Next Article in Journal
Variant Prediction by Analyzing RdRp/S Gene Double or Low Amplification Pattern in Allplex SARS-CoV-2 Assay
Next Article in Special Issue
Combining FORGE Score and Histopathological Diagnostic Criteria of Atypical Meningioma Enables Risk Stratification of Tumor Progression
Previous Article in Journal
New Treatment Strategy Using Repetitive Transcranial Magnetic Stimulation for Post-Stroke Aphasia
Previous Article in Special Issue
Human TERT Promoter Mutations in Atypical and Anaplastic Meningiomas
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Discovering the Molecular Landscape of Meningioma: The Struggle to Find New Therapeutic Targets

1
Medical Oncology Department, Azienda USL, Via Altura n. 3, 40139 Bologna, Italy
2
Nervous System Medical Oncology Department, IRCSS Istituto di Scienze Neurologiche di Bologna, 40139 Bologna, Italy
3
IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2021, 11(10), 1852; https://doi.org/10.3390/diagnostics11101852
Submission received: 24 July 2021 / Revised: 27 September 2021 / Accepted: 4 October 2021 / Published: 8 October 2021
(This article belongs to the Special Issue Molecular Diagnostics of Brain Tumours)

Abstract

:
Meningiomas are the most common primary CNS tumors. They are usually benign but can present aggressive behavior in about 20% of cases. The genetic landscape of meningioma is characterized by the presence (in about 60% of cases) or absence of NF2 mutation. Low-grade meningiomas can also present other genetic alterations, particularly affecting SMO, TRAF7, KLF4 AKT1 and PI3KCA. In higher grade meningiomas, mutations of TERT promoter and deletion of CDKN2A/B seem to have a prognostic value. Furthermore, other genetic alterations have been identified, such as BAP1, DMD and PBRM1. Different subgroups of DNA methylation appear to be correlated with prognosis. In this review, we explored the genetic landscape of meningiomas and the possible therapeutic implications.

1. Introduction

Meningiomas are central nervous system (CNS) tumors that originate from arachnoid cells in the inner surface of the dura. With an incidence of 7.86 cases per 100,000 people per year [1], meningiomas represent the most frequent CNS tumor, accounting for about 36% of all cases and 53% of benign lesions [1]. They are often benign and mostly diagnosed incidentally [2].
Many risk factors have been associated with development of meningioma. Among these, diabetes mellitus, arterial hypertension, smoking, and mobile phone use have been investigated, but results are inconclusive [3,4]. A recognized risk factor is ionizing radiation to the skull, with an increased risk of developing meningiomas six-fold to ten-fold greater [5,6]. Meningiomas that originated after irradiation are often multiple and present aggressive behavior [7]. In addition, sex hormones have been investigated as risk factors since there is a higher incidence of meningiomas in post-puberal women (2:1 versus men), with a ratio of about 3:1 in the reproductive period, but there is no definitive evidence [4,8,9,10,11,12].
Meningiomas can also be genetically determined and are present in several genetic syndromes such as: neurofibromatosis type 2 (NF2), which is most frequently associated with meningiomas, Gorlin, Li Fraumeni, Cowden, von Hippel–Lindau, and multiple endocrine neoplasia type 1 (MEN1). In these genetic syndromes, meningiomas are often multiple and arise in children [13].
Ten-year overall survival (OS) of benign meningiomas is about 81.4%, while for malignant ones, it is 57.1%, with about 53% for grade II and 0% for grade III. The recurrence rate for grade II is about 50%, and for grade III, it is 90% [14]. Disease progression is defined not only by the growth of the residual tumor, but also by the transformation into a higher-grade tumor.
In the 2016 WHO classification, meningiomas are distinct in different grades [15], as reported in Table 1.
Different grades present distinct histological and clinical features, but molecular alterations are not included due to the scant evidence about this topic [16].
We performed a review of the literature to summarize the current knowledge on meningiomas with a specific insight on the molecular alterations and possible therapeutic strategies. We carried out research on Pubmed/Medline, Cochrane library and Scopus using the keywords “meningioma” or “anaplastic” or “atypical” or “CNS” or “brain tumor” and we selected pivotal studies, while considering the quality of the study, how it was conducted, and the strength of the results.

2. Molecular Alterations

Meningiomas present immunohistochemical markers that are epithelial membrane antigens, in particular: somatostatin receptor 2A (SSTR2A) and progesterone receptor in 70–80% of cases, and estrogen receptor in about 5–30% of cases [17,18].
New genomic analysis techniques such as whole genomic sequencing (WGS), transcriptome analysis, whole exome sequencing (WES), DNA methylation assay, and chromatin immunoprecipitation sequencing allowed the characterization of the mutational landscape of meningiomas, with consequent identification of possible druggable targets [19].

2.1. Genomic Analysis Alterations

A common molecular alteration in meningiomas is 22q12 chromosome deletion that encodes for the tumor suppressor gene Merlin (also called neurofibromin 2, NF2), present in about half of all tumors [20,21]. NF2 mutation predispose to the development of meningiomas, and it seems to be associated with fibroblastic/transitional meningiomas [20,22]. NF2 inactivation gives to the tumor a genomic instability and a peculiar multiple localization in the hemispheres [23]. It is not yet well understood how the inactivation of NF2 can guide the development of meningiomas. Some hypotheses suggest that Merlin can inhibit cell proliferation with a contact-dependent regulation including Hippo, Notch and Patched pathway [24] and that it can activate the mammalian target of rapamycin (mTOR) pathway during tumor development [25,26]. The high frequency of this mutation can create a subdivision of meningiomas into NF2 and non-NF2 mutated.
Four mutually exclusive pathways that can guide the development of meningiomas have been identified through genomic analysis [27,28].

2.1.1. Increased Hedgehog Signaling (through Mutation of SMO, SUFU or PRKAR1A)

Smoothened, frizzled class receptor (SMO) is a gene that encodes the transmembrane receptor protein smoothened homolog. This receptor is responsible for the transmission of signals from Hedgehog ligands to cells, leading to constitutive activation of SMO. SMO and v-akt murine thymoma viral oncogene homolog 1 (AKT1) can lead to the activation of PI3K–AKT–mTOR pathway; it is relatively common and is associated with meningiomas localized in the skull base and with genomic stability [20,29].
AKT1 mutations seems to be associated with grade I meningothelial meningiomas of the spine and skull base [20,30]. Somatic mutations at codon 17 are associated with higher risk of recurrence [30].
Suppressor of fused (SUFU) and patched 1 mutations can activate the SHH pathway, and somatic mutations of SUFU are present in 1% of sporadic meningiomas [30]. These alterations are commonly found in Gorlin syndrome [31].

