Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 This entry provides up-to-date knowledge on current state, challenges, and future perspectives on immunotherapy for glioblastoma. + 4917 word(s) 4917 2020-08-20 06:32:59 |
2 format correct -93 word(s) 4824 2020-08-25 10:48:11 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Yang, M.; Oh, I.Y.; Mahanty, A.; Jin, W.; Yoo, J.S. Immunotherapy for Glioblastoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/1738 (accessed on 26 April 2024).
Yang M, Oh IY, Mahanty A, Jin W, Yoo JS. Immunotherapy for Glioblastoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/1738. Accessed April 26, 2024.
Yang, Minfeng, In Young Oh, Arpan Mahanty, Wei-Lin Jin, Jung Sun Yoo. "Immunotherapy for Glioblastoma" Encyclopedia, https://encyclopedia.pub/entry/1738 (accessed April 26, 2024).
Yang, M., Oh, I.Y., Mahanty, A., Jin, W., & Yoo, J.S. (2020, August 20). Immunotherapy for Glioblastoma. In Encyclopedia. https://encyclopedia.pub/entry/1738
Yang, Minfeng, et al. "Immunotherapy for Glioblastoma." Encyclopedia. Web. 20 August, 2020.
Immunotherapy for Glioblastoma
Edit

       Glioblastoma (GBM) is the deadliest and most aggressive neuroepithelial cancer of the central nervous system (CNS) with an abysmal median survival of 14.6-month despite the multiple forms of intervention. In the United States, the total annual incidence rate of glioma has been ~6 cases per 100,000 individuals, of which GBM accounts for about 50% of the cases, with a higher predominance in males. Clinical studies have indicated that most of the GBM patients present an intact blood–brain barrier (BBB) for certain brain regions, capable of blocking the delivery of agents to cancer sites. The BBB is considered to prevent diffusion of 98% of small-molecule and 100% of large-molecule agents into the brain from blood circulation. Given the aggressive and heterogeneous nature of GBM and the blocking capability of BBB, a very limited number of medications for patients with GBM is available in clinics. In addition, due to the existence of other cellular and extracellular barriers, as well as the development of drug resistance over the treatment course, the efficacy of many current therapeutic approaches has been compromised.

       Currently available standards of care for GBM include maximal tumor resection followed by radiotherapy, chemotherapy, and corticosteroids, all of which have immune suppressive characteristics. Unfortunately, complete surgical removal of the whole tumor is almost impossible due to their diffusive characteristics into normal brain tissue. Some reports indicated that ~65% of the post-surgery cases still showed residual tumor cells, which eventually contributed to a high relapse rate of GBM . Therefore, GBM patients may undergo repeated surgical resection, radiotherapy, chemotherapy, or additional bevacizumab treatment. Eventually, most of the patients suffering from GBM will relapse despite an ample set of interventional approaches. According to the data from Surveillance and Epidemiology, the median overall survival (OS) of GBM patients was normally less than 2 years from the time of first progression or relapse. An international phase III randomized trial, conducted by the European Organization for Research and Treatment of Cancer/National Cancer Institute of Canada (EORTC/NCIC), has shown that the median OS of GBM patients who received radiotherapy and Temozolomide therapy remains poor (14.6 months). Moreover, Grossman and colleagues found that the utilization of systemic chemotherapy and hyperfractionated radiation therapy with corticosteroids is likely to disable immune activity. Immune-suppressive characteristics, high toxicity, and lower OS of traditional care made a considerable number of GBM patients (~50%) not accept any second-line of anti-tumor treatment. In addition, there is no evidence that traditional intervention can significantly impact the OS rate under a recurrence setting. Accordingly, given the poor prognosis and limited therapy regimens for patients affected by GBM, there is an urgent need to develop novel therapeutic approaches.

