The CNS and the Brain Tumor Microenvironment: Implications for Glioblastoma Immunotherapy
Abstract
1. Introduction
2. Immune Activation and CNS Privilege
3. CNS Lymphatics
4. Immune and Stromal Landscape in GBM
4.1. Blood–Brain Barrier
4.2. Glia and Neurons
4.3. Resident Microglia and Monocyte-Derived Macrophages
5. Preclinical Models of GBM
6. Enhancing the Adaptive Immune Response in GBM
6.1. Oncolytic Viral Therapy
6.2. DC Vaccination
6.3. Checkpoint Inhibition
6.4. CAR-T Cell Therapy
6.5. BiTEs
6.6. CNS Lymphatic System Modulation
7. Conclusions and Perspectives
Author Contributions
Funding
Conflicts of Interest
References
- Hegi, M.E.; Diserens, A.C.; Gorlia, T.; Hamou, M.F.; de Tribolet, N.; Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L.; et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 2005, 352, 997–1003. [Google Scholar] [CrossRef] [PubMed]
- Friedmann-Morvinski, D. Glioblastoma heterogeneity and cancer cell plasticity. Crit. Rev. Oncog. 2014, 19, 327–336. [Google Scholar] [CrossRef] [PubMed]
- Parker, N.R.; Hudson, A.L.; Khong, P.; Parkinson, J.F.; Dwight, T.; Ikin, R.J.; Zhu, Y.; Cheng, Z.J.; Vafaee, F.; Chen, J.; et al. Intratumoral heterogeneity identified at the epigenetic, genetic and transcriptional level in glioblastoma. Sci. Rep. 2016, 6, 22477. [Google Scholar] [CrossRef] [PubMed]
- Patel, A.P.; Tirosh, I.; Trombetta, J.J.; Shalek, A.K.; Gillespie, S.M.; Wakimoto, H.; Cahill, D.P.; Nahed, B.V.; Curry, W.T.; Martuza, R.L.; et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 2014, 344, 1396–1401. [Google Scholar] [CrossRef]
- Ohgaki, H.; Kleihues, P. The definition of primary and secondary glioblastoma. Clin. Cancer Res. 2013, 19, 764–772. [Google Scholar] [CrossRef]
- Nobusawa, S.; Watanabe, T.; Kleihues, P.; Ohgaki, H. IDH1 mutations as molecular signature and predictive factor of secondary glioblastomas. Clin. Cancer Res. 2009, 15, 6002–6007. [Google Scholar] [CrossRef]
- Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef]
- Chen, Z.; Hambardzumyan, D. Immune Microenvironment in Glioblastoma Subtypes. Front. Immunol. 2018, 9, 1004. [Google Scholar] [CrossRef]
- Park, A.K.; Kim, P.; Ballester, L.Y.; Esquenazi, Y.; Zhao, Z. Subtype-specific signaling pathways and genomic aberrations associated with prognosis of glioblastoma. Neuro-Oncology 2019, 21, 59–70. [Google Scholar] [CrossRef]
- Guan, X.; Vengoechea, J.; Zheng, S.; Sloan, A.E.; Chen, Y.; Brat, D.J.; O’Neill, B.P.; de Groot, J.; Yust-Katz, S.; Yung, W.K.; et al. Molecular subtypes of glioblastoma are relevant to lower grade glioma. PLoS ONE 2014, 9, e91216. [Google Scholar] [CrossRef]
- Verhaak, R.G.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef]
- Clark, O.; Yen, K.; Mellinghoff, I.K. Molecular Pathways: Isocitrate Dehydrogenase Mutations in Cancer. Clin. Cancer Res. 2016, 22, 1837–1842. [Google Scholar] [CrossRef]
- Aldape, K.; Zadeh, G.; Mansouri, S.; Reifenberger, G.; von Deimling, A. Glioblastoma: Pathology, molecular mechanisms and markers. Acta Neuropathol. 2015, 129, 829–848. [Google Scholar] [CrossRef]
- Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; Taphoorn, M.J.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef]
- Touat, M.; Idbaih, A.; Sanson, M.; Ligon, K.L. Glioblastoma targeted therapy: Updated approaches from recent biological insights. Ann. Oncol. 2017, 28, 1457–1472. [Google Scholar] [CrossRef] [PubMed]
- Heye, A.K.; Culling, R.D.; Valdes Hernandez Mdel, C.; Thrippleton, M.J.; Wardlaw, J.M. Assessment of blood-brain barrier disruption using dynamic contrast-enhanced MRI. A systematic review. Neuroimage Clin. 2014, 6, 262–274. [Google Scholar] [CrossRef] [PubMed]
- Fecci, P.E.; Mitchell, D.A.; Whitesides, J.F.; Xie, W.; Friedman, A.H.; Archer, G.E.; Herndon, J.E., 2nd; Bigner, D.D.; Dranoff, G.; Sampson, J.H. Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res. 2006, 66, 3294–3302. [Google Scholar] [CrossRef] [PubMed]
- Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Medawar, P.B. Immunity to homologous grafted skin; the fate of skin homografts transplanted to the brain, to subcutaneous tissue, and to the anterior chamber of the eye. Br. J. Exp. Pathol. 1948, 29, 58–69. [Google Scholar]
- Shirai, Y. On the transplantation of the rat sarcoma in adult heterogeneous animals. Jpn. Med. World 1921, 1, 14–15. [Google Scholar]
- Padera, T.P.; Meijer, E.F.; Munn, L.L. The Lymphatic System in Disease Processes and Cancer Progression. Annu. Rev. Biomed. Eng. 2016, 18, 125–158. [Google Scholar] [CrossRef] [PubMed]
