Sie sind auf Seite 1von 41

13 

Meningiomas
Arie Perry, MD

meningiomas arising at sites of prior skull fracture, cranioplasty, or


Introduction and Proposed Etiologies  259
lodged foreign material, such as shrapnel, implicating wound healing
Definition and Analogies With Meningothelial Cells  259 and arachnoidal proliferation as a possible mechanism.4 However, most
Brief Historical Overview of Nomenclature and Histogenetic epidemiologic studies have not found a significant correlation.5,6 Similarly,
Notions 260 evidence to date argues against the common notion that cell phones
Incidence and Demographics  260 cause meningiomas and other brain tumors, although admittedly this
is a relatively new technology and more follow-up may still be needed.7,8
Localization and Clinical Manifestations  260 In contrast, meningiomas caused by irradiation are well documented,
Radiologic Features and Gross Pathology  263 and this is an irrefutable risk factor. As with other radiation-associated
Histopathology 266 neoplasms (see Chapter 21), these tumors must arise in the prior radiation
field, develop after long latency (typically years), differ histologically
Histologic Variants and Grading  267
from the originally irradiated disease process, and arise more frequently
Differential Diagnosis  289 in irradiated than in control patients. Radiation-induced meningiomas
Ancillary Diagnostic Studies  292 are sometimes divided into low-dose (<10 Gy), medium-dose and
Genetics 294 miscellaneous (10 to 20 Gy), and high-dose (>20 Gy) categories. The
low-dose group has been most extensively studied, mostly in children
Treatment and Prognosis  296
receiving scalp irradiation for tinea capitis in Israel during the 1960s,1
although recent studies have similarly implicated prior exposures to
computed tomography (CT) imaging and dental x-rays (mostly the
older and somewhat higher dose Panorex films in young children).9–11
The miscellaneous group includes direct application of radium, thorotrast
ventriculography, and myelography, whereas high-dose examples mostly
Introduction and Proposed Etiologies follow therapeutic irradiation for cancer. Radiation-induced meningiomas
Meningiomas are among the most common of central nervous system typically afflict younger patients, with median onset being 45, 32, and
(CNS) neoplasms and are by far the most common of the extra-axial 34 years for the high-, medium-, and low-dose groups, respectively.
tumors. Our understanding of meningioma classification, grading, and The average latencies are 35, 26, and 19.5 years, respectively. Children
molecular genetics has evolved greatly over time and continues to be appear to be particularly sensitive (“window of vulnerability”), with
refined.1 Both environmental and genetic factors have been implicated the latency often being shorter than in adults. Providing further evidence,
in meningioma formation, although most examples are idiopathic. atomic bomb survivors are also at increased risk of developing menin-
Meningioma genetics is covered later in this chapter, whereas the giomas, the distance from the epicenter of the explosion being inversely
meningioma-associated syndrome NF2 is discussed further in Chapter associated with overall risk.
22. Proposed environmental factors include irradiation (discussed later), Lastly, it is of interest that allergic diseases, such as asthma and
hormone exposures (discussed later), and head trauma, although only eczema, have been confirmed in epidemiologic studies as protective
the first of these has been unequivocally established. The latter is (i.e., lower risk) for subsequent meningioma development.12
occasionally the basis for medicolegal cases, though it is hardly a proven
cause. In Cushing and Eisenhardt’s 1938 monograph, a surprising 32% Definition and Analogies With
of intracranial meningiomas had a prior history of head injury,2 and a Meningothelial Cells
modern epidemiologic study estimated a 4.33 odds ratio for developing Meningiomas are a clinicopathologically diverse group of neoplasms
meningiomas 10 to 19 years after head trauma.3 Rare case reports feature with morphologic, immunohistochemical, and ultrastructural features

259
Meningiomas

Abstract
Accounting for over a third of all primary CNS neoplasms, meningiomas aggressive features. Current grading criteria within the 2016 WHO
13
are amongst the most common diagnoses encountered in neurosurgical scheme largely derive from the findings in two large Mayo Clinic series,
practice. Like their non-neoplastic counterparts, meningiomas show which have been subsequently validated. Nevertheless, predicting clinical
some overlapping features with both epithelial and mesenchymal cell behavior in individual patients can still be challenging. As such, this
types. As such, many histologic variants are recognized, some of which revised chapter incorporates (and illustrates) recent immunohistochemical
commonly engender diagnostic confusion due to their mimicry of other and molecular advances that enhance our diagnostic accuracy and our
tumor types. It has long been appreciated that the two most important understanding of meningioma tumorigenesis and malignant progression,
prognostic variables are extent of surgical resection and tumor grade. including some of the rarer variants.
While most are histologically and clinically benign, 20%–30% show

Keywords
meningioma
classification
grading
immunohistochemistry
molecular genetics

259.e1
Practical Surgical Neuropathology
resembling normal meningothelial cells. More specifically, the majority solitary fibrous tumor/hemangiopericytoma) have been removed as a
resemble the arachnoidal cap cell, although rare variants such as the biologically distinct tumor type (see Chapter 14), grading criteria have
microcystic meningioma resemble other cell types (arachnoidal trabecular been extensively revised, our understanding of the molecular pathology
cell). Like their normal meningothelial counterparts, meningiomas can has greatly increased, and therapeutic modalities have evolved. The
display a wide range of epithelial- and mesenchymal-like properties. classification scheme utilized by most pathologists today is the World
Mesenchymal features include a prominent spindled morphology in the Health Organization (WHO) 2016 edition.1
fibroblastic and sarcoma-like meningiomas, an ability to produce collagen As with other neoplasms, the issue of meningioma histogenesis has
and other extracellular matrix proteins, and the finding of mesenchymal long been debated.13 The earliest terms attempted to lump these tumors
“metaplasia” in a subset of otherwise classic meningiomas (discussed in with better known extracranial categories, resulting in clumsy and
further under Histologic Variants and Grading). Epithelial features include inaccurate diagnoses, such as fungus of the dura mater, sarcoma, car-
epithelioid morphology in meningothelial (and other variant) menin- cinoma, mesothelioma, endothelioma, meningoexothelioma, and
giomas, the presence of desmosomes and other intercellular junctions, arachnoidal/meningeal fibroblastoma. In 1922, recognizing that these
epithelial membrane antigen (EMA) positivity, the capacity for papillary tumors were distinct from systemic tumor types, Cushing proposed
growth, the formation of gland-like lumens in the secretory variant, the term meningioma, a clinically practical alternative based primarily
and occasional carcinoma-like histology in anaplastic meningiomas. on anatomy, rather than unsubstantiated histogenetic notions. Various
As an appropriate candidate cell of origin, arachnoidal cap cells are hypotheses on the cell of origin were posited nonetheless, including
essentially found in the same locations as meningiomas (see Chapter dural, endothelial, fibroblastic, and epithelial cell types. In 1831, Dr.
2). These include Richard Bright raised the possibility that they might originate from the
• the external layer of the leptomeninges and arachnoid granulations inner arachnoidal lining of the dura, rather than the dense fibrous tissue
attaching loosely to the inner surface of the dura (e.g., dural-based of the dura proper. Similarly, Cleland and, later on, Schmidt emphasized
and, less often, leptomeningeal-based meningiomas); the similar histology of most meningiomas with the cell clusters that
• extensions along perivascular Virchow-Robin spaces into the line arachnoid villi. Since then, these arachnoidal cap cells have been
underlying brain (rare intracerebral meningiomas); considered the most likely, though still unproven, cell of origin for
• the tela choroidea (meningeal invagination) at the stromal base meningiomas.
of the choroid plexus (intraventricular meningiomas); and
• rare meningothelial rests/heterotopias, traumatic displacements, Incidence and Demographics
metaplasias/aberrant differentiation, and perineural extensions Meningioma accounts for about 36% of over 356,000 primary brain
(calvarial/intraosseous, scalp, sinonasal, pulmonary, and other tumors reported to the Central Brain Tumor Registry of the United
extradural meningiomas). States in the 2008 to 2012 census; it is the single most common tumor
The light- and electron-microscopic similarities include frequent subtype overall and also accounts for over half of all nonmalignant
intranuclear inclusions, interdigitating cell membranes, intercellular CNS tumors.14 The incidence is roughly 7.86 per 100,000 person-years
junctions, and cytoplasmic filaments. Arachnoidal cap cells are unique (4.68 in males and 10.62 in females), with a mean diagnostic age of 65
cell types that are also functionally similar to meningiomas. Their known years. Meningioma incidence figures have progressively risen over time,
functions include: possibly reflecting ever-increasing radiologic detection of incidental
• ensheathing or wrapping around each other, the surface of tumors. In fact, incidental meningiomas are quite common, especially
the CNS, and proximal portions of nerve roots, forming part in the elderly where they are reported in up to 2.3% of autopsies.15 The
of the CNS–cerebrospinal fluid (CSF) and blood-CSF barriers prevalence of meningioma is much higher in families with NF2 and
(recapitulated in the whorls of meningiomas); rarely in those with Gorlin, Cowden, Li-Fraumeni, Turcot, or von
• helping to maintain CSF homeostasis, both in terms of protein Hippel-Lindau syndromes as well (see Chapter 22). It also occasionally
secretion (recapitulated in the pseudopsammoma bodies of secre- runs in families without any well-established tumor predisposition
tory meningiomas) and the regulation of intracranial pressure; syndromes.
• providing trophic support for migrating neuroglial cells during Meningiomas are most common in middle-age to elderly adults and
development; and vary in incidence with race in the United States, affecting Blacks, Whites,
• taking on macrophage-like functions rarely, including some limited and Asians/Pacific Islanders in decreasing order, respectively.14 However,
capacity for phagocytosis and rheumatoid nodule formation; they are considerably more common in women than men, with a roughly
the most macrophage-like meningioma is the metaplastic- 2 to 1 female–male ratio. This sex predilection is even higher in thoracic
xanthomatous subtype. spinal meningiomas, where the ratio is nearly 9 to 1.13 Pediatric menin-
giomas are distinctly rare, but are more likely than adult counterparts
Brief Historical Overview of Nomenclature and to present with large tumor size, cyst formation, lack of dural attachment,
Histogenetic Notions high-grade histology, aggressive behavior, and/or aggressive variant
The wide-ranging biologic and histologic continuum of meningiomas histology, particularly the clear cell or papillary subtypes.16–18 They lack
has fascinated physicians since the beginning of the 20th century. One any female predominance and are more likely to present in unusual
of the greatest single advances in our understanding of these tumors locations, such as lateral ventricles, posterior fossa, and spinal epidural
came in 1938 when Drs. Harvey Cushing and Louise Eisenhardt published regions. Predisposing factors often include NF2 or a history of prior
their detailed monograph on 313 meningiomas accumulated over nearly radiation, although over half are still sporadic. In comparison to adult
30 years.2 Over the next few decades, variants became better characterized cases, the clinical behavior is more difficult to predict.
and new techniques such as karyotyping, electron microscopy, and
immunohistochemistry were subsequently applied. In 1982, a second Localization and Clinical Manifestations
major monograph by John Kepes summarized the many advances of Localization
that time, along with personal observations obtained from a review of Meningiomas occur all along the craniospinal axis (Fig. 13.1). Intracrani-
approximately 1300 cases.13 Since then, a few more aggressive variants ally, they are most common over the convexity and parasagittal regions,
have been described or clarified, hemangiopericytomas (now termed with approximately half extending to the superior sagittal sinus and

260
Meningiomas

Olfactory groove

Tuberculum 13
sellae

Lateral Cavernous
sphenoid sinus
Medial
wing
sphenoid
wing

A Foramen magnum B Petroclival


Parasagittal
Falcine

Convexity

C D
Fig. 13.1  Common locations for meningiomas. Common sites of tumor growth in relationship to adjacent skull, brain, and dural reflections (A–D). Illustrations created by MedPIC
at Washington University School of Medicine with additional input from Dr. Michael Chicoine, Department of Neurosurgery.

half affecting more lateral areas (Fig. 13.2A). Falcine meningiomas attach extramedullary tumors similar to schwannomas. Less commonly, however,
to the dense fibrous tissue of the falx rather than the sinus per se, while they present as leptomeningeal or epidural masses.
parasagittal meningiomas are located slightly more laterally (Fig. 13.1C Intraventricular meningiomas are relatively rare (0.5% to 3%) and
and D). “Skull base meningiomas” are often technically challenging to most likely originate from the tela choroidea of the choroid plexus (Fig.
resect and variably involve olfactory groove, parasellar region, lateral 13.2D). Lateral ventricles are most often involved, and there is an
or medial sphenoid wing (Fig. 13.2B), foramen magnum, cavernous unexplained left-sided predilection. Most reach a large size by the time
sinus, or tuberculum sella (Fig. 13.1A and B). Orbital (optic pathway) they present, making complete resection difficult.
meningiomas or those that involve the anterior visual pathway in general Extracranial or extradural meningiomas have been reported in the
are often highly invasive and frequently recur, despite a benign histologic orbit (see previous discussion), scalp, paranasal sinuses, nasal cavities,
appearance. Recent studies suggest some heterogeneity in this location, and the skull (“intraosseous meningioma”). In the latter, one must first
with those involving the spheno-orbital region being most clinically rule out the possibility of bone invasion from a thin en plaque or carpet-
aggressive, especially those with 1p and 6q chromosomal losses.19,20 like dural-based meningioma (Fig. 13.3).22 The origin of other extracranial
Nevertheless, advances in radiation therapy have generally improved meningiomas is controversial. In cases where extension of intracranial
local control for these tumors.21 In the posterior fossa, they arise in meningioma has been ruled out, meningothelial heterotopias or rests
regions such as the petrous bone, tentorium cerebelli, and foramen seem most plausible. Peripheral nerve, lung, salivary gland, and the
magnum. Tentorial meningiomas may be supratentorial, infratentorial, soft tissue of the little finger are even rarer sites of supposed involvement.
or both. In the lung, minute meningothelial pulmonary nodules are thought to
Spinal meningiomas occur mostly in the thoracic region (Fig. 13.2C), arise from reactive or metaplastic changes and provide one viable
although males are less likely to show this regional predilection. Cervical histogenetic explanation (Fig. 13.4).23 The function of these unusual
and thoracic examples mostly present anterior and posterior to the structures is unclear, but may involve drainage of pulmonary edema
cord, respectively. Most cover multiple segments and present as intradural fluid. Some of the older case reports could also include misdiagnosed

