Dear Editor,
High-grade meningiomas (HGM) remain a formidable clinical challenge due to their aggressive biological behavior, high recurrence rates, and limited responsiveness to conventional therapies. Standard approaches such as surgical resection, radiation, and chemotherapy often fail to achieve durable disease control or significantly improve progression-free survival (PFS) in hemodialysis and non-dialysis patients alike [1-4]. These limitations underscore the urgent need for novel therapeutic strategies, particularly immunotherapy, which aims to harness and modulate the host immune system to recognize and eliminate tumor cells.
Immunological Barriers in CNS Tumors
The CNS has long been considered an immune- privileged site due to the presence of the blood- brain barrier (BBB), limited lymphatic drainage, and immunosuppressive microenvironment [5]. GBM, in particular, is categorized as an immunologically “cold” tumor, characterized by poor T cell infiltration, low mutational burden, and upregulation of immune-inhibitory signals [6, 7]. In the CheckMate-143 trial, nivolumab, an anti-PD-1 agent, yielded an objective response in only 8% of recurrent GBM patients, highlighting the formidable barriers to immunotherapy in this setting [8].
Interestingly, meningiomas especially HGM do not reside entirely within the BBB, allowing for greater immune cell access. Nonetheless, these tumors have evolved mechanisms to evade immune surveillance, including the expression of immune checkpoint ligands (PD-L1, PD-L2, CTLA-4, B7-H3), the recruitment of immunosuppressive regulatory T cells (Tregs), tumor- associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), and the promotion of a tolerogenic microenvironment [9-13].
Immunological Landscape of High-Grade Meningiomas
Recent immunogenomic and transcriptomic profiling studies have revealed significant heterogeneity in immune infiltration patterns among meningiomas of different WHO grades [14-16]. Grade III meningiomas exhibit a higher density of immune infiltrates, particularly CD8+ T cells, but also an increased presence of immunosuppressive elements, including PD-1+ exhausted T cells, FOXP3+ Tregs, and M2-polarized TAMs [17, 18].
Fang et al. conducted an in-depth analysis of meningioma-infiltrating immune cells, revealing that both CD4+ and CD8+ T cells exhibited antigen experience and checkpoint receptor expression (PD-1, TIM-3), indicative of functional exhaustion [19]. B cell infiltration was rare but notable for somatic hypermutation and clonal expansion, implying active antigen presentation and participation in intratumoral immunity [20].
Interestingly, WHO grade III tumors exhibit elevated levels of MDSCs and PD-L1-expressing macrophages, which correlate with poor prognosis [21-23]. The presence of chromosomal 22q deletions, frequently observed in high-grade tumors, has also been associated with enhanced M1/M2 macrophage polarization and increased immune infiltration [24, 25].
Opportunities for Immunotherapeutic Intervention
The expression of immune checkpoints in HGM makes them attractive candidates for ICIs. PD-L1 is particularly overexpressed in WHO grade II and III meningiomas, correlating with tumor progression and recurrence [26]. Preclinical studies suggest that blocking PD-1/PD-L1 interactions can restore T cell function and enhance anti-tumor immunity [27-29]. Moreover, CAR-T cells targeting antigens such as IL13Rα2, HER2, and EphA2 though more commonly studied in gliomas are being investigated for their applicability in meningioma [30-32].
Vaccination strategies utilizing tumor-specific peptides or dendritic cells (DCs) loaded with meningioma antigens have shown early promise in generating robust cytotoxic T lymphocyte (CTL) responses [33-35]. Similarly, oncolytic viral therapies are being explored to lyse tumor cells directly while inducing systemic anti-tumor immunity [36].
Novel Perspectives and Future Directions
Recent efforts have focused on improving patient stratification using immunohistochemical, genomic, and proteomic biomarkers to predict responsiveness to immunotherapy [37-40]. For example, immune gene expression profiling has identified “immune hot” and “cold” meningiomas, analogous to the classification used in melanoma and lung cancer [41, 42].
The integration of immunotherapy with radiation therapy is another promising avenue. Radiation may enhance antigen presentation and upregulate checkpoint molecule expression, potentially synergizing with ICIs or CAR-T therapy [43, 44]. Additionally, modulation of the gut microbiome has emerged as a novel means of enhancing systemic immunity in CNS tumors [45-47].