- Kida, S.; Pantazis, A.; Weller, R.O. CSF drains directly from the subarachnoid space into nasal lymphatics in the rat. Anatomy, histology and immunological significance. Neuropathol. Appl. Neurobiol. 1993, 19, 480–488. [Google Scholar] [CrossRef] [PubMed]
- Cserr, H.F.; Cooper, D.N.; Milhorat, T.H. Flow of cerebral interstitial fluid as indicated by the removal of extracellular markers from rat caudate nucleus. Exp. Eye Res. 1977, 25 (Suppl. S1), 461–473. [Google Scholar] [CrossRef]
- Iliff, J.J.; Wang, M.; Liao, Y.; Plogg, B.A.; Peng, W.; Gundersen, G.A.; Benveniste, H.; Vates, G.E.; Deane, R.; Goldman, S.A.; et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci. Transl. Med. 2012, 4, 147ra111. [Google Scholar] [CrossRef] [PubMed]
- Koh, L.; Zakharov, A.; Johnston, M. Integration of the subarachnoid space and lymphatics: Is it time to embrace a new concept of cerebrospinal fluid absorption? Cerebrospinal Fluid Res. 2005, 2, 6. [Google Scholar] [CrossRef] [PubMed]
- Andres, K.H.; von During, M.; Muszynski, K.; Schmidt, R.F. Nerve fibres and their terminals of the dura mater encephali of the rat. Anat. Embryol. 1987, 175, 289–301. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef]
- Aspelund, A.; Antila, S.; Proulx, S.T.; Karlsen, T.V.; Karaman, S.; Detmar, M.; Wiig, H.; Alitalo, K. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 2015, 212, 991–999. [Google Scholar] [CrossRef]
- Ahn, J.H.; Cho, H.; Kim, J.H.; Kim, S.H.; Ham, J.S.; Park, I.; Suh, S.H.; Hong, S.P.; Song, J.H.; Hong, Y.K.; et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature 2019, 572, 62–66. [Google Scholar] [CrossRef]
- Harling-Berg, C.J.; Knopf, P.M.; Cserr, H.F. Myelin basic protein infused into cerebrospinal fluid suppresses experimental autoimmune encephalomyelitis. J. Neuroimmunol. 1991, 35, 45–51. [Google Scholar] [CrossRef]
- Phillips, M.J.; Needham, M.; Weller, R.O. Role of cervical lymph nodes in autoimmune encephalomyelitis in the Lewis rat. J. Pathol. 1997, 182, 457–464. [Google Scholar] [CrossRef]
- Van Zwam, M.; Huizinga, R.; Heijmans, N.; van Meurs, M.; Wierenga-Wolf, A.F.; Melief, M.J.; Hintzen, R.Q.; t Hart, B.A.; Amor, S.; Boven, L.A.; et al. Surgical excision of CNS-draining lymph nodes reduces relapse severity in chronic-relapsing experimental autoimmune encephalomyelitis. J. Pathol. 2009, 217, 543–551. [Google Scholar] [CrossRef] [PubMed]
- Murphy, J.B.; Sturm, E. Conditions Determining the Transplantability of Tissues in the Brain. J. Exp. Med. 1923, 38, 183–197. [Google Scholar] [CrossRef] [PubMed]
- Mason, D.W.; Charlton, H.M.; Jones, A.J.; Lavy, C.B.; Puklavec, M.; Simmonds, S.J. The fate of allogeneic and xenogeneic neuronal tissue transplanted into the third ventricle of rodents. Neuroscience 1986, 19, 685–694. [Google Scholar] [CrossRef]
- Nicholas, M.K.; Antel, J.P.; Stefansson, K.; Arnason, B.G. Rejection of fetal neocortical neural transplants by H-2 incompatible mice. J. Immunol. 1987, 139, 2275–2283. [Google Scholar]
- Galea, I.; Bechmann, I.; Perry, V.H. What is immune privilege (not)? Trends Immunol. 2007, 28, 12–18. [Google Scholar] [CrossRef]
- Bechmann, I.; Galea, I.; Perry, V.H. What is the blood–brain barrier (not)? Trends Immunol. 2007, 28, 5–11. [Google Scholar] [CrossRef]
- Pachter, J.S.; de Vries, H.E.; Fabry, Z. The blood-brain barrier and its role in immune privilege in the central nervous system. J. Neuropathol. Exp. Neurol. 2003, 62, 593–604. [Google Scholar] [CrossRef]
- Daneman, R.; Prat, A. The blood-brain barrier. Cold Spring Harb. Perspect. Biol. 2015, 7, a020412. [Google Scholar] [CrossRef]
- Daneman, R.; Zhou, L.; Agalliu, D.; Cahoy, J.D.; Kaushal, A.; Barres, B.A. The mouse blood-brain barrier transcriptome: A new resource for understanding the development and function of brain endothelial cells. PLoS ONE 2010, 5, e13741. [Google Scholar] [CrossRef]
- Omari, K.I.; Dorovini-Zis, K. Expression and function of the costimulatory molecules B7-1 (CD80) and B7-2 (CD86) in an in vitro model of the human blood—Brain barrier. J. Neuroimmunol. 2001, 113, 129–141. [Google Scholar] [CrossRef]
- Wheway, J.; Obeid, S.; Couraud, P.O.; Combes, V.; Grau, G.E. The brain microvascular endothelium supports T cell proliferation and has potential for alloantigen presentation. PLoS ONE 2013, 8, e52586. [Google Scholar] [CrossRef]
