Next Article in Journal
EGFR-Mutant Non-Small-Cell Lung Cancer at Surgical Stages: What Is the Place for Tyrosine Kinase Inhibitors?
Next Article in Special Issue
Meningioma Radiomics: At the Nexus of Imaging, Pathology and Biomolecular Characterization
Previous Article in Journal
Cell Free Methylated Tumor DNA in Bronchial Lavage as an Additional Tool for Diagnosing Lung Cancer—A Systematic Review
Previous Article in Special Issue
The Simpson Grading: Is It Still Valid?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Significance of Molecular Alterations and Systemic Therapy for Meningiomas: Where Do We Stand?

1
Division of Neuro-Oncology, Department Neuroscience, University and City of Health and Science Hospital, 10126 Turin, Italy
2
Department of Neurology Unit, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
3
Pathology Unit, Department of Medical Sciences, University and City of Health and Science Hospital, 10126 Turin, Italy
4
Department of Neurology, Castelfranco Veneto and Treviso Hospital, 31100 Treviso, Italy
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(9), 2256; https://doi.org/10.3390/cancers14092256
Submission received: 13 April 2022 / Revised: 28 April 2022 / Accepted: 29 April 2022 / Published: 30 April 2022
(This article belongs to the Special Issue Meningiomas: Update on the Diagnosis and Management)

Abstract

:

Simple Summary

Meningiomas are the most frequent intracranial tumors and comprise a heterogeneous spectrum of diseases, ranging from small, asymptomatic tumors that do not need treatment to large, symptomatic ones causing seizures or neurological deficits that require surgery and/or radiotherapy. Systemic therapy is reserved for progressive or recurrent meningiomas when surgery and/or radiotherapy options have been exhausted, with only modest activity in terms of disease control and survival. Novel molecular alterations are correlated with grading, location, and prognosis of meningiomas. Moreover, some of these driver alterations regulate meningioma growth and progression and may be targeted by specific drugs that are under investigation in clinical trials. Lastly, the microenvironment surrounding meningiomas may also contribute to regulating tumor growth: in particular, PD-L1 and/or M2 macrophage expression may represent a target for immunotherapy.

Abstract

Meningiomas are common intracranial tumors that can be treated successfully in most cases with surgical resection and/or adjuvant radiotherapy. However, approximately 20% of patients show an aggressive clinical course with tumor recurrence or progressive disease, resulting in significant morbidity and increased mortality. Despite several studies that have investigated different cytotoxic agents in aggressive meningiomas in the past several years, limited evidence of efficacy and clinical benefit has been reported thus far. Novel molecular alterations have been linked to a particular clinicopathological phenotype and have been correlated with grading, location, and prognosis of meningiomas. In this regard, SMO, AKT, and PIK3CA mutations are typical of anterior skull base meningiomas, whereas KLF4 mutations are specific for secretory histology, and BAP1 alterations are common in progressive rhabdoid meningiomas. Alterations in TERT, DMD, and BAP1 correlate with poor outcomes. Moreover, some actionable mutations, including SMO, AKT1, and PIK3CA, regulate meningioma growth and are under investigation in clinical trials. PD-L1 and/or M2 macrophage expression in the microenvironment provides evidence for the investigation of immunotherapy in progressive meningiomas.

1. Introduction

Meningiomas are the most frequent intracranial tumors, with an annual age-adjusted incidence rate of 9.12 per 100,000 population in 2014–2018 according to the CBTRUS report of 2021 [1]. The incidence of meningiomas increases with age, with a major prevalence after the age of 66 years. Meningiomas comprise a heterogenous spectrum of diseases with significant variability in tumor biology and clinical outcome, ranging from small and asymptomatic incidental meningiomas that are observed and do not need treatment to large, symptomatic meningiomas causing seizures or neurological deficits that require surgery and/or radiotherapy [2]. Most meningiomas are grade 1 tumors (94.6%), where the gross total resection is curative, with excellent long-term control rates; however, some clinical series with follow-up ranging from 5 to 10 years suggest the risk of underreporting late meningioma recurrences that can occur decades after primary treatment [3]. Grades 2 and 3 meningiomas represent 4.2% and 1.2%, respectively [4], with early and multiple relapses that require multimodality treatment, including repeated surgery and/or radiation therapy, and, in selected cases, chemotherapy or experimental clinical trials. In general, systemic therapy is reserved for grade 2 or 3 meningiomas as a last chance of treatment, when surgery and/or radiotherapy options have been exhausted, with only modest efficacy.
This review discusses the emerging role of genetic and epigenetic features as relevant biomarkers for outcome prediction, as well as advances in systemic therapies for intracranial meningiomas.

2. Molecular Features and Correlation with Histology and Grading

The WHO classification of 2021 identifies 15 different meningioma subtypes. Some diagnostic difficulties may occur when achieving a histological diagnosis of meningioma: for instance, different histological patterns can co-occur within the same tumor sample, posing challenges in terms of diagnostic interpretation and resulting in prognostic assessment [5]. Grade 1 meningiomas comprise nine variants: Meningothelial and fibroblastic variants are very common, while other variants, such as metaplastic- or lymphoplasmacyte-rich meningioma, are exceptionally rare. Grade 2 meningiomas include three histological subtypes, characterized by increased proliferation, nuclear pleomorphism, tumor necrosis, brain invasion, and an increased risk of recurrence. Grade 3 tumors comprise three histological subtypes—namely, papillary, rhabdoid, and anaplastic—and present a highly aggressive behavior and poor clinical course [6]. The histopathological criterion alone still leaves some uncertainty regarding the risk assessment in meningiomas [7]: It is now emphasized that the criteria defining atypical or anaplastic (i.e., grades 2 and 3) meningioma should be applied regardless of the underlying subtype. The inclusion of some novel molecular alterations in the diagnostic assessment may improve the identification of patients with a higher risk of recurrence who need close surveillance and/or more aggressive treatment.
Higher rates of copy-number alterations and karyotypic abnormalities are reported in anaplastic/malignant meningiomas, while fewer copy-number alterations are common in grade 1 meningiomas, although a small subset of these tumors harbors complex genomic rearrangements [8].
The most frequent alteration in meningioma is the loss of the neurofibromin 2 (NF2) gene on chromosome 22 [9,10]. The tumor suppressor gene NF2 encodes the protein merlin (or schwannomin), which is correlated with the onset of schwannomas and meningiomas in the familial syndrome neurofibromatosis 2 [11] and is found in >50% of sporadic meningiomas [12,13,14]. The NF2 inactivation is mostly due to LOH of chr22q and NF2 mutation in other alleles, mitotic recombinations, or single or multiexon deletions, causing chromosomal instability that drives meningiomagenesis in a way that remains poorly understood [15]. Merlin negatively acts on multiple signaling pathways, including Hippo, Patched, and Notch pathways, and negatively regulates mammalian target of rapamycin complex 1 (mTORC1) [16] but positively influences the kinase activity of mTORC2 [17].
A gene involved in the growth of NF2-negative meningiomas is TRAF7, an E3 ubiquitin ligase that interacts with MEKK3/MAP3K3 (mitogen-activated protein kinase kinase 3) and regulates apoptosis. TRAF7 mutations were detected in up to 25% of grades 1 and 2 meningiomas [18,19]. A frequent aberration that can co-occur with TRAF7 is KLF4 mutations in up to 50% of NF2-nonmutated meningiomas of grade 1 [18,20,21]. Brastianos et al., using next-generation sequencing, showed that other genetic aberrations may be found in NF2-negative meningiomas, including mutations in KDM5C, KDM6A, and SMARCB1 in 8% of patients, and the other six patients exhibited mutations of the PI3K–AKT–mTOR pathway, of whom five harbored AKT1 mutations, and one, a novel MTOR mutation (p.Glu17Lys) [8]. Another study in 300 grades 1 and 2 meningiomas has found that 13% harbored the AKT1 p.Glu17Lys mutation [18] and displayed immunohistochemical evidence of PI3K–AKT–mTOR pathway activation. In addition, 1–5% of meningiomas without alterations in NF2 and AKT1, harbor mutations in the SMO gene, which encodes smoothened homolog, a member of the Hedgehog signaling pathway [8,18,19]. SMO interacts with the suppressor of fused homolog (SUFU), causing the nuclear translocation of zinc-finger protein GLI1 (GLI1) and activation of target genes involved in cellular proliferation and angiogenesis [22]. Notably, the PIK3CA-mutant meningiomas lacking mutations in NF2, AKT1, and SMO, tend to express TRAF7 mutations. Lastly, in the absence of any of the previously mentioned mutations, somatic mutations in POLR2A (encoding the DNA-directed RNA polymerase II subunit RPB1) may be found in about 6% of meningiomas. Typically, POLR2A is correlated with meningothelial histology, tumor location in the tuberculum sellae, and exclusive presence in grade 1 meningiomas [23]. In fact, the majority of genetic alterations listed above are found in grade 1 meningiomas only, while NF2 mutations are the dominant molecular events (75%) in grade 2 meningiomas, followed by 9% harboring TRAF7 or PI3K mutations, and 16% that do not contain any mutation [24].
Another important finding is the different role played by TERT mutations in favoring the progression of meningioma. In particular, the lack of TERT promoter mutations is the main characteristic of de novo grade 2 meningiomas, in contrast with the occurrence of TERT promoter mutations in secondary grade 2 meningiomas that have recurred from grade 1 [25]. NF2 alterations are the main finding also in grade 3 meningiomas. Vaubel et al. investigated the molecular features of rhabdoid meningiomas, which are designated as WHO grade 3 tumors, and found that the presence of a rhabdoid phenotype in the absence of other features of malignancy, such as high mitotic count and necroses, seems to define a clinical course comparable to grade 1 meningiomas [26]. Moreover, the identification of the inactivation of BAP1 in rhabdoid meningiomas may differentiate between aggressive and less-aggressive rhabdoid-appearing meningiomas, where the loss of BAP1 protein expression indicates early tumor recurrence [27]. A genomic survey in a large, multi-institutional cohort of high-grade/progressive meningiomas has revealed at least three distinct patterns. The most common subtype was the NF2- mutated (NF2-associated pattern) which frequently harbored CDKN2A/B alterations and may be eligible for targeted therapies; in addition, the NF2-mutated pathway partly associated with BAP1/PBRM1 alterations (rhabdoid/papillary histology) or skull-base disease (NF2-exclusive); lastly, the NF2-agnostic group harbored frequent TERTp and TP53 mutations [28]. Recently, these genetic aberrations have been correlated with methylation classes in order to provide prognostic information not captured by previously established clinical and molecular factors [29,30,31]. In particular, Sahm et al. have shown that the classification of meningiomas based on DNA methylation profiling provides a more precise prediction of clinical behaviour than the WHO classification and grading system. Six methylation classes were identified, three of which were benign (MC ben-1, MC ben-2, MC ben-3), two were intermediate (MC int-A and MC int-B), and one was malignant (MC mal). Interestingly, WHO grade 1 meningiomas clustered in MC ben-1, MC ben-2, and MC ben-3 subgroups, while WHO grade 3 meningiomas fell into the MC mal subgroup, and WHO grade 2 meningiomas were scattered across all methylation classes [29]. Regarding histology, MC ben-1 contained mainly fibroblastic and psammomatous meningiomas, MC ben-2 was highly enriched for meningothelial meningiomas and almost all secretory meningiomas, and MC ben-3 included several subtypes but was particularly enriched for angiomatous meningiomas. Transitional meningiomas, and other rare entities, such as microcystic, chordoid, clear-cell, and metaplastic meningiomas, were distributed into several methylation classes. MC int-A and MC int-B mainly comprised atypical meningiomas, while anaplastic meningiomas predominantly fell into MC mal, and in a small proportion, in MC int-B or MC int-A. NF2 inactivation alone was found in MC ben-1 and MC int A, or in association with TERT mutations in MC int-B and MC mal, while TRAF7, KLF4, AKT1, and SMO mutation were found in MC ben-2 only. MC ben-3 was not associated with any known mutation [29].