2.1.2. TRAF7 (through KLF4 Mutation or PI3K Pathway Activation)

TRAF7 mutations is present in about 25% of WHO grade I and II meningiomas. The most frequent alteration is in the WD40 domain, which is involved in the regulation of JUN N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) signaling [23,27,32]. TRAF7 mutation can be associated with AKT1, Krupple-like factor 4 (KFL4) or phosphatidylinositol-4,5- bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) alteration, but it is mutually exclusive with NF2 and SMO mutations [30].
PIK3CA mutations are mutually exclusive with AKT1, SMO and NF2, and they can rarely be co-present to TRAF7 mutation [30].
KLF4 K409Q missense mutations are present in up to 50% of NF2 nonmutated meningiomas, often co-present with TRAF7 mutations, and lead to upregulation of HIF-1a pathway. Furthermore, KLF4 and TRAF7 seem to be associated with secretory meningiomas with more aggressive behavior due to increased swelling of the brain [23,33].

2.1.3. POLR2A Mutations

Mutations on two hotspots of RNA polymerase II subunit A (POLR2A) are associated with meningothelial histology, usually localized in the tuberculum sellae [28].

2.1.4. Other Rarer Mutations

Knowledge of molecular alterations of WHO grade III meningiomas is limited because of the rarity of the disease. In rhabdoid meningiomas, the frequent inactivation of breast cancer-associated protein 1 (BAP1) has been identified, and it seems to be associated with early tumor recurrence [34]. In papillary meningiomas, the biallelic inactivation of polybromo 1 (PBRM1) gene has been described, which can overlap with BAP1 mutations, but its role is not yet understood [35].
In atypical meningiomas, the upregulation of enhancer of zeste homolog 2 (EZH2) that interacts with epigenetic mechanisms can be present [36,37], and it seems to be a marker of aggressiveness and higher grade [38]. In particular, the expression of EZH2 and H3K27me3 negativity has been associated with shorter progression-free survival (PFS), thus highlighting a potential prognostic role for these biomarkers [38].
Duchenne muscular dystrophy (DMD) gene encodes for the protein dystrophin and its mutation has also been pointed out in meningiomas. Somatic DMD deletions seem to be associated with worse prognosis [39].
Mutations of KDM5C (Lysine Demethylase 5C), KDM6A or Somatic SWI/SNF-related matrix associated actin-dependent regulation of chromatin subfamily B member 1 protein (SMARCB1) can lead to epigenetic modifications that are present in about 10% of non-NF2 meningiomas [20].
Furthermore, telomerase reverse transcriptase (TERT) promoter mutations have never been detected in de novo atypical meningiomas but are present in secondary atypical meningiomas progressed from lower-grade primary tumors [40,41]. These findings highlight the role of TERT promoter alteration as a possible prognostic marker of recurrence and survival. Several studies have shown that TERT promoter mutation in meningiomas is associated with worse prognosis and shorter overall survival independent of WHO grade [42,43,44,45].
Mixed-lineage leukemia 1/Lysine (K)-specific MethylTransferase 2 (MLL/KMT2) mutation has been identified in WHO grade 2 chordoid meningioma, associated with NF2 mutations and with worse prognosis [46].
Cyclin-dependent kinase Inhibitor 2A/B (CDKN2A/B) plays an important role in cell cycle regulation, acting as a tumor suppressor. It has been found to be alternated in meningiomas, and its alteration can lead to the progression from grade 2 to grade 3 meningioma [38,47]. Somatic mutations and homozygous losses of CDKN2A and CDKN2B have been identified in anaplastic meningiomas [48,49,50]. A retrospective analysis of 67 atypical meningiomas showed that overexpression of p16, CDK6, and pRB protein predicted recurrence [49]. Moreover, a next-generation sequencing analysis of 17 recurrent and 13 non-recurrent meningiomas identified three CDKN2A alteration (p.Ala148Thr) mutation, whole homozygous or heterozygous gene loss, or promotor methylation (>8) strongly correlated with recurrence and a Ki67 labeling index > 7% [51].
The main molecular alterations are provided in Table 2.

2.2. Copy Number Alterations

Overall, copy number aberrations (CNA) have been associated with risk of recurrence in patients with resected atypical meningiomas, and it can represent a marker to help in the decision-making process on whether to start adjuvant radiotherapy [30]. The most frequent CNAs are 22q deletion, 1p, 14q, 9p loss, which are associated with higher grade meningiomas [53], and 6q, 10q, and 18q loss, or the gain of 1q, 9q, 12q, 15q, 17q, 19q, 20q and 5 chromosomes [36,54]. In recurrent tumors, CNAs overlap in about 75% the primary tumors, suggesting that they may play a key role in tumor development [36,54].
In addition to NF2, another gene located on chromosome 22q that can be altered is CHEK2, which has been described in NF2 mutated meningiomas. It is a tumor suppressor involved in DNA repair, and its mutations seem to contribute to genome instability [52].