- Zhang, Y.; Chen, K.; Sloan, S.A.; Bennett, M.L.; Scholze, A.R.; O’Keeffe, S.; Phatnani, H.P.; Guarnieri, P.; Caneda, C.; Ruderisch, N.; et al. An RNA-Sequencing Transcriptome and Splicing Database of Glia, Neurons, and Vascular Cells of the Cerebral Cortex. J. Neurosci. 2014, 34, 11929–11947. [Google Scholar] [CrossRef]
- Zhang, Y.; Sloan, S.A.; Clarke, L.E.; Caneda, C.; Plaza, C.A.; Blumenthal, P.D.; Vogel, H.; Steinberg, G.K.; Edwards, M.S.B.; Li, G.; et al. Purification and Characterization of Progenitor and Mature Human Astrocytes Reveals Transcriptional and Functional Differences with Mouse. Neuron 2016, 89, 37–53. [Google Scholar] [CrossRef] [PubMed]
- Pober, J.S.; Gimbrone, M.A.; Cotran, R.S.; Reiss, C.S.; Burakoff, S.J.; Fiers, W.; Ault, K.A. Ia expression by vascular endothelium is inducible by activated T cells and by human gamma interferon. J. Exp. Med. 1983, 157, 1339–1353. [Google Scholar] [CrossRef] [PubMed]
- Hughes, C.C.; Savage, C.O.; Pober, J.S. Endothelial cells augment T cell interleukin 2 production by a contact-dependent mechanism involving CD2/LFA-3 interaction. J. Exp. Med. 1990, 171, 1453–1467. [Google Scholar] [CrossRef] [PubMed]
- Epperson, D.E.; Pober, J.S. Antigen-presenting function of human endothelial cells. Direct activation of resting CD8 T cells. J. Immunol. 1994, 153, 5402–5412. [Google Scholar]
- Hickey, W.F.; Hsu, B.L.; Kimura, H. T-lymphocyte entry into the central nervous system. J. Neurosci. Res. 1991, 28, 254–260. [Google Scholar] [CrossRef]
- Harris, M.G.; Hulseberg, P.; Ling, C.; Karman, J.; Clarkson, B.D.; Harding, J.S.; Zhang, M.; Sandor, A.; Christensen, K.; Nagy, A.; et al. Immune privilege of the CNS is not the consequence of limited antigen sampling. Sci. Rep. 2014, 4, 4422. [Google Scholar] [CrossRef]
- Lyck, R.; Engelhardt, B. Going against the tide--how encephalitogenic T cells breach the blood-brain barrier. J. Vasc. Res. 2012, 49, 497–509. [Google Scholar] [CrossRef]
- Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef] [PubMed]
- Alexander, J.J. Blood-brain barrier (BBB) and the complement landscape. Mol. Immunol. 2018, 102, 26–31. [Google Scholar] [CrossRef] [PubMed]
- Pittet, C.L.; Newcombe, J.; Prat, A.; Arbour, N. Human brain endothelial cells endeavor to immunoregulate CD8 T cells via PD-1 ligand expression in multiple sclerosis. J. Neuroinflamm. 2011, 8, 155. [Google Scholar] [CrossRef] [PubMed]
- Mazanet, M.M.; Hughes, C.C. B7-H1 is expressed by human endothelial cells and suppresses T cell cytokine synthesis. J. Immunol. 2002, 169, 3581–3588. [Google Scholar] [CrossRef] [PubMed]
- Di Tacchio, M.; Macas, J.; Weissenberger, J.; Sommer, K.; Bahr, O.; Steinbach, J.P.; Senft, C.; Seifert, V.; Glas, M.; Herrlinger, U.; et al. Tumor Vessel Normalization, Immunostimulatory Reprogramming, and Improved Survival in Glioblastoma with Combined Inhibition of PD-1, Angiopoietin-2, and VEGF. Cancer Immunol. Res. 2019, 7, 1910–1927. [Google Scholar] [CrossRef]
- McLaughlin, M.; Patin, E.C.; Pedersen, M.; Wilkins, A.; Dillon, M.T.; Melcher, A.A.; Harrington, K.J. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nature Reviews Cancer 2020, 20, 203–217. [Google Scholar] [CrossRef]
- Bechmann, I.; Mor, G.; Nilsen, J.; Eliza, M.; Nitsch, R.; Naftolin, F. FasL (CD95L, Apo1L) is expressed in the normal rat and human brain: Evidence for the existence of an immunological brain barrier. Glia 1999, 27, 62–74. [Google Scholar] [CrossRef]
- Hickey, W.F. Basic principles of immunological surveillance of the normal central nervous system. Glia 2001, 36, 118–124. [Google Scholar] [CrossRef]
- Delgado, M.; Ganea, D. Vasoactive intestinal peptide: A neuropeptide with pleiotropic immune functions. Amino Acids 2013, 45, 25–39. [Google Scholar] [CrossRef]
- Gorczynski, L.; Chen, Z.; Hu, J.; Kai, Y.; Lei, J.; Ramakrishna, V.; Gorczynski, R.M. Evidence that an OX-2-positive cell can inhibit the stimulation of type 1 cytokine production by bone marrow-derived B7-1 (and B7-2)-positive dendritic cells. J. Immunol. 1999, 162, 774–781. [Google Scholar]
- Gorczynski, R.M.; Cattral, M.S.; Chen, Z.; Hu, J.; Lei, J.; Min, W.P.; Yu, G.; Ni, J. An immunoadhesin incorporating the molecule OX-2 is a potent immunosuppressant that prolongs allo- and xenograft survival. J. Immunol. 1999, 163, 1654–1660. [Google Scholar]
- Ransohoff, R.M.; Engelhardt, B. The anatomical and cellular basis of immune surveillance in the central nervous system. Nat. Rev. Immunol. 2012, 12, 623–635. [Google Scholar] [CrossRef] [PubMed]
- Brandao, M.; Simon, T.; Critchley, G.; Giamas, G. Astrocytes, the rising stars of the glioblastoma microenvironment. Glia 2019, 67, 779–790. [Google Scholar] [CrossRef] [PubMed]
