Skip to main content

Review of meningioma diagnosis and management

Abstract

Meningiomas are the most common intracranial tumors in adult patients. Although the majority of meningiomas are diagnosed as benign, approximately 20% of cases are high-grade tumors that require significant clinical treatment. The gold standard for grading central nervous system tumors comes from the World Health Organization Classification of Tumors of the central nervous system. Treatment options also depend on the location, imaging, and histopathological features of the tumor. This review will cover diagnostic strategies for meningiomas, including 2021 updates to the World Health Organization’s grading of meningiomas. Meningioma treatment plans are variable and highly dependent on tumor grading. This review will also update the reader on developments in the treatment of meningiomas, including surgery, radiation therapy and monoclonal antibody treatment.

Background

Accounting for approximately one-third of all primary central nervous system tumors in adult patients, meningiomas are the most prevalent primary brain tumor [1, 2]. With the median age of diagnosis being 65, 36.6% of all adult brain tumors are diagnosed as meningiomas, and as few as 3–5% of pediatric primary brain tumors are believed to be meningiomas [2, 3]. With the exception of rare high-grade variants and pediatric cases, primary brain meningiomas are more common in female patients at an incidence rate of 3:1 compared to males [4, 5]. Furthermore, the female to male ratio is approximately 9:1 for all primary meningiomas of the spine [4]. Neurofibromatosis type 2 (NF2), schwannomatosis, multiple endocrine neoplasia type 1 (MEN1), and numerous other familial syndromes are associated with an increased risk of meningioma occurrence [6]. This genetic predisposition is especially notable in NF2, with as many as 50% of patients presenting with meningiomas [6, 7].

Originating from meningothelial or arachnoid cap cells of dura tissue, meningiomas are commonly observed at the vault of the skull, the skull base, and locations of dural reflections (e.g., tentorium cerebelli, falx cerebri, and adjacent to dural venous sinuses) [8, 9]. In 12% of all cases, meningiomas occur as primary spinal lesions [10, 11]. Although less common, meningiomas can occur in the optic nerve sheath and the choroid plexus of ventricles [8, 12, 13]. Despite their benign nature in the vast majority of cases, meningiomas can cause symptoms due to mass effect displacement of surrounding tissue [14]. The presence of pre-operative seizures can be observed in a wide range of supratentorial intracranial meningiomas, while focal symptoms are often specific to the site of the lesion [15]. Some of the most common self-reported symptoms at the time of diagnosis include headache due to increased intracranial pressure, fatigue, vision changes, altered cognition, and extremity weakness or numbness [16, 17]. While many meningiomas present with symptoms, a significant portion are asymptomatic when first diagnosed, representing one of the most common incidental brain tumor findings on imaging [18, 19].

Recent discoveries have identified genetic markers that have a high correlation with aggressive meningiomas. Telomerase reverse transcriptase (TERT) promoter gene alterations, which increase TERT expression and telomere length, conferring cell immortality, are linked to high-grade meningiomas with elevated rates of recurrence and poor clinical outcomes [20,21,22,23,24]. Similarly, homologous deletion of the CDKN2A/B tumor suppressor genes has been identified as a marker of aggressive meningioma clinical course [25, 26]. While uncommon (< 5%), loss of trimethylation expression of lysine 27 of histone H3 (H3K27me3) is believed to be correlated with an increased risk of meningioma tumor recurrence [27, 28].

Meningioma diagnostic strategies

With the identification of several high-risk mutations and molecular markers in higher grade meningiomas (Fig. 1), interest has grown for the use of molecular markers as a method of risk stratification [33]. Biopsy or resection, however, remains the only methods of definitive meningioma diagnosis [17]. Nevertheless, meningiomas are primarily first seen and diagnosed as a result of imaging [19]. In some cases, to rule out other types of tumors, a biopsy is taken and analyzed through histopathology [32].

Fig. 1
figure 1

Meningiomas arise from meningothelial and arachnoid cap cells of the leptomeninges and may progress to invade the dura mater [29,30,31,32]

The World Health Organization (WHO) grading is considered to be the “gold standard” in classifying histological and etiological meningioma factors [34]. 2016 WHO guidelines categorized meningiomas into 15 subtypes encompassed by 3 grades; benign (grade I), atypical (grade II), and anaplastic (grade III) (Fig. 2). The 2016 version of the WHO meningioma classifications was hallmark because it combined genetic/molecular alterations, along with tumor histopathology to categorize meningiomas [35]. This directly led to more detailed subtypes of meningiomas, which improved clinical estimation of recurrence and prognosis of meningioma patients. [36]. The number of subtypes increased specifically for grade II and grade III meningiomas. [37]

Fig. 2
figure 2

2016 WHO Subtypes and Grades [35, 38]

Grade I meningiomas make up approximately 80% of all meningioma cases [38]. Hence, meningiomas are regarded as being mostly benign and having a routine clinical course. The remaining approximately 20% (~ 17% grade II and ~ 3% grade III) of cases, however, tend to face more severe clinical challenges such as local recurrence, brain invasion, and/or progression to higher tumor grade [35].