3. Molecular Features and Correlation with Location

The association between histological subtypes of meningiomas and their location are explained by embryonic reasons, as meninges at the skull base arise from the mesoderm, while meninges of the convexity derive from the neural crest. Therefore, meningothelial meningiomas preferentially develop from the skull base, while fibroblastic meningiomas arise primarily from convexity [32,33]. Grading is linked to the location of meningiomas, as grades 2 and 3 meningiomas are often located at the convexity or at parasagittal areas, whereas grade 1 meningiomas are mainly located at the skull base [34]. Meningiomas with AKT1 p.Glu17Lys mutations tend to have a skull-base or basal localization [35,36], SMO-mutated meningiomas predominate in the medial anterior skull base [18,37], PIK3CA-mutant meningiomas are preferentially localized at the skull base [19], and POLR2A-mutated meningiomas are mainly found in the tuberculum sellae region [23]. The presence of loss-of-function SMARCE1 mutations is significantly associated with clear-cell histology in the spinal cord and cranial meningiomas [38,39]. Most intraventricular meningiomas (44%) harbor NF2 mutations in the series of Jungwirth et al., while in non-NF2-mutated intraventricular meningiomas, genetic alterations including TRAF7, AKT1, SMO, KLF4, PIK3CA, and TERT are lacking, thus suggesting a role for alternative genes in the pathogenesis of non-NF2 intraventricular meningiomas. In fact, mutations of APC, GABRA6, GSE1, KDR, and two SMO missense mutations different from those previously reported have been found. Notably, all WHO grade 2 intraventricular meningiomas (n = 3) harbored SMARCB1 and SMARCA4 mutations [40]. An open question is whether the different embryological origin of meningiomas affects the sensitivity to drugs [41].
In a small proportion of patients, meningiomas arise as multiple and spatially distinct lesions and not as solitary tumors [42]. Multiple meningiomas may be associated with familial syndromes, such as neurofibromatosis type 2 (NF2) and familial meningiomatosis in patients with germline NF2 and SMARCB1 mutations [43]; however, Juratli et al. have reported a significantly lower frequency of NF2 mutations in a series of 17 multiple meningiomas. All patients, with the exception of two cases, expressed TRAF7, AKT1, SMO, or PIK3CA mutations. In particular, the most frequent driver mutations were TRAF7 (n  =  5); PIK3CA, H1047R, and E545G (n  =  3); AKT1 E17K (n  =  3); NF2 (n  =  2); SMO L412F (n = 1); and NF1 (n = 1), and one patient only did not harbor any driver mutation. Interestingly, the same mutation was not detected in different tumors from the same patient, suggesting genomically distinct molecular drivers and an independent origin of multiple meningiomas [44].

4. Molecular Features and Correlation with Prognosis

TERT promoter mutations have been correlated with shorter, progression-free survival in a retrospective series of 252 meningiomas, with 10.1 months in patients with TERT promoter mutations, compared with 179 months in patients without a TERT promoter mutations regardless of the histological grading [25]. Other studies have demonstrated the negative prognostic role of TERT mutations regardless of the WHO grading, suggesting that TERT-mutated meningiomas should be followed carefully, or treated aggressively, and include TERT analysis in the routine diagnostic assessment [45,46]. Furthermore, the loss of function of dystrophin-encoding and muscular dystrophy-associated gene (DMD) has been considered an additional negative prognostic factor in TERT-mutated meningiomas [47]. SMO- and AKT-1-mutated meningiomas have shown to recur more frequently, compared with meningiomas lacking SMO and AKT1 mutations [37]. Furthermore, AKT1 p.Glu17Lys mutation confers a reduced time to tumor recurrence [20]. Conversely, a larger study on 3031 meningioma samples from 514 individual cases has shown that TRAF7, AKT1, and/or KLF4 mutations were significantly associated with a lower risk of progression [48].
DNA methylation analyses distinguish six clinically different meningioma groups—three with favorable outcomes, two with intermediate outcomes, and one with a poor outcome—representing a new approach for decisions regarding postoperative therapeutic interventions, in particular, whether to treat patients with adjuvant radiotherapy versus observation alone [29,30,31]. Recently, Berghoff et al. have investigated meningioma-relevant mutations and their correlation with DNA methylation clusters and patient survival. TRAKL pattern (any of the following mutations: TRAF7, AKT1, and KLF4) was predominantly found in methylation classes with favorable outcomes, while NF2 was associated with methylation classes with poor outcomes. TRAF7, KLF4, and TRAKL mutation genotypes were associated with improved progression-free survival (PFS) and overall survival (OS), whereas TERT promoter methylation, and intermediate- and poor-outcome methylation classes, were associated with impaired PFS and OS. Methylation clustering showed better prognostic discrimination for PFS and OS than each of the individual mutations, where TERT mutation remained the unique independent significant prognostic factor for PFS in multivariable analysis [49]. Loss of H3K27me3 has been reported as a prognostically unfavorable alteration in meningiomas: 13.9% (21/151) of meningiomas displayed the H3K27me3 loss by immunohistochemistry (IHC) in a multicenter study and identified a subset of WHO grades 1 and 2 meningiomas with increased risk of recurrence [50]. In Table 1, a summary of correlations of WHO grading with histology, molecular alterations, methylation classes, prognosis, and location of meningiomas is presented.