- Henrik Heiland, D.; Ravi, V.M.; Behringer, S.P.; Frenking, J.H.; Wurm, J.; Joseph, K.; Garrelfs, N.W.C.; Strahle, J.; Heynckes, S.; Grauvogel, J.; et al. Tumor-associated reactive astrocytes aid the evolution of immunosuppressive environment in glioblastoma. Nat. Commun. 2019, 10, 2541. [Google Scholar] [CrossRef] [PubMed]
- Sin, W.C.; Aftab, Q.; Bechberger, J.F.; Leung, J.H.; Chen, H.; Naus, C.C. Astrocytes promote glioma invasion via the gap junction protein connexin43. Oncogene 2016, 35, 1504–1516. [Google Scholar] [CrossRef]
- Osswald, M.; Jung, E.; Sahm, F.; Solecki, G.; Venkataramani, V.; Blaes, J.; Weil, S.; Horstmann, H.; Wiestler, B.; Syed, M.; et al. Brain tumour cells interconnect to a functional and resistant network. Nature 2015, 528, 93–98. [Google Scholar] [CrossRef]
- Quail, D.F.; Bowman, R.L.; Akkari, L.; Quick, M.L.; Schuhmacher, A.J.; Huse, J.T.; Holland, E.C.; Sutton, J.C.; Joyce, J.A. The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas. Science 2016, 352, aad3018. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. The Microenvironmental Landscape of Brain Tumors. Cancer Cell 2017, 31, 326–341. [Google Scholar] [CrossRef]
- Greter, M.; Lelios, I.; Pelczar, P.; Hoeffel, G.; Price, J.; Leboeuf, M.; Kundig, T.M.; Frei, K.; Ginhoux, F.; Merad, M.; et al. Stroma-derived interleukin-34 controls the development and maintenance of langerhans cells and the maintenance of microglia. Immunity 2012, 37, 1050–1060. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Kana, V.; Desland, F.A.; Casanova-Acebes, M.; Ayata, P.; Badimon, A.; Nabel, E.; Yamamuro, K.; Sneeboer, M.; Tan, I.L.; Flanigan, M.E.; et al. CSF-1 controls cerebellar microglia and is required for motor function and social interaction. J. Exp. Med. 2019, 216, 2265–2281. [Google Scholar] [CrossRef] [PubMed]
- Colonna, M.; Butovsky, O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu. Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef] [PubMed]
- Krasemann, S.; Madore, C.; Cialic, R.; Baufeld, C.; Calcagno, N.; El Fatimy, R.; Beckers, L.; O’Loughlin, E.; Xu, Y.; Fanek, Z.; et al. The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity 2017, 47, 566–581. [Google Scholar] [CrossRef] [PubMed]
- Kwidzinski, E.; Bechmann, I. IDO expression in the brain: A double-edged sword. J. Mol. Med. 2007, 85, 1351–1359. [Google Scholar] [CrossRef]
- Buttgereit, A.; Lelios, I.; Yu, X.; Vrohlings, M.; Krakoski, N.R.; Gautier, E.L.; Nishinakamura, R.; Becher, B.; Greter, M. Sall1 is a transcriptional regulator defining microglia identity and function. Nat. Immunol. 2016, 17, 1397–1406. [Google Scholar] [CrossRef]
- Sorensen, M.D.; Dahlrot, R.H.; Boldt, H.B.; Hansen, S.; Kristensen, B.W. Tumour-associated microglia/macrophages predict poor prognosis in high-grade gliomas and correlate with an aggressive tumour subtype. Neuropathol. Appl. Neurobiol. 2018, 44, 185–206. [Google Scholar] [CrossRef]
- Hambardzumyan, D.; Gutmann, D.H.; Kettenmann, H. The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci. 2016, 19, 20–27. [Google Scholar] [CrossRef]
- 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]
- Yan, D.; Kowal, J.; Akkari, L.; Schuhmacher, A.J.; Huse, J.T.; West, B.L.; Joyce, J.A. Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene 2017, 36, 6049–6058. [Google Scholar] [CrossRef]
- Ries, C.H.; Cannarile, M.A.; Hoves, S.; Benz, J.; Wartha, K.; Runza, V.; Rey-Giraud, F.; Pradel, L.P.; Feuerhake, F.; Klaman, I.; et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 2014, 25, 846–859. [Google Scholar] [CrossRef]
- Cannarile, M.A.; Weisser, M.; Jacob, W.; Jegg, A.M.; Ries, C.H.; Ruttinger, D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J. Immunother. Cancer 2017, 5, 53. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Bowman, R.L.; Klemm, F.; Akkari, L.; Pyonteck, S.M.; Sevenich, L.; Quail, D.F.; Dhara, S.; Simpson, K.; Gardner, E.E.; Iacobuzio-Donahue, C.A.; et al. Macrophage Ontogeny Underlies Differences in Tumor-Specific Education in Brain Malignancies. Cell Rep. 2016, 17, 2445–2459. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Herndon, J.M.; Sojka, D.K.; Kim, K.W.; Knolhoff, B.L.; Zuo, C.; Cullinan, D.R.; Luo, J.; Bearden, A.R.; Lavine, K.J.; et al. Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 2017, 47, 323–338. [Google Scholar] [CrossRef] [PubMed]