2.3. Methylation Profiles

Through analysis of DNA methylation profiles, six subclasses of meningiomas have been identified, characterized by different histology, cytogenetic, mutational and gene expression patterns, and associated with different risk of recurrence [55]:
-
group A: four methylation classes (MC benign (ben)-1 (n = 113), MC ben-2 (n = 118), and MC ben-3 (n = 73), MC intermediate (int)-A (n = 105));
-
group B: two methylation classes (MC int-B (n = 47), MC malignant (mal) (n = 41)).
This classification is superior to WHO grading in terms of predicting PFS. In particular, MC intermediate patients with WHO grade I meningiomas resulted in a worse prognosis than patients characterized as grade I only by histology. Furthermore, grade I MC intermediate patients seem to have similar prognoses to WHO grade II ones, while grade II MC benign meningiomas had a better prognosis than WHO grade II ones.
Olar and colleagues identified 64 CpG loci methylation predictors correlated with different recurrence risks [56]. In this study, two subgroups of patients were identified: a prognostically unfavorable and a prognostically favorable molecular methylation subgroup. The prognostically unfavorable subgroup presented a higher proportion of copy number aberrations, including losses of 1p, 6q, 14q and 18q, and a gain of 1q, all associated with poor outcomes, with 185 hypermethylated CpG loci and a median RFS of 12.07 years (range 0.31–17.61 years). The favorable subgroup presented 98 hypermethylated CpG loci and had a median recurrence-free survival (RFS) not reached (range 0.27–16.6 years).
A nomogram integrating clinical factors and DNA methylation profiles has been developed to predict early meningioma recurrence. The 5-year methylome predictor subdivided patients into lower and higher risk groups with different FRS in all three validation cohorts of the study. In particular, patients in the higher risk group had a median RFS of 2.1, 8.1, and 4.2 years in the first, second, and third validation cohorts, respectively, compared with patients in the lower risk groups, which presented a median RFS of unreached in the first and second validation cohorts and of 7.2 years in the third validation cohort while higher risk groups had a median RFS of 2.1, 8.1, and 4.2 years in the three validation cohorts, respectively [57].
Molecular analysis allowed to clarify the biological landscape of two other types of meningiomas, with possible therapeutic implications: radiation-induced and progestin-associated meningiomas [58,59]. Progestine-associated meningiomas, which are generally grade 1 and 2, present NF2 alteration only in 7.5% of cases, while a PIK3CA mutation can be found in 35% of cases [59,60]. Otherwise, radiation-induced meningiomas often present NF2 alterations and deficit of DNA double strand repair genes [61].
The increasing knowledge of the molecular landscape of meningiomas has allowed individuation of prognostic and predictive markers that can guide therapeutic decision-making processes and timing of follow-up. Nonetheless, molecular features need to be integrated with histopathologic and clinical characteristics to better help predict the risk of recurrence.

3. Treatment Options

The choice of the more appropriate treatment approach depends on growth rate, grade, localization and symptoms of meningioma.

3.1. Surgery

Surgery is the cornerstone treatment for growing and symptomatic meningiomas, while for asymptomatic ones, radiological surveillance can be considered [62]. It should be as radical as possible, and the radicality of resection is measured by the Simpson grade, reported in Table 3 [63]. The risk of recurrence is higher if residual disease is present, and this can happen if the localization of meningioma limits the surgical approach. After gross total resection, the 5-year recurrence rate is 7–23% for grade I meningiomas, 50–55% for grade II and 72–78% for grade III ones [14].

3.2. Radiotherapy

Meningiomas not suitable for surgery can be irradiated, but radiotherapy is mainly used in an adjuvant setting after surgical resection or to treat recurrent disease. Currently, there is no definitive evidence on timing, dose and fractioning of radiotherapy in meningiomas because of the lack of randomized controlled trials. It is not indicated in radically resected WHO grade I meningiomas, but it can be proposed in cases of residual disease [13]. In grade II meningiomas, it still plays a controversial role, but it can be considered in cases of incomplete resection [64]. In grade III meningiomas, adjuvant radiotherapy should be used to improve local control [65].

3.3. Systemic Treatment

The use of systemic treatment is largely experimental since several drugs have been tested with limited results. Currently, there is not a standard of care treatment or definitive evidence on preferable therapies and their sequence. Enrollment in clinical trial is highly recommended [66].
Some prospective and retrospective studies investigated different types of treatments, including target therapies such as vascular endothelial growth factor (VEGF)/receptor (VEGFR) inhibitors [67,68,69,70] or epidermal growth factor receptor (EGFR) inhibitors [71] and chemotherapies such as as hydroxyurea [72,73,74], temozolomide [75], irinotecan [76], and trabectedin [77,78]. These latter compounds are not recommended due to their scant efficacy. Molecular alteration presented by meningiomas suggests the investigation of targeted therapy agents. Meningiomas are highly vascularized tumors and present an upregulated expression of VEGF [79], supporting the study of anti-angiogenic agents in phase II trials [80]. Among these, there are monoclonal antibody against VEGF bevacizumab [68,69,70,81,82,83], the tyrosine kinase inhibitor (TKI) directed against VEGFR sunitinib, the TKI against platelet-derived growth factor receptor-beta (PDGFR-beta) c-kit, VEGFR-2 vatalanib [84], the TKI against PDGFR c-kit, and BCR-ABL imatinib [85].
The inactivation of NF2 and its interaction with the mTOR pathway, with a negative regulation of mTORC1, which results in overexpression and a positive regulation of mTORC2 [86,87,88], supported the investigation of mTOR inhibitors. Furthermore, KLF4 K409Q mutation, evidenced in 10–14% of meningiomas and found in combination with TRAF7 mutations in the secretory subtype, upregulates hypoxia-inducible factor-1a (HIF-1a) pathway, thus creating a state of cellular hypoxia with the subsequent activation of mTOR pathway. Consequently, mTOR inhibitors have been experimented as possible treatment strategies [89].
Everolimus has been tested in combination with octreotide [90] and with bevacizumab [70]. In the phase II CEVOREM trial, the combination of everolimus 10 mg daily and octreotide 30 mg on day 1 was investigated in 20 patients with recurrent progressive meningiomas ineligible for further surgery or radiotherapy (WHO grade I, II, or III). The experimental combination showed clinical and radiological activity. Everolimus 10 mg daily was also investigated in a phase II trial in combination with bevacizumab (10 mg/kg IV days 1 and 15) in 17 patients with recurrent meningioma (WHO grade I, II, or III) following standard treatments with surgical resection and radiotherapy. The combination resulted to be well tolerated and obtained stable disease in 88% of patients.
Vistusertib (AZD2014), an inhibitor of mTORC1 and mTORC2, is being testing in NCT03071874 trial, based on promising results of preclinical studies, in both recurrent and progressive meningiomas [91]. Activating mutations of PIK3-AKT-mTOR pathway is being evaluated as a therapeutic target in a phase II trial in progressive refractory meningiomas investigating the PI3K inhibitor alpelisib in combination with the MEK inhibitor trametinib (NCT03631953 trial) [27].
Dasatinib, an inhibitor of Src kinases, erythropoietin-producing hepatocellular receptor tyrosine kinases (EPH RTKs) and c-KIT, has been tested in glioblastoma [92], and it also showed an inhibition of growth in NF2-deficient meningiomas when associated with dual mTORC1/2 inhibitor [93].
The alteration of AKT1 can be targeted by AKT inhibitors such as capivasertinib (AZD5363). A patient with meningotheliomatous meningioma presenting AKT1E17K mutation, who progressed to surgery, radiotherapy and two previous lines of systemic therapies, was treated with capivasertinib with a durable disease control, thus suggesting this molecular alteration may be a potential therapeutic target [94].
Preclinical studies also showed the sensibility of NF2-mutated meningiomas to focal adhesion kinase (FAK) inhibition, which seem to have a synthetic lethal relationship with NF2 loss [95]. On the basis of this finding, a FAK inhibitor (GSK2256098) has been tested in a phase II trial (Alliance A071401, NCT02523014) in recurrent or progressive meningiomas (WHO I-III grade) [96]. The experimental drug resulted to improve 6-months PFS (one of the primary endpoints) and to be well tolerated, thus supporting further investigation of this treatment. The same FAK inhibitor (GSK2256098) is also being tested in a two-arm phase II trial (NCT02523014) in progressive meningiomas harboring NF2 mutations. In the second arm of this trial, patients with SMO mutations received an SMO and PTCH1 inhibitor (vismodegib).
BAP1-mutated meningiomas are associated with DNA damage repair defects [34]. In this subgroup, Poly ADP ribose polymerase (PARP) inhibitors are being investigated [97]. Furthermore, a phase II study (NCT02860286) is evaluating Tazemetostat, an EZH2 inhibitor, in tumors with BAP1 loss of function.
CDK inhibitors are another class of drugs being tested in meningiomas, given their role in suppressing tumor cell survival by depleting proteins such as MCL-1 and c-MYC. Classifying patients based on the genomic methylation profiling [55], the oral CDK inhibitor TG02 has been tested in meningioma cell cultures [98]. Another CDK inhibitor, ribociclib, has been tested in vivo, demonstrating to achieve pharmacologically active concentrations in aggressive meningioma tissue [99].
In progestin-associated meningiomas, the anti-progesterone drug mifepristone is being tested in a phase III trial against placebo in unresectable meningiomas (NCT03015701). In radiation-induced meningiomas, the alteration of DNA double strand break repair genes through the homologous recombination repair system could suggest the investigation of PARP inhibitors [58,100].
The identification of the expression of somatostatin receptor (SSTR) subtype 2 by [101] meningiomas led to investigating the role somatostatin analogs. In vitro studies showed that octreotide, a somatostatin analog, presented an antiproliferative effect with reduced tumor growth, but its effect did not result in tumor shrinkage. Further trials confirmed the absence of clinical effect of these compounds in meningioma [102].