5. Driver Signaling Mutations in Meningiomas: Potential New Targets of Therapy

Different growth factor receptors and kinases may promote the meningioma growth, including epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor β (PDGFRβ), vascular endothelial growth factor receptor (VEGFR), and insulin-like growth factor receptor (IGFR) [41]. PDGFR, EGFR, and VEGFR may have a dual activity on the RAS–RAF–MEK–MAPK or FAK–PI3K–AKT pathway, resulting in growth-favoring signals in meningiomas. Moreover, after the activation of AKT, an intracellular signaling pathway acts on mTORC1 and 2 and regulates DNA replication. Other signaling pathways shown to be activated in meningiomas are the phospholipase A2–arachidonic acid–cyclooxygenase pathway [51], the phospholipase C γ1 (PLCγ1)–protein kinase C pathway (PKC) [52], and the transforming growth factor-β (TGFβ)–SMAD signaling pathway [53,54], which act as inhibitory mechanisms of meningioma growth, thus representing potential targets of treatment.

6. Role of Tumor Microenvironment in Meningiomas: Is It Druggable?

The meningioma microenvironment seems to play a role in tumor growth, and some data suggest that WHO grades 2 and 3 meningiomas represent a relatively immunosuppressed status. In particular, grading in meningioma negatively correlates with the amount of CD4+, CD8+, and PD-1+ lymphocytes, along with increased numbers of Treg (FOXP3+) cells in the tumor. Moreover, programmed death-ligand 1 (PD-L1) expression has been correlated with the grading of meningiomas, with PD-L1 protein detection in 40% of grade 1, 60% of grade 2, and 77–88% of grade 3 meningiomas [55]. Karimi et al. reported that PD-L1 protein expression had a patchy pattern, along with peri-vascular and peri-necrotic, membranous, and cytoplasmic immunoreactivity in both tumor and immune cells of the microenvironment [56]. Furthermore, PD-L1 expression was confined to a small subpopulation of cells (median  <  1% of cells, range 0–20% of cells), and correlated with a higher risk of early recurrence, regardless of grading, the extent of resection, and tumor diameter. Tumor-infiltrating T lymphocytes (TILs) around meningiomas may influence the prognosis. Rapp et al. analyzed the presence of TILs in 97 newly diagnosed and 62 recurrent high-grade meningiomas, reporting that a higher number of cytotoxic TILs (CD3+ CD8+ FOXP3-) were associated with an improved PFS, while recurrent meningiomas were characterized by lower numbers of TILs and proportions of PD-1+CD8+ T cells [57]. Some findings support the hypothesis that somatic genetic alterations in meningioma can potentially affect PD-L1 or other checkpoint protein expression. For instance, an increased PD-L1 expression was found in TRAF7-mutated, compared with wild-type skull-base meningiomas [58], and an increased number of CTLA4+/CD3+ lymphocytes was found in grades 2 and 3 meningiomas harboring the PI3K–AKT–mTOR pathway or SMO mutations [59]. Lastly, 40% of NF2-mutated meningiomas express PD-L1 in the surrounding microenvironment [60], suggesting that the therapeutic role of checkpoint inhibitors is worth investigating in progressive/refractory meningiomas after the failure of surgery and/or radiation therapy [61].
As M2 macrophages are the most prevalent immune cell type in meningiomas, Yeung et al. have targeted with a specific monoclonal antibody the colony-stimulating factor 1 (CSF1) and CFS1 receptor (CSF1R) expressed in myeloid cells, reporting a significant reduction in tumor growth in a murine meningioma model. This provides a strong rationale for future human clinical trials targeting the CSF1–CSF1R pathway in malignant meningiomas [62].

7. Systemic Therapy for Progressive/Recurrent Meningiomas: Present and Future

Patients with progressive/recurrent meningiomas, in whom surgery and/or radiation are not feasible anymore, have a limited PFS, ranging from 6-month PFS of 29% for grade 1 to 26% for grades 2 and 3 tumors [63]. To date, there is no evidence regarding the standard of care, and enrollment in clinical trials is recommended in case of disease progression [2]. A matter of debate was the choice of the best endpoint in clinical trials in surgery- and radiation-refractory meningioma: in 2019, the Response Assessment in Neuro-Oncology (RANO) group stated that an appropriate endpoint for medical therapy trials is either a 6-month PFS rate alone or in combination with radiological response [64].
Different cytotoxic chemotherapies and targeted agents have been investigated, with poor results in terms of disease control and survival, including hydroxyurea [65,66,67], temozolomide [68], irinotecan [69], trabectedin [70,71], IFN-α [72,73,74], somatostatin analogs (pasireotide [75] and octreotide [76,77]), VEGF/VEGFR inhibitors [78,79,80,81], EGFR inhibitors (erlotinib and gefitinib) [82], imatinib [83], and mifepristone [84] (Table 2).
As meningiomas are highly vascularized, anti-VEGF drugs have been largely investigated. The most employed compound was bevacizumab, which has displayed some benefit in terms of PFS (median-PFS 16.8 months, range 6.5–22 months; 6-month PFS: 73%, range 44%–93%), with particular advantage in patients with high-grade and/or multiple and/or radiation-induced meningiomas [86]. The clinical and radiological benefit of bevacizumab may derive from a pronounced inhibitory effect on tumor growth, as well as some anti-edema activity, in comparison with other targeted therapies and cytotoxic agents [87]. Given these favorable properties, bevacizumab was investigated in combination with other compounds. Shih et al. evaluated the activity of bevacizumab and the mTORC1 inhibitor everolimus; the authors reported the best radiological response for stable disease (SD) in 15 patients (88%), and 6 of these patients had SD for >12 months. Median PFS was 22 months (95% CI 4.5–26.8) and was longer for patients with grades 2 and 3 than for those with grade 1 meningiomas (22.0 months vs. 17.5 months, respectively) [79].
The Combination of Everolimus and Octreotide LAR in Aggressive Recurrent Meningiomas (CEVOREM) phase 2 trial reported a 6-month PFS of 55% (95% CI 31.3%–73.5%), and 6-month- and 12-month OS of 90% (95% CI 65.6%–97.4%) and 75% (95% CI, 50.0%–88.7%), respectively, in 20 patients with progressive meningiomas. A radiological response (decrease >50%) was achieved in 78% of patients, with a median tumor growth rate decreasing from 16.6% 3 months before inclusion to 0.02% after 3 months and 0.48% at 6 months after treatment [85].
In the era of precision medicine, we may select appropriate therapy based on specific genetic mutations. In this regard, the Alliance/NCI A071401 study initiated a genomically driven meningioma phase 2 trial in which the targeted therapy is delivered according to the mutation found in the tissue. Thus, different compounds are under investigation, including the SMO inhibitor vismodegib in SMO-mutant tumors, the AKT inhibitor capivasertib (AZD5363) for AKT/PIK3CA-mutant tumors, the CDK inhibitor abemaciclib for NF2 or CDK-mutant tumors, and the FAK inhibitor GSK2256098 for SMO/PTCH1-mutant and NF2-mutant meningiomas, respectively (NCT02523014). The FAK inhibitor arm has already completed the accrual with 37 patients enrolled (12 grade 1 and 25 grades 2 and 3 meningiomas) in the trial [88]. Most patients received prior radiotherapy (75.7%) and chemotherapy (40.5%) before the start of the FAK inhibitor. One patient had a partial response, and 24 had SD as the best response to treatment. In grade 1 meningiomas, the 6-month PFS was 83% (10/12 patients; 95% CI: 52–98%). In grades 2 and 3 meningiomas, the 6–month PFS was 33% (8/24 patients; 95% CI: 16–55%). The study met the 6-month PFS endpoint both for the grade 1 and the grades 2 and 3 cohorts with excellent tolerability. However, a major concern for all targeted therapies is the resistance owing to tumor heterogeneity. Indeed, malignant meningiomas have been shown significant molecular heterogeneity within the original tumor and recurrence [89].
High-grade meningiomas may harbor an immunosuppressive microenvironment. In this regard, some studies reported that a subset of high-grade meningiomas have a high somatic mutation burden [90] that could represent a predisposing factor for a response to immune-checkpoint inhibitors [55,91]. Brastianos et al. have designed a phase 2 trial evaluating pembrolizumab in 26 patients with recurrent high-grade meningiomas (23 grade 2, and 3 grade 3 tumors). The study met the primary endpoint and achieved a 6-month PFS of 48% (90% CI 31–66), a median PFS of 7.6 months (90% CI 3.4–12.9 months), and a median OS of 20.2 months (90% CI 14.8–25.8 months). Eighteen patients had SD as the best radiological response, while no patients had complete or partial response according to RANO criteria. PD-L1 expression in pretreatment tissue was not correlated with outcome. Notably, the trial enrolled seven patients with metastatic extracranial meningiomas, of whom four achieved 6-month PFS and one patient had PFS lasting for nearly 20 months. While the trial met the primary endpoint, these results will require additional validation, and further studies are needed to identify which meningioma subtypes or tumor microenvironment patterns are correlated with the efficacy of immune-checkpoint inhibitors [92]. Conversely, a phase 2 trial on 20 patients using anti-PD1 nivolumab failed to improve 6-month PFS (42.4%, 95% CI 22.8, 60.7), although a subset of patients appeared to derive benefit (one patient obtained a partial response) [93].
Novel targeted agents are under investigation in clinical trials, including MEK inhibitor alone (selumetinib, NCT03095248) or in combination with Pi3Kα inhibitor (alpelisib, NCT03631953), VEGF inhibitor apatinib (NCT04501705), CDK-p16-Rb inhibitor ribociclib (NCT02933736), immune checkpoint inhibitors, such as nivolumab alone or in combination with ipilimumab (NCT02648997) or with stereotactic radiosurgery (NCT03604978), or sintilimab (NCT04728568) (Table 3).