glioblastoa immune-checkpoint inhibitors tumor microenvironment tumor-associated macrophages and microglia immune-related adverse events

1. Immune Microenvironment of Glioblastoma

       It has been regarded that the CNS lacks dedicated lymphatic channels for a long time. The CNS was considered as an immune privileged system, devoid of any immune cells. This overstated historical notion was mainly based on the experimental data reported by Peter Medawar, where foreign grafts transplanted into the brains of rodents did not induce any immune response and the same foreign grafts transplanted into other tissues or organs were rejected [1][2][3][4]. However, this perception has been challenged recently since several studies showed vigorous immunosurveillance and meaningful immune response in the CNS [5][6]. For example, the discovery of a novel route of lymphatic-based channels, reported by Louveau and colleagues [5] in 2015 and the findings of robust immune responses in multiple inflammatory conditions [6] have both demonstrated the CNS as a region for active immunosurveillance. Such findings prompted an increase in studies for the feasibility of cancer immunotherapy towards brain tumors. Although immunotherapy holds great potential for treatment of malignant GBM, unique GBM-associated immune suppression and immune escape still provide challenges to generate efficient anti-tumor responses [4][7]. GBM can form a highly immunosuppressive milieu, mediated by distinct immune or tumor cells (Figure 1). Tumor cells normally express plenty of immunosuppressive factors, such as programmed cell death 1 ligand 1 (PD-L1) and indolamine 2,3-dioxygenase (IDO), while reducing antigen presentation by diminishing major histocompatibility complex (MHC) expression [4]. Notably, gliomas produce IDO, whose function relates to the recruitment of regulatory T (Treg) cells and the inhibition of effector T cells through tryptophan depletion [4][8]. In the context of microglial cells, these often secrete transforming growth factor β (TGFβ) and/or interleukin 10 (IL-10) to decrease the amount of myeloid and lymphoid immune cells to boost systemic immunosuppression [4][9][10] (Figure 1). The lymphoid compartment also mediates immunosuppressive effects with Treg cells through upregulation of different soluble factors and some immune-checkpoint molecules [4]. These immunosuppressive factors may ultimately block T-cell proliferation and activation. One unique factor of GBM is its relatively low tumor mutational burden (TMB) which reduces the responding T cell clones resulting in poor adaptive immunity [11][12][13]. High TMB often suggested as a reliable biomarker for ICIs [11][12][13]. Other variables, including chemotherapy, corticosteroids, and patient age-related factors may also lead to immunosuppression in GBM patients [10]. Overall, GBM is considered as a highly immunosuppressive CNS-related tumor.

Figure 1. Immunity-related microenvironment of glioblastoma. (1) The immune microenvironment involving glioblastoma (GBM) is characterized by large amounts of CD8+ and CD4+ T cells, M1 and M2 polarized macrophages, microglia, and regulatory T (Treg) cells in addition to a limited number of natural killer (NK) cells. Tumor-associated macrophages and microglia (TAMs) have considerable plasticity toward anti-tumor M1 (inflammatory TAMs) and pro-tumor M2 (anti-inflammatory TAMs) phenotypes. Pharmacological strategies to re-educate tumorigenic M2 TAMs to tumoricidal M1 TAMs may help to relieve immune suppression in the tumor microenvironment (TME), as well as enhance the related anti-tumor activity. (2) GBM normally expressed high levels of immunosuppressive factors, such as programmed cell death 1 ligand 1 (PD-L1) and indolamine 2,3-dioxygenase (IDO), while limiting the presentation of antigens by decreasing major histocompatibility complex (MHC) presentation. The application of IDO inhibitors has effects on Treg cell accumulation. (3) CD47 is highly expressed in various types of tumors. Signal regulatory protein α (SIRPα) is an inhibitory receptor expressed on macrophages and other myeloid immune cells. Upon CD47 binding to SIRPα, src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) and SHP-2 phosphatases are activated to further abrogate phagocytosis via downstream mediators. Disruption of the CD47/SIRPα axis using anti-CD47 antibody (CD47 Ab) can interrupt the inhibitory signaling mediated by SIRPα, thereby promoting phagocytosis of tumor cells. (4) T-cell immunoglobulin and mucin domain-containing protein-3 (TIM3) is a strong negative regulator of lymphocyte function and survival, acting as a marker of CD4+ and CD8+ T-cell exhaustion similarly to programmed cell death 1 (PD-1). It has been verified that the co-expression of PD-1 and TIM3 in lymphocytes is positively correlated with the tumor grade, but it is negatively correlated with progression-free survival (PFS) in different types of tumors including GBM. (5) In the context of microglial cells, these often secrete transforming growth factor β (TGFβ) and/or interleukin 10 (IL-10) to decrease the amount of myeloid and/or lymphoid immune cells, resulting in a systemic immunosuppression and immune evasion of GBM cells. Th, helper T cell; ADCC, antibody-dependent-cell-mediated cytotoxicity; Treg, regulatory T cell; CTL, cytotoxic T lymphocyte; CAR T, chimeric antigen receptor T cell; DC, dendritic cell.