4. Conclusions

Novel molecular characterization techniques allow understanding of the biology underlying the development and progression of meningiomas. Several molecular alterations have been shown, with prognostic and therapeutic implications. Novel therapeutic approaches are being tested to target peculiar mutations. This biological evidence and the new treatments under investigation could possibly change the approach to meningioma relapse or refractory to surgery and radiotherapy.

Author Contributions

Conceptualization, A.A.B.; investigation, A.A.B., I.M., E.F., V.D.N., L.G., A.T., D.A., S.B. and R.L.; methodology: A.A.B.; supervision, A.A.B.; writing—original draft, A.A.B., E.F. and I.M.; writing—review and editing, A.A.B. and E.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gittleman, H.R.; Ostrom, Q.T.; Rouse, C.D.; Dowling, J.A.; de Blank, P.M.; Kruchko, C.A.; Elder, J.B.; Rosenfeld, S.S.; Selman, W.R.; Sloan, A.E.; et al. Trends in central nervous system tumor incidence relative to other common cancers in adults, adolescents, and children in the United States, 2000 to 2010. Cancer 2015, 121, 102–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Maggio, I.; Franceschi, E.; Tosoni, A.; Nunno, V.D.; Gatto, L.; Lodi, R.; Brandes, A.A. Meningioma: Notalways a benigntumor. A review of advances in the treatment of meningiomas. CNS Oncol. 2021, 10, Cns72. [Google Scholar] [CrossRef] [PubMed]
  3. Flint-Richter, P.; Mandelzweig, L.; Oberman, B.; Sadetzki, S. Possible interaction between ionizing radiation, smoking, and gender in the causation of meningioma. Neuro-Oncology 2011, 13, 345–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Schneider, B.; Pülhorn, H.; Röhrig, B.; Rainov, N.G. Predisposing conditions and risk factors for development of symptomatic meningioma in adults. Cancer Detect. Prev. 2005, 29, 440–447. [Google Scholar] [CrossRef]
  5. Ron, E.; Modan, B.; Boice, J.D., Jr.; Alfandary, E.; Stovall, M.; Chetrit, A.; Katz, L. Tumors of the brain and nervous system after radiotherapy in childhood. N. Engl. J. Med. 1988, 319, 1033–1039. [Google Scholar] [CrossRef]
  6. Sadetzki, S.; Flint-Richter, P.; Starinsky, S.; Novikov, I.; Lerman, Y.; Goldman, B.; Friedman, E. Genotyping of patients with sporadic and radiation-associated meningiomas. Cancer Epidemiol. Biomark. Prev. 2005, 14, 969–976. [Google Scholar] [CrossRef] [Green Version]
  7. Al-Mefty, O.; Topsakal, C.; Pravdenkova, S.; Sawyer, J.R.; Harrison, M.J. Radiation-induced meningiomas: Clinical, pathological, cytokinetic, and cytogenetic characteristics. J. Neurosurg. 2004, 100, 1002–1013. [Google Scholar] [CrossRef]
  8. Blitshteyn, S.; Crook, J.E.; Jaeckle, K.A. Is there an association between meningioma and hormone replacement therapy? J. Clin. Oncol. 2008, 26, 279–282. [Google Scholar] [CrossRef] [Green Version]
  9. Claus, E.B.; Black, P.M.; Bondy, M.L.; Calvocoressi, L.; Schildkraut, J.M.; Wiemels, J.L.; Wrensch, M. Exogenous hormone use and meningioma risk: What do we tell our patients? Cancer 2007, 110, 471–476. [Google Scholar] [CrossRef]
  10. Claus, E.B.; Park, P.J.; Carroll, R.; Chan, J.; Black, P.M. Specific genes expressed in association with progesterone receptors in meningioma. Cancer Res. 2008, 68, 314–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Korhonen, K.; Salminen, T.; Raitanen, J.; Auvinen, A.; Isola, J.; Haapasalo, H. Female predominance in meningiomas can not be explained by differences in progesterone, estrogen, or androgen receptor expression. J. Neurooncol. 2006, 80, 1–7. [Google Scholar] [CrossRef]
  12. Vadivelu, S.; Sharer, L.; Schulder, M. Regression of multiple intracranial meningiomas after cessation of long-term progesterone agonist therapy. J. Neurosurg. 2010, 112, 920–924. [Google Scholar] [CrossRef]
  13. 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]
  14. Buerki, R.A.; Horbinski, C.M.; Kruser, T.; Horowitz, P.M.; James, C.D.; Lukas, R.V. An overview of meningiomas. Future Oncol. 2018, 14, 2161–2177. [Google Scholar] [CrossRef]
  15. 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] [PubMed] [Green Version]
  16. 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]
  17. Hsu, D.W.; Efird, J.T.; Hedley-Whyte, E.T. Progesterone and estrogen receptors in meningiomas: Prognostic considerations. J. Neurosurg. 1997, 86, 113–120. [Google Scholar] [CrossRef] [PubMed]
  18. Menke, J.R.; Raleigh, D.R.; Gown, A.M.; Thomas, S.; Perry, A.; Tihan, T. Somatostatin receptor 2a is a more sensitive diagnostic marker of meningioma than epithelial membrane antigen. Acta Neuropathol. 2015, 130, 441–443. [Google Scholar] [CrossRef] [Green Version]
  19. Birzu, C.; Peyre, M.; Sahm, F. Molecular alterations in meningioma: Prognostic and therapeutic perspectives. Curr. Opin. Oncol. 2020, 32, 613–622. [Google Scholar] [CrossRef]
  20. 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]
  21. Choy, W.; Kim, W.; Nagasawa, D.; Stramotas, S.; Yew, A.; Gopen, Q.; Parsa, A.T.; Yang, I. The molecular genetics and tumor pathogenesis of meningiomas and the future directions of meningioma treatments. Neurosurg. Focus 2011, 30, E6. [Google Scholar] [CrossRef] [PubMed]
