Novel Advances in Treatment of Meningiomas: Comparison
Please note this is a comparison between Version 2 by Camila Xu and Version 1 by Maria Caffo.

Meningiomas are extra-axial, slow-growing, and (usually) benign tumors. These tumors arise from meningothelial cells of the arachnoid layer, so they can be encountered anywhere this type of cell is localized.

  • biomarkers
  • genetics
  • meningioma
  • molecular therapy

1. Introduction

Meningiomas are the most frequent histotype of tumors of the central nervous system (CNS). Their incidence is approximately 35% of all primary brain tumors and approximately 50% of all benign cerebral tumors [1]. These neoplasms are classified into three grades according to the World Health Organization (WHO) [2]. Commonly, patients with meningioma may present with headache, onset of critical episodes, altered mental status, speech impairment, strength impairment, and cranial nerve deficits. The neurological signs are clearly related to the site of the lesion. Initial treatment involves observation and surgical treatment, while radiotherapy and radiosurgery should be considered in cases of atypical and anaplastic meningioma. The site of the lesion as well as close proximity to vital structures can cause complications during surgical procedures. The treatment of recurrent tumors includes radiotherapy and repeated surgery. Some histotypes, despite surgical removal, show an aggressive trend with an early tendency toward recurrence.

2. Meningiomas

Meningiomas are extra-axial, slow-growing, and (usually) benign tumors. These tumors arise from meningothelial cells of the arachnoid layer, so they can be encountered anywhere this type of cell is localized. The most common locations (which make up about 50% of cases) are para-sagittal (28.8%), convexity (15.2%) and tuberculum sellae (12.8%). According to the statistical USA report by Ostrom et al. [1], meningiomas represent 36.6% of all primary CNS tumors and 53.2% of non-malignant primary CNS tumors. Although they have the reputation of benign lesions, meningiomas are often associated with a decreased quality of life (QoL) due to focal neurological deficits that may be related, and in 20% of cases, they display an aggressive behavior, even when the best standard of care is provided [3]. Meningiomas are most common in the elderly population (the median age at diagnosis is 66 years [1], and the occurrence risk increases with age) and have a female preponderance (1.8:1) [4]; however, grades II and III occur more often in males [5]. Only 1.5% of meningiomas occur in childhood or adolescence, and about 30% of these have an intra-ventricular localization [4]. Overall 5-year survival varies with age between 97% and 87.3% for non-malignant cases and between 85 and 50.2% for the malignant meningiomas [6]. Meningioma is considered a single type in the WHO CNS, and its broad morphological spectrum is reflected in 15 subtypes [2]. It is now emphasized that the criteria defining atypical or anaplastic (i.e., grade 2 and 3) meningioma should be applied regardless of the underlying subtype. As in prior classifications, chordoid and clear cell meningioma are noted to have a higher likelihood of recurrence than the average CNS WHO grade 1 meningioma and have hence been assigned to CNS WHO grade 2; however, larger and prospective studies would be helpful to validate these suggested CNS WHO grade 2 assignments and to suggest additional prognostic biomarkers. While papillary and rhabdoid features are often seen in combination with other aggressive features, more recent studies suggest that the grading of these tumors should not be carried out on the basis of rhabdoid cytology or papillary architecture alone. Several molecular biomarkers are associated with classification and grading of meningiomas, including SMARCE1 (clear cell subtype), BAP1 (rhabdoid and papillary subtypes), KLF4/TRAF (secretory subtype), TERT promoter mutation and/or homozygous deletion of CDKN2A/B (WHO grade 3), H3K27me3 loss of nuclear expression (potentially worse prognosis), and methylome profiling (prognostic subtyping) [7,8,9,10,11][7][8][9][10][11]. In meningiomas with a high risk of recurrence, meningiomas with a high proliferation index may be included, characterized by frequent mitotic figures and recurrence even after an apparently total removal, as well as particular subtypes of meningiomas such as atypical or rhabdoid meningiomas [4].

2.1. Risk Factors

The onset of meningiomas can be linked to environmental factors such as ionizing radiation. Ionizing radiation is the only established environmental risk factor, with higher risk in patients who have undergone radiation therapy [4]. It seems there is a genetic susceptibility to the development of radiation-induced meningiomas [12]. Numerous studies searching for the correlation between meningiomas and environmental factors (e.g., hormonal influence, diet, allergies, or phone use) have been conducted, but none with statistically significant results. Meningiomas occur with greater frequency in patients with germline mutations of genes such as type-2 neurofibromatosis (NF2) [13], type-1 neurofibromatosis (NF1, 19–24% of adolescent meningiomas) [4], type-1 multiple endocrine neoplasia (MEN1) [14], SMARCB1, LZTR1, or SMARCE1 genes [5].

2.2. Diagnosis

The gold standard for diagnosis and surveillance of meningiomas is magnetic resonance imaging (MRI). In MRI sequences, meningiomas appear as well-circumscribed and dural-based lesions, isointense to gray matter on non-contrast sequences, and homogeneously enhanced with gadolinium. Meningiomas may or not be associated with brain edema. A common but non-pathognomonic finding, especially in benign lesions, is the so-called dural tail: an enhancement of the dura due to a thickening of the dural layer adjacent to the mass. Moreover, MRI is very accurate in predicting venous sinus involvement (accuracy of about 90%) [15]. Meningiomas are usually single lesions. However, sometimes multiple lesions may suggest a genetic syndrome (e.g., NF2) or the presence of metastases. Other possible diseases to keep in consideration in differential diagnosis are [4,16][4][16]: pleomorphic xanthoastrocytoma which tend to be peripherally located and may have dural tail, dural metastases, gliosarcoma, Rosai-Dorfman disease, a connective tissue disorder with sinus histiocytosis, massive painless lymphadenopathy and dural based enhancing masses with characteristics similar to meningioma.