The 2021 revision of the WHO guidelines further emphasizes the review of genomic alterations to support tumor classification and assist clinicians with meningioma management. However, this revision did change how meningiomas and other CNS tumors are graded. Within the 2016 guidelines, meningiomas were graded based on histopathological subtype. For example, an anaplastic subtype meningioma would automatically be classified as a grade III meningioma. Anaplastic lesions of other CNS tumor types would also be classified as grade III [35, 36, 38, 39]. This method of grading attempted to classify different categories of CNS tumors by expected clinical course. The problem with the clinical approach to grading was that it assumed different tumor types with similar histological findings behaved relatively the same. This was, however, not always true and did not conform to grading used for other non-CNS tumors [39]. 2021 WHO guidelines retained the 15 subtypes of meningioma, but shifted to within-tumor-type grading, allowing for the criteria of grade 2 or 3 to be applied to tumors regardless of subtype. This change gives clinicians more flexibility with tumor classification and puts further emphasis on the biological similarities between tumor types [38, 39]. The grading also changed from the use of Roman numerals to Arabic numerals to align CNS tumors with other systems. The use of Roman numerals throughout this paper refers to 2016 WHO grading.

The clinicopathological relevance of genetic alterations in meningiomas is still being studied, but certain alterations are seen more frequently in varying subtypes and locations of meningioma [38]. It has also been observed that higher grade meningiomas contain a higher frequency of abnormalities [40]. Alteration of the NF2 gene is the most common and is seen in approximately 60% of all sporadic meningiomas, along with other additional modifications [41] For example, meningiomas that occur on the surface of the brain (convexity meningiomas) are found to have abnormalities in NF2 as well as SMARCB1 (SWItch/Sucrose Non-Fermentable Related, Matrix Associated, Actin Dependent Regulator Of Chromatin, Subfamily B, Member 1), TERT and CDKN2A. Convexity meningiomas are predominantly of the fibrous and transitional subtypes and are more commonly grade 2 and 3 [42,43,44,45]. Contrastingly, meningiomas that occur along the base of the skull (skull base meningiomas) are predominantly of the meningothelial, microcystic and secretory subtypes [40]. Meningiomas located on the spinal cord are frequently associated with rhabdoid and clear cell subtypes [46]. Table 1 outlines common molecular biomarkers utilized in meningioma diagnosis per the 2021 WHO guidelines.

Table 1 Common genetic alteration in meningiomas [42,43,44,45,46,47,48,49,50,51,52,53,54]

Meningioma imaging modalities

While contrast-enhanced computed tomography (CT) may offer advantages in the identification of characteristic meningioma lesion calcification (15–20% of cases) and hyperostosis (25–49% of cases), magnetic resonance imaging (MRI) offers significant advantages in tumor tissue and edema analysis [55,56,57]. Compared to the cerebral cortex, meningiomas appear hypointense to isointense on T1-weighted MRI sequences and isointense to hyperintense on T2-weighted sequences [58]. Fig. 3 displays T2-weighted MRI images of a meningioma with an encasement partial occluding the superior sagittal sinus. The left is without contrast, while the right images are with contrast. The addition of gadolinium contrast markedly enhances the visibility of meningiomas on MRI [58]. Characteristics typical of more benign meningiomas include a dural tail, calcification, homogenous enhancement, and a uniform border. While not pathognomonic for meningiomas, dural tails are a common feature found in 72% of meningiomas [59]. MRI radiographic features that correlate with more aggressive and higher-grade meningiomas include intra-tumoral necrosis, cortex invasion, intertumoral cystic changes, edema volume, and tumor extension across skull base foramina [56, 60,61,62,63,64,65]. Additional radiographic features are detailed in Table 2.