- Loyher, P.L.; Hamon, P.; Laviron, M.; Meghraoui-Kheddar, A.; Goncalves, E.; Deng, Z.; Torstensson, S.; Bercovici, N.; Baudesson de Chanville, C.; Combadiere, B.; et al. Macrophages of distinct origins contribute to tumor development in the lung. J. Exp. Med. 2018, 215, 2536–2553. [Google Scholar] [CrossRef]
- Muller, S.; Kohanbash, G.; Liu, S.J.; Alvarado, B.; Carrera, D.; Bhaduri, A.; Watchmaker, P.B.; Yagnik, G.; Di Lullo, E.; Malatesta, M.; et al. Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome Biol. 2017, 18, 234. [Google Scholar] [CrossRef]
- Tamura, R.; Ohara, K.; Sasaki, H.; Morimoto, Y.; Kosugi, K.; Yoshida, K.; Toda, M. Difference in Immunosuppressive Cells Between Peritumoral Area and Tumor Core in Glioblastoma. World Neurosurg. 2018, 120, e601–e610. [Google Scholar] [CrossRef]
- Darmanis, S.; Sloan, S.A.; Croote, D.; Mignardi, M.; Chernikova, S.; Samghababi, P.; Zhang, Y.; Neff, N.; Kowarsky, M.; Caneda, C.; et al. Single-Cell RNA-Seq Analysis of Infiltrating Neoplastic Cells at the Migrating Front of Human Glioblastoma. Cell Rep. 2017, 21, 1399–1410. [Google Scholar] [CrossRef]
- Shono, K.; Yamaguchi, I.; Mizobuchi, Y.; Kagusa, H.; Sumi, A.; Fujihara, T.; Nakajima, K.; Kitazato, K.T.; Matsuzaki, K.; Saya, H.; et al. Downregulation of the CCL2/CCR2 and CXCL10/CXCR3 axes contributes to antitumor effects in a mouse model of malignant glioma. Sci. Rep. 2020, 10, 15286. [Google Scholar] [CrossRef]
- Flores-Toro, J.A.; Luo, D.; Gopinath, A.; Sarkisian, M.R.; Campbell, J.J.; Charo, I.F.; Singh, R.; Schall, T.J.; Datta, M.; Jain, R.K.; et al. CCR2 inhibition reduces tumor myeloid cells and unmasks a checkpoint inhibitor effect to slow progression of resistant murine gliomas. Proc. Natl. Acad. Sci. USA 2020, 117, 1129–1138. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Robertson, F.L.; Marques-Torrejon, M.A.; Morrison, G.M.; Pollard, S.M. Experimental models and tools to tackle glioblastoma. Dis. Models Mech. 2019, 12, dmm040386. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.; Kotliarova, S.; Kotliarov, Y.; Li, A.; Su, Q.; Donin, N.M.; Pastorino, S.; Purow, B.W.; Christopher, N.; Zhang, W.; et al. Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 2006, 9, 391–403. [Google Scholar] [CrossRef] [PubMed]
- Hubert, C.G.; Rivera, M.; Spangler, L.C.; Wu, Q.; Mack, S.C.; Prager, B.C.; Couce, M.; McLendon, R.E.; Sloan, A.E.; Rich, J.N. A Three-Dimensional Organoid Culture System Derived from Human Glioblastomas Recapitulates the Hypoxic Gradients and Cancer Stem Cell Heterogeneity of Tumors Found In Vivo. Cancer Res. 2016, 76, 2465–2477. [Google Scholar] [CrossRef] [PubMed]
- Szatmari, T.; Lumniczky, K.; Desaknai, S.; Trajcevski, S.; Hidvegi, E.J.; Hamada, H.; Safrany, G. Detailed characterization of the mouse glioma 261 tumor model for experimental glioblastoma therapy. Cancer Sci. 2006, 97, 546–553. [Google Scholar] [CrossRef] [PubMed]
- Oh, T.; Fakurnejad, S.; Sayegh, E.T.; Clark, A.J.; Ivan, M.E.; Sun, M.Z.; Safaee, M.; Bloch, O.; James, C.D.; Parsa, A.T. Immunocompetent murine models for the study of glioblastoma immunotherapy. J. Transl. Med. 2014, 12, 107. [Google Scholar] [CrossRef]
- Behnan, J.; Isakson, P.; Joel, M.; Cilio, C.; Langmoen, I.A.; Vik-Mo, E.O.; Badn, W. Recruited brain tumor-derived mesenchymal stem cells contribute to brain tumor progression. Stem Cells 2014, 32, 1110–1123. [Google Scholar] [CrossRef]
- Chen, J.; McKay, R.M.; Parada, L.F. Malignant glioma: Lessons from genomics, mouse models, and Stem Cells. Cell 2012, 149, 36–47. [Google Scholar] [CrossRef]
- Szulzewsky, F.; Pelz, A.; Feng, X.; Synowitz, M.; Markovic, D.; Langmann, T.; Holtman, I.R.; Wang, X.; Eggen, B.J.; Boddeke, H.W.; et al. Glioma-associated microglia/macrophages display an expression profile different from M1 and M2 polarization and highly express Gpnmb and Spp1. PLoS ONE 2015, 10, e0116644. [Google Scholar] [CrossRef]
- Hambardzumyan, D.; Parada, L.F.; Holland, E.C.; Charest, A. Genetic modeling of gliomas in mice: New tools to tackle old problems. Glia 2011, 59, 1155–1168. [Google Scholar] [CrossRef]
- Chow, L.M.; Endersby, R.; Zhu, X.; Rankin, S.; Qu, C.; Zhang, J.; Broniscer, A.; Ellison, D.W.; Baker, S.J. Cooperativity within and among Pten, p53, and Rb pathways induces high-grade astrocytoma in adult brain. Cancer Cell 2011, 19, 305–316. [Google Scholar] [CrossRef] [PubMed]
- Holland, E.C.; Hively, W.P.; DePinho, R.A.; Varmus, H.E. A constitutively active epidermal growth factor receptor cooperates with disruption of G1 cell-cycle arrest pathways to induce glioma-like lesions in mice. Genes Dev. 1998, 12, 3675–3685. [Google Scholar] [CrossRef] [PubMed]