3. Treatment

3.1. Surgery

While radiological surveillance may be an acceptable strategy for many patients who present with asymptomatic incidental meningiomas [17], for growing and symptomatic tumors the standard of care remains maximal safe surgical resection. Surgical treatment allows for the removal of the lesion and, consequently, of the bulking effect; clinically, the improvement of neurological functions and the resolution of seizures can be seen [18,19][18][19]. In meningioma surgery, the optimal goal is, whenever safely possible, the complete removal of the lesion and affected tissue. This minimizes neurological morbidity and allows for greater long-term control. Surgical treatment also makes it possible to obtain the correct histological diagnosis. Minimally invasive neurosurgery refers to the technological advances made in improving surgical access, thereby enabling neurosurgeons to reduce morbidity and greatly improve the precision of neurosurgical procedures. The introduction of the operating microscope and advances in neuroendoscopy have allowed for the refinement of lighting and the magnification of deep structures [20]. The use of micro-tools allows for the fine dissection of the neoplastic lesion from the nerves and vascular structures. Furthermore, advances in surgical neuronavigation and neuromonitoring allow, during the surgical approach, for minimum brain retraction, as well as the ability to highlight ischemic alterations and pressure variations in eloquent areas. In any case, surgical removal is limited by various factors, such as the location of the tumor and the incorporation within the lesion of nerves, venous sinuses, veins, and arteries. Simpson’s grading scale (Table 1) correlates to the extent of tumor resection, associated dural attachments, and any hyperostotic bone to local recurrence risk and five defined grades of resection, which are associated with distinct rates of recurrence [21].
Table 1.
The Simpson Grade.
In cases of partial occlusion of a venous sinus, the tumor component is not removed due to the high risk of hemorrhage, thrombosis, and gas embolism. Removal of meningiomas at the base of the skull, due to their relationship to bony anatomy (sphenoid wing, olfactory sulcus, saddle tubercle, ponto-cerebellar angle, or petroclival region) or those involving blood vessels and cranial nerves are at higher risk and require more advanced surgical techniques in order to minimize brain retraction and protect neurovascular structures [22]. Several midline anterior skull-base tumors are resected via an endoscopic endonasal approach. The advantages of this approach are the visualization of the ventral side of the deep skull-base tumor and safer resection which avoids traction of the brain tissue during the operation.

3.2. Radiation Therapy

Radiation therapy (RT) has commonly been utilized following subtotal resection surgery as an adjuvant therapy for recurrence of previously resected WHO grade II or grade III meningiomas [23]. Radiation therapy is individualized and must be chosen depending on meningioma size, proximity to critical structures, and any prior radiation to the same site. The goal of RT is to reduce meningioma’s proliferation and control its progress. Stereotactic radiotherapy (SRT) is defined as a method of external beam radiotherapy, in which a defined target volume is treated with a high radiation dose in up to 12 fractions, delivered on separate days of treatment. By means of SRT technology, it is possible to irradiate a specific target with a high dose in a single time; the dose of radiation absorbed outside the target area is inversely proportional to the distance from it, and the peripheral tissues around the focus are not damaged. Stereotactic radiosurgery (SRS) is the precise, single-session delivery of a therapeutically effective radiation dose to a certain target. SRS utilizes a single fraction of high-dose radiation but is associated with peculiar toxicities such as radiation-induced brain necrosis or intralesional hemorrhage [24]. Gamma knife radiosurgery (also known as stereotactic radiosurgery) focuses many tiny beams of radiation with extreme accuracy on a target. Each beam has very little effect on the brain tissue it passes through. Fractional stereotactic radiation therapy (FSRT) is administered over the course of several days, rather than in a single dose, reducing the dose exposure to normal brain tissue. SRS was developed by combining radiotherapy and stereotaxis. SRS is a widely accepted technique for small grade I or II lesions, while EBRT is recommended for grade III meningiomas, which require larger doses (50–60 Gy) to achieve local control [25,26][25][26]. Single-fraction SRS is typically used in meningiomas with a maximum diameter of <3 cm, located more than 3 mm from radiosensitive structures [27]. Although surgery remains the primary option, radiotherapy has become a first-line option for some meningiomas, particularly lesions of the cranial base that enclose vascular-nerve structures such as the optic nerve sheath or the cavernous sinus. Radiation therapy and fractional and hypofractionated stereotaxic radiosurgery, in single or multiple doses, have been shown to be beneficial for patients with a high rate of tumor control ranging from 85 to 100% at 5 years [28]. Side effects of stereotaxic radiotherapy for small tumors are mild [29[29][30],30], but cases of radionecrosis have been reported, and pituitary function should also be monitored after skull-base irradiation [31]. A long-term study of 290 consecutive patients showed progression-free control rates of 88.7 and 87.2% at 10 years and 20 years at follow-up, respectively, with adverse radiation effects in 3.1% of patients [32]. With the advent of SRS, surgical goals may be modified with plans for less-aggressive surgical resections, with the knowledge that radiosurgery offers an excellent option for long-term management of incompletely resected meningiomas. This aspect is particularly recommended for large meningiomas in surgically inaccessible locations. Such lesions may require pre-surgical planning for subtotal surgical resections, with planned adjuvant gamma knife radiosurgery to treat residual tumor due to its capacity to provide excellent long-term control of tumor growth while minimizing injury associated with tumor removal.