Fig. 3
figure 3

Representative images of meningioma with encasement and partial occlusion of superior sagittal sinus. Recommended treatment is surgical resection given location

Table 2 Typical MRI radiographic features of meningioma

Another useful tool in the risk stratification of meningiomas is MRI diffusion-weighted imaging (DWI), which quantifies water diffusion levels in tissue through a reported apparent diffusion coefficient (ADC) value [66]. A lower intertumoral ADC may correlate with higher Ki-67 levels, increased cell proliferation, and higher-grade meningiomas [67,68,69,70,71,72]. Relative cerebral blood volume (rCBV), a measure generated through MR perfusion analysis, is an estimate of blood volume in a given space and is elevated with increased vasculature [73]. Studies have demonstrated that lower intertumoral rCBV and a higher peritumoral edema rCBV correlate with higher-grade meningiomas and may be effective in distinguishing meningiomas from schwannomas [74,75,76,77,78,79].

Meningioma treatment

Most meningiomas are routinely treated as a neurosurgical disease. Despite its reputation as a commonly benign disease, the associated clinical symptoms, risk of recurrence, and unfavorable course of outcomes are far from indolent [80]. Fortunately, since Harvey Cushing first coined the term “meningioma” in 1922, there have been significant advances made in systemic treatment and monitoring [81].

The current treatment strategies can be determined based on two main types of meningiomas, asymptomatic and symptomatic [17]. For small, asymptomatic meningiomas, a watchful waiting strategy is usually recommended. Clinical observation and MRI screening are performed every 6 months following initial diagnosis [17]. Patients that remain asymptomatic after 5 years are then seen for annual observation-only [17].

In contrast, symptomatic meningiomas are treated with surgical intervention. Surgical resection remains the first line of treatment. Symptomatic meningiomas are classified according to the WHO grading system [82]. Recurrence risk, survival rates, and morbidity all correlate with WHO grades, holding major consideration into the choice of treatment [82]. However, patients who are not fit for surgery, including elderly or disabled individuals, have the option to choose either stereotactic radiotherapy/radiosurgery (SRT/SRS) or chemotherapy as a primary treatment [83].

Patients with grade I meningiomas typically undergo gross total resection (GTR) with routine follow-ups or subtotal resection (STR) followed by rounds of SRT/SRS therapy [82]. Patients with grade II meningiomas also either undergo GTR or STR. For those patients, intimate follow-up is recommended after GTR, and SRT/SRS follow-up is recommended after STR. Grade III meningiomas require adjuvant radiotherapy following surgical resection, regardless of the degree of resection [82]. There is debate on how long after diagnosis radiotherapy should start [83].

Advancements in operating techniques including surgical microscopy, neuronavigation, intraoperative monitoring, imaging, and endovascular approaches have allowed for more radical resections [84]. However, depending on factors like surgical approach, tumor location, the extent of dural attachment, and the proximity to neurovascular structures, total tumor resection is not always possible [81]. In 1957, Donald Simpson classified the extent of surgical tumor resection into Simpson Grades I–V [84]. Typically, Simpson Grades I-III are designated as GTR, whereas Simpson Grades IV-V are designated as STR. Simpson Grading remains a reliable tool for classifying the extent of surgical tumor resections [82]. The characterization of recurrence rates of meningiomas has a high correlation with the Simpson Grading. Studies show the recurrence rate of Simpson grade I surgery patients is 9%, grade II is 19%, and grade III is 29% [85]. Similar to other neoplastic entities, meningiomas make up a range of marked variation; therefore, the different grading criteria discussed remains broad.

For total surgical resection, the tumor and its dural base are removed. Resection of the dura was found to be important for the prevention of recurrence [86]. The dura is replaced with a dural patch or graft. When the tumor includes the involvement of the skull, that portion of the bone is removed, and replaced post-surgically through cranioplasty [87]. Aside from these general techniques, the surgical approach differs depending on the location and extent of the tumor [87]. For example, for tumors that invade and obstruct the superior sagittal sinus, attempts are made to remove the entire tumor along with that portion of the sinus. This is followed by venous reconstruction [88]. Contrastingly, for parasellar meningiomas, a conservative approach is taken due to the anatomical complexity of the area. The preference is STR followed by irradiation to reduce the chance of neurological injury or deficit [89]. Meningiomas comprise a spectrum of disease types, so treatment plans should be individualized for each patient, especially when predicting risk stratifications [80].

Current treatment developments for meningioma

With the development of monoclonal antibody-based pharmacotherapies that effectively treat other oncologic conditions, there have been a host of new cell cycle regulators and antibody-based drugs which are currently in clinical trials for the treatment and management of varying severities of meningioma [90,91,92]. Palbociclib is a CDK4/6 inhibitor that blocks the cell cycle and prevents rapid cell replication [90, 91]. Palbociclib, in combination with radiation, has been shown to diminish cell growth in vivo using mouse models with anaplastic and radiation-induced meningioma cells [90].