- Hambardzumyan, D.; Amankulor, N.M.; Helmy, K.Y.; Becher, O.J.; Holland, E.C. Modeling Adult Gliomas Using RCAS/t-va Technology. Transl. Oncol. 2009, 2, 89–95. [Google Scholar] [CrossRef] [PubMed]
- Uhrbom, L.; Hesselager, G.; Nister, M.; Westermark, B. Induction of brain tumors in mice using a recombinant platelet-derived growth factor B-chain retrovirus. Cancer Res. 1998, 58, 5275–5279. [Google Scholar]
- Zhu, H.; Acquaviva, J.; Ramachandran, P.; Boskovitz, A.; Woolfenden, S.; Pfannl, R.; Bronson, R.T.; Chen, J.W.; Weissleder, R.; Housman, D.E.; et al. Oncogenic EGFR signaling cooperates with loss of tumor suppressor gene functions in gliomagenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 2712–2716. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Hu, B.; Hu, X.; Kim, H.; Squatrito, M.; Scarpace, L.; deCarvalho, A.C.; Lyu, S.; Li, P.; Li, Y.; et al. Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment. Cancer Cell 2018, 33, 152. [Google Scholar] [CrossRef]
- Herting, C.J.; Chen, Z.; Pitter, K.L.; Szulzewsky, F.; Kaffes, I.; Kaluzova, M.; Park, J.C.; Cimino, P.J.; Brennan, C.; Wang, B.; et al. Genetic driver mutations define the expression signature and microenvironmental composition of high-grade gliomas. Glia 2017, 65, 1914–1926. [Google Scholar] [CrossRef]
- Khalsa, J.K.; Cheng, N.; Keegan, J.; Chaudry, A.; Driver, J.; Bi, W.L.; Lederer, J.; Shah, K. Immune phenotyping of diverse syngeneic murine brain tumors identifies immunologically distinct types. Nat. Commun. 2020, 11, 3912. [Google Scholar] [CrossRef]
- Woroniecka, K.I.; Rhodin, K.E.; Chongsathidkiet, P.; Keith, K.A.; Fecci, P.E. T-cell Dysfunction in Glioblastoma: Applying a New Framework. Clin. Cancer Res. 2018, 24, 3792–3802. [Google Scholar] [CrossRef]
- 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]
- Dock, G. The influence of complicating diseases upon leukemia. Am. J. Med. Sci. 1902, 127, 563–593. [Google Scholar] [CrossRef]
- Kelly, E.; Russell, S.J. History of oncolytic viruses: Genesis to genetic engineering. Mol. Ther. 2007, 15, 651–659. [Google Scholar] [CrossRef] [PubMed]
- Lawler, S.E.; Speranza, M.C.; Cho, C.F.; Chiocca, E.A. Oncolytic Viruses in Cancer Treatment: A Review. JAMA Oncol. 2017, 3, 841–849. [Google Scholar] [CrossRef] [PubMed]
- van den Bossche, W.B.L.; Kleijn, A.; Teunissen, C.E.; Voerman, J.S.A.; Teodosio, C.; Noske, D.P.; van Dongen, J.J.M.; Dirven, C.M.F.; Lamfers, M.L.M. Oncolytic virotherapy in glioblastoma patients induces a tumor macrophage phenotypic shift leading to an altered glioblastoma microenvironment. Neuro-Oncology 2018, 20, 1494–1504. [Google Scholar] [CrossRef]
- Goradel, N.H.; Baker, A.T.; Arashkia, A.; Ebrahimi, N.; Ghorghanlu, S.; Negahdari, B. Oncolytic virotherapy: Challenges and solutions. Curr. Probl. Cancer 2020, 100639. [Google Scholar] [CrossRef]
- Belcaid, Z.; Berrevoets, C.; Choi, J.; van Beelen, E.; Stavrakaki, E.; Pierson, T.; Kloezeman, J.; Routkevitch, D.; van der Kaaij, M.; van der Ploeg, A.; et al. Low-dose oncolytic adenovirus therapy overcomes tumor-induced immune suppression and sensitizes intracranial gliomas to anti-PD-1 therapy. Neurooncol. Adv. 2020, 2, vdaa011. [Google Scholar] [CrossRef]
- Rahman, M.; Dastmalchi, F.; Karachi, A.; Mitchell, D. The role of CMV in glioblastoma and implications for immunotherapeutic strategies. Oncoimmunology 2019, 8, e1514921. [Google Scholar] [CrossRef]
- Gromeier, M.; Nair, S.K. Recombinant Poliovirus for Cancer Immunotherapy. Annu. Rev. Med. 2018, 69, 289–299. [Google Scholar] [CrossRef]
- Totsch, S.K.; Schlappi, C.; Kang, K.D.; Ishizuka, A.S.; Lynn, G.M.; Fox, B.; Beierle, E.A.; Whitley, R.J.; Markert, J.M.; Gillespie, G.Y.; et al. Oncolytic herpes simplex virus immunotherapy for brain tumors: Current pitfalls and emerging strategies to overcome therapeutic resistance. Oncogene 2019, 38, 6159–6171. [Google Scholar] [CrossRef]
- Martuza, R.L.; Malick, A.; Markert, J.M.; Ruffner, K.L.; Coen, D.M. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science 1991, 252, 854–856. [Google Scholar] [CrossRef]
- Lang, F.F.; Conrad, C.; Gomez-Manzano, C.; Yung, W.K.A.; Sawaya, R.; Weinberg, J.S.; Prabhu, S.S.; Rao, G.; Fuller, G.N.; Aldape, K.D.; et al. Phase I Study of DNX-2401 (Delta-24-RGD) Oncolytic Adenovirus: Replication and Immunotherapeutic Effects in Recurrent Malignant Glioma. J. Clin. Oncol. 2018, 36, 1419–1427. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Batich, K.A.; Reap, E.A.; Archer, G.E.; Sanchez-Perez, L.; Nair, S.K.; Schmittling, R.J.; Norberg, P.; Xie, W.; Herndon, J.E., 2nd; Healy, P.; et al. Long-term Survival in Glioblastoma with Cytomegalovirus pp65-Targeted Vaccination. Clin. Cancer Res. 2017, 23, 1898–1909. [Google Scholar] [CrossRef] [PubMed]