3.3. Chemotherapy

At long-term follow-up, up to 60% of meningiomas can recur after 15 years and exhibit aggressive behavior. RTOG 0539, a phase II trial that stratified meningioma risk based on pathologic grade and extent of resection, outlines options for postoperative management [33]. In RTOG 0539, meningiomas were stratified as follows: low risk—grade I and gross total resection (GTR) or subtotal resection (STR); intermediate risk—recurrent grade I or grade II after GTR; and high risk—STR or recurrent grade II and any grade III. Low-risk tumors demonstrated a progression-free survival (PFS) at 3 years of 92%, intermediate risk PFS of 94%, and high-risk PFS of 59% [34]. Due to their limited effectiveness, systemic therapies should be considered once all surgical and radiotherapy possibilities have been excluded and should be planned on an individual basis. There is little evidence in the literature to support systemic therapeutic treatment, and numerous clinical trials and case series have shown that chemotherapy has a minimal role and does not improve patients’ outcomes [35,36][35][36]. A major problem in interpreting the published literature on medical therapies for recurrent meningioma is the inclusion of different histology in reports of patients at various stages of their disease, ranging from newly diagnosed tumors to tumors that have relapsed after multiple surgeries and radiotherapy treatments and, in some cases, multiple chemotherapy regimens. Classic chemotherapeutic agents, including temozolomide, irinotecan, doxorubicin, ifosfamide, adriamycin, and vincristine, have not been shown to be effective against meningiomas [37,38][37][38]. Hydroxyurea, a ribonucleotide reductase inhibitor, can induce apoptosis and cell cycle arrest in the S-phase. An Italian randomized study showed the association between hydroxyurea, with or without imatinib, and recurrent or progressive meningiomas without, however, reaching conclusive data due to the small number of patients enrolled [39]. Hydroxyurea has shown stabilizing activity in only a few cases, but this has not been fully confirmed. It can be considered another adjuvant tool for atypical meningiomas if postoperative adjuvant radiotherapy cannot be applied. A recent study suggested that adjuvant treatment after STR of atypical meningiomas correlates with a longer PFS than conservative treatment and that there are no significant differences in PFS between hydroxyurea chemotherapy and radiotherapy after surgery [40]. In addition, hydroxyurea chemotherapy was shown to be effective in a retrospective study that analyzed 19 patients with atypical meningiomas. These were treated with hydroxyurea after GKR. The results of the present study suggest the safety and efficacy of HU after GKR with stabilization or shrinkage of atypical (grade II) meningiomas [41]. The possibility of systemic treatment as adjuvant therapy after surgery was also evaluated in a prospective study that enrolled 14 patients with malignant meningioma. After surgery and 2–4 weeks of radiotherapy (median dose 60 Gy), all patients were treated with cyclophosphamide, Adriamycin, and vincristine. This approach demonstrated moderate efficacy with partial response and disease stability in 3 and 11 patients, respectively, resulting in a median overall survival of 5.3 years and progression-free survival of 4.6 years [42].

4. Genetic and Biomarkers for Meningiomas

In the past years, thanks to new technologies in genomics, knowledge of genetic factors underlying the development of meningiomas has been significantly improved. With the identification of neurofibromin 2 (NF2) located on 22q12.2 (NF2 codes for the Merlin protein, which shows tumor suppressor properties) and the high percentage of patients (about 50–75%) with NF2 who develop one or more meningiomas, meningiomas represent one of the first types of tumors linked to a genomic driver [14]. Recently, thanks to the advent of next-generation sequencing (NGS), numerous additional somatic mutations have been identified (Table 2), and this has promising implications for new frontiers in target therapy. For this reason, to date, the mutational landscape can be divided into NF2-mutated (approximately 40–60% of cases) and non-NF2-mutated meningioma [43]. Among the non-NF2-mutated group, the most common oncogene in WHO grade I meningiomas is TNF receptor-activated factor 7 (TRAF7), located on chromosome 16p13 (mutated in nearly 25% of all meningiomas), followed by the mutation in codon K409Q of KLF4 (encountered in about 15% of benign meningiomas) [44,45][44][45]. The mutation of TRAF 7 is frequently associated with mutations of AKT1 (which encodes for a kinase that regulates cell proliferation) or KLF4, and this combination is often linked to grade 1 meningiomas [44,45][44][45]. AKT1 mutation occurs at the E17 location, triggering activation of the mTOR and ERK1/2 pathways. The AKT1 p.Glu 17 Lys mutation triggers the abnormal activation of the PI3K pathway, indicating a key role in meningiomas proliferation [46]. POLR2A mutant tumors show dysregulation of key meningeal identity genes, including WNT6 and ZIC1/ZIC4 [47]. Still, smoothened mutations (SMOs) trigger the Sonic Hedgehog signaling pathway promoting angiogenesis and tumor progression [48]. Mutations in Krüppel-like factor 4 (KLF4), a transcription factor in oncogenic activation, have been detected in about 50% of NF2-nonmutated meningiomas [45,49][45][49]. As shown in Table 1, in 20% of cases, the oncogenic mutations remain unclear. Notably, mutations of these genes in meningiomas occur to a large degree without concurrent alteration of NF2 or loss of chromosome 22 [50]. Other rarer mutations include SWI/SNF-related matrix-associated actin-dependent regulator of chromatin, subfamily B member 1 (SMARCB1), SMARCE1, and SUFU genes [51,52,53][51][52][53]. SMARCB1 mutation has also been shown to co-occur in NF2-mutated meningiomas [47]. In higher grade meningiomas, other genomic alterations with independent prognostic value have been reported, namely mutation of TERT promoter and deletions of CDKN2A/B [54,55][54][55]. The CDKN2A and BAP1 mutation seem to be associated with aggressive forms of meningioma [8[8][56],56], while the SMASRCE1 mutation is attributable to the onset of spinal meningiomas [52].
Table 2.
Most common mutations in WHO grade I meningiomas.