  22. 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]
  23. 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] [PubMed] [Green Version]
  24. Curto, M.; McClatchey, A.I. Nf2/Merlin: A coordinator of receptor signalling and intercellular contact. Br. J. Cancer 2008, 98, 256–262. [Google Scholar] [CrossRef] [Green Version]
  25. 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]
  26. 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]
  27. 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-Oncology 2016, 18, 649–655. [Google Scholar] [CrossRef] [Green Version]
  28. 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] [Green Version]
  29. 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-Oncology 2017, 19, 345–351. [Google Scholar] [CrossRef] [Green Version]
  30. Aizer, A.A.; Abedalthagafi, M.; Bi, W.L.; Horvath, M.C.; Arvold, N.D.; Al-Mefty, O.; Lee, E.Q.; Nayak, L.; Rinne, M.L.; Norden, A.D.; et al. A prognostic cytogenetic scoring system to guide the adjuvant management of patients with atypical meningioma. Neuro-Oncology 2016, 18, 269–274. [Google Scholar] [CrossRef] [Green Version]
  31. Narang, A.; Maheshwari, C.; Aggarwal, V.; Bansal, P.; Singh, P. Gorlin-Goltz Syndrome with Intracranial Meningioma: Case Report and Review of Literature. World Neurosurg. 2020, 133, 324–330. [Google Scholar] [CrossRef]
  32. Yuzawa, S.; Nishihara, H.; Yamaguchi, S.; Mohri, H.; Wang, L.; Kimura, T.; Tsuda, M.; Tanino, M.; Kobayashi, H.; Terasaka, S.; et al. Clinical impact of targeted amplicon sequencing for meningioma as a practical clinical-sequencing system. Mod. Pathol. 2016, 29, 708–716. [Google Scholar] [CrossRef]
  33. 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]
  34. 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] [PubMed] [Green Version]
  35. Williams, E.A.; Wakimoto, H.; Shankar, G.M.; Barker, F.G., 2nd; Brastianos, P.K.; Santagata, S.; Sokol, E.S.; Pavlick, D.C.; Shah, N.; Reddy, A.; et al. Frequent inactivating mutations of the PBAF complex gene PBRM1 in meningioma with papillary features. Acta Neuropathol. 2020, 140, 89–93. [Google Scholar] [CrossRef] [PubMed]
  36. Harmancı, A.S.; Youngblood, M.W.; Clark, V.E.; Coşkun, 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, 1–14. [Google Scholar] [CrossRef] [PubMed]
  37. Viré, E.; Brenner, C.; Deplus, R.; Blanchon, L.; Fraga, M.; Didelot, C.; Morey, L.; Van Eynde, A.; Bernard, D.; Vanderwinden, J.M.; et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature 2006, 439, 871–874. [Google Scholar] [CrossRef] [PubMed]
  38. Samal, S.; Patnaik, A.; Sahu, F.; Purkait, S. Altered expression of epigenetic modifiers EZH2, H3K27me3, and DNA methyltransferases in meningiomas—prognostic biomarkers for routine practice. Folia Neuropathol. 2020, 58, 133–142. [Google Scholar] [CrossRef]
  39. 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] [PubMed]
  40. Goutagny, S.; Nault, J.C.; Mallet, M.; Henin, D.; Rossi, J.Z.; Kalamarides, M. High incidence of activating TERT promoter mutations in meningiomas undergoing malignant progression. Brain Pathol. 2014, 24, 184–189. [Google Scholar] [CrossRef]
  41. 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, djv377. [Google Scholar] [CrossRef]
  42. Lu, V.M.; Goyal, A.; Lee, A.; Jentoft, M.; Quinones-Hinojosa, A.; Chaichana, K.L. The prognostic significance of TERT promoter mutations in meningioma: A systematic review and meta-analysis. J. Neuro-Oncol. 2019, 142, 1–10. [Google Scholar] [CrossRef] [PubMed]
  43. 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] [PubMed]
  44. Spiegl-Kreinecker, S.; Lötsch, D.; Neumayer, K.; Kastler, L.; Gojo, J.; Pirker, C.; Pichler, J.; Weis, S.; Kumar, R.; Webersinke, G.; et al. TERT promoter mutations are associated with poor prognosis and cell immortalization in meningioma. Neuro-Oncology 2018, 20, 1584–1593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Mellai, M.; Porrini Prandini, O.; Mustaccia, A.; Fogazzi, V.; Allesina, M.; Krengly, M.; Boldorini, R. Human TERT Promoter Mutations in Atypical and Anaplastic Meningiomas. Diagnostics 2021, 11, 1624. [Google Scholar] [CrossRef] [PubMed]
  46. Georgescu, M.M.; Nanda, A.; Li, Y.; Mobley, B.C.; Faust, P.L.; Raisanen, J.M.; Olar, A. Mutation Status and Epithelial Differentiation Stratify Recurrence Risk in Chordoid Meningioma-A Multicenter Study with High Prognostic Relevance. Cancers 2020, 12, 225. [Google Scholar] [CrossRef] [Green Version]
  47. 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] [Green Version]
  48. Kheirollahi, M.; Mehr-Azin, M.; Kamalian, N.; Mehdipour, P. Expression of cyclin D2, P53, Rb and ATM cell cycle genes in brain tumors. Med. Oncol. 2011, 28, 7–14. [Google Scholar] [CrossRef]
  49. Kim, M.S.; Kim, K.H.; Lee, E.H.; Lee, Y.M.; Lee, S.H.; Kim, H.D.; Kim, Y.Z. Results of immunohistochemical staining for cell cycle regulators predict the recurrence of atypical meningiomas. J. Neurosurg. 2014, 121, 1189–1200. [Google Scholar] [CrossRef] [Green Version]