261
Practical Surgical Neuropathology

A B

C D
Fig. 13.2  Common meningioma sites and appearances on MRI. (A) Convexity meningioma on contrast T1-weighted MRI. Note the extra-axial location, uniform contrast enhancement,
and “dural tail” sign, with mass effect causing compression of occipital horn of lateral ventricle. (B) Sphenoid wing meningioma on contrast T1-weighted MRI. Uniformly contrast-
enhancing extra-axial mass centered on sphenoid wing and pushing adjacent frontal and temporal lobes. (C) Spinal cord meningioma on T2-weighted MRI. This thoracic cord
meningioma (lower image) pushes and compresses the spinal cord posteriorly. The normal rim of bright CSF is displaced by the tumor and compressed cord. (D) Intraventricular
meningioma on contrast T1-weighted MRI. A large homogeneously enhancing intraventricular meningioma fills the left lateral ventricle.

perineuriomas, given the extensive morphologic and immunohistochemi- The WHO grade II meningiomas (atypical, clear cell, chordoid)
cal overlap of this rare entity (see Chapter 15). are characterized by considerably increased risks of recurrence when
compared with the benign (WHO grade I) counterparts, even after a
Clinical Manifestations gross total resection. They also have a modest but statistically significant
Clinical manifestations of meningiomas vary with location and may increased mortality rate. Their accurate identification is therefore of
also include nonspecific symptoms of mass effect. Headaches, seizures, critical importance. The estimated 5-year recurrence rate for atypical
personality changes, and localizing signs such as hemiparesis, sensory meningiomas is 40% even after gross total resection, as compared with
deficits, and ataxia are most common. Parasagittal meningiomas may 5% for similarly treated benign meningiomas.24 After subtotal resection,
cause leg weakness combined with urinary incontinence. Parasellar the vast majority of atypical meningiomas recur. At a minimum, these
and orbital meningiomas often produce visual symptoms that occa- tumors require careful radiologic follow-up; whether they should be
sionally lead to blindness. Similarly, hearing loss is common in the treated with ancillary radiation therapy remains controversial and often
cerebellopontine angle meningiomas. Spinal examples compress the depends on other variables as well, such as patient age and extent of
cord, leading to motor and sensory deficits. Those in locations with resection.25,26
greater room for growth (e.g., lateral ventricles) can be asymptomatic for In addition to increased recurrence rates, WHO grade III tumors
years, finally reaching a critical mass that brings it to clinical attention. (anaplastic, papillary, rhabdoid) are associated with significant risks
It is also well known that patient morbidity and mortality increase of highly aggressive local disease, widespread dissemination, and
with peritumoral cerebral edema, as does the likelihood of psychiatric tumor-related patient death. As such, they are considered malignant
manifestations. meningiomas. They lack the usual female predominance of benign

262
Meningiomas

13

1
LAH

PFL
A 5cms B C
Fig. 13.3  (A) En plaque meningioma on contrast T1-weighted MRI. Thin, carpet-like meningioma involving a large portion of the left convexity. There is marked associated cerebral
edema with midline shift. (B) Skull x-ray from a meningioma specimen with bone invasion. Note the extensive hyperostosis associated with bone thickening and a “sunburst”
pattern of new bone formation. (C) Hyperostotic bone. Thickened lamellar bone with replacement of marrow space by aggregates of meningioma.

Fig. 13.5  “Multicentric meningioma” resection specimen. This large dural resection
specimen showed a primary tumor nodule (far right) with a nearby satellite nodule,
likely representing intradural spread of tumor.

Fig. 13.4  Minute meningothelial pulmonary nodules. Interstitial lung nodules with removed from a single patient, consistent with a monoclonal origin
aggregates of meningothelial-like cells. despite the multicentricity.30 This suggests intradural spread as a
mechanistic explanation (see Fig. 13.10C and D), which is further
meningiomas and present either as anaplastic from the outset (de novo) supported by the common finding of microscopic tumor foci in otherwise
or via progression from atypical or benign meningiomas. Either way, grossly unremarkable dura samples from meningioma patients.31 In the
the prognosis is poor with median survival of 2 to 5 years.27 Roughly rarer case of a child with multiple meningiomas, this is nearly pathog-
a quarter of patients live considerably longer, and most of their tumors nomonic for NF2.
lack 9p21 deletions (see Molecular Genetics section).1,28
Radiologic Features and Gross Pathology
Multiple Meningiomas Radiology and Cerebral Edema
Multiple meningiomas are typical of NF2, but are also seen in sporadic Neuroimaging plays a critical role in the diagnosis of meningiomas and
adult patients, representing roughly 3% of surgical and up to 8% of their therapeutic planning. Meningiomas are commonly isodense to
postmortem cases. In some patients, there is one predominant menin- gray matter on noncontrast CT and T1-weighted magnetic resonance
gioma with one or more satellite masses (Fig. 13.5), either in close imaging (MRI) studies, making it difficult to appreciate their borders.
proximity or at a distance; they often show the same variant type However, they are avidly contrast enhancing, such that meningiomas
morphology, many being transitional or psammomatous. The number as small as 3 mm are often detectable. Radiologists use the “dural tail”
of multiple meningiomas varies, with as many as 47 histologically similar sign as a helpful feature to diagnose meningioma or to confirm that the
tumors reported in one non-NF2 patient.29 Often, the same NF2 gene lesion is indeed extra-axial, rather than parenchymal in origin (see Fig.
mutation is found in multiple meningioma patients in all the tumors 13.2A). This refers to the tail-like extension of contrast enhancement into

263
Practical Surgical Neuropathology
adjacent dura. In fact, this is a nonspecific finding commonly encountered coefficient (ADC) values on diffusion-weighted imaging.33–35 However,
in nearly all dural-based masses, but given that meningioma is so much these radiopathologic correlations are far from perfect and there are
more common, it often leads to the correct diagnosis. Microscopically, this many exceptions.
dural tail often consists merely of reactive hypervascular dura, although Extent of radiologically detected peritumoral cerebral edema (dark
small foci of tumor can also be seen occasionally.1 T2-weighted images on T1, bright on T2 MRI) is highly variable, and, not surprisingly, it is
vary with tumoral consistency, such that firm, fibroblastic meningiomas often more prominent in symptomatic versus incidentally detected
often appear hypointense, whereas hyperintensity is often associated meningiomas. Extensive edema has also been associated with large
with soft, vascular, and/or meningothelial meningiomas.32 Calcifications tumor size, recruitment of pial vascular supply, location in the convexity
appear dark on T1 MRI and bright on CT. Necrosis is typically also or middle fossa, an irregular (often “mushroom-like”) brain–tumor
dark on T1, but bright on T2. interface, T2 bright signal intensity, microscopic brain invasion, histologic
Most meningiomas display a smooth, rounded edge that appears to malignancy, and variant histology that includes secretory, microcystic
compress rather than invade the adjacent brain (Fig. 13.6). An irregular angiomatous, and/or lymphoplasmacyte-rich features (see Histologic
tumor–brain interface has been associated with increased risk of finding Variants and Grading section).
brain invasion and other high-grade features on histology (Fig. 13.7).
Other features that have been associated with more aggressive menin- Gross Pathology
giomas include increased signals on both T1- and T2-weighted MRI, Meningiomas show a diverse range of gross appearances, depending
“mushrooming” into the adjacent brain (i.e., irregular contour), extensive mostly on their locations, growth patterns, and histologic subtypes (see
edema, lack of calcifications, central necrosis, and low apparent diffusion Histologic Variants and Grading section). Most are rubbery, well

A B

C D
Fig. 13.6  (A) Incidental meningioma on T2-weighted MRI. The extra-axial appearance of this meningioma is well demonstrated by the thin layer of T2 bright CSF between the
tumor and the adjacent indented, but noninvaded brain parenchyma. (B) Benign meningioma on autopsy. This meningioma showed a classic attachment to the inner surface of
the dura, with the adjacent compressed brain molded to the rounded contours of the slowly growing tumor. (C) Resected meningioma. The tumor is attached to the resected sleeve
of dura and shows a smooth external surface. (D) Resected meningioma. The tumor shows central attachment to the dura below and a bosselated cut surface.

264
Meningiomas

13

A B

C D
Fig. 13.7  (A) Atypical meningioma on contrast T1-weighted MRI. The enhancing extra-axial mass with dural tail sign is typical of meningioma, although the irregular brain–tumor
interface is worrisome for a more aggressive subtype. (B) Anaplastic meningioma on contrast T1-weighted MRI. The irregular contour, inhomogeneous signal characteristics, and
central hypodensity suggestive of necrosis are worrisome for higher grade. (C) Atypical meningioma. The soft fleshy appearance with focal papillary (cauliflower-like) configuration
is worrisome for higher grade meningioma. (D) Anaplastic meningioma. Features worrisome of high grade include the soft, almost gelatinous cut surface suggesting high cellularity
and the large yellow region of tumor necrosis evident in the upper fragment.

circumscribed, spherical, and firmly attached to the inner surface of the to be softer and more fleshy, the latter often being extensively necrotic
dura (see Fig. 13.6). The tumor surface is usually smooth or bosselated, on cut surface (see Figs. 13.5, 13.7C and D).
like cauliflower. The cut surface is white, yellow, or tan, often with The cerebral cortex adjacent to meningiomas often appears com-
intersecting white streaky bands of fibrous tissue. Xanthomatous or pressed and distorted, sometimes forming a cavity that molds perfectly
lipomatous “metaplasia” imparts a yellow color, whereas foci of metaplastic around the protuberant surface of the meningioma (see Fig. 13.6B).
cartilage and bone appear white. Psammoma bodies (“sand-like” from Nevertheless, the brain–tumor interface is typically distinct, with most
the Greek word psammos) impart a gritty cut surface. Decalcification meningiomas easily peeling away. Those that don’t are either brain
is often needed for the psammomatous variant, ossified tumors, and invasive or merely adherent to the leptomeninges. In contrast to true
those with bone invasion. The microcystic variant has a moist glistening brain invasion, infiltration of adjacent dura, venous sinuses, bones, and
cut surface, consistent with Masson’s original name of “meningioma soft tissue is relatively frequent, even in benign examples. For example,
humide.” Secretory meningiomas have a tan-red gland-like appearance. orbital meningiomas are often highly invasive of adjacent bone and
Hemorrhage and necrosis are relatively rare findings in benign menin- soft tissue, typically surrounding and compressing the optic nerve with
giomas, but are seen more commonly after therapeutic embolization. resulting loss of vision (Fig. 13.8). Other cranial and spinal nerves/
More cellular tumors such as atypical and anaplastic meningiomas tend ganglia are occasionally invaded by meningioma as well.

265
Practical Surgical Neuropathology

A B C
Fig. 13.8  Optic pathway meningioma in a patient with progressive visual loss/blindness. (A) Eye and optic nerve resection specimen. The optic nerve was markedly expanded by
a circumferentially placed meningioma. (B) Eye and optic nerve resection specimen. Cross sections reveal a large meningioma surrounding and compressing a central atrophic optic
nerve. (C) Optic nerve resection specimen. The optic nerve on the left is compressed, but not directly invaded by the meningioma on the right.

Some meningiomas are dumbbell shaped with tumor masses growing


out on both sides of a dural reflection, such as the falx or tentorium.
Intraventricular meningiomas are often large, potentially expanding and
distorting the ventricular cavity, often with resultant obstructive
hydrocephalus (see Fig. 13.2D). The en plaque meningiomas are defined
by a carpet-like growth pattern, resulting in long stretches of thickened,
sometimes rough or shaggy dura (see Fig. 13.3A). They are most com-
monly encountered in the skull base, especially the sphenoid wing,
where they tend to invade the adjacent bone, with associated hyperostosis
(Fig. 13.3B and C).
Cystic meningiomas represent 2% to 4% of all cases (Fig. 13.9). They
are mostly benign and are seen more commonly in children.36 Any
meningioma can develop grossly evident cysts, although the microcystic
variant is most frequent, with macrocysts likely forming from coalescence
of microscopic ones. In some of these cases, the dural attachment is
subtle and there may even be ring enhancement, mimicking glioblastoma.
Cystic elements are intratumoral and/or peritumoral with suspected
etiologies including secretory, reactive, and degenerative changes. In
rare cases, a cystic meningioma may form within a preexisting malforma-
tion, such as an arachnoid cyst.

