene therapy [30]. Yet another Phase I trial evaluated the use of a human interferon-β-expressing adenovirus vector (Ad.hIFN-β). Intratumoral injection of Ad.hIFN-β was associated with a dose-related induction of apoptosis within tumors, but several patients experienced adverse effects and one patient experienced two serious dose-related adverse effects [56]. Ultimately, further investigation into adenovirus vectors is required.
The use of a live attenuated form of poliovirus has recently been studied as well. A Phase II clinical trial demonstrated that PVSRIPO, a live attenuated poliovirus Type 1 vaccine with its cognate internal ribosome entry site replaced by that of human rhinovirus Type 2 conferred an overall survival benefit [31]. Specifically, this randomized controlled trial (RCT) showed that the group treated with PVSRIPO had an overall survival rate of 21% at both 24 and 36 months, compared with 14% and 4% in the control group, respectively [31]. The foreign ribosomal entry site on PVSRIPO causes neuronal incompetence and ablates neurovirulence [57]. The effects of PVSRIPO are mediated by CD155, a Type 1 transmembrane glycoprotein receptor that is more commonly known as the poliovirus receptor [31,58,59,60]. CD155 is almost ubiquitously upregulated in solid tumors, including GBM, and it regulates natural killer (NK) cells and is part of the Ig-superfamily adhesion family response for cell motility and invasiveness [58,60,61]. When the PV capsid binds to CD155, the capsid protein is extruded and ultimately initiates the transfer of the viral RNA genome to the cytoplasm, then subsequently allows for the translation of the RNA and mediates the viral oncolytic effects [62]. Additional Phase II studies for PVSRIPO in conjunction with additional drugs are underway, with Phase III studies likely to commence in the foreseeable future.
Translating the success of early Phase I and Phase II trials to widespread clinical use has been challenging. Phase I and Phase II trials of the drug Toca 511 (Vocimagene amiretrorepvec), a γ retroviral replicating vector encoding a transgene for an optimized yeast cytosine deaminase, demonstrated both early safety and efficacy, with prolonged overall survival and complete responses in recurrent high-grade glioma and GBM compared with accepted survival rates in the literature [32]. However, in the Phase III arm of the clinical trial, the overall survival for patients treated with Toca 511 was 11 months compared with 12 months in the patient group receiving the standard-of-care treatment, with no significant difference between the two groups [33]. Toca 511′s Phase III failure underscores how challenging the introduction of new GBM therapies into the market has been. Several obstacles underlie these challenges in translating OVT into widespread clinical use. Pre-existing antibodies and the circulating complement in the peripheral vasculature may neutralize OVT particles before they are successfully delivered into the TME [63]. Moreover, uptake into nontarget organs (e.g., the liver) is a common barrier to efficient delivery [63]. As with the other therapeutic modalities, the BBB is a major obstacle to the effective delivery of any exogenous therapeutics.

3. Challenges to Immunotherapy

The equivocal results of clinical studies testing the four aforementioned immunotherapeutic agents in glioblastoma, compared with other solid tumor types, are largely due to three key factors of immune resistance: the blood–brain barrier and the brain–tumor barrier (BTB), the immunosuppressive microenvironment, and the low tumor mutational burden (TMB) of glioblastoma.

3.1. Blood–Brain/Brain–Tumor Barriers

The BBB is a semipermeable physiologic border that isolates the blood from the cerebrospinal fluid and the internal environment of the central nervous system (CNS) to preserve homeostasis and maintain normal brain function. The BBB comprises endothelial cells of the capillary wall, astrocyte endfeet wrapping the capillary, and pericytes of the capillary basement membrane (Figure 2) [64,65]. The endothelial cells making up the BBB are tightly linked through a series of tight junctions which prevent the paracellular passage of most large molecules. Astrocytes are a glial population that is well-known for regulating the synaptic junction but play a diverse set of roles in the CNS, one of which is regulation of the BBB. The insulation provided by astrocytic endfeet has shown to be compromised in various neural proteinopathies (e.g., Parkinson′s disease), where the lack of integrity of the BBB can lead to the accumulation of pathogenic solutes [66].
A subpopulation of GBM stem cells is localized in proximity to microvascular capillaries and subvert cerebrovascular tissue function to turn the BBB into a BTB. Even after a growing tumor damages the BBB, the newly formed BTB prohibits the optimal accumulation of drugs in the tumor (Figure 2). The BBB–BTB acts to shield the TME from therapeutics [67,68].

3.2. The Immune-Suppressive Microenvironment

The glioblastoma microenvironment is dominated by immunosuppressive tumor-associated macrophages (TAMs), which eliminate the effect of immunotherapy and promote tumor growth. Colony-stimulating factor 1 (CSF-1) and chemokine (C-C motif) ligand 2 (CCL2), which are overexpressed in glioblastoma, attract macrophages and determine their behavior. TAMs suppress antitumor immunity via at least two mechanisms: (1) the production of arginase and inducible nitric oxide synthase (iNOS); (2) surface expression of IL-4Rα. Arginase and iNOS restrict the proliferation of T-cells by depleting essential amino acids from the extracellular space [69]. Activation of IL-4Rα leads to the overexpression of transforming growth factor (TGF)-β, which, in turn, suppresses the IL-2-dependent survival of CD8+ T-cells and diminishes their activity by curbing the production of several effectors and immune-stimulatory molecules, including granzymes A and B, perforin, IL-6, IL-10, and IFN-γ [70]. Furthermore, TGF-β promotes the differentiation of naïve T-cells into (suppressor) Tregs. In GBM patients, the adenosine receptor pathway (A2aR/CD39/CD73), followed by PD-1, was found to be the most frequent immunomodulatory target in CD8+ cytotoxic T-cells obtained from the TME. Among various other immune markers profiled in GBM patients, A2aR expression was higher in TILs compared with the peripheral blood mononuclear cells (PBMCs) of GBM patients and PBMCs obtained from healthy donors [71]. The GBM TME induces hypoxia and cellular stress, leading to increased production of ATP, followed by its conversion to AMP by ectonucleoside CD39, ultimately resulting in the production of adenosine [72]. Increased levels of adenosine in the GBM TME suppress the effector function of TILs and recruit TAMs, contributing to immunosuppression [71,72].
Typically, leukocytes are absent from the brain parenchyma. However, a small number of T-cells can be found in the cerebrospinal fluid, choroid plexus stroma, and subarachnoid and perivascular spaces. Some of these T-cells escape from the capillaries in the event of a primary malignant brain tumor. Higher numbers of intratumoral CD8+ T-cells have been shown to correlate with better prognosis in several cancer types, including glioblastoma [73,74,75]. However, in glioblastoma, CD8+ T-cells comprise only 0–12% of all cells in the tumor. Besides, a significant fraction of these CD8+ T-cells show signs of exhaustion [76,77].
Cancer stem cells (CSCs) are a cellular subpopulation of GBMs which exhibit a unique form of immunosuppression. These CSCs are capable of self-renewal and differentiation, thereby repopulating the tumor niche [78]. They serve as a particularly complex barrier to treatment, as they are difficult to completely resect and display chemo/radioresistance [79]. CSCs can be isolated through the expression of the surface markers CD24, CD34, CD44, CD47, CD90, and CD133 [80]. CSCs have been shown to alter the immune microenvironment through the recruitment of TAMs, which help to maintain the self-renewal and maintenance capabilities of CSCs [81,82]. Additionally, CSCs display the ability to inhibit the proliferation of TILs through the upregulation of PD-L1 [78,81]. Conversely, CSCs may serve as a viable vaccine target, given that CSC lysates are more effective at generating a dendritic cell vaccine compared with whole tumor cell lysates [81,83]. The double-edged nature of CSCs warrants further investigation in regards to immunotherapy.
Further complicating the inherent GBM immunosuppressive environment, patient-dependent lifestyle choices can impact the immune system′s ability to mount a successful immune response. For example, in obesity, the immune system cellular profile changes from an anti-inflammatory/regulatory to a pro-inflammatory profile [84]. Interestingly, the data has been equivocal as to whether this shift to a pro-inflammatory state in obesity is beneficial, with studies both showing increased survival in obese patients with melanoma treated with ICIs, and obesity leading to tumor progression through T-cell aging [85,86,87]. Another common confounding lifestyle choice is the patient’s smoking history. Smoking both increases the TMB of tumors and alters the immunogenic microenvironment in a site-dependent manner [85,88,89,90]. Patient lifestyle choices must be contextualized in both the specific tumor subtype and anatomical localization.

3.3. Low Tumor Mutational Burden

TMB, defined as the total number of nonsynonymous mutations per coding area of a tumor′s genome, is a promising predictor of the response to treatment with immune checkpoint inhibitors in various cancers, including melanoma, renal cell carcinoma, and non-small-cell lung cancer [91,92]. Initially, TMB was determined using whole-exome sequencing of tumor samples, with targeted panel sequencing being currently explored [93,94]. The neoantigen and antigen burden in glioblastoma is generally low. Glioblastoma harbors a relatively insignificant number of mutations compared with immunogenic tumors, such as non-small cell lung cancer or melanoma. Only a few mutation-derived neoantigens have been predicted in glioma [13,95,96,97,98]. The expression levels of other, non-mutated targets (e.g., cancer germline antigens) are usually low. Recently, a study using multi-omics data from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) found that TMB was an independent marker of prognosis in diffuse glioma [99].
Based on a cut-off value between 0.64 and 0.67 mutations/Mb, Wang et al. classified 654 primary glioma patients from the TCGA database into TMB-high and TMB-low groups and revealed an inverse correlation between TMB and glioma grade [99]. As expected, an analysis of the distribution of nonsynonymous mutations showed that the TMB-high group had a higher incidence of mutations typical for glioblastoma (PTEN: 29% vs. 5%; EGFR: 17% vs. 5%), while the opposite was true for mutations associated with low-grade gliomas (IDH1: 77% vs. 7%). The patients with elevated TMB had, on average, less favorable outcomes than the patients with decreased TMB. The MOS was 23 months in the TMB-high group. Gene set enrichment analysis in the TMB-high group revealed enrichment in transcriptional programs associated with DNA replication and the cell cycle, indicating increased proliferative activity in high-TMB gliomas, which may, in part, explain the lack of treatment effect in these tumors. Further complicating these findings is the fact that glioblastoma TMB can increase following SOC treatment [100].
Gromeier et al. performed a genomic analysis of recurrent glioblastoma biopsy samples and determined that tumors harboring low TMB were more responsive to subsequent treatment with recombinant polio virotherapy (PVSRIPO) or immune checkpoint inhibitors [101]. They found that the patients who survived longer than 20 months after PVSRIPO treatment carried a TMB of less than 0.6 mutations/Mb. Stratifying overall survival following treatment with PVSRIPO or checkpoint inhibitors based on the median TMB (1.3 mutations/Mb) verified a more favorable response in patients carrying a below-median TMB in both cohorts. The difference remained significant even after excluding patients with hypermutation (>10 mutations/Mb). Notably, a correlation between survival and TMB has not been observed in immunotherapy-naïve primary or recurrent glioblastoma.

