Next Article in Journal
High OCT4 Expression Might Be Associated with an Aggressive Phenotype in Rectal Cancer
Next Article in Special Issue
Targeting Cytokines and Their Pathways for the Treatment of Cancer
Previous Article in Journal
Histology-Validated Dielectric Characterisation of Lung Carcinoma Tissue for Microwave Thermal Ablation Applications
Previous Article in Special Issue
Immunobiology and Cytokine Modulation of the Pediatric Brain Tumor Microenvironment: A Scoping Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Use of Targeted Cytokines as Cancer Therapeutics in Glioblastoma

1
Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
2
Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
3
Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94143, USA
4
Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
5
Department of Neurosurgery, Northwestern University, Chicago, IL60611, USA
6
Simpson Querrey Biomedical Research Center, 303 E. Superior Street, 6-516, Chicago, IL 60611, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(14), 3739; https://doi.org/10.3390/cancers15143739
Submission received: 2 June 2023 / Revised: 19 July 2023 / Accepted: 21 July 2023 / Published: 23 July 2023
(This article belongs to the Special Issue The Use of Targeted Cytokine for Novel Cancer Therapeutics)

Abstract

:

Simple Summary

Despite multi-modal treatment consisting of surgery, chemotherapy, and radiation, glioblastoma inevitably recurs due to its diffuse infiltrative nature. Anti-tumor immune responses, supported by pro-inflammatory cytokines, that can seek out remote cancer vestiges will likely become part of the therapeutic armamentarium but will require thoughtful selection, combinatorial vetting, and innovative delivery strategies.

Abstract

Cytokines play an important role in regulating the immune response. Although there is great interest in exploiting cytokines for cancer immunotherapy, their clinical potential is limited by their pleiotropic properties and instability. A variety of cancer cell-intrinsic and extrinsic characteristics pose a barrier to effective treatments including cytokines. Recent studies using gene and cell therapy offer new opportunities for targeting cytokines or their receptors, demonstrating that they are actionable targets. Current efforts such as virotherapy, systemic cytokine therapy, and cellular and gene therapy have provided novel strategies that incorporate cytokines as potential therapeutic strategies for glioblastoma. Ongoing research on characterizing the tumor microenvironment will be informative for prioritization and combinatorial strategies of cytokines for future clinical trials. Unique therapeutic opportunities exist at the convergence of cytokines that play a dual role in tumorigenesis and immune modulation. Here, we discuss the underlying strategies in pre- and clinical trials aiming to enhance treatment outcomes in glioblastoma patients.

1. Introduction

Glioblastoma isocitrate dehydrogenase wild type (GBM IDHwt) is a highly infiltrative malignancy that is poorly controlled by the standard of care that includes surgery, radiotherapy, chemotherapy, and alternating electrical fields [1,2,3,4]. Objective response rates (ORR) are very low and are influenced, in large part, by the specific mechanism of action of the therapeutics and their effects on imaging parameters more than on direct tumor cytotoxicity [5]. Current treatment approaches for GBM remain challenging due to tumor heterogeneity [6], an immune-suppressive tumor microenvironment (TME) [7], and the highly infiltrative nature of these tumors [8]. Cytokines are soluble small molecules that mediate the interactions between immune and non-immune cells in the TME and either support pro- or anti-inflammatory responses [9]. Targeted delivery of immune modulatory cytokines through either gene- or cell-based strategies [10,11,12,13] may limit adverse effects related to the systemic administration and enhance the efficacy of the treatment [13]. Herein, we focus on cytokine-targeted therapy that mediates crosstalk between cancer and immune cells that have yet to be fully investigated or integrated into treatment strategies.
Efforts to use immune responses to control cancer date back to 1891, when William Coley attempted treatment of sarcoma patients using mixtures of live and inactivated Serratia marcescens and Streptococcus pyogenes [14]. More recently, immunotherapy has become a well-integrated component of the standard of care. This success is grounded in an understanding of the specific mechanisms of immune dysregulation in cancer. Work from Allison et al. has been foundational for inducing immune responses to overcome disseminated cancer and provide prolonged duration of responses [15,16]. Multiple immunotherapy strategies have been and continue to be investigated in clinical trials including combinations utilizing immune checkpoint inhibitors [17,18,19,20] and oncolytic viruses [21,22,23,24,25,26]. However, current immunotherapy strategies have limited benefits in GBM [27]. This lack of responsiveness indicates the need to expand the current approaches designed to treat GBM.

2. Modulation of Tumor Immunogenicity

GBM is a heterogeneous disease that develops a complex TME composed of infiltrating immune cells, vasculature, and fibroblasts exposed to various soluble factors affecting tumor growth [28]. These various factors within the TME determine phenotypic features and treatment outcomes. Cancer cells create an immunosuppressive microenvironment through a variety of mechanisms including inducing immune-suppressive macrophages/microglia [29] and downregulation of antigen presentation [30]. The presence of myeloid-derived suppressor cells (MDSCs) is one of the mechanisms that promote immunosuppressive TME and likely inhibits effective immunotherapy [31]. MDSCs migrate as immature cells from the bone marrow to tumors, where they differentiate into mature macrophages and dendritic cells [32,33]. MDSCs inhibit activation and proliferation of cytotoxic T cells [34] through increased expression of arginase-1 [35], resulting in increased secretion of IL-10 [36] and TGF-β [37]. Tumor-associated microglia/macrophages (TAM) impose additional constraints on anti-tumor immunity [38] by secreting low levels of pro-inflammatory cytokines [39] and compromising T cell function as summarized in Figure 1 and Figure 2 [40]. This immune suppression is further compounded by a paucity of T cells within the TME through sequestration in the bone marrow [41] and irreversible T cell exhaustion [42].

3. Cytokine Biology

Cytokines are secreted proteins that engage the extracellular domains of cell surface receptors and regulate immune response and homeostasis [9]. Cytokines can be classified based on their roles as pro- or anti-inflammatory cytokines [43] or on cellular origin (Table 1). Type 1 (cellular response) cytokines are secreted by CD4+ Th1 and type 2 (humoral response) cytokines are produced by CD4+ Th2 cells [44]. Although the immune regulatory effects of cytokines make them compelling candidates for cancer immunotherapy, undesirable side effects and short serum half-life can restrict clinical implementation [45]. Cytokine pleiotropy, which refers to the ability of cytokines to act on different cell types in the immune system and peripheral tissues, is also a challenge for clinical translation because of off-target effects [46]. Multiple immunomodulatory cytokines have or are being investigated for clinical use, including TGF-β, CSF-1, IL-2, IL-7, IL-10, IL-12, IL-18, IL-21, IL-22, and IFN-α, some of which include glioma patients (Table 2). Only IFN-α and IL-2 have received U.S. Food and Drug Administration (FDA) approval for cancer treatment [47]. There has been limited experience with high-dose IL-2 in GBM patients after one subject had a fatal outcome secondary to herniation associated with marked T cell tumor infiltration that has not been reported. Human interferon alpha 2b (IFN-α2b) was approved for the treatment of hairy cell leukemia in 1986 and recombinant IL-2 for treating melanoma and renal cancers in 1992 [47]. With these treatments, severe side effects can include capillary leak syndrome and cytokine release syndrome, leading to death in some patients. In many instances, the concentration of the cytokine leads to different effects including unwanted off-target toxicities. As opposed to conventional chemotherapy in which the highest tolerated doses are typically used, efforts need to be directed at the identification of the appropriate dose for the desired physiological result in the case of cytokines. As such, the management of cytokines, including toxicities, is a more subtle process with titration of the dose in contrast to more standard pharmacologic management of an “on/off switch” approach. As such, the management of cytokines is a different concept when juxtaposed with cytotoxic chemotherapy where the intention is to maximize cytotoxicity. Given the toxicity of cytokine-based therapies, considerable effort has been focused on targeting cytokines through cytokine-producing viral vector gene therapy and adoptive transfer of cytokine-producing cells. Below, we discuss specific promising cytokine-based approaches undergoing investigation in GBM. We first discuss the targeting of pro-tumoral cytokines followed by a discussion of approaches using pro-inflammatory anti-tumoral cytokines.

4. Targeting Pro-Tumoral Cytokines

4.1. Targeting Transforming Growth Factor β (TGF-β)

TGF-β is a cytokine with pleiotropic effects which may play an important role in anti-tumor immune responses [77]. TGF-β supports stem-like self-renewal and suppression of immune response [78]. TGF-β expression, presumably in the context of the above-described effects, is associated with glioma development and progression [79]. In turn, targeting this cytokine is a rational therapeutic approach. A non-randomized phase 1/2 clinical trial (NCT01220271) showed the safety and tolerability of LY2157299, a small molecule inhibitor of TGF-β receptor type I, in combination with temozolomide and radiation in newly diagnosed high-grade gliomas [80]. However, treatment of patients with LY2157299 and lomustine did not improve the overall survival (OS) relative to monotherapeutic lomustine in patients with recurrent GBM [81]. Another approach for targeting TGF- β involves the use of bintrafusp alfa, a bifunctional protein consisting of an antibody blocking PD-L1 and TGF-β trap [82]. Because PD-L1 can be expressed on some types of cancer cells which prevents T cells from killing, targeting two distinct mechanisms of tumor-mediated immune suppression may show an additive or synergistic effect. Partial responses were observed in a phase 1 trial of this agent in conjunction with radiation and temozolomide in patients with recurrent GBM [82]. Because PD-L1 is not frequently expressed on GBM [83,84], this strategy likely needs to be considered in the context of selected patients. In addition, the size of the therapeutic molecule requires consideration with respect to its ability to adequately cross the blood–brain barrier (BBB) at adequate concentrations to treat the tumor. Antisense nucleotides are another means for targeting TGF-β. These (AP12009) have been investigated in a non-randomized phase 2 trial in which they were directly administered into recurrent tumors using convection-enhanced delivery (CED). Partial and complete responses were observed [85]. There are a number of technical challenges currently associated with CED [86] which limit scalability and dampen the enthusiasm for later-stage clinical investigations.

4.2. CSF-1

Colony-stimulating factor-1 (CSF-1) is a glycoprotein cytokine that functions through the receptor CSF1R [87] and regulates the differentiation of myeloid progenitors into dendritic cells, monocytes, and macrophages [88]. One of the most frequent immune cells within the TME are TAMs. The cells can become polarized to the M1 and M2 states [89,90] in which the M1 state exerts a pro-inflammatory, anti-tumor response [91] and the M2 state promotes tumor growth, invasion, metastasis, and resistance to therapy [92]. TAM-directed therapies using CSF-1 and CSF1R inhibitors have been tested in preclinical models of gliomas [93,94], as well as in clinical studies. A phase 2 trial (NCT01349036) of pexidartinib (PLX3397), a CSF1R inhibitor, in recurrent GBM was well tolerated but did not improve progression-free survival (PFS) [95]. Similarly, a combination of pexidartinib, radiation therapy, and temozolomide did not improve median PFS or OS in newly diagnosed GBM [96]. This lack of effect may be due to, at least in part, compensatory mechanisms such as CSF2-driven macrophage resistance or phosphatidylinositol 3-kinase [97].