Another monoclonal antibody, Nivolumab, a programmed cell death protein 1 (PD-1) blocker, underwent Phase II clinical trials but was not shown to bolster six-month progression-free survival in patients with recurrent atypical/anaplastic meningioma [92]. Positively, however, Nivolumab treatments did not show significant side effects and were generally well tolerated by patients [92]. The ability to attack specific tumor cells without causing significant harm to healthy cells is essential in cancer treatment. Further research and clinical trials of these drugs can revolutionize how we treat and manage meningioma. Some current clinical trials involving cell cycle inhibitors and antibody therapy in the treatment of various forms/grades of meningioma are listed in Fig. 4.

Fig. 4
figure 4

Monoclonal Antibody Studies for Meningioma [93]

Currently, MRI is the standard of care for the thorough assessment of meningiomas from an imaging standpoint. With further advancement in artificial intelligence, radiomics could play a role in the diagnosis of and classification of meningioma [94]. Radiomics is the use of MRI, CT, or PET/CT to produce mathematical models which allow for a more detailed analysis of bodily structures based on the texture, shape, and intensity of lesions provided by basic imaging [95,96,97]. When radiomics was applied to basic MRI, it was shown to distinguish between Grade I, II, and III meningiomas between 76 and 93% based on the features of the lesion [98,99,100,101]. This accuracy can be further increased when radiomics is applied to diffusion-weighted imaging (advanced MRI) [102]. Radiomics, therefore, presents a new way to assess and diagnose meningiomas [94].

Availability of data and materials

Not applicable.

Abbreviations

AKT1:

AKT serine/threonine kinase 1

ADC:

Apparent diffusion coefficient

BAP1:

BRCA1 associated protein 1

CT:

Contrast-enhanced computed tomography

DWI:

Diffusion-weighted imaging

GTR:

Gross total resection

KLF4:

Krüppel-like factor 4

MRI:

Magnetic resonance imaging

MEN1:

Multiple endocrine neoplasia type 1

NF2:

Neurofibromatosis type 2

PIK3CA:

Phosphatidylinositol-4,5-Bisphosphate 3-Kinase catalytic subunit alpha

rCBV:

Relative cerebral blood volume

POLR2A:

RNA polymerase II subunit A

SMO:

Smoothened, frizzled class receptor

SRT/SRS:

Stereotactic radiotherapy/radiosurgery

STR:

Subtotal resection

SMARCB1:

SWItch/sucrose non-fermentable related, matrix associated, actin dependent regulator of chromatin, subfamily b, member 1

TERT:

Telomerase reverse transcriptase

TRAF7:

TNF receptor associated factor 7

WHO:

World Health Organization

References

  1. Baldi I, et al. Epidemiology of meningiomas. Neurochirurgie. 2018;64:5–14.

    Article  CAS  PubMed  Google Scholar 

  2. Kirches E, et al. Molecular profiling of pediatric meningiomas shows tumor characteristics distinct from adult meningiomas. Acta Neuropathol. 2021;142:873–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kikuchi K, et al. Arterial spin-labeling is useful for the diagnosis of residual or recurrent meningiomas. Eur Radiol. 2018;28:4334–42.

    Article  PubMed  Google Scholar 

  4. Huntoon K, Toland AMS, Dahiya S. Meningioma: a review of clinicopathological and molecular aspects. Front Oncol 10, (2020).

  5. Toland A, Huntoon K, Dahiya SM. Meningioma: a pathology perspective. Neurosurgery. 2021;89:11–21.

    Article  PubMed  Google Scholar 

  6. Kerr K, Qualmann K, Esquenazi Y, Hagan J, Kim DH. Familial syndromes involving meningiomas provide mechanistic insight into sporadic disease. Neurosurgery. 2018. https://doi.org/10.1093/neuros/nyy121.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Bachir S, et al. Neurofibromatosis type 2 (NF2) and the Implications for vestibular schwannoma and meningioma pathogenesis. Int J Mol Sci. 2021;22:690.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Whittle IR, Smith C, Navoo P, Collie D. Meningiomas. Lancet. 2004;363:1535–43.

    Article  PubMed  Google Scholar 

  9. Kunimatsu A, et al. Variants of meningiomas: a review of imaging findings and clinical features. Jpn J Radiol. 2016;34:459–69.

    Article  PubMed  Google Scholar 

  10. Kwee LE, et al. Spinal meningiomas: treatment outcome and long-term follow-up. Clin Neurol Neurosurg. 2020;198:106238.