       One distinctive aspect of the brain’s microenvironment is related to the bulk of myeloid cell population, which is capable of manipulating the immune microenvironment and GBM progression by producing immunosuppressive and anti-inflammatory cytokines and growth factors, as well as promoting T-cell apoptosis, thus suggesting a new strategy for immunotherapy [14][15]. A considerable population of brain myeloid cells are microglia, which are equivalent to macrophages from other tissues [16]. In the absence of any inflammatory stimulation, the microglia normally arise from the yolk sac and are maintained by continuous replication during our whole life [17]. Upon pro-inflammatory stimulation in the GBM tissue, microglial cells may undergo significant phenotypic changes, while extensive additional macrophages can also be recruited from peripheral monocytes into the tumor site [18][19][20]. Notably, the GBM microenvironment has a surprisingly high composition of tumor-associated macrophages and microglia (TAMs), ranging between 30 and 50% [21] of tumor mass. Such a percentage of TAMs is much higher than the ones observed in other major malignancies such as melanoma [22]. One notable feature of TAMs is that they have considerable plasticity toward anti-tumor M1 (inflammatory TAMs) and pro-tumor M2 (anti-inflammatory TAMs) phenotypes (Figure 1). Redirecting TAMs from immunoinhibitory M2 to immunostimulatory M1 phenotype is a promising approach to elicit an immune response and to inhibit GBM progression since this can reduce immunosuppressive restrains and thus boost immunity driven by cytotoxic T lymphocytes (CTLs) [23]. More recently, research evidence has indicated that pharmacological inhibition provided by certain soluble factors, such as colony-stimulating factor-1 receptor, can dramatically decrease M2 polarization and significantly improve OS [23]. Moreover, several reports have confirmed a strong association between the survival of high-grade glioma patients and M1 or M2 polarization. For instance, M1 polarization has been positively correlated with improved patient survival [24]. In contrast, M2 polarization (assessed by F11R marker) has been negatively correlated with patient survival [25]. Therefore, strategies to target TAMs have emerged as alternate routes for GBM therapy [26]. In this sense, a number of studies have pursued ways to (i) inhibit monocyte recruitment into the CNS, (ii) deplete M2 TAMs, and (iii) reprogram tumorigenic M2 to M1 phenotype [27]. One recent report has also demonstrated TAM-mediated resistance of programmed cell death 1 (PD-1) immunotherapy, thus providing a strong rationale towards TAM targeting as a reliable approach to enhance PD-1-inhibitor treatment response [28]. Of note, TAM-targeted immunotherapy has received particular attention in recent years although investigations related to this promising therapeutic area are still in progress.

2. Overview of Current Immunotherapy Modalities for Glioblastoma

       As a paradigm shift in cancer treatment, immunotherapy has recently gained enormous attention and also achieved a rapid expansion in the context of GBM. Immunotherapy approaches for GBM have been focused on ICIs, oncolytic viruses, chimeric T-cell receptors, and dendritic cell (DC) vaccines [4][9]Figure 2 outlines four distinct immunotherapy modalities available for GBM. We can notice that a successful vaccine for GBM treatment depends on DC-mediated presentation of GBM-related antigens as well as peptides for T-cell activation in the adaptive immune system. Among the pathways involved in these processes, one is related to the combination of T-cell receptors and MHC, while another pathway involves the interaction between CD80/CD86 and CD28. Cytotoxic T lymphocytes (CTLs) can be subsequently activated to kill GBM cells having specific antigens for MHC I presentation [4][29]. In general, tumor cells avoid this disruption by upregulating PD-L1, which binds to its complementary receptor, PD-1 along the T-cell surface to further inhibit the activation of CTLs [30]. We can utilize different approaches of immune-checkpoint blockage to effectively prevent the interaction between PD-L1 and PD-1 in GBM. However, a phase III trial result to compare therapeutic efficacy of nivolumab and bevacizumab in recurrent GBM was disappointing with no improvement in OS (Clinical trial identifier: NCT02017717, Table 1). Cytotoxic T lymphocyte protein 4 (CTLA-4) is another important immune regulatory molecule that binds to CD80 or CD86 and inhibits their combination with CD28 to prevent T-cell activation [30]. Epidermal growth factor receptor variant III (EGFRvIII), IL-13 receptor subunit-α2 (IL-13R α2), and human epidemic growth factor receptor 2 (HER2) are expressed on the surface of GBM cells and may also be targeted by a genetically modified chimeric antigen receptor (CAR) T cell to promote GBM cell death [4][9][29]. Given the promising role of cancer immunotherapies towards GBM pathophysiology, a substantial number of clinical trials have been performed or planned to explore the potential roles and efficacy of targeting these three antigens [4][9][29] (Table 1). These clinical trials have demonstrated feasibility, safety, and efficacy of CAR T cell therapy for GBM. For example, treatment constituted of virus-specific T cells (VSTs) expressing HER2-specific CAR (HER2-CAR VST)in progressive GBM patients resulted in an median OS of up to 11.1 months from the first T cell infusion and 24.5 months from the first diagnosis (Clinical trial identifier: NCT01109095, Table 1).Interestingly, genetic engineering has also been applied in oncolytic viral treatment to produce viruses that may infect tumor cells, trigger tumor cell lysis, and hijack tumor cell replication, which ultimately leads to tumor cell death [4][9] (Table 1). This particular treatment has enabled the breakage of shackles from many tumors and also triggered a higher immune backlash, thus shifting GBM from cold to hot tumor types [4][9]. Promisingly, data from phase II trial have verified the high clinical response in GBM patients after intratumoral inoculation of Polio/Rhinovirus Recombinant (PVSRIPO), with an increase in OS up to 12.5 months from the time of inoculation and higher survival rate at 24 and 36 months over historical controls (Clinical trial identifier: NCT01491893, Table 1). These results show that oncolytic-based therapy has a high potential to improve OS and quality of life for patients affected by GBM [29]. Although oncolytic-based therapies may provide significant immunostimulatory effects, including the depletion of regulatory T cells, the induction of immunogenic cell death, and abscopal effects, these therapeutic approaches still carry some intrinsic limitations. For instance, pro-inflammatory responses caused by oncolytic viruses may potentially limit the application of oncolytic viruses as a single-modality immunotherapy [29]. Besides, CAR T-cell treatment for GBM relies on the identification of stably expressed and sufficient tumor-related antigens, which might eventually limit the clinical application of this therapy [9][29]. Considering the highly heterogeneous characteristics of GBM, one could postulate that targeting one antigen in GBM might not be sufficient to eradicate all the GBM cells. Overall, these obstacles have promoted the development of alternate immunotherapy modalities, which may better recapitulate tumor immunology with improved accuracy. 