Histopathology
The histopathologic spectrum of meningiomas is extremely wide (see
next section), although there are several characteristic features, seen in Fig. 13.9  Cystic meningioma on contrast T1-weighted MRI. This cystic meningioma
most tumors regardless of subtype or grade. Key features include whorl was difficult to distinguish from a primary CNS neoplasm.
formation, psammoma bodies, and characteristic cytologic features such
as nuclear clearings (“holes”) and pseudoinclusions. As stated earlier,
one of the common characteristics of meningothelial cells is to wrap cytoplasm and oval nuclei with delicate chromatin, often displaying
around various structures including themselves, resulting in cellular numerous clear vacuoles (“holes”) and pseudoinclusions (Fig. 13.10B).
whorls (Fig. 13.10A). Over time, the cells within these whorls deposit The latter represent cytoplasmic invaginations into the nuclei, such that
intercellular collagen, progressively leading to hyalinized whorls, typically on cross section, they appear membrane bound with identical tinctorial
with concentric laminations. As these structures mineralize, one sees qualities to that of the cytoplasm (i.e., eosinophilic, rather than clear).
concentric calcifications known as psammoma bodies. Cytologically, These cytologic features are by no means specific to meningiomas, but
most meningioma cells are moderately sized with ample eosinophilic are helpful for the differential diagnosis nonetheless, particularly in

266
Meningiomas

13

A B

C D
Fig. 13.10  Common histology in meningiomas. (A) Prominent whorl formation gives way to concentric rings of hyalinization and psammoma bodies. (B) Meningioma with frequent
clear nuclear holes and pseudoinclusions (eosinophilic like the cytoplasm). (C, D) Foci of dural invasion with aggregates of meningioma cells between bands of dense collagen.

variants that do not otherwise show classic architectural features, such Table 13.1  WHO 2016 Classification Scheme for Meningiomas
as whorling. The vast majority of meningiomas, even the most benign, Meningioma Variants That Are
invade the dura, and this likely represents a common mode of spread Mostly Benign (WHO Grade I) WHO Grade II Meningiomas
beyond the point of dural attachment (Fig. 13.10C and D). Other Meningothelial Atypical (any variant plus criteria)
histologic features such as cytoplasmic characteristics, degree of col-
lagenization, stromal background, architectural growth patterns, vascular Fibrous (fibroblastic) Clear cell
changes, and invasiveness into bone, soft tissue, and brain are highly Transitional (mixed) Chordoid
variable and discussed in greater detail in the next section.
Psammomatous
Histologic Variants and Grading Angiomatous (vascular)
The vast histopathologic spectrum encountered in meningiomas is
Microcystic WHO Grade III Meningiomas
astonishing and has led to several complex classification schemes
with numerous variants described over the years. In 1938, Cushing Secretory Anaplastic (any variant plus criteria)
and Eisenhardt stratified meningiomas into 9 main types and 20
Lymphoplasmacyte rich Papillary
subtypes.2 Since that time, several variants have been reclassified as
distinct entities. For example, the meningeal solitary fibrous tumor/ Metaplastic Rhabdoid
hemangiopericytoma is now considered a distinct form of sarcoma
(see Chapter 14). However, in the WHO 2016 classification scheme,
there remain nine mostly benign (WHO grade I), three intermediate (clear cell, chordoid, papillary, rhabdoid), it has been suggested that the
category (WHO grade II), and three malignant (WHO grade III) subtypes WHO grade II or III designation only be assigned if the majority of
(Table 13.1).37 For the more aggressive subtypes, it remains unclear what the tumor (>50%) shows the appropriate morphology38; nevertheless,
percentage of the variant histology must be present before the higher a study of rhabdoid meningiomas lacking overt features of malignancy
grade is automatically assigned. For the four subtypes in this category was associated with a mostly favorable prognosis even when more than

267
Practical Surgical Neuropathology
Table 13.2  WHO 2016 Grading Criteria for Meningioma
Benign Meningioma (WHO Grade I)

• Any predominant histology other than clear cell, chordoid, papillary, or rhabdoid
• Lacks criteria of atypical and anaplastic meningioma

Atypical Meningioma (WHO Grade II) (Any of Three Major Criteria)

• Mitotic index ≥4 mitoses/10 HPF


• Brain invasion
• At least three of the following five parameters:
Sheeting architecture (two-dimensional sheets with loss of whorls and fascicles)
Small cell formation (clusters of lymphocyte-like cells with high N/C ratio)
Hypercellularity
Macronucleoli
Spontaneous necrosis (i.e., not induced by embolization or radiation)

Anaplastic (Malignant) Meningioma (WHO Grade III) (Either of Two Criteria)


• Mitotic index ≥20 mitoses/10 HPF
• Frank anaplasia (sarcoma-, carcinoma-, or melanoma-like histology)
A

50% of cells appeared rhabdoid.39 In contrast to these rare variants,


any of the more common meningioma subtypes may qualify as either
atypical (WHO grade II) or anaplastic (WHO grade III) if diagnostic
criteria are met even focally (Table 13.2), either at initial presentation
or in subsequent recurrences. These subtypes are discussed in greater
detail in this chapter.

Meningiomas That Are Mostly Benign (WHO Grade I)


Meningothelial Meningioma
These are defined by lobulated, nested, or whorled aggregates of polygonal
to epithelioid cells with fuzzy or indistinct cell borders (Fig. 13.11A).
The latter imparts a syncytial appearance, explaining the older term,
“syncytial meningioma.” Large nests or islands of cells are often surrounded
by variable quantities of collagen or fibrous septae. Nuclear atypia may
occur and by itself has no prognostic bearing. Occasional examples of
B
meningothelial meningoma (or other subtypes) feature bizarre mono-
nucleated or multinucleated giant cells with dark smudgy chromatin Fig. 13.11  (A) Meningothelial meningioma. Large nests of epithelioid cells with fuzzy
and abundant cytoplasm (Fig. 13.11B). This type of nuclear atypia is intercellular borders imparting a syncytium-like growth pattern. There are also numerous
likely degenerative in nature, analogous to that encountered in “ancient nuclear clear holes. (B) Meningothelial meningioma with “ancient changes.” The bizarre
schwannomas.” nuclei in this meningioma are most likely degenerative in nature, since there are no
mitoses or other associated high-grade features.
Fibrous (Fibroblastic) Meningioma
These fibroblast-like spindle cell tumors predominantly form intersecting that develop so many individual and confluent psammoma bodies over
fascicles and storiform growth patterns, with variable but often prominent time that the calcifications obscure the underlying meningothelial cells.
collagen deposition (Fig. 13.12). The nuclei are oval to elongate, but Psammoma bodies should constitute over half of the tumor mass (Fig.
retain the overall features of meningothelial cells, including the presence 13.13B–D), with eventual dystrophic ossification (“osseous metaplasia”)
of clear holes and pseudoinclusions. Whorls and psammoma bodies also being common.
are often only present focally, but are helpful in excluding schwannoma
(see Differential Diagnosis section). Angiomatous (Vascular) Meningioma
This subtype is defined by its hypervascularity, with tumoral blood
Transitional Meningioma vessels exceeding 50% of the total volume40 (Fig. 13.14). Larger vessels
This variant is perhaps the most common and readily recognized form are often markedly hyalinized (Fig. 13.14B), while smaller, capillary-sized
of meningioma; it contains a mixture of the meningothelial and the vessels are often surrounded by bubbly or foamy tumor cells with
fibrous types, as well as cells with intermediate features (see Fig. 13.10A). degenerative nuclear atypia (Fig. 13.14C), closely mimicking the histology
Syncytial islands intermix with fascicles of collagen-rich, spindle-shaped of hemangioblastoma (see Differential Diagnosis section). This variant
cells. Whorl formation is prominent and well developed, often with often coexists with the microcystic subtype and is often associated with
numerous psammoma bodies. cerebral edema beyond that expected for its size (Fig. 13.14A).40,41 Special
studies are occasionally needed to confirm the meningothelial nature
Psammomatous Meningioma of this variant, including somatostatin receptor 2a (SSTR2a; see later
These tumors manifest commonly in the spinal cord of older women, sections) (Fig. 13.14D). Genetic studies also suggest that these tumors
where they are usually slow growing and biologically indolent. The frequently harbor polysomies (chromosomal gains), especially polysomy
marked calcifications are readily evident on both plain x-rays and CT 5, rather than the losses of chromosome 22 more commonly encountered
scans (Fig. 13.13A). They likely start out as transitional meningiomas in conventional meningiomas.42

268
Meningiomas

13

A B

C D
Fig. 13.12  Fibrous meningiomas. (A, B) Fibrous meningiomas with intersecting fascicles of spindled cells and variable collagen deposition. (C) Verocay-like bodies in a fibrous
meningioma, mimicking schwannoma. Other regions had whorls and occasional psammoma bodies. The tumor was also EMA positive. (D) Patchy EMA immunoreactivity in a fibrous
meningioma.

Microcystic Meningioma Secretory Meningioma


Microcystic meningiomas are typically T1 hypodense, T2 bright tumors This morphologic variant virtually never occurs in pure form, but most
on MRI, often with marked cerebral edema.43,44 Grossly, they are soft often develops within an otherwise meningothelial or transitional
and occasionally form macrocysts. They have a “wet” glistening cut meningioma. It represents an advanced form of epithelial differentiation
surface, justifying the original name of humid meningioma. Microscopi- characterized by the formation of gland-like lumens with eosinophilic
cally, the tumor has a “cobweb-like” appearance due to intercellular clear secretions (Fig. 13.16). Female predominance is even more pronounced
fluid collections (sometimes containing blue to pale eosinophilic mucin) in secretory meningioma than that of more common subtypes. They
between spider-like cells with long thin processes (Fig. 13.15). In fact, are most commonly encountered in the frontal convexity or sphenoid
this morphology (and ultrastructural features) more closely resembles ridge. Like the angiomatous and microcystic subtypes, secretory
the normal arachnoidal trabecular, rather than the cap cells.45 The meningiomas often induce cerebral edema out of proportion to their
pathogenesis of microcyst formation remains unclear, with proposed size.46,47 The gland-like spaces contain brightly eosinophilic, smooth or
mechanisms including secretory activity by the tumor, cystic degenera- granular PAS-positive secretions known as pseudopsammoma bodies
tion, ischemia, and penetration of the CSF into the tumor. Tumor due to their superficial resemblance to calcifications at first glance (Fig.
cytoplasm appears faintly eosinophilic or finely vacuolated. Whorls and 13.16B–F). They vary in size and may be single or multiple. Pseudopsam-
psammoma bodies are rare. Scattered bizarre nuclei and hypervascularity moma bodies may be evenly distributed, but are more often concentrated
are relatively common, as they are with the angiomatous variant; in in smaller foci. The tumor cells associated with the gland-like spaces
fact, these two meningioma subtypes are often seen together in the are usually strongly immunoreactive for cytokeratin, while carcinoem-
same tumor. In cases where EMA expression is weak or absent (given bryonic antigen (CEA) is found within the pseudopsammoma bodies
wispy processes), an SSTR2a immunostain can be helpful in establishing and/or the tumor cells surrounding them (Fig. 13.16E and F). Occasion-
the meningothelial nature. ally, there is sufficient production of CEA to elevate serum levels. When

269
Practical Surgical Neuropathology

A B

C D
Fig. 13.13  Psammomatous meningioma. (A) This heavily calcified upper cervical meningioma appears hyperintense on this CT scan and was also visible on plain x-ray. (B, C) The
majority of the tumor mass was composed of psammoma bodies, which appear red rather than purple due to acid decalcification. (D) Meningioma cells in between the psammoma
bodies are EMA positive.

compared with other meningiomas, studies have found increased have had associated monoclonal gammopathies and/or anemia. Other
pericytic, mast cell, and histiocytic elements, as well as extracellular common features reminiscent of an inflammatory process include young
matrix proteins in the secretory subtype.46,48–50 Of additional interest, age of onset, en plaque growth pattern, multicentricity, spontaneous
nearly all cases are associated with combined KLF4 K409Q and TRAF7 regression, and recurrence over multiple sites. Therefore, it is likely that
gene mutations.51 some examples diagnosed as “lymphoplasmacyte-rich meningioma”
are actually inflammatory disorders with associated meningothelial
Lymphoplasmacyte-Rich Meningioma hyperplasia. Features favoring meningioma over the latter include a
Of all meningiomas, this one is the most enigmatic and controversial. definite mass on imaging (rather than en plaque or pachymeningitis
It is characterized by dense infiltrates of lymphocytes and plasma cells pattern), larger meningothelial aggregates, and dural invasion by the
that partially obscure the underlying meningioma (Fig. 13.17).52,53 Using tumor. In the absence of these features, one should always exclude
criteria similar to other variants, the inflammatory component should other lymphoplasmacyte-rich meningeal disorders, such as infection,
comprise over half of the tumor cells; in some cases, it’s entirely intermixed Rosai-Dorfman disease, IgG4-associated pachymeningitis, inflammatory
with the meningioma, whereas in others, the inflammation is mainly at myofibroblastic tumor, Castleman disease, collagen vascular disorders,
the periphery of the tumor. Some of the previously reported patients and either primary or secondary low-grade lymphomas. Also, given

270
Meningiomas

13

A B

C D
Fig. 13.14  Angiomatous meningioma. (A) This angiomatous meningioma was associated with abundant cerebral edema (increased signal) on FLAIR MRI, despite its relatively small
size. (B, C) Marked hypervascularity with extensive hyalinization of both large and small vessels. As is often the case, the intervening tumor cells are only subtly meningothelial with
foamy spider-like cells (overlaps with microcystic) and often prominent degenerative atypia. (D) The meningioma cells are strongly and diffusely positive for somatostatin receptor 2a.

A B C
Fig. 13.15  Microcystic meningioma. (A, B) A small nest of more classic meningothelial cells is seen within this predominantly microcystic meningioma (A). The latter is characterized
by thin tumor processes and a cobweb-like lattice. (C) Focal EMA positivity outlines the thin tumor processes surrounding microcystic spaces. 271
Practical Surgical Neuropathology

A B

C D

E F
Fig. 13.16  Secretory meningiomas. (A) Secretory meningioma with a tan, gland-like appearance on cut surface. (B, C) Gland-like spaces with brightly eosinophilic pseudopsammoma
bodies seen on both intraoperative smear (B) and permanent sections (C). The latter range from smooth to granular or particulate. (D) The pseudopsammoma bodies are strongly
PAS positive. (E) The epithelioid cells forming gland-like spaces are strongly cytokeratin positive. (F) The pseudopsammoma bodies and, sometimes, the cells surrounding them show
CEA positivity.

272
Meningiomas

13

A B

C D
Fig. 13.17  (A–D) Lymphoplasmacyte-rich meningioma. On postcontrast T1-weighted MRI, the tumor appears heterogeneous, possibly representing pockets of inflammation (A).
The meningothelial component is partially obscured by dense inflammation (B). The lymphoid and meningothelial components are highlighted by the CD45 (C) and somatostatin
receptor 2a (D) stains, respectively.

that macrophages often predominate and plasma cells are not always WHO Grade II Meningiomas
conspicuous, “inflammatory-rich meningioma” has been proposed as WHO grade II meningiomas are characterized primarily by a significantly
an alternate term.52 increased risk of tumor recurrence, but also a small yet statistically
significant decrease in survival. Difficulties with local control of these
Metaplastic Meningiomas tumors account for their higher morbidity and mortality.
Virtually any type of meningioma (most often meningothelial, transi-
tional, or fibroblastic) can develop areas of mesenchymal “metaplasia,” Atypical Meningioma
including cartilaginous/chondroid, osseous, lipomatous, xanthomatous, Atypical meningioma may either arise in a recurrence of one of the
and myxoid patterns that alternate with more classic histology (Fig. more commonly benign histologic patterns already described or de
13.18). However, careful scrutiny suggests that in most examples, it is novo. Clinical risk factors include male sex, nonskull base location, and
not true metaplasia per se, but rather cytoplasmic fat accumulation, prior surgery.57 The diagnosis is based on the presence of one or more
vacuolization, dystrophic ossification, or other changes within otherwise established microscopic criteria (see Table 13.2). These 2016 WHO
EMA (Fig. 13.18D) and/or SSTR2a (Fig. 13.18E) immunoreactive criteria1 are based mostly on data from two large clinicopathologic
meningothelial cells that also retain their characteristic meningothelial series from the Mayo Clinic.24,27 In these series, the estimated 5-year
features ultrastructurally. As such, Roncaroli et al. have proposed alternate recurrence rate for gross totally resected atypical meningiomas was
terms, such as lipidized rather than lipomatous meningioma.54 Also, in 40%, as compared to 5% for similarly treated benign meningiomas. This
the rare cases of myxoid and chondroid meningiomas, one must take considerably increased risk of recurrence justifies the WHO grade II
care to distinguish them from chordoid meningiomas (see upcoming designation. Despite the name “atypical meningioma,” cytologic atypia
discussion), given some overlapping features.55,56 is neither required nor reliable given that benign meningiomas may

273
Practical Surgical Neuropathology

A B

C D

E F
Fig. 13.18  “Metaplastic meningiomas.” (A) Dystrophic ossification or “bone metaplasia” in the center of an otherwise typical meningioma. (B–E) Lipidized meningioma (or
“lipomatous metaplasia”) with accumulation of large globules of fat within meningioma cells, many of which still demonstrate EMA (D) and somatostatin receptor 2a (E) positivity.
(F) Chondroid (cartilaginous) metaplasia with islands of cells containing basophilic matrix resembling cartilage.