References

  1. Ostrom, Q.T.; Gittleman, H.; Xu, J.; Kromer, C.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2009–2013. Neuro-Oncology 2016, 18, v1–v75.
  2. 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.
  3. Birzu, C.; Peyre, M.; Sahm, F. Molecular Alterations in Meningioma: Prognostic and Therapeutic Perspectives. Curr. Opin. Oncol. 2020, 32, 613–622.
  4. Greenberg, M.S. Handbook of Neurosurgery, 9th ed.; Thieme: Stuttgart, Germany, 2019; ISBN 978-1-68420-137-2.
  5. Kim, L. A Narrative Review of Targeted Therapies in Meningioma. Chin. Clin. Oncol. 2020, 9, 76.
  6. Ostrom, Q.T.; Truitt, G.; Gittleman, H.; Brat, D.J.; Kruchko, C.; Wilson, R.; Barnholtz-Sloan, J.S. Relative Survival after Diagnosis with a Primary Brain or Other Central Nervous System Tumor in the National Program of Cancer Registries, 2004 to 2014. Neuro-Oncol. Pract. 2020, 7, 306–312.
  7. Tauziede-Espariat, A.; Parfait, B.; Besnard, A.; Lacombe, J.; Pallud, J.; Tazi, S.; Puget, S.; Lot, G.; Terris, B.; Cohen, J.; et al. Loss of SMARCE1 Expression Is a Specific Diagnostic Marker of Clear Cell Meningioma: A Comprehensive Immunophenotypical and Molecular Analysis. Brain Pathol. Zurich Switz. 2018, 28, 466–474.
  8. Shankar, G.M.; Abedalthagafi, M.; Vaubel, R.A.; Merrill, P.H.; Nayyar, N.; Gill, C.M.; Brewester, 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.
  9. Williams, E.A.; Wakimoto, H.; Shankar, G.M.; Barker, F.G.; 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.
  10. Lee, Y.S.; Lee, Y.S. Molecular Characteristics of Meningiomas. J. Pathol. Transl. Med. 2020, 54, 45–63.
  11. Nassiri, F.; Wang, J.Z.; Singh, O.; Karimi, S.; Dalcourt, T.; Ijad, N.; Pirouzmand, N.; Ng, H.K.; Saladino, A.; Pollo, B.; et al. Loss of H3K27me3 in Meningiomas. Neuro Oncol. 2021, 23, 1282–1291.
  12. Flint-Richter, P.; Sadetzki, S. Genetic Predisposition for the Development of Radiation-Associated Meningioma: An Epidemiological Study. Lancet Oncol. 2007, 8, 403–410.
  13. Perry, A.; Giannini, C.; Raghavan, R.; Scheithauer, B.W.; Banerjee, R.; Margraf, L.; Bowers, D.C.; Lytle, R.A.; Newsham, I.F.; Gutmann, D.H. Aggressive Phenotypic and Genotypic Features in Pediatric and NF2-Associated Meningiomas: A Clinicopathologic Study of 53 Cases. J. Neuropathol. Exp. Neurol. 2001, 60, 994–1003.
  14. Asgharian, B.; Chen, Y.-J.; Patronas, N.J.; Peghini, P.L.; Reynolds, J.C.; Vortmeyer, A.; Zhuang, Z.; Venzon, D.J.; Gibril, F.; Jensen, R.T. Meningiomas May Be a Component Tumor of Multiple Endocrine Neoplasia Type 1. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2004, 10, 869–880.
  15. Zimmerman, R.D.; Fleming, C.A.; Saint-Louis, L.A.; Lee, B.C.; Manning, J.J.; Deck, M.D. Magnetic Resonance Imaging of Meningiomas. AJNR Am. J. Neuroradiol. 1985, 6, 149–157.
  16. Caffo, M.; Curcio, A.; Laera, R.; Barresi, V.; Caruso, G. Intracranial Dural Metastases and Diagnostic Misunderstandings. J. Analyt. Oncol. 2022, 11, 54–62.
  17. Islim, A.I.; Kolamunnage-Dona, R.; Mohan, M.; Moon, R.D.C.; Crofton, A.; Haylock, B.J.; Rathi, N.; Brodbelt, A.R.; Mills, S.J.; Jenkinson, M.D. A Prognostic Model to Personalize Monitoring Regimes for Patients with Incidental Asymptomatic Meningiomas. Neuro-Oncology 2020, 22, 278–289.
  18. Englot, D.J.; Magill, S.T.; Han, S.J.; Chang, E.F.; Berger, M.S.; McDermott, M.W. Seizures in Supratentorial Meningioma: A Systematic Review and Meta-Analysis. J. Neurosurg. 2016, 124, 1552–1561.
  19. Chen, W.C.; Magill, S.T.; Englot, D.J.; Baal, J.D.; Wagle, S.; Rick, J.W.; McDermott, M.W. Factors Associated with Pre- and Postoperative Seizures in 1033 Patients Undergoing Supratentorial Meningioma Resection. Neurosurgery 2017, 81, 297–306.
  20. Bailo, M.; Gagliardi, F.; Boari, N.; Castellano, A.; Spina, A.; Mortini, P. The Role of Surgery in Meningiomas. Curr. Treat. Options Neurol. 2019, 21, 51.
  21. Rogers, L.; Barani, I.; Chamberlain, M.; Kaley, T.; 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.
  22. Zada, G.; Başkaya, M.K.; Shah, M.V. Introduction: Surgical Management of Skull Base Meningiomas. Neurosurg. Focus 2017, 43.
  23. Goldbrunner, R.; Minniti, G.; Preusser, M.; Jenkinson, M.D.; Sallabanda, K.; Houdart, E.; von Diemling, 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.