Figure 2. Current immunotherapy strategies for glioblastoma. (1) Vaccines for glioblastoma (GBM) treatment have been relied on dendritic cell (DC)-mediated presentation of GBM-related antigens and peptides for T-cell activation in the adaptive immune system. (2) The immunosuppression status of cytotoxic T lymphocytes (CTLs) can be relieved by the application of immune-checkpoint inhibitors (ICIs), including anti-programmed cell death protein 1 (PD-1), anti-cytotoxic T lymphocyte protein 4 (CTLA-4) and anti-programmed cell death 1 ligand 1 (PD-L1). (3) Genetically engineered chimeric antigen receptor (CAR) T cells can generate artificial T-cell receptors with high affinity to cancer-specific antigens. (4) Genetic engineering has also been applied in oncolytic viral treatment to medicate cancer cell lysis and promote tumor necrosis. MHC, Major histocompatibility complex; TCR, T-cell receptor; EGFRvIII, Epidermal growth factor receptor variant III.

Table 1. Recent clinical studies with immune-checkpoint inhibitors and some combinational therapies targeting glioblastoma.

References

  1. Billingham, R.E.; Brent, L.; Medawar, P.B. 'Actively acquired tolerance' of foreign cells. Nature 1953, 172, 603–606, doi:10.1038/172603a0.
  2. Billingham, R.E.; Brent, L.; Medawar, P.B.; Sparrow, E.M. Quantitative studies on tissue transplantation immunity. I. The survival times of skin homografts exchanged between members of different inbred strains of mice. Proc. R. Soc. Lond. B Biol. Sci. 1954, 143, 43–58, doi:10.1098/rspb.1954.0053.
  3. Medawar, P.B. Immunity to homologous grafted skin. III. The fate of skin homographs transplanted to the brain, to subcutaneous tissue, and to the anterior chamber of the eye. Br. J. Exp. Pathol. 1948, 29, 58–69.
  4. Lim, M.; Xia, Y.X.; Bettegowda, C.; Weller, M. Current state of immunotherapy for glioblastoma. Nat. Rev. Clin. Oncol. 2018, 15, 422–442, doi:10.1038/s41571-018-0003-5.
  5. Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.S.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341, doi:10.1038/nature14432.
  6. Kipnis, J. Multifaceted Interactions Between Adaptive Immunity and the Central Nervous System. Science 2016, 353, 766–771, doi: 10.1126/science.aag2638.
  7. Bloch, O.; Crane, C.A.; Kaur, R.; Safaee, M.; Rutkowski, M.J.; Parsa, A.T. Gliomas promote immunosuppression through induction of B7-H1 expression in tumor-associated macrophages. Clin. Cancer Res. 2013, 19, 3165–3175, doi:10.1158/1078-0432.CCR-12-3314.
  8. Wainwright, D.A.; Chang, A.L.; Dey, M.; Balyasnikova, I.V.; Kim, C.K.; Tobias, A.; Cheng, Y.; Kim, J.W.; Qiao, J.; Zhang, L.; et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4 and PD-L1 in mice with brain tumors. Clin. Cancer Res. 2014, 20, 5290–5301, doi:10.1158/1078-0432.CCR-14-0514.
  9. McGranahan, T.; Li, G.; Nagpal, S. History and current state of immunotherapy in glioma and brain metastasis. Ther. Adv. Med. Oncol. 2017, 9, 347–368, doi:10.1177/1758834017693750.