4. Strategies to Enhance Immunotherapy’s Effectiveness

The failure to achieve a meaningful clinical benefit through immunotherapy exposes the flaws of the current immunotherapeutic approaches in glioblastoma. The need for strategies to increase the sensitivity of glioblastoma to immunotherapeutic agents is evident. Finding ways to increase the influx of cytotoxic T-cells to the tumor, downregulate the immunosuppressive microenvironment, and target the low immunogenicity of glioblastoma could be some of the potential next steps. In addition, reflecting on and revising disease management is warranted. Specifically, developing alternatives to steroids (such as the glucocorticoid dexamethasone) for the effective control of edema in GBM patients is potentially crucial, because this would allow us to avoid steroid-induced immunosuppression [102,103].
One of the most logical targets for improving immunotherapy’s effectiveness is the BBB, given that the BBB is known to limit immune cell infiltration and antigen presentation in glioma. Recent findings have suggested promising strategies to mitigate this limitation. A compromised BBB increases the expression of tumor-associated antigens, as evidenced by the improved responses in glioblastoma patients with both pre- and post-surgical administration of PD-L1 blockers compared with adjuvant administration alone [20].
However, although the BBB is breached in glioblastoma, the disruption is heterogeneous, and thus sufficient delivery of an intravenously administered drug, such as nivolumab, to the entire TME has not been achieved [104]. Notably, the BBB restricts the general passage of compounds heavier than 400–600 Da and those that have a charge that is not intermediate or low, significantly hampering the treatment of brain tumors and CNS diseases [105]. For reference, the molecular mass of nivolumab is 146 kDa.
Physical modalities, such as noninvasive microbubble-enhanced focused ultrasound (MB-FUS) (Figure 3), can safely and transiently alter the permeability of the BBB/BTB without directly causing changes in the tumor cells. This technology has been demonstrated preclinically in numerous species, including nonhuman primates [3,106,107,108,109,110,111,112,113,114,115], and in multiple successful Phase I and IIa clinical trials executed by several different groups [4,5,116,117,118,119,120,121,122,123]. Ultrasound-mediated BBB disruption has been observed in normal brains [3,124], brains affected by neurodegenerative diseases (e.g., Parkinson′s disease and Alzheimer′s disease) [123], and brains with tumors [4,5,6]. Together, these studies have demonstrated the robustness of the technique. The temporary increase in permeability lasts between a few hours and several days, and depends on the type and dose of the microbubbles used and the ultrasound parameters [125,126,127,128,129,130,131]. The increased permeability occurs both through the opening of the tight junctions of the endothelium and through increased transcytosis [132]. These effects are nucleated by the gentle volumetric oscillation of the microbubbles when they are exposed to low-amplitude ultrasound, with the ultrasound amplitude being within the range used for diagnostic ultrasound imaging. Care must be taken to identify the appropriate ultrasound amplitude. If the amplitudes are too low, the barrier will not be disrupted, and for amplitudes that are too high, petechial hemorrhage may occur [133]. The emissions from the oscillating microbubbles can be used to identify the appropriate amplitudes in real time, providing patient- and treatment-specific guidance and control [134,135,136,137,138,139]. Phase I clinical trials have demonstrated the safety of this technology. Oscillation of the microbubbles not only increases the permeability of the blood–brain/tumor barrier but can also establish a convective flow that enhances the delivery of chemotherapeutics [140,141,142,143].
Because of the cavitation-dependent nature of barrier disruption, specific locations of disruption in the brain can be controlled with high precision based on where the ultrasound is focused in the brain. Multiple approaches have been pursued to obtain precise ultrasound insonation. The most common approach is magnetic resonance imaging-guided focused ultrasound (MRgFUS). This approach uses a stereotactic system and concurrent MR imaging to perform a real-time guided treatment, optimizing precision. In small animal preclinical models, the focused ultrasound transducer is typically a single element and targeting is achieved by physically moving the transducer [3,144,145,146]. More advanced systems use arrays of transducer elements and ultrasound beamforming to electronically steer the beam throughout the regions of interest in the brain [147,148,149]. A key advantage of these systems is that they can account for the effects of the skull when focusing the ultrasound. Additionally, the procedure avoids mechanical brain tissue shifts and eliminates the risk of infection. This approach has been used in clinical trials with the ExAblate Neuro system from Insightec [4,5,119,123,150]. A drawback of this approach is the financial expense associated with using the MR imaging system. An alternative methodology uses neuronavigation systems for targeting focused ultrasound transducers (either single-element or multi-element arrays) [124,151,152,153]. The NaviFUS system is testing this methodology clinically [154,155]. A recent Phase I immunostimulation study demonstrated safe delivery across the BBB [6]. The study was designed to determine if the ultrasound insonation induced immunostimulation without other therapeutics being administered (e.g., a chemotherapeutic). No immunostimulation was observed in the clinical trial participants. A follow-on preclinical study determined that immunostimulation occurred if the ultrasound’s pressure amplitude was increased above those used in the clinical trial [6]. Cavitation-mediated inflammation has also been observed in other preclinical studies using neuronavigation [156]. A third clinically investigated methodology uses surgically implanted ultrasound transducers [157,158,159]. The SonoCloud systems from CarThera have demonstrated increased barrier permeability in patients with recurrent glioblastoma and a trend toward increased survival with the co-administration of carboplatin [116,117,160].
Delivery of a wide range of potential therapeutics has been demonstrated in preclinical models, including chemotherapeutics [121,161,162], adenoviruses [163,164], antibodies [165,166], nanoparticles (NPs) [142,167,168,169], and whole cells [170,171]. Guo et al. demonstrated that NPs as large as 50 nm can achieve significant extravasation into the TME with the application of focused ultrasound [142]. NPs have a wide variety of formulations. Guo et al. used them as a lipid-based encapsulation method to protect therapeutic payloads from degradation as they traversed the vasculature to the TME. Their study also demonstrated that focused ultrasound delivery of RNA-loaded NPs significantly downregulated the expression of an oncogenic mRNA [169,172]. NPs have a use in immunotherapy, as they can be combined with anti-PD-L1 antibodies to focally target drug delivery to the TME [173,174,175]. Similarly, groups have used NPs to deliver CAR-T-cells in a mouse model of glioma [174,176]. NPs could also have a use in the delivery of vaccines or OVT, given the previously discussed barriers to the effective delivery of these therapies. Ultrasound-mediated delivery to specifically induce immune modulation and therapy has been previously described [146]. Approaches include the passage of IL-12 [177], immune checkpoint inhibitors [106,116,178,179,180,181], and natural killer cells [182]. In addition to transient disruption facilitating the diffusion of therapeutics into the brain, disruption of the BBB can also enable the release of tumor biomarkers, which can assist in assessing the treatment response [183].
A concern of ultrasound-mediated therapy is the potential adverse effects of multiple sonications. Park et al. performed repeated MRgFUS on patients with GBM receiving TMZ and found no clinical adverse effects during six ultrasound insonations [117]. Another concern is the risk of RBC extravasation due to permeabilization of the brain’s vasculature. However, fine control of the FUS parameters can avoid this risk [184,185]. Finally, FUS-induced mild inflammatory responses have been reported, with some variability in the literature [111,186]. In fact, some of these groups reported an increase in IL-12-mediated immune recognition, which could enhance an immunotherapeutic approach [177,187].
While there has been a significant and deserved emphasis on focused ultrasound to transiently permeabilize the blood–brain barrier, ablative ultrasound therapies can also enhance immune checkpoint inhibition [188]. Thermal ablative ultrasound therapy uses high-intensity focused ultrasound to increase the local temperature to 60 °C or higher to induce coagulative necrosis. It has been safely used in the brain to ablate neuronal tracks underlying the pathogenesis of essential tremor [189,190,191] and also to treat chronic neuropathic pain [192]. Preclinical evidence has indicated that ultrasound thermal ablation may work adjunctively with immune checkpoint inhibitors [193,194]. Furthermore, mechanically ablative ultrasound therapy (histotripsy) can also potentially enhance immune checkpoint inhibitors by stimulating nonimmunogenic “cold” tumors into becoming “hot” immunogenic tumors [195,196]. Common obstacles to this treatment approach are the interference of uniform ultrasound wave propagation through bone and gas, and organ movement during treatment, leading to collateral tissue damage [197].

5. Conclusions and Future Directions

Immunotherapy has recently become a highly researched potential therapeutic avenue for glioblastoma. The four main approaches are immune checkpoint inhibition, T-cell transfer therapy, vaccination, and oncolytic viral therapy. In regards to immune checkpoint inhibition, CTLA-4 and PD-L1 inhibitors have entered clinical trials, but inconsistent effects on prolonging the median survival time have slowed progress in this area of immunotherapy. T-cell transfer therapy has similarly drawn interest due to its success in hematogenous tumors. However, in solid cell tumors, clinical trials have shown equivocal results, likely due to a combination of low T-cell penetration of the TME and the need for a more diverse array of GBM molecular targets. Research is already underway for a bi- or tri-CAR T-cell approach in which a single T-cell can have multiple antigenic targets. Groups have started developing trivalent CAR T-cells simultaneously targeting HER2, IL-13Rα2, and EphA2 in murine models of GBM [198]. A vaccine approach has gained public attention due to the recent success of the SARS-CoV-2 mRNA vaccine. The SurVaxM vaccine against the oncoprotein survivin has had moderate success in recurrent GBM and warrants further exploration of other vaccine targets. Finally, several OVT therapies have entered clinical trials, with some exciting successes due to OVT′s unique ability to both induce tumor lysis and promote an immunogenic response. The main concern with OVT is the potential for normal cell infection.
Thus far, the clinical improvements achieved with the aforementioned immunotherapies for treating several extracranial solid tumors have been modest. The lack of clinical improvements can be attributed to several factors, including the low TMB of GBM, the immunosuppressive features of GBM, tumor heterogeneity [199], and the BBB and BTB. Critical advances need to be made in finding GBM-specific antigens for targeted immunotherapy, which may suggest the need for a combinatorial approach. Moreover, given the heterogeneity of GBM, specific subsets of GBM patients may preferentially benefit from certain immunotherapies; the challenge lies in determining which subpopulations would best benefit from which immunotherapies. Furthermore, advances in noninvasive MB-FUS may provide the transient permeabilization necessary to deliver these therapeutics while bypassing the BBB and directly targeting the TME. Advances have been made in NPs delivering mRNA payloads via a technique termed “selective organ targeting” (SORT) [200]. SORT molecules are added to the lipid nanoparticles’ outer layer, which aids in their delivery to specific organs. These targeting mechanisms would allow for better delivery of therapeutics to the TME. While advances need to be made before widespread clinical success can be achieved, immunotherapy combined with ultrasound-mediated delivery serves as a highly promising avenue for treating GBM and ultimately improving patient outcomes.

Author Contributions

M.L., R.R., S.S. (Sanjit Shah), M.M. and K.J.H. were involved in the drafting of the manuscript. D.B. and D.K.T. were involved in the editing of the manuscript. A.K. helped with designing figures for the manuscript. D.P.K. was involved in the drafting and editing of the manuscript. S.S. (Soma Sengupta) was involved in planning, detailing the content, and editing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

S.S.G. is supported by the Harold C. Schott Endowment and the Pam and Tom Mischell Funds. K.J.H. is supported by NIH. grant R01HL148451. D.P.K. and K.J.H. are supported by the UCGNI and Brain Tumor Center Pilot Funds.

Conflicts of Interest

D.P.K. is the president/CEO of Amlal Pharmaceuticals, Inc. Other authors have no conflict of interest to declare.