  50. Lee, Y.; Liu, J.; Patel, S.; Cloughesy, T.; Lai, A.; Farooqi, H.; Seligson, D.; Dong, J.; Liau, L.; Becker, D.; et al. Genomic landscape of meningiomas. Brain Pathol. 2010, 20, 751–762. [Google Scholar] [CrossRef]
  51. Guyot, A.; Duchesne, M.; Robert, S.; Lia, A.S.; Derouault, P.; Scaon, E.; Lemnos, L.; Salle, H.; Durand, K.; Labrousse, F. Analysis of CDKN2A gene alterations in recurrent and non-recurrent meningioma. J. Neuro-Oncol. 2019, 145, 449–459. [Google Scholar] [CrossRef]
  52. Yang, H.W.; Kim, T.M.; Song, S.S.; Shrinath, N.; Park, R.; Kalamarides, M.; Park, P.J.; Black, P.M.; Carroll, R.S.; Johnson, M.D. Alternative splicing of CHEK2 and codeletion with NF2 promote chromosomal instability in meningioma. Neoplasia 2012, 14, 20–28. [Google Scholar] [CrossRef] [Green Version]
  53. Collord, G.; Tarpey, P.; Kurbatova, N.; Martincorena, I.; Moran, S.; Castro, M.; Nagy, T.; Bignell, G.; Maura, F.; Young, M.D.; et al. An integrated genomic analysis of anaplastic meningioma identifies prognostic molecular signatures. Sci. Rep. 2018, 8, 13537. [Google Scholar] [CrossRef] [Green Version]
  54. 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]
  55. 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]
  56. 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]
  57. 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] [Green Version]
  58. Paramasivam, N.; Hübschmann, D.; Toprak, U.H.; Ishaque, N.; Neidert, M.; Schrimpf, D.; Stichel, D.; Reuss, D.; Sievers, P.; Reinhardt, A.; et al. Mutational patterns and regulatory networks in epigenetic subgroups of meningioma. Acta Neuropathol. 2019, 138, 295–308. [Google Scholar] [CrossRef] [PubMed]
  59. Peyre, M.; Gaillard, S.; de Marcellus, C.; Giry, M.; Bielle, F.; Villa, C.; Boch, A.L.; Loiseau, H.; Baussart, B.; Cazabat, L.; et al. Progestin-associated shift of meningioma mutational landscape. Ann. Oncol. 2018, 29, 681–686. [Google Scholar] [CrossRef] [PubMed]
  60. Shahin, M.N.; Magill, S.T.; Dalle Ore, C.L.; Viner, J.A.; Peters, P.N.; Solomon, D.A.; McDermott, M.W. Fertility treatment is associated with multiple meningiomas and younger age at diagnosis. J. Neuro-Oncol. 2019, 143, 137–144. [Google Scholar] [CrossRef] [PubMed]
  61. Viaene, A.N.; Zhang, B.; Martinez-Lage, M.; Xiang, C.; Tosi, U.; Thawani, J.P.; Gungor, B.; Zhu, Y.; Roccograndi, L.; Zhang, L.; et al. Transcriptome signatures associated with meningioma progression. Acta Neuropathol. Commun. 2019, 7, 67. [Google Scholar] [CrossRef] [Green Version]
  62. Aghi, M.K.; Carter, B.S.; Cosgrove, G.R.; Ojemann, R.G.; Amin-Hanjani, S.; Martuza, R.L.; Curry, W.T., Jr.; Barker, F.G., 2nd. Long-term recurrence rates of atypical meningiomas after gross total resection with or without postoperative adjuvant radiation. Neurosurgery 2009, 64, 56–60. [Google Scholar] [CrossRef]
  63. Simpson, D. The recurrence of intracranial meningiomas after surgical treatment. J. Neurol. Neurosurg. Psychiatry 1957, 20, 22–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Goldbrunner, R.; Minniti, G.; Preusser, M.; Jenkinson, M.D.; Sallabanda, K.; Houdart, E.; von Deimling, A.; Stavrinou, P.; Lefranc, F.; Lund-Johansen, M.; et al. EANO guidelines for the diagnosis and treatment of meningiomas. Lancet Oncol. 2016, 17, e383–e391. [Google Scholar] [CrossRef] [Green Version]
  65. Hug, E.B.; Devries, A.; Thornton, A.F.; Munzenride, J.E.; Pardo, F.S.; Hedley-Whyte, E.T.; Bussiere, M.R.; Ojemann, R. Management of atypical and malignant meningiomas: Role of high-dose, 3D-conformal radiation therapy. J. Neuro-Oncol. 2000, 48, 151–160. [Google Scholar] [CrossRef] [PubMed]
  66. Gupta, S.; Bi, W.L.; Dunn, I.F. Medical management of meningioma in the era of precision medicine. Neurosurg. Focus 2018, 44, E3. [Google Scholar] [CrossRef] [PubMed]
  67. 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]
  68. 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] [PubMed] [Green Version]
  69. 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]
  70. Shih, K.C.; Chowdhary, S.; Rosenblatt, P.; Weir, A.B., 3rd; Shepard, G.C.; Williams, J.T.; Shastry, M.; Burris, H.A., 3rd; 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]
  71. Norden, A.D.; Raizer, J.J.; Abrey, L.E.; Lamborn, K.R.; Lassman, A.B.; Chang, S.M.; Yung, W.K.; 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. 2010, 96, 211–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chamberlain, M.C. Hydroxyurea for recurrent surgery and radiation refractory high-grade meningioma. J. Neuro-Oncol. 2012, 107, 315–321. [Google Scholar] [CrossRef] [PubMed]
  73. 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] [PubMed]
  74. 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 ItalianoCooperativo di Neuro-Oncologia (GICNO). Cancer Chemother. Pharmacol. 2016, 77, 115–120. [Google Scholar] [CrossRef]
  75. Chamberlain, M.C.; Tsao-Wei, D.D.; Groshen, S. Temozolomide for treatment-resistant recurrent meningioma. Neurology 2004, 62, 1210–1212. [Google Scholar] [CrossRef]