4.3. The Paradoxical Targeting of the Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) for Glioblastoma

GM-CSF is a hemopoietic growth factor and is responsible for the expansion and activation of macrophages and granulocytes [98]. GM-CSF modulates cell maturation proliferation and survival. GM-CSF boosts immune responses by promoting T and B cell expansion and differentiation and dendritic cell maturation, proliferation, and migration. It is from this immunological perspective that GM-CSF has been used in oncology clinical trials including a wide variety of peptide vaccine strategies for GBM patients. Notably, GM-CSF is elevated in cancer patients [99]. In glioblastoma, GM-CSF and its receptor can promote tumor progression likely through upregulating anti-apoptotic and pro-angiogenic signals via the activation of the signal transducer and activator of transcription 3 (STAT3) signaling pathway or by increasing the expression of VEGF and its receptor [100,101]. In the tumor environment, tumor cells, and tumor-associated microglial cells secrete GM-CSF [102,103,104]. Inhibiting GM-CSF thereby can suppress cancer cell growth and metastasis [103]. GM-CSF has been used in multiple large vaccine trials for GBM which could have had both beneficial and detrimental effects [1,72]. Given the dual pro-cancer and pro-inflammatory roles of GM-CSF, monotherapy inhibitors will likely not be tested in the context of glioma.

5. Utilizing Anti-Tumoral Cytokines

5.1. Virus-Based Cytokine Expression

Virotherapy is an evolving class of immunotherapies based on the selective replication of these viruses in cancer cells to trigger tumor antigen presentation, immune activation, and subsequent tumor cytotoxicity [20,21,22]. Initiation and activation of apoptosis in the cancer cells and the induction of type I IFN is the underlying mechanism of these types of viruses. Viruses can also be devised to elaborate a variety of cytokines to modulate the immune system that thereby mediates the anti-tumor effect. The first oncolytic virus approved by the FDA in 2015 for the treatment of metastatic melanoma was talimogene laherparepvec (T-VEC), an engineered herpes simplex virus-1 that expresses human GM-CSF [105,106]. A series of preclinical studies have shown that cytokine-armed viruses can enhance immune response and provide additional survival benefits in glioma-bearing mice. For example, a virus expressing IL-4 prolonged survival in tumor-bearing mice [107] and one expressing a single-chain variable fragment of the epidermal growth factor receptor (EGFR) antibody conjugated to CCL5 increased the infiltration of innate and adaptive immune cells [108].
A number of cytokine-elaborating viruses have been tested in GBM [21,22,23,24] but tumor heterogeneity and the immune-suppressive TME have likely compromised clinical effectiveness thus far. Ad–RTS–hIL-12 is an adenoviral vector expressing IL-12 controlled by binding of an orally administered ligand, veledimex [62]. Safety, tolerability, and feasibility were demonstrated in a phase 1 monotherapy trial in recurrent high-grade glioma. The ability to measure extra-CNS spill-over of IL-12 and its downstream product IFN-γ was demonstrated via elevated serum concentrations. Based on preclinical studies, the intracranial concentration of cytokines was likely substantially higher than what could be measured in the serum. Post-treatment resected tumor tissue demonstrated an increase in T cell infiltration of the tumor. This approach has been further investigated in conjunction with PD-1 blockade in the phase 1 [61] and phase 2 settings [109]. As discussed earlier, the highest level of IL-12 production did not appear to be the optimal dose for impacting survival and in turn was not utilized as the phase 2 dose. Two other IL-12-based viral vector gene therapy approaches are currently under investigation in gliomas. Ad-TD-nsIL12, a human adenovirus with three genes deleted and expressing human non-secretory IL-12, was developed to minimize IL-12 toxic effects [110]. A phase I Ad-TD-nsIL12 trial (NCT05717699, NCT05717712) in pediatric patients with diffuse intrinsic pontine glioma is currently recruiting patients in China. Another phase 1/2 trial (NSC 733972) is now enrolling patients with high-grade gliomas to study the combination of M032, a genetically engineered HSV-1 expressing IL-12, with pembrolizumab.

5.2. The Addition of IFN-α with the Standard of Care Temozolomide

IFN-α can inhibit tumor cell proliferation, enhance the cytotoxic activity of macrophages and natural killer (NK) cells, and prevent the formation of blood vessels in tumors [111]. A multi-center randomized phase 3 clinical trial enrolled 199 patients with high-grade gliomas. After receiving standard radiation therapy with concurrent temozolomide, patients were randomized to receive either temozolomide or temozolomide with IFN-α. The median OS of patients in the temozolomide plus IFN-α group was 26.7 months, which was longer than that in the standard of care group of 18.8 months (p = 0.005). Seizure and influenza-like symptoms were more common in the combination group [48]. The potential benefit was consistent with a prior study that demonstrated that a pegylated formulation had some benefit in addition to temozolomide [112]. However, a prior phase III study of 275 randomized high-grade glioma patients had demonstrated that IFN-α did not improve time to disease progression or OS when added to treatment with radiation therapy and carmustine. Patients treated with IFN-α experienced more fevers, chills, myalgia, somnolence, confusion, and neurological deficits [49]. The differences in outcomes between these trials may have been a function of the combination with the type of chemotherapy.

5.3. Systemic Cytokine Therapy in Conjunction with Brain Tumor Vaccines

The objective of cancer vaccines is to stimulate adaptive immunity against tumor antigens to control tumor growth [113]. The first cancer vaccine approved by the FDA was sipuleucel-T (Provenge), which is a personalized vaccine developed using ex vivo activated peripheral-blood mononuclear cells co-incubated with a recombinant fusion protein (PA2024) to control asymptomatic metastatic castration-resistant prostate cancer [114]. Various types of GBM vaccines have been developed that are usually administered in conjunction with GM-CSF [71,115,116,117,118,119,120,121,122,123]. Thus far, they have not demonstrated an improvement in survival. Newer strategies involve the co-administration of additional cytokines to augment the potential activity of glioma vaccines. For example, IL-12 was shown to improve the therapeutic efficacy in preclinical murine models bearing intracranial gliomas treated with dendritic cells loaded with GL261 mRNA [124]. Several different approaches are being investigated with all appearing safe and having acceptable tolerability thus far.

5.4. Cell-Based Therapies

Cell-based therapies rely on genetically modified immune cells such as T, NK, and B cells. Adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells demonstrated success in hematologic malignancies and melanoma with six CAR T cell therapies having received FDA approval [125]. While preclinical studies of CAR T therapy were effective in brain tumor control [126,127], overall response rates have been low, likely because of antigen heterogeneity [128,129] and the immune-suppressive TME [130,131]. CAR T therapy may have the ability to reprogram TME and thus may be a compelling partnering approach with other treatment modalities [131]. Improving CAR T therapy can be achieved by engineered expression of cytokines or their receptors to enhance T cell activation, proliferation, and trafficking. In preclinical testing, disialoganglioside (GD2)-targeting CARs engineered with constitutively active IL-7 receptor or IL-15, enhanced survival in GBM xenograft models [132,133]. In another approach, the expression of CXCR1 or CXCR2 in CAR T cells improved trafficking in a GBM model [134]. An upcoming phase 1 trial (NCT05353530) has been designed to assess the safety and feasibility of IL-8 receptor-modified CD70 CAR T treatment in CD70+ and MGMT-unmethylated GBM patients. IL13 receptor alpha 2 (IL13Ra2) is a monomeric receptor of IL-13 [135] that is expressed in ~70% of GBM patients. IL-13Ra2 is associated with higher-grade glioma and poor prognosis [136]. Data from the clinical experience of IL-13Ra2 CAR T intracranial administration supported the safety of CAR T in patients with recurrent GBM [137].
NK cells have also been evaluated in the treatment of gliomas [138,139]. NK cells, a key component of innate immunity, facilitate cell lysis by degranulation achieved by the activating receptor NK group 2 member D (NKG2D) [140], killer cell immunoglobulin-like receptor (KIR), and coactivating/adhesion DNAX-activating molecule (DNAM-1) [141]. Because NK cells become deactivated by TGF-β in the immune-suppressive TME of GBM [138], these cells are co-administered with IL-2 and a TGF-βR1 inhibitor (NCT05400122) or are genetically modified so that the TGF-βR is deleted (NCT04991870) in ongoing clinical trials for colorectal adenocarcinoma and GBM patients, respectively.

5.5. Cytokines Associated with Toxicity in GLIOMA Patients

Distinct elevated serum cytokines may be associated with side effects in glioma patients. In one study, plasma profiling of patients treated with the antiangiogenic agent aflibercept in 28 patients with recurrent GBM revealed that changes in IL-13 from baseline to 24 h predicted on-target toxicities. Increases in IL-1β, IL-6, and IL-10 at 24 h were significantly associated with fatigue [142].

5.6. The Modern Era of Monitoring Intratumoral Cytokines

Under most circumstances, cytokines of CNS tumor patients are measured in the periphery, and these are likely not fully representative of intra-CNS, including intratumoral, concentrations. To determine both the absolute intratumoral concentrations and to follow the longitudinal kinetics, microdialysis catheters can be implanted with minimal risk [143]. This type of analysis is important since it may also identify those subjects that are showing early signs of response, whereas those who do not demonstrate immune effector responses could be spared further ineffective therapy or an alternative therapy based on the changes in the tumor microenvironment. This is contingent on the conditions that cytokines alone would be biologically meaningful as a biomarker of response and that the captured time point for analysis coincides with the therapeutic monitoring period.

5.7. Modulating Cytokines in Glioma Preclinical Model

There are substantial preclinical efforts to use cytokines, especially in adoptive cellular strategies. For example, IL-7 expressed by CAR T improved the survival outcome in a GBM murine model [144]. In another model, IL-15-modified CAR T also improved median survival [127]. Thus far, it is unclear in what specific contexts these cytokine modifications of CAR T cells should be optimally used, the prioritization of which ones, or the combinations. A key limitation is the distribution of adaptive immune therapies through a complex heterogeneous TME. In addition to the delivery of cytokines using viral vectors, an alternative strategy would be the deposition of cells elaborating cytokines and/or chemokines in the TME using BBB opening ultrasound [26]. This type of strategy allows for large molecules to be deposited into the glioblastoma TME. Our group engineered antigen-presenting cells to express CXCL10. These were deposited into the TME of gliomas and markedly increased the number of T cells in the TME and increased median survival [145]. Moving forward, one could engineer off-the-shelf cells that have been transduced with a variety of pro-inflammatory cytokines that are deposited into the TME using BBB opening ultrasound for sustained delivery.