    Article  CAS  PubMed  Google Scholar 

  11. Wang N, Osswald M. Meningiomas: overview and new directions in therapy. Semin Neurol. 2018;38:112–20.

    Article  CAS  PubMed  Google Scholar 

  12. Nowosielski M, et al. Diagnostic challenges in meningioma. Neuro Oncol. 2017;19:1588–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Parker R, Ovens CA, Fraser CL, Samarawickrama C. Optic nerve sheath meningiomas: prevalence, impact, and management strategies. Eye Brain. 2018;10:85–99.

    Article  PubMed  PubMed Central  Google Scholar 

  14. de Baene W, et al. Lesion symptom mapping at the regional level in patients with a meningioma. Neuropsychology. 2019;33:103–10.

    Article  PubMed  Google Scholar 

  15. Chen WC, et al. Factors associated with pre- and postoperative seizures in 1033 patients undergoing supratentorial meningioma resection. Neurosurgery. 2017;81:297–306.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Nassiri F, et al. How to live with a meningioma: experiences, symptoms, and challenges reported by patients. Neuro-Oncology Adv. 2, (2020).

  17. Buerki RA, et al. An overview of meningiomas. Future Oncol. 2018;14:2161–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Surov A, et al. Use of diffusion weighted imaging in differentiating between maligant and benign meningiomas: a multicenter analysis. World Neurosurg. 2016;88:598–602.

    Article  PubMed  Google Scholar 

  19. Islim AI, et al. A prognostic model to personalize monitoring regimes for patients with incidental asymptomatic meningiomas. Neuro Oncol. 2020;22:278–89.

    Article  PubMed  Google Scholar 

  20. Mirian C, 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;91:378–87.

    Article  PubMed  Google Scholar 

  21. Juratli TA, et al. Intratumoral heterogeneity and TERT promoter mutations in progressive/higher-grade meningiomas. Oncotarget. 2017;8:109228–37.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Bell RJA, et al. Understanding TERT promoter mutations: a common path to immortality. Mol Cancer Res. 2016;14:315–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sahm F, et al. TERT promoter mutations and risk of recurrence in meningioma. J Natl Cancer Inst. 108, djv377 (2016).

  24. Goutagny S, et al. High incidence of activating TERT promoter mutations in meningiomas undergoing malignant progression. Brain Pathol. 2014;24:184–9.

    Article  CAS  PubMed  Google Scholar 

  25. Birzu C, Peyre M, Sahm F. Molecular alterations in meningioma: prognostic and therapeutic perspectives. Curr Opin Oncol. 2020;32:613–22.

    Article  CAS  PubMed  Google Scholar 

  26. Sievers P, et al. CDKN2A/B homozygous deletion is associated with early recurrence in meningiomas. Acta Neuropathol. 2020;140:409–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nassiri F, et al. Loss of H3K27me3 in meningiomas. Neuro Oncol. 2021;23:1282–91.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Behling F, et al. H3K27me3 loss indicates an increased risk of recurrence in the Tübingen meningioma cohort. Neuro Oncol. 2021;23:1273–81.

    Article  PubMed  Google Scholar 

  29. Bhat A, Wani M, Kirmani A, Ramzan A. Histological-subtypes and anatomical location correlated in meningeal brain tumors (meningiomas). J Neurosci Rural Pract. 2014;5:244.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Nagashima G, et al. Dural invasion of meningioma: a histological and immunohistochemical study. Brain Tumor Pathol. 2006;23:13–7.

    Article  CAS  PubMed  Google Scholar 

  31. SMART. Servier medical art. https://smart.servier.com/.

  32. Solomon DA, Pekmezci M. Pathology of meningiomas. in 87–99 (2020). doi:https://doi.org/10.1016/B978-0-12-804280-9.00005-6.

  33. Juratli TA, et al. DMD genomic deletions characterize a subset of progressive/higher-grade meningiomas with poor outcome. Acta Neuropathol. 2018;136:779–92.

    Article  CAS  PubMed  Google Scholar 

  34. Apra C, Peyre M, Kalamarides M. Current treatment options for meningioma. Expert Rev Neurother. 2018;18:241–9.

    Article  CAS  PubMed  Google Scholar 

  35. Harter PN, Braun Y, Plate KH. Classification of meningiomas—advances and controversies. Chin Clin Oncol. 2017;6:S2–S2.

    Article  PubMed  Google Scholar 

  36. Backer-Grøndahl T, Moen BH, Torp SH. The histopathological spectrum of human meningiomas. Int J Clin Exp Pathol. 2012;5:231–42.