  10. Sampson, J.H.; Maus, M.V.; June, C.H. Immunotherapy for brain tumors. J. Clin. Oncol. 2017, 35, 2450–2456, doi:10.1200/JCO.2017.72.8089.
  11. Hodges, T.R.; Ott, M.; Xiu, J.; Gatalica, Z.; Swensen, J.; Zhou, S.; Huse, J.T.; de Groot, J.; Li, S.L.; 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, doi:10.1093/neuonc/nox026.
  12. Bouffet, E.; Larouche, V.; Campbell, B.B.; Merico, D.; de Borja, R.; Aronson, M.; Durno, C.; Krueger, J.; Cabric, V.; Ramaswamy, V.; et al. Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J. Clin. Oncol. 2016, 34, 2206–2211, doi:10.1200/JCO.2016.66.6552.
  13. Johanns, T.M.; Miller, C.A.; Dorward, I.G.; Tsien, C.; Chang, E.; Perry, A.; Uppaluri, R.; Ferguson, C.; Schmidt, R.E.; Dahiya, S.; et al. Immunogenomics of hypermutated glioblastoma: A patient with germline POLE deficiency treated with checkpoint blockade immunotherapy. Cancer Discov. 2016, 6, 1230–1236, doi:10.1158/2159-8290.CD-16-0575.
  14. Feng, M.; Jiang, W.; Kim, B.Y.S.; Zhang, C.C.; Fu, Y.X.; Weissman, I.L. Phagocytosis Checkpoints as New Targets for Cancer Immunotherapy. Nat Rev Cancer. 2019, 19, 568-586, doi: 10.1038/s41568-019-0183-z.
  15. Zhao, P.F.; Wang, Y.H.; Kang, X.J.; Wu, A.H.; Yin, W.M.; Tang, Y.S.; Wang, J.Y.; Zhang, M.; Duan, Y.F.; Huang, Y.Z. Dual-targeting biomimetic delivery for anti-glioma activity via remodeling the tumor microenvironment and directing macrophage-mediated immunotherapy. Chem. Sci. 2018, 9, 2674–2689, doi:10.1039/c7sc04853j.
  16. Li, Q.Y.; Barres, B.A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 2018, 18, 225–242, doi:10.1038/nri.2017.125.
  17. Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845, doi:10.1126/science.1194637.
  18. Ajami, B.; Bennett, J.L.; Krieger, C.; McNagny, K.M.; Rossi, F.M.V. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat. Neurosci. 2011, 14, 1142–1149, doi:10.1038/nn.2887.
  19. Bennett, F.C.; Bennett, M.L.; Yaqoob, F.; Mulinyawe, S.B.; Grant, G.A.; Hayden Gephart, M.; Plowey, E.D.; Barres, B.A. A combination of ontogeny and CNS environment establishes microglial identity. Neuron 2018, 98, 1170–1183, doi:10.1016/j.neuron.2018.05.014.
  20. Graeber, M.B.; Scheithauer, B.W.; Kreutzberg, G.W. Microglia in brain tumors. Glia 2002, 40, 252–259, doi:10.1002/glia.10147.
  21. Matias, D.; Balça-Silva, J.; Da Graça, G.C.; Wanjiru, C.M.; Macharia, L.W.; Nascimento, C.P.; Roque, N.R.; Coelho-Aguiar, J.M.; Pereira, C.M.; Dos Santos, M.F.; et al. Microglia/Astrocytes–glioblastoma crosstalk: Crucial molecular mechanisms and microenvironmental factors. Front. Cell. Neurosci. 2018, 12, 235, doi:10.3389/fncel.2018.00235.
  22. Hussein, M.R. Tumour-associated macrophages and melanoma tumourigenesis: Integrating the complexity. Int. J. Exp. Pathol. 2006, 87, 163–176, doi:10.1111/j.1365-2613.2006.00478.x.
  23. 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, doi:10.1038/nm.3337.
  24. Zeiner, P.S.; Preusse, C.; Blank, A.E.; Zachskorn, C.; Baumgarten, P.; Caspary, L.; Braczynski, A.K.; Weissenberger, J.; Bratzke, H.; Reiß, S.; et al. MIF receptor CD74 is restricted to Microglia/Macrophages, associated with a m1‐polarized immune milieu and prolonged patient survival in gliomas. Brain Pathol. 2015, 25, 491–504, doi:10.1111/bpa.12194.
  25. Pong, W.W.; Walker, J.; Wylie, T.; Magrini, V.; Luo, J.; Emnett, R.J.; Choi, J.; Cooper, M.L.; Griffith, M.; Griffith, O.L.; et al. F11R is a novel monocyte prognostic biomarker for malignant glioma. PLoS ONE 2013, 8, e77571, doi:10.1371/journal.pone.0077571.