6. Future Directions

Clinical trials investigating the therapeutic effect of cytokines in glioblastoma patients have demonstrated signals of biological response. Cytokine-modulated therapy in GBM will likely evolve as a combinatorial strategy with other immune therapies. The pathophysiology of the cancer and the mechanisms of resistance will prioritize the selection of the cytokines likely in the context of the specific types of immune therapy. Although studies have provided significant insights into the outcomes of GBM patients treated with cytokine modulation, there are significant areas of investigation needed to fully optimize this strategy. One could argue that targeting key hubs such as p-STAT3 that control many immune-suppressive cytokines might be a more rational strategy in GBM. Recent studies have highlighted the importance of IL-33 in GBM progression. Secretion of IL-33 from glioma cells recruits TAM and microglia and promotes a pro-tumorigenic environment [146]. Phospho-proteomic analysis revealed that IL-33+ tumors have a high expression of p-STAT3. STAT3 is a transcriptional regulator of IL-10 [147] and TGF-β [129]. STAT3 inhibits proinflammatory cytokines and dampens the generation of antigen-dependent T cells and T cell proliferation [148]. A BBB penetrant inhibitor of STAT3 is being advanced into phase II studies in combination with radiation [149,150].
Given the multiplicity and various roles of cytokines in the GBM TME, it is unlikely that a single cytokine-focused therapy will result in patient benefits greater than the well-established standard of care. As such, strategically targeting the most immunosuppressive cytokines while pairing an immune checkpoint blockade is the next logical step. To specifically focus on the biology of the TME, particularly the abundance of myeloid lineage cells preventing adaptive immune responses, would enhance the potential for this therapeutic approach to achieve success [151]. As such, pairing established inhibitors of immunosuppressive signaling in myeloid cells with pro-inflammatory checkpoint blockades could be a worthy avenue. Designing brain-penetrant, homing, and myeloid-specific combinatorial approaches is a daunting task and requires the combined collaboration of vastly different areas of scientific expertise. Nanoparticles modified with mannose residues, the binding partner of the canonical immunosuppressive M2 surface receptor CD206, have been shown to target immunosuppressive TAMs while also carrying a payload capable of reversing their immunosuppression [152]. This strategy is proof-of-principle that targeting and reversing immunosuppression in the TME can be cell-specific. To apply this to GBM, we then need to address the spatial challenges of treating the tumor and overcoming the BBB. BBB opening delivery strategies could be a way to deposit cytokine-elaborating cell factories. The contents and cytokines were selected based on specific TME components assayed during biopsy or through circulating biomarkers, to precisely address the TME of the individual tumor and to work in synergy with therapeutic response to existing checkpoint blockades [153]. Novel cytokines that relate to tumor-associated myeloid cells including osteopontin (OPN) [154], and macrophage inhibitory factor (MIF) [155,156], seem to address the above parameters in that they both regulate myeloid cell trafficking and the immunosuppressive phenotypes in the TME of GBM.

Key Strategic Decisions for the Scientific Community

  • Which cytokines should be prioritized for use and why?
  • How can optimal cytokine concentration/dose be established and what is the best strategy for modulation?
  • Are there some contexts in which certain cytokines should be used relative to others?
  • If we were to devise a cellular biofactory for the deposition of various cytokines into the TME, what should be prioritized?
  • Are there some cytokines that should be explored next for GBM that have not been thus far?

7. Conclusions

Although various studies have provided valuable insight into cytokine-based therapy, significant efforts need to be directed toward selection of cytokine(s) in various indications, optimization of combinatorial strategies, delivery strategies, and companion biomarkers.

Author Contributions

Writing—original manuscript: M.S. and A.B.H.; writing—review and editing: M.S., S.T., C.D., H.N., J.d.G., R.V.L. and A.B.H. All authors have read and agreed to the published version of the manuscript.

Funding

Support was provided by the Lurie Cancer Center and NIH grants P30CA060553, P50CA221747, NS120547, R01CA272639, and CA120813.

Conflicts of Interest

A.B.H. serves on the advisory board of Caris Life Sciences and WCG Oncology Advisory Board, receives royalty and milestone payments from DNAtrix, and has been supported by research grants from Alnylam, Celularity, and AbbVie. Other material support has been provided by Moleculin, Carthera, and Takeda. A.B.H. has received consulting fees from BlueRock Therapeutics, NovoCure. The rest of the authors declare no conflict of interest.

References

  1. Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F.; et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Lukas, R.V.; Wainwright, D.A.; Ladomersky, E.; Sachdev, S.; Sonabend, A.M.; Stupp, R. Newly Diagnosed Glioblastoma: A Review on Clinical Management. Oncology 2019, 33, 91–100. [Google Scholar] [PubMed]
  3. Lukas, R.V.; Mrugala, M.M. Pivotal therapeutic trials for infiltrating gliomas and how they affect clinical practice. Neurooncol. Pract. 2017, 4, 209–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Stupp, R.; Taillibert, S.; Kanner, A.; Read, W.; Steinberg, D.; Lhermitte, B.; Toms, S.; Idbaih, A.; Ahluwalia, M.S.; Fink, K.; et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA 2017, 318, 2306–2316. [Google Scholar] [CrossRef] [Green Version]
  5. Ellingson, B.M.; Wen, P.Y.; Chang, S.M.; van den Bent, M.; Vogelbaum, M.A.; Li, G.; Li, S.; Kim, J.; Youssef, G.; Wick, W.; et al. Objective response rate (ORR) targets for recurrent glioblastoma clinical trials based on the historic association between ORR and median overall survival. Neuro-Oncology 2023, 25, 1017–1028. [Google Scholar] [CrossRef]
  6. Sottoriva, A.; Spiteri, I.; Piccirillo, S.G.; Touloumis, A.; Collins, V.P.; Marioni, J.C.; Curtis, C.; Watts, C.; Tavare, S. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl. Acad. Sci. USA 2013, 110, 4009–4014. [Google Scholar] [CrossRef]
  7. Cui, X.; Ma, C.; Vasudevaraja, V.; Serrano, J.; Tong, J.; Peng, Y.; Delorenzo, M.; Shen, G.; Frenster, J.; Morales, R.T.; et al. Dissecting the immunosuppressive tumor microenvironments in Glioblastoma-on-a-Chip for optimized PD-1 immunotherapy. eLife 2020, 9, e52253. [Google Scholar] [CrossRef]
  8. Drumm, M.R.; Dixit, K.S.; Grimm, S.; Kumthekar, P.; Lukas, R.V.; Raizer, J.J.; Stupp, R.; Chheda, M.G.; Kam, K.L.; McCord, M.; et al. Extensive brainstem infiltration, not mass effect, is a common feature of end-stage cerebral glioblastomas. Neuro-Oncology 2020, 22, 470–479. [Google Scholar] [CrossRef]
  9. Briukhovetska, D.; Dörr, J.; Endres, S.; Libby, P.; Dinarello, C.A.; Kobold, S. Interleukins in cancer: From biology to therapy. Nat. Rev. Cancer 2021, 21, 481–499. [Google Scholar] [CrossRef] [PubMed]
  10. Zhang, D.Y.; Singer, L.; Sonabend, A.M.; Lukas, R.V. Gene Therapy for the Treatment of Malignant Glioma. Adv. Oncol. 2021, 1, 189–202. [Google Scholar] [CrossRef]
  11. Waldmann, T.A. Cytokines in Cancer Immunotherapy. Cold Spring Harb. Perspect. Biol. 2018, 10, a028472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Yang, F.; He, Z.; Duan, H.; Zhang, D.; Li, J.; Yang, H.; Dorsey, J.F.; Zou, W.; Nabavizadeh, S.A.; Bagley, S.J.; et al. Synergistic immunotherapy of glioblastoma by dual targeting of IL-6 and CD40. Nat. Commun. 2021, 12, 3424. [Google Scholar] [CrossRef]
  13. Birocchi, F.; Cusimano, M.; Rossari, F.; Beretta, S.; Rancoita, P.M.V.; Ranghetti, A.; Colombo, S.; Costa, B.; Angel, P.; Sanvito, F.; et al. Targeted inducible delivery of immunoactivating cytokines reprograms glioblastoma microenvironment and inhibits growth in mouse models. Sci. Transl. Med. 2022, 14, eabl4106. [Google Scholar] [CrossRef] [PubMed]
  14. McCarthy, E.F. The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthop. J. 2006, 26, 154–158. [Google Scholar] [PubMed]
  15. van Elsas, A.; Hurwitz, A.A.; Allison, J.P. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med. 1999, 190, 355–366. [Google Scholar] [CrossRef]
  16. Hurwitz, A.A.; Foster, B.A.; Kwon, E.D.; Truong, T.; Choi, E.M.; Greenberg, N.M.; Burg, M.B.; Allison, J.P. Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade. Cancer Res. 2000, 60, 2444–2448. [Google Scholar]
  17. Lukas, R.V.; Rodon, J.; Becker, K.; Wong, E.T.; Shih, K.; Touat, M.; Fasso, M.; Osborne, S.; Molinero, L.; O’Hear, C.; et al. Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma. J. Neurooncol. 2018, 140, 317–328. [Google Scholar] [CrossRef]
  18. Lim, M.; Weller, M.; Idbaih, A.; Steinbach, J.; Finocchiaro, G.; Raval, R.R.; Ansstas, G.; Baehring, J.; Taylor, J.W.; Honnorat, J.; et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro-Oncology 2022, 24, 1935–1949. [Google Scholar] [CrossRef]
  19. Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bahr, O.; et al. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef]
  20. Omuro, A.; Brandes, A.A.; Carpentier, A.F.; Idbaih, A.; Reardon, D.A.; Cloughesy, T.; Sumrall, A.; Baehring, J.; van den Bent, M.; Bähr, O.; et al. Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: An international randomized phase III trial. Neuro-Oncology 2023, 25, 123–134. [Google Scholar] [CrossRef]
  21. Fares, J.; Ahmed, A.U.; Ulasov, I.V.; Sonabend, A.M.; Miska, J.; Lee-Chang, C.; Balyasnikova, I.V.; Chandler, J.P.; Portnow, J.; Tate, M.C.; et al. Neural stem cell delivery of an oncolytic adenovirus in newly diagnosed malignant glioma: A first-in-human, phase 1, dose-escalation trial. Lancet Oncol. 2021, 22, 1103–1114. [Google Scholar] [CrossRef] [PubMed]
  22. Tobias, A.L.; Thaci, B.; Auffinger, B.; Rincon, E.; Balyasnikova, I.V.; Kim, C.K.; Han, Y.; Zhang, L.; Aboody, K.S.; Ahmed, A.U.; et al. The timing of neural stem cell-based virotherapy is critical for optimal therapeutic efficacy when applied with radiation and chemotherapy for the treatment of glioblastoma. Stem Cells Transl. Med. 2013, 2, 655–666. [Google Scholar] [CrossRef] [PubMed]
  23. Gesundheit, B.; Ben-David, E.; Posen, Y.; Ellis, R.; Wollmann, G.; Schneider, E.M.; Aigner, K.; Brauns, L.; Nesselhut, T.; Ackva, I.; et al. Effective Treatment of Glioblastoma Multiforme With Oncolytic Virotherapy: A Case-Series. Front. Oncol. 2020, 10, 702. [Google Scholar] [CrossRef] [PubMed]
  24. Todo, T.; Ino, Y.; Ohtsu, H.; Shibahara, J.; Tanaka, M. A phase I/II study of triple-mutated oncolytic herpes virus G47∆ in patients with progressive glioblastoma. Nat. Commun. 2022, 13, 4119. [Google Scholar] [CrossRef]
  25. Frampton, J.E. Teserpaturev/G47Delta: First Approval. BioDrugs 2022, 36, 667–672. [Google Scholar] [CrossRef]
  26. Nassiri, F.; Patil, V.; Yefet, L.S.; Singh, O.; Liu, J.; Dang, R.M.A.; Yamaguchi, T.N.; Daras, M.; Cloughesy, T.F.; Colman, H.; et al. Oncolytic DNX-2401 virotherapy plus pembrolizumab in recurrent glioblastoma: A phase 1/2 trial. Nat. Med. 2023, 29, 1370–1378. [Google Scholar] [CrossRef]
  27. de Groot, J.; Penas-Prado, M.; Alfaro-Munoz, K.; Hunter, K.; Pei, B.L.; O’Brien, B.; Weathers, S.P.; Loghin, M.; Kamiya Matsouka, C.; Yung, W.K.A.; et al. Window-of-opportunity clinical trial of pembrolizumab in patients with recurrent glioblastoma reveals predominance of immune-suppressive macrophages. Neuro-Oncology 2020, 22, 539–549. [Google Scholar] [CrossRef]
  28. Dahlberg, D.; Rummel, J.; Distante, S.; De Souza, G.A.; Stensland, M.E.; Mariussen, E.; Rootwelt, H.; Voie, O.; Hassel, B. Glioblastoma microenvironment contains multiple hormonal and non-hormonal growth-stimulating factors. Fluids Barriers CNS 2022, 19, 45. [Google Scholar] [CrossRef]
  29. Wu, A.; Wei, J.; Kong, L.Y.; Wang, Y.; Priebe, W.; Qiao, W.; Sawaya, R.; Heimberger, A.B. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro-Oncology 2010, 12, 1113–1125. [Google Scholar] [CrossRef] [Green Version]
  30. Morandi, F.; Fainardi, E.; Rizzo, R.; Rouas-Freiss, N. The role of HLA-class Ib molecules in immune-related diseases, tumors, and infections. J. Immunol. Res. 2014, 2014, 231618. [Google Scholar] [CrossRef]
  31. Kohanbash, G.; Okada, H. Myeloid-derived suppressor cells (MDSCs) in gliomas and glioma-development. Immunol. Investig. 2012, 41, 658–679. [Google Scholar] [CrossRef]
  32. Bronte, V.; Brandau, S.; Chen, S.H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Raber, P.L.; Thevenot, P.; Sierra, R.; Wyczechowska, D.; Halle, D.; Ramirez, M.E.; Ochoa, A.C.; Fletcher, M.; Velasco, C.; Wilk, A.; et al. Subpopulations of myeloid-derived suppressor cells impair T cell responses through independent nitric oxide-related pathways. Int. J. Cancer 2014, 134, 2853–2864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ostrand-Rosenberg, S.; Sinha, P. Myeloid-derived suppressor cells: Linking inflammation and cancer. J. Immunol. 2009, 182, 4499–4506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Rodriguez, P.C.; Ochoa, A.C.; Al-Khami, A.A. Arginine Metabolism in Myeloid Cells Shapes Innate and Adaptive Immunity. Front. Immunol. 2017, 8, 93. [Google Scholar] [CrossRef] [Green Version]
  36. Zhang, Z.; Huang, X.; Li, J.; Fan, H.; Yang, F.; Zhang, R.; Yang, Y.; Feng, S.; He, D.; Sun, W.; et al. Interleukin 10 promotes growth and invasion of glioma cells by up-regulating KPNA 2 in vitro. J. Cancer Res. Ther. 2019, 15, 927–932. [Google Scholar] [CrossRef]
  37. Li, H.; Han, Y.; Guo, Q.; Zhang, M.; Cao, X. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-β 1. J. Immunol. 2009, 182, 240–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Dumas, A.A.; Pomella, N.; Rosser, G.; Guglielmi, L.; Vinel, C.; Millner, T.O.; Rees, J.; Aley, N.; Sheer, D.; Wei, J.; et al. Microglia promote glioblastoma via mTOR-mediated immunosuppression of the tumour microenvironment. EMBO J. 2020, 39, e103790. [Google Scholar] [CrossRef]
  39. Zheng, S.; Hedl, M.; Abraham, C. TAM receptor-dependent regulation of SOCS3 and MAPKs contributes to proinflammatory cytokine downregulation following chronic NOD2 stimulation of human macrophages. J. Immunol. 2015, 194, 1928–1937. [Google Scholar] [CrossRef] [Green Version]
  40. Dannenmann, S.R.; Thielicke, J.; Stockli, M.; Matter, C.; von Boehmer, L.; Cecconi, V.; Hermanns, T.; Hefermehl, L.; Schraml, P.; Moch, H.; et al. Tumor-associated macrophages subvert T-cell function and correlate with reduced survival in clear cell renal cell carcinoma. Oncoimmunology 2013, 2, e23562. [Google Scholar] [CrossRef] [Green Version]
  41. Chongsathidkiet, P.; Jackson, C.; Koyama, S.; Loebel, F.; Cui, X.; Farber, S.H.; Woroniecka, K.; Elsamadicy, A.A.; Dechant, C.A.; Kemeny, H.R.; et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat. Med. 2018, 24, 1459–1468. [Google Scholar] [CrossRef]
  42. Woroniecka, K.; Chongsathidkiet, P.; Rhodin, K.; Kemeny, H.; Dechant, C.; Farber, S.H.; Elsamadicy, A.A.; Cui, X.; Koyama, S.; Jackson, C.; et al. T-Cell Exhaustion Signatures Vary with Tumor Type and Are Severe in Glioblastoma. Clin. Cancer Res. 2018, 24, 4175–4186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chi, H.; Barry, S.P.; Roth, R.J.; Wu, J.J.; Jones, E.A.; Bennett, A.M.; Flavell, R.A. Dynamic regulation of pro- and anti-inflammatory cytokines by MAPK phosphatase 1 (MKP-1) in innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 2274–2279. [Google Scholar] [CrossRef] [PubMed]
  44. Lucey, D.R.; Clerici, M.; Shearer, G.M. Type 1 and type 2 cytokine dysregulation in human infectious, neoplastic, and inflammatory diseases. Clin. Microbiol. Rev. 1996, 9, 532–562. [Google Scholar] [CrossRef]
  45. Aziz, N.; Detels, R.; Quint, J.J.; Li, Q.; Gjertson, D.; Butch, A.W. Stability of cytokines, chemokines and soluble activation markers in unprocessed blood stored under different conditions. Cytokine 2016, 84, 17–24. [Google Scholar] [CrossRef] [Green Version]
  46. Ozaki, K.; Leonard, W.J. Cytokine and cytokine receptor pleiotropy and redundancy. J. Biol. Chem. 2002, 277, 29355–29358. [Google Scholar] [CrossRef] [Green Version]
  47. Floros, T.; Tarhini, A.A. Anticancer Cytokines: Biology and Clinical Effects of Interferon-alpha2, Interleukin (IL)-2, IL-15, IL-21, and IL-12. Semin. Oncol. 2015, 42, 539–548. [Google Scholar] [CrossRef] [Green Version]
  48. Guo, C.; Yang, Q.; Xu, P.; Deng, M.; Jiang, T.; Cai, L.; Li, J.; Sai, K.; Xi, S.; Ouyang, H.; et al. Adjuvant Temozolomide Chemotherapy With or Without Interferon Alfa Among Patients with Newly Diagnosed High-grade Gliomas: A Randomized Clinical Trial. JAMA Netw. Open 2023, 6, e2253285. [Google Scholar] [CrossRef] [PubMed]
  49. Buckner, J.C.; Schomberg, P.J.; McGinnis, W.L.; Cascino, T.L.; Scheithauer, B.W.; O’Fallon, J.R.; Morton, R.F.; Kuross, S.A.; Mailliard, J.A.; Hatfield, A.K.; et al. A phase III study of radiation therapy plus carmustine with or without recombinant interferon-alpha in the treatment of patients with newly diagnosed high-grade glioma. Cancer 2001, 92, 420–433. [Google Scholar] [CrossRef]
  50. Brandes, A.A.; Scelzi, E.; Zampieri, P.; Rigon, A.; Rotilio, A.; Amista, P.; Berti, F.; Fiorentino, M.V. Phase II trial with BCNU plus alpha-interferon in patients with recurrent high-grade gliomas. Am. J. Clin. Oncol. 1997, 20, 364–367. [Google Scholar] [CrossRef]
  51. Buckner, J.C.; Brown, L.D.; Kugler, J.W.; Cascino, T.L.; Krook, J.E.; Mailliard, J.A.; Kardinal, C.G.; Tschetter, L.K.; O’Fallon, J.R.; Scheithauer, B.W. Phase II evaluation of recombinant interferon alpha and BCNU in recurrent glioma. J. Neurosurg. 1995, 82, 430–435. [Google Scholar] [CrossRef] [PubMed]
  52. Dillman, R.O.; Shea, W.M.; Tai, D.F.; Mahdavi, K.; Barth, N.M.; Kharkar, B.R.; Poor, M.M.; Church, C.K.; DePriest, C. Interferon-alpha2a and 13-cis-retinoic acid with radiation treatment for high-grade glioma. Neuro-Oncology 2001, 3, 35–41. [Google Scholar] [CrossRef] [Green Version]
  53. Warren, K.; Bent, R.; Wolters, P.L.; Prager, A.; Hanson, R.; Packer, R.; Shih, J.; Camphausen, K. A phase 2 study of pegylated interferon α-2b (PEG-Intron(®)) in children with diffuse intrinsic pontine glioma. Cancer 2012, 118, 3607–3613. [Google Scholar] [CrossRef] [PubMed]
  54. Wakabayashi, T.; Natsume, A.; Mizusawa, J.; Katayama, H.; Fukuda, H.; Sumi, M.; Nishikawa, R.; Narita, Y.; Muragaki, Y.; Maruyama, T.; et al. JCOG0911 INTEGRA study: A randomized screening phase II trial of interferonβ plus temozolomide in comparison with temozolomide alone for newly diagnosed glioblastoma. J. Neurooncol. 2018, 138, 627–636. [Google Scholar] [CrossRef] [Green Version]
  55. Wakabayashi, T.; Kayama, T.; Nishikawa, R.; Takahashi, H.; Hashimoto, N.; Takahashi, J.; Aoki, T.; Sugiyama, K.; Ogura, M.; Natsume, A.; et al. A multicenter phase I trial of combination therapy with interferon-β and temozolomide for high-grade gliomas (INTEGRA study): The final report. J. Neurooncol. 2011, 104, 573–577. [Google Scholar] [CrossRef] [PubMed]
  56. Fine, H.A.; Wen, P.Y.; Robertson, M.; O’Neill, A.; Kowal, J.; Loeffler, J.S.; Black, P.M. A phase I trial of a new recombinant human beta-interferon (BG9015) for the treatment of patients with recurrent gliomas. Clin. Cancer Res. 1997, 3, 381–387. [Google Scholar]
  57. Packer, R.J.; Prados, M.; Phillips, P.; Nicholson, H.S.; Boyett, J.M.; Goldwein, J.; Rorke, L.B.; Needle, M.N.; Sutton, L.; Zimmerman, R.A.; et al. Treatment of children with newly diagnosed brain stem gliomas with intravenous recombinant beta-interferon and hyperfractionated radiation therapy: A childrens cancer group phase I/II study. Cancer 1996, 77, 2150–2156. [Google Scholar] [CrossRef]
  58. Allen, J.; Packer, R.; Bleyer, A.; Zeltzer, P.; Prados, M.; Nirenberg, A. Recombinant interferon beta: A phase I-II trial in children with recurrent brain tumors. J. Clin. Oncol. 1991, 9, 783–788. [Google Scholar] [CrossRef]
  59. Lillehei, K.O.; Mitchell, D.H.; Johnson, S.D.; McCleary, E.L.; Kruse, C.A. Long-term follow-up of patients with recurrent malignant gliomas treated with adjuvant adoptive immunotherapy. Neurosurgery 1991, 28, 16–23. [Google Scholar] [CrossRef] [PubMed]
  60. Wolff, J.E.; Wagner, S.; Reinert, C.; Gnekow, A.; Kortmann, R.D.; Kuhl, J.; Van Gool, S.W. Maintenance treatment with interferon-gamma and low-dose cyclophosphamide for pediatric high-grade glioma. J. Neurooncol. 2006, 79, 315–321. [Google Scholar] [CrossRef] [PubMed]
  61. Chiocca, E.A.; Gelb, A.B.; Chen, C.C.; Rao, G.; Reardon, D.A.; Wen, P.Y.; Bi, W.L.; Peruzzi, P.; Amidei, C.; Triggs, D.; et al. Combined immunotherapy with controlled interleukin-12 gene therapy and immune checkpoint blockade in recurrent glioblastoma: An open-label, multi-institutional phase I trial. Neuro-Oncology 2022, 24, 951–963. [Google Scholar] [CrossRef]
  62. Chiocca, E.A.; Yu, J.S.; Lukas, R.V.; Solomon, I.H.; Ligon, K.L.; Nakashima, H.; Triggs, D.A.; Reardon, D.A.; Wen, P.; Stopa, B.M.; et al. Regulatable interleukin-12 gene therapy in patients with recurrent high-grade glioma: Results of a phase 1 trial. Sci. Transl. Med. 2019, 11, eaaw5680. [Google Scholar] [CrossRef]
  63. Patel, D.M.; Foreman, P.M.; Nabors, L.B.; Riley, K.O.; Gillespie, G.Y.; Markert, J.M. Design of a Phase I Clinical Trial to Evaluate M032, a Genetically Engineered HSV-1 Expressing IL-12, in Patients with Recurrent/Progressive Glioblastoma Multiforme, Anaplastic Astrocytoma, or Gliosarcoma. Hum. Gene Ther. Clin. Dev. 2016, 27, 69–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lee, E.Q.; Duda, D.G.; Muzikansky, A.; Gerstner, E.R.; Kuhn, J.G.; Reardon, D.A.; Nayak, L.; Norden, A.D.; Doherty, L.; LaFrankie, D.; et al. Phase I and Biomarker Study of Plerixafor and Bevacizumab in Recurrent High-Grade Glioma. Clin. Cancer Res. 2018, 24, 4643–4649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Butowski, N.; Colman, H.; De Groot, J.F.; Omuro, A.M.; Nayak, L.; Wen, P.Y.; Cloughesy, T.F.; Marimuthu, A.; Haidar, S.; Perry, A.; et al. Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: An Ivy Foundation Early Phase Clinical Trials Consortium phase II study. Neuro-Oncology 2016, 18, 557–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Capper, D.; von Deimling, A.; Brandes, A.A.; Carpentier, A.F.; Kesari, S.; Sepulveda-Sanchez, J.M.; Wheeler, H.R.; Chinot, O.; Cher, L.; Steinbach, J.P.; et al. Biomarker and Histopathology Evaluation of Patients with Recurrent Glioblastoma Treated with Galunisertib, Lomustine, or the Combination of Galunisertib and Lomustine. Int. J. Mol. Sci. 2017, 18, 995. [Google Scholar] [CrossRef] [Green Version]
  67. Rodon, J.; Carducci, M.; Sepulveda-Sanchez, J.M.; Azaro, A.; Calvo, E.; Seoane, J.; Brana, I.; Sicart, E.; Gueorguieva, I.; Cleverly, A.; et al. Pharmacokinetic, pharmacodynamic and biomarker evaluation of transforming growth factor-beta receptor I kinase inhibitor, galunisertib, in phase 1 study in patients with advanced cancer. Investig. New Drugs 2015, 33, 357–370. [Google Scholar] [CrossRef] [Green Version]
  68. Bogdahn, U.; Hau, P.; Stockhammer, G.; Venkataramana, N.K.; Mahapatra, A.K.; Suri, A.; Balasubramaniam, A.; Nair, S.; Oliushine, V.; Parfenov, V.; et al. Targeted therapy for high-grade glioma with the TGF-β2 inhibitor trabedersen: Results of a randomized and controlled phase IIb study. Neuro-Oncology 2011, 13, 132–142. [Google Scholar] [CrossRef] [Green Version]
  69. Oshiro, S.; Tsugu, H.; Komatsu, F.; Ohnishi, H.; Ueno, Y.; Sakamoto, S.; Fukushima, T.; Soma, G. Evaluation of intratumoral administration of tumor necrosis factor-alpha in patients with malignant glioma. Anticancer. Res. 2006, 26, 4027–4032. [Google Scholar]
  70. Colman, H.; Berkey, B.A.; Maor, M.H.; Groves, M.D.; Schultz, C.J.; Vermeulen, S.; Nelson, D.F.; Mehta, M.P.; Yung, W.K.; Radiation Therapy Oncology, G. Phase II Radiation Therapy Oncology Group trial of conventional radiation therapy followed by treatment with recombinant interferon-beta for supratentorial glioblastoma: Results of RTOG 9710. Int. J. Radiat. Oncol. Biol. Phys. 2006, 66, 818–824. [Google Scholar] [CrossRef]
  71. Curry, W.T., Jr.; Gorrepati, R.; Piesche, M.; Sasada, T.; Agarwalla, P.; Jones, P.S.; Gerstner, E.R.; Golby, A.J.; Batchelor, T.T.; Wen, P.Y.; et al. Vaccination with Irradiated Autologous Tumor Cells Mixed with Irradiated GM-K562 Cells Stimulates Antitumor Immunity and T Lymphocyte Activation in Patients with Recurrent Malignant Glioma. Clin. Cancer Res. 2016, 22, 2885–2896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Reardon, D.A.; Desjardins, A.; Vredenburgh, J.J.; O’Rourke, D.M.; Tran, D.D.; Fink, K.L.; Nabors, L.B.; Li, G.; Bota, D.A.; Lukas, R.V.; et al. Rindopepimut with Bevacizumab for Patients with Relapsed EGFRvIII-Expressing Glioblastoma (ReACT): Results of a Double-Blind Randomized Phase II Trial. Clin. Cancer Res. 2020, 26, 1586–1594. [Google Scholar] [CrossRef] [PubMed]
  73. Sankhla, S.K.; Nadkarni, J.S.; Bhagwati, S.N. Adoptive immunotherapy using lymphokine-activated killer (LAK) cells and interleukin-2 for recurrent malignant primary brain tumors. J. Neurooncol. 1996, 27, 133–140. [Google Scholar] [CrossRef]
  74. Boiardi, A.; Silvani, A.; Ruffini, P.A.; Rivoltini, L.; Parmiani, G.; Broggi, G.; Salmaggi, A. Loco-regional immunotherapy with recombinant interleukin-2 and adherent lymphokine-activated killer cells (A-LAK) in recurrent glioblastoma patients. Cancer Immunol. Immunother. 1994, 39, 193–197. [Google Scholar] [CrossRef]
  75. Merchant, R.E.; McVicar, D.W.; Merchant, L.H.; Young, H.F. Treatment of recurrent malignant glioma by repeated intracerebral injections of human recombinant interleukin-2 alone or in combination with systemic interferon-alpha. Results of a phase I clinical trial. J. Neurooncol. 1992, 12, 75–83. [Google Scholar] [CrossRef]
  76. Jacobs, S.K.; Wilson, D.J.; Kornblith, P.L.; Grimm, E.A. Interleukin-2 or autologous lymphokine-activated killer cell treatment of malignant glioma: Phase I trial. Cancer Res. 1986, 46, 2101–2104. [Google Scholar] [PubMed]
  77. Batlle, E.; Massague, J. Transforming Growth Factor-beta Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef]
  78. Matsuda, S.; Revandkar, A.; Dubash, T.D.; Ravi, A.; Wittner, B.S.; Lin, M.; Morris, R.; Burr, R.; Guo, H.; Seeger, K.; et al. TGF-β in the microenvironment induces a physiologically occurring immune-suppressive senescent state. Cell Rep. 2023, 42, 112129. [Google Scholar] [CrossRef] [PubMed]
  79. Chao, M.; Liu, N.; Sun, Z.; Jiang, Y.; Jiang, T.; Xv, M.; Jia, L.; Tu, Y.; Wang, L. TGF-β Signaling Promotes Glioma Progression Through Stabilizing Sox9. Front. Immunol. 2020, 11, 592080. [Google Scholar] [CrossRef]
  80. Wick, A.; Desjardins, A.; Suarez, C.; Forsyth, P.; Gueorguieva, I.; Burkholder, T.; Cleverly, A.L.; Estrem, S.T.; Wang, S.; Lahn, M.M.; et al. Phase 1b/2a study of galunisertib, a small molecule inhibitor of transforming growth factor-beta receptor I, in combination with standard temozolomide-based radiochemotherapy in patients with newly diagnosed malignant glioma. Investig. New Drugs 2020, 38, 1570–1579. [Google Scholar] [CrossRef] [Green Version]
  81. Brandes, A.A.; Carpentier, A.F.; Kesari, S.; Sepulveda-Sanchez, J.M.; Wheeler, H.R.; Chinot, O.; Cher, L.; Steinbach, J.P.; Capper, D.; Specenier, P.; et al. A Phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro-Oncology 2016, 18, 1146–1156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Khasraw, M.; Weller, M.; Lorente, D.; Kolibaba, K.; Lee, C.K.; Gedye, C.; de La Fuente, M.I.; Vicente, D.; Reardon, D.A.; Gan, H.K.; et al. Bintrafusp alfa (M7824), a bifunctional fusion protein targeting TGF-β and PD-L1: Results from a phase I expansion cohort in patients with recurrent glioblastoma. Neuro-Oncol. Adv. 2021, 3, vdab058. [Google Scholar] [CrossRef] [PubMed]
  83. Nduom, E.K.; Wei, J.; Yaghi, N.K.; Huang, N.; Kong, L.Y.; Gabrusiewicz, K.; Ling, X.; Zhou, S.; Ivan, C.; Chen, J.Q.; et al. PD-L1 expression and prognostic impact in glioblastoma. Neuro-Oncology 2016, 18, 195–205. [Google Scholar] [CrossRef] [Green Version]
  84. Hodges, T.R.; Ott, M.; Xiu, J.; Gatalica, Z.; Swensen, J.; Zhou, S.; Huse, J.T.; de Groot, J.; Li, S.; Overwijk, W.W.; et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: Implications for immune checkpoint immunotherapy. Neuro-Oncology 2017, 19, 1047–1057. [Google Scholar] [CrossRef] [Green Version]
  85. Uckun, F.M.; Qazi, S.; Hwang, L.; Trieu, V.N. Recurrent or Refractory High-Grade Gliomas Treated by Convection-Enhanced Delivery of a TGFbeta2-Targeting RNA Therapeutic: A Post-Hoc Analysis with Long-Term Follow-Up. Cancers 2019, 11, 1892. [Google Scholar] [CrossRef] [Green Version]
  86. Lonser, R.R.; Sarntinoranont, M.; Morrison, P.F.; Oldfield, E.H. Convection-enhanced delivery to the central nervous system. J. Neurosurg. 2015, 122, 697–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Hume, D.A.; MacDonald, K.P. Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood 2012, 119, 1810–1820. [Google Scholar] [CrossRef] [PubMed]
  88. Rojo, R.; Raper, A.; Ozdemir, D.D.; Lefevre, L.; Grabert, K.; Wollscheid-Lengeling, E.; Bradford, B.; Caruso, M.; Gazova, I.; Sanchez, A.; et al. Deletion of a Csf1r enhancer selectively impacts CSF1R expression and development of tissue macrophage populations. Nat. Commun. 2019, 10, 3215. [Google Scholar] [CrossRef] [Green Version]
  89. Jayasingam, S.D.; Citartan, M.; Thang, T.H.; Mat Zin, A.A.; Ang, K.C.; Ch’ng, E.S. Evaluating the Polarization of Tumor-Associated Macrophages Into M1 and M2 Phenotypes in Human Cancer Tissue: Technicalities and Challenges in Routine Clinical Practice. Front. Oncol. 2019, 9, 1512. [Google Scholar] [CrossRef] [Green Version]
  90. Wu, M.; Wu, L.; Wu, W.; Zhu, M.; Li, J.; Wang, Z.; Li, J.; Ding, R.; Liang, Y.; Li, L.; et al. Phagocytosis of Glioma Cells Enhances the Immunosuppressive Phenotype of Bone Marrow-Derived Macrophages. Cancer Res. 2023, 83, 771–785. [Google Scholar] [CrossRef]
  91. Wang, F.; Zhang, S.; Jeon, R.; Vuckovic, I.; Jiang, X.; Lerman, A.; Folmes, C.D.; Dzeja, P.D.; Herrmann, J. Interferon Gamma Induces Reversible Metabolic Reprogramming of M1 Macrophages to Sustain Cell Viability and Pro-Inflammatory Activity. eBioMedicine 2018, 30, 303–316. [Google Scholar] [CrossRef] [Green Version]
  92. Vidyarthi, A.; Agnihotri, T.; Khan, N.; Singh, S.; Tewari, M.K.; Radotra, B.D.; Chatterjee, D.; Agrewala, J.N. Predominance of M2 macrophages in gliomas leads to the suppression of local and systemic immunity. Cancer Immunol. Immunother. 2019, 68, 1995–2004. [Google Scholar] [CrossRef] [PubMed]
  93. Akkari, L.; Bowman, R.L.; Tessier, J.; Klemm, F.; Handgraaf, S.M.; de Groot, M.; Quail, D.F.; Tillard, L.; Gadiot, J.; Huse, J.T.; et al. Dynamic changes in glioma macrophage populations after radiotherapy reveal CSF-1R inhibition as a strategy to overcome resistance. Sci. Transl. Med. 2020, 12, eaaw7843. [Google Scholar] [CrossRef] [PubMed]
  94. Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [Green Version]
  95. Butowski, N.A.; Colman, H.; Groot, J.F.D.; Omuro, A.M.P.; Nayak, L.; Cloughesy, T.F.; Marimuthu, A.; Perry, A.; Phillips, J.J.; West, B.; et al. A phase 2 study of orally administered PLX3397 in patients with recurrent glioblastoma. J. Clin. Oncol. 2014, 32, 2023. [Google Scholar] [CrossRef]
  96. Colman, H.; Raizer, J.J.; Walbert, T.; Plotkin, S.R.; Chamberlain, M.C.; Wong, E.T.; Puduvalli, V.K.; Reardon, D.A.; Iwamoto, F.M.; Mrugala, M.M.; et al. Phase 1b/2 study of pexidartinib (PEX) in combination with radiation therapy (XRT) and temozolomide (TMZ) in newly diagnosed glioblastoma. J. Clin. Oncol. 2018, 36, 2015. [Google Scholar] [CrossRef]
  97. Klemm, F.; Mockl, A.; Salamero-Boix, A.; Alekseeva, T.; Schaffer, A.; Schulz, M.; Niesel, K.; Maas, R.R.; Groth, M.; Elie, B.T.; et al. Compensatory CSF2-driven macrophage activation promotes adaptive resistance to CSF1R inhibition in breast-to-brain metastasis. Nat. Cancer 2021, 2, 1086–1101. [Google Scholar] [CrossRef]
  98. Lotfi, N.; Thome, R.; Rezaei, N.; Zhang, G.X.; Rezaei, A.; Rostami, A.; Esmaeil, N. Roles of GM-CSF in the Pathogenesis of Autoimmune Diseases: An Update. Front. Immunol. 2019, 10, 1265. [Google Scholar] [CrossRef]
  99. Albulescu, R.; Codrici, E.; Popescu, I.D.; Mihai, S.; Necula, L.G.; Petrescu, D.; Teodoru, M.; Tanase, C.P. Cytokine patterns in brain tumour progression. Mediat. Inflamm. 2013, 2013, 979748. [Google Scholar] [CrossRef]
  100. Jung, K.H.; Chu, K.; Lee, S.T.; Kim, S.J.; Sinn, D.I.; Kim, S.U.; Kim, M.; Roh, J.K. Granulocyte colony-stimulating factor stimulates neurogenesis via vascular endothelial growth factor with STAT activation. Brain Res. 2006, 1073–1074, 190–201. [Google Scholar] [CrossRef]
  101. Ohki, Y.; Heissig, B.; Sato, Y.; Akiyama, H.; Zhu, Z.; Hicklin, D.J.; Shimada, K.; Ogawa, H.; Daida, H.; Hattori, K.; et al. Granulocyte colony-stimulating factor promotes neovascularization by releasing vascular endothelial growth factor from neutrophils. FASEB J. 2005, 19, 2005–2007. [Google Scholar] [CrossRef] [PubMed]
  102. Gabrusiewicz, K.; Ellert-Miklaszewska, A.; Lipko, M.; Sielska, M.; Frankowska, M.; Kaminska, B. Characteristics of the alternative phenotype of microglia/macrophages and its modulation in experimental gliomas. PLoS ONE 2011, 6, e23902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Curran, C.S.; Evans, M.D.; Bertics, P.J. GM-CSF production by glioblastoma cells has a functional role in eosinophil survival, activation, and growth factor production for enhanced tumor cell proliferation. J. Immunol. 2011, 187, 1254–1263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kucerova, L.; Matuskova, M.; Hlubinova, K.; Altanerova, V.; Altaner, C. Tumor cell behaviour modulation by mesenchymal stromal cells. Mol. Cancer 2010, 9, 129. [Google Scholar] [CrossRef] [Green Version]
  105. Dummer, R.; Gyorki, D.E.; Hyngstrom, J.; Berger, A.C.; Conry, R.; Demidov, L.; Sharma, A.; Treichel, S.A.; Radcliffe, H.; Gorski, K.S.; et al. Neoadjuvant talimogene laherparepvec plus surgery versus surgery alone for resectable stage IIIB-IVM1a melanoma: A randomized, open-label, phase 2 trial. Nat. Med. 2021, 27, 1789–1796. [Google Scholar] [CrossRef]
  106. Andtbacka, R.H.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef]
  107. Andreansky, S.; He, B.; van Cott, J.; McGhee, J.; Markert, J.M.; Gillespie, G.Y.; Roizman, B.; Whitley, R.J. Treatment of intracranial gliomas in immunocompetent mice using herpes simplex viruses that express murine interleukins. Gene Ther. 1998, 5, 121–130. [Google Scholar] [CrossRef] [Green Version]
  108. Tian, L.; Xu, B.; Chen, Y.; Li, Z.; Wang, J.; Zhang, J.; Ma, R.; Cao, S.; Hu, W.; Chiocca, E.A.; et al. Specific targeting of glioblastoma with an oncolytic virus expressing a cetuximab-CCL5 fusion protein via innate and adaptive immunity. Nat. Cancer 2022, 3, 1318–1335. [Google Scholar] [CrossRef]
  109. Lukas, R.; Oberheim-Bush, N.A.; Cavaliere, R.; Landolfi, J.; Yu, J.S.; Chen, C.; Cordova, C.; Amidei, C.; Buck, J.Y.; Hadar, N.; et al. CTIM-20. Final Results of Controlled IL-12 Monotherapy and in Combination with PD-1 Inhibitor in Adult Subjects with Recurrent Glioblastoma. Neuro-Oncology 2021, 23, vi54. [Google Scholar] [CrossRef]
  110. Wang, P.; Li, X.; Wang, J.; Gao, D.; Li, Y.; Li, H.; Chu, Y.; Zhang, Z.; Liu, H.; Jiang, G.; et al. Re-designing Interleukin-12 to enhance its safety and potential as an anti-tumor immunotherapeutic agent. Nat. Commun. 2017, 8, 1395. [Google Scholar] [CrossRef] [Green Version]
  111. Vidal, P. Interferon α in cancer immunoediting: From elimination to escape. Scand. J. Immunol. 2020, 91, e12863. [Google Scholar] [CrossRef] [PubMed]
  112. Groves, M.D.; Puduvalli, V.K.; Gilbert, M.R.; Levin, V.A.; Conrad, C.A.; Liu, V.H.; Hunter, K.; Meyers, C.; Hess, K.R.; Alfred Yung, W.K. Two phase II trials of temozolomide with interferon-alpha2b (pegylated and non-pegylated) in patients with recurrent glioblastoma multiforme. Br. J. Cancer 2009, 101, 615–620. [Google Scholar] [CrossRef] [Green Version]
  113. Ott, P.A.; Wu, C.J. Cancer Vaccines: Steering T Cells Down the Right Path to Eradicate Tumors. Cancer Discov. 2019, 9, 476–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Small, E.J.; Schellhammer, P.F.; Higano, C.S.; Redfern, C.H.; Nemunaitis, J.J.; Valone, F.H.; Verjee, S.S.; Jones, L.A.; Hershberg, R.M. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J. Clin. Oncol. 2006, 24, 3089–3094. [Google Scholar] [CrossRef] [Green Version]
  115. Liau, L.M.; Ashkan, K.; Brem, S.; Campian, J.L.; Trusheim, J.E.; Iwamoto, F.M.; Tran, D.D.; Ansstas, G.; Cobbs, C.S.; Heth, J.A.; et al. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination With Extension of Survival Among Patients With Newly Diagnosed and Recurrent Glioblastoma: A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol. 2023, 9, 112–121. [Google Scholar] [CrossRef] [PubMed]
  116. Ahluwalia, M.S.; Reardon, D.A.; Abad, A.P.; Curry, W.T.; Wong, E.T.; Figel, S.A.; Mechtler, L.L.; Peereboom, D.M.; Hutson, A.D.; Withers, H.G.; et al. Phase IIa Study of SurVaxM Plus Adjuvant Temozolomide for Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2023, 41, 1453–1465. [Google Scholar] [CrossRef] [PubMed]
  117. Bota, D.A.; Taylor, T.H.; Piccioni, D.E.; Duma, C.M.; LaRocca, R.V.; Kesari, S.; Carrillo, J.A.; Abedi, M.; Aiken, R.D.; Hsu, F.P.K.; et al. Phase 2 study of AV-GBM-1 (a tumor-initiating cell targeted dendritic cell vaccine) in newly diagnosed Glioblastoma patients: Safety and efficacy assessment. J. Exp. Clin. Cancer Res. 2022, 41, 344. [Google Scholar] [CrossRef]
  118. 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]
  119. Rampling, R.; Peoples, S.; Mulholland, P.J.; James, A.; Al-Salihi, O.; Twelves, C.J.; McBain, C.; Jefferies, S.; Jackson, A.; Stewart, W.; et al. A Cancer Research UK First Time in Human Phase I Trial of IMA950 (Novel Multipeptide Therapeutic Vaccine) in Patients with Newly Diagnosed Glioblastoma. Clin. Cancer Res. 2016, 22, 4776–4785. [Google Scholar] [CrossRef] [Green Version]
  120. Heimberger, A.B.; Crotty, L.E.; Archer, G.E.; Hess, K.R.; Wikstrand, C.J.; Friedman, A.H.; Friedman, H.S.; Bigner, D.D.; Sampson, J.H. Epidermal growth factor receptor VIII peptide vaccination is efficacious against established intracerebral tumors. Clin. Cancer Res. 2003, 9, 4247–4254. [Google Scholar]
  121. Sampson, J.H.; Archer, G.E.; Mitchell, D.A.; Heimberger, A.B.; Herndon, J.E., 2nd; Lally-Goss, D.; McGehee-Norman, S.; Paolino, A.; Reardon, D.A.; Friedman, A.H.; et al. An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme. Mol. Cancer Ther. 2009, 8, 2773–2779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Sampson, J.H.; Heimberger, A.B.; Archer, G.E.; Aldape, K.D.; Friedman, A.H.; Friedman, H.S.; Gilbert, M.R.; Herndon, J.E., 2nd; McLendon, R.E.; Mitchell, D.A.; et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 2010, 28, 4722–4729. [Google Scholar] [CrossRef] [Green Version]
  123. Schuster, J.; Lai, R.K.; Recht, L.D.; Reardon, D.A.; Paleologos, N.A.; Groves, M.D.; Mrugala, M.M.; Jensen, R.; Baehring, J.M.; Sloan, A.; et al. A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: The ACT III study. Neuro-Oncology 2015, 17, 854–861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Ciesielski, M.J.; Apfel, L.; Barone, T.A.; Castro, C.A.; Weiss, T.C.; Fenstermaker, R.A. Antitumor effects of a xenogeneic survivin bone marrow derived dendritic cell vaccine against murine GL261 gliomas. Cancer Immunol. Immunother. 2006, 55, 1491–1503. [Google Scholar] [CrossRef] [PubMed]
  125. Sadelain, M.; Riviere, I.; Riddell, S. Therapeutic T cell engineering. Nature 2017, 545, 423–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Pituch, K.C.; Miska, J.; Krenciute, G.; Panek, W.K.; Li, G.; Rodriguez-Cruz, T.; Wu, M.; Han, Y.; Lesniak, M.S.; Gottschalk, S.; et al. Adoptive Transfer of IL13Ralpha2-Specific Chimeric Antigen Receptor T Cells Creates a Pro-inflammatory Environment in Glioblastoma. Mol. Ther. 2018, 26, 986–995. [Google Scholar] [CrossRef] [Green Version]
  127. Zannikou, M.; Duffy, J.T.; Levine, R.N.; Seblani, M.; Liu, Q.; Presser, A.; Arrieta, V.A.; Chen, C.J.; Sonabend, A.M.; Horbinski, C.M.; et al. IL15 modification enables CAR T cells to act as a dual targeting agent against tumor cells and myeloid-derived suppressor cells in GBM. J. Immunother. Cancer 2023, 11, e006239. [Google Scholar] [CrossRef]
  128. 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] [Green Version]
  129. Brown, C.E.; Alizadeh, D.; Starr, R.; Weng, L.; Wagner, J.R.; Naranjo, A.; Ostberg, J.R.; Blanchard, M.S.; Kilpatrick, J.; Simpson, J.; et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N. Engl. J. Med. 2016, 375, 2561–2569. [Google Scholar] [CrossRef]
  130. Chae, M.; Peterson, T.E.; Balgeman, A.; Chen, S.; Zhang, L.; Renner, D.N.; Johnson, A.J.; Parney, I.F. Increasing glioma-associated monocytes leads to increased intratumoral and systemic myeloid-derived suppressor cells in a murine model. Neuro-Oncology 2015, 17, 978–991. [Google Scholar] [CrossRef] [Green Version]
  131. Ott, M.; Prins, R.M.; Heimberger, A.B. The immune landscape of common CNS malignancies: Implications for immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 729–744. [Google Scholar] [CrossRef]
  132. Shum, T.; Omer, B.; Tashiro, H.; Kruse, R.L.; Wagner, D.L.; Parikh, K.; Yi, Z.; Sauer, T.; Liu, D.; Parihar, R.; et al. Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells. Cancer Discov. 2017, 7, 1238–1247. [Google Scholar] [CrossRef] [Green Version]
  133. Gargett, T.; Ebert, L.M.; Truong, N.T.H.; Kollis, P.M.; Sedivakova, K.; Yu, W.; Yeo, E.C.F.; Wittwer, N.L.; Gliddon, B.L.; Tea, M.N.; et al. GD2-targeting CAR-T cells enhanced by transgenic IL-15 expression are an effective and clinically feasible therapy for glioblastoma. J. Immunother. Cancer 2022, 10, e005187. [Google Scholar] [CrossRef]
  134. Jin, L.; Tao, H.; Karachi, A.; Long, Y.; Hou, A.Y.; Na, M.; Dyson, K.A.; Grippin, A.J.; Deleyrolle, L.P.; Zhang, W.; et al. CXCR1- or CXCR2-modified CAR T cells co-opt IL-8 for maximal antitumor efficacy in solid tumors. Nat. Commun. 2019, 10, 4016. [Google Scholar] [CrossRef] [Green Version]
  135. Lupardus, P.J.; Birnbaum, M.E.; Garcia, K.C. Molecular basis for shared cytokine recognition revealed in the structure of an unusually high affinity complex between IL-13 and IL-13Ralpha2. Structure 2010, 18, 332–342. [Google Scholar] [CrossRef] [Green Version]
  136. Brown, C.E.; Warden, C.D.; Starr, R.; Deng, X.; Badie, B.; Yuan, Y.C.; Forman, S.J.; Barish, M.E. Glioma IL13Ralpha2 is associated with mesenchymal signature gene expression and poor patient prognosis. PLoS ONE 2013, 8, e77769. [Google Scholar] [CrossRef]
  137. Brown, C.E.; Badie, B.; Barish, M.E.; Weng, L.; Ostberg, J.R.; Chang, W.C.; Naranjo, A.; Starr, R.; Wagner, J.; Wright, C.; et al. Bioactivity and Safety of IL13Ralpha2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2015, 21, 4062–4072. [Google Scholar] [CrossRef] [Green Version]
  138. Shaim, H.; Shanley, M.; Basar, R.; Daher, M.; Gumin, J.; Zamler, D.B.; Uprety, N.; Wang, F.; Huang, Y.; Gabrusiewicz, K.; et al. Targeting the alphav integrin/TGF-β axis improves natural killer cell function against glioblastoma stem cells. J. Clin. Investig. 2021, 131, e142116. [Google Scholar] [CrossRef]
  139. Mitwasi, N.; Feldmann, A.; Arndt, C.; Koristka, S.; Berndt, N.; Jureczek, J.; Loureiro, L.R.; Bergmann, R.; Mathe, D.; Hegedus, N.; et al. “UniCAR”-modified off-the-shelf NK-92 cells for targeting of GD2-expressing tumour cells. Sci. Rep. 2020, 10, 2141. [Google Scholar] [CrossRef] [Green Version]
  140. Raulet, D.H. Roles of the NKG2D immunoreceptor and its ligands. Nat. Rev. Immunol. 2003, 3, 781–790. [Google Scholar] [CrossRef]
  141. Sayitoglu, E.C.; Georgoudaki, A.M.; Chrobok, M.; Ozkazanc, D.; Josey, B.J.; Arif, M.; Kusser, K.; Hartman, M.; Chinn, T.M.; Potens, R.; et al. Boosting Natural Killer Cell-Mediated Targeting of Sarcoma Through DNAM-1 and NKG2D. Front. Immunol. 2020, 11, 40. [Google Scholar] [CrossRef]
  142. Shonka, N.; Piao, Y.; Gilbert, M.; Yung, A.; Chang, S.; DeAngelis, L.M.; Lassman, A.B.; Liu, J.; Cloughesy, T.; Robins, H.I.; et al. Cytokines associated with toxicity in the treatment of recurrent glioblastoma with aflibercept. Target. Oncol. 2013, 8, 117–125. [Google Scholar] [CrossRef] [Green Version]