    PubMed  PubMed Central  Google Scholar 

  37. Weber RG, et al. Analysis of genomic alterations in benign, atypical, and anaplastic meningiomas: toward a genetic model of meningioma progression. Proc Natl Acad Sci USA. 1997;94:14719–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gritsch S, Batchelor TT, Gonzalez Castro LN. Diagnostic, therapeutic, and prognostic implications of the 2021 World Health Organization classification of tumors of the central nervous system. Cancer 128, 47–58 (2022).

  39. Louis DN, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol. 2021;23:1231–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mawrin C, Perry A. Pathological classification and molecular genetics of meningiomas. J Neurooncol. 2010;99:379–91.

    Article  CAS  PubMed  Google Scholar 

  41. Zang KD. Meningioma: a cytogenetic model of a complex benign human tumor, including data on 394 karyotyped cases. Cytogenet Genome Res. 2001;93:207–20.

    Article  CAS  Google Scholar 

  42. Kros J, et al. NF2 status of meningiomas is associated with tumour localization and histology. J Pathol. 2001;194:367–72.

    Article  CAS  PubMed  Google Scholar 

  43. Lee JH, Sade B, Choi E, Golubic M, Prayson R. Meningothelioma as the predominant histological subtype of midline skull base and spinal meningioma. J Neurosurg. 2006;105:60–4.

    Article  PubMed  Google Scholar 

  44. Ketter R, et al. Correspondence of tumor localization with tumor recurrence and cytogenetic progression in meningiomas. Neurosurgery. 2008;62:61–70.

    Article  PubMed  Google Scholar 

  45. Goutagny S, et al. Genomic profiling reveals alternative genetic pathways of meningioma malignant progression dependent on the underlying NF2 status. Clin Cancer Res. 2010;16:4155–64.

    Article  CAS  PubMed  Google Scholar 

  46. Smith MJ, et al. Germline SMARCE1 mutations predispose to both spinal and cranial clear cell meningiomas. J Pathol. 2014;234:436–40.

    Article  CAS  PubMed  Google Scholar 

  47. Smith MJ, et al. Loss-of-function mutations in SMARCE1 cause an inherited disorder of multiple spinal meningiomas. Nat Genet. 2013;45:295–8.

    Article  CAS  PubMed  Google Scholar 

  48. Abedalthagafi M, et al. Oncogenic PI3K mutations are as common as AKT1 and SMO mutations in meningioma. Neuro Oncol. 2016;18:649–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Reuss DE, et al. Secretory meningiomas are defined by combined KLF4 K409Q and TRAF7 mutations. Acta Neuropathol. 2013;125:351–8.

    Article  CAS  PubMed  Google Scholar 

  50. Clark VE, et al. Genomic analysis of non-NF2 meningiomas reveals mutations in TRAF7, KLF4, AKT1, and SMO. Science. 2013;339:1077–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. 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 now276 (2017) doi:https://doi.org/10.1093/neuonc/now276.

  52. Sahm F, et al. AKT1E17K mutations cluster with meningothelial and transitional meningiomas and can be detected by SFRP1 immunohistochemistry. Acta Neuropathol. 2013;126:757–62.

    Article  CAS  PubMed  Google Scholar 

  53. Brastianos PK, et al. Genomic sequencing of meningiomas identifies oncogenic SMO and AKT1 mutations. Nat Genet. 2013;45:285–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Strickland MR, et al. Targeted sequencing of SMO and AKT1 in anterior skull base meningiomas. J Neurosurg. 2017;127:438–44.

    Article  CAS  PubMed  Google Scholar 

  55. Bikmaz K, Mrak R, Al-Mefty O. Management of bone-invasive, hyperostotic sphenoid wing meningiomas. J Neurosurg. 2007;107:905–12.

    Article  PubMed  Google Scholar 

  56. Salah F, et al. Can CT and MRI features differentiate benign from malignant meningiomas? Clin Radiol. 2019;74(898):e15-898.e23.

    Google Scholar 

  57. Sheporaitis LA, et al. Intracranial meningioma. AJNR. Am J Neuroradiol. 13, 29–37.

  58. Huang RY, et al. Imaging and diagnostic advances for intracranial meningiomas. Neuro Oncol. 2019;21:i44–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. O’Leary S, Adams WM, Parrish RW, Mukonoweshuro W. Atypical imaging appearances of intracranial meningiomas. Clin Radiol. 2007;62:10–7.