  26. Hambardzumyan, D.; Gutmann, D.H.; Kettenmann, H. The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci. 2016, 19, 20–27, doi:10.1038/nn.4185.
  27. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Onco.l 2017, 14, 399–416, doi:10.1038/nrclinonc.2016.217.
  28. Arlauckas, S.P.; Garris, C.S.; Kohler, R.H.; Kitaoka, M.; Cuccarese, M.F.; Yang, K.S.; Miller, M.A.; Carlson, J.C.; Freeman, G.J.; Anthony, R.M.; et al. In vivo imaging reveals a tumor-associated macrophage-mediated resistance pathway in anti-PD-1 therapy. Sci. Transl. Med. 2017, 9, doi:10.1126/scitranslmed.aal3604
  29. Tarhini, A.A.; Iqbal, F. CTLA-4 blockade: Therapeutic potential in cancer treatments. Oncotargets Ther. 2010, 3, 15–25, doi:10.2147/ott.s4833
  30. Wang, H.X.; Xu, T.; Huang, Q.L.; Jin, W.L.; Chen, J.X. Immunotherapy for malignant glioma: Current status and future directions. Trends Pharmacol. Sci. 2020, 41, 123–138, doi:10.1016/j.tips.2019.12.003.
  31. ClinicalTrials.gov. A Study of the Effectiveness and Safety of Nivolumab Compared to Bevacizumab and of Nivolumab with or without Ipilimumab in Glioblastoma Patients (CheckMate 143). 2013. Available online: https://clinicaltrials.gov/show/NCT02017717 (accessed on 6 June 2020).
  32. ClinicalTrials.gov. Neoadjuvant Nivolumab in Glioblastoma (Neo-nivo). 2015. Available online: https://clinicaltrials.gov/show/NCT02550249 (accessed on 6 June 2020).
  33. ClinicalTrials.gov. Nivolumab with Radiation Therapy and Bevacizumab for Recurrent MGMT Methylated Glioblastoma. 2018. Available online: https://clinicaltrials.gov/show/NCT03743662 (accessed on 6 June 2020).
  34. Reiss, S.N.; Yerram, P.; Modelevsky, L.; Grommes, C. Retrospective review of safety and efficacy of programmed cell death-1 inhibitors in refractory high grade gliomas. J. Immunother. Cancer 2017, 5, 99, doi:10.1186/s40425-017-0302-x.
  35. ClinicalTrials.gov. Nivolumab and Temozolomide Versus Temozolomide alone in Newly Diagnosed Elderly Patients with GBM (NUTMEG). 2018. Available online: https://clinicaltrials.gov/show/NCT04195139 (accessed on 6 June 2020).
  36. ClinicalTrials.gov. IL13Ralpha2-Targeted Chimeric Antigen Receptor (CAR) t Cells with or without Nivolumab and Ipilimumab in Treating Patients with Recurrent or Refractory Glioblastoma. 2019. Available online: https://clinicaltrials.gov/show/NCT04003649 (accessed on 6 June 2020).
  37. ClinicalTrials.gov. Intra-tumoral Ipilimumab Plus Intravenous Nivolumab Following the Resection of Recurrent Glioblastoma (GlitIpNi). 2016. Available online: https://clinicaltrials.gov/show/NCT03233152 (accessed on 6 June 2020).
  38. ClinicalTrials.gov. A Study Testing the Effect of Immunotherapy (Ipilimumab and Nivolumab) in Patients with Recurrent Glioblastoma with Elevated Mutational Burden. 2020. Available online: https://clinicaltrials.gov/show/NCT04145115 (accessed on 6 June 2020).
  39. ClinicalTrials.gov. Nivolumab with DC Vaccines for Recurrent Brain Tumors (AVERT). 2016. Available online: https://clinicaltrials.gov/show/NCT02529072 (accessed on 6 June 2020).
  40. ClinicalTrials.gov. Combination Adenovirus + Pembrolizumab to Trigger Immune Virus Effects (CAPTIVE). 2016. Available online: https://clinicaltrials.gov/show/NCT02798406 (accessed on 6 June 2020).
  41. ClinicalTrials.gov. Trial of Anti-Tim-3 in Combination with Anti-PD-1 and SRS in Recurrent GBM. 2020. Available online: https://clinicaltrials.gov/show/NCT03961971 (accessed on 6 June 2020).
  42. ClinicalTrials.gov. Nivolumab, BMS-986205, and Radiation Therapy with or without Temozolomide in Treating Patients with Newly Diagnosed Glioblastoma. 2019. Available online: https://clinicaltrials.gov/show/NCT04047706 (accessed on 6 June 2020).