References

  1. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed]
  2. Melenhorst, J.J.; Chen, G.M.; Wang, M.; Porter, D.L.; Chen, C.; Collins, M.A.; Gao, P.; Bandyopadhyay, S.; Sun, H.; Zhao, Z.; et al. Decade-Long Leukaemia Remissions with Persistence of CD4+ CAR T Cells. Nature 2022, 602, 503–509. [Google Scholar] [CrossRef]
  3. Hynynen, K.; McDannold, N.; Vykhodtseva, N.; Jolesz, F.A. Noninvasive MR Imaging-Guided Focal Opening of the Blood-Brain Barrier in Rabbits. Radiology 2001, 220, 640–646. [Google Scholar] [CrossRef] [PubMed]
  4. Mainprize, T.; Lipsman, N.; Huang, Y.; Meng, Y.; Bethune, A.; Ironside, S.; Heyn, C.; Alkins, R.; Trudeau, M.; Sahgal, A.; et al. Blood-Brain Barrier Opening in Primary Brain Tumors with Non-Invasive MR-Guided Focused Ultrasound: A Clinical Safety and Feasibility Study. Sci. Rep. 2019, 9, 321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Abrahao, A.; Meng, Y.; Llinas, M.; Huang, Y.; Hamani, C.; Mainprize, T.; Aubert, I.; Heyn, C.; Black, S.E.; Hynynen, K.; et al. First-in-Human Trial of Blood–Brain Barrier Opening in Amyotrophic Lateral Sclerosis Using MR-Guided Focused Ultrasound. Nat. Commun. 2019, 10, 4373. [Google Scholar] [CrossRef] [Green Version]
  6. Chen, K.-T.; Chai, W.-Y.; Lin, Y.-J.; Lin, C.-J.; Chen, P.-Y.; Tsai, H.-C.; Huang, C.-Y.; Kuo, J.S.; Liu, H.-L.; Wei, K.-C. Neuronavigation-Guided Focused Ultrasound for Transcranial Blood-Brain Barrier Opening and Immunostimulation in Brain Tumors. Sci. Adv. 2021, 7, eabd0772. [Google Scholar] [CrossRef]
  7. Delves, P.J.; Roitt, I.M. The Immune System. N. Engl. J. Med. 2000, 343, 37–49. [Google Scholar] [CrossRef]
  8. Khattri, R.; Cox, T.; Yasayko, S.-A.; Ramsdell, F. An Essential Role for Scurfin in CD4+CD25+ T Regulatory Cells. Nat. Immunol. 2003, 4, 337–342. [Google Scholar] [CrossRef]
  9. He, X.; Xu, C. Immune Checkpoint Signaling and Cancer Immunotherapy. Cell Res. 2020, 30, 660–669. [Google Scholar] [CrossRef]
  10. Brahm, C.G.; van Linde, M.E.; Enting, R.H.; Schuur, M.; Otten, R.H.J.; Heymans, M.W.; Verheul, H.M.W.; Walenkamp, A.M.E. The Current Status of Immune Checkpoint Inhibitors in Neuro-Oncology: A Systematic Review. Cancers 2020, 12, 586. [Google Scholar] [CrossRef] [Green Version]
  11. Majd, N.; Dasgupta, P.; de Groot, J. Immunotherapy for Neuro-Oncology. Adv. Exp. Med. Biol. 2020, 1244, 183–203. [Google Scholar] [CrossRef] [PubMed]
  12. Maggs, L.; Cattaneo, G.; Dal, A.E.; Moghaddam, A.S.; Ferrone, S. CAR T Cell-Based Immunotherapy for the Treatment of Glioblastoma. Front. Neurosci. 2021, 15, 662064. [Google Scholar] [CrossRef] [PubMed]
  13. Keskin, D.B.; Anandappa, A.J.; Sun, J.; Tirosh, I.; Mathewson, N.D.; Li, S.; Oliveira, G.; Giobbie-Hurder, A.; Felt, K.; Gjini, E.; et al. Neoantigen Vaccine Generates Intratumoral T Cell Responses in Phase Ib Glioblastoma Trial. Nature 2019, 565, 234–239. [Google Scholar] [CrossRef] [PubMed]
  14. Jiang, Y.; Chen, M.; Nie, H.; Yuan, Y. PD-1 and PD-L1 in Cancer Immunotherapy: Clinical Implications and Future Considerations. Hum. Vaccines Immunother. 2019, 15, 1111–1122. [Google Scholar] [CrossRef] [PubMed]
  15. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 Pathway: Current Researches in Cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar]
  16. Khasraw, M.; Reardon, D.A.; Weller, M.; Sampson, J.H. PD-1 Inhibitors: Do They Have a Future in the Treatment of Glioblastoma? Clin. Cancer Res. 2020, 26, 5287–5296. [Google Scholar] [CrossRef]
  17. Liu, F.; Huang, J.; Liu, X.; Cheng, Q.; Luo, C.; Liu, Z. CTLA-4 Correlates with Immune and Clinical Characteristics of Glioma. Cancer Cell Int. 2020, 20, 7. [Google Scholar] [CrossRef]
  18. Reardon, D.A.; Omuro, A.; Brandes, A.A.; Rieger, J.; Wick, A.; Sepulveda, J.; Phuphanich, S.; de Souza, P.; Ahluwalia, M.S.; Lim, M.; et al. OS10.3 Randomized Phase 3 Study Evaluating the Efficacy and Safety of Nivolumab vs Bevacizumab in Patients with Recurrent Glioblastoma: CheckMate 143. Neuro-Oncology 2017, 19, iii21. [Google Scholar] [CrossRef] [Green Version]
  19. Sampson, J.H.; Omuro, A.M.P.; Preusser, M.; Lim, M.; Butowski, N.A.; Cloughesy, T.F.; Strauss, L.C.; Latek, R.R.; Paliwal, P.; Weller, M.; et al. A Randomized, Phase 3, Open-Label Study of Nivolumab versus Temozolomide (TMZ) in Combination with Radiotherapy (RT) in Adult Patients (Pts) with Newly Diagnosed, O-6-Methylguanine DNA Methyltransferase (MGMT)-Unmethylated Glioblastoma (GBM): CheckMate-498. J. Clin. Oncol. 2016, 34, TPS2079. [Google Scholar] [CrossRef]
  20. 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]
  21. Neyns, B.; Ben Salama, L.; Awada, G.; De Cremer, J.; Schwarze, J.K.; Seynaeve, L.; Du Four, S.; Fischbuch, L.; Vanbinst, A.-M.; Everaert, H.; et al. GLIAVAX: A Stratified Phase II Clinical Trial of Avelumab and Axitinib in Patients with Recurrent Glioblastoma. J. Clin. Oncol. 2019, 37, 2034. [Google Scholar] [CrossRef]
  22. Reardon, D.A.; Kaley, T.J.; Dietrich, J.; Clarke, J.L.; Dunn, G.; Lim, M.; Cloughesy, T.F.; Gan, H.K.; Park, A.J.; Schwarzenberger, P.; et al. Phase II Study to Evaluate Safety and Efficacy of MEDI4736 (Durvalumab) + Radiotherapy in Patients with Newly Diagnosed Unmethylated MGMT Glioblastoma (New Unmeth GBM). J. Clin. Oncol. 2019, 37, 2032. [Google Scholar] [CrossRef]
  23. Goff, S.L.; Morgan, R.A.; Yang, J.C.; Sherry, R.M.; Robbins, P.F.; Restifo, N.P.; Feldman, S.A.; Lu, Y.-C.; Lu, L.; Zheng, Z.; et al. Pilot Trial of Adoptive Transfer of Chimeric Antigen Receptor-Transduced T Cells Targeting EGFRvIII in Patients with Glioblastoma. J. Immunother. 2019, 42, 126–135. [Google Scholar] [CrossRef] [PubMed]
  24. Platten, M.; Bunse, L.; Wick, A.; Bunse, T.; Le Cornet, L.; Harting, I.; Sahm, F.; Sanghvi, K.; Tan, C.L.; Poschke, I.; et al. A Vaccine Targeting Mutant IDH1 in Newly Diagnosed Glioma. Nature 2021, 592, 463–468. [Google Scholar] [CrossRef] [PubMed]
  25. Fenstermaker, R.A.; Ciesielski, M.J.; Qiu, J.; Yang, N.; Frank, C.L.; Lee, K.P.; Mechtler, L.R.; Belal, A.; Ahluwalia, M.S.; Hutson, A.D. Clinical Study of a Survivin Long Peptide Vaccine (SurVaxM) in Patients with Recurrent Malignant Glioma. Cancer Immunol. Immunother. 2016, 65, 1339–1352. [Google Scholar] [CrossRef] [Green Version]
  26. MimiVax, LLC. Prospective Randomized Placebo-Controlled Trial of SurVaxM Plus Adjuvant Temozolomide for Newly Diagnosed Glioblastoma (SURVIVE). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT05163080 (accessed on 28 February 2022).
  27. Markert, J.M.; Razdan, S.N.; Kuo, H.-C.; Cantor, A.; Knoll, A.; Karrasch, M.; Nabors, L.B.; Markiewicz, M.; Agee, B.S.; Coleman, J.M.; et al. A Phase 1 Trial of Oncolytic HSV-1, G207, given in Combination with Radiation for Recurrent GBM Demonstrates Safety and Radiographic Responses. Mol. Ther. 2014, 22, 1048–1055. [Google Scholar] [CrossRef] [Green Version]
  28. Friedman, G.K.; Johnston, J.M.; Bag, A.K.; Bernstock, J.D.; Li, R.; Aban, I.; Kachurak, K.; Nan, L.; Kang, K.-D.; Totsch, S.; et al. Oncolytic HSV-1 G207 Immunovirotherapy for Pediatric High-Grade Gliomas. N. Engl. J. Med. 2021, 384, 1613–1622. [Google Scholar] [CrossRef]
  29. Wheeler, L.A.; Manzanera, A.G.; Bell, S.D.; Cavaliere, R.; McGregor, J.M.; Grecula, J.C.; Newton, H.B.; Lo, S.S.; Badie, B.; Portnow, J.; et al. Phase II Multicenter Study of Gene-Mediated Cytotoxic Immunotherapy as Adjuvant to Surgical Resection for Newly Diagnosed Malignant Glioma. Neuro-Oncology 2016, 18, 1137–1145. [Google Scholar] [CrossRef] [Green Version]
  30. Westphal, M.; Ylä-Herttuala, S.; Martin, J.; Warnke, P.; Menei, P.; Eckland, D.; Kinley, J.; Kay, R.; Ram, Z.; ASPECT Study Group. Adenovirus-Mediated Gene Therapy with Sitimagene Ceradenovec Followed by Intravenous Ganciclovir for Patients with Operable High-Grade Glioma (ASPECT): A Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2013, 14, 823–833. [Google Scholar] [CrossRef]
  31. 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]
  32. Cloughesy, T.F.; Landolfi, J.; Vogelbaum, M.A.; Ostertag, D.; Elder, J.B.; Bloomfield, S.; Carter, B.; Chen, C.C.; Kalkanis, S.N.; Kesari, S.; et al. Durable Complete Responses in Some Recurrent High-Grade Glioma Patients Treated with Toca 511 + Toca FC. Neuro-Oncology 2018, 20, 1383–1392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Cloughesy, T.F.; Petrecca, K.; Walbert, T.; Butowski, N.; Salacz, M.; Perry, J.; Damek, D.; Bota, D.; Bettegowda, C.; Zhu, J.-J.; et al. Effect of Vocimagene Amiretrorepvec in Combination with Flucytosine vs Standard of Care on Survival Following Tumor Resection in Patients with Recurrent High-Grade Glioma: A Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1939–1946. [Google Scholar] [CrossRef] [PubMed]
  34. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Cohen, C.J.; Gartner, J.J.; Horovitz-Fried, M.; Shamalov, K.; Trebska-McGowan, K.; Bliskovsky, V.V.; Parkhurst, M.R.; Ankri, C.; Prickett, T.D.; Crystal, J.S.; et al. Isolation of Neoantigen-Specific T Cells from Tumor and Peripheral Lymphocytes. J. Clin. Investig. 2015, 125, 3981–3991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rosenberg, S.A.; Packard, B.S.; Aebersold, P.M.; Solomon, D.; Topalian, S.L.; Toy, S.T.; Simon, P.; Lotze, M.T.; Yang, J.C.; Seipp, C.A.; et al. Use of Tumor-Infiltrating Lymphocytes and Interleukin-2 in the Immunotherapy of Patients with Metastatic Melanoma. N. Engl. J. Med. 1988, 319, 1676–1680. [Google Scholar] [CrossRef]
  37. Wang, S.; Sun, J.; Chen, K.; Ma, P.; Lei, Q.; Xing, S.; Cao, Z.; Sun, S.; Yu, Z.; Liu, Y.; et al. Perspectives of Tumor-Infiltrating Lymphocyte Treatment in Solid Tumors. BMC Med. 2021, 19, 140. [Google Scholar] [CrossRef]
  38. Mathewson, N.D.; Ashenberg, O.; Tirosh, I.; Gritsch, S.; Perez, E.M.; Marx, S.; Jerby-Arnon, L.; Chanoch-Myers, R.; Hara, T.; Richman, A.R.; et al. Inhibitory CD161 Receptor Identified in Glioma-Infiltrating T Cells by Single-Cell Analysis. Cell 2021, 184, 1281–1298. [Google Scholar] [CrossRef]
  39. Brown, C.E.; Mackall, C.L. CAR T Cell Therapy: Inroads to Response and Resistance. Nat. Rev. Immunol. 2019, 19, 73–74. [Google Scholar] [CrossRef]
  40. Watanabe, K.; Kuramitsu, S.; Posey, A.D.; June, C.H. Expanding the Therapeutic Window for CAR T Cell Therapy in Solid Tumors: The Knowns and Unknowns of CAR T Cell Biology. Front. Immunol. 2018, 9, 2486. [Google Scholar] [CrossRef] [Green Version]
  41. Majzner, R.G.; Mackall, C.L. Clinical Lessons Learned from the First Leg of the CAR T Cell Journey. Nat. Med. 2019, 25, 1341–1355. [Google Scholar] [CrossRef]