8. Conclusions

Despite multiple studies on different cytotoxic agents performed on recurrent meningiomas in the past several years, limited evidence of efficacy and clinical benefit has been reported thus far. Hence, there is no evidence of effective systemic therapy for meningiomas. Since 2013, a genomic revolution in the biology and genomic landscape of meningiomas is underway, where the identification of molecular alterations driving the aggressiveness is translated into more reliable preclinical models that allow for rapid translation of discoveries into clinical trials. These key molecular alterations are refining the histological and molecular classification of meningiomas and allow for the stratification of patients with different outcomes and tailored treatments.

Author Contributions

Conceptualization, A.P.; data curation, A.P., F.B. and L.B.; writing—original draft preparation, A.P.; writing—review and editing, A.P., F.B., R.P., E.P., L.B. and R.R.; supervision, R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

A.P. declares no conflicts of interest; F.B. declares no conflicts of interest; R.P. declares no conflicts of interest; E.P. declares no conflicts of interest; L.B. declares no conflicts of interest; R.S declares the following financial disclosure: AstraZeneca, Merck and Orbus e Agios Therapeutics.; R.R. declares the following financial disclosure: UCB, Mundipharma, Bayer, and Novocure.

References

  1. Ostrom, Q.T.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2014–2018. Neuro-Oncology 2021, 23, iii1–iii105. [Google Scholar] [CrossRef] [PubMed]
  2. Goldbrunner, R.; Stavrinou, P.; Jenkinson, M.D.; Sahm, F.; Mawrin, C.; Weber, D.C.; Preusser, M.; Minniti, G.; Lund-Johansen, M.; Lefranc, F.; et al. EANO guideline on the diagnosis and management of meningiomas. Neuro-Oncology 2021, 23, 1821–1834. [Google Scholar] [CrossRef] [PubMed]
  3. Nakasu, S.; Fukami, T.; Jito, J.; Matsuda, M. Microscopic anatomy of the brain–meningioma interface. Brain Tumor Pathol. 2005, 22, 53–57. [Google Scholar] [CrossRef] [PubMed]
  4. Kshettry, V.R.; Ostrom, Q.; Kruchko, C.; Al-Mefty, O.; Barnett, G.H.; Barnholtz-Sloan, J.S. Descriptive epidemiology of World Health Organization grades II and III intracranial meningiomas in the United States. Neuro-Oncology 2015, 17, 1166–1173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Nowosielski, M.; Galldiks, N.; Iglseder, S.; Kickingereder, P.; von Deimling, A.; Bendszus, M.; Wick, W.; Sahm, F. Diagnostic challenges in meningioma. Neuro-Oncology 2017, 19, 1588–1598. [Google Scholar] [CrossRef]
  6. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro-Oncology 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed]
  7. Rogers, L.; Barani, I.; Chamberlain, M.; Kaley, T.J.; McDermott, M.; Raizer, J.; Schiff, D.; Weber, D.C.; Wen, P.Y.; Vogelbaum, M.A. Meningiomas: Knowledge base, treatment outcomes, and uncertainties. A RANO review. J. Neurosurg. 2015, 122, 4–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Brastianos, P.K.; Horowitz, P.M.; Santagata, S.; Jones, R.T.; McKenna, A.; Getz, G.; Ligon, K.L.; Palescandolo, E.; Van Hummelen, P.; Ducar, M.D.; et al. Genomic sequencing of meningiomas identifies oncogenic SMO and AKT1 mutations. Nat. Genet. 2013, 45, 285–289. [Google Scholar] [CrossRef]
  9. Zang, K.D. Meningioma: A cytogenetic model of a complex benign human tumor, including data on 394 karyotyped cases. Cytogenet. Genome Res. 2001, 93, 207–220. [Google Scholar] [CrossRef]
  10. Mawrin, C.; Perry, A. Pathological classification and molecular genetics of meningiomas. J. Neuro-Oncol. 2010, 99, 379–391. [Google Scholar] [CrossRef]
  11. Petrilli, A.M.; Fernández-Valle, C. Role of Merlin/NF2 inactivation in tumor biology. Oncogene 2015, 35, 537–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ruttledge, M.H.; Sarrazin, J.; Rangaratnam, S.; Phelan, C.M.; Twist, E.; Merel, P.; Delattre, O.; Thomas, G.; Nordenskjöld, M.; Collins, V.P.; et al. Evidence for the complete inactivation of the NF2 gene in the majority of sporadic meningiomas. Nat. Genet. 1994, 6, 180–184. [Google Scholar] [CrossRef] [PubMed]
  13. Wellenreuther, R.; Kraus, J.A.; Lenartz, D.; Menon, A.G.; Schramm, J.; Louis, D.N.; Ramesh, V.; Gusella, J.F.; Wiestler, O.D.; Von Deimling, A. Analysis of the neurofibromatosis 2 gene reveals molecular variants of meningioma. Am. J. Pathol. 1995, 146, 827–832. [Google Scholar] [PubMed]
  14. Hartmann, C.; Sieberns, J.; Gehlhaar, C.; Simon, M.; Paulus, W.; von Deimling, A. NF2 Mutations in Secretory and Other Rare Variants of Meningiomas. Brain Pathol. 2006, 16, 15–19. [Google Scholar] [CrossRef] [PubMed]
  15. Goutagny, S.; Yang, H.W.; Zucman-Rossi, J.; Chan, J.; Dreyfuss, J.M.; Park, P.J.; Black, P.M.; Giovannini, M.; Carroll, R.S.; Kalamarides, M. Genomic Profiling Reveals Alternative Genetic Pathways of Meningioma Malignant Progression Dependent on the Underlying NF2 Status. Clin. Cancer Res. 2010, 16, 4155–4164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. James, M.F.; Han, S.; Polizzano, C.; Plotkin, S.R.; Manning, B.D.; Stemmer-Rachamimov, A.O.; Gusella, J.F.; Ramesh, V. NF2/merlin is a novel negative regulator of mTOR complex 1, and activation of mTORC1 is associated with meningioma and schwannoma growth. Mol. Cell. Biol. 2009, 29, 4250–4261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. James, M.F.; Stivison, E.; Beauchamp, R.; Han, S.; Li, H.; Wallace, M.R.; Gusella, J.F.; Stemmer-Rachamimov, A.O.; Ramesh, V. Regulation of mTOR Complex 2 Signaling in Neurofibromatosis 2–Deficient Target Cell Types. Mol. Cancer Res. 2012, 10, 649–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Clark, V.E.; Erson-Omay, E.Z.; Serin, A.; Yin, J.; Cotney, J.; Ozduman, K.; Avşar, T.; Li, J.; Murray, P.B.; Henegariu, O.; et al. Genomic analysis of non-NF2 meningiomas reveals mutations in TRAF7, KLF4, AKT1, and SMO. Science 2013, 339, 1077–1080. [Google Scholar] [CrossRef] [Green Version]
  19. Abedalthagafi, M.; Bi, W.L.; Aizer, A.A.; Merrill, P.H.; Brewster, R.; Agarwalla, P.K.; Listewnik, M.L.; Dias-Santagata, D.; Thorner, A.R.; Van Hummelen, P.; et al. Oncogenic PI3K mutations are as common as AKT1 and SMO mutations in meningioma. Neuro Oncol. 2016, 18, 649–655. [Google Scholar] [CrossRef] [Green Version]
  20. Yesilöz, Ü.; Kirches, E.; Hartmann, C.; Scholz, J.; Kropf, S.; Sahm, F.; Nakamura, M.; Mawrin, C. Frequent AKT1E17K mutations in skull base meningiomas are associated with mTOR and ERK1/2 activation and reduced time to tumor recurrence. Neuro Oncol. 2017, 19, 1088–1096. [Google Scholar] [CrossRef]