    Article  PubMed  Google Scholar 

  60. Boukobza M, et al. Cystic meningioma: radiological, histological, and surgical particularities in 43 patients. Acta Neurochir. 2016;158:1955–64.

    Article  PubMed  Google Scholar 

  61. Hsu C-C, et al. Do aggressive imaging features correlate with advanced histopathological grade in meningiomas? J Clin Neurosci. 2010;17:584–7.

    Article  PubMed  Google Scholar 

  62. Mattei TA, et al. Edema and malignancy in meningiomas. Clinics. 2005;60:201–6.

    Article  PubMed  Google Scholar 

  63. Ressel A, Fichte S, Brodhun M, Rosahl SK, Gerlach R. WHO grade of intracranial meningiomas differs with respect to patient’s age, location, tumor size and peritumoral edema. J Neurooncol. 2019;145:277–86.

    Article  CAS  PubMed  Google Scholar 

  64. Soon WC, et al. Correlation of volumetric growth and histological grade in 50 meningiomas. Acta Neurochir. 2017;159:2169–77.

    Article  PubMed  Google Scholar 

  65. Spille DC, et al. Predicting the risk of postoperative recurrence and high-grade histology in patients with intracranial meningiomas using routine preoperative MRI. Neurosurg Rev. 2021;44:1109–17.

    Article  PubMed  Google Scholar 

  66. Surov A, Meyer HJ, Wienke A. Associations between apparent diffusion coefficient (ADC) and KI 67 in different tumors: a meta-analysis. Part 1: ADC mean. Oncotarget 8, 75434–75444 (2017).

  67. Atalay B, Ediz SS, Ozbay NO. Apparent diffusion coefficient in predicting the preoperative grade of meningiomas. J Coll Phys Surg-Pak: JCPSP. 2020;30:1126–32.

    Google Scholar 

  68. Baskan O, et al. Relation of apparent diffusion coefficient with Ki-67 proliferation index in meningiomas. Br J Radiol. 2016;89:20140842.

    Article  PubMed  Google Scholar 

  69. Hakyemez B, Yıldırım N, Gokalp G, Erdogan C, Parlak M. The contribution of diffusion-weighted MR imaging to distinguishing typical from atypical meningiomas. Neuroradiology. 2006;48:513–20.

    Article  PubMed  Google Scholar 

  70. Hwang WL, et al. Imaging and extent of surgical resection predict risk of meningioma recurrence better than WHO histopathological grade. Neuro Oncol. 2016;18:863–72.

    Article  PubMed  Google Scholar 

  71. Nagar VA, et al. Diffusion-weighted MR imaging: diagnosing atypical or malignant meningiomas and detecting tumor dedifferentiation. Am J Neuroradiol. 2008;29:1147–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Toh C-H, et al. Differentiation between classic and atypical meningiomas with use of diffusion tensor imaging. Am J Neuroradiol. 2008;29:1630–5.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Kickingereder P, et al. Relative cerebral blood volume is a potential predictive imaging biomarker of bevacizumab efficacy in recurrent glioblastoma. Neuro Oncol. 2015;17:1139–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Cebeci H, et al. Precise discrimination between meningiomas and schwannomas using time-to-signal intensity curves and percentage signal recoveries obtained from dynamic susceptibility perfusion imaging. J Neuroradiol. 2021;48:157–63.

    Article  PubMed  Google Scholar 

  75. Kang Y, Wei K-C, Toh CH. Can we predict intraoperative blood loss in meningioma patients? Application of dynamic susceptibility contrast-enhanced magnetic resonance imaging. J Neuroradiol. 2021;48:254–8.

    Article  PubMed  Google Scholar 

  76. Rohilla S, Garg HK, Singh I, Yadav RK, Dhaulakhandi DB. rCBV- and ADC-based grading of meningiomas with glimpse into emerging molecular diagnostics. Basic Clin Neurosci J. 417–428 (2018) doi:https://doi.org/10.32598/bcn.9.6.417.

  77. Shi R, Jiang T, Si L, Li M. Correlations of magnetic resonance, perfusion-weighed imaging parameters and microvessel density in meningioma. J B.U.ON. : Off J Balkan Union of Oncol 21, 709–13.

  78. Todua F, Chedia S. Differentiation between benign and malignant meningiomas using diffusion and perfusion MR imaging. Georgian Med News 16–22 (2012).

  79. Zhang H, Rödiger LA, Shen T, Miao J, Oudkerk M. Perfusion MR imaging for differentiation of benign and malignant meningiomas. Neuroradiology. 2008;50:525–30.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Wilson TA, et al. Review of atypical and anaplastic meningiomas: classification, molecular biology, and management. Front Oncol. 2020;10:565582.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Rogers L, et al. Meningiomas: knowledge base, treatment outcomes, and uncertainties: a RANO review. J Neurosurg. 2015;122:4–23.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Louis DN, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131:803–20.