  43. ClinicalTrials.gov. Study of the IDO Pathway Inhibitor, Indoximod, and Temozolomide for Pediatric Patients with Progressive Primary Malignant Brain Tumors. 2015. Available online: https://clinicaltrials.gov/show/NCT02502708 (accessed on 6 June 2020).
  44. ClinicalTrials.gov. Safety and Efficacy of the ONCOlytic VIRus Armed for Local Chemotherapy, TG6002/5-FC, in Recurrent Glioblastoma Patients (ONCOVIRAC). 2017. Available online: https://clinicaltrials.gov/show/NCT03294486 (accessed on 6 June 2020).
  45. ClinicalTrials.gov. Safety and Effectiveness Study of g207, a Tumor-Killing Virus, in Patients with Recurrent Brain Cancer. 2017. Available online: https://clinicaltrials.gov/show/NCT00028158 (accessed on 6 June 2020).
  46. ClinicalTrials.gov. DNX-2401 with Interferon Gamma (IFN-γ) for Recurrent Glioblastoma or Gliosarcoma Brain Tumors (TARGET-I). 2014. Available online: https://clinicaltrials.gov/show/NCT02197169 (accessed on 6 June 2020).
  47. ClinicalTrials.gov. PVSRIPO for Recurrent Glioblastoma (GBM) (PVSRIPO). 2012. Available online: https://clinicaltrials.gov/show/NCT01491893 (accessed on 6 June 2020).
  48. Desjardins, A.; Gromeier, M.; Herndon, J.E. 2nd.; Beaubier, N.; Bolognesi, D.P.; Friedman, A.H.; Friedman, H.S.; McSherry, F.; Muscat, A.M.; Nair, S.; et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N. Engl. J. Med. 2018, 379, 150–161. doi: 10.1056/NEJMoa1716435.
  49. ClinicalTrials.gov. Pembrolizumab and a Vaccine (ATL-DC) for the Treatment of Surgically Accessible Recurrent Glioblastoma. 2020. Available online: https://clinicaltrials.gov/show/NCT04201873 (accessed on 6 June 2020).
  50. ClinicalTrials.gov. DC Migration Study for Newly-Diagnosed GBM (ELEVATE). 2015. Available online: https://clinicaltrials.gov/show/NCT02366728 (accessed on 6 June 2020).
  51. ClinicalTrials.gov. Pilot Study of Autologous Anti-EGFRvIII CAR t cells in Recurrent Glioblastoma Multiforme. 2016. Available online: https://clinicaltrials.gov/show/NCT02844062 (accessed on 6 June 2020).
  52. ClinicalTrials.gov. Cellular Adoptive Immunotherapy Using Genetically Modified T-Lymphocytes in Treating Patients with Recurrent or Refractory High-Grade Malignant Glioma. 2002. Available online: https://clinicaltrials.gov/show/NCT00730613 (accessed on 6 June 2020).
  53. ClinicalTrials.gov. CMV-Specific Cytotoxic t Lymphocytes Expressing CAR Targeting HER2 in Patients with GBM (HERT-GBM). 2010. Available online: https://clinicaltrials.gov/show/NCT01109095 (accessed on 6 June 2020).
  54. Koyama, S.; Akbay, E.A.; Li, Y.Y.; Herter-Sprie, G.S.; Buczkowski, K.A.; Richards, W.G.; Gandhi, L.; Redig, A.J.; Rodig, S.J.; Asahina, H.; et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 2016, 7, e10501, doi:10.1038/ncomms10501.
  55. Kim, J.E.; Patel, M.A.; Mangraviti, A.; Kim, E.S.; Theodros, D.; Velarde, E.; Liu, A.; Sankey, E.W.; Tam, A.; Xu, H.Y.; et al. Combination therapy with Anti-PD-1, Anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin. Cancer Res. 2017, 23, 124–136, doi:10.1158/1078-0432.CCR-15-1535.
  56. Sun, F.; Guo, Z.S.; Gregory, A.D.; Shapiro, S.D.; Xiao, G.; Qu, Z. Dual but not single PD-1 or TIM-3 blockade enhances oncolytic virotherapy in refractory lung cancer. J Immunother. Cancer. 2019, 8, e000294, doi:10.1136/jitc-2019-000294.
  57. Patel, M.A.; Kim, J.E.; Theodros, D.; Tam, A.; Velarde, E.; Kochel, C.M.; Francica, B.; Nirschl, T.R.; Ghasemzadeh, A.; Mathios, D.; et al. Agonist anti-GITR monoclonal antibody and stereotactic radiation induce immune-mediated survival advantage in murine intracranial glioma. J. Immunother. Cancer 2016, 4, e28, doi:10.1186/s40425-016-0132-2.
  58. Washington University School of Medicine; Sharp, M.; Corp, D. MK-3475 in Combination with MRI-Guided Laser Ablation in Recurrent Malignant Gliomas. Available online: https://clinicaltrials.gov/ct2/show/NCT02311582 (accessed on 15 July 2020).