In conclusion, this article underscores the growing promise of immunotherapy as a transformative approach in the management of high-grade meningiomas (HGMs), which have long demonstrated resistance to conventional treatment modalities. In contrast to glioblastomas, HGMs exhibit partial immune accessibility and harbor distinct immunogenomic characteristics, including elevated PD-L1 expression and activation of immune checkpoints. These features render HGMs viable candidates for immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T-cell (CAR-T) therapies. Furthermore, emerging insights into the tumor immune microenvironment particularly the regulatory roles of T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages highlight critical mechanisms of immune evasion and potential targets for therapeutic intervention. Integrative treatment strategies, encompassing the combination of immunotherapy with radiotherapy, the use of personalized immune biomarkers, and modulation of the microbiome, represent the forefront of precision oncology for patients with high-grade meningiomas.
Author Contribution Statement
Conceptualization, funding acquisition, Investigation, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing original draft, Writing review and editing, Final approval of manuscript for publication: All authors.
Acknowledgements
We thank all those who assisted us in the course of this work.
Ethical approval
Ethical approval was not required for this Editorial Article
Consent
Informed consent was not required for this manuscript since it was an Editorial Article.
Conflict of interest
There was not any conflict of interest among the authors of this editorial article.
References
References
- Goldbrunner R, Minniti G, Preusser M, Jenkinson MD, Sallabanda K, Houdart E, Deimling A, et al. EANO guidelines for the diagnosis and treatment of meningiomas. The Lancet. Oncology. 2016; 17(9)DOI
- Mawrin C, Perry A. Pathological classification and molecular genetics of meningiomas. Journal of Neuro-Oncology. 2010; 99(3)DOI
- Kaley TJ, et al. Management of recurrent meningiomas: A clinical review. J Neurooncol. 2017; 133(3):545-553.
- Magill ST, et al. Defining aggressive meningiomas: Updates from the WHO Classification. Brain Pathol. 2021; 31(6):e12990.
- Boussiotis VA, Charest A. Immunotherapies for malignant glioma. Oncogene. 2018; 37(9)DOI
- Sun C, et al. Tumor immune microenvironment in glioblastoma: mechanisms and potential therapeutic strategies. Cancer Lett. 2022; 545:215820.
- Han S, et al. Tumor-associated macrophages in meningiomas. Brain Tumor Pathol. 2020; 37(4):134-42.
- Han S, et al. PD-L1 expression and prognostic implications in meningiomas. Neurosurg Rev. 2021; 44(2):943-952.
- Wang YC, et al. PD-L1 expression in meningiomas and implications for immunotherapy. J Neurooncol. 2018; 139(3):483-90.
- Domenico F, et al. Tumor microenvironment and immune checkpoints in meningioma: new frontiers for targeted therapy. Cancers (Basel). 2021; 13(4):689.
- Dutoit V, et al. Exploiting the immune system for glioma therapy: current status and future perspectives. Brain Pathol. 2020; 30(3):613-634.
- Sayour EJ, et al. Immunotherapy for pediatric brain tumors: a review. J Neurooncol. 2021; 151(1):1-13.
- Lehrer E, et al. Proton and photon therapy for primary CNS tumors: A systematic review. Curr Treat Options Oncol. 2020; 21(2):10.
- Nassiri F, et al. Integrated molecular characterization of meningiomas identifies the prognostic significance of immune signatures. Nat Commun. 2021; 12(1):7095.
- Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, Cahill DP, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science (New York, N.Y.). 2014; 344(6190)DOI
- Lopez-Rivera E, et al. Landscape of tumor-infiltrating T cell repertoire of human cancers. Nat Commun. 2021; 12(1):1740.
- Choudhury A, et al. Role of immune checkpoint blockade in CNS tumors: current evidence and future directions. Neurooncol Adv. 2021; 3(1):vdab059.
- Gieryng A, Pszczolkowska D, Walentynowicz KA, Rajan WD, Kaminska B. Immune microenvironment of gliomas. Laboratory Investigation; a Journal of Technical Methods and Pathology. 2017; 97(5)DOI
- Fang AS, et al. Immune profiling of human meningiomas reveals a paracrine loop promoting tumor proliferation and immune evasion. Cell Rep. 2021; 36(10):109628.
- Schittenhelm J, et al. Expression of immune checkpoints in meningiomas: stratification by WHO grade and mutational profile. Cancers (Basel). 2020; 12(2):362.