  24. Jimenez, R.B.; Alexander, B.M.; Mahadevan, A.; Niemierko, A.; Rajakesari, S.; Arvold, N.D.; Floyd, S.R.; Oh, K.S.; Loeffler, J.S.; Shih, H.A. The impact of Different Stereotactic Radiation Therapy Regimens for Brain Metastases on Local Control and Toxicity. Adv. Rad. Oncol. 2017, 2, 391–397.
  25. Cohen-Inbar, O.; Lee, C.C.; Sheehan, J.P. The Contemporary Role of Stereotactic Radiosurgery in the Treatment of Meningiomas. Neurosurg. Clin. N. Am. 2016, 27, 215–228.
  26. Buerki, R.A.; Horbinski, C.M.; Kruser, T.; Horowitz, P.M.; James, C.D.; Lukas, R.M. An Overview of Meningiomas. Future Oncol. 2018, 14, 2161–2177.
  27. Lee, J.Y.K.; Niranjan, A.; McInerney, J.; Kondziolka, D.; Flickinger, J.C.; Lunsford, L.D. Stereotactic Radiosurgery Providing Long-Term Tumor Control of Cavernous Sinus Meningiomas. J. Neurosurg. 2002, 97, 65–72.
  28. Patibandla, M.R.; Lee, C.-C.; Sheehan, J. Stereotactic Radiosurgery of Central Skull Base Meningiomas-Volumetric Evaluation and Long-Term Outcomes. World Neurosurg. 2017, 108, 176–184.
  29. Valery, C.A.; Faillot, M.; Lamproglou, I.; Golmard, J.-L.; Jenny, C.; Peyre, M.; Mokhtari, K.; Mazeron, J.; Cornu, P.; Kalamarides, M. Grade II Meningiomas and Gamma Knife Radiosurgery: Analysis of Success and Failure to Improve Treatment Paradigm. J. Neurosurg. 2016, 125, 89–96.
  30. Cho, M.; Joo, J.-D.; Kim, I.A.; Han, J.H.; Oh, C.W.; Kim, C.-Y. The Role of Adjuvant Treatment in Patients with High-Grade Meningioma. J. Korean Neurosurg. Soc. 2017, 60, 527–533.
  31. Minniti, G.; Clarke, E.; Cavallo, L.; Osti, M.F.; Esposito, V.; Cantore, G.; Cappabianca, P.; Enrici, R.M. Fractionated Stereotactic Conformal Radiotherapy for Large Benign Skull Base Meningiomas. Radiat. Oncol. Lond. Engl. 2011, 6, 36.
  32. Santacroce, A.; Walier, M.; Régis, J.; Liščák, R.; Motti, E.; Lindquist, C.; Kemeny, A.; Kitz, K.; Lippitz, B.; Martinez Alvarez, R.; et al. Long-Term Tumor Control of Benign Intracranial Meningiomas after Radiosurgery in a Series of 4565 Patients. Neurosurgery 2012, 70, 32–39; discussion 39.
  33. Rogers, L.; Zhang, P.; Vogelbaum, M.A.; Perry, A.; Ashby, L.S.; Modi, J.M.; Alleman, A.M.; Brachman, D.; Jenrette, J.M.; De Groot, J.; et al. Intermediate-Risk Meningioma: Initial Outcomes from NRG Oncology RTOG 0539. J. Neurosurg. 2017, 129, 35–47.
  34. Farooqi, A.; Li, J.; de Groot, J.; Yeboa, D.N. Current Role of Radiation Therapy in the Management of Malignant Central Nervous System Tumors. Hematol. Oncol. Clin. North Am. 2020, 34, 13–28.
  35. Sioka, C.; Kyritsis, A.P. Chemotherapy, Hormonal Therapy, and Immunotherapy for Recurrent Meningiomas. J. Neurooncol. 2009, 92, 1–6.
  36. 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. Neurooncol. 2010, 96, 211–217.
  37. Wilson, T.A.; Huang, L.; Ramanathan, D.; Lopez-Gonzalez, M.; Pillai, P.; De Los Reyes, K.; Kumal, M.; Boling, W. Review of Atypical and Anaplastic Meningiomas: Classification, Molecular Biology, and Management. Front. Oncol. 2020, 10, 565582.
  38. Chamberlain, M.C.; Tsao-Wei, D.D.; Groshen, S. Salvage Chemotherapy with CPT-11 for Recurrent Meningioma. J. Neurooncol. 2006, 78, 271–276.
  39. 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.
  40. Kim, J.; Kim, K.H.; Kim, Y.Z. The Clinical Outcome of Hydroxyurea Chemotherapy after Incomplete Resection of Atypical Meningiomas. Brain Tumor Res. Treat. 2017, 5, 77–86.
  41. Abdel Karim, K.; El Shehaby, A.; Emad, R.; Reda, W.; El Mahdy, M.; Ghali, R.; Nabeel, A. Role of Hydroxyurea as an Adjuvant Treatment after Gamma Knife Radiosurgery for Atypical (WHO Grade II) Meningiomas. J. Egypt. Natl. Cancer Inst. 2018, 30, 69–72.
  42. Chamberlain, M.C. Adjuvant Combined Modality Therapy for Malignant Meningiomas. J. Neurosurg. 1996, 84, 733–736.
  43. Dumanski, J.P.; Rouleau, G.A.; Nordenskjold, M.; Collins, V.P. Molecular Genetic Analysis of Chromosome 22 in 81 Cases of Meningioma. Cancer Res. 1990, 50, 5863–5867.
  44. Clark, V.E.; Erson-Omay, E.Z.; Serin, A.; Yin, J.; Cotney, J.; Ozduman, K.; Avsar, 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.
  45. 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.
  46. Pachow, D.; Wick, W.; Gutmann, D.H.; Mawrin, C. The mTOR Signaling Pathway as a Treatment Target for Intracranial Neoplasms. Neuro-Oncology 2015, 17, 189–199.