  21. Reuss, D.E.; Piro, R.M.; Jones, D.T.; Simon, M.; Ketter, R.; Kool, M.; Becker, A.; Sahm, F.; Pusch, S.; Meyer, J.; et al. Secretory meningiomas are defined by combined KLF4 K409Q and TRAF7 mutations. Acta Neuropathol. 2013, 125, 351–358. [Google Scholar] [CrossRef] [PubMed]
  22. Ng, J.M.; Curran, T. The Hedgehog’s tale: Developing strategies for targeting cancer. Nat. Rev. Cancer 2011, 11, 493–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Clark, V.E.; Harmancı, A.S.; Bai, H.; Youngblood, M.W.; Lee, T.I.; Baranoski, J.F.; Ercan-Sencicek, A.G.; Abraham, B.J.; Weintraub, A.S.; Hnisz, D.; et al. Recurrent somatic mutations in POLR2A define a distinct subset of meningiomas. Nat. Genet. 2016, 48, 1253–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Harmancı, A.S.; Youngblood, M.W.; Clark, V.E.; Coskun, S.; Henegariu, O.; Duran, D.; Erson-Omay, E.Z.; Kaulen, L.D.; Lee, T.I.; Abraham, B.J.; et al. Integrated genomic analyses of de novo pathways underlying atypical meningiomas. Nat. Commun. 2017, 8, 14433. [Google Scholar] [CrossRef]
  25. Sahm, F.; Schrimpf, D.; Olar, A.; Koelsche, C.; Reuss, D.; Bissel, J.; Kratz, A.; Capper, D.; Schefzyk, S.; Hielscher, T.; et al. TERT Promoter Mutations and Risk of Recurrence in Meningioma. J. Natl. Cancer Inst. 2016, 108, djv370. [Google Scholar] [CrossRef]
  26. Vaubel, R.A.; Chen, S.G.; Raleigh, D.R.; Link, M.J.; Chicoine, M.R.; Barani, I.; Jenkins, S.M.; Aleff, P.A.; Rodriguez, F.J.; Burger, P.C.; et al. Meningiomas With Rhabdoid Features Lacking Other Histologic Features of Malignancy: A Study of 44 Cases and Review of the Literature. J. Neuropathol. Exp. Neurol. 2016, 75, 44–52. [Google Scholar] [CrossRef] [Green Version]
  27. Shankar, G.M.; Abedalthagafi, M.; Vaubel, R.A.; Merrill, P.H.; Nayyar, N.; Gill, C.M.; Brewster, R.; Bi, W.L.; Agarwalla, P.K.; Thorner, A.R.; et al. Germline and somatic BAP1 mutations in high-grade rhabdoid meningiomas. Neuro-Oncology 2017, 19, 535–545. [Google Scholar] [CrossRef] [Green Version]
  28. Williams, E.A.; Santagata, S.; Wakimoto, H.; Shankar, G.M.; Barker, F.G., II; Sharaf, R.; Reddy, A.; Spear, P.; Alexander, B.M.; Ross, J.S.; et al. Distinct genomic subclasses of high-grade/progressive meningiomas: NF2-associated, NF2-exclusive, and NF2-agnostic. Acta Neuropathol. Commun. 2020, 8, 171. [Google Scholar] [CrossRef]
  29. Sahm, F.; Schrimpf, D.; Stichel, D.; Jones, D.T.W.; Hielscher, T.; Schefzyk, S.; Okonechnikov, K.; Koelsche, C.; Reuss, D.E.; Capper, D.; et al. DNA methylation-based classification and grading system for meningioma: A multicentre, retrospective analysis. Lancet Oncol. 2017, 18, 682–694. [Google Scholar] [CrossRef] [Green Version]
  30. Olar, A.; Wani, K.M.; Wilson, C.D.; Zadeh, G.; Demonte, F.; Jones, D.T.; Pfister, S.M.; Sulman, E.P.; Aldape, K.D. Global epigenetic profiling identifies methylation subgroups associated with recurrence-free survival in meningioma. Acta Neuropathol. 2017, 133, 431–444. [Google Scholar] [CrossRef] [Green Version]
  31. Nassiri, F.; Mamatjan, Y.; Suppiah, S.; Badhiwala, J.H.; Mansouri, S.; Karimi, S.; Saarela, O.; Poisson, L.; Gepfner-Tuma, I.; Schittenhelm, J.; et al. DNA methylation profiling to predict recurrence risk in meningioma: Development and validation of a nomogram to optimize clinical management. Neuro-Oncology 2019, 21, 901–910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kros, J.; de Greve, K.; van Tilborg, A.; Hop, W.; Pieterman, H.; Avezaat, C.; Deprez, R.L.D.; Zwarthoff, E. NF2 status of meningiomas is associated with tumour localization and histology. J. Pathol. 2001, 194, 367–372. [Google Scholar] [CrossRef] [PubMed]
  33. Lee, J.H.; Sade, B.; Choi, E.; Golubic, M.; Prayson, R. Meningothelioma as the predominant histological subtype of midline skull base and spinal meningioma. J. Neurosurg. 2006, 105, 60–64. [Google Scholar] [CrossRef] [PubMed]
  34. Ketter, R.; Rahnenführer, J.; Henn, W.; Kim, Y.J.; Feiden, W.; Steudel, W.I.; Zang, K.D.; Urbschat, S. Correspondence of tumor localization with tumor recurrence and cytogenetic progression in meningiomas. Neurosurgery 2008, 62, 61–70. [Google Scholar] [CrossRef] [Green Version]
  35. Strickland, M.R.; Gill, C.M.; Nayyar, N.; D’Andrea, M.R.; Thiede, C.; Juratli, T.A.; Schackert, G.; Borger, D.R.; Santagata, S.; Frosch, M.P.; et al. Targeted sequencing of SMO and AKT1 in anterior skull base meningiomas. J. Neurosurg. 2017, 127, 438–444. [Google Scholar] [CrossRef] [Green Version]
  36. Williams, S.R.; Juratli, T.A.; Castro, B.A.; Lazaro, T.T.; Gill, C.M.; Nayyar, N.; Strickland, M.R.; Babinski, M.; Johnstone, S.E.; Frosch, M.P.; et al. Genomic Analysis of Posterior Fossa Meningioma Demonstrates Frequent AKT1 E17K Mutations in Foramen Magnum Meningiomas. J. Neurol. Surg B. Skull Base 2019, 80, 562–567. [Google Scholar] [CrossRef]
  37. Boetto, J.; Bielle, F.; Sanson, M.; Peyre, M.; Kalamarides, M. SMO mutation status defines a distinct and frequent molecular subgroup in olfactory groove meningiomas. Neuro Oncol. 2017, 19, 345–351. [Google Scholar]
  38. Smith, M.J.; O’Sullivan, J.; Bhaskar, S.S.; Hadfield, K.D.; Poke, G.; Caird, J.; Sharif, S.; Eccles, D.; FitzPatrick, D.; Rawluk, D.; et al. Loss-of-function mutations in SMARCE1 cause an inherited disorder of multiple spinal meningiomas. Nat. Genet. 2013, 45, 295–298. [Google Scholar] [CrossRef]
  39. Smith, M.J.; Wallace, A.J.; Bennett, C.; Hasselblatt, M.; Elert-Dobkowska, E.; Evans, L.T.; Hickey, W.F.; van Hoff, J.; Bauer, D.; Lee, A.; et al. GermlineSMARCE1mutations predispose to both spinal and cranial clear cell meningiomas. J. Pathol. 2014, 234, 436–440. [Google Scholar] [CrossRef]
  40. Jungwirth, G.; Warta, R.; Beynon, C.; Sahm, F.; Von Deimling, A.; Unterberg, A.; Herold-Mende, C.; Jungk, C. Intraventricular meningiomas frequently harbor NF2 mutations but lack common genetic alterations in TRAF7, AKT1, SMO, KLF4, PIK3CA, and TERT. Acta Neuropathol. Commun. 2019, 7, 140. [Google Scholar] [CrossRef] [Green Version]
  41. Preusser, M.; Brastianos, P.K.; Mawrin, C. Advances in meningioma genetics: Novel therapeutic opportunities. Nat. Rev. Neurol. 2018, 14, 106–115. [Google Scholar] [CrossRef] [PubMed]
  42. Tsermoulas, G.; Turel, M.K.; Wilcox, J.T.; Shultz, D.; Farb, R.; Zadeh, G.; Bernstein, M. Management of multiple meningiomas. J. Neurosurg. 2018, 128, 1403–1409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Shen, Y.; Nunes, F.; Stemmer-Rachamimov, A.; James, M.; Mohapatra, G.; Plotkin, S.; Betensky, R.A.; Engler, D.A.; Roy, J.; Ramesh, V.; et al. Genomic profiling distinguishes familial multiple and sporadic multiple meningiomas. BMC Med. Genom. 2009, 2, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Juratli, T.A.; Prilop, I.; Saalfeld, F.C.; Herold, S.; Meinhardt, M.; Wenzel, C.; Zeugner, S.; Aust, D.E.; Barker, F.G.; Cahill, D.P.; et al. Sporadic multiple meningiomas harbor distinct driver mutations. Acta Neuropathol. Commun. 2021, 9, 8. [Google Scholar] [CrossRef]