  76. 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]
  77. Preusser, M.; Silvani, A.; Rhun, E.L.; Soffietti, R.; Lombardi, G.; Sepúlveda, J.M.; Brandal, P.; Beaney, R.P.; Bonneville-Levard, A.; Lorgis, V.; et al. Trabectedin for recurrent WHO grade II or III meningioma: A randomized phase II study of the EORTC Brain Tumor Group (EORTC-1320-BTG). J. Clin. Oncol. 2019, 37, 2007. [Google Scholar] [CrossRef]
  78. 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]
  79. Le Rhun, E.; Taillibert, S.; Chamberlain, M.C. Systemic therapy for recurrent meningioma. Expert Rev. Neurother. 2016, 16, 889–901. [Google Scholar] [CrossRef]
  80. Nabors, L.B.; Portnow, J.; Ahluwalia, M.; Baehring, J.; Brem, H.; Brem, S.; Butowski, N.; Campian, J.L.; Clark, S.W.; Fabiano, A.J.; et al. Central Nervous System Cancers, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2020, 18, 1537–1570. [Google Scholar] [CrossRef]
  81. Dasanu, C.A.; Alvarez-Argote, J.; Limonadi, F.M.; Codreanu, I. Bevacizumab in refractory higher-grade and atypical meningioma: The current state of affairs. Expert Opin. Biol. Ther. 2019, 19, 99–104. [Google Scholar] [CrossRef]
  82. Goutagny, S.; Raymond, E.; Sterkers, O.; Colombani, J.M.; Kalamarides, M. Radiographic regression of cranial meningioma in a NF2 patient treated by bevacizumab. Ann. Oncol. 2011, 22, 990–991. [Google Scholar] [CrossRef]
  83. 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] [Green Version]
  84. Raizer, J.J.; Grimm, S.A.; Rademaker, A.; Chandler, J.P.; Muro, K.; Helenowski, I.; Rice, L.; McCarthy, K.; Johnston, S.K.; Mrugala, M.M.; et al. A phase II trial of PTK787/ZK 222584 in recurrent or progressive radiation and surgery refractory meningiomas. J. Neuro-Oncol. 2014, 117, 93–101. [Google Scholar] [CrossRef]
  85. 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]
  86. LekanneDeprez, R.H.; Bianchi, A.B.; Groen, N.A.; Seizinger, B.R.; Hagemeijer, A.; van Drunen, E.; Bootsma, D.; Koper, J.W.; Avezaat, C.J.; Kley, N.; et al. Frequent NF2 gene transcript mutations in sporadic meningiomas and vestibular schwannomas. Am. J. Hum. Genet. 1994, 54, 1022–1029. [Google Scholar]
  87. Pachow, D.; Andrae, N.; Kliese, N.; Angenstein, F.; Stork, O.; Wilisch-Neumann, A.; Kirches, E.; Mawrin, C. mTORC1 inhibitors suppress meningioma growth in mouse models. Clin. Cancer Res. 2013, 19, 1180–1189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. 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]
  89. von Spreckelsen, N.; Waldt, N.; Poetschke, R.; Kesseler, C.; Dohmen, H.; Jiao, H.K.; Nemeth, A.; Schob, S.; Scherlach, C.; Sandalcioglu, I.E.; et al. KLF4(K409Q)-mutated meningiomas show enhanced hypoxia signaling and respond to mTORC1 inhibitor treatment. Acta Neuropathol. Commun. 2020, 8, 41. [Google Scholar] [CrossRef] [PubMed]
  90. 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]
  91. Beauchamp, R.L.; James, M.F.; DeSouza, P.A.; Wagh, V.; Zhao, W.N.; Jordan, J.T.; Stemmer-Rachamimov, A.; Plotkin, S.R.; Gusella, J.F.; Haggarty, S.J.; et al. A high-throughput kinome screen reveals serum/glucocorticoid-regulated kinase 1 as a therapeutic target for NF2-deficient meningiomas. Oncotarget 2015, 6, 16981–16997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Franceschi, E.; Stupp, R.; van den Bent, M.J.; van Herpen, C.; LaigleDonadey, F.; Gorlia, T.; Hegi, M.; Lhermitte, B.; Strauss, L.C.; Allgeier, A.; et al. EORTC 26083 phase I/II trial of dasatinib in combination with CCNU in patients with recurrent glioblastoma. Neuro-Oncology 2012, 14, 1503–1510. [Google Scholar] [CrossRef] [Green Version]
  93. Angus, S.P.; Oblinger, J.L.; Stuhlmiller, T.J.; DeSouza, P.A.; Beauchamp, R.L.; Witt, L.; Chen, X.; Jordan, J.T.; Gilbert, T.S.K.; Stemmer-Rachamimov, A.; et al. EPH receptor signaling as a novel therapeutic target in NF2-deficient meningioma. Neuro-Oncology 2018, 20, 1185–1196. [Google Scholar] [CrossRef] [Green Version]
  94. Weller, M.; Roth, P.; Sahm, F.; Burghardt, I.; Schuknecht, B.; Rushing, E.J.; Regli, L.; Lindemann, J.P.; von Deimling, A. Durable Control of Metastatic AKT1-Mutant WHO Grade 1 Meningothelial Meningioma by the AKT Inhibitor, AZD5363. J. Natl. Cancer Inst. 2017, 109, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Shapiro, I.M.; Kolev, V.N.; Vidal, C.M.; Kadariya, Y.; Ring, J.E.; Wright, Q.; Weaver, D.T.; Menges, C.; Padval, M.; McClatchey, A.I.; et al. Merlin deficiency predicts FAK inhibitor sensitivity: A synthetic lethal relationship. Sci. Transl. Med. 2014, 6, 237ra68. [Google Scholar] [CrossRef] [Green Version]
  96. 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]
  97. Shankar, G.M.; Santagata, S. BAP1 mutations in high-grade meningioma: Implications for patient care. Neuro-Oncology 2017, 19, 1447–1456. [Google Scholar] [CrossRef] [Green Version]
  98. von Achenbach, C.; Le Rhun, E.; Sahm, F.; Wang, S.S.; Sievers, P.; Neidert, M.C.; Rushing, E.J.; Lawhon, T.; Schneider, H.; von Deimling, A.; et al. Sensitivity of human meningioma cells to the cyclin-dependent kinase inhibitor, TG02. Transl. Oncol. 2020, 13, 100852. [Google Scholar] [CrossRef]