  47. Clark, V.E.; Harmanci, A.S.; Bai, H.; Youngblood, M.W.; Lee, T.I.; Baranoski, J.F.; Erkan-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.
  48. 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.
  49. Moussalem, C.; Massaad, E.; Minassian, G.B.; Ftouni, L.; Bsat, S.; El Houshiemy, M.N.; Alomari, S.; Sarieddine, R.; Kobeissy, F.; Omeis, I. Meningioma Genomics: A Therapeutic Challenge for Clinicians. J. Integr. Neurosci. 2021, 20, 463–469.
  50. Bi, W.L.; Zhang, M.; Wu, W.W.; Mei, Y.; Dunn, I.F. Meningioma Genomics: Diagnostic, Prognostic, and Therapeutic Applications. Front. Surg. 2016, 3, 40.
  51. Wang, A.S.; Jamshidi, A.O.; Oh, N.; Sahyouni, R.; Nowroozizadeh, B.; Kim, R.; Hsu, F.P.K.; Bota, D. Somatic SMARCB1 Mutation in Sporadic Multiple Meningiomas: Case Report. Front. Neurol. 2018, 9, 919.
  52. 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.
  53. Aavikko, M.; Li, S.; Saarinen, S.; Alhopuro, P.; Kaasinen, E.; Morgunova, E.; Li, Y.; Vesanen, K.; Smith, M.J.; Gareth, D.; et al. Loss of SUFU Function in Familial Multiple Meningioma. Am. J. Hum. Genet. 2012, 91, 520–526.
  54. 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.
  55. Barresi, V.; Simbolo, M.; Fioravanzo, A.; Piredda, M.L.; Caffo, M.; Ghimenton, C.; Pinna, G.; Longhi, M.; Nicolato, A.; Scarpa, A. Molecular Profiling of 22 Primary Atypical Meningiomas Shows the Prognostic Significance of 18q Heterozygous Loss and CDKN2A/B Homozygous Deletion on Recurrence-Free Survival. Cancers 2021, 13, 903.
  56. Wang, J.Z.; Patil, V.; Liu, J.; Dogan, H.; Tabatabai, G.; Yefet, L.S.; Behling, F.; Hoffman, E.; Bunda, S.; Yakubov, R.; et al. Increased mRNA Expression of CDKN2A is a Transcriptomic Marker of Clinically Aggressive Meningiomas. Acta Neuropathol. 2023, 46, 145–162.
  57. 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.
  58. Okano, A.; Miyawaki, S.; Hongo, H.; Dofuku, S.; Teranishi, Y.; Mitsui, J.; Tanaka, M.; Shin, M.; Nakatomi, H.; Saito, N. Associations of Pathological Diagnosis and Genetic Abnormalities in Meningiomas with the Embryological Origins of the Meninges. Sci. Rep. 2021, 11, 6987.
  59. 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.
  60. Youngblood, M.W.; Duran, D.; Montejo, J.D.; Li, C.; Omay, S.B.; Ozduman, K.; Sheth, A.H.; Zhao, A.Y.; Tyrtova, E.; Miyagishima, D.F.; et al. Correlations Between Genomic Subgroup and Clinical Features in a Cohort of More Than 3000 Meningiomas. J. Neurosurg. 2019, 133, 1345–1354.
  61. Sievers, P.; Sill, M.; Blume, C.; Tauziede-Espariat, A.; Schrimpf, D.; Stichel, D.; Reuss, D.E.; Dogan, H.; Hartmann, C.; Mawrin, C.; et al. Clear Cell Meningiomas are Defined by a Highly Distinct DNA Methylation Profile and Mutations in SMARCE1. Acta Neuropathol. 2021, 141, 281–290.
  62. Nassiri, F.; Liu, J.; Patil, V.; Mamatjan, Y.; Wang, J.Z.; Hugh-White, R.; Macklin, A.M.; Khan, S.; Singh, O.; Karimi, S.; et al. A Clinically Applicable Integrative Molecular Classification of Meningiomas. Nature 2021, 597, 119–125.
  63. Deng, J.; Hua, L.; Bian, L.; Chen, H.; Chen, L.; Cheng, H.; Dou, C.; Geng, D.; Hong, T.; Ji, H.; et al. Molecular Diagnosis and Treatment of Meningiomas: An Expert Consensus (2022). Chin. Med. J. 2022, 135, 1894–1912.
  64. Caffo, M.; Pino, M.A.; Caruso, G.; Tomasello, F. Antisense Molecular Therapy in Cerebral Gliomas. J. Analyt. Oncol. 2012, 1, 135–144.
  65. Yamasaki, F.; Yoshioka, H.; Hama, S.; Sugiyama, K.; Arita, K.; Kurisu, K. Recurrence of Meningiomas. Cancer 2000, 89, 1102–1110.
  66. Sakuma, T.; Nakagawa, T.; Ido, K.; Takeuchi, H.; Sato, K.; Kubota, T. Expression of Vascular Endothelial Growth Factor-A and mRNA Stability Factor HuR in Human Meningiomas. J. Neurooncol. 2008, 88, 143–155.
  67. Barresi, V.; Tuccari, G. Increased Ratio of Vascular Endothelial Growth Factor to Semaphorin3A is a Negative Prognostic Factor in Human Meningiomas. Neuropathology 2010, 30, 537–546.
  68. Baxter, D.S.; Orrego, A.; Rosenfeld, J.V.; Mathiesen, T. An Audit of Immunohistochemical Marker Patterns in Meningioma. J. Clin. Neurosci. 2014, 21, 421–426.
  69. Pistolesi, S.; Fontanini, G.; Camacci, T.; de Ieso, K.; Boldrini, L.; Lupi, G.; Caniglia, M.; Mariani, G.; Boni, G.; Suriano, S.; et al. Meningioma-Associated Brain Oedema: The Role of Angiogenic Factors and Pial Blood Supply. J. Neurooncol. 2002, 60, 159–164.