  45. Mirian, C.; Duun-Henriksen, A.K.; Juratli, T.; Sahm, F.; Spiegl-Kreinecker, S.; Peyre, M.; Biczok, A.; Tonn, J.-C.; Goutagny, S.; Bertero, L.; et al. Poor prognosis associated with TERT gene alterations in meningioma is independent of the WHO classification: An individual patient data meta-analysis. J. Neurol. Neurosurg. Psychiatry 2020, 91, 378–387. [Google Scholar] [CrossRef]
  46. Mirian, C.; Grell, K.; Juratli, T.A.; Sahm, F.; Spiegl-Kreinecker, S.; Peyre, M.; Biczok, A.; Tonn, J.C.; Goutagny, S.; Bertero, L.; et al. Implementation of TERT promoter mutations improve prognostication of the WHO classification in meningioma. Neuropathol. Appl. Neurobiol. 2022, 48, e12773. [Google Scholar] [CrossRef]
  47. Juratli, T.A.; McCabe, D.; Nayyar, N.; Williams, E.A.; Silverman, I.M.; Tummala, S.S.; Fink, A.L.; Baig, A.; Martinez-Lage, M.; Selig, M.K.; et al. DMD genomic deletions characterize a subset of progressive/higher-grade meningiomas with poor outcome. Acta Neuropathol. 2018, 136, 779–792. [Google Scholar] [CrossRef]
  48. Maas, S.L.N.; Stichel, D.; Hielscher, T.; Sievers, P.; Berghoff, A.S.; Schrimpf, D.; Sill, M.; Euskirchen, P.; Blume, C.; Patel, A.; et al. Integrated Molecular-Morphologic Meningioma Classification: A Multicenter Retrospective Analysis, Retrospectively and Prospectively Validated. J. Clin. Oncol. 2021, 39, 3839–3852. [Google Scholar] [CrossRef]
  49. Berghoff, A.S.; Hielscher, T.; Ricken, G.; Furtner, J.; Schrimpf, D.; Widhalm, G.; Rajky, U.; Marosi, C.; Hainfellner, J.A.; Deimling, A.; et al. Prognostic impact of genetic alterations and methylation classes in meningioma. Brain Pathol. 2022, 32, e12970. [Google Scholar] [CrossRef]
  50. Nassiri, F.; Wang, J.Z.; Singh, O.; Karimi, S.; Dalcourt, T.; Ijad, N.; Pirouzmand, N.; Ng, H.K.; Saladino, A.; Pollo, B.; et al. International Consortium on Meningiomas. Loss of H3K27me3 in meningiomas. Neuro Oncol. 2021, 23, 1282–1291. [Google Scholar] [CrossRef]
  51. Kang, H.-C.; Kim, I.H.; Park, C.I.; Park, S.-H. Immunohistochemical analysis of cyclooxygenase-2 and brain fatty acid binding protein expression in grades I-II meningiomas: Correlation with tumor grade and clinical outcome after radiotherapy. Neuropathology 2014, 34, 446–454. [Google Scholar] [CrossRef] [PubMed]
  52. Johnson, M.D.; Horiba, M.; Winnier, A.R.; Arteaga, C.L. The epidermal growth factor receptor is associated with phospholipase C-gamma 1 in meningiomas. Hum. Pathol. 1994, 25, 146–153. [Google Scholar] [CrossRef]
  53. Johnson, M.D.; Shaw, A.K.; O’Connell, M.J.; Sim, F.J.; Moses, H.L. Analysis of transforming growth factor β receptor expression and signaling in higher grade meningiomas. J. Neuro-Oncol. 2010, 103, 277–285. [Google Scholar] [CrossRef] [PubMed]
  54. Johnson, M.D.; O’Connell, M.J.; Vito, F.; Pilcher, W. Bone Morphogenetic Protein 4 and Its Receptors Are Expressed in the Leptomeninges and Meningiomas and Signal via the Smad Pathway. J. Neuropathol. Exp. Neurol. 2009, 68, 1177–1183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Du, Z.; Abedalthagafi, M.; Aizer, A.A.; McHenry, A.R.; Sun, H.H.; Bray, M.-A.; Viramontes, O.; Machaidze, R.; Brastianos, P.K.; Reardon, D.A.; et al. Increased expression of the immune modulatory molecule PD-L1 (CD274) in anaplastic meningioma. Oncotarget 2014, 6, 4704–4716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Karimi, S.; Mansouri, S.; Mamatjan, Y.; Liu, J.; Nassiri, F.; Suppiah, S.; Singh, O.; Aldape, K.; Zadeh, G. Programmed death ligand-1 (PD-L1) expression in meningioma; prognostic significance and its association with hypoxia and NFKB2 expression. Sci. Rep. 2020, 10, 14115. [Google Scholar] [CrossRef]
  57. Rapp, C.; Dettling, S.; Liu, F.; Ull, A.T.; Warta, R.; Jungk, C.; Roesch, S.; Mock, A.; Sahm, F.; Schmidt, M.; et al. Cytotoxic T Cells and their Activation Status are Independent Prognostic Markers in Meningiomas. Clin. Cancer Res. 2019, 25, 5260–5270. [Google Scholar] [CrossRef]
  58. Hao, S.; Huang, G.; Feng, J.; Li, D.; Wang, K.; Wang, L.; Wu, Z.; Wan, H.; Zhang, L.; Zhang, J. Non-NF2 mutations have a key effect on inhibitory immune checkpoints and tumor pathogenesis in skull base meningiomas. J. Neuro-Oncol. 2019, 144, 11–20. [Google Scholar] [CrossRef]
  59. Proctor, D.T.; Patel, Z.; Lama, S.; Resch, L.; van Marle, G.; Sutherland, G.R. Identification of PD-L2, B7-H3 and CTLA-4 immune checkpoint proteins in genetic subtypes of meningioma. OncoImmunology 2018, 8, e1512943. [Google Scholar] [CrossRef] [Green Version]
  60. Wang, S.; Liechty, B.; Patel, S.; Weber, J.S.; Hollmann, T.J.; Snuderl, M.; Karajannis, M.A. Programmed death ligand 1 expression and tumor infiltrating lymphocytes in neurofibromatosis type 1 and 2 associated tumors. J. Neuro-Oncol. 2018, 138, 183–190. [Google Scholar] [CrossRef]
  61. Karimi, S.; Mansouri, S.; Nassiri, F.; Bunda, S.; Singh, O.; Brastianos, P.K.; Dunn, I.F.; Zadeh, G. Clinical significance of checkpoint regulator “Programmed death ligand-1 (PD-L1)” expression in meningioma: Review of the current status. J. Neuro-Oncol. 2021, 151, 443–449. [Google Scholar] [CrossRef] [PubMed]
  62. Yeung, J.; Yaghoobi, V.; Miyagishima, D.; Vesely, M.D.; Zhang, T.; Badri, T.; Nassar, A.; Han, X.; Sanmamed, M.F.; Youngblood, M.; et al. Targeting the CSF1/CSF1R axis is a potential treatment strategy for malignant meningiomas. Neuro-Oncology 2021, 23, 1922–1935. [Google Scholar] [CrossRef] [PubMed]
  63. Kaley, T.; Barani, I.; Chamberlain, M.; McDermott, M.; Panageas, K.; Raizer, J.; Rogers, L.; Schiff, D.; Vogelbaum, M.; Weber, D.; et al. Historical benchmarks for medical therapy trials in surgery- and radiation-refractory meningioma: A RANO review. Neuro-Oncology 2014, 16, 829–840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Huang, R.Y.; Bi, W.L.; Weller, M.; Kaley, T.; Blakeley, J.; Dunn, I.; Galanis, E.; Preusser, M.; McDermott, M.; Rogers, L.; et al. Proposed response assessment and endpoints for meningioma clinical trials: Report from the Response Assessment in Neuro-Oncology Working Group. Neuro-Oncology 2019, 21, 26–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Chamberlain, M.C.; Johnston, S.K. Hydroxyurea for recurrent surgery and radiation refractory meningioma: A retrospective case series. J. Neuro-Oncol. 2011, 104, 765–771. [Google Scholar] [CrossRef]
  66. Chamberlain, M.C. Hydroxyurea for recurrent surgery and radiation refractory high-grade meningioma. J. Neuro-Oncol. 2012, 107, 315–321. [Google Scholar] [CrossRef]