274
Meningiomas

13

G H
Fig. 13.18, cont’d  (G, H) Meningioma with “xanthomatous metaplasia” due to accumulations of lysosomes and/or small lipid vacuoles. The similarity to macrophages is highlighted
in G, whereas H demonstrates earlier metaplasia consisting of clear vacuoles but retained spindled cytology of fibrous meningioma.

A B
Fig. 13.19  (A, B) Embolized meningioma. In this case, the meningioma was embolized with polyvinyl alcohol (PVA) imparting a striped “tigroid” appearance in dural (A) and
intratumoral (B) vessels, due to intermixed PVA and fibrin thrombus. The induced tumor necrosis often appears acute with nuclear debris (B) and should not be used for meningioma
grading.

show considerable degenerative nuclear atypia (see Fig. 13.11B), while whereas the other features require greater time spent at medium to
mitotically active meningiomas can be completely monomorphic and high magnifications.
cytologically bland. Likewise, although it makes the surgery more chal- Based on these WHO criteria, the diagnosis of atypical menin-
lenging, invasion of adjacent dura, bone, and soft tissue do not warrant gioma has become more common. Whereas most studies before
a higher grade. Lastly, embolization-induced necrosis (Fig. 13.19) should 2000 quoted frequencies of 5% to 7%, they comprise 15% to 25% in
not be utilized as evidence of higher grade, since it is iatrogenically modern series.24,58,59 For instance, increased mitotic count was the most
induced. The same applies to radiation-induced ischemia. In other words, common reason for “upgrading” in a careful review of 314 previously
only “spontaneous necrosis” is considered when grading meningiomas diagnosed meningiomas, emphasizing the importance of review at
(Table 13.2). Instead, atypical meningiomas are currently defined by higher magnification, even when “worrisome features” are not seen on
at least focal presence of either high mitotic index (≥4 mitoses per 10 initial scan.58
high powered fields [HPF]), brain invasion (discussed later), or at least
three of the following five features: hypercellularity (originally defined Clear Cell Meningioma
as >53 nuclei/HPF diameter, but mostly a subjective impression in daily Clear cell meningiomas are rare (<1% of meningiomas), have a strong
practice), small cell formation (clusters of lymphocyte-like tumor cells predilection for the spinal cord and posterior fossa, and often present at a
with high nuclear/cytoplasmic ratio), macronucleoli (e.g., visible at 100×), younger age, with many reported cases encountered in infants, children,
sheeting architecture (loss of whorled or fascicular growth patterns), and young adults (Fig. 13.21A and B). They are composed of sheets
and spontaneous necrosis (Fig. 13.20). Often, the two-dimensional of polygonal cells with clear cytoplasm (Fig. 13.21C), resulting from
sheeting and small cell formation are evident at low magnification, glycogen accumulation which can be demonstrated as diastase-digestible

275
Practical Surgical Neuropathology

A B

C D

E F
Fig. 13.20  Atypical meningiomas. (A) Sheeting architecture with loss of whorl and fascicle formation. (B) Small cell formation (and sheeting) with clusters of lymphocyte-like tumor
cells that have lost most of their cytoplasm (i.e., high NC ratio). (C) Macronucleoli. (D) Focus of “spontaneous necrosis.” Note the lack of nuclear debris that is more commonly
seen in embolization-induced necrosis (see Fig. 13.19B). (E) Increased proliferative index (>4% labeling index) on Ki-67 immunostaining. (F) Only a rare PR positive cell is seen,
providing further (albeit nonspecific) support for high-grade histology.

276
Meningiomas
PAS positivity (Fig. 13.21E and F).60,61 Whorl formation is often subtle originally reported 61% recurrence and 23% mortality rates in their 14
and psammoma bodies are typically absent, making it more difficult
to recognize the meningothelial nature of this tumor. A distinctive
cases, with one also showing CSF dissemination (Fig. 13.21B).61 Others
have similarly reported both local and distant forms of progression.63 Rare
13
feature, however, is the often prominent interstitial and perivascular examples have featured prominent inflammatory components.64 Both
hyalinization. In some, this collagenization becomes confluent, forming familial and sporadic clear cell meningiomas have strong associations
large acellular zones of hyalinization, overlapping with reported cases of with SMARCE1 mutations and loss of protein expression.65,66
the so-called “sclerosing meningioma” (Fig. 13.21D).62 Thick, brightly
eosinophilic amianthoid-type fibers may also be seen. Despite a bland Chordoid Meningioma
cytology in most cases, the behavior is surprisingly aggressive, justifying As one might guess, chordoid meningioma was named for its striking
its WHO grade II designation. For instance, Zorludemir and colleagues resemblance to the bone tumor, chordoma. It is a rare subtype, accounting

A B

C D
Fig. 13.21  Clear cell meningiomas. (A, B) Postcontrast MR images showing a large posterior fossa/cervicomedullary junction primary (A) and a spinal drop metastasis (B). (C)
Predominantly clear cytoplasm and only vague whorling. (D) Focus of advanced interstitial and perivascular hyalinization forming hypocellular sclerotic zone on trichrome stain.
Continued

277
Practical Surgical Neuropathology

E F

G H

Fig. 13.21, cont’d  (E, F) Increased cytoplasmic glycogen confirmed by PAS without (E) and with diastase (F) treatment, the latter eliminating PAS staining. (G, H) The tumor shows
patchy EMA (G) and diffuse somatostatin receptor 2a (H) expression.

for roughly 0.6% of meningiomas in one large series.67 Most cases present Brain Invasive Meningioma
as large, difficult to resect, supratentorial masses (Fig. 13.22A).68–71 The The issue of brain invasion in meningioma grading has been a contro-
tumor is often vaguely lobulated with abundant basophilic mucin and versial one for many decades. It is histologically defined as extension of
epithelioid to spindled cells arranged in ribbons or cords (Fig. 13.22B–D). tumor cells beyond the pial surface into the adjacent brain parenchyma
The mimicry of chordoma is further enhanced by the presence of foamy (Fig. 13.23). It is typically found at the periphery of larger meningio-
or vacuolated “physaliferous-like” cells. A prominent lymphoplasmacytic mas, as irregular tongue-like protrusions associated with gliosis in the
infiltrate is occasionally seen and was stressed in the original descrip- adjacent brain, and especially in entrapped fragments of brain within
tion in 1988.70 Foci of conventional meningothelial or transitional the tumor. Extension of tumor along leptomeningeal Virchow–Robin
meningioma may be seen, although others are composed entirely of spaces is occasionally seen, particularly in pediatric examples with or
chordoid morphology, making it difficult to recognize the tumor as without associated meningioangiomatosis; however, this should not
meningioma, especially since psammoma bodies are also uncommon. The be misconstrued as true brain invasion. The previously published view
original series of seven cases reported children with Castleman disease, that brain invasion in meningiomas represents the most reliable sign
anemia, and a prominent inflammatory response to the tumor; systemic of malignancy is no longer tenable.27 Careful studies of brain invasive
findings disappeared upon tumor resection and reappeared with tumor meningiomas that were otherwise benign or otherwise atypical showed
recurrence.70 However, larger series have found these clinicopathologic recurrence and mortality rates that were virtually identical to those of
associations to be relatively rare.69 Nonetheless, chordoid meningiomas atypical meningiomas (WHO grade II) defined by other criteria. As such,
are particularly likely to recur, particularly following subtotal resection the 2016 classification scheme now considers brain invasion to represent
(nearly 100% in this setting), justifying the WHO grade II designation. another criterion for atypical meningioma, WHO grade II, even in the
As with other meningiomas, however, the prognosis is considerably absence of any other worrisome features.1 Nevertheless, this remains a
better with complete resection.72 Rare examples of CSF dissemination controversial issue given that some studies still suggest that such cases are
have been reported.73 less aggressive than conventionally defined atypical meningiomas.74 This

278
Meningiomas

13

A B

C D

E F
Fig. 13.22  Chordoid meningiomas. (A) Large supratentorial chordoid meningioma (clinically recurrent) with extensive frontal sinus invasion on contrast T1-weighted MRI. (B) Gross
pathology shows mucoid and glistening cut surface, which may be similar to that seen in microcystic meningiomas. (C, D) Chordoma-like histology with cords of small epithelioid
cells and bubbly physaliferous-like clear vacuoles (C), set within an Alcian blue–positive mucin-rich stroma (D). (E, F) Meningothelial features include EMA (E), somatostatin receptor
2a (not shown), and, sometimes, progesterone receptor (F) expression.

279
Practical Surgical Neuropathology

A B C
Fig. 13.23  (A–C) Brain invasive meningioma. Brain invasion is characterized by tongue-like protrusions of meningioma into adjacent often gliotic brain (A, B), with entrapped
parenchyma highlighted on GFAP stains (C).

distinction between brain invasive and other high-grade meningiomas • Ultrastructural features of meningioma
is also based in part on molecular genetic observations, showing that • Imbricating cellular processes
otherwise benign brain invasive meningiomas less commonly harbor • Intercellular junctions, including desmosomes
classic “progression-associated alterations” (see Genetics section).75 As • Genetic features of high-grade meningioma (see later discussion)
such, additional studies with larger numbers and longer follow-up times • 22q, 1p, 6q, 10, 14q, 18q, and/or 9p (p16 gene) deletions (Fig.
are still needed to resolve the remaining controversies. 13.24F–H)
• NF2 and TERT promoter region mutations
WHO Grade III (Malignant) Meningiomas Other common features in anaplastic meningiomas include extensive
The WHO grade III meningiomas are those that have a considerably zones of geographic necrosis, brain invasion, atypical mitotic figures,
increased risk of not only recurrence, but also metastatic dissemination and Ki-67 labeling indices exceeding 20% (Fig. 13.24E). Whereas
and patient death from disease. As such, they are considered overtly the revised WHO criteria have led to increased numbers of atypical
malignant. meningiomas, the more stringent definition of malignancy has led to a
less dramatic but noticeable decrease in anaplastic meningiomas, with
Anaplastic (Malignant) Meningioma current estimates for the latter being 1% to 6%.1,27,58,76 They should
These highly aggressive WHO grade III neoplasms are defined primarily not be confused with a variety of true meningeal sarcomas; thus,
by the presence of cellular anaplasia and/or excessive mitotic activity the older term “sarcomatous meningioma” should not be used for
(see Table 13.2). Earlier definitions of anaplastic meningioma were anaplastic meningiomas that are predominantly spindled or collagen
problematic, since they were often vague, sometimes depended mainly rich (Fig. 13.24B). Nevertheless, there are rare anaplastic meningiomas
on the presence of brain invasion (see prior discussion), and were not with compelling carcinoma-like (e.g., malignant glands) or sarcoma-
always associated with a particularly poor prognosis. However, since like (e.g., rhabdomyoblastic, myofibroblastic) foci of metaplasia
the 2000 version, the WHO schemes adopted stricter criteria based on (Fig. 13.25).77–79
previously published statistical associations with overall patient survival:
either at least 20 mitoses per 10 HPF or overtly malignant sarcoma-, Papillary Meningioma
carcinoma-, or melanoma-like cytology (Fig. 13.24A and B).1,27 These Papillary meningiomas are rare (<1% of all meningiomas) cellular
features may be encountered focally or diffusely. In the latter case, a tumors characterized, at least in part, by (1) a papillary or more often
firm diagnosis of meningioma requires additional supporting evidence, a pseudopapillary pattern resulting from discohesive tumor cells (Fig.
such as: 13.26B), and (2) a perivascular arrangement of epithelioid tumor cells
• History of prior meningioma at the same site (often better resembling the pseudorosettes of ependymoma (Fig. 13.26C and E). When
differentiated) the architectural features are well developed, it may appear grossly or
• Immunohistochemical features of meningioma (see later radiologically similar to other papillary neoplasms with a cauliflower-
discussion) like growth pattern (Fig. 13.26A). In cases with abundant cytoplasm,
• Strong diffuse vimentin positivity (Fig. 13.24C) tumor cells often appear epithelioid (Fig. 13.26E and F) to rhabdoid (see
• Strong diffuse SSTR2a immunoreactivity upcoming discussion), suggesting metastatic carcinoma, melanoma, and
• Patchy EMA expression (Fig. 13.24D) other malignancies, but nevertheless maintaining immunoprofiles typical
• Progesterone receptor usually negative or only focally expressed of meningioma. This variant was originally described by Cushing and
• Limited or no cytokeratin expression Eisenhardt in a patient who underwent 17 operations, and eventually
• Lack of nuclear STAT6 staining (i.e., rule out SFT/HPC) died with pulmonary metastases.2 Tumor nuclei often resemble those

280
Meningiomas

13

A B

C D

E F
Fig. 13.24  Anaplastic meningioma. (A) Carcinoma-like region with excessive mitotic activity (>20/10 HPF). (B) Fibrosarcoma-like spindled region in the same tumor. (C) Strong
and diffuse vimentin positivity. (D) Patchy EMA positivity and lack of cytokeratin (not shown) supported a meningothelial derivation. (E) The Ki-67 labeling index in this case was
similarly more than 20%. (F–H) FISH studies showed typical genetic features of high-grade meningioma, including 22q loss (F: BCR gene in red, NF2 gene in green; only one signal
Continued

281
Practical Surgical Neuropathology

G H
Fig. 13.24, cont’d  in most cells) and 1p deletion (G: 1p32 in green, 14q32 in red; most nuclei have one green and two red signals). FISH also showed 9p21 deletion (centromere
9 in green and 9p21 in red; one red and two green signals in most nuclei), consistent with the more aggressive subset of anaplastic meningioma.