  70. Denizot, Y.; de Armas, R.; Caire, F.; Moreau, J.J.; Pommepuy, I.; Truffinet, V.; Labrousse, F. The Quantitative Analysis of bFGF and VEGF by ELISA in Human Meningiomas. Mediators Inflamm. 2006, 2006, 36376.
  71. Winter, R.C.; Antunes, A.C.M.; de Oliveira, F.H. The Relationship Between Vascular Endothelial Growth Factor and Histological Grade in Intracranial Meningioma. Surg. Neurol. Int. 2020, 11, 328.
  72. Hou, J.; Kshettry, V.R.; Selman, W.R.; Bambakidis, N.C. Peritumoral Brain Edema in Intracranial Meningiomas: The Emergence of Vascular Endothelial Growth Factor-Directed Therapy. Neurosurg. Focus 2013, 35, E2.
  73. Salokorpi, N.; Yrjänä, S.; Tuominen, H.; Karttunen, A.; Heljasvaara, R.; Pihlajaniemi, T.; Heikkinen, E.; Koivukangas, J. Expression of VEGF and Collagen XVIII in Meningiomas: Correlations with Histopathological and MRI Characteristics. Acta Neurochir. 2013, 155, 989–996; discussion 996.
  74. Shih, K.C.; Chowdhary, S.; Rosenblatt, P.; Weir, A.B.; Shepard, G.C.; Williams, J.T.; Shastry, M.; Burris, H.A.; Hainsworth, J.D. A Phase II Trial of Bevacizumab and Everolimus as Treatment for Patients with Refractory, Progressive Intracranial Meningioma. J. Neurooncol. 2016, 129, 281–288.
  75. 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 2016, 18, 401–407.
  76. 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.
  77. 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. Neurooncol. 2014, 117, 93–101.
  78. Lou, E.; Sumrall, A.L.; Turner, S.; Peters, K.B.; Desjardins, A.; Vredenburgh, J.J.; McLendon, R.E.; Herndon, J.E.; McSherry, F.; Norfleet, J.; et al. Bevacizumab Therapy for Adults with Recurrent/Progressive Meningioma: A Retrospective Series. J. Neurooncol. 2012, 109, 63–70.
  79. 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. Neurooncol. 2012, 109, 187–193.
  80. Franke, A.J.; Skelton, W.P.; 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.
  81. Pinker, B.; Barciszewska, A.M. mTOR Signaling and Potential Therapeutic Targeting in Meningioma. Int. J. Mol. Sci. 2022, 23, 1978.
  82. 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.
  83. Graillon, T.; Sanson, M.; Campello, C.; Idbaih, A.; Peyre, M.; Peyrière, H.; Basset, N.; Autran, D.; Roche, D.; Kalamarides, M.; et al. Everolimus and Octreotide for Patients with Recurrent Meningioma: Results from the Phase II CEVOREM Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 552–557.
  84. Caltabiano, R.; Barbagallo, G.M.V.; Castaing, M.; Cassenti, A.; Senetta, R.; Cassoni, P.; Albanese, V.; Lanzafame, S. Prognostic Value of EGFR Expression in de Novo and Progressed Atypical and Anaplastic Meningiomas: An Immunohistochemical and Fluorescence in Situ Hybridization Pilot Study. J. Neurosurg. Sci. 2013, 57, 139–151.
  85. Osorio, D.S.; Hu, J.; Mitchell, C.; Allen, J.C.; Stanek, J.; Hagiwara, M.; Karajannis, M.A. Effect of Lapatinib on Meningioma Growth in Adults with Neurofibromatosis Type 2. J. Neurooncol. 2018, 139, 749–755.
  86. Wen, P.Y.; Yung, W.K.A.; 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.
  87. Brastianos, P.K.; Twohy, E.L.; Gerstner, E.R.; Kaufmann, T.J.; Iafrate, A.J.; Lennerz, J.; Jeyapalan, S.; Piccioni, D.E.; Monga, V.; Fadul, C.E.; et al. Alliance A071401: Phase II Trial of Focal Adhesion Kinase Inhibition in Meningiomas With Somatic NF2 Mutations. J. Clin. Oncol. 2023, 41, 618–628.
  88. Shapiro, I.M.; Kolev, V.N.; Vidal, C.M.; Kadariya, Y.; Ring, J.E.; Wright, Q.; Weaver, D.T.; Menges, C.; Padval, M.; NcClatchey, A.I.; et al. Merlin Deficiency Predicts FAK Inhibitor Sensitivity: A Synthetic Lethal Relationship. Sci. Transl. Med. 2014, 6, 237ra268.
  89. Lamszus, K. Meningioma Pathology, Genetics, and Biology. J. Neuropathol. Exp. Neurol. 2004, 63, 275–286.
  90. Klaeboe, L.; Lonn, S.; Scheie, D.; Auvinen, A.; Christensen, H.C.; Feychting, M.; Johansen, C.; Salminen, T.; Tynes, T. Incidence of Intracranial Meningiomas in Denmark, Finland, Norway and Sweden, 1968-1997. Int. J. Cancer 2005, 117, 996–1001.
  91. Wigertz, A.; Lönn, S.; Hall, P.; Auvinen, A.; Christensen, H.C.; Johansen, C.; Klaeboe, L.; Salminen, T.; Schoemaker, M.J.; Swerdlow, A.J.; et al. Reproductive Factors and Risk of Meningioma and Glioma. Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol. 2008, 17, 2663–2670.