  67. Mazza, E.; Brandes, A.; Zanon, S.; Eoli, M.; Lombardi, G.; Faedi, M.; Franceschi, E.; Reni, M. Hydroxyurea with or without imatinib in the treatment of recurrent or progressive meningiomas: A randomized phase II trial by Gruppo Italiano Cooperativo di Neuro-Oncologia (GICNO). Cancer Chemother. Pharmacol. 2016, 77, 115–120. [Google Scholar] [CrossRef]
  68. Chamberlain, M.C.; Tsao-Wei, D.D.; Groshen, S. Temozolomide for treatment-resistant recurrent meningioma. Neurology 2004, 62, 1210–1212. [Google Scholar] [CrossRef]
  69. Chamberlain, M.C.; Tsao-Wei, D.D.; Groshen, S. Salvage chemotherapy With CPT-11 for recurrent meningioma. J. Neuro-Oncol. 2006, 78, 271–276. [Google Scholar] [CrossRef]
  70. Preusser, M.; Spiegl-Kreinecker, S.; Lötsch, D.; Wöhrer, A.; Schmook, M.; Dieckmann, K.; Saringer, W.; Marosi, C.; Berger, W. Trabectedin has promising antineoplastic activity in high-grade meningioma. Cancer 2012, 118, 5038–5049. [Google Scholar] [CrossRef] [Green Version]
  71. Preusser, M.; Silvani, A.; Le Rhun, E.; Soffietti, R.; Lombardi, G.; Sepulveda, J.M.; Brandal, P.; Brazil, L.; Bonneville-Levard, A.; Lorgis, V.; et al. Trabectedin for recurrent WHO grade 2 or 3 meningioma: A randomized phase II study of the EORTC Brain Tumor Group (EORTC-1320-BTG). Neuro-Oncology 2021, noab243. [Google Scholar] [CrossRef] [PubMed]
  72. Koper, J.W.; Zwarthoff, E.C.; Hagemeijer, A.; Braakman, R.; Avezaat, C.J.; Bergström, M.; Lamberts, S.W. Inhibition of the growth of cultured human meningioma cells by recombinant interferon-α. Eur. J. Cancer Clin. Oncol. 1991, 27, 416–419. [Google Scholar] [CrossRef] [Green Version]
  73. Chamberlain, M.C.; Glantz, M.J. Interferon-α for recurrent World Health Organization grade 1 intracranial meningiomas. Cancer 2008, 113, 2146–2151. [Google Scholar] [CrossRef] [PubMed]
  74. Chamberlain, M.C. IFN-α for recurrent surgery- and radiation-refractory high-grade meningioma: A retrospective case series. CNS Oncol. 2013, 2, 227–235. [Google Scholar] [CrossRef] [PubMed]
  75. Norden, A.D.; Ligon, K.L.; Hammond, S.N.; Muzikansky, A.; Reardon, D.A.; Kaley, T.J.; Batchelor, T.T.; Plotkin, S.R.; Raizer, J.J.; Wong, E.T.; et al. Phase II study of monthly pasireotide LAR (SOM230C) for recurrent or progressive meningioma. Neurology 2015, 84, 280–286. [Google Scholar] [CrossRef] [Green Version]
  76. Chamberlain, M.C.; Glantz, M.J.; Fadul, C.E. Recurrent meningioma: Salvage therapy with long-acting somatostatin analogue. Neurology 2007, 69, 969–973. [Google Scholar] [CrossRef]
  77. Simó, M.; Argyriou, A.A.; Macià, M.; Plans, G.; Majós, C.; Vidal, N.; Gil, M.; Bruna, J. Recurrent high-grade meningioma: A phase II trial with somatostatin analogue therapy. Cancer Chemother. Pharmacol. 2014, 73, 919–923. [Google Scholar] [CrossRef]
  78. Kaley, T.J.; Wen, P.; Schiff, D.; Ligon, K.; Haidar, S.; Karimi, S.; Lassman, A.B.; Nolan, C.P.; DeAngelis, L.M.; Gavrilovic, I.; et al. Phase II trial of sunitinib for recurrent and progressive atypical and anaplastic meningioma. Neuro-Oncology 2015, 17, 116–121. [Google Scholar] [CrossRef] [Green Version]
  79. Shih, K.C.; Chowdhary, S.; Rosenblatt, P.; Weir, A.B.; Shepard, G.C.; Williams, J.T.; Shastry, M.; Burris, H.A., III; Hainsworth, J.D. A phase II trial of bevacizumab and everolimus as treatment for patients with refractory, progressive intracranial meningioma. J. Neuro-Oncol. 2016, 129, 281–288. [Google Scholar] [CrossRef]
  80. Lou, E.; Sumrall, A.L.; Turner, S.; Peters, K.B.; Desjardins, A.; Vredenburgh, J.J.; McLendon, R.E.; Herndon, J.E., 2nd; McSherry, F.; Norfleet, J.; et al. Bevacizumab therapy for adults with recurrent/progressive meningioma: A retrospective series. J. Neuro-Oncol. 2012, 109, 63–70. [Google Scholar] [CrossRef] [Green Version]
  81. Nayak, L.; Iwamoto, F.M.; Rudnick, J.D.; Norden, A.D.; Lee, E.Q.; Drappatz, J.; Omuro, A.; Kaley, T.J. Atypical and anaplastic meningiomas treated with bevacizumab. J. Neuro-Oncol. 2012, 109, 187–193. [Google Scholar] [CrossRef] [PubMed]
  82. Norden, A.D.; Raizer, J.J.; Abrey, L.E.; Lamborn, K.R.; Lassman, A.B.; Chang, S.M.; Yung, W.K.A.; Gilbert, M.R.; Fine, H.A.; Mehta, M.; et al. Phase II trials of erlotinib or gefitinib in patients with recurrent meningioma. J. Neuro-Oncol. 2009, 96, 211–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Wen, P.Y.; Yung, W.K.; Lamborn, K.R.; Norden, A.D.; Cloughesy, T.F.; Abrey, L.E.; Fine, H.A.; Chang, S.M.; Robins, H.I.; Fink, K.; et al. Phase II study of imatinib mesylate for recurrent meningiomas (North American Brain Tumor Consortium study 01–08). Neuro-Oncology 2009, 11, 853–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Ji, Y.; Rankin, C.; Grunberg, S.; Sherrod, A.E.; Ahmadi, J.; Townsend, J.J.; Feun, L.G.; Fredericks, R.K.; Russell, C.A.; Kabbinavar, F.F.; et al. Double-Blind Phase III Randomized Trial of the Antiprogestin Agent Mifepristone in the Treatment of Unresectable Meningioma: SWOG S9005. J. Clin. Oncol. 2015, 33, 4093–4098. [Google Scholar] [CrossRef]
  85. Graillon, T.; Sanson, M.; Campello, C.; Idbaih, A.; Peyre, M.; Peyrière, H.; Basset, N.; Autran, D.; Roche, C.; Kalamarides, M.; et al. Everolimus and Octreotide for Patients with Recurrent Meningioma: Results from the Phase II CEVOREM Trial. Clin. Cancer Res. 2020, 26, 552–557. [Google Scholar] [CrossRef]
  86. Franke, A.J.; Skelton, W.P., IV; Woody, L.E.; Bregy, A.; Shah, A.H.; Vakharia, K.; Komotar, R.J. Role of bevacizumab for treatment-refractory meningiomas: A systematic analysis and literature review. Surg. Neurol. Int. 2018, 9, 133. [Google Scholar] [CrossRef]
  87. Furtner, J.; Schöpf, V.; Seystahl, K.; Le Rhun, E.; Rudà, R.; Roelcke, U.; Koeppen, S.; Berghoff, A.S.; Marosi, C.; Clement, P.; et al. Kinetics of tumor size and peritumoral brain edema before, during, and after systemic therapy in recurrent WHO grade II or III meningioma. Neuro-Oncology 2015, 18, 401–407. [Google Scholar] [CrossRef] [Green Version]
  88. Brastianos, P.K.; Twohy, E.; Gerstner, E.R.; Kaufmann, T.J.; Iafrate, A.J.; Jeyapalan, S.A.; Piccioni, D.E.; Lassman, A.B.; Fadul, C.E.; Schiff, D.; et al. Alliance A071401: Phase II trial of FAK inhibition in meningiomas with somatic NF2 mutations. J. Clin. Oncol. 2020, 38, 2502. [Google Scholar] [CrossRef]
  89. Bi, W.L.; Greenwald, N.F.; Abedalthagafi, M.; Wala, J.; Gibson, W.J.; Agarwalla, P.K.; Horowitz, P.; Schumacher, S.E.; Esaulova, E.; Mei, Y.; et al. Genomic landscape of high-grade meningiomas. npj Genom. Med. 2017, 2, 15. [Google Scholar] [CrossRef]
  90. Dunn, I.F.; Du, Z.; Touat, M.; Sisti, M.B.; Wen, P.Y.; Umeton, R.; Dubuc, A.M.; Ducar, M.; Canoll, P.D.; Severson, E.; et al. Mismatch Repair Deficiency in High-Grade Meningioma: A Rare but Recurrent Event Associated With Dramatic Immune Activation and Clinical Response to PD-1 Blockade. JCO Precis. Oncol. 2018, 2, PO.18.00190. [Google Scholar] [CrossRef]