A B

C D
Fig. 13.25  Anaplastic meningiomas with metaplastic features. (A, B) Anaplastic meningioma with adenocarcinoma-like foci. This case had CK7 immunoreactive (B) epithelioid cells
forming gland-like structures intermixed with anaplastic meningioma. Both components showed classic meningioma-associated deletions by FISH (not shown). (C–F) Anaplastic
meningioma with rhabdomyosarcoma-like foci. Predominantly spindled anaplastic meningioma with scattered brightly eosinophilic cells suggestive of rhabdomyoblasts (C, D).

282
Meningiomas

13

E F
Fig. 13.25, cont’d  The latter were positive for desmin (E), whereas larger meningioma-like nests expressed vimentin most avidly (F). Both components showed classic meningioma-
associated deletions by FISH (not shown).

A B

C D
Fig. 13.26  (A–F) Papillary meningiomas. Occasional examples where papillary architecture is well developed will show a “cauliflower-like” gross or postcontrast MRI appearance
(A). This tumor was characterized by a pseudopapillary growth pattern due to poor tumor cohesion (B). Remaining cells cling to blood vessels with perivascular nuclear-free zones
reminiscent of ependymal pseudorosettes (C). The Ki-67 labeling index was elevated (D). Continued

283
Practical Surgical Neuropathology

E F
Fig. 13.26, cont’d  Another example shows papillary architecture combined with epithelioid cytology (E) and strong vimentin positivity (F).

of meningothelial cells, and regions of more classic variant histology are therefore, it is recommended such cases be designated as “meningioma
commonly found; in fact, the papillary pattern predominates in only with focal rhabdoid features, WHO grade I,” with a comment that closer
about a fourth of cases. However, the diagnosis of “papillary meningioma, follow-up may be warranted given at least a potential for more aggressive
WHO grade III” should only be rendered when this architectural pattern behavior.39 As mentioned previously, rhabdoid cytology is occasionally
predominates (i.e., >50% of tumor). Otherwise, normal grading criteria combined with papillary architecture. Most recently, it has been shown
should be applied followed by “with focal papillary features” in the that the most aggressive rhabdoid meningiomas often lose BAP1 expres-
diagnostic line and a comment regarding the need for close clinical sion (Fig. 13.27D), a finding that can be associated with germline as
follow-up due to a potential for more aggressive behavior. In cases well as somatic BAP1 mutations.88 Lastly, it should be noted that rare
with multiple recurrences, the papillary morphology often becomes “pseudo-rhabdoid” meningiomas show complex interdigitating cell
more prominent over time. The vascular walls may occasionally show processes on electron microscopy, rather than whorled bundles of
hyaline thickening. Mitotic activity is variable, although some high-grade intermediate filaments; the significance of this rare variant is unknown
histologic features are seen in the majority of cases (Fig. 13.26D). at this time.89
Papillary meningiomas correspond to WHO grade III; they are highly
aggressive and have a propensity to invade, recur, and metastasize.80,81 Other Meningioma Patterns Not Included in the WHO
In one review of 46 cases, 75% invaded the brain or other adjacent A small series of six oncocytic meningiomas was initially reported in
structures, 55% recurred, and 20 metastasized.82 Roughly half of the 1997.90 Histologically, these were characterized by granular eosinophilic
patients die of disease. They also tend to occur in younger patients, cytoplasm that corresponded to mitochondrial accumulations (Fig.
roughly half presenting in children.80 Occasional examples combine 13.28). These tumors had both aggressive histologic features and frequent
papillary architecture with rhabdoid cytology (Fig. 13.27A).83 recurrences. However, a second group subsequently reported benign
histology and clinical behavior in five additional cases.91 Therefore,
Rhabdoid Meningioma additional experience is needed and, to date, it has not been officially
Representing another rare variant (<1%), rhabdoid meningiomas contain recognized by the WHO. Meningiomas with granular cytoplasm due
discohesive aggregates of classic rhabdoid cells (see Fig. 13.27).84,85 When to lysosome accumulation or other cellular structures are even rarer92,93
fully developed, the latter contain large eccentric vesicular nuclei, variably and perhaps overlap somewhat with the metaplastic-xanthomatous
prominent nucleoli, and densely eosinophilic cytoplasm with paranuclear variant. Similarly, a “sclerosing” or “sclerosing whorling” variant has been
globular or whorled filamentous inclusions (Fig. 13.27B and C). Ultra- reported and is defined by extensive hyalinization that obscures
structurally, the paranuclear inclusions correspond to whorled bundles the underlying meningothelial cytology (Fig. 13.29).62,94–96 Originally
of intermediate filaments, the latter most commonly being vimentin described as a pediatric variant, it has subsequently been reported in
(Fig. 13.27C), although GFAP and desmin immunoreactivity have also adults, sometimes associated with more conventional variant histology
been reported occasionally.85–87 Most meningiomas with fully developed and sometimes with the clear cell variant. A few have shown the unusual
rhabdoid cells have additional features of malignancy, such as high feature of GFAP positivity in meningothelial cells. Therefore, it remains
mitotic indices, cellular anaplasia, and necrosis. Such examples are highly unclear whether this is truly a distinct variant or merely an exaggerated
aggressive clinically and have therefore been assigned a WHO grade of form of the normal hyalinization encountered to lesser degrees in nearly
III. In one series of 15 cases, 13 (87%) had recurrences, 2 (13%) had all types of meningioma. Another rare histologic pattern reported in
extracranial metastasis, and 8 (53%) had died of disease.85 Occasionally, the literature is the mucinous meningioma, although it overlaps with
though, rhabdoid cells are seen focally in otherwise benign (or atypical) both the chordoid and metaplastic-myxoid subtypes.97 Occasionally,
meningiomas, and often the rhabdoid features are not fully developed meningiomas will form diagnostically confusing structures such as
(e.g., still cohesive, eccentric eosinophilic cytoplasm without paranuclear nuclear palisades resembling the Verocay bodies of schwannoma (Fig.
inclusions). Such cases do not automatically behave more aggressively; 13.30) or meningothelial rosettes simulating those of embryonal neoplasms

284
Meningiomas

13

A B

C D
Fig. 13.27  (A–D) Rhabdoid meningiomas. Rhabdoid features include large cells with vesicular nuclei, variably prominent nucleoli, eccentrically placed eosinophilic cytoplasm, and
globular paranuclear inclusions (A, B), the latter of which are often vimentin immunoreactive (C). A subset of rhabdoid meningiomas show loss of BAP1 expression, often accompanied
by aggressive clinical behavior (D, Complements of Dr. Sandro Santagata, Brigham and Women’s Hospital, Boston, MA.)

A B
Fig. 13.28  (A, B) “Oncocytic meningioma.” Enlarged cells with coarsely granular cytoplasm (A) due to accumulation of mitochondria, the latter highlighted on an antimitochondrial
immunostain (B).

285
Practical Surgical Neuropathology

A B

C D
Fig. 13.29  (A–D) “Sclerotic whorling meningioma.” This meningioma had small foci of classic cytology, but was predominantly composed of large hypocellular zones replaced by
dense collagenization (A–C). Meningothelial cells showed strong PR positivity (D).

A B C
Fig. 13.30  (A–C) Meningioma with extensive nuclear palisades. Nuclear palisades in meningioma (A) may mimic the Verocay bodies of schwannoma, but retain expression of
meningothelial markers such as EMA (B) and progesterone receptor (C).

286
Meningiomas

13

A B C
Fig. 13.31  (A–C) Meningothelial rosettes. Meningothelial rosettes feature central fibrillar material (A) or lumens (not shown) and are primarily composed of vimentin-positive cell
processes (B) and/or collagen (C; trichrome).

A B C
Fig. 13.32  (A–C) Granulofilamentous inclusions. Granulofilamentous meningiomas feature eosinophilic inclusions (A) that may superficially resemble those of rhabdoid meningioma,
but are more irregular and contain clear vacuoles with one or more central granules. The inclusions appear red on trichrome stain (B) and the vacuoles can be vimentin positive (C).

(Fig. 13.31); the central zones of the latter may be variably composed without any aggressive histologic features. These “benign metastasiz-
of cell processes and/or collagen.98 Finally, there are rare meningiomas ing meningiomas” spread to the lungs and pleura most often (Fig.
with unusual inclusions or extracellular deposits, such as the granulo- 13.34), followed in frequency by the musculoskeletal system, CSF,
filamentous meningioma, containing eosinophilic cytoplasmic inclusions liver, reticuloendothelial system, and kidneys; the prognosis for such
with one or more vacuoles (Fig. 13.32), or the meningioma with tyrosine patients can be surprisingly favorable; and, in some, the metastatic foci
crystals (Fig. 13.33)99; the granulofilamentous variant may be confused are clinically silent.102,103 In general, factors associated with increased
with rhabdoid meningiomas but has generally been considered benign risk of metastasis include prior craniotomy, venous sinus invasion, local
in the few cases reported to date.100,101 recurrence, WHO grade III, and papillary and/or rhabdoid variant
morphology.102
Metastasis of and to Meningiomas For reasons that remain enigmatic, meningioma is also one of the
Metastasis is an exceedingly rare event, estimated to involve only 0.1% most common receptor neoplasms for systemic metastases. Breast and
of meningiomas. Forms of spread include both systemic and CSF lung carcinomas are particularly common primaries to metastasize to
(e.g., drop metastases). Most metastatic meningiomas are histologi- meningioma, although other malignancies have also been reported.104 In
cally malignant, although in up to 34% of all examples (especially if most cases, the meningioma is histologically benign, although sometimes
including incidentally identified metastases), it occurs in tumors the cytology of the metastasis and the meningioma are similar enough

287
Practical Surgical Neuropathology

A B
Fig. 13.33  (A, B) Tyrosine crystals. These rare deposits feature petal-shaped eosinophilic crystals (A) that appear red on trichrome stain (B).

A B

C D
Fig. 13.34  (A–D) Benign metastasizing meningioma. This patient with a previously resected intracranial meningioma was found to have multiple lung nodules years later, one of
which was PET avid (A; arrow) and underwent needle biopsy. The histology resembled a benign meningioma (B) with patchy EMA positivity (not shown), lack of keratin expression
(not shown), diffuse somatostatin receptor 2a expression (C), extensive progesterone receptor positivity (D), and a low Ki-67 labeling index (not shown).

288
Meningiomas
that only careful inspection reveals the malignant component (Fig. 13.35). 13.30). Of note, up to 70% of fibrous meningiomas are S-100 positive,
Immunohistochemical studies are useful in this scenario. although this is often patchy, rather than the diffuse pattern encountered
in schwannomas. A similar pitfall is that SSTR2a can be positive in
13
Differential Diagnosis both.105,106 Unlike meningothelial cells, Schwann cells lay down a continu-
Most meningiomas are readily diagnosed from routine H & E–stained ous basement membrane. Therefore, reticulin histochemical and type
sections alone. However, the remarkably wide morphologic spectrum IV collagen immunohistochemical stains are useful to highlight the
that can be encountered causes considerable diagnostic difficulties in widespread peritumoral basement membrane network of schwannomas.
selected cases (Table 13.3), particularly with some of the rarer or higher The basal lamina expression is also demonstrable by electron microscopy,
grade variants. The majority of differential diagnoses can be resolved whereas fibrous meningiomas show classic ultrastructural features of
using immunohistochemistry (see Ancillary Diagnostic Studies section), meningiomas in general (see Ancillary Diagnostic Studies section). Also,
although in rare and often poorly differentiated meningiomas, ultra- schwannomas are strongly SOX10 positive, while meningiomas are
structural or genetic studies may be required to establish the diagnosis. typically negative.107 Lastly, schwannomas are consistently negative for
The differential between fibrous meningioma and schwannoma is a EMA, whereas most fibrous meningiomas will show at least focal
common one, especially in the cerebellopontine angle and in the spinal immunoreactivity.
cord, where both tumors are regularly encountered. This is also a common In small tumors, the differential diagnosis between meningothelial
differential in the setting of NF2, where patients are predisposed to meningioma and meningothelial hyperplasia can occasionally be chal-
both tumor types (see Chapter 22). Whorls and psammoma bodies are lenging, though the latter never invades into the dura and is often
found at least focally in most fibrous meningiomas, but one is occasionally associated with other pathologic features, such as hemorrhage, inflam-
surprised by schwannomas that contain well-developed whorls and mation, or the presence of another neoplasm.108 Most cases of hyperplasia
meningiomas with Verocay body–like structures (see Figs. 13.12C and are also detected incidentally.

A B

C D
Fig. 13.35  Benign meningioma with metastatic lung carcinoma. (A, B) Typical dural-based meningioma (A) with slightly enlarged, mitotically active epithelioid cells in central
portion of tumor (B; left side). (C) Strong cytokeratin was seen in the central focus of metastatic carcinoma, as well as CK7, CEA, and TTF-1 (not shown). (D) In contrast, the
peripheral zones of benign meningioma were strongly vimentin positive.