  92. Sanson, M.; Cornu, P. Biology of Meningiomas. Acta Neurochir. 2000, 142, 493–505.
  93. Wolfsberger, S.; Doostkam, S.; Boecher-Schwarz, H.-G.; Roessler, K.; van Trotsenburg, M.; Hainfellner, J.A.; Knosp, E. Progesterone-Receptor Index in Meningiomas: Correlation with Clinico-Pathological Parameters and Review of the Literature. Neurosurg. Rev. 2004, 27, 238–245.
  94. Markwalder, T.M.; Seiler, R.W.; Zava, D.T. Antiestrogenic Therapy of Meningiomas--a Pilot Study. Surg. Neurol. 1985, 24, 245–249.
  95. Goodwin, J.W.; Crowley, J.; Eyre, H.J.; Stafford, B.; Jaeckle, K.A.; Townsend, J.J. A Phase II Evaluation of Tamoxifen in Unresectable or Refractory Meningiomas: A Southwest Oncology Group Study. J. Neurooncol. 1993, 15, 75–77.
  96. Grunberg, S.M.; Weiss, M.H.; Spitz, I.M.; Ahmadi, J.; Sadun, A.; Russell, C.A.; Lucci, L.; Stevenson, L.L. Treatment of Unresectable Meningiomas with the Antiprogesterone Agent Mifepristone. J. Neurosurg. 1991, 74, 861–866.
  97. Touat, M.; Lombardi, G.; Farina, P.; Kalamarides, M.; Sanson, M. Successful Treatment of Multiple Intracranial Meningiomas with the Antiprogesterone Receptor Agent Mifepristone (RU486). Acta Neurochir. 2014, 156, 1831–1835.
  98. Ji, Y.; Rankin, C.; Grunberg, S.; Sherrod, A.E.; Ahmadi, J.; Townsend, J.J.; Feun, L.G.; Fredericks, R.K.; Russel, 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. Off. J. Am. Soc. Clin. Oncol. 2015, 33, 4093–4098.
  99. Chamberlain, M.C. IFN-α for Recurrent Surgery- and Radiation-Refractory High-Grade Meningioma: A Retrospective Case Series. CNS Oncol. 2013, 2, 227–235.
  100. Chamberlain, M.C.; Glantz, M.J. Interferon-Alpha for Recurrent World Health Organization Grade 1 Intracranial Meningiomas. Cancer 2008, 113, 2146–2151.
  101. Chamberlain, M.C.; Glantz, M.J.; Fadul, C.E. Recurrent Meningioma: Salvage Therapy with Long-Acting Somatostatin Analogue. Neurology 2007, 69, 969–973.
  102. 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.
  103. Marincek, N.; Radojewski, P.; Dumont, R.A.; Brunner, P.; Müller-Brand, J.; Maecke, H.R.; Briel, M.; Walter, M.A. Somatostatin Receptor-Targeted Radiopeptide Therapy with 90Y-DOTATOC and 177Lu-DOTATOC in Progressive Meningioma: Long-Term Results of a Phase II Clinical Trial. J. Nucl. Med. 2015, 56, 171–176.
  104. Brastianos, P.K.; Kim, A.E.; Giobbie-Hurder, A.; Lee, E.Q.; Wang, N.; Eichler, A.F.; Chukwueke, U.; Forst, D.A.; Arillaga-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.
  105. 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, 1–12.
  106. Giles, A.J.; Hao, S.; Padget, M.; Song, H.; Zhang, W.; Lynes, J.; Sanchez, V.; Liu, Y.; Jung, J.; Cao, X.; et al. Efficient ADCC Killing of Meningioma by Avelumab and a High-Affinity Natural Killer Cell Line, HaNK. JCI Insight 2019, 4, e130688.
  107. Wang, L.; Chen, S.; Liu, Y.; Zhang, H.; Ren, N.; Ma, R.; He, Z. The Biological and Diagnostic Roles of MicroRNAs in Meningiomas. Rev. Neurosci. 2020, 31, 771–778.
  108. Caruso, G.; Elbabaa, S.K.; Gonzalez-Lopez, P.; Barresi, V.; Passalacqua, M.; Caffo, M. Innovative Therapeutic Strategies in the Treatment of Meningioma. Anticancer Res. 2015, 35, 6391–6400.
  109. Zhi, F.; Zhou, G.; Wang, S.; Shi, Y.; Peng, Y.; Shao, N.; Guan, W.; Qu, H.; Zhang, Y.; Wang, Q.; et al. A MicroRNA Expression Signature Predicts Meningioma Recurrence. Int. J. Cancer 2013, 132, 128–136.
  110. Shi, L.; Jiang, D.; Sun, G.; Wan, Y.; Zhang, S.; Zeng, Y.; Pan, T.; Wang, Z. MiR-335 Promotes Cell Proliferation by Directly Targeting Rb1 in Meningiomas. J. Neurooncol. 2012, 110, 155–162.
  111. Katar, S.; Baran, O.; Evran, S.; Cevik, S.; Akkaya, E.; Baran, G.; Antar, V.; Hanimoglu, H.; Kaynar, M.Y. Expression of MiRNA-21, MiRNA-107, MiRNA-137 and MiRNA-29b in Meningioma. Clin. Neurol. Neurosurg. 2017, 156, 66–70.
  112. Ludwig, N.; Kim, Y.-J.; Mueller, S.C.; Backes, C.; Werner, T.V.; Galata, V.; Sartorius, E.; Bohle, R.M.; Keller, A.; Meese, E. Posttranscriptional Deregulation of Signaling Pathways in Meningioma Subtypes by Differential Expression of MiRNAs. Neuro-Oncology 2015, 17, 1250–1260.
  113. Hu, S.; Wei, W.; Yuan, J.; Cheng, J. Resveratrol Inhibits Proliferation in HBL-52 Meningioma Cells. OncoTargets Ther. 2019, 12, 11579–11586.
More
Video Production Service