  42. 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, eaaa0984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Li, F.; Ambrosini, G.; Chu, E.Y.; Plescia, J.; Tognin, S.; Marchisio, P.C.; Altieri, D.C. Control of Apoptosis and Mitotic Spindle Checkpoint by Survivin. Nature 1998, 396, 580–584. [Google Scholar] [CrossRef]
  44. Ambrosini, G.; Adida, C.; Altieri, D.C. A Novel Anti-Apoptosis Gene, Survivin, Expressed in Cancer and Lymphoma. Nat. Med. 1997, 3, 917–921. [Google Scholar] [CrossRef] [PubMed]
  45. Kajiwara, Y.; Yamasaki, F.; Hama, S.; Yahara, K.; Yoshioka, H.; Sugiyama, K.; Arita, K.; Kurisu, K. Expression of Survivin in Astrocytic Tumors: Correlation with Malignant Grade and Prognosis. Cancer 2003, 97, 1077–1083. [Google Scholar] [CrossRef] [PubMed]
  46. Satoh, K.; Kaneko, K.; Hirota, M.; Masamune, A.; Satoh, A.; Shimosegawa, T. Expression of Survivin Is Correlated with Cancer Cell Apoptosis and Is Involved in the Development of Human Pancreatic Duct Cell Tumors. Cancer 2001, 92, 271–278. [Google Scholar] [CrossRef]
  47. Galbo, P.M.; Ciesielski, M.J.; Figel, S.; Maguire, O.; Qiu, J.; Wiltsie, L.; Minderman, H.; Fenstermaker, R.A. Circulating CD9+/GFAP+/Survivin+ Exosomes in Malignant Glioma Patients Following Survivin Vaccination. Oncotarget 2017, 8, 114722–114735. [Google Scholar] [CrossRef]
  48. Chiocca, E.A.; Nassiri, F.; Wang, J.; Peruzzi, P.; Zadeh, G. Viral and Other Therapies for Recurrent Glioblastoma: Is a 24-Month Durable Response Unusual? Neuro-Oncology 2019, 21, 14–25. [Google Scholar] [CrossRef]
  49. Marelli, G.; Howells, A.; Lemoine, N.R.; Wang, Y. Oncolytic Viral Therapy and the Immune System: A Double-Edged Sword Against Cancer. Front. Immunol. 2018, 9, 866. [Google Scholar] [CrossRef] [Green Version]
  50. Rainov, N.G. A Phase III Clinical Evaluation of Herpes Simplex Virus Type 1 Thymidine Kinase and Ganciclovir Gene Therapy as an Adjuvant to Surgical Resection and Radiation in Adults with Previously Untreated Glioblastoma Multiforme. Hum. Gene Ther. 2000, 11, 2389–2401. [Google Scholar] [CrossRef]
  51. Mineta, T.; Rabkin, S.D.; Yazaki, T.; Hunter, W.D.; Martuza, R.L. Attenuated Multi-Mutated Herpes Simplex Virus-1 for the Treatment of Malignant Gliomas. Nat. Med. 1995, 1, 938–943. [Google Scholar] [CrossRef]
  52. Markert, J.M.; Medlock, M.D.; Rabkin, S.D.; Gillespie, G.Y.; Todo, T.; Hunter, W.D.; Palmer, C.A.; Feigenbaum, F.; Tornatore, C.; Tufaro, F.; et al. Conditionally Replicating Herpes Simplex Virus Mutant, G207 for the Treatment of Malignant Glioma: Results of a Phase I Trial. Gene Ther. 2000, 7, 867–874. [Google Scholar] [CrossRef] [Green Version]
  53. Chiocca, E.A.; Nakashima, H.; Kasai, K.; Fernandez, S.A.; Oglesbee, M. Preclinical Toxicology of RQNestin34.5v.2: An Oncolytic Herpes Virus with Transcriptional Regulation of the ICP34.5 Neurovirulence Gene. Mol. Ther.-Methods Clin. Dev. 2020, 17, 871–893. [Google Scholar] [CrossRef] [PubMed]
  54. Chiocca, E.A. A Phase I Study of the Treatment of Recurrent Malignant Glioma with RQNestin34.5v.2, a Genetically Engineered HSV-1 Virus, and Immunomodulation with Cyclophosphamide. 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT03152318 (accessed on 28 February 2022).
  55. Chiocca, E.A.; Aguilar, L.K.; Bell, S.D.; Kaur, B.; Hardcastle, J.; Cavaliere, R.; McGregor, J.; Lo, S.; Ray-Chaudhuri, A.; Chakravarti, A.; et al. Phase IB Study of Gene-Mediated Cytotoxic Immunotherapy Adjuvant to up-Front Surgery and Intensive Timing Radiation for Malignant Glioma. J. Clin. Oncol. 2011, 29, 3611–3619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chiocca, E.A.; Smith, K.M.; McKinney, B.; Palmer, C.A.; Rosenfeld, S.; Lillehei, K.; Hamilton, A.; DeMasters, B.K.; Judy, K.; Kirn, D. A Phase I Trial of Ad.HIFN-β Gene Therapy for Glioma. Mol. Ther. 2008, 16, 618–626. [Google Scholar] [CrossRef] [PubMed]
  57. Gromeier, M.; Alexander, L.; Wimmer, E. Internal Ribosomal Entry Site Substitution Eliminates Neurovirulence in Intergeneric Poliovirus Recombinants. Proc. Natl. Acad. Sci. USA 1996, 93, 2370–2375. [Google Scholar] [CrossRef] [Green Version]
  58. Sloan, K.E.; Eustace, B.K.; Stewart, J.K.; Zehetmeier, C.; Torella, C.; Simeone, M.; Roy, J.E.; Unger, C.; Louis, D.N.; Ilag, L.L.; et al. CD155/PVR Plays a Key Role in Cell Motility during Tumor Cell Invasion and Migration. BMC Cancer 2004, 4, 73. [Google Scholar] [CrossRef] [Green Version]
  59. Lupo, K.B.; Matosevic, S. CD155 Immunoregulation as a Target for Natural Killer Cell Immunotherapy in Glioblastoma. J. Hematol. Oncol. 2020, 13, 76. [Google Scholar] [CrossRef]
  60. Brown, M.C.; Gromeier, M. Cytotoxic and Immunogenic Mechanisms of Recombinant Oncolytic Poliovirus. Curr. Opin. Virol. 2015, 13, 81–85. [Google Scholar] [CrossRef] [Green Version]
  61. Carlsten, M.; Norell, H.; Bryceson, Y.T.; Poschke, I.; Schedvins, K.; Ljunggren, H.-G.; Kiessling, R.; Malmberg, K.-J. Primary Human Tumor Cells Expressing CD155 Impair Tumor Targeting by Down-Regulating DNAM-1 on NK Cells. J. Immunol. 2009, 183, 4921–4930. [Google Scholar] [CrossRef] [Green Version]
  62. Strauss, M.; Filman, D.J.; Belnap, D.M.; Cheng, N.; Noel, R.T.; Hogle, J.M. Nectin-like Interactions between Poliovirus and Its Receptor Trigger Conformational Changes Associated with Cell Entry. J. Virol. 2015, 89, 4143–4157. [Google Scholar] [CrossRef] [Green Version]
  63. Wong, H.H.; Lemoine, N.R.; Wang, Y. Oncolytic Viruses for Cancer Therapy: Overcoming the Obstacles. Viruses 2010, 2, 78–106. [Google Scholar] [CrossRef] [PubMed]
  64. Sofroniew, M.V.; Vinters, H.V. Astrocytes: Biology and Pathology. Acta Neuropathol. 2010, 119, 7–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Brown, L.S.; Foster, C.G.; Courtney, J.-M.; King, N.E.; Howells, D.W.; Sutherland, B.A. Pericytes and Neurovascular Function in the Healthy and Diseased Brain. Front. Cell Neurosci. 2019, 13, 282. [Google Scholar] [CrossRef] [Green Version]
  66. Cabezas, R.; Ávila, M.; Gonzalez, J.; El-Bachá, R.S.; Báez, E.; García-Segura, L.M.; Jurado Coronel, J.C.; Capani, F.; Cardona-Gomez, G.P.; Barreto, G.E. Astrocytic Modulation of Blood Brain Barrier: Perspectives on Parkinson’s Disease. Front. Cell Neurosci. 2014, 8, 211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. 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]
  68. Dubois, L.G.; Campanati, L.; Righy, C.; D’Andrea-Meira, I.; de Sampaio e Spohr, T.C.L.; Porto-Carreiro, I.; Pereira, C.M.; Balça-Silva, J.; Kahn, S.A.; DosSantos, M.F.; et al. Gliomas and the Vascular Fragility of the Blood Brain Barrier. Front. Cell Neurosci. 2014, 8, 418. [Google Scholar] [CrossRef] [Green Version]
  69. Kamran, N.; Kadiyala, P.; Saxena, M.; Candolfi, M.; Li, Y.; Moreno-Ayala, M.A.; Raja, N.; Shah, D.; Lowenstein, P.R.; Castro, M.G. Immunosuppressive Myeloid Cells’ Blockade in the Glioma Microenvironment Enhances the Efficacy of Immune-Stimulatory Gene Therapy. Mol. Ther. 2017, 25, 232–248. [Google Scholar] [CrossRef] [Green Version]
  70. Bouffet, E.; Larouche, V.; Campbell, B.B.; Merico, D.; de Borja, R.; Aronson, M.; Durno, C.; Krueger, J.; Cabric, V.; Ramaswamy, V.; et al. Immune Checkpoint Inhibition for Hypermutant Glioblastoma Multiforme Resulting From Germline Biallelic Mismatch Repair Deficiency. J. Clin. Oncol. 2016, 34, 2206–2211. [Google Scholar] [CrossRef] [Green Version]
  71. Ott, M.; Tomaszowski, K.-H.; Marisetty, A.; Kong, L.-Y.; Wei, J.; Duna, M.; Blumberg, K.; Ji, X.; Jacobs, C.; Fuller, G.N.; et al. Profiling of Patients with Glioma Reveals the Dominant Immunosuppressive Axis Is Refractory to Immune Function Restoration. JCI Insight 2020, 5, 134386. [Google Scholar] [CrossRef]
  72. Takenaka, M.C.; Gabriely, G.; Rothhammer, V.; Mascanfroni, I.D.; Wheeler, M.A.; Chao, C.-C.; Gutiérrez-Vázquez, C.; Kenison, J.; Tjon, E.C.; Barroso, A.; et al. Control of Tumor-Associated Macrophages and T Cells in Glioblastoma via AHR and CD39. Nat. Neurosci. 2019, 22, 729–740. [Google Scholar] [CrossRef]
  73. Yang, I.; Tihan, T.; Han, S.J.; Wrensch, M.R.; Wiencke, J.; Sughrue, M.E.; Parsa, A.T. CD8+ T-Cell Infiltrate in Newly Diagnosed Glioblastoma Is Associated with Long-Term Survival. J. Clin. Neurosci. 2010, 17, 1381–1385. [Google Scholar] [CrossRef] [Green Version]
  74. Kmiecik, J.; Poli, A.; Brons, N.H.C.; Waha, A.; Eide, G.E.; Enger, P.Ø.; Zimmer, J.; Chekenya, M. Elevated CD3+ and CD8+ Tumor-Infiltrating Immune Cells Correlate with Prolonged Survival in Glioblastoma Patients despite Integrated Immunosuppressive Mechanisms in the Tumor Microenvironment and at the Systemic Level. J. Neuroimmunol. 2013, 264, 71–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Mostafa, H.; Pala, A.; Högel, J.; Hlavac, M.; Dietrich, E.; Westhoff, M.A.; Nonnenmacher, L.; Burster, T.; Georgieff, M.; Wirtz, C.R.; et al. Immune Phenotypes Predict Survival in Patients with Glioblastoma Multiforme. J. Hematol. Oncol. 2016, 9, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Varn, F.S.; Wang, Y.; Mullins, D.W.; Fiering, S.; Cheng, C. Systematic Pan-Cancer Analysis Reveals Immune Cell Interactions in the Tumor Microenvironment. Cancer Res. 2017, 77, 1271–1282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Orrego, E.; Castaneda, C.A.; Castillo, M.; Bernabe, L.A.; Casavilca, S.; Chakravarti, A.; Meng, W.; Garcia-Corrochano, P.; Villa-Robles, M.R.; Zevallos, R.; et al. Distribution of Tumor-Infiltrating Immune Cells in Glioblastoma. CNS Oncol. 2018, 7, CNS21. [Google Scholar] [CrossRef] [Green Version]
  78. Lathia, J.D.; Mack, S.C.; Mulkearns-Hubert, E.E.; Valentim, C.L.L.; Rich, J.N. Cancer Stem Cells in Glioblastoma. Genes Dev. 2015, 29, 1203–1217. [Google Scholar] [CrossRef] [Green Version]
  79. Wang, Z.-F.; Liao, F.; Wu, H.; Dai, J. Glioma Stem Cells-Derived Exosomal MiR-26a Promotes Angiogenesis of Microvessel Endothelial Cells in Glioma. J. Exp. Clin. Cancer Res. 2019, 38, 201. [Google Scholar] [CrossRef] [Green Version]
  80. Taniguchi, H.; Suzuki, Y.; Natori, Y. The Evolving Landscape of Cancer Stem Cells and Ways to Overcome Cancer Heterogeneity. Cancers 2019, 11, 532. [Google Scholar] [CrossRef] [Green Version]
  81. Lei, M.M.L.; Lee, T.K.W. Cancer Stem Cells: Emerging Key Players in Immune Evasion of Cancers. Front. Cell Dev. Biol. 2021, 9, 692940. [Google Scholar] [CrossRef]
  82. Sainz, B.; Carron, E.; Vallespinós, M.; Machado, H.L. Cancer Stem Cells and Macrophages: Implications in Tumor Biology and Therapeutic Strategies. Mediat. Inflamm. 2016, 2016, 9012369. [Google Scholar] [CrossRef] [Green Version]