  143. Lynes, J.; Jackson, S.; Sanchez, V.; Dominah, G.; Wang, X.; Kuek, A.; Hayes, C.P.; Benzo, S.; Scott, G.C.; Chittiboina, P.; et al. Cytokine Microdialysis for Real-Time Immune Monitoring in Glioblastoma Patients Undergoing Checkpoint Blockade. Neurosurgery 2019, 84, 945–953. [Google Scholar] [CrossRef] [Green Version]
  144. Swan, S.L.; Mehta, N.; Ilich, E.; Shen, S.H.; Wilkinson, D.S.; Anderson, A.R.; Segura, T.; Sanchez-Perez, L.; Sampson, J.H.; Bellamkonda, R.V. IL7 and IL7 Flt3L co-expressing CAR T cells improve therapeutic efficacy in mouse EGFRvIII heterogeneous glioblastoma. Front. Immunol. 2023, 14, 1085547. [Google Scholar] [CrossRef]
  145. 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]
  146. De Boeck, A.; Ahn, B.Y.; D’Mello, C.; Lun, X.; Menon, S.V.; Alshehri, M.M.; Szulzewsky, F.; Shen, Y.; Khan, L.; Dang, N.H.; et al. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat. Commun. 2020, 11, 4997. [Google Scholar] [CrossRef]
  147. Benkhart, E.M.; Siedlar, M.; Wedel, A.; Werner, T.; Ziegler-Heitbrock, H.W. Role of Stat3 in lipopolysaccharide-induced IL-10 gene expression. J. Immunol. 2000, 165, 1612–1617. [Google Scholar] [CrossRef]
  148. Oh, H.M.; Yu, C.R.; Golestaneh, N.; Amadi-Obi, A.; Lee, Y.S.; Eseonu, A.; Mahdi, R.M.; Egwuagu, C.E. STAT3 protein promotes T-cell survival and inhibits interleukin-2 production through up-regulation of Class O Forkhead transcription factors. J. Biol. Chem. 2011, 286, 30888–30897. [Google Scholar] [CrossRef] [Green Version]
  149. Ott, M.; Kassab, C.; Marisetty, A.; Hashimoto, Y.; Wei, J.; Zamler, D.; Leu, J.S.; Tomaszowski, K.H.; Sabbagh, A.; Fang, D.; et al. Radiation with STAT3 Blockade Triggers Dendritic Cell-T cell Interactions in the Glioma Microenvironment and Therapeutic Efficacy. Clin. Cancer Res. 2020, 26, 4983–4994. [Google Scholar] [CrossRef]
  150. Groot, J.; Ott, M.; Wei, J.; Kassab, C.; Fang, D.; Najem, H.; O’Brien, B.; Weathers, S.P.; Matsouka, C.K.; Majd, N.K.; et al. A first-in-human Phase I trial of the oral p-STAT3 inhibitor WP1066 in patients with recurrent malignant glioma. CNS Oncol. 2022, 11, CNS87. [Google Scholar] [CrossRef]
  151. Charles, N.A.; Holland, E.C.; Gilbertson, R.; Glass, R.; Kettenmann, H. The brain tumor microenvironment. Glia 2012, 60, 1169–1180. [Google Scholar] [CrossRef]
  152. Wei, Z.; Zhang, X.; Yong, T.; Bie, N.; Zhan, G.; Li, X.; Liang, Q.; Li, J.; Yu, J.; Huang, G.; et al. Boosting anti-PD-1 therapy with metformin-loaded macrophage-derived microparticles. Nat. Commun. 2021, 12, 440. [Google Scholar] [CrossRef]
  153. Mullick Chowdhury, S.; Lee, T.; Willmann, J.K. Ultrasound-guided drug delivery in cancer. Ultrasonography 2017, 36, 171–184. [Google Scholar] [CrossRef] [Green Version]
  154. Wei, J.; Marisetty, A.; Schrand, B.; Gabrusiewicz, K.; Hashimoto, Y.; Ott, M.; Grami, Z.; Kong, L.Y.; Ling, X.; Caruso, H.; et al. Osteopontin mediates glioblastoma-associated macrophage infiltration and is a potential therapeutic target. J. Clin. Investig. 2019, 129, 137–149. [Google Scholar] [CrossRef]
  155. Lee, S.H.; Kwon, H.J.; Park, S.; Kim, C.I.; Ryu, H.; Kim, S.S.; Park, J.B.; Kwon, J.T. Macrophage migration inhibitory factor (MIF) inhibitor 4-IPP downregulates stemness phenotype and mesenchymal trans-differentiation after irradiation in glioblastoma multiforme. PLoS ONE 2021, 16, e0257375. [Google Scholar] [CrossRef]
  156. Mangano, K.; Mazzon, E.; Basile, M.S.; Di Marco, R.; Bramanti, P.; Mammana, S.; Petralia, M.C.; Fagone, P.; Nicoletti, F. Pathogenic role for macrophage migration inhibitory factor in glioblastoma and its targeting with specific inhibitors as novel tailored therapeutic approach. Oncotarget 2018, 9, 17951–17970. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The dynamics of cytokines in the glioblastoma tumor microenvironment (TME). A cartoon depiction of the cytokines that modulate anti-tumor immune responses. Production of immune-suppressive cytokines shown in red are counterbalanced by pro-inflammatory cytokines shown in green. Cytokines that have different immunological roles depending on context are shown in black. A variety of cells within the TME elaborate these cytokines with some, such as macrophages, being abundant, whereas T cells are relatively rare.
Figure 1. The dynamics of cytokines in the glioblastoma tumor microenvironment (TME). A cartoon depiction of the cytokines that modulate anti-tumor immune responses. Production of immune-suppressive cytokines shown in red are counterbalanced by pro-inflammatory cytokines shown in green. Cytokines that have different immunological roles depending on context are shown in black. A variety of cells within the TME elaborate these cytokines with some, such as macrophages, being abundant, whereas T cells are relatively rare.
Cancers 15 03739 g001
Figure 2. Immunological features of tumors based on cellular and cytokine composition within the tumor microenvironment (TME). Tumors that are devoid of cytotoxic T cells and pro-inflammatory cytokines such as IL-2, IFN-γ, and TNF-α, but with immune-suppressive cytokines such as TGF-β and immune-suppressive cells such as tumor-associated macrophages (TAMs), are designated as immunologically cold. This cold TME is associated with microglia infiltration. In a hot TME, which is rare in glioblastoma, there would be abundant CD8 cytotoxic T cells and dendritic cells alongside pro-inflammatory cytokines.
Figure 2. Immunological features of tumors based on cellular and cytokine composition within the tumor microenvironment (TME). Tumors that are devoid of cytotoxic T cells and pro-inflammatory cytokines such as IL-2, IFN-γ, and TNF-α, but with immune-suppressive cytokines such as TGF-β and immune-suppressive cells such as tumor-associated macrophages (TAMs), are designated as immunologically cold. This cold TME is associated with microglia infiltration. In a hot TME, which is rare in glioblastoma, there would be abundant CD8 cytotoxic T cells and dendritic cells alongside pro-inflammatory cytokines.
Cancers 15 03739 g002
Table 1. Cytokine control of the immune system.
Table 1. Cytokine control of the immune system.
MediatorCellular SourceFunction
IL-1Macrophages, epithelial cellsPro-inflammatory, macrophage, and Th17 cell activation
IL-2T cellsEffector T cell and regulatory T cell growth factor
IL-4Th-cellsT and B cell proliferation and B cell differentiation
IL-6Macrophages, T cells, endothelial cellsBoth pro-inflammatory and immune suppressive, increased antibody production
IL-8Macrophages, epithelial cellsRecruitment of neutrophils
IL-9Th9 cellsActivation of mast cells
IL-10Regulatory T cells, Th9 cellsImmune suppressive, inhibition of Th1 cells
IL-11Fibroblasts, neurons Immune suppression
IL-12Dendritic cells, macrophagesActivation of Th1, induction of interferon from cytotoxic T cells and NK cells
IL-15CD8 T cells, NK cells Expansion of memory CD8 and NK cells
IL-17Th17 cells, NK cellsPromotes neutrophilic inflammation
IL-18Monocytes, macrophages, dendritic cellsPro-inflammatory, activation of the Th1 pathway
IL-33Macrophages, dendritic cells, mast cells, epithelial cellsPro-inflammatory, amplification of Th1 and Th2 cells, activation of NK cells
IFN-γTh1 cells, cytotoxic T and NK cells Pro-inflammatory and activates macrophages
Tumor necrosis factorMacrophages, T cells, NK cells Pro-inflammatory increases vascular permeability
GM-CSF Macrophages, T cells, NK cells, and endothelial cells Pro-inflammatory but glioma propagating
VEGFMacrophages Angiogenesis
TGF-βMacrophages, T cellsImmune suppressive
CXCL9Monocytes, endothelial cells Recruitment of Th1, NK, and dendritic cells
CXCL10Monocytes, endothelial cellsRecruitment of macrophages, Th1, and NK cells
CXCL12 Mesenchymal stem cells Chemotactic for T cells
CCL2Macrophages, dendritic cells Recruitment of Th2, monocytes, and dendritic cells
CCL3 Monocytes, neutrophils, dendritic cells Recruitment of macrophages, Th2, NK, and dendritic cells
CCL4Macrophages, neutrophils, endothelium Recruitment of macrophages, Th1 cells, NK, and dendritic cells
CXCL13B cellsRecruitment of B cells, CD4 T, and dendritic cells
Table 2. Chemokine clinical trials in glioma patients.
Table 2. Chemokine clinical trials in glioma patients.
MediatorPhaseTherapeutic BenefitSide Effects Reference
IFN-α3Increase in overall survival in combination with the current standard of careSeizures and flu-like symptoms[48]
3No benefit in combination with radiation and carmustine Fevers, chills, myalgia, somnolence, confusion, and neurological deficits [49,50,51]
IFN- α-2a2No benefit Dermatological effects [52]
IFN-α-2b (PEG-Intron) 2No benefit in DIPG patients Well tolerated [53]
IFN-β2No benefit in combination with the current standard of care Increased neutropenia [54,55,56,57,58,59]
IFN-γ2No benefit Well tolerated [60]
IL-121Safety Well tolerated [61,62,63]
CXCR4 inhibitor 1Safety Well tolerated [64]
CSF-1 inhibitor2No benefit Well tolerated [65]
TGF-βR12Safety Preserved T cell counts[66,67]
TGF- βR22No benefit Seizures, edema [68]
TNF-α1Safety Well tolerated [69,70]
GM-CSF 3No benefit Well tolerated [1,71,72]
IL-2 1No benefit Fatigue, edema [73,74,75,76]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sooreshjani, M.; Tripathi, S.; Dussold, C.; Najem, H.; de Groot, J.; Lukas, R.V.; Heimberger, A.B. The Use of Targeted Cytokines as Cancer Therapeutics in Glioblastoma. Cancers 2023, 15, 3739. https://doi.org/10.3390/cancers15143739

AMA Style

Sooreshjani M, Tripathi S, Dussold C, Najem H, de Groot J, Lukas RV, Heimberger AB. The Use of Targeted Cytokines as Cancer Therapeutics in Glioblastoma. Cancers. 2023; 15(14):3739. https://doi.org/10.3390/cancers15143739

Chicago/Turabian Style

Sooreshjani, Moloud, Shashwat Tripathi, Corey Dussold, Hinda Najem, John de Groot, Rimas V. Lukas, and Amy B. Heimberger. 2023. "The Use of Targeted Cytokines as Cancer Therapeutics in Glioblastoma" Cancers 15, no. 14: 3739. https://doi.org/10.3390/cancers15143739

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