- Hildner, K.; Edelson, B.T.; Purtha, W.E.; Diamond, M.; Matsushita, H.; Kohyama, M.; Calderon, B.; Schraml, B.U.; Unanue, E.R.; Diamond, M.S.; et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell. Immun. Sci. 2008, 322, 1097–1100. [Google Scholar] [CrossRef]
- Salmon, H.; Idoyaga, J.; Rahman, A.; Leboeuf, M.; Remark, R.; Jordan, S.; Casanova-Acebes, M.; Khudoynazarova, M.; Agudo, J.; Tung, N.; et al. Expansion and Activation of CD103(+) Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition. Immunity 2016, 44, 924–938. [Google Scholar] [CrossRef]
- Liau, L.M.; Black, K.L.; Prins, R.M.; Sykes, S.N.; DiPatre, P.L.; Cloughesy, T.F.; Becker, D.P.; Bronstein, J.M. Treatment of intracranial gliomas with bone marrow-derived dendritic cells pulsed with tumor antigens. J. Neurosurg. 1999, 90, 1115–1124. [Google Scholar] [CrossRef]
- Heimberger, A.B.; Crotty, L.E.; Archer, G.E.; McLendon, R.E.; Friedman, A.; Dranoff, G.; Bigner, D.D.; Sampson, J.H. Bone marrow-derived dendritic cells pulsed with tumor homogenate induce immunity against syngeneic intracerebral glioma. J. Neuroimmunol. 2000, 103, 16–25. [Google Scholar] [CrossRef]
- Pellegatta, S.; Poliani, P.L.; Corno, D.; Grisoli, M.; Cusimano, M.; Ubiali, F.; Baggi, F.; Bruzzone, M.G.; Finocchiaro, G. Dendritic cells pulsed with glioma lysates induce immunity against syngeneic intracranial gliomas and increase survival of tumor-bearing mice. Neurol. Res. 2006, 28, 527–531. [Google Scholar] [CrossRef]
- Lim, M.; Xia, Y.; Bettegowda, C.; Weller, M. Current state of immunotherapy for glioblastoma. Nat. Rev. Clin. Oncol. 2018, 15, 422–442. [Google Scholar] [CrossRef]
- Mitchell, D.A.; Batich, K.A.; Gunn, M.D.; Huang, M.N.; Sanchez-Perez, L.; Nair, S.K.; Congdon, K.L.; Reap, E.A.; Archer, G.E.; Desjardins, A.; et al. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature 2015, 519, 366–369. [Google Scholar] [CrossRef]
- King, G.D.; Muhammad, A.K.; Larocque, D.; Kelson, K.R.; Xiong, W.; Liu, C.; Sanderson, N.S.; Kroeger, K.M.; Castro, M.G.; Lowenstein, P.R. Combined Flt3L/TK gene therapy induces immunological surveillance which mediates an immune response against a surrogate brain tumor neoantigen. Mol Ther. 2011, 19, 1793–1801. [Google Scholar] [CrossRef] [PubMed]
- Garzon-Muvdi, T.; Theodros, D.; Luksik, A.S.; Maxwell, R.; Kim, E.; Jackson, C.M.; Belcaid, Z.; Ganguly, S.; Tyler, B.; Brem, H.; et al. Dendritic cell activation enhances anti-PD-1 mediated immunotherapy against glioblastoma. Oncotarget 2018, 9, 20681–20697. [Google Scholar] [CrossRef] [PubMed]
- Eoli, M.; Corbetta, C.; Anghileri, E.; Di Ianni, N.; Milani, M.; Cuccarini, V.; Musio, S.; Paterra, R.; Frigerio, S.; Nava, S.; et al. Expansion of effector and memory T cells is associated with increased survival in recurrent glioblastomas treated with dendritic cell immunotherapy. Neurooncol. Adv. 2019, 1, vdz022. [Google Scholar] [CrossRef] [PubMed]
- Mahoney, K.M.; Freeman, G.J.; McDermott, D.F. The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in Melanoma. Clin. Ther. 2015, 37, 764–782. [Google Scholar] [CrossRef] [PubMed]
- Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Reardon, D.A.; Gokhale, P.C.; Klein, S.R.; Ligon, K.L.; Rodig, S.J.; Ramkissoon, S.H.; Jones, K.L.; Conway, A.S.; Liao, X.; Zhou, J.; et al. Glioblastoma Eradication Following Immune Checkpoint Blockade in an Orthotopic, Immunocompetent Model. Cancer Immunol. Res. 2016, 4, 124–135. [Google Scholar] [CrossRef]
- Fecci, P.E.; Ochiai, H.; Mitchell, D.A.; Grossi, P.M.; Sweeney, A.E.; Archer, G.E.; Cummings, T.; Allison, J.P.; Bigner, D.D.; Sampson, J.H. Systemic CTLA-4 blockade ameliorates glioma-induced changes to the CD4+ T cell compartment without affecting regulatory T-cell function. Clin. Cancer Res. 2007, 13, 2158–2167. [Google Scholar] [CrossRef]
- Ito, H.; Nakashima, H.; Chiocca, E.A. Molecular responses to immune checkpoint blockade in glioblastoma. Nat. Med. 2019, 25, 359–361. [Google Scholar] [CrossRef]
- Zheng, H.; Zeltsman, M.; Zauderer, M.G.; Eguchi, T.; Vaghjiani, R.G.; Adusumilli, P.S. Chemotherapy-induced immunomodulation in non-small-cell lung cancer: A rationale for combination chemoimmunotherapy. Immunotherapy 2017, 9, 913–927. [Google Scholar] [CrossRef]