289
Practical Surgical Neuropathology
Table 13.3  Differential Diagnoses for Meningioma Variants The primary diagnostic consideration for psammomatous meningioma
Variant DDx is a rare entity known as calcifying pseudoneoplasm of the neuraxis
(CAPNON). This lesion is considered an unusual reactive process that
Meningothelial or Transitional Metastatic Carcinoma
Meningothelial Hyperplasia
involves the meninges and/or adjacent bone of the skull or spine.109–111
Pituitary Adenoma/Neuroendocrine Tumor MR imaging often reveals a lobulated, rim-enhancing mass, the center
of which is hypodense on all sequences, suggesting calcification (Fig.
Fibroblastic Schwannoma 13.36A and B). An older term for this lesion was fibro-osseous lesion of the
Solitary Fibrous Tumor/Hemangiopericytoma (SFT/HPC)
neuraxis, and some have amusingly referred to this lesion as “CRUDoma”
Psammomatous Reactive Process With Calcification due to its distinctive basophilic “crud” (Fig. 13.36C). Most reported cases
Calcifying Pseudoneoplasm of the Neuraxis have been sporadic and idiopathic in nature. The basophilic material is
Calcium Pyrophosphate Deposition Disease (Tophaceous nonetheless fairly distinctive in terms of a fibrillated quality at least focally,
Pseudogout) which resembles “chicken feet or footprints.” It is variably calcified or
Angiomatous (Vascular) Hemangioblastoma even ossified. The etiology of this material remains a mystery, but often
Vascular Malformation is associated with reactive epithelioid macrophages and meningothelial
cells (Fig. 13.36D). When arranged in multilayered nests or whorls, the
Microcystic Diffuse or Pilocytic Astrocytoma
latter constitutes a form of meningothelial hyperplasia and should not
Clear Cell Meningioma
be mistaken for an underlying meningioma. Inflammation and foreign
Secretory Metastatic Adenocarcinoma body reaction may also be encountered, as well as crystalline deposits
resembling the calcium pyrophosphate of pseudogout (see upcoming
Lymphoplasmacyte Rich Inflammatory/Infectious Process
IgG4 Disease/Collagen Vascular Disorders
discussion).112 Surgery is usually curative. Another rare differential
Marginal Zone (MALT) Lymphoma with psammomatous meningioma is indeed tophaceous pseudogout or
Meningothelial Hyperplasia calcium pyrophosphate disease (CPPD) (Fig. 13.37); such cases have
Angiomatoid Fibrous Histiocytoma (AFH) been reported primarily in the spine, though the distinctive rhomboidal
calcium pyrophosphate crystals (Fig. 13.37B) appear quite different than
Metaplastic (Osseous, Soft Tissue Tumors
the concentric layers of calcium in psammoma bodies.113,114
Cartilaginous, Xanthomatous, Histiocytic Disorders
Myxoid, Lipidized, etc.)
Of the rare benign histologic variants, the secretory, microcystic,
lymphoplasmacyte-rich, and chordoid meningiomas are most likely to
Clear Cell (WHO Grade II) Metastatic Renal Cell Carcinoma cause diagnostic difficulties. Secretory meningiomas may be confused with
Microcystic Meningioma adenocarcinomas, since they form gland-like spaces with both cytokeratin
Hemangioblastoma
and CEA positivity (see Fig. 13.16). However, the pseudopsammoma
Chordoid (WHO Grade II) Chordoma bodies are fairly distinctive and regions of more typical meningioma
Chordoid Glioma of Third Ventricle are usually also present. Microcystic meningiomas superficially resemble
Epithelioid Hemangioendothelioma low-grade astrocytomas (see Fig. 13.15B), a mimicry that may actually be
Papillary (WHO Grade III) Papillary Ependymoma
more than just superficial at the time of frozen section. However, the dural
Astroblastoma attachment is an important clue and areas of more classic meningioma
Meningeal SFT/HPC are often found as well (Fig. 13.15A). Nonetheless, if it remains unclear
Metastatic Malignancy even on permanent sections, the lack of GFAP, OLIG2, and SOX10
immunoreactivities argue against this diagnostic consideration. The
Rhabdoid (WHO Grade III) Metastatic Malignancy
controversial nature of the lymphoplasmacyte-rich meningiomas has
Atypical Teratoid/Rhabdoid Tumor
GBM/Gliosarcoma
already been discussed. Care is needed to avoid misinterpretation of
Granulofilamentous Meningioma scattered hyperplastic meningothelial nests as being neoplastic and a
Rhabdoid-like Meningioma With Complex Interdigitating number of other lymphoplasmacyte-rich disorders should be excluded,
Cell Processes (Inclusions) including various infections, sarcoidosis, IgG4-associated pachymeningitis
(Fig. 13.38), inflammatory myofibroblastic tumor, meningeal rheumatoid
Anaplastic (WHO Grade III) Metastatic Carcinoma/Melanoma
nodules, Wegener granulomatosis, Rosai-Dorfman disease (extranodal
Meningeal SFT/HPC
Other Meningeal Sarcomas
sinus histiocytosis), Castleman disease, and low-grade lymphomas, such
as marginal zone lymphoma (see also Chapters 14, 17, and 23).
The differential between chordoid meningioma and chordoma may
The occasional difficulty with the differential diagnosis with astro- be problematic in diagnosing a midline, skull base tumor. However, this
cytoma can usually be resolved by immunostaining for glial markers is rare and most chordoid meningiomas present as large, dural-based
such as GFAP, OLIG2, and SOX10, which are typically positive in supratentorial masses (see Fig. 13.22A), without involvement of the sites
astrocytomas and nearly always negative in meningiomas. Meningiomas, most commonly implicated in chordoma, such as the clivus or sacrum.
particularly the meningothelial type, may superficially mimic metastatic In addition to neuroimaging, immunocytochemistry may be helpful.
carcinoma, especially in higher grade examples (see Fig. 13.24A). EMA Chordomas are typically much more strongly and diffusely cytokeratin
is positive in both, although usually stronger in carcinomas, most of and S-100 positive than chordoid meningiomas, as well as expressing
which also strongly express various cytokeratins (see Fig. 13.35C). In the more specific marker, brachyury.115
contrast, even poorly differentiated meningiomas are typically strongly Clear cell meningiomas may superficially resemble metastatic renal
and diffusely positive for vimentin, a nonspecific finding that would cell carcinoma (see Fig. 13.21), although they have a fairly unique
nevertheless be uncommon in metastatic carcinomas (see Fig. 13.30D). pattern of dense perivascular and interstitial collagen deposition that
Lastly, SSTR2a is typically diffusely positive in even the most anaplastic is not a feature of classic renal cell carcinoma.61 Both tumors may
meningiomas, whereas metastatic carcinomas lack this pattern with have abundant cytoplasmic PAS-positive diastase digestible glycogen,
the exception of neuroendocrine tumors.106 although this is not true of microcystic meningioma, which also

290
Meningiomas

13

A B

C D
Fig. 13.36  Calcifying pseudoneoplasm of the neuraxis. T1-weighted MRI with gadolinium shows a meningeal-based ring-enhancing mass with a hypodense center (A). This center
remains hypodense on other modalities, including T2-weighted images (B). On histology, the center of the lesion consists of variably calcified, fibrillated basophilic material (C),
which is surrounded by epithelioid macrophages and reactive meningothelial cells (D).

A B C
Fig. 13.37  (A–C) Calcium pyrophosphate deposition disease (tophaceous pseudogout). This spinal contrast-enhancing mass was favored to represent a meningioma on MRI (A).
Histology showed typical rhomboid crystalline deposits (B, C) that were birefringent on polarized light (not shown).
291
Practical Surgical Neuropathology

A B

C D
Fig. 13.38  (A–D) IgG4-associated pachymeningitis. Postcontrast MR images showed both diffuse dural thickening and meningioma-like tumor masses (A). On histology, there is
a lymphoplasmacytic infiltrate as well as meningothelial hyperplasia (upper half), which does not invade the underlying dura (B). Germinal centers and numerous plasma cells were
evident (C), with many of the latter staining for IgG4 (D).

occasionally enters the differential. Renal cell carcinomas are more (“angiomatous meningiomas”). However, the clinicopathologic features
likely to be positive for carbonic anhydrase IX (CA IX), cytokeratin, of these two tumor types differ greatly (see Chapter 14) and those with
CD10, and RCC and negative for progesterone receptor than clear cell otherwise classic features of meningioma should be diagnosed as such.
meningiomas.61,116,117 Most cases of SFT/HPC do not express EMA or show diffuse SSTR2a
Papillary meningioma should be distinguished from other papillary positivity, although occasional examples are encountered with focal
neoplasms, including ependymoma, choroid plexus tumors, and astro- staining of these markers.119 In contrast, only the SFT/HPCs feature
blastoma (see Fig. 13.26). Strong GFAP positivity should aid in establish- nuclear STAT6 positivity,120 often in addition to extensive positivity for
ing a diagnosis of ependymoma or astroblastoma. Choroid plexus tumors both CD99 and bcl-2. Lastly, these tumors are associated with NAB2-
are strongly cytokeratin reactive. Rhabdoid meningioma (see Fig. 13.27) STAT6 fusions,120,121 rather than the typical genetic features of menin-
may look similar to other rhabdoid neoplasms; however, atypical teratoid/ giomas (see Genetics section).
rhabdoid tumors (AT/RTs) are almost exclusively encountered in infants,
whereas rhabdoid meningiomas mostly afflict adults. The latter also do Ancillary Diagnostic Studies
not inactivate the INI1/hSNF5 (SMARCB1) gene, and, therefore, nuclear Immunohistochemistry
protein expression is retained in rhabdoid meningiomas and lost in Immunohistochemistry supports the dual mesenchymal and epithelial-
AT/RTs118; in contrast, a subset of rhabdoid meningiomas lose BAP1 like features of meningothelial cells. The vast majority of meningiomas are
expression.88 strongly positive for vimentin (see Figs. 13.24C, 13.25F, 13.26F, 13.27C,
Occasionally, anaplastic meningiomas develop branching, thin-walled 13.31B, 13.32C, 13.35D) and show patchy, often modest EMA expres-
gaping blood vessels, similar to those of solitary fibrous tumor/ sion, the latter representing the most commonly utilized antibody for
hemangiopericytoma (SFT/HPC; Fig. 13.39). In the past, such cases supporting a meningioma diagnosis (see Figs. 13.12D, 13.13D, 13.15C,
were used as evidence that these two tumor types were related 13.18D, 13.21G, 13.22E, 13.24D, 13.30B). Nevertheless, it has long been

292
Meningiomas
an independent prognostic variable has been debated, since it usually
increases proportionally to both the mitotic index and the histologic
grade in general.124,125 Another difficulty comes from interlaboratory
13
variability in staining and counting methods, making it difficult to
extrapolate cutoff values from one lab to the next. Nonetheless, the
Ki-67 LI is particularly useful in borderline cases falling between either
benign and atypical or atypical and anaplastic meningioma; it is mainly
in these borderline cases that Ki-67 and PR studies add value. Those
with higher than expected LI may behave more aggressively and those
with lower than expected LI more indolently. Although it is not formally
used in histologic grading, this information may nonetheless be useful
in guiding patient management. For instance, a patient with a “benign
meningioma with increased proliferative index, WHO grade I” may need
to be followed more carefully for potential tumor recurrence, especially
if subtotally resected. Of interest (yet perhaps not surprisingly), one
study suggests that very focal elevations in proliferative index may not
A be as biologically relevant as more diffuse ones.126

Hormone Studies
The study of sex hormones and their receptors in meningiomas has
been of great interest for many years, particularly given the well-known
female patient predominance, reports of rapidly worsening visual
symptoms during pregnancy, and modest epidemiologic links between
the incidence of meningioma and breast carcinoma, another hormone-
associated tumor of women.127,128 In terms of enhanced pregnancy-
associated growth, this is probably more the exception than the rule.
Nonetheless, these rare examples can be clinically dramatic with
symptoms presenting typically in the third trimester most often from
optic nerve compression; of interest, the symptoms often resolve
postpartum and recur in subsequent pregnancies.129–132 Based on this
unusual behavior and a general lack of increased mitotic figures on
histology, the “growth” is thought to primarily result from pregnancy-
associated vascular congestion and cytotoxic edema, rather than true
tumor cell proliferation due to hormone stimulation, a finding supported
B
by the increased vascularity, hemorrhage, and intracellular/extracellular
Fig. 13.39  (A, B) Anaplastic meningioma. Focal hemangiopericytoma-like vascular pattern edema found histologically in some of the resected cases.131 Nevertheless,
in an anaplastic meningioma (A). Other regions showed classic features of meningioma (B). reports of PR expression in 50% to 80% of meningiomas (see Figs.
13.22F, 13.29D, 13.30C, 13.34D) have generated great interest in both
the clinical and scientific (not to mention medicolegal) communities.
appreciated that EMA suffers from insufficient sensitivity and specificity. In comparison, estrogen receptor expression is relatively rare and, typically,
As such, another marker, somatostatin receptor 2a (SSTR2a), has been low level.133
reported as a more sensitive and relatively specific marker (see Figs. Degrees of PR expression are highly variable, but they are roughly
13.14D, 13.17D, 13.18E, 13.21H, 13.34C), an exception being its strong inversely proportional to grade, such that the majority of benign menin-
immunoreactivity in neuroendocrine tumors.106 Cytokeratin is usually giomas and even reactive meningothelial proliferations are strongly
negative or only focally positive, with the exception of secretory menin- immunoreactive. A patchy or mosaic pattern of staining is also common
gioma, which strongly expresses cytokeratin in the cells surrounding the and the significance of this intratumoral heterogeneity remains unclear
pseudopsammoma bodies (see Fig. 13.16E). The latter structures (and (Fig. 13.40A and B). In contrast, most atypical, anaplastic, and other
sometimes the cells surrounding them) are also immunoreactive for clinically aggressive meningiomas are completely negative or show only
CEA (Fig. 13.16F), CA 19-9, alpha-1-antitrypsin, and immunoglobulins, focal staining (see Fig. 13.20F).123,134,135 Unfortunately, there remains a
which are typically negative in other types of meningioma.122 Variable great overlap in PR status among WHO grades and clinical subsets.
S-100 protein expression is seen most commonly in the fibrous subtype. Whether or not PR status is an independent prognosticator remains
Except in very rare cases (most often rhabdoid meningiomas), GFAP uncertain. Therefore, its routine use is not warranted. Nonetheless, it
is negative; however, this stain is useful for highlighting foci of brain may be particularly useful in borderline atypical cases, especially when
invasion in equivocal cases (see Fig. 13.23C). Additional meningothelial utilized in combination with MIB-1 (Ki-67) labeling indices. In other
markers that may be useful and are commonly positive include cathepsin words, “loss of expression” is worrisome for more aggressive behavior.
D, E-cadherin, claudin-1, and platelet-derived growth factor receptor The precise biologic role of PR also remains unclear. Arguing in
beta.123 Unfortunately, most of these markers are not terribly specific. favor of a tumorigenic or growth stimulatory function is the female
Other particularly useful ancillary markers for grading and prognosis predominance of meningiomas and epidemiologic data suggesting a
include progesterone receptor (PR; see Hormone Studies section) and minimally increased risk of meningioma for women using oral contracep-
Ki-67 (see Figs. 13.20E, 13.24E, 13.26D). As with many other tumor tives or hormone replacement therapy.1 Against a critical role, however,
types, Ki-67 labeling provides additional information regarding the is the common presence of PR in the meningiomas of men and children,
proliferation index. Whether or not this labeling index (LI) represents a lack of compelling growth stimulation by progesterone in vitro, and