- Uddin MN, et al. Myeloid-derived suppressor cells in cancer: Understanding their regulatory mechanisms and therapeutic implications. Front Immunol. 2020; 11:1879.
- Acar A, et al. Immunogenomic profiling of meningiomas identifies distinct immune phenotypes correlating with histological grade. Acta Neuropathol Commun. 2021; 9(1):78.
- Grauer OM, et al. CD4+FoxP3+ regulatory T cells in gliomas: the jury is still out. J Neuroimmunol. 2007; 189((1–2)):1-5.
- Aizer AA, et al. Meningioma recurrence rate following resection: systematic review and meta-analysis. J Neurosurg. 2014; 120(5):1186-1193.
- Shankar GM, et al. Combined histologic and molecular WHO grading for meningiomas. Acta Neuropathol. 2022; 144(2):191-201.
- Simonetti G, et al. The immune microenvironment in meningiomas: Implications for immunotherapy. Int J Mol Sci. 2021; 22(20):10851.
- Dunn GP, Rinne ML, Wykosky J, Genovese G, Quayle SN, Dunn IF, Agarwalla PL, et al. Emerging insights into the molecular and cellular basis of glioblastoma. Genes & Development. 2012; 26(8)DOI
- Dunn IF, et al. Molecular profiling of meningioma: guiding patient care, neurosurgery, and research. Neurosurgery. 2020; 87(4):769-780.
- Johnson BE, et al. Comprehensive genomic characterization of meningiomas reveals the prognostic role of epigenetic subgroup. Cell Rep. 2021; 36(6):109422.
- Sampson JH, et al. EGFRvIII peptide vaccination is associated with improved survival in glioblastoma. J Clin Oncol. 2010; 28(31):4722-4729.
- Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, Ostberg JR, et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. The New England Journal of Medicine. 2016; 375(26)DOI
- Weller M, et al. Immunotherapy for brain tumors. Neuro Oncol. 2015; 17(Suppl 7):vii75–87.
- Platten M, et al. Cancer immunotherapy targeting IDH1(R132H), a neoantigen in glioma. Nature. 2021; 597(7878):662-667.
- Lim M, et al. Current approaches for glioblastoma treatment: converging on immunotherapy. J Clin Invest. 2018; 128(1):40-47.
- Ramakrishna R, et al. Immune checkpoint blockade in CNS tumors: rationale, challenges, and future directions. Neuro Oncol. 2020; 22(7):933-944.
- Zeng J, et al. Oncolytic viruses: promising tools for cancer immunotherapy. Transl Cancer Res. 2021; 10(4):1889-1898.
- Reuss DE, et al. A comprehensive analysis of DNA methylation profiles identifies novel meningioma subgroups. Acta Neuropathol. 2015; 130(3):309-321.
- Sahm F, Schrimpf D, Stichel D, Jones DTW, Hielscher T, Schefzyk S, Okonechnikov K, et al. DNA methylation-based classification and grading system for meningioma: a multicentre, retrospective analysis. The Lancet. Oncology. 2017; 18(5)DOI
- Stichel D, et al. DNA methylation-based classification of meningiomas highlights the prognostic relevance of epigenetic profiling. Acta Neuropathol. 2021; 141(3):363-379.
- Schmid T, et al. Radiation-induced immune modulation: implications for immunotherapy of glioblastoma. Curr Treat Options Oncol. 2021; 21(7):62.
- Sahoo SS, et al. Immune cell profiling in glioblastoma: insights from single-cell technologies. Neurooncol Adv. 2020; 2(Suppl_1):i50–60.
- Zhai L, et al. Immunosuppressive tumor microenvironment in gliomas: potential targets for immunotherapy. Front Immunol. 2020; 11:603121.
- van den Bent MJ, et al. Challenges in integrating immunotherapy in the treatment of glioblastoma. Curr Opin Neurol. 2020; 33(6):726-733.
- Lim M, et al. Radiation and immunotherapy for brain tumors: current status and future directions. J Neurooncol. 2021; 151(3):451-464.
- Davar D, et al. The microbiome and cancer immunotherapy: diagnostic tools and therapeutic strategies. Science. 2021; 371(6536):eabc4552.
- Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell. 2018; 33(4)DOI
- Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science (New York, N.Y.). 2018; 359(6371)DOI