- Bagley, S.J.; Desai, A.S.; Linette, G.P.; June, C.H.; O’Rourke, D.M. CAR T-cell therapy for glioblastoma: Recent clinical advances and future challenges. Neuro-Oncology 2018, 20, 1429–1438. [Google Scholar] [CrossRef]
- Brown, C.E.; Aguilar, B.; Starr, R.; Yang, X.; Chang, W.C.; Weng, L.; Chang, B.; Sarkissian, A.; Brito, A.; Sanchez, J.F.; et al. Optimization of IL13Ralpha2-Targeted Chimeric Antigen Receptor T Cells for Improved Anti-tumor Efficacy against Glioblastoma. Mol. Ther. 2018, 26, 31–44. [Google Scholar] [CrossRef] [PubMed]
- Kahlon, K.S.; Brown, C.; Cooper, L.J.; Raubitschek, A.; Forman, S.J.; Jensen, M.C. Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells. Cancer Res. 2004, 64, 9160–9166. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, N.; Salsman, V.S.; Kew, Y.; Shaffer, D.; Powell, S.; Zhang, Y.J.; Grossman, R.G.; Heslop, H.E.; Gottschalk, S. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin. Cancer Res. 2010, 16, 474–485. [Google Scholar] [CrossRef] [PubMed]
- Sampson, J.H.; Choi, B.D.; Sanchez-Perez, L.; Suryadevara, C.M.; Snyder, D.J.; Flores, C.T.; Schmittling, R.J.; Nair, S.K.; Reap, E.A.; Norberg, P.K.; et al. EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebral glioma and generates host immunity against tumor-antigen loss. Clin. Cancer Res. 2014, 20, 972–984. [Google Scholar] [CrossRef]
- June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science 2018, 359, 1361–1365. [Google Scholar] [CrossRef]
- Slaney, C.Y.; Wang, P.; Darcy, P.K.; Kershaw, M.H. CARs versus BiTEs: A Comparison between T Cell-Redirection Strategies for Cancer Treatment. Cancer Discov. 2018, 8, 924–934. [Google Scholar] [CrossRef]
- Ellerman, D. Bispecific T-cell engagers: Towards understanding variables influencing the in vitro potency and tumor selectivity and their modulation to enhance their efficacy and safety. Methods 2019, 154, 102–117. [Google Scholar] [CrossRef]
- Einsele, H.; Borghaei, H.; Orlowski, R.Z.; Subklewe, M.; Roboz, G.J.; Zugmaier, G.; Kufer, P.; Iskander, K.; Kantarjian, H.M. The BiTE (bispecific T-cell engager) platform: Development and future potential of a targeted immuno-oncology therapy across tumor types. Cancer 2020, 126, 3192–3201. [Google Scholar] [CrossRef]
- Yang, J.; Yan, J.; Liu, B. Targeting EGFRvIII for glioblastoma multiforme. Cancer Lett. 2017, 403, 224–230. [Google Scholar] [CrossRef]
- Gedeon, P.C.; Choi, B.D.; Hodges, T.R.; Mitchell, D.A.; Bigner, D.D.; Sampson, J.H. An EGFRvIII-targeted bispecific T-cell engager overcomes limitations of the standard of care for glioblastoma. Expert Rev. Clin. Pharmacol. 2013, 6, 375–386. [Google Scholar] [CrossRef][Green Version]
- Ma, Q.; Ineichen, B.V.; Detmar, M.; Proulx, S.T. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat. Commun. 2017, 8, 1434. [Google Scholar] [CrossRef] [PubMed]
- Song, E.; Mao, T.; Dong, H.; Boisserand, L.S.B.; Antila, S.; Bosenberg, M.; Alitalo, K.; Thomas, J.L.; Iwasaki, A. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 2020, 577, 689–694. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Deng, Q.; Ma, L.; Li, Q.; Chen, Y.; Liao, Y.; Zhou, F.; Zhang, C.; Shao, L.; Feng, J.; et al. Meningeal lymphatic vessels regulate brain tumor drainage and Immunity. Cell Res. 2020, 30, 229–243. [Google Scholar] [CrossRef] [PubMed]
- Sampson, J.H.; Gunn, M.D.; Fecci, P.E.; Ashley, D.M. Brain immunology and immunotherapy in brain tumours. Nat. Rev. Cancer 2020, 20, 12–25. [Google Scholar] [CrossRef] [PubMed]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Desland, F.A.; Hormigo, A. The CNS and the Brain Tumor Microenvironment: Implications for Glioblastoma Immunotherapy. Int. J. Mol. Sci. 2020, 21, 7358. https://doi.org/10.3390/ijms21197358
Desland FA, Hormigo A. The CNS and the Brain Tumor Microenvironment: Implications for Glioblastoma Immunotherapy. International Journal of Molecular Sciences. 2020; 21(19):7358. https://doi.org/10.3390/ijms21197358
Chicago/Turabian StyleDesland, Fiona A., and Adília Hormigo. 2020. "The CNS and the Brain Tumor Microenvironment: Implications for Glioblastoma Immunotherapy" International Journal of Molecular Sciences 21, no. 19: 7358. https://doi.org/10.3390/ijms21197358
APA StyleDesland, F. A., & Hormigo, A. (2020). The CNS and the Brain Tumor Microenvironment: Implications for Glioblastoma Immunotherapy. International Journal of Molecular Sciences, 21(19), 7358. https://doi.org/10.3390/ijms21197358