  91. Nebot-Bral, L.; Brandao, D.; Verlingue, L.; Rouleau, E.; Caron, O.; Despras, E.; El-Dakdouki, Y.; Champiat, S.; Aoufouchi, S.; Leary, A.; et al. Hypermutated tumours in the era of immunotherapy: The paradigm of personalised medicine. Eur. J. Cancer 2017, 84, 290–303. [Google Scholar] [CrossRef] [PubMed]
  92. Brastianos, P.K.; Kim, A.E.; Giobbie-Hurder, A.; Lee, E.Q.; Wang, N.; Eichler, A.F.; Chukwueke, U.; Forst, D.A.; Arrillaga-Romany, I.C.; Dietrich, J.; et al. Phase 2 study of pembrolizumab in patients with recurrent and residual high-grade meningiomas. Nat. Commun. 2022, 13, 1325. [Google Scholar] [CrossRef] [PubMed]
  93. Bi, W.L.; Nayak, L.; Meredith, D.M.; Driver, J.; Du, Z.; Hoffman, S.; Li, Y.; Lee, E.Q.; Beroukhim, R.; Rinne, M.; et al. Activity of PD-1 blockade with nivolumab among patients with recurrent atypical/anaplastic meningioma: Phase II trial results. Neuro-Oncology 2021, 24, 101–113. [Google Scholar] [CrossRef] [PubMed]
Table 1. Correlations of WHO grading with histology, methylation classes, molecular alterations, location, and prognosis of meningiomas.
Table 1. Correlations of WHO grading with histology, methylation classes, molecular alterations, location, and prognosis of meningiomas.
WHO
Grading
HistologyMethylation
Classes
Molecular
Alterations
LocationPrognosis
Grade 1Fibroblastic
Psammomatous
MC ben-1NF2Convexity
Parasagittal areas
Hemispheric meninges
Intraventricular Space
Good
(≥95%)
Meningothelial
Secretory
MC ben-2TRAF7, KLF4, AKT1, SMO1, PIK3CA, POLR2ASkull base
Basal location
Tuberculum sellae for POLR2A mutation
Good
(≥95%)
Angiomatous
Transitional
Rare entities
(metaplastic,
microcystic, rhabdoid)
MC ben-3Not knownConvexity
Parasagittal areas
Hemispheric Meninges
Good
(≥95%)
Grade 2Clear cell
Chordoid
Atypical
MC int-A
(atypical, clear cell)
NF2, SMARCE1, SMARCB1, SMARCA4Convexity
Parasagittal areas
Hemispheric meninges
Intraventricular space
Cranial and spinal location for SMARCE1 mutation
Intermediate
(~88–90%)
MC int-B
(atypical, chordoid)
NF2, TERT mutations, CDKN2A deletionIntermediate
(~45–47%)
Grade 3Anaplastic
Rhabdoid
MC int-BNF2, TERT mutations, CDKN2A deletionConvexity
Parasagittal areas
Hemispheric meninges
Intraventricular space
Intermediate
(~45–47%)
MC malNF2, TERT mutations, CDKN2A deletion
BAP1 (Rhabdoid)
Poor
(~18–20%)
Table 2. Studies of systemic therapies in meningiomas.
Table 2. Studies of systemic therapies in meningiomas.
TreatmentType of StudynResults
Hydroxyurea [65]Retrospective606-month PFS: 10%
Hydroxyurea [66]Retrospective356-month PFS: 3%
Median OS: 8 months
Hydroxyurea
plus imatinib [67]
Phase 215Early interrupted for slow accrual
No significant activity
Temozolomide [68]Phase 2166-month PFS: 0%
Median OS: 7.5 months
Irinotecan [69]Phase 2166-month PFS: 6%
Median OS: 7 months
Trabectedin [71]Randomized phase 2 (EORTC-1320-BTG)90No improvement of median PFS or median OS
Interferon-α [73]Phase 2356-month PFS: 54%
Median OS: 8 months
Interferon-α [74]Retrospective series356-month PFS: 17%
Median OS: 8 months
Pasireotide [75]Phase 234Grade 1: 6-month PFS: 50%: median OS: 104 weeks
Grade 2–3: 6-month-PFS: 17%; median OS: 26 weeks
Octreotide [76]Phase 2166-month PFS: 44%
Median OS: 7.5 months
Octreotide [77]Phase 296-month PFS: 44%
Median OS: 18.7 months
Bevacizumab [80]Retrospective series146-month PFS: 86%
Median OS: not reached
Bevacizumab [81]Retrospective series156-month PFS: 44%
Median OS: 15 months
Bevacizumab plus
everolimus [79]
Phase 217Stable disease: 88%
6-month PFS: 69%
Median OS: 23.8 months
Everolimus plus octreotide [85]Phase 2
(CEVOREM trial)
206-month PFS: 55%
6-month OS: 90%
12-month OS: 75%
Partial response in 78% of patients
Erlotinib or gefitinib [82]Phase 225Grade 1: 6-month PFS: 25%; 12-month OS: 50%
Grade 2–3: 6-month PFS: 29%; 12-month OS: 65%
Imatinib [83]Phase 223Grade 1: 6-month PFS: 45%
Grade 2–3: 6-month PFS: 0%
Sunitinib [78]Phase 2366-month PFS: 42%
Median PFS: 5.2 months
Median OS: 24.6 months
Mifepristone [84]Randomized phase 3 (SWOG-S9005)164No statistical difference between
mifepristone and placebo in terms of
PFS and OS
PFS: progression-free survival; OS: overall survival.
Table 3. Ongoing clinical trials on systemic treatments in meningiomas.
Table 3. Ongoing clinical trials on systemic treatments in meningiomas.
Trial IDType of StudyArm of TreatmentnEndpoints
NCT02648997Phase 2Nivolumab alone (Cohort 1) or in combination with ipilimumab (Cohort 2)50Primary: 6-month PFS
Secondary: median PFS, median OS, ORR, safety
NCT03631953Phase 1Alpelisib in combination with trametinib25Primary: DLT
NCT04728568ProspectiveSintilimab15Primary: PFS
Secondary: OS
NCT04501705ProspectiveApatinib29Primary: 6-month PFS
Secondary: ORR, OS
NCT03604978Phase 1–2Nivolumab alone or plus ipilimumab in combination with fractionated SRS15Primary: DLT, safety, ORR
Secondary: median PFS, median OS, changes in peripheral T-cells
NCT02933736Early phase 1Ribociclib48Primary: plasma exposure, CSF penetration, brain accumulation of ribociclib
NCT02523014Phase 2Vismodegib or
FAK inhibitor, or GSK2256098 or capivasertib, or abemaciclib based on molecular screening
124Primary: 6-month PFS, ORR
Secondary: median PFS, median OS, safety
NCT04659811Phase 2Pembrolizumab plus SRS90Primary: 12-month PFS
Secondary: median PFS, median OS
NCT04374305Phase 2Brigatinib80Primary: radiological response rate
Secondary: safety
NCT03095248Phase 2Selumetinib34Primary: change in hearing response, response rate of other NF2-related tumors (including meningiomas)
NCT04541082Phase 1ONC206102Primary: MTD
PFS: progression-free survival; OS: overall survival; ORR: objective response rate; DLT: dose-limiting toxicity; SRS: stereotactic radiosurgery; MTD: maximum tolerated dose.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pellerino, A.; Bruno, F.; Palmiero, R.; Pronello, E.; Bertero, L.; Soffietti, R.; Rudà, R. Clinical Significance of Molecular Alterations and Systemic Therapy for Meningiomas: Where Do We Stand? Cancers 2022, 14, 2256. https://doi.org/10.3390/cancers14092256

AMA Style

Pellerino A, Bruno F, Palmiero R, Pronello E, Bertero L, Soffietti R, Rudà R. Clinical Significance of Molecular Alterations and Systemic Therapy for Meningiomas: Where Do We Stand? Cancers. 2022; 14(9):2256. https://doi.org/10.3390/cancers14092256

Chicago/Turabian Style

Pellerino, Alessia, Francesco Bruno, Rosa Palmiero, Edoardo Pronello, Luca Bertero, Riccardo Soffietti, and Roberta Rudà. 2022. "Clinical Significance of Molecular Alterations and Systemic Therapy for Meningiomas: Where Do We Stand?" Cancers 14, no. 9: 2256. https://doi.org/10.3390/cancers14092256

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