293
Practical Surgical Neuropathology
Like normal arachnoidal cap cells, meningioma cells are interconnected
by intercellular junctions, including desmosomes, hemidesmosomes,
and gap junctions. The abundance of 10-nm intermediate filaments is
usually due to abundant vimentin expression. The intermediate filaments
tend to converge on the cytoplasmic side of desmosomes or form a
tight whorling pattern in the cytoplasm, most commonly in the rhabdoid
variant. As with routine light microscopy, nuclear pseudoinclusions are
often prominent and represent cytoplasmic invaginations. In contrast,
the clear intranuclear vacuoles occasionally contain glycogen particles,
but most often represent empty nonmembrane bound spaces.142 The
presence of classic meningothelial features by EM is particularly useful
in establishing the lineage in poorly differentiated meningiomas, such
as the anaplastic and papillary subtypes, each of which often generates
extensive differential diagnostic considerations.27,143 Lastly, “metaplastic
meningiomas” typically do not show evidence of true metaplasia, but
rather retain meningothelial features; they simply accumulate cytoplasmic
A lipid, lysosomes, and more.54
In addition to the nearly universal ultrastructural features described,
some meningioma variants may show distinctive features. For instance,
spider-like cytoplasmic processes line the intercellular spaces of microcystic
meningiomas. There are also scattered desmosomes, and the widened
extracellular spaces are filled with finely granular material, reminiscent
of the EM findings in normal arachnoidal trabecular cells.45 Despite
the presence of xanthoma-like cells by light microscopy, they typically
do not contain lipid droplets. The pseudopsammoma bodies of secretory
meningiomas are seen within gland-like spaces lined by microvilli.144 The
inclusions may appear amorphous, particulate, vesicular, or lamellar.
The cells surrounding the inclusions appear epithelial-like with electron
dense cytoplasm, abundant filaments, and numerous desmosomes. The
glycogen-rich cytoplasm of clear cell meningiomas is easily confirmed
by EM, and occasional amianthoid-type collagen fibers may be encoun-
tered.60 Rhabdoid meningiomas have the classic whorled bundles of
intermediate filaments, although entrapped organelles are also common
B within these paranuclear inclusions.85 As the name implies, oncocytic
meningiomas have cytoplasm stuffed with mitochondria,90 whereas the
Fig. 13.40  (A, B) Progesterone receptor (PR) immunohistochemistry. Meningiomas that even rarer granular subtypes have lysosomal accumulations93 or other
are positive for PR often display a mosaic or heterogeneous pattern of immunoreactivity membrane bound inclusions.92
with clusters of strongly positive nuclei alternating with negative ones.
Genetics
A detailed discussion of meningioma genetics is beyond the scope of
a peak incidence in women at roughly 60 years of age (i.e., beyond this book, and the interested reader is referred to recent reviews of this
childbearing years or periods of life when circulating progesterone levels topic.75,145 Nonetheless, much has been learned over the last few decades
are normally high). Despite these observations, it was hoped that therapy and a putative model of molecular tumorigenesis and malignant progres-
with antiprogestational agents such as mifepristone (RU-486) might be sion of meningiomas is illustrated in Fig. 13.41.
useful as an ancillary therapy for unresectable meningiomas; unfortu-
nately, this has not borne out clinically, perhaps because the most biologi- Cytogenetics
cally aggressive meningiomas are the ones least likely to express PR.136 Consistent cytogenetic abnormalities are found in most meningiomas
Additional hormone receptors detected in meningiomas include the and mostly involve common chromosomal losses that result in a progres-
androgen, prolactin, growth hormone, thyroid hormone, and cholecys- sive state of hypodiploidy.146,147 The most frequent chromosomal
tokinin receptors.137–141 The potential biologic and therapeutic roles for abnormality is monosomy 22 or, less often, a partial 22q deletion; these
these receptors have yet to be fully elucidated. changes are encountered in about 40% to 70% of cases. Following
monosomy 22, losses involving chromosomes 6, 10, 14, and 18 are most
Electron Microscopy common and often seen in higher grade tumors. In terms of structural
Electron microscopy (EM) has mostly been replaced by immunohis- alterations, loss of chromosome 1p is by far the most common, again
tochemistry (see earlier discussion) for the diagnosis of meningioma being associated with more aggressive meningiomas. In general, the
and its variants. Nevertheless, classic ultrastructural features of menin- more aggressive and higher grade the meningioma, the more abnormal
gioma, regardless of subtype, include complex intercellular junctions, the karyotype becomes.
interdigitating cell processes, and abundant intracellular filaments. The
syncytium-like pattern of meningothelial meningiomas seen at the light Molecular Genetics
microscopic level is due to an abundance of interdigitating cell processes, Molecular genetic and molecular cytogenetic techniques, such as loss of
creating fuzzy rather than sharp intercellular borders. Ultrastructurally, heterozygosity (LOH) or fluorescence in situ hybridization (FISH), have
they have been likened to the interlocking pieces of a jigsaw puzzle. also demonstrated frequent 22q losses in benign meningiomas, with

294
Meningiomas

Meningothelial Hyperplasia Arachnoid Cap Cells


13

Gain of PR expression (65%)

Benign Meningioma Atypical Meningioma Anaplastic Meningioma

Chromosome 22q loss (40–70%) TERT promoter mutation (5–10%) TERT promoter mutation (20–30%)
NF2 mutations (30–60%) SMARCE1 mutation (clear cell subtype) Chromosome 1p loss (70–100%)
TRAF7 and KLF4 mutations (10–25%; Chromosome 1p loss (40–75%) Chromosome 6q loss (50%)
Chromosome 6q loss (30%) 9p21 (CDKN2A/B, p14ARF) loss (60–80%)
skull base and secretory subtype)
Chromosome 10 loss (30–40%) Chromosome 10 loss (40–70%)
AKT1 mutations (10%; skull base)
Chromosome 14q loss (40–60%) Chromosome 14q loss (60–100%)
SMO mutations (5%; skull base)
Chromosome 18q loss (40%) Chromosome 18q loss (60–70%)
PIK3CA mutations (5%; skull base)
Gains of 1q, 9q, 12q, 15q, 17q, 20q NDRG2 hypermethylation (70%)
SMARCB1 mutations (rare;
(30–50% each) Loss of TSLC1 expression (70%)
schwannomatosis)
Loss of TSLC1 expression (70%)
SUFU or PTCH1 mutations (rare; Gorlin Loss of PR expression (80–90%)
Loss of PR expression (60–80%)
/nevoid basal cell carcinoma syndrome) 17q23 amplification (RPS6KB1, others)
Activation of Notch, WNT, IGF, or
Gain of PR expression (50–90%)
VEGF pathways
Loss of TSLC1 expression (30–50%)
EGFR activation
PDGFRB activation

Fig. 13.41  Genetic model of meningioma tumorigenesis and malignant progression (modified from WHO 2016 scheme). Pathways that are almost certain are shown with solid
arrows, whereas those that are less certain are displayed using dashed arrows. The most common alterations and their frequencies are shown at each stage of development.

deletions of other chromosomal regions as meningiomas progress to base.150,151 This suggests that many meningothelial meningiomas could
WHO grades II and III.1 As such, this provides a potential ancillary test arise from alternate (i.e., non-NF2 associated) tumorigenic pathways.
to support a suspicion of meningioma, as opposed to a meningothelial Recently, a number of other driver mutations have also been identified,
hyperplasia or nonmeningothelial neoplasm (see Fig. 13.24F). Deletions mostly in skull base meningiomas lacking NF2 mutations. Notably,
of 1p, 10, and 14q are especially frequent in the high-grade meningiomas, mutations of TRAF7 and KLF4 are nearly always combined, are present
potentially providing ancillary support for the likelihood of aggressive in nearly all of the secretory meningiomas, and are found in roughly
behavior in a known meningioma or for the diagnosis of high-grade 10% to 25% of meningiomas overall.1,51,152 AKT1, SMO, and PIK3CA
meningioma versus other malignant neoplasms (Fig. 13.24G). Of interest, mutations are each detected in about 5% to 10% of meningiomas and
such deletions in recurrent high-grade meningiomas are often already are of additional interest due to the possible use of targeted therapies
detectable in the original tumors at a time when they may have appeared for patients with mutant-specific meningiomas that have failed con-
otherwise benign.148 In fact, a comparison of benign meningiomas with ventional surgical and radiation approaches.1,152–155 SMARCB1, SUFU,
and without subsequent recurrences revealed a statistically increased and PTCH1 mutations are each rare, but associated with familial tumor
likelihood of 14q deletions.149 predisposition syndromes such as schwannomatosis and Gorlin (nevoid
These common chromosomal losses strongly suggest the presence basal cell carcinoma) syndromes (see Chapter 22).145,156–158
of meningioma tumor suppressor genes. The most studied has been For most of the progression-associated losses, the relevant tumor
chromosome 22q, with the NF2 gene being well established as the main suppressor genes have yet to be identified (see Fig. 13.41). However,
target of inactivation for both sporadic and NF2-associated meningiomas.1 an important association has been found with promoter region mutations
Genetic studies suggest that other chromosome 22q loci may also play of the TERT gene (same mutations as seen in glioblastomas and oligo-
a role in rare examples. In atypical and anaplastic meningiomas, NF2 dendrogliomas) and aggressive clinical behavior.159–161 As such, testing
gene mutations occur in approximately 70% of cases, consistent with a may be useful in grade II and III meningiomas, as well as clinically
role for NF2 inactivation in the formation, rather than progression, of worrisome grade I examples. Additionally, mutations of SMARCE1
meningiomas. Interestingly, the frequency of NF2 gene mutations varies (another member of the SWI/SNF chromatin remodeling complex with
somewhat among the three most common variants of meningioma. The SMARCB1) have been identified in both familial and sporadic clear
fibroblastic and transitional meningiomas have detectable NF2 gene muta- cell meningiomas.65,66,145 Investigations of known tumor suppressor genes
tions in roughly 70% to 80%, whereas they are found in only about 25% of have revealed only rare alterations in meningiomas. Of interest, however,
meningothelial meningiomas, particularly those encountered at the skull losses of the CDKN2A/p16 region on 9p21 are seen in the majority of