  99. Tien, A.-C.; Li, J.; Bao, X.; DeRogatis, A.; Fujita, Y.; Pennington-Krygier, C.; Kim, S.; Mehta, S.; Sanai, N. MNGI-01. A phase 0 trial of ribociclib in aggressive meningioma patients incorporating a tumor pharmacodynamic- and pharmacokinetic-guided expansion cohort. Neuro-Oncology 2019, 21, vi139. [Google Scholar] [CrossRef]
  100. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [Green Version]
  101. Durand, A.; Champier, J.; Jouvet, A.; Labrousse, F.; Honnorat, J.; Guyotat, J.; Fèvre-Montange, M. Expression of c-Myc, neurofibromatosis Type 2, somatostatin receptor 2 and erb-B2 in human meningiomas: Relation to grades or histotypes. Clin. Neuropathol. 2008, 27, 334–345. [Google Scholar] [CrossRef]
  102. Graillon, T.; Romano, D.; Defilles, C.; Saveanu, A.; Mohamed, A.; Figarella-Branger, D.; Roche, P.H.; Fuentes, S.; Chinot, O.; Dufour, H.; et al. Octreotide therapy in meningiomas: In vitro study, clinical correlation, and literature review. J. Neurosurg. 2017, 127, 660–669. [Google Scholar] [CrossRef]
Table 1. Meningiomas grading and frequency [15].
Table 1. Meningiomas grading and frequency [15].
WHO GradeDescriptionFrequency (%)
Grade INo brain invasion
Mitotic rate < 4 per 10 HPFs
80–85
Grade II
Atypical
Brain invasion
Or
Mitotic rate 4–19 per 10 HPFs
Or
≥3 or 5 specific histological characteristics:
- spontaneous or geographic necrosis
- patternless sheet-like growth
- prominent nucleoli
- high cellularity
- small cells with high n:c ratio
15–20
Grade III
Anaplastic
Malignant
Mitotic rate > 20 per 10 HPFs
Or
Papillary or rhabdoid histologies
1–2
HPF: high-power field.
Table 2. Molecular alteration in meningiomas and correlated clinical characteristics.
Table 2. Molecular alteration in meningiomas and correlated clinical characteristics.
GeneMolecular AlterationPathwayTumor Histology and GradeGradeTumor LocalizationFrequency
NF2 [20,21,22,23,24,25,26]- 22q12 chromosome deletion
- Genomic instability
Activation of PI3K–AKT–mTOR pathway and Hippo pathwayFibrous and transitionalI–IIIHemispheres, often bilateral40–60%
SMO [20,29]- Leu412Pheand Trp535Leu mutations
- Genomic stability
Activation of PI3K–AKT–mTOR and SHH pathwayMeningothelialISkull base1–5%
AKT1 [20,30]p.Glu17Lys mutationActivation of PI3K–AKT–mTOR pathwayMeningothelialISpine and skull base7–12%
SUFU [30,31]Locus 10q24.32SHH pathwayNANANA1%
TRAF7 [23,27,30,32]WD40 domains mutationsJUN N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) signalingSecretoryI > II, IIIAnterior and middle medial skull base15–26%
PIK3CA [30]Locus 3q26.32, especially in H1047R, E542K and E545KActivation of PI3K–AKT–mTOR pathwayMeningothelial, transitionalI > II, IIISkull base4–7%
KLF4 [23,33]Locus 9p31Activation of PI3K–AKT–mTOR pathwaySecretoryIMiddle and lateral skull base9–12%
Up to half of NF2 nonmutated meningiomas
POLAR2 [28]- p.Gln403Lys mutation
- p.Leu438_His439del
TranscriptionMeningothelialITuberculum sellae6%
BAP1 [34,35]Multiple mutationsDNA repairRhabdoid, papillaryII, IIIConvexity<1%
PBRM1 [35]Locus 3p21.1Chromatin remodelingPapillary, rhabdoidNANA2.8%
Epigenetic alterations [20,36,37,38]Mutations in KDM5C, KDM6A, SMARCB1, EZH2Chromatin remodelingMultiple pathogenic variant
EZH2: High grade
NANA10% of non-NF2 meningiomas
DMD [39]Locus Xp21.1CytoskeletonNAII, IIINANA
CDKNA2A/B [38,47,48,49,50]Somatic mutations and homozygous lossesCell cycle regulationNAII, IIINA<5%
TERTp [40,41,42,43,44]Locus sp15.33 in C228T and C250TTelomerase activityNAI-IIINA4.7–15.4%
CHECK2 [52]Locus 22q12.1Cell cycle regulationNAINARare
FAK [19]Locus 8q24.3Cell motilityNANANARare
NA: Not available.
Table 3. Simpson grade [63].
Table 3. Simpson grade [63].
1Complete resection, with dural and bone resection
2Gross total resection with dural coagulation
3Macroscopic resection, without dural excision or coagulation
4Subtotal resection
5Biopsy
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Maggio, I.; Franceschi, E.; Di Nunno, V.; Gatto, L.; Tosoni, A.; Angelini, D.; Bartolini, S.; Lodi, R.; Brandes, A.A. Discovering the Molecular Landscape of Meningioma: The Struggle to Find New Therapeutic Targets. Diagnostics 2021, 11, 1852. https://doi.org/10.3390/diagnostics11101852

AMA Style

Maggio I, Franceschi E, Di Nunno V, Gatto L, Tosoni A, Angelini D, Bartolini S, Lodi R, Brandes AA. Discovering the Molecular Landscape of Meningioma: The Struggle to Find New Therapeutic Targets. Diagnostics. 2021; 11(10):1852. https://doi.org/10.3390/diagnostics11101852

Chicago/Turabian Style

Maggio, Ilaria, Enrico Franceschi, Vincenzo Di Nunno, Lidia Gatto, Alicia Tosoni, Daniele Angelini, Stefania Bartolini, Raffaele Lodi, and Alba Ariela Brandes. 2021. "Discovering the Molecular Landscape of Meningioma: The Struggle to Find New Therapeutic Targets" Diagnostics 11, no. 10: 1852. https://doi.org/10.3390/diagnostics11101852

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