  59. Pardridge, W.M. Blood–brain barrier delivery. Drug Discov. Today 2007, 12, 54–61, doi:10.1016/j.drudis.2006.10.013.
  60. Weiss, N.; Miller, F.; Cazaubon, S.; Couraud, P. The blood-brain barrier in brain homeostasis and neurological diseases. Biochim. et Biophys. Acta (BBA)-Biomembr. 2009, 1788, 842–857, doi:10.1016/j.bbamem.2008.10.022.
  61. Da Ros, M.; De Gregorio, V.; Iorio, A.L.; Giunti, L.; Guidi, M.; De Martino, M.; Genitori, L.; Sardi, I. Glioblastoma chemoresistance: The double play by microenvironment and blood-brain barrier. Int. J. Mol. Sci. 2018, 19, e2879, doi:10.3390/ijms19102879.
  62. Liu, Y.Y.; Ran, R.; Chen, J.T.; Kuang, Q.F.; Tang, J.; Mei, L.; Zhang, Q.Y.; Gao, H.L.; Zhang, Z.R.; He, Q. Paclitaxel loaded liposomes decorated with a multifunctional tandem peptide for glioma targeting. Biomaterials 2014, 35, 4835–4847, doi:10.1016/j.biomaterials.2014.02.031.
  63. Shi, J.L.; Hou, S.Q.; Huang, J.F.; Wang, S.S.; Huan, W.; Huang, C.J.; Liu, X.J.; Jiang, R.; Qian, W.B.; Lu, J.J.; et al. An MSN-PEG-IP drug delivery system and IL13Rα\alphaα2 as targeted therapy for glioma. Nanoscale 2017, 9, 8970–8981, doi:10.1039/c6nr08786h.
  64. Strauss, L.; Mahmoud, M.A.A.; Weaver, J.D.; Tijaro-Ovalle, N.M.; Christofides, A.; Wang, Q.; Pal, R.; Yuan, M.; Asara, J.; Patsoukis, N.; et al. Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci. Immunol. 2020, 5, doi:10.1126/sciimmunol.aay1863.
  65. Goswami, S.; Walle, T.; Cornish, A.E.; Basu, S.; Anandhan, S.; Fernandez, I.; Vence, L.; Blando, J.; Zhao, H.; Yadav, S.S.; et al. Immune profiling of human tumors identifies CD73 as a combinatorial target in glioblastoma. Nat. Med. 2020, 26, 39–46, doi:10.1038/s41591-019-0694-x
  66. Goswami, S.; Walle, T.; Cornish, A.E.; Basu, S.; Anandhan, S.; Fernandez, I.; Vence, L.; Blando, J.; Zhao, H.; Yadav, S.S.; et al. Immune profiling of human tumors identifies CD73 as a combinatorial target in glioblastoma. Nat. Med. 2020, 26, 39–46, doi:10.1038/s41591-019-0694-x
  67. Filley, A.C.; Henriquez, M.; Dey, M. Recurrent glioma clinical trial, CheckMate-143: The game is not over yet. Oncotarget 2017, 8, e91779, doi:10.18632/oncotarget.21586.
  68. Cloughesy, T.F.; Mochizuki, A.Y.; Orpilla, J.R.; Hugo, W.; Lee, A.H.; Davidson, T.B.; Wang, A.C.; Ellingson, B.M.; Rytlewski, J.A.; Sanders, C.M.; et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 2019, 25, 477–486, doi:10.1038/s41591-018-0337-7.
  69. Forde, P.M.; Chaft, J.E.; Smith, K.N.; Anagnostou, V.; Cottrell, T.R.; Hellmann, M.D.; Zahurak, M.; Yang, S.C.; Jones, D.R.; Broderick, S.; et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N. Engl. J. Med. 2018, 378, 1976–1986, doi:10.1056/NEJMoa1716078.
  70. Schalper, K.A.; Rodriguez-Ruiz, M.E.; Diez-Valle, R.; López-Janeiro, A.; Porciuncula, A.; Idoate, M.A.; Inogés, S.; de Andrea, C.; López-Diaz De Cerio, A.; Tejada, S.; et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat. Med. 2019, 25, 470–476, doi:10.1038/s41591-018-0339-5.
  71. Schalper, K.A.; Rodriguez-Ruiz, M.E.; Diez-Valle, R.; López-Janeiro, A.; Porciuncula, A.; Idoate, M.A.; Inogés, S.; de Andrea, C.; López-Diaz De Cerio, A.; Tejada, S.; et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat. Med. 2019, 25, 470–476, doi:10.1038/s41591-018-0339-5.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 482
Revisions: 2 times (View History)
Update Date: 25 Aug 2020
1000/1000