  83. Xu, Q.; Liu, G.; Yuan, X.; Xu, M.; Wang, H.; Ji, J.; Konda, B.; Black, K.L.; Yu, J.S. Antigen-Specific T-Cell Response from Dendritic Cell Vaccination Using Cancer Stem-like Cell-Associated Antigens. Stem Cells 2009, 27, 1734–1740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. de Frel, D.L.; Atsma, D.E.; Pijl, H.; Seidell, J.C.; Leenen, P.J.M.; Dik, W.A.; van Rossum, E.F.C. The Impact of Obesity and Lifestyle on the Immune System and Susceptibility to Infections Such as COVID-19. Front. Nutr. 2020, 7, 597600. [Google Scholar] [CrossRef] [PubMed]
  85. Deshpande, R.P.; Sharma, S.; Watabe, K. The Confounders of Cancer Immunotherapy: Roles of Lifestyle, Metabolic Disorders and Sociological Factors. Cancers 2020, 12, 2983. [Google Scholar] [CrossRef] [PubMed]
  86. Lewis, D.E.; Lysaght, J.; Wu, H. Editorial: T Cell Alterations in Adipose Tissue During Obesity, HIV, and Cancer. Front. Immunol. 2019, 10, 1190. [Google Scholar] [CrossRef]
  87. McQuade, J.L.; Daniel, C.R.; Hess, K.R.; Mak, C.; Wang, D.Y.; Rai, R.R.; Park, J.J.; Haydu, L.E.; Spencer, C.; Wongchenko, M.; et al. Association of Body-Mass Index and Outcomes in Patients with Metastatic Melanoma Treated with Targeted Therapy, Immunotherapy, or Chemotherapy: A Retrospective, Multicohort Analysis. Lancet Oncol. 2018, 19, 310–322. [Google Scholar] [CrossRef] [Green Version]
  88. Desrichard, A.; Kuo, F.; Chowell, D.; Lee, K.-W.; Riaz, N.; Wong, R.J.; Chan, T.A.; Morris, L.G.T. Tobacco Smoking-Associated Alterations in the Immune Microenvironment of Squamous Cell Carcinomas. J. Natl. Cancer Inst. 2018, 110, 1386–1392. [Google Scholar] [CrossRef]
  89. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Mutational Landscape Determines Sensitivity to PD-1 Blockade in Non–Small Cell Lung Cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [Green Version]
  90. Norum, J.; Nieder, C. Tobacco Smoking and Cessation and PD-L1 Inhibitors in Non-Small Cell Lung Cancer (NSCLC): A Review of the Literature. ESMO Open 2018, 3, e000406. [Google Scholar] [CrossRef] [Green Version]
  91. Devarakonda, S.; Rotolo, F.; Tsao, M.-S.; Lanc, I.; Brambilla, E.; Masood, A.; Olaussen, K.A.; Fulton, R.; Sakashita, S.; McLeer-Florin, A.; et al. Tumor Mutation Burden as a Biomarker in Resected Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 2995–3006. [Google Scholar] [CrossRef]
  92. Hwang, W.L.; Wolfson, R.L.; Niemierko, A.; Marcus, K.J.; DuBois, S.G.; Haas-Kogan, D. Clinical Impact of Tumor Mutational Burden in Neuroblastoma. J. Natl. Cancer Inst. 2018, 111, 695–699. [Google Scholar] [CrossRef]
  93. Meléndez, B.; Van Campenhout, C.; Rorive, S.; Remmelink, M.; Salmon, I.; D’Haene, N. Methods of Measurement for Tumor Mutational Burden in Tumor Tissue. Transl. Lung Cancer Res. 2018, 7, 661–667. [Google Scholar] [CrossRef] [PubMed]
  94. Serratì, S.; De Summa, S.; Pilato, B.; Petriella, D.; Lacalamita, R.; Tommasi, S.; Pinto, R. Next-Generation Sequencing: Advances and Applications in Cancer Diagnosis. OncoTargets Ther. 2016, 9, 7355–7365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Charoentong, P.; Finotello, F.; Angelova, M.; Mayer, C.; Efremova, M.; Rieder, D.; Hackl, H.; Trajanoski, Z. Pan-Cancer Immunogenomic Analyses Reveal Genotype-Immunophenotype Relationships and Predictors of Response to Checkpoint Blockade. Cell Rep. 2017, 18, 248–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Hilf, N.; Kuttruff-Coqui, S.; Frenzel, K.; Bukur, V.; Stevanović, S.; Gouttefangeas, C.; Platten, M.; Tabatabai, G.; Dutoit, V.; van der Burg, S.H.; et al. Actively Personalized Vaccination Trial for Newly Diagnosed Glioblastoma. Nature 2019, 565, 240–245. [Google Scholar] [CrossRef] [PubMed]
  97. Weenink, B.; Draaisma, K.; Ooi, H.Z.; Kros, J.M.; Sillevis Smitt, P.A.E.; Debets, R.; French, P.J. Low-Grade Glioma Harbors Few CD8 T Cells, Which Is Accompanied by Decreased Expression of Chemo-Attractants, Not Immunogenic Antigens. Sci. Rep. 2019, 9, 14643. [Google Scholar] [CrossRef] [Green Version]
  98. Weenink, B.; French, P.J.; Sillevis Smitt, P.A.E.; Debets, R.; Geurts, M. Immunotherapy in Glioblastoma: Current Shortcomings and Future Perspectives. Cancers 2020, 12, 751. [Google Scholar] [CrossRef] [Green Version]
  99. Wang, L.; Ge, J.; Lan, Y.; Shi, Y.; Luo, Y.; Tan, Y.; Liang, M.; Deng, S.; Zhang, X.; Wang, W.; et al. Tumor Mutational Burden Is Associated with Poor Outcomes in Diffuse Glioma. BMC Cancer 2020, 20, 213. [Google Scholar] [CrossRef]
  100. Valero, C.; Lee, M.; Hoen, D.; Wang, J.; Nadeem, Z.; Patel, N.; Postow, M.A.; Shoushtari, A.N.; Plitas, G.; Balachandran, V.P.; et al. The Association between Tumor Mutational Burden and Prognosis Is Dependent on Treatment Context. Nat. Genet. 2021, 53, 11–15. [Google Scholar] [CrossRef]
  101. Gromeier, M.; Brown, M.C.; Zhang, G.; Lin, X.; Chen, Y.; Wei, Z.; Beaubier, N.; Yan, H.; He, Y.; Desjardins, A.; et al. Very Low Mutation Burden Is a Feature of Inflamed Recurrent Glioblastomas Responsive to Cancer Immunotherapy. Nat. Commun. 2021, 12, 352. [Google Scholar] [CrossRef]
  102. Schiff, D.; Lee, E.Q.; Nayak, L.; Norden, A.D.; Reardon, D.A.; Wen, P.Y. Medical Management of Brain Tumors and the Sequelae of Treatment. Neuro-Oncology 2015, 17, 488–504. [Google Scholar] [CrossRef] [Green Version]
  103. Murayi, R.; Chittiboina, P. Glucocorticoids in the Management of Peritumoral Brain Edema: A Review of Molecular Mechanisms. Child’s Nerv. Syst. 2016, 32, 2293–2302. [Google Scholar] [CrossRef]
  104. Sarkaria, J.N.; Hu, L.S.; Parney, I.F.; Pafundi, D.H.; Brinkmann, D.H.; Laack, N.N.; Giannini, C.; Burns, T.C.; Kizilbash, S.H.; Laramy, J.K.; et al. Is the Blood-Brain Barrier Really Disrupted in All Glioblastomas? A Critical Assessment of Existing Clinical Data. Neuro-Oncology 2018, 20, 184–191. [Google Scholar] [CrossRef] [PubMed]
  105. Banks, W.A. Characteristics of Compounds That Cross the Blood-Brain Barrier. BMC Neurol. 2009, 9, S3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. 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]
  107. Poon, C.; McMahon, D.; Hynynen, K. Noninvasive and Targeted Delivery of Therapeutics to the Brain Using Focused Ultrasound. Neuropharmacology 2017, 120, 20–37. [Google Scholar] [CrossRef] [PubMed]
  108. Aryal, M.; Vykhodtseva, N.; Zhang, Y.-Z.; Park, J.; McDannold, N. Multiple Treatments with Liposomal Doxorubicin and Ultrasound-Induced Disruption of Blood-Tumor and Blood-Brain Barriers Improve Outcomes in a Rat Glioma Model. J. Control. Release 2013, 169, 103–111. [Google Scholar] [CrossRef] [Green Version]
  109. McDannold, N.; Arvanitis, C.D.; Vykhodtseva, N.; Livingstone, M.S. Temporary Disruption of the Blood-Brain Barrier by Use of Ultrasound and Microbubbles: Safety and Efficacy Evaluation in Rhesus Macaques. Cancer Res. 2012, 72, 3652–3663. [Google Scholar] [CrossRef] [Green Version]
  110. Lynch, M.; Heinen, S.; Markham-Coultes, K.; O’Reilly, M.; Van Slyke, P.; Dumont, D.J.; Hynynen, K.; Aubert, I. Vasculotide Restores the Blood-Brain Barrier after Focused Ultrasound-Induced Permeability in a Mouse Model of Alzheimer’s Disease. Int. J. Med. Sci. 2021, 18, 482–493. [Google Scholar] [CrossRef]
  111. Kovacs, Z.I.; Kim, S.; Jikaria, N.; Qureshi, F.; Milo, B.; Lewis, B.K.; Bresler, M.; Burks, S.R.; Frank, J.A. Disrupting the Blood-Brain Barrier by Focused Ultrasound Induces Sterile Inflammation. Proc. Natl. Acad. Sci. USA 2017, 114, E75–E84. [Google Scholar] [CrossRef] [Green Version]
  112. Shi, L.; Palacio-Mancheno, P.; Badami, J.; Shin, D.W.; Zeng, M.; Cardoso, L.; Tu, R.; Fu, B.M. Quantification of Transient Increase of the Blood-Brain Barrier Permeability to Macromolecules by Optimized Focused Ultrasound Combined with Microbubbles. Int. J. Nanomed. 2014, 9, 4437–4448. [Google Scholar] [CrossRef] [Green Version]
  113. Marquet, F.; Tung, Y.-S.; Teichert, T.; Ferrera, V.P.; Konofagou, E.E. Noninvasive, Transient and Selective Blood-Brain Barrier Opening in Non-Human Primates In Vivo. PLoS ONE 2011, 6, e22598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. McDannold, N.; Zhang, Y.; Supko, J.G.; Power, C.; Sun, T.; Vykhodtseva, N.; Golby, A.J.; Reardon, D.A. Blood-Brain Barrier Disruption and Delivery of Irinotecan in a Rat Model Using a Clinical Transcranial MRI-Guided Focused Ultrasound System. Sci. Rep. 2020, 10, 8766. [Google Scholar] [CrossRef] [PubMed]
  115. Todd, N.; Angolano, C.; Ferran, C.; Devor, A.; Borsook, D.; McDannold, N. Secondary Effects on Brain Physiology Caused by Focused Ultrasound-Mediated Disruption of the Blood-Brain Barrier. J. Control. Release 2020, 324, 450–459. [Google Scholar] [CrossRef] [PubMed]
  116. Carpentier, A.; Canney, M.; Vignot, A.; Reina, V.; Beccaria, K.; Horodyckid, C.; Karachi, C.; Leclercq, D.; Lafon, C.; Chapelon, J.-Y.; et al. Clinical Trial of Blood-Brain Barrier Disruption by Pulsed Ultrasound. Sci. Transl. Med. 2016, 8, 343re2. [Google Scholar] [CrossRef] [PubMed]
  117. Idbaih, A.; Canney, M.; Belin, L.; Desseaux, C.; Vignot, A.; Bouchoux, G.; Asquier, N.; Law-Ye, B.; Leclercq, D.; Bissery, A.; et al. Safety and Feasibility of Repeated and Transient Blood-Brain Barrier Disruption by Pulsed Ultrasound in Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2019, 25, 3793–3801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Meng, Y.; MacIntosh, B.J.; Shirzadi, Z.; Kiss, A.; Bethune, A.; Heyn, C.; Mithani, K.; Hamani, C.; Black, S.E.; Hynynen, K.; et al. Resting State Functional Connectivity Changes after MR-Guided Focused Ultrasound Mediated Blood-Brain Barrier Opening in Patients with Alzheimer’s Disease. Neuroimage 2019, 200, 275–280. [Google Scholar] [CrossRef]
  119. Rezai, A.R.; Ranjan, M.; D’Haese, P.-F.; Haut, M.W.; Carpenter, J.; Najib, U.; Mehta, R.I.; Chazen, J.L.; Zibly, Z.; Yates, J.R.; et al. Noninvasive Hippocampal Blood−brain Barrier Opening in Alzheimer’s Disease with Focused Ultrasound. Proc. Natl. Acad. Sci. USA 2020, 117, 9180–9182. [Google Scholar] [CrossRef] [Green Version]
  120. Deng, Z.; Sheng, Z.; Yan, F. Ultrasound-Induced Blood-Brain-Barrier Opening Enhances Anticancer Efficacy in the Treatment of Glioblastoma: Current Status and Future Prospects. J. Oncol. 2019, 2019, e2345203. [Google Scholar] [CrossRef]
  121. Alli, S.; Figueiredo, C.A.; Golbourn, B.; Sabha, N.; Wu, M.Y.; Bondoc, A.; Luck, A.; Coluccia, D.; Maslink, C.; Smith, C.; et al. Brainstem Blood Brain Barrier Disruption Using Focused Ultrasound: A Demonstration of Feasibility and Enhanced Doxorubicin Delivery. J. Control. Release 2018, 281, 29–41. [Google Scholar] [CrossRef]
  122. Gasca-Salas, C.; Fernández-Rodríguez, B.; Pineda-Pardo, J.A.; Rodríguez-Rojas, R.; Obeso, I.; Hernández-Fernández, F.; del Álamo, M.; Mata, D.; Guida, P.; Ordás-Bandera, C.; et al. Blood-Brain Barrier Opening with Focused Ultrasound in Parkinson’s Disease Dementia. Nat. Commun. 2021, 12, 779. [Google Scholar] [CrossRef]
  123. Lipsman, N.; Meng, Y.; Bethune, A.J.; Huang, Y.; Lam, B.; Masellis, M.; Herrmann, N.; Heyn, C.; Aubert, I.; Boutet, A.; et al. Blood–Brain Barrier Opening in Alzheimer’s Disease Using MR-Guided Focused Ultrasound. Nat. Commun. 2018, 9, 2336. [Google Scholar] [CrossRef] [Green Version]
  124. Pouliopoulos, A.N.; Kwon, N.; Jensen, G.; Meaney, A.; Niimi, Y.; Burgess, M.T.; Ji, R.; McLuckie, A.J.; Munoz, F.A.; Kamimura, H.A.S.; et al. Safety Evaluation of a Clinical Focused Ultrasound System for Neuronavigation Guided Blood-Brain Barrier Opening in Non-Human Primates. Sci. Rep. 2021, 11, 15043. [Google Scholar] [CrossRef] [PubMed]
  125. Conti, A.; Mériaux, S.; Larrat, B. About the Marty Model of Blood-Brain Barrier Closure after Its Disruption Using Focused Ultrasound. Phys. Med. Biol. 2019, 64, 14NT02. [Google Scholar] [CrossRef] [PubMed]
  126. McDannold, N.; Vykhodtseva, N.; Hynynen, K. Effects of Acoustic Parameters and Ultrasound Contrast Agent Dose on Focused-Ultrasound Induced Blood-Brain Barrier Disruption. Ultrasound Med. Biol. 2008, 34, 930–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Marty, B.; Larrat, B.; Van Landeghem, M.; Robic, C.; Robert, P.; Port, M.; Le Bihan, D.; Pernot, M.; Tanter, M.; Lethimonnier, F.; et al. Dynamic Study of Blood-Brain Barrier Closure after Its Disruption Using Ultrasound: A Quantitative Analysis. J. Cereb. Blood Flow Metab. 2012, 32, 1948–1958. [Google Scholar] [CrossRef]
  128. Park, J.; Zhang, Y.; Vykhodtseva, N.; Jolesz, F.A.; McDannold, N.J. The Kinetics of Blood Brain Barrier Permeability and Targeted Doxorubicin Delivery into Brain Induced by Focused Ultrasound. J. Control. Release 2012, 162, 134–142. [Google Scholar] [CrossRef] [Green Version]
  129. O’Reilly, M.A.; Waspe, A.C.; Ganguly, M.; Hynynen, K. Focused-Ultrasound Disruption of the Blood-Brain Barrier Using Closely-Timed Short Pulses: Influence of Sonication Parameters and Injection Rate. Ultrasound Med. Biol. 2011, 37, 587–594. [Google Scholar] [CrossRef] [Green Version]
  130. Samiotaki, G.; Vlachos, F.; Tung, Y.-S.; Konofagou, E.E. A Quantitative Pressure and Microbubble-Size Dependence Study of Focused Ultrasound-Induced Blood-Brain Barrier Opening Reversibility in Vivo Using MRI. Magn. Reson. Med. 2012, 67, 769–777. [Google Scholar] [CrossRef] [Green Version]
  131. Chen, K.-T.; Wei, K.-C.; Liu, H.-L. Theranostic Strategy of Focused Ultrasound Induced Blood-Brain Barrier Opening for CNS Disease Treatment. Front. Pharmacol. 2019, 10, 86. [Google Scholar] [CrossRef] [Green Version]
  132. Aryal, M.; Arvanitis, C.D.; Alexander, P.M.; McDannold, N. Ultrasound-Mediated Blood-Brain Barrier Disruption for Targeted Drug Delivery in the Central Nervous System. Adv. Drug Deliv. Rev. 2014, 72, 94–109. [Google Scholar] [CrossRef] [Green Version]
  133. McDannold, N.; Vykhodtseva, N.; Hynynen, K. Use of Ultrasound Pulses Combined with Definity for Targeted Blood-Brain Barrier Disruption: A Feasibility Study. Ultrasound Med. Biol. 2007, 33, 584–590. [Google Scholar] [CrossRef] [Green Version]
  134. Chu, P.-C.; Liu, H.-L.; Lai, H.-Y.; Lin, C.-Y.; Tsai, H.-C.; Pei, Y.-C. Neuromodulation Accompanying Focused Ultrasound-Induced Blood-Brain Barrier Opening. Sci. Rep. 2015, 5, 15477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. McDannold, N.; Vykhodtseva, N.; Hynynen, K. Targeted Disruption of the Blood-Brain Barrier with Focused Ultrasound: Association with Cavitation Activity. Phys. Med. Biol. 2006, 51, 793–807. [Google Scholar] [CrossRef]
  136. O’Reilly, M.A.; Hynynen, K. Blood-Brain Barrier: Real-Time Feedback-Controlled Focused Ultrasound Disruption by Using an Acoustic Emissions-Based Controller. Radiology 2012, 263, 96–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Sun, T.; Samiotaki, G.; Wang, S.; Acosta, C.; Chen, C.C.; Konofagou, E.E. Acoustic Cavitation-Based Monitoring of the Reversibility and Permeability of Ultrasound-Induced Blood-Brain Barrier Opening. Phys. Med. Biol. 2015, 60, 9079–9094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Sun, T.; Zhang, Y.; Power, C.; Alexander, P.M.; Sutton, J.T.; Aryal, M.; Vykhodtseva, N.; Miller, E.L.; McDannold, N.J. Closed-Loop Control of Targeted Ultrasound Drug Delivery across the Blood–Brain/Tumor Barriers in a Rat Glioma Model. Proc. Natl. Acad. Sci. USA 2017, 114, E10281–E10290. [Google Scholar] [CrossRef] [Green Version]
  139. Patel, A.; Schoen, S.J.; Arvanitis, C.D. Closed-Loop Spatial and Temporal Control of Cavitation Activity with Passive Acoustic Mapping. IEEE Trans. Biomed. Eng. 2019, 66, 2022–2031. [Google Scholar] [CrossRef]
  140. Collis, J.; Manasseh, R.; Liovic, P.; Tho, P.; Ooi, A.; Petkovic-Duran, K.; Zhu, Y. Cavitation Microstreaming and Stress Fields Created by Microbubbles. Ultrasonics 2010, 50, 273–279. [Google Scholar] [CrossRef]
  141. Sutton, J.T.; Haworth, K.J.; Pyne-Geithman, G.; Holland, C.K. Ultrasound-Mediated Drug Delivery for Cardiovascular Disease. Expert Opin. Drug Deliv. 2013, 10, 573–592. [Google Scholar] [CrossRef] [Green Version]
  142. Guo, Y.; Lee, H.; Fang, Z.; Velalopoulou, A.; Kim, J.; Ben Thomas, M.; Liu, J.; Abramowitz, R.G.; Kim, Y.; Coskun, A.F.; et al. Single-Cell Analysis Reveals Effective SiRNA Delivery in Brain Tumors with Microbubble-Enhanced Ultrasound and Cationic Nanoparticles. Sci. Adv. 2021, 7, eabf7390. [Google Scholar] [CrossRef]
  143. Anastasiadis, P.; Gandhi, D.; Guo, Y.; Ahmed, A.-K.; Bentzen, S.M.; Arvanitis, C.; Woodworth, G.F. Localized Blood–Brain Barrier Opening in Infiltrating Gliomas with MRI-Guided Acoustic Emissions–Controlled Focused Ultrasound. Proc. Natl. Acad. Sci. USA 2021, 118, e2103280118. [Google Scholar] [CrossRef] [PubMed]
  144. Choi, J.J.; Pernot, M.; Small, S.A.; Konofagou, E.E. Noninvasive, Transcranial and Localized Opening of the Blood-Brain Barrier Using Focused Ultrasound in Mice. Ultrasound Med. Biol. 2007, 33, 95–104. [Google Scholar] [CrossRef] [PubMed]
  145. Morse, S.V.; Pouliopoulos, A.N.; Chan, T.G.; Copping, M.J.; Lin, J.; Long, N.J.; Choi, J.J. Rapid Short-Pulse Ultrasound Delivers Drugs Uniformly across the Murine Blood-Brain Barrier with Negligible Disruption. Radiology 2019, 291, 459–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Beccaria, K.; Sabbagh, A.; de Groot, J.; Canney, M.; Carpentier, A.; Heimberger, A.B. Blood-Brain Barrier Opening with Low Intensity Pulsed Ultrasound for Immune Modulation and Immune Therapeutic Delivery to CNS Tumors. J. Neuro-Oncol. 2021, 151, 65–73. [Google Scholar] [CrossRef]
  147. Hynynen, K.; McDannold, N.; Clement, G.; Jolesz, F.A.; Zadicario, E.; Killiany, R.; Moore, T.; Rosen, D. Pre-Clinical Testing of a Phased Array Ultrasound System for MRI-Guided Noninvasive Surgery of the Brain—A Primate Study. Eur. J. Radiol. 2006, 59, 149–156. [Google Scholar] [CrossRef]
  148. Song, J.; Hynynen, K. Feasibility of Using Lateral Mode Coupling Method for a Large Scale Ultrasound Phased Array for Noninvasive Transcranial Therapy. IEEE Trans. Biomed. Eng. 2010, 57, 124–133. [Google Scholar] [CrossRef] [Green Version]
  149. O’Reilly, M.A.; Jones, R.M.; Hynynen, K. Three-Dimensional Transcranial Ultrasound Imaging of Microbubble Clouds Using a Sparse Hemispherical Array. IEEE Trans. Biomed. Eng. 2014, 61, 1285–1294. [Google Scholar] [CrossRef] [Green Version]
  150. Park, S.H.; Kim, M.J.; Jung, H.H.; Chang, W.S.; Choi, H.S.; Rachmilevitch, I.; Zadicario, E.; Chang, J.W. One-Year Outcome of Multiple Blood–Brain Barrier Disruptions with Temozolomide for the Treatment of Glioblastoma. Front. Oncol. 2020, 10, 1663. [Google Scholar] [CrossRef]
  151. Wei, K.-C.; Tsai, H.-C.; Lu, Y.-J.; Yang, H.-W.; Hua, M.-Y.; Wu, M.-F.; Chen, P.-Y.; Huang, C.-Y.; Yen, T.-C.; Liu, H.-L. Neuronavigation-Guided Focused Ultrasound-Induced Blood-Brain Barrier Opening: A Preliminary Study in Swine. AJNR Am. J. Neuroradiol. 2013, 34, 115–120. [Google Scholar] [CrossRef] [Green Version]
  152. Wu, S.-Y.; Aurup, C.; Sanchez, C.S.; Grondin, J.; Zheng, W.; Kamimura, H.; Ferrera, V.P.; Konofagou, E.E. Efficient Blood-Brain Barrier Opening in Primates with Neuronavigation-Guided Ultrasound and Real-Time Acoustic Mapping. Sci. Rep. 2018, 8, 7978. [Google Scholar] [CrossRef]
  153. Pouliopoulos, A.N.; Wu, S.-Y.; Burgess, M.T.; Karakatsani, M.E.; Kamimura, H.A.S.; Konofagou, E.E. A Clinical System for Non-Invasive Blood-Brain Barrier Opening Using a Neuronavigation-Guided Single-Element Focused Ultrasound Transducer. Ultrasound Med. Biol. 2020, 46, 73–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Chen, K.-T.; Lin, Y.-J.; Chai, W.-Y.; Lin, C.-J.; Chen, P.-Y.; Huang, C.-Y.; Kuo, J.S.; Liu, H.-L.; Wei, K.-C. Neuronavigation-Guided Focused Ultrasound (NaviFUS) for Transcranial Blood-Brain Barrier Opening in Recurrent Glioblastoma Patients: Clinical Trial Protocol. Ann. Transl. Med. 2020, 8, 673. [Google Scholar] [CrossRef] [PubMed]
  155. Wu, C.-C. A Feasibility Study Examining the Use of Non-Invasive Focused Ultrasound (FUS) with Oral Panobinostat Administration in Children with Progressive Diffuse Midline Glioma (DMG). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT04804709 (accessed on 28 February 2022).
  156. Ji, R.; Karakatsani, M.E.; Burgess, M.; Smith, M.; Murillo, M.F.; Konofagou, E.E. Cavitation-Modulated Inflammatory Response Following Focused Ultrasound Blood-Brain Barrier Opening. J. Control. Release 2021, 337, 458–471. [Google Scholar] [CrossRef] [PubMed]
  157. Assistance Publique—Hôpitaux de Paris. A Study to Evaluate the Safety of Transient Opening of the Blood-Brain Barrier by Low Intensity Pulsed Ultrasound with the SonoCloud Implantable Device in Patients with Recurrent Glioblastoma before Chemotherapy Administration. 2018. Available online: https://clinicaltrials.gov/ct2/show/NCT02253212 (accessed on 28 February 2022).
  158. CarThera. A Study to Evaluate the Safety and the Efficacy of Transient Opening of the Blood-Brain Barrier (BBB) by Low Intensity Pulsed Ultrasound with the SonoCloud-9 Implantable Device in Recurrent Glioblastoma Patients Eligible for Surgery and for Carboplatin Chemotherapy. 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT03744026 (accessed on 28 February 2022).
  159. Assistance Publique–Hôpitaux de Paris. Safety and Efficacy of Blood Brain Barrier Opening with Implantable Device Sonocloud® Combined with Nivolumab Used Alone or an Association with Ipilimumab in Brain Metastases from Patients with Malignant Melanoma. 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT04021420 (accessed on 28 February 2022).
  160. Asquier, N.; Bouchoux, G.; Canney, M.; Martin, C.; Law-Ye, B.; Leclercq, D.; Chapelon, J.-Y.; Lafon, C.; Idbaih, A.; Carpentier, A. Blood-Brain Barrier Disruption in Humans Using an Implantable Ultrasound Device: Quantification with MR Images and Correlation with Local Acoustic Pressure. J. Neurosurg. 2019, 132, 875–883. [Google Scholar] [CrossRef]
  161. Liu, H.-L.; Hsu, P.-H.; Lin, C.-Y.; Huang, C.-W.; Chai, W.-Y.; Chu, P.-C.; Huang, C.-Y.; Chen, P.-Y.; Yang, L.-Y.; Kuo, J.S.; et al. Focused Ultrasound Enhances Central Nervous System Delivery of Bevacizumab for Malignant Glioma Treatment. Radiology 2016, 281, 99–108. [Google Scholar] [CrossRef]
  162. Coluccia, D.; Figueiredo, C.A.; Wu, M.Y.; Riemenschneider, A.N.; Diaz, R.; Luck, A.; Smith, C.; Das, S.; Ackerley, C.; O’Reilly, M.; et al. Enhancing Glioblastoma Treatment Using Cisplatin-Gold-Nanoparticle Conjugates and Targeted Delivery with Magnetic Resonance-Guided Focused Ultrasound. Nanomedicine 2018, 14, 1137–1148. [Google Scholar] [CrossRef]
  163. Thévenot, E.; Jordão, J.F.; O’Reilly, M.A.; Markham, K.; Weng, Y.-Q.; Foust, K.D.; Kaspar, B.K.; Hynynen, K.; Aubert, I. Targeted Delivery of Self-Complementary Adeno-Associated Virus Serotype 9 to the Brain, Using Magnetic Resonance Imaging-Guided Focused Ultrasound. Hum. Gene Ther. 2012, 23, 1144–1155. [Google Scholar] [CrossRef]
  164. Noroozian, Z.; Xhima, K.; Huang, Y.; Kaspar, B.K.; Kügler, S.; Hynynen, K.; Aubert, I. MRI-Guided Focused Ultrasound for Targeted Delivery of RAAV to the Brain. Methods Mol. Biol. 2019, 1950, 177–197. [Google Scholar] [CrossRef]
  165. Jordão, J.F.; Ayala-Grosso, C.A.; Markham, K.; Huang, Y.; Chopra, R.; McLaurin, J.; Hynynen, K.; Aubert, I. Antibodies Targeted to the Brain with Image-Guided Focused Ultrasound Reduces Amyloid-Beta Plaque Load in the TgCRND8 Mouse Model of Alzheimer’s Disease. PLoS ONE 2010, 5, e10549. [Google Scholar] [CrossRef] [Green Version]
  166. Kobus, T.; Zervantonakis, I.K.; Zhang, Y.; McDannold, N.J. Growth Inhibition in a Brain Metastasis Model by Antibody Delivery Using Focused Ultrasound-Mediated Blood-Brain Barrier Disruption. J. Control. Release 2016, 238, 281–288. [Google Scholar] [CrossRef] [Green Version]
  167. Morse, S.V.; Mishra, A.; Chan, T.G.; de Rosales, R.M.; Choi, J.J.; Choi, J.J. Liposome Delivery to the Brain with Rapid Short-Pulses of Focused Ultrasound and Microbubbles. J. Control. Release 2022, 341, 605–615. [Google Scholar] [CrossRef]
  168. Chan, T.G.; Morse, S.V.; Copping, M.J.; Choi, J.J.; Vilar, R. Targeted Delivery of DNA-Au Nanoparticles across the Blood-Brain Barrier Using Focused Ultrasound. ChemMedChem 2018, 13, 1311–1314. [Google Scholar] [CrossRef] [PubMed]
  169. Zhao, G.; Huang, Q.; Wang, F.; Zhang, X.; Hu, J.; Tan, Y.; Huang, N.; Wang, Z.; Wang, Z.; Cheng, Y. Targeted ShRNA-Loaded Liposome Complex Combined with Focused Ultrasound for Blood Brain Barrier Disruption and Suppressing Glioma Growth. Cancer Lett. 2018, 418, 147–158. [Google Scholar] [CrossRef] [PubMed]
  170. Burgess, A.; Ayala-Grosso, C.A.; Ganguly, M.; Jordão, J.F.; Aubert, I.; Hynynen, K. Targeted Delivery of Neural Stem Cells to the Brain Using MRI-Guided Focused Ultrasound to Disrupt the Blood-Brain Barrier. PLoS ONE 2011, 6, e27877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Alkins, R.; Burgess, A.; Kerbel, R.; Wels, W.S.; Hynynen, K. Early Treatment of HER2-Amplified Brain Tumors with Targeted NK-92 Cells and Focused Ultrasound Improves Survival. Neuro-Oncology 2016, 18, 974–981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. McDannold, N.; Zhang, Y.; Supko, J.G.; Power, C.; Sun, T.; Peng, C.; Vykhodtseva, N.; Golby, A.J.; Reardon, D.A. Acoustic Feedback Enables Safe and Reliable Carboplatin Delivery across the Blood-Brain Barrier with a Clinical Focused Ultrasound System and Improves Survival in a Rat Glioma Model. Theranostics 2019, 9, 6284–6299. [Google Scholar] [CrossRef]
  173. Zhang, P.; Miska, J.; Lee-Chang, C.; Rashidi, A.; Panek, W.K.; An, S.; Zannikou, M.; Lopez-Rosas, A.; Han, Y.; Xiao, T.; et al. Therapeutic Targeting of Tumor-Associated Myeloid Cells Synergizes with Radiation Therapy for Glioblastoma. Proc. Natl. Acad. Sci. USA 2019, 116, 23714–23723. [Google Scholar] [CrossRef]
  174. Taiarol, L.; Formicola, B.; Magro, R.D.; Sesana, S.; Re, F. An Update of Nanoparticle-Based Approaches for Glioblastoma Multiforme Immunotherapy. Nanomedicine 2020, 15, 1861–1871. [Google Scholar] [CrossRef]
  175. Mathew, E.N.; Berry, B.C.; Yang, H.W.; Carroll, R.S.; Johnson, M.D. Delivering Therapeutics to Glioblastoma: Overcoming Biological Constraints. Int. J. Mol. Sci. 2022, 23, 1711. [Google Scholar] [CrossRef]
  176. Zhang, F.; Stephan, S.B.; Ene, C.I.; Smith, T.T.; Holland, E.C.; Stephan, M.T. Nanoparticles That Reshape the Tumor Milieu Create a Therapeutic Window for Effective T-Cell Therapy in Solid Malignancies. Cancer Res. 2018, 78, 3718–3730. [Google Scholar] [CrossRef] [Green Version]
  177. Chen, P.-Y.; Hsieh, H.-Y.; Huang, C.-Y.; Lin, C.-Y.; Wei, K.-C.; Liu, H.-L. Focused Ultrasound-Induced Blood–Brain Barrier Opening to Enhance Interleukin-12 Delivery for Brain Tumor Immunotherapy: A Preclinical Feasibility Study. J. Transl. Med. 2015, 13, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Curley, C.T.; Sheybani, N.D.; Bullock, T.N.; Price, R.J. Focused Ultrasound Immunotherapy for Central Nervous System Pathologies: Challenges and Opportunities. Theranostics 2017, 7, 3608–3623. [Google Scholar] [CrossRef] [PubMed]
  179. Sevenich, L. Turning “Cold” Into “Hot” Tumors-Opportunities and Challenges for Radio-Immunotherapy Against Primary and Metastatic Brain Cancers. Front. Oncol. 2019, 9, 163. [Google Scholar] [CrossRef] [PubMed]
  180. Steeg, P.S. The Blood-Tumour Barrier in Cancer Biology and Therapy. Nat. Rev. Clin. Oncol. 2021, 18, 696–714. [Google Scholar] [CrossRef]
  181. Sabbagh, A.; Beccaria, K.; Ling, X.; Marisetty, A.; Ott, M.; Caruso, H.; Barton, E.; Kong, L.-Y.; Fang, D.; Latha, K.; et al. Opening of the Blood-Brain Barrier Using Low-Intensity Pulsed Ultrasound Enhances Responses to Immunotherapy in Preclinical Glioma Models. Clin. Cancer Res. 2021, 27, 4325–4337. [Google Scholar] [CrossRef]
  182. Alkins, R.; Burgess, A.; Ganguly, M.; Francia, G.; Kerbel, R.; Wels, W.S.; Hynynen, K. Focused Ultrasound Delivers Targeted Immune Cells to Metastatic Brain Tumors. Cancer Res. 2013, 73, 1892–1899. [Google Scholar] [CrossRef] [Green Version]
  183. Pacia, C.P.; Yuan, J.; Yue, Y.; Xu, L.; Nazeri, A.; Desai, R.; Gach, H.M.; Wang, X.; Talcott, M.R.; Chaudhuri, A.A.; et al. Sonobiopsy for Minimally Invasive, Spatiotemporally-Controlled, and Sensitive Detection of Glioblastoma-Derived Circulating Tumor DNA. Theranostics 2022, 12, 362–378. [Google Scholar] [CrossRef]
  184. Liu, H.-L.; Hsu, P.-H.; Chu, P.-C.; Wai, Y.-Y.; Chen, J.-C.; Shen, C.-R.; Yen, T.-C.; Wang, J.-J. Magnetic Resonance Imaging Enhanced by Superparamagnetic Iron Oxide Particles: Usefulness for Distinguishing between Focused Ultrasound-Induced Blood-Brain Barrier Disruption and Brain Hemorrhage. J. Magn. Reson. Imaging 2009, 29, 31–38. [Google Scholar] [CrossRef]
  185. Wu, S.-K.; Tsai, C.-L.; Huang, Y.; Hynynen, K. Focused Ultrasound and Microbubbles-Mediated Drug Delivery to Brain Tumor. Pharmaceutics 2020, 13, 15. [Google Scholar] [CrossRef]
  186. McMahon, D.; Hynynen, K. Acute Inflammatory Response Following Increased Blood-Brain Barrier Permeability Induced by Focused Ultrasound Is Dependent on Microbubble Dose. Theranostics 2017, 7, 3989–4000. [Google Scholar] [CrossRef]
  187. McMahon, D.; Mah, E.; Hynynen, K. Angiogenic Response of Rat Hippocampal Vasculature to Focused Ultrasound-Mediated Increases in Blood-Brain Barrier Permeability. Sci. Rep. 2018, 8, 12178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Yuan, J.; Ye, D.; Chen, S.; Chen, H. Therapeutic Ultrasound-Enhanced Immune Checkpoint Inhibitor Therapy. Front. Phys. 2021, 9, 102. [Google Scholar] [CrossRef]
  189. Elias, W.J.; Huss, D.; Voss, T.; Loomba, J.; Khaled, M.; Zadicario, E.; Frysinger, R.C.; Sperling, S.A.; Wylie, S.; Monteith, S.J.; et al. A Pilot Study of Focused Ultrasound Thalamotomy for Essential Tremor. N. Engl. J. Med. 2013, 369, 640–648. [Google Scholar] [CrossRef] [PubMed]
  190. Elias, W.J.; Lipsman, N.; Ondo, W.G.; Ghanouni, P.; Kim, Y.G.; Lee, W.; Schwartz, M.; Hynynen, K.; Lozano, A.M.; Shah, B.B.; et al. A Randomized Trial of Focused Ultrasound Thalamotomy for Essential Tremor. N. Engl. J. Med. 2016, 375, 730–739. [Google Scholar] [CrossRef]
  191. Iorio-Morin, C.; Yamamoto, K.; Sarica, C.; Zemmar, A.; Levesque, M.; Brisebois, S.; Germann, J.; Loh, A.; Boutet, A.; Elias, G.J.B.; et al. Bilateral Focused Ultrasound Thalamotomy for Essential Tremor (BEST-FUS Phase 2 Trial). Mov. Disord. 2021, 36, 2653–2662. [Google Scholar] [CrossRef]
  192. Martin, E.; Jeanmonod, D.; Morel, A.; Zadicario, E.; Werner, B. High-Intensity Focused Ultrasound for Noninvasive Functional Neurosurgery. Ann. Neurol. 2009, 66, 858–861. [Google Scholar] [CrossRef] [Green Version]
  193. Silvestrini, M.T.; Ingham, E.S.; Mahakian, L.M.; Kheirolomoom, A.; Liu, Y.; Fite, B.Z.; Tam, S.M.; Tucci, S.T.; Watson, K.D.; Wong, A.W.; et al. Priming Is Key to Effective Incorporation of Image-Guided Thermal Ablation into Immunotherapy Protocols. JCI Insight 2017, 2, e90521. [Google Scholar] [CrossRef]
  194. Chavez, M.; Silvestrini, M.T.; Ingham, E.S.; Fite, B.Z.; Mahakian, L.M.; Tam, S.M.; Ilovitsh, A.; Monjazeb, A.M.; Murphy, W.J.; Hubbard, N.E.; et al. Distinct Immune Signatures in Directly Treated and Distant Tumors Result from TLR Adjuvants and Focal Ablation. Theranostics 2018, 8, 3611–3628. [Google Scholar] [CrossRef]
  195. Eranki, A.; Srinivasan, P.; Ries, M.; Kim, A.; Lazarski, C.A.; Rossi, C.T.; Khokhlova, T.D.; Wilson, E.; Knoblach, S.M.; Sharma, K.V.; et al. High-Intensity Focused Ultrasound (HIFU) Triggers Immune Sensitization of Refractory Murine Neuroblastoma to Checkpoint Inhibitor Therapy. Clin. Cancer Res. 2020, 26, 1152–1161. [Google Scholar] [CrossRef] [Green Version]
  196. Qu, S.; Worlikar, T.; Felsted, A.E.; Ganguly, A.; Beems, M.V.; Hubbard, R.; Pepple, A.L.; Kevelin, A.A.; Garavaglia, H.; Dib, J.; et al. Non-Thermal Histotripsy Tumor Ablation Promotes Abscopal Immune Responses That Enhance Cancer Immunotherapy. J. Immunother. Cancer 2020, 8, e000200. [Google Scholar] [CrossRef] [Green Version]
  197. Elhelf, I.A.S.; Albahar, H.; Shah, U.; Oto, A.; Cressman, E.; Almekkawy, M. High Intensity Focused Ultrasound: The Fundamentals, Clinical Applications and Research Trends. Diagn. Interv. Imaging 2018, 99, 349–359. [Google Scholar] [CrossRef] [PubMed]
  198. Huang, B.; Li, X.; Li, Y.; Zhang, J.; Zong, Z.; Zhang, H. Current Immunotherapies for Glioblastoma Multiforme. Front. Immunol. 2021, 11, 3890. [Google Scholar] [CrossRef] [PubMed]
  199. Dirkse, A.; Golebiewska, A.; Buder, T.; Nazarov, P.V.; Muller, A.; Poovathingal, S.; Brons, N.H.C.; Leite, S.; Sauvageot, N.; Sarkisjan, D.; et al. Stem Cell-Associated Heterogeneity in Glioblastoma Results from Intrinsic Tumor Plasticity Shaped by the Microenvironment. Nat. Commun. 2019, 10, 1787. [Google Scholar] [CrossRef] [PubMed]
  200. Cheng, Q.; Wei, T.; Farbiak, L.; Johnson, L.T.; Dilliard, S.A.; Siegwart, D.J. Selective Organ Targeting (SORT) Nanoparticles for Tissue-Specific MRNA Delivery and CRISPR–Cas Gene Editing. Nat. Nanotechnol. 2020, 15, 313–320. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) Immune checkpoint inhibitors bind to and inhibit immunosuppressive molecules on either T-cells or tumor cells. This dampens tumor cells′ ability to evade the immune system. (B) (1) In tumor-infiltrating lymphocyte (TIL) therapy, T-cells from the tumor microenvironment are isolated following surgical resection. (2) Isolated T-cells are clonally expanded by using IL-2 stimulation. (3) Expanded T-cells are reintroduced to the patient. (C) (1) In the vaccine approach, a resected tumor biopsy is taken from the patient and sequenced to identify neoantigens. (2) Neoantigens are then delivered via a vaccine. (3) At the site of injection, neoantigens stimulate antigen-presenting cells (APCs). (4) In the lymph node, APCs present T-cells with neoantigens. (5) Activated T-cells attack cancer cells. Created with BioRender.com.
Figure 1. (A) Immune checkpoint inhibitors bind to and inhibit immunosuppressive molecules on either T-cells or tumor cells. This dampens tumor cells′ ability to evade the immune system. (B) (1) In tumor-infiltrating lymphocyte (TIL) therapy, T-cells from the tumor microenvironment are isolated following surgical resection. (2) Isolated T-cells are clonally expanded by using IL-2 stimulation. (3) Expanded T-cells are reintroduced to the patient. (C) (1) In the vaccine approach, a resected tumor biopsy is taken from the patient and sequenced to identify neoantigens. (2) Neoantigens are then delivered via a vaccine. (3) At the site of injection, neoantigens stimulate antigen-presenting cells (APCs). (4) In the lymph node, APCs present T-cells with neoantigens. (5) Activated T-cells attack cancer cells. Created with BioRender.com.
Cancers 14 01627 g001
Figure 2. The left panel shows the normal anatomy of the blood–brain barrier in which tight junctions exist between endothelial cells to prevent the passage of most therapeutics into the brain parenchyma. This basic structure is supported by astrocytes and pericytes, which help maintain and regulate these tight junctions. The right panel shows the pathology of the BBB induced by tumor growth. For one, there is increased permeability of the endothelial cells’ tight junctions, permitting tumor cell extravasation. There is an atrophied basal lamina, which contributes to anergic endothelial cells. Finally, pericytes are both fewer and display an abnormal morphology. The combination of these factors can promote tumor migration and growth. Created with BioRender.com.
Figure 2. The left panel shows the normal anatomy of the blood–brain barrier in which tight junctions exist between endothelial cells to prevent the passage of most therapeutics into the brain parenchyma. This basic structure is supported by astrocytes and pericytes, which help maintain and regulate these tight junctions. The right panel shows the pathology of the BBB induced by tumor growth. For one, there is increased permeability of the endothelial cells’ tight junctions, permitting tumor cell extravasation. There is an atrophied basal lamina, which contributes to anergic endothelial cells. Finally, pericytes are both fewer and display an abnormal morphology. The combination of these factors can promote tumor migration and growth. Created with BioRender.com.
Cancers 14 01627 g002
Figure 3. Cartoon illustrating how microbubbles can induce a focal disruption or opening of the blood–brain barrier (BBB), thus enabling the delivery of a biologic such as a monoclonal antibody. Microbubbles flow through the normal vasculature or vasculature supplying the glioblastoma tumor microenvironment (TME). Only the microbubbles in the vasculature exposed to ultrasound insonation enable BBB/BTB disruption following ultrasound insonation. Created with BioRender.com.
Figure 3. Cartoon illustrating how microbubbles can induce a focal disruption or opening of the blood–brain barrier (BBB), thus enabling the delivery of a biologic such as a monoclonal antibody. Microbubbles flow through the normal vasculature or vasculature supplying the glioblastoma tumor microenvironment (TME). Only the microbubbles in the vasculature exposed to ultrasound insonation enable BBB/BTB disruption following ultrasound insonation. Created with BioRender.com.
Cancers 14 01627 g003
Table 1. Overview of current and in-progress immunotherapeutic clinical trials.
Table 1. Overview of current and in-progress immunotherapeutic clinical trials.
Trial NumberStudy DesignTrial DetailsPatient NumberReference
NCT02017717Phase III
R, PA
Comparison of the PD-1 inhibitor nivolumab, with and without the CTLA-4 inhibitor ipilimumab, versus the VEGF inhibitor bevacizumab. The primary outcome is overall survival. Study is in progress. 530[18]
NCT03291314Phase II
PA
A study of the combination of the anti-PD-L1 molecule avelumab and the VEGF inhibitor axitinib on the progression of GBM. The 6-month progression-free survival (PFS) was 18% (95% CI 4–33, n = 27), which did not meet the threshold for justifying further investigation.52[21]
NCT02336165Phase II
PA
A study of the anti-PD-L1 molecule durvalumab in subjects with glioblastoma. Patients were enrolled into 5 non-comparative cohorts receiving either durvalumab monotherapy or durvalumab and bevacizumab combotherapy. MOS was 15.1 months (95% CI 12.0–18.4). 159[22]
NCT01454596Phase I/II
SA
A study to determine the safety and effects of CART-EGFRvIII therapy in patients with recurrent GBM. CAR T-cell therapy was given in combination with a synthetic IL-2 molecule, aldesleukin, and a lymphodepleting preparative regimen of cyclophosphamide and fludarabine. MOS was 6.9 months (IQR 2.8–10.0), with 3 instances of adverse effects.18[23]
NCT02454634Phase I
SGA
A study to identify the safety and tolerability of the first in-human mutant IDH1 peptide vaccine in patients with WHO Grade III–IV gliomas. Vaccine-induced immune responses were observed in 93.3% of patients. No regime-limiting toxicity was observed.32[24]
NCT01250470Phase I
SGA
A study of the side effects of a vaccine therapy directed against the tumorigenic molecule survivin, in combination with the synthetic granulocyte-macrophage colony-stimulating factor sargramostim. The therapy was well tolerated with no serious adverse events attributable to the therapy; 6 out of 8 immunologically evaluable patients developed immune responses to the vaccine.9[25]
NCT05163080Phase II
R, PA, DB
A Phase II clinical trial analyzing whether an antisurvivin vaccine treatment combined with SOC TMZ treatment is better than TMZ treatment alone for GBM patients. The primary outcome is OS. The trial is in progress.265[26]
NCT00157703Phase I
SGA
A study to determine the safety of oncolytic HSV-1, G207, given in combination with radiation for recurrent GBM. Three serious AEs were reported (seizures after administration), possibly related to G207 administration. The estimated median survival time from G207 inoculation was 7.5 months (95% CI 3.0–12.7).9[27]
NCT02457845Phase I
SGA
A study to determine the safety of G207 treatment in combination with radiotherapy for pediatric patients with recurrent supratentorial brain tumors. Twenty Grade 1 AEs were reported, possibly related to G207. MOS was 12.2 months (95% CI 8.0–16.4) as of 6/2020. The trial is in progress.12[28]
NCT00589875Phase II
SGA
A study to determine the safety and potential efficacy of adenoviral vector expressing HSV1-tk (aglatimagene besadonevac, AdV-tk) followed by valacyclovir in combination with the SOC treatment. MOS was 17.1 month for treatment + SOC vs. 13.5 months for SOC alone (p = 0.0417)52[29]
EudraCT 2004-000464-28Phase III
R, PA
A study comparing the adenovirus vector-mediated delivery of HSV1-tk (AdV-tk) followed by IV ganciclovir with the SOC treatment versus SOC treatment alone in newly diagnosed GBM. No difference in MOS was found in the experimental (497 days, 95% CI 369–574) versus the control group (452 days, 95% CI 437–558) (HR 1.18, 95% CI 0.86–1.61, p = 0.31).250[30]
NCT01491893Phase I
SA
A study to determine the maximum tolerated dose of a live attenuated polio–rhinovirus chimera (PVSRIPO) on GBM; 19% of patients treated with PVSRIPO had a Grade 3 or higher adverse event.61[31]
NCT02414165Phase II/III
R, PA
A study of a gamma retroviral replicating vector encoding a yeast cytosine deaminase, vocimagene amiretrorepvec, combined with 5-fluorocytosine treatment versus SOC in recurrent GBM. MOS was 11.10 months for the experimental group compared to 12.22 months for the control group (HR = 1.06; 95% CI 0.83, 1.35; p  =  0.62).58[32,33]
DB, double-blind; PA, parallel assignment; R, randomized; SA, sequential assignment; SGA, single group assignment.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lechpammer, M.; Rao, R.; Shah, S.; Mirheydari, M.; Bhattacharya, D.; Koehler, A.; Toukam, D.K.; Haworth, K.J.; Pomeranz Krummel, D.; Sengupta, S. Advances in Immunotherapy for the Treatment of Adult Glioblastoma: Overcoming Chemical and Physical Barriers. Cancers 2022, 14, 1627. https://doi.org/10.3390/cancers14071627

AMA Style

Lechpammer M, Rao R, Shah S, Mirheydari M, Bhattacharya D, Koehler A, Toukam DK, Haworth KJ, Pomeranz Krummel D, Sengupta S. Advances in Immunotherapy for the Treatment of Adult Glioblastoma: Overcoming Chemical and Physical Barriers. Cancers. 2022; 14(7):1627. https://doi.org/10.3390/cancers14071627

Chicago/Turabian Style

Lechpammer, Mirna, Rohan Rao, Sanjit Shah, Mona Mirheydari, Debanjan Bhattacharya, Abigail Koehler, Donatien Kamdem Toukam, Kevin J. Haworth, Daniel Pomeranz Krummel, and Soma Sengupta. 2022. "Advances in Immunotherapy for the Treatment of Adult Glioblastoma: Overcoming Chemical and Physical Barriers" Cancers 14, no. 7: 1627. https://doi.org/10.3390/cancers14071627

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