295
Practical Surgical Neuropathology
anaplastic meningiomas (see Fig. 13.24H), where they are known to be 2. Cushing H, Eisenhardt L. Meningiomas: Their Classification, Regional Behaviour, Life History,
associated with poor survival.28,162 The alternate (“optimist”) viewpoint and Surgical End Results. Springfield, IL: CC Thomas; 1938.
3. Phillips LE, Koepsell TD, van Belle G, et al. History of head trauma and risk of intracranial
is that the roughly one-third of patients whose anaplastic meningiomas meningioma: population-based case-control study. Neurology. 2002;58:1849–1852.
lack 9p21 deletions have surprisingly long survival times, with tumor 4. Francois P, N’Dri D, Bergemer-Fouquet AM, et al. Post-traumatic meningioma: three case
biology more closely resembling that of atypical meningioma. For this reports of this rare condition and a review of the literature. Acta Neurochir (Wien). 2010;152:
reason, there may be prognostic value in the analysis of 9p21 by FISH 1755–1760.
5. Inskip PD, Mellemkjaer L, Gridley G, Olsen JH. Incidence of intracranial tumors following
in anaplastic meningiomas.
hospitalization for head injuries (Denmark). Cancer Causes Control. 1998;9:109–116.
6. Bondy M, Ligon BL. Epidemiology and etiology of intracranial meningiomas: a review. J Neurooncol.
Treatment and Prognosis 1996;29:197–205.
Given the slow growth and indolent behavior of most meningiomas, 7. Frei P, Poulsen AH, Johansen C, et al. Use of mobile phones and risk of brain tumours: update
a “watch and wait” approach is commonly utilized for many patients, of Danish cohort study. BMJ. 2011;343:d6387.
8. Swerdlow AJ, Feychting M, Green AC, et al. Mobile phones, brain tumors, and the interphone
especially the elderly and those who had their meningioma identified as study: where are we now? Environ Health Perspect. 2011;119:1534–1538.
an incidental finding on neuroimaging. For patients with symptomatic 9. Mathews JD, Forsythe AV, Brady Z, et al. Cancer risk in 680,000 people exposed to computed
disease or evidence of tumor growth on serial imaging, however, definitive tomography scans in childhood or adolescence: data linkage study of 11 million Australians.
therapy is often necessary. Surgery is the standard and most effective BMJ. 2013;346:f2360.
10. Claus EB, Calvocoressi L, Bondy ML, et al. Dental x-rays and risk of meningioma. Cancer.
therapy for meningiomas, although radiation is also a major treatment
2012;118:4530–4537.
modality, particularly for patients where surgery carries high risk.25,163 11. Pearce MS, Salotti JA, Little MP, et al. Radiation exposure from CT scans in childhood and
Adjuvant radiotherapy has been shown to significantly reduce recurrence subsequent risk of leukaemia and brain tumours: a retrospective cohort study. Lancet.
rates in subtotally resected or higher grade meningiomas. Additionally, 2012;380:499–505.
newer forms of delivery have become increasingly popular, especially for 12. Wiemels JL, Wrensch M, Sison JD, et al. Reduced allergy and immunoglobulin E among adults
with intracranial meningioma compared to controls. Int J Cancer. 2011;129:1932–1939.
smaller tumors or postsurgical residual masses. These include stereotactic 13. Kepes J. Meningiomas: Biology, Pathology, and Differential Diagnosis. New York: Masson; 1982.
radiosurgery (e.g., “gamma knife”), as well as fractionated stereotactic, 14. Ostrom QT, Gittleman H, Fulop J, et al. CBTRUS Statistical Report: Primary Brain and Central
intensity modulated, and proton beam forms of radiotherapy. As with Nervous System Tumors Diagnosed in the United States in 2008-2012. Neuro Oncol. 2015;17
any form of radiation therapy, however, the potential benefits must Suppl 4:iv1–iv62.
15. Nakasu S, Hirano A, Shimura T, Llena JF. Incidental meningiomas in autopsy study. Surg Neurol.
be weighed against the low but significant risks of side effects, such
1987;319–322.
as radiation-induced secondary neoplasms and leukoencephalopathy- 16. Perry A, Giannini C, Raghavan R, et al. Aggressive phenotypic and genotypic features in pediatric
associated cognitive decline, particularly in younger patients with long and NF2-associated meningiomas: a clinicopathologic study of 53 cases. J Neuropathol Exp
life expectancies (see Chapter 21). Neurol. 2001;60:994–1003.
Using the 2016 WHO scheme (see Table 13.1), roughly 75% to 80% 17. Ravindranath K, Vasudevan MC, Pande A, Symss N. Management of pediatric intracranial
meningiomas: an analysis of 31 cases and review of literature. Childs Nerv Syst. 2013;29:
of meningiomas are benign (WHO grade I), 15% to 25% are WHO grade 573–582.
II, and 1% to 3% are malignant (WHO grade III). Gross totally resected 18. Thuijs NB, Uitdehaag BM, Van Ouwerkerk WJ, et al. Pediatric meningiomas in The Netherlands
WHO grade I meningiomas are appropriately designated “benign,” since 1974-2010: a descriptive epidemiological case study. Childs Nerv Syst. 2012;28:1009–1015.
they show no increased mortality when compared with age- and sex- 19. Stafford SL, Perry A, Leavitt JA, et al. Anterior visual pathway meningiomas primarily resected
between 1978 and 1988: the Mayo Clinic Rochester experience. J Neuroophthalmol. 1998;18:
matched controls, but have a roughly 5% recurrence rate at 5 years, which
206–210.
continues to increase steadily with longer follow-up times.24,27 In subtotally 20. Ho CY, Mosier S, Safneck J, et al. Genetic profiling by single-nucleotide polymorphism-based
resected cases, this risk of recurrence increases considerably (~30% to array analysis defines three distinct subtypes of orbital meningioma. Brain Pathol. 2015;25:
40% at 5 years), with extent of resection itself being highly linked to 193–201.
tumor location, such that meningiomas in surgically challenging regions 21. Eddleman CS, Liu JK. Optic nerve sheath meningioma: current diagnosis and treatment. Neurosurg
Focus. 2007;23:E4.
(e.g., skull base) are more likely to be limited to subtotal resection. The 22. Lang FF, Macdonald OK, Fuller GN, DeMonte F. Primary extradural meningiomas: a report on
high-grade variants are associated with statistically significant increased nine cases and review of the literature from the era of computerized tomography scanning. J
risks of both recurrence and death, although in general, the WHO grade Neurosurg. 2000;93:940–950.
II meningiomas often recur even with gross total resection, while the 23. Weissferdt A, Tang X, Suster S, et al. Pleuropulmonary Meningothelial Proliferations: Evidence
for a Common Histogenesis. Am J Surg Pathol. 2015;39:1673–1678.
WHO grade III tumors have high mortality rates.
24. Perry A, Stafford SL, Scheithauer BW, et al. Meningioma grading: an analysis of histologic
Meningiomas that can no longer be treated with any additional parameters. Am J Surg Pathol. 1997;21:1455–1465.
surgery or radiation constitute a considerable therapeutic challenge. 25. Sun SQ, Hawasli AH, Huang J, et al. An evidence-based treatment algorithm for the management
Standard chemotherapy and hormonal therapy approaches have been of WHO Grade II and III meningiomas. Neurosurg Focus. 2015;38:E3.
disappointing to date, although potential strategies currently being 26. Jenkinson MD, Javadpour M, Haylock BJ, et al. The ROAM/EORTC-1308 trial: Radiation versus
Observation following surgical resection of Atypical Meningioma: study protocol for a randomised
explored in early stages include targeted therapies and immunother- controlled trial. Trials. 2015;16:519.
apy.155,164–169 Additional experience in large clinical trials will be needed 27. Perry A, Scheithauer BW, Stafford SL, et al. “Malignancy” in meningiomas: a clinicopathologic
to further assess their efficacies. study of 116 patients, with grading implications. Cancer. 1999;85:2046–2056.
28. Perry A, Banerjee R, Lohse CM, et al. A role for chromosome 9p21 deletions in the malignant
Suggested Readings progression of meningiomas and the prognosis of anaplastic meningiomas. Brain Pathol.
2002;12:183–190.
Perry A, Louis DN, Budka H, et al. Meningioma. In: Louis DN, Ohgaki H, Wiestler OD, et al., eds.
29. Chaparro MJ, Young RF, Smith M, et al. Multiple spinal meningiomas: a case of 47 distinct lesions
WHO Classification of Tumours of the Central Nervous System. Revised 4th ed. Lyon: IARC;
in the absence of neurofibromatosis or identified chromosomal abnormality. Neurosurgery.
2016:232–245 [Chapter 10].
1993;32:298–301, discussion 301–302.
Perry A, Scheithauer BW, Stafford SL, et al. “Malignancy” in meningiomas: a clinicopathologic study
30. Stangl AP, Wellenreuther R, Lenartz D, et al. Clonality of multiple meningiomas. J Neurosurg.
of 116 patients, with grading implications. Cancer. 1999;85:2046–2056.
1997;86:853–858.
Rogers CL, Perry A, Pugh S, et al. Pathology concordance levels for meningioma classification and
31. Borovich B, Doron Y. Recurrence of intracranial meningiomas: the role played by regional
grading in NRG Oncology RTOG Trial 0539. Neuro Oncol. 2016;18:565–574.
multicentricity. J Neurosurg. 1986;64:58–63.
32. Engelhard HH. Progress in the diagnosis and treatment of patients with meningiomas. Part I:
References diagnostic imaging, preoperative embolization. Surg Neurol. 2001;55:89–101.
1. Perry A, Louis DN, Budka H, et al. Meningioma. In: Louis DN, Ohgaki H, Wiestler OD, et al., 33. Filippi CG, Edgar MA, Ulug AM, et al. Appearance of meningiomas on diffusion-weighted
eds. WHO Classification of Tumours of the Central Nervous System. Revised 4th ed. Lyon: images: correlating diffusion constants with histopathologic findings. AJNR Am J Neuroradiol.
IARC; 2016:232–245 [Chapter 10]. 2001;22:65–72.

296
Meningiomas
34. Hakyemez B, Yildirim N, Gokalp G, et al. The contribution of diffusion-weighted MR imaging 68. Kozler P, Benes V, Netuka D, et al. Chordoid meningioma: presentation of two case reports,
to distinguishing typical from atypical meningiomas. Neuroradiology. 2006;48:513–520.
35. Zhang H, Rodiger LA, Shen T, et al. Perfusion MR imaging for differentiation of benign and
review of the literature, and plea for data standardisation. J Neurooncol. 2008;88:115–120.
69. Couce ME, Aker FV, Scheithauer BW. Chordoid meningioma: a clinicopathologic study of 42 13
malignant meningiomas. Neuroradiology. 2008;50:525–530. cases. Am J Surg Pathol. 2000;24:899–905.
36. Weber J, Gassel AM, Hoch A, et al. Intraoperative management of cystic meningiomas. Neurosurg 70. Kepes JJ, Chen WY, Connors MH, Vogel FS. “Chordoid” meningeal tumors in young individuals
Rev. 2003;26:62–66. with peritumoral lymphoplasmacellular infiltrates causing systemic manifestations of the Castleman
37. Louis DN, Ohgaki H, Wiestler OD, et al. WHO Classification of Tumours of the Central Nervous syndrome. A report of seven cases. Cancer. 1988;62:391–406.
System. Revised 4th ed. Lyon, France: IARC; 2016. 71. Epari S, Sharma MC, Sarkar C, et al. Chordoid meningioma, an uncommon variant of meningioma:
38. Trembath D, Miller CR, Perry A. Gray zones in brain tumor classification: evolving concepts. a clinicopathologic study of 12 cases. J Neurooncol. 2006;78:263–269.
Adv Anat Pathol. 2008;15:287–297. 72. Choy W, Ampie L, Lamano JB, et al. Predictors of recurrence in the management of chordoid
39. Vaubel RA, Chen SG, Raleigh DR, et al. Meningiomas With Rhabdoid Features Lacking Other meningioma. J Neurooncol. 2016;126:107–116.
Histologic Features of Malignancy: A Study of 44 Cases and Review of the Literature. J Neuropathol 73. Mullassery D, O’Brien DF, Williams D, et al. Malignant disseminated chordoid meningioma
Exp Neurol. 2016;75:44–52. in a 12-year-old child: a role for early cranial and spinal radiation treatment after subtotal
40. Hasselblatt M, Nolte KW, Paulus W. Angiomatous meningioma: a clinicopathologic study of resection. Childs Nerv Syst. 2006;22:1344–1350.
38 cases. Am J Surg Pathol. 2004;28:390–393. 74. Baumgarten P, Gessler F, Schittenhelm J, et al. Brain invasion in otherwise benign meningiomas
41. Tamiya T, Ono Y, Matsumoto K, Ohmoto T. Peritumoral brain edema in intracranial meningiomas: does not predict tumor recurrence. Acta Neuropathol. 2016;132:479–481.
effects of radiological and histological factors. Neurosurgery. 2001;49:1046–1051, discussion 75. Riemenschneider MJ, Perry A, Reifenberger G. Histological classification and molecular genetics
51–52. of meningiomas. Lancet Neurol. 2006;5:1045–1054.
42. Abedalthagafi MS, Merrill PH, Bi WL, et al. Angiomatous meningiomas have a distinct genetic 76. Lohmann CM, Brat DJ. A conceptual shift in the grading of meningiomas. Adv Anat Pathol.
profile with multiple chromosomal polysomies including polysomy of chromosome 5. Oncotarget. 2000;7:153–157.
2014;5:10596–10606. 77. Lach B, Benoit BG. Myofibroblastic sarcoma in meningioma: a new variant of “metaplastic”
43. Chen CJ, Tseng YC, Hsu HL, Jung SM. Microcystic meningioma: importance of obvious hypointensity meningioma. Ultrastruct Pathol. 2007;31:357–363.
on T1-weighted magnetic resonance images. J Comput Assist Tomogr. 2008;32:130–134. 78. Fudaba H, Abe T, Morishige M, et al. Dedifferentiated chordoid meningioma with rhabdomyo-
44. Paek SH, Kim SH, Chang KH, et al. Microcystic meningiomas: radiological characteristics of 16 sarcomatous differentiation on the middle cranial fossa. Neuropathology. 2016;36:579–583.
cases. Acta Neurochir (Wien). 2005;147:965–972, discussion 972. 79. Patil S, Scheithauer BW, Strom RG, et al. Malignant meningiomas with epithelial (adenocarcinoma-
45. Nishio S, Takeshita I, Fukui M. Microcystic meningioma: tumors of arachnoid cap vs trabecular like) metaplasia: a study of 3 cases. Neurosurgery. 2011;69:884–892.
cells. Clin Neuropathol. 1994;13:197–203. 80. Ludwin SK, Rubinstein LJ, Russell DS. Papillary meningioma: a malignant variant of meningioma.
46. Tirakotai W, Mennel HD, Celik I, et al. Secretory meningioma: immunohistochemical findings Cancer. 1975;36:1363–1373.
and evaluation of mast cell infiltration. Neurosurg Rev. 2006;29:41–48. 81. Wang XQ, Chen H, Zhao L, et al. Intracranial papillary meningioma: a clinicopathologic study
47. Buhl R, Hugo HH, Mehdorn HM. Brain oedema in secretory meningiomas. J Clin Neurosci. of 30 cases at a single institution. Neurosurgery. 2013;73:777–790, discussion 789.
2001;8(suppl 1):19–21. 82. Pasquier B, Gasnier F, Pasquier D, et al. Papillary meningioma. Clinicopathologic study of seven
48. Caffo M, Caruso G, Germano A, et al. CD68 and CR3/43 immunohistochemical expression in cases and review of the literature. Cancer. 1986;58:299–305.
secretory meningiomas. Neurosurgery. 2005;57:551–557, discussion 551–557. 83. Wu YT, Ho JT, Lin YJ, Lin JW. Rhabdoid papillary meningioma: a clinicopathologic case series
49. Colakoglu N, Demirtas E, Oktar N, et al. Secretory meningiomas. J Neurooncol. 2003;62: study. Neuropathology. 2011;31:599–605.
233–241. 84. Kepes JJ, Moral LA, Wilkinson SB, et al. Rhabdoid transformation of tumor cells in meningiomas:
50. Caffo M, Caruso G, Galatioto S, et al. Immunohistochemical study of the extracellular matrix a histologic indication of increased proliferative activity: report of four cases. Am J Surg Pathol.
proteins laminin, fibronectin and type IV collagen in secretory meningiomas. J Clin Neurosci. 1998;22:231–238.
2008;15:806–811. 85. Perry A, Scheithauer BW, Stafford SL, et al. “Rhabdoid” meningioma: an aggressive variant.
51. Reuss DE, Piro RM, Jones DT, et al. Secretory meningiomas are defined by combined KLF4 Am J Surg Pathol. 1998;22:1482–1490.
K409Q and TRAF7 mutations. Acta Neuropathol. 2013;125:351–358. 86. Nozza P, Raso A, Rossi A, et al. Rhabdoid meningioma of the tentorium with expression of
52. Lal A, Dahiya S, Gonzales M, et al. IgG4 overexpression is rare in meningiomas with a prominent desmin in a 12-year-old Turner syndrome patient. Acta Neuropathol (Berl). 2005;110:205–206.
inflammatory component: a review of 16 cases. Brain Pathol. 2014;24:352–359. 87. Al-Habib A, Lach B, Al Khani A. Intracerebral rhabdoid and papillary meningioma with
53. Zhu HD, Xie Q, Gong Y, et al. Lymphoplasmacyte-rich meningioma: our experience with 19 leptomeningeal spread and rapid clinical progression. Clin Neuropathol. 2005;24:1–7.
cases and a systematic literature review. Int J Clin Exp Med. 2013;6:504–515. 88. Shankar GM, Abedalthagafi M, Vaubel RA, et al. Germline and somatic BAP1 mutations in
54. Roncