Abstract
Purpose of review:
Chimeric Antigen Receptor (CAR) T cell therapy has been successful in some hematologic malignancies, but the central nervous system (CNS) presents unique obstacles to its use against tumors arising therein. This review discusses recent improvements in the delivery and design of these cells to improve the efficacy and safety of this treatment against malignant gliomas.
Recent Findings:
The immunosuppressive environment of the CNS affects the functionality of CAR T cells but recent developments using metabolic manipulation and cytokine delivery have shown that the performance of CAR T cells can be improved in this environment. Emerging techniques can improve the delivery of CAR T cells to the CNS parenchyma, which is normally well-protected from peripheral immune cells. The implementation of novel antigens and CAR-expression regulation strategies will improve the specificity and efficacy of these cells. Finally, while autologous T cells have historically been the standard, recent developments have made the use of allogeneic T cells or Natural Killer (NK) cells more clinically feasible.
Summary:
The discoveries highlighted in this review will aid the development of CAR cells that are safer, more resilient against immunosuppressive signals in the CNS, and able to specifically target intracranial tumor cells.
Keywords: CAR T, glioma, central nervous system
INTRODUCTION
Chimeric antigen receptor (CAR)-T cells represent a groundbreaking therapy for many blood cancers, such as B-cell lymphoma, but thus far have failed to demonstrate the same efficacy against non-hematological malignancies, including gliomas. One of the key challenges is an immunosuppressive tumor microenvironment (TME) (1). The mechanisms by which gliomas achieve immune evasion remain to be fully elucidated. However, it has been shown that malignant gliomas exhibit stable epitranscriptomic changes which facilitate the recruitment of tumor-associated macrophages (TAMs) and secretion of anti-inflammatory cytokines (2–4). The release of anti-inflammatory cytokines via TAMs and other pro-tumorigenic myeloid cells causes T cell dysfunction, which reduces the functionality of CAR-T cells (5). Tumor cells and TAMs also deplete metabolic fuels and release catabolic byproducts into the TME, which similarly impairs CAR-T cell persistence and survival (6). Furthermore, CAR-T cell trafficking to central nervous system (CNS) tumors is often poor owing to some extent to the presence of the blood-brain barrier (BBB) (7). In this review, we discuss strategies to overcome an immunosuppressive TME, enhance CAR-T persistence, and improve CAR-T cell delivery to gliomas.
Other obstacles pertain to the manufacturing of CAR-T cells and the design of CAR circuits for gliomas. Firstly, there is a lack of antigen targets that are both homogenously expressed by tumors and have absolute tumor-specificity. Heterogenous antigen expression can lead to CAR-T resistance via antigen escape (8, 9). However, targeting abundantly expressed antigens in gliomas can cause “on-target, off-tumor” toxicities (10). Secondly, because many glioma patients have insufficient T cells to produce therapeutic doses of autologous CAR-T cells (11), the development of allogeneic, off-the-shelf CAR-T cells would be beneficial. Proper engineering of allogeneic CAR-T cells should mitigate the issues, such as graft-versus-host disease (GvHD) (12). We describe gaps in the literature, next-generation CAR design, and how genetic editing could facilitate safer, more effective “off-the-shelf” CAR-T cells.
Programming metabolic fitness of CAR T cells
Clinical response is still very rare in patients with malignant glioma following CAR T cell therapy. The immunosuppressive microenvironment of glioma is one of the major factors that limits the functionality of CAR T cells. In addition to the presence of immunosuppressive cytokines and factors (PGE2, TGFb, IL10, VEGF), the glioma microenvironment is also hypoxic, hypoglycemic, lactic acid-rich, and depleted of amino acids such as glutamine, arginine, and tryptophan (6).
Tumor cells and CAR T cells compete for the limited amount of metabolic fuels in the tumor microenvironment (TME). To maintain the fitness of CAR T cells in metabolically altered and nutrient-deprived TME, CAR T cells can be manipulated either pharmacologically or by selective knocking out of genes using CRISPR. In recent years, several studies have focused on identifying the key regulators of metabolic pathways in T cells that can serve as therapeutic targets for enhancing the antitumor function and persistence of T cells (13–18). Hamaidi et al (19) investigated the role of Sirtuin-2(Sirt2), an NAD+ dependent deacetylase, in T cell metabolism and anti-tumor immunity. Selective inhibition or genetic deletion of Sirt2 in T cells results in reprogramming towards a hyper-metabolic state characterized by an enhanced capacity for aerobic glycolysis and oxidative phosphorylation. At the operational level, Sirt2 suppresses T cell metabolism by targeting multiple enzymes involved in glycolysis, TCA cycle, fatty acid oxidation, and glutaminolysis. Importantly, Sirt2 deficient T cells display enhanced proliferation, survival, and effector function, thus Sirt2 can serve as an actionable target for metabolic manipulation of CAR T cells. Inhibition of the MAPK pathway by MEK1/MEK2 inhibitors generates CD8+ stem cell-like memory cells that have enhanced mitochondrial respiration fueled by fatty acid oxidation and superior antitumor activity (20).
In addition to the engineering approaches applied at the cellular level, engineering the CAR construct itself can also significantly impact the fitness of the T cells. A dual antigen CAR T cells with split costimulatory signaling domains and shared CD3ζ chain exhibit a rapid effector function via glycolysis, which is supported by CD28 signaling (21). Additionally, 4-1BB costimulatory signaling preserves the oxidative function which is critical for long-term persistence and memory formation (22). Next-generation CAR T cells called TRUCKs (T-cell redirected for unrestricted cytokine-mediated killing) incorporate a transgenic cytokine release machinery to augment the proliferation and persistence in vivo (23, 24). The incorporated cytokines also have a metabolic effect on the T cells. For example, IL-15 treated CAR T cells have elevated levels of oxidative phosphorylation (OXPHOS) and fatty acid oxidation-related genes (25). Methods that can further boost the metabolic and phenotypic fitness of CAR T cells in the TME need to be evaluated in the glioma setting.
Advances in regulation of CAR expression
We and others have shown that spatial and temporal regulation of CAR expression protects T cells from exhaustion and favors a memory stem-like phenotype that improved their in vivo long-term persistence (26–29). Numerous approaches are being evaluated to achieve conditional expression of CAR (Figure 1), such as recognition of tumor-specific antigen, sensing of hypoxia (30, 31), small molecule drug-mediated transient inhibition of CAR expression, and by use of “degrons” (32).
Figure 1. Schematic representation of the approaches for controlling and regulating the expression of CAR.

First panel: SynNotch receptors are engineered to sense tumor- or tissue-specific antigens and induce the expression of a CAR to a second tumor-associated antigen. Second panel: Dynamic on/off oxygen sensing safety switch is engineered in the CAR construct to minimize off-tumor toxicities. Third panel: Engineering of CAR construct to include a drug-responsive domain (degron) such that the activity of CAR T cells can be controlled by small molecule drugs such as lenalidomide. Fourth Panel: Drug-dependent assembly of split CAR where membrane signaling domain is separated from scFV. The split CAR is switched “on” only in the presence of the drug.
A Synthetic Notch (SynNotch) receptor recognizes a tumor- or organ-specific antigen to induce and confine the expression of CAR to the tumor site. Additionally, transient expression of CAR prevents tonic signaling and exhaustion of T cells(26). Mechanistically, Synthetic Intramembrane Proteolysis Receptors (SNIPRs) are similar to SynNotch receptors, both of which can be tuned to match the antigen density on the tumor cells(27). Another approach is where CAR is fused to a ligand-induced degradation domain (LID) for reversible and tunable CAR expression (33). The addition of a small molecule ligand causes exposure of cryptic degron within the LID domain and subsequent proteasomal degradation of CAR-LID fusion protein and the loss of CAR on the T cells. Alternatively, drug-induced dimerization can serve as an ON-switch for split CAR circuit design (34). The strategies that allow CAR expression to be controlled via drug, may not work for glioma if the drug has poor BBB penetration.
Improvements in delivery of CAR T Cells
The BBB presents a unique challenge for treatment delivery to tumors that arise in the parenchyma of the CNS. While intravenous (IV) delivery has been the most frequently used method, locoregional delivery modes, such as the delivery of CAR T cells into the cerebral spinal fluid (CSF), are developing as an alternative, especially when the targeted antigens are also expressed in non-CNS tissues. This technique was previously efficacious in a case with glioblastoma (35), and has been implemented in recent clinical trials delivering CAR T cells to pediatric CNS tumors (36, 37). Patients exhibited local inflammation and no dose-limiting toxicities, although most patients still progressed. A caveat of the intra-CSF delivery of CAR T cells is that the CSF space is still separated from the CNS parenchyma by the glia-limitans and the tight network of the astrocyte end-feet (Figure 2).
Figure 2: Delivery Methods of CAR T cells.

Intratumoral delivery completely bypasses the Blood-Brain barrier (BBB) through the delivery of the agent directly to the tumor site. Intracerebroventricular (ICV) delivery partially bypasses the BBB as cells enter the CSF, but the glia-limitans still separates the CNS parenchyma. Locoregional delivery encompasses intratumoral and intracerebroventricular delivery. Intravenous delivery is the least invasive option of the three, but CAR T cells must penetrate the endothelial cells surrounding capillaries as well as the glia-limitans to enter the CNS. Vector images from vecteezy.com
Focused ultrasound-mediated BBB disruption (FUS-BBBD) has been rapidly emerging as a way to disrupt the BBB (38). In this system, FUS induces vibrations of IV-injected microbubbles, thereby safely and transiently disrupting the BBB. In glioma-bearing mice, FUS-BBBD resulted in significantly increased CAR T cell delivery to the CNS and extended survival (39). FUS-BBBD can also have effects beyond delivery, such as activating microglia or releasing tumor antigens into the peripheral circulation (40). These effects may have therapeutic potential but require stringent tuning of the ultrasound parameters to ensure that they do not contribute to neuroinflammation.
New Antigen Targets
Identifying an antigen that is both abundantly expressed by tumor cells and not expressed in the normal brain is a challenge when treating malignant gliomas. Several antigens are currently being investigated in clinical trials (Table 1). EphA2 is one promising target as a clinical trial using EphA2-directed CAR T cells in glioblastoma demonstrated transient efficacy, although all patients experienced pulmonary edema during treatment (10). This is likely an “on-target, off-tumor” effect as EphA2 is expressed in the lungs and the intravenous delivery of these cells allowed them to traffic to the lung. This toxicity indicates a need for local delivery of EphA2-directed CAR T cells or restricted expression of the CAR.
Table 1:
Antigens currently investigated for CAR T cells therapy in intracranial tumors.
Other new targets that have emerged in pre-clinical studies are CD70 and B7-H3. A flow cytometry panel of patient-derived intracranial tumor cells showed that B7-H3 was abundantly expressed, showing greater expression than EphA2, IL-13Rα2 and HER2. B7-H3-directed CAR T cells showed preclinical efficacy in murine glioma models. Although B7-H3 is expressed at low levels in the brain, thus far, no neurotoxicites have been observed in preclinical models or phase I clinical trials(41, 42). CD70-directed CAR T cells recently demonstrated efficacy in a murine glioblastoma model, although efficacy varied in vivo due to the heterogeneity of CD70 expression in patient samples (8). Such heterogeneity might be overcome by a tandem CAR. A tandem CAR against CD70 and B7-H3 has been developed and tested in several solid tumor types although remains to be tested in intracranial tumors (43).
Advancements in Manufacturing Allogeneic CAR-T Therapies
There are inherent challenges associated with the use of patient-derived T cells which are pertinent to gliomas. Firstly, the timeline of autologous CAR-T cell production from leukapheresis to autologous CAR-T administration is approximately 3 to 6 weeks (44, 45). Tumor progression is likely to occur during the course of autologous CAR-T cell manufacturing, especially aggressive tumors, such as glioblastoma (46, 47). Progression may induce new neurological symptoms and deficits (48), which would require management with corticosteroids (49). Secondly, newly diagnosed, treatment-naïve glioma patients display a reduced number of circulating CD4+ T cells. Gliomas and other intracranial tumors cause sequestration of T cells in the bone marrow, thus limiting their availability for therapeutic purposes (11).
Although the use of allogeneic CAR-T cells may circumvent the issues of long manufacturing periods and inadequate T cells, other challenges related to their allogeneic nature may arise. Though the CNS and tumors therein used to be described as immune-privileged (50), active immune responses have been documented within the brain following immunotherapy and allogeneic transplantation (51, 52). Therefore, GvHD and allogeneic rejection due to human leukocyte antigen (HLA)-mismatch represent key obstacles to developing allogeneic CAR-T therapies for gliomas. Additionally, the large-scale expansion of off-the-shelf products for multiple patients may induce exhaustion in allogeneic CAR-T cell products (53).
One common strategy to reduce the risk of GvHD following allogeneic CAR-T cells is disrupting the endogenous T cell receptor (TCR) alpha constant (TRAC) with genomic editing techniques (e.g. CRISPR/Cas9, TAL effector nucleases) (54–56). Although not related to the mitigation of GvHD, Brown et al. generated steroid-resistance allogeneic CAR-T cells via glucocorticoid receptor knockout, which enabled corticosteroid administration without reducing CAR-T cell efficacy (53). Furthermore, it is feasible to combine these methods with additional genetic modifications that enhance allogeneic CAR-T cell persistence. For instance, Choi et al performed multiplexed gene disruption of TRAC, beta-2 microglobulin, and PD-1 to create universal CAR-T cells with enhanced antitumor activity in vivo (56). Lastly, induced pluripotent stem cells (iPSCs) may represent a future strategy for readily available, off-the-shelf CAR-T products. Research has shown the feasibility of producing large-scale, functional, expandable T cell-derived iPSCs (T-iPSCs) (57). Genetically editing T-iPSCs via the aforementioned strategies could facilitate safer, more effective off-the-shelf CAR-T therapies.
CAR NK Cells
Natural Killer (NK) cells are another attractive source for allogeneic cells as they lack a T cell receptor (TCR) and are less likely to cause GvHD. Several clinical trials are leveraging NK CAR cells against high-grade gliomas (58). One limitation of these cells is their limited persistence compared to T cells. Delivery of cytokines has been shown to improve the persistence of NK CAR cells, motivating the development of cytokine-delivery systems that will synergize with these cells (59).
Rudek et al developed a system for NK cells whereby activation of a GD2 CAR induces the expression of cytokines in a mechanism like TRUCKS (60). The authors found that, while NFAT is the promotor of choice for the cytokine package in TRUCKS, NFAT-driven promoters behave differently in NK cells than in T cells, leading the authors to select NFkB as their promotor instead. Thus, the principles of CAR T cells cannot always be directly applied to NK cells.
The application of CAR NK cells will benefit from more mechanistic studies on how NK cells are affected by the tumor microenvironment. Recently, Wang et al demonstrated that an autophagy inhibitor, chloroquine, increased the trafficking of NK cells to GBM, mediated by the NFkB signaling pathway and the release of CCL2 and CXCL12. A combination treatment of chloroquine and NK CAR cells synergistically diminished tumor volume in murine GBM (61). Further studies to examine the persistence of these NK cells will have translational importance.
Adverse Events of CAR- T Therapy
CAR-T cell therapies can incur acute and chronic adverse events. One notable example is cytokine release syndrome (CRS), defined as a systemic immune response to pro-inflammatory cytokines released by CAR-T cells (62). Most of the literature on CRS pertains to B-cell malignancies, as up to one-third of patients treated with CD19-directed CAR-T cells develop severe symptoms (e.g. hypotension, hypoxia requiring intubation) (62). Comparatively few clinical trials have tested CAR-T cells in gliomas, with most reporting no cases or mild symptoms that typically resolved with corticosteroids and/or anticytokines (9, 10, 35, 36). However, one severe case of CRS led to a mortality in a Phase 1 trial on EGFRvIII-CAR-T cells (63).
Immune-effector cell-associated neurotoxicity syndrome (ICANS) represents another major side effect of CAR-T therapies (51). ICANS case reports most commonly describe CD19-CAR-T cells, some of which are associated with the expression of CD19 in human brain mural cells (64). The neurological symptoms of ICANS may overlap with those of tumor inflammation-associated neurotoxicity (TIAN), a recently defined phenomenon involving peritumoral oedema and/or CSF obstruction as well as the transient worsening of existing deficits (36). A GD2- CAR-T therapy caused symptoms of TIAN (e.g. transient ataxia, sensory loss, impaired motor function) and potentially ICANS in H3K27M-mutated diffuse midline glioma patients. Further research should explore the clinical manifestations of ICANS versus TIAN in gliomas to elucidate their unique etiologies and facilitate better symptomatic management.
While the immediate risks associated with CAR-T therapies are well-characterized, the long-term effects remain poorly understood. It is possible that repeated CAR-T cell administrations could increase patients’ risks of developing neurodegenerative diseases, such as Alzheimer’s or Parkinson’s. A growing body of evidence suggests that neuroinflammation is a hallmark feature of the pathogenesis of Alzheimer’s. Inhibition of CD4+ T cells is known to ameliorate Alzheimer’s and Parkinson’s pathology in mouse models, while Treg depletion–a crucial step preceding leukapheresis for CAR-T generation–predicts the exacerbation of symptoms (65, 66). Furthermore, some adverse events associated with CAR-T therapies (e.g. CRS, increased production of reactive oxygen species, reduced immunosuppressive Treg cells) (67, 68) have been implicated in the pathogenesis of Alzheimer’s (69, 70). The potential for CAR-T therapies to increase long-term neurodegeneration warrants further attention, as brain tumors may independently predispose patients to CNS diseases (71). Indeed, GBM and Alzheimer’s both involve similar patterns of expression in a significant number of genes–these genes hypothetically dampen oxidative phosphorylation and establish a local chronic inflammatory state (71). We should gain a more in-depth understanding of the possible long-term impacts of CAR-T-associated inflammation on CNS function as these therapies continue to advance in safety and efficacy.
CONCLUSION
Many of the immunological features of the CNS and CNS tumors are not favorable for eliciting a successful anti-tumor immune responses. Most CNS tumors bear low mutations (72). While dural/meningeal lymphatic systems should allow the presentation of CSF-derived antigens (73), it is still unclear whether the antigens derived from tumors in the CNS parenchyma could be effectively presented by functional antigen-presenting cells. The unique anatomical challenges also include the presence of BBB which may limit both antigen-presentation and homing of the effector cells to the tumor. Furthermore, standard therapeutic modalities for these patients, such as the use of corticosteroids to alleviate CNS inflammation, significantly impact the immune system. Due to these challenges, immunotherapy strategies that rely on the induction of endogenous anti-tumor immune responses, such as checkpoint blockade and cancer vaccines, may have limited efficacy as monotherapy. By integrating our understanding of these unique challenges and novel technologies, the adoptive transfer of genetically engineered immune cells may hold a promise as discussed in this review. Selection of antigens remains fundamental to minimizing the tumor antigen-loss escape and off-tumor toxicity, and the use of novel engineered circuits, such as SynNotch receptors and SNIPRs, may allow us to improve both efficacy and safety. The therapeutic cells could be also engineered to mitigate metabolic challenges and immunosuppression in the tumor microenvironment. Improvement of the manufacturing process and the use of allogeneic CAR T cells can circumvent the current issues, such as the fitness and long manufacturing and release time of the cell products for patients with aggressive tumor.
KEY POINTS.
Advances of cell engineering techniques will be coupled with in-depth understanding of the immunological and metabolic characteristics of malignant gliomas, thereby allowing the development of novel CAR T strategies that will overcome the unique challenges.
The ideal selection of antigen targets and delivery routes should be based on our emerging understanding of glioma cell heterogeneity and CNS immunology.
The development of the off-the-shelf CAR T cells, including T-iPSC cells, may allow timely and feasible delivery of the therapeutic cells in patients with rapidly growing tumors, such as glioblastoma.
ACKNOWLEDGMENT
Funding Support:
This work was supported by NIH/NINDS R35 NS105068 and NIH/NCI R01CA222965
Footnotes
Conflicts of Interest: Hideho Okada is an inventor of IL-13Ra2 (345–353:1A9V) peptide and EGFRvIII-CAR for which an exclusive licensing agreement has been executed with Stemline, Inc. and Novartis Pharma, respectively.
References
- 1.Grabowski M, Sankey E, Ryan K, Chongsathidkiet P, Lorrey S, Wilkinson D, et al. Immune suppression in gliomas. Journal of neuro-oncology. 2021;151(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Gangoso E, Southgate B, Bradley L, Rus S, Galvez-Cancino F, McGivern N, et al. Glioblastomas acquire myeloid-affiliated transcriptional programs via epigenetic immunoediting to elicit immune evasion. Cell. 2021;184(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Dong F, Qin X, Wang B, Li Q, Hu J, Cheng X, et al. ALKBH5 Facilitates Hypoxia-Induced Paraspeckle Assembly and IL8 Secretion to Generate an Immunosuppressive Tumor Microenvironment. Cancer research. 2021;81(23). [DOI] [PubMed] [Google Scholar]
- 4.Kohanbash G, McKaveney K, Sakaki M, Ueda R, Mintz AH, Amankulor N, et al. GM-CSF Promotes the Immunosuppressive Activity of Glioma-Infiltrating Myeloid Cells through Interleukin-4 Receptor-alpha. Cancer Res. 2013;73(21):6413–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ravi V, Neidert N, Will P, Joseph K, Maier J, Kückelhaus J, et al. T-cell dysfunction in the glioblastoma microenvironment is mediated by myeloid cells releasing interleukin-10. Nature communications. 2022;13(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Di Ianni N, Musio S, Pellegatta S. Altered Metabolism in Glioblastoma: Myeloid-Derived Suppressor Cell (MDSC) Fitness and Tumor-Infiltrating Lymphocyte (TIL) Dysfunction. International journal of molecular sciences. 2021;22(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Beccaria K, Sabbagh A, de Groot J, Canney M, Carpentier A, Heimberger A. Blood-brain barrier opening with low intensity pulsed ultrasound for immune modulation and immune therapeutic delivery to CNS tumors. Journal of neuro-oncology. 2021;151(1). [DOI] [PubMed] [Google Scholar]
- 8.Seyfrid M, Maich W, Shaikh V, Tatari N, Upreti D, Piyasena D, et al. CD70 as an actionable immunotherapeutic target in recurrent glioblastoma and its microenvironment. Journal for immunotherapy of cancer. 2022;10(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.O’Rourke D, Nasrallah M, Desai A, Melenhorst J, Mansfield K, Morrissette J, 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. Science translational medicine. 2017;9(399). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.*.Lin Q, Ba T, Ho J, Chen D, Cheng Y, Wang L, et al. First-in-Human Trial of EphA2-Redirected CAR T-Cells in Patients With Recurrent Glioblastoma: A Preliminary Report of Three Cases at the Starting Dose. Front Oncol. 2021;11:694941. [DOI] [PMC free article] [PubMed] [Google Scholar]; The first-reported use of EphA2 CAR T cells in human glioblastoma demonstrates that this treatment is tolerable and transiently efficacious, but the lung toxicities indicate that on-target, off-tumor toxicities are a significant concern for constituitively-active EphA2 CAR T cells
- 11.Chongsathidkiet P, Jackson C, Koyama S, Loebel F, Cui X, Farber S, et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nature medicine. 2018;24(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Liu S, Deng B, Yin Z, Lin Y, An L, Liu D, et al. Combination of CD19 and CD22 CAR-T cell therapy in relapsed B-cell acute lymphoblastic leukemia after allogeneic transplantation. American journal of hematology. 2021;96(6). [DOI] [PubMed] [Google Scholar]
- 13.Kong W, Dimitri A, Wang W, Jung IY, Ott CJ, Fasolino M, et al. BET bromodomain protein inhibition reverses chimeric antigen receptor extinction and reinvigorates exhausted T cells in chronic lymphocytic leukemia. J Clin Invest. 2021;131(16). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Good CR, Aznar MA, Kuramitsu S, Samareh P, Agarwal S, Donahue G, et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell. 2021;184(25):6081–100 e26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ye L, Park JJ, Peng L, Yang Q, Chow RD, Dong MB, et al. A genome-scale gain-of-function CRISPR screen in CD8 T cells identifies proline metabolism as a means to enhance CAR-T therapy. Cell Metab. 2022;34(4):595–614 e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hermans D, Gautam S, Garcia-Canaveras JC, Gromer D, Mitra S, Spolski R, et al. Lactate dehydrogenase inhibition synergizes with IL-21 to promote CD8(+) T cell stemness and antitumor immunity. Proc Natl Acad Sci U S A. 2020;117(11):6047–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Klein Geltink RI, Edwards-Hicks J, Apostolova P, O’Sullivan D, Sanin DE, Patterson AE, et al. Metabolic conditioning of CD8(+) effector T cells for adoptive cell therapy. Nat Metab. 2020;2(8):703–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.*.Wenes M, Jaccard A, Wyss T, Maldonado-Pérez N, Teoh ST, Lepez A, et al. The mitochondrial pyruvate carrier regulates memory T cell differentiation and antitumor function. Cell Metabolism. 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper describes the role of mitochondrial pyurvate carrier (MPC) in determining the fate and function of CD8+T cells. Inhibition of MPC can be implemented during CAR T cell manfacturing process thus imprinting memory like phenotype prior to adoptive transfer.
- 19.Hamaidi I, Zhang L, Kim N, Wang MH, Iclozan C, Fang B, et al. Sirt2 Inhibition Enhances Metabolic Fitness and Effector Functions of Tumor-Reactive T Cells. Cell Metab. 2020;32(3):420–36 e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Verma V, Jafarzadeh N, Boi S, Kundu S, Jiang Z, Fan Y, et al. MEK inhibition reprograms CD8(+) T lymphocytes into memory stem cells with potent antitumor effects. Nat Immunol. 2021;22(1):53–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hirabayashi K, Du H, Xu Y, Shou P, Zhou X, Fuca G, et al. Dual Targeting CAR-T Cells with Optimal Costimulation and Metabolic Fitness enhance Antitumor Activity and Prevent Escape in Solid Tumors. Nat Cancer. 2021;2(9):904–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kawalekar OU, O’Connor RS, Fraietta JA, Guo L, McGettigan SE, Posey AD Jr., et al. Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells. Immunity. 2016;44(2):380–90. [DOI] [PubMed] [Google Scholar]
- 23.Hombach A, Barden M, Hannappel L, Chmielewski M, Rappl G, Sachinidis A, et al. IL12 integrated into the CAR exodomain converts CD8(+) T cells to poly-functional NK-like cells with superior killing of antigen-loss tumors. Mol Ther. 2022;30(2):593–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Chmielewski M, Abken H. CAR T Cells Releasing IL-18 Convert to T-Bet(high) FoxO1(low) Effectors that Exhibit Augmented Activity against Advanced Solid Tumors. Cell Rep. 2017;21(11):3205–19. [DOI] [PubMed] [Google Scholar]
- 25.Alizadeh D, Wong RA, Yang X, Wang D, Pecoraro JR, Kuo CF, et al. IL15 Enhances CAR-T Cell Antitumor Activity by Reducing mTORC1 Activity and Preserving Their Stem Cell Memory Phenotype. Cancer Immunol Res. 2019;7(5):759–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.*.Choe J, Watchmaker P, Simic M, Gilbert R, Li A, Krasnow N, et al. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Science translational medicine. 2021;13(591). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study describes a multi-antigen recognition platform where synthetic Notch receptor senses tumor or brain-specific antigen and drives the expression of tandem CAR to a tumor-associated antigens. This helps to mitigate risk of toxicity and preserves naive state of T cells.
- 27.Hyrenius-Wittsten A, Su Y, Park M, Garcia JM, Alavi J, Perry N, et al. SynNotch CAR circuits enhance solid tumor recognition and promote persistent antitumor activity in mouse models. Sci Transl Med. 2021;13(591). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Srivastava S, Salter AI, Liggitt D, Yechan-Gunja S, Sarvothama M, Cooper K, et al. Logic-Gated ROR1 Chimeric Antigen Receptor Expression Rescues T Cell-Mediated Toxicity to Normal Tissues and Enables Selective Tumor Targeting. Cancer Cell. 2019;35(3):489–503 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Weber EW, Parker KR, Sotillo E, Lynn RC, Anbunathan H, Lattin J, et al. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science. 2021;372(6537). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Juillerat A, Marechal A, Filhol JM, Valogne Y, Valton J, Duclert A, et al. An oxygen sensitive self-decision making engineered CAR T-cell. Sci Rep. 2017;7:39833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kosti P, Opzoomer JW, Larios-Martinez KI, Henley-Smith R, Scudamore CL, Okesola M, et al. Hypoxia-sensing CAR T cells provide safety and efficacy in treating solid tumors. Cell Rep Med. 2021;2(4):100227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Murawska GM, Vogel C, Jan M, Lu X, Schild M, Slabicki M, et al. Repurposing the Damage Repair Protein Methyl Guanine Methyl Transferase as a Ligand Inducible Fusion Degron. ACS Chem Biol. 2022;17(1):24–31. [DOI] [PubMed] [Google Scholar]
- 33.Richman SA, Wang LC, Moon EK, Khire UR, Albelda SM, Milone MC. Ligand-Induced Degradation of a CAR Permits Reversible Remote Control of CAR T Cell Activity In Vitro and In Vivo. Mol Ther. 2020;28(7):1600–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jan M, Scarfo I, Larson RC, Walker A, Schmidts A, Guirguis AA, et al. Reversible ON- and OFF-switch chimeric antigen receptors controlled by lenalidomide. Sci Transl Med. 2021;13(575). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N Engl J Med. 2016;375(26):2561–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.*.Majzner RG, Ramakrishna S, Yeom KW, Patel S, Chinnasamy H, Schultz LM, et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature. 2022;603(7903):934–41. [DOI] [PMC free article] [PubMed] [Google Scholar]; This clinical trial is the first to use GD2-directed CAR T cells against diffuse midline glioma and incorporates a novel lymphodepleting chemotherapy regimen. The clinical improvements experienced by 3 of the 4 patients demonstrate the promise of this treatment in pediatric brainstem gliomas, and the lack of neurotoxicities show that even an antigen with some expression in the brain can be safely used to target glioma cells.
- 37.Vitanza NA, Johnson AJ, Wilson AL, Brown C, Yokoyama JK, Kunkele A, et al. Locoregional infusion of HER2-specific CAR T cells in children and young adults with recurrent or refractory CNS tumors: an interim analysis. Nat Med. 2021;27(9):1544–52. [DOI] [PubMed] [Google Scholar]
- 38.Chen KT, Wei KC, Liu HL. Focused Ultrasound Combined with Microbubbles in Central Nervous System Applications. Pharmaceutics. 2021;13(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.*.Sabbagh A, Beccaria K, Ling X, Marisetty A, Ott M, Caruso H, 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(15):4325–37. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper demonstrates that LIPU-mediated BBB disruption can increase trafficking of immune cells to the CNS, expanding the possible applications of this therapy and raising interesting questions about the mechanism of the BBB disruption
- 40.Kovacs ZI, Kim S, Jikaria N, Qureshi F, Milo B, Lewis BK, et al. Disrupting the blood-brain barrier by focused ultrasound induces sterile inflammation. Proc Natl Acad Sci U S A. 2017;114(1):E75–E84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.*.Haydar D, Houke H, Chiang J, Yi Z, Ode Z, Caldwell K, et al. Cell-surface antigen profiling of pediatric brain tumors: B7-H3 is consistently expressed and can be targeted via local or systemic CAR T-cell delivery. Neuro Oncol. 2021;23(6):999–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper includes a flow panel that assays for the expression some of the most widely-studied CAR antigens in patient samples of several different CNS tumor types. They find that many of these antigens are more heterogeneously expressed in certain tumor types than previously thought, raising questions about the general applicability of these antigen targets.
- 42.Vitanza JG Nicholas, Wilson Ashley, Huang Wenjun, Perez Francisco, Wright Jason, Leary Sarah, Cole Bonnie, Albert Catherine, Pinto Navin, Orentas Rimas, Jensen Michael, Park Julie. IMMU-03. UPDATES ON BRAINCHILD-01, −02, AND −03: PHASE 1 LOCOREGIONAL CAR T CELL TRIALS TARGETING HER2, EGFR, AND B7-H3 FOR CHILDREN WITH RECURRENT CNS TUMORS AND DIPG Neuro-Oncology. 2020;22:Page iii360. [Google Scholar]
- 43.Yang M, Tang X, Zhang Z, Gu L, Wei H, Zhao S, et al. Tandem CAR-T cells targeting CD70 and B7-H3 exhibit potent preclinical activity against multiple solid tumors. Theranostics. 2020;10(17):7622–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ahmed N, Brawley V, Hegde M, Bielamowicz K, Kalra M, Landi D, et al. HER2-Specific Chimeric Antigen Receptor-Modified Virus-Specific T Cells for Progressive Glioblastoma: A Phase 1 Dose-Escalation Trial. JAMA Oncol. 2017;3(8):1094–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.*.Durgin JS, Henderson F, Nasrallah MP, Mohan S, Wang S, Lacey SF, et al. Case Report: Prolonged Survival Following EGFRvIII CAR T Cell Treatment for Recurrent Glioblastoma. Front Oncol. 2021;11:669071. [DOI] [PMC free article] [PubMed] [Google Scholar]; Prolonged survival for 36 months was observed in a patient with recurrent glioblastoma following a single infusion of autologous EGFRvIII-directed CAR-T cells. The patient developed mild CRS symptoms which resolved with dexamethasone and acetaminophen, and no other toxicities were reported. The rate of tumor growth slowed significantly in response to the therapy
- 46.Huang R, Young R, Ellingson B, Veeraraghavan H, Wang W, Tixier F, et al. Volumetric analysis of IDH-mutant lower-grade glioma: a natural history study of tumor growth rates before and after treatment. Neuro-oncology. 2020;22(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jiang H, Yu K, Li M, Cui Y, Ren X, Yang C, et al. Classification of Progression Patterns in Glioblastoma: Analysis of Predictive Factors and Clinical Implications. Frontiers in oncology. 2020;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Katrib A, Jeong H, Fransen N, Henzel K, Miller J. An Inflammatory Landscape for Preoperative Neurologic Deficits in Glioblastoma. Frontiers in genetics. 2019;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Palombi L, Marchetti P, Salvati M, Osti MF, Frati L, Frati A. Interventions to Reduce Neurological Symptoms in Patients with GBM Receiving Radiotherapy: From Theory to Clinical Practice. Anticancer Res. 2018;38(4):2423–7. [DOI] [PubMed] [Google Scholar]
- 50.Harris MG, Hulseberg P, Ling C, Karman J, Clarkson BD, Harding JS, et al. Immune privilege of the CNS is not the consequence of limited antigen sampling. Scientific Reports. 2014;4(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lee D, Santomasso B, Locke F, Ghobadi A, Turtle C, Brudno J, et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2019;25(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vinnakota J, Zeiser R. Acute Graft- Versus-Host Disease, Infections, Vascular Events and Drug Toxicities Affecting the Central Nervous System. Frontiers in immunology. 2021;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.*.Brown C, Rodriguez A, Palmer J, Ostberg J, Naranjo A, Wagner J, et al. Off-the-shelf, Steroid Resistant, IL13Rα2-Specific CAR T Cells for Treatment of Glioblastoma. Neuro-oncology. 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper generated steroid-resistant CAR-T cells which did not exhibit reduced efficacy in the presence of concurrent dexamethasone adminstration. These findings validate gene disruption of the glucocorticoid receptor as a viable strategy to enable corticosteroid administration to patients with GvHD and/or allorejection symptoms without reducing the efficacy of CAR-T therapies
- 54.Sommer C, Boldajipour B, Kuo T, Bentley T, Sutton J, Chen A, et al. Preclinical Evaluation of Allogeneic CAR T Cells Targeting BCMA for the Treatment of Multiple Myeloma. Molecular therapy : the journal of the American Society of Gene Therapy. 2019;27(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Legut M, Dolton G, Mian A, Ottmann O, Sewell A. CRISPR-mediated TCR replacement generates superior anticancer transgenic T cells. Blood. 2018;131(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.*.Choi J, Medikonda R, Saleh L, Kim T, Pant A, Srivastava S, et al. Combination checkpoint therapy with anti-PD-1 and anti-BTLA results in a synergistic therapeutic effect against murine glioblastoma. Oncoimmunology. 2021;10(1). [DOI] [PMC free article] [PubMed] [Google Scholar]; This group genetically disrupted TRAC, beta-2 microglobulin, and PD-1 to generate universal CAR-T cells with enhanced anti-tumor activity in vivo. These findings illustrate the feasibility of leveraging genomic engineering techniques (e.g. CRISPR/Cas9) to both mitigate the risks of adverse effects associated with CAR-T therapies and improve the efficacy/persistence of CAR-T cells via immune checkpoint blockade.
- 57.Nagano S, Maeda T, Ichise H, Kashima S, Ohtaka M, Nakanishi M, et al. High Frequency Production of T Cell-Derived iPSC Clones Capable of Generating Potent Cytotoxic T Cells. Molecular therapy Methods & clinical development. 2019;16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Pan C, Zhai Y, Li G, Jiang T, Zhang W. NK Cell-Based Immunotherapy and Therapeutic Perspective in Gliomas. Front Oncol. 2021;11:751183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ma R, Lu T, Li Z, Teng KY, Mansour AG, Yu M, et al. An Oncolytic Virus Expressing IL15/IL15Ralpha Combined with Off-the-Shelf EGFR-CAR NK Cells Targets Glioblastoma. Cancer Res. 2021;81(13):3635–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rudek LS, Zimmermann K, Galla M, Meyer J, Kuehle J, Stamopoulou A, et al. Generation of an NFkappaB-Driven Alpharetroviral “All-in-One” Vector Construct as a Potent Tool for CAR NK Cell Therapy. Front Immunol. 2021;12:751138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.*.Wang J, Toregrosa-Allen S, Elzey BD, Utturkar S, Lanman NA, Bernal-Crespo V, et al. Multispecific targeting of glioblastoma with tumor microenvironment-responsive multifunctional engineered NK cells. Proc Natl Acad Sci U S A. 2021;118(45). [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper uncovers a novel mechanism for trafficking of NK cells to the tumor microenvironment and identifies a potent combination treatment that may improve the efficacy of CAR NK cells.
- 62.Morris EC, Neelapu SS, Giavridis T, Sadelain M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nature Reviews Immunology. 2021;22(2):85–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Goff S, Morgan R, Yang J, Sherry R, Robbins P, Restifo N, et al. Pilot Trial of Adoptive Transfer of Chimeric Antigen Receptor-transduced T Cells Targeting EGFRvIII in Patients With Glioblastoma. Journal of immunotherapy (Hagerstown, Md : 1997). 2019;42(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Parker KR, Migliorini D, Perkey E, Yost KE, Bhaduri A, Bagga P, et al. Single-Cell Analyses Identify Brain Mural Cells Expressing CD19 as Potential Off-Tumor Targets for CAR-T Immunotherapies. Cell. 2020;183(1):126–42 e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.González H, Pacheco R. T-cell-mediated regulation of neuroinflammation involved in neurodegenerative diseases. Journal of Neuroinflammation. 2014;11(1):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Williams G, Schonhoff A, Jurkuvenaite A, Gallups N, Standaert D, Harms A. CD4 T cells mediate brain inflammation and neurodegeneration in a mouse model of Parkinson’s disease. Brain : a journal of neurology. 2021;144(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Truong NTH, Gargett T, Brown MP, Ebert LM. Effects of Chemotherapy Agents on Circulating Leukocyte Populations: Potential Implications for the Success of CAR-T Cell Therapies. Cancers. 2021;13(9):2225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Yoo H, Liu Y, Wang L, Schubert M, Hoffmann J, Wang S, et al. Tumor-Specific Reactive Oxygen Species Accelerators Improve Chimeric Antigen Receptor T Cell Therapy in B Cell Malignancies. International journal of molecular sciences. 2019;20(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bhatt S, Puli L, Patil CR. Role of reactive oxygen species in the progression of Alzheimer’s disease. Drug discovery today. 2021;26(3). [DOI] [PubMed] [Google Scholar]
- 70.Ciccocioppo F, Lanuti P, Pierdomenico L, Simeone P, Bologna G, Ercolino E, et al. The Characterization of Regulatory T-Cell Profiles in Alzheimer’s Disease and Multiple Sclerosis. Scientific Reports. 2019;9(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sánchez-Valle J, Tejero H, Ibáñez K, Portero JL, Krallinger M, Al-Shahrour F, et al. A molecular hypothesis to explain direct and inverse co-morbidities between Alzheimer’s Disease, Glioblastoma and Lung cancer. Scientific Reports. 2017;7(1):1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523(7560):337–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