    Article  PubMed  Google Scholar 

  83. Goldbrunner R, et al. EANO guidelines for the diagnosis and treatment of meningiomas. Lancet Oncol. 2016;17:e383–91.

    Article  PubMed  Google Scholar 

  84. Brastianos PK, et al. Advances in multidisciplinary therapy for meningiomas. Neuro Oncol. 2019;21:i18–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Zhao L, et al. An overview of managements in meningiomas. Front Oncol. 2020;10:1523.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Simpson D. The recurrence of intracranial meningiomas after surgical treatment. J Neurol Neurosurg Psychiatry. 1957;20:22–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Alexiou GA, Gogou P, Markoula S, Kyritsis AP. Management of meningiomas. Clin Neurol Neurosurg. 2010;112:177–82.

    Article  PubMed  Google Scholar 

  88. DiMeco F, et al. Meningiomas invading the superior sagittal sinus. Neurosurgery. 2008;62:1263–73.

    Article  Google Scholar 

  89. Wilson CB. Meningiomas: genetics, malignancy, and the role of radiation in induction and treatment. J Neurosurg. 1994;81:666–75.

    Article  CAS  PubMed  Google Scholar 

  90. Das A, et al. Evaluating anti-tumor activity of palbociclib plus radiation in anaplastic and radiation-induced meningiomas: pre-clinical investigations. Clin Transl Oncol. 2020;22:2017–25.

    Article  CAS  PubMed  Google Scholar 

  91. Horbinski, C, et al. The effects of palbociclib in combination with radiation in preclinical models of aggressive meningioma. Neuro-Oncol Adv 3 (2021).

  92. Bi WL, et al. Activity of PD-1 blockade with nivolumab among patients with recurrent atypical/anaplastic meningioma: phase II trial results. Neuro Oncol. 2022;24:101–13.

    Article  CAS  PubMed  Google Scholar 

  93. Maggio I, et al. Meningioma: not always a benign tumor: a review of advances in the treatment of meningiomas. CNS Oncol 10, CNS72 (2021).

  94. Galldiks N, et al. Use of advanced neuroimaging and artificial intelligence in meningiomas. Brain Pathol. 32 (2022).

  95. Amorim BJ, et al. PET/MRI radiomics in rectal cancer: a pilot study on the correlation between PET- and MRI-derived image features with a clinical interpretation. Mol Imag Biol. 2020;22:1438–45.

    Article  CAS  Google Scholar 

  96. Kumar V, et al. Radiomics: the process and the challenges. Magn Reson Imaging. 2012;30:1234–48.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Schick U, et al. MRI-derived radiomics: methodology and clinical applications in the field of pelvic oncology. Br J Radiol. 2019;92:20190105.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Fatima K, Arooj A, Majeed H. A new texture and shape based technique for improving meningioma classification. Microsc Res Tech. 2014;77:862–73.

    Article  PubMed  Google Scholar 

  99. Chu H, et al. Value of MRI radiomics based on enhanced T1WI images in prediction of meningiomas grade. Acad Radiol. 2021;28:687–93.

    Article  PubMed  Google Scholar 

  100. Hale AT, Stonko DP, Wang L, Strother MK, Chambless LB. Machine learning analyses can differentiate meningioma grade by features on magnetic resonance imaging. Neurosurg Focus. 2018;45:E4.

    Article  PubMed  Google Scholar 

  101. Han Y, et al. Meningiomas: preoperative predictive histopathological grading based on radiomics of MRI. Magn Reson Imaging. 2021;77:36–43.

    Article  PubMed  Google Scholar 

  102. Filippidis A, et al. Intracranial venous malformation masquerading as a meningioma in PI3KCA -related overgrowth spectrum disorder. Am J Med Genet A. 2022;188:907–10.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

C.H. Jr. contributed to diagnostic strategies. M.W. contributed to background. D.C. contributed to current developments in treatment. J.W. contributed to imaging. Y.M. contributed to treatment approaches. S.L. contributed to treatment approaches. B.L.-W. is the senior author. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Chadwin Hanna Jr.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hanna, C., Willman, M., Cole, D. et al. Review of meningioma diagnosis and management. Egypt J Neurosurg 38, 16 (2023). https://doi.org/10.1186/s41984-023-00195-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41984-023-00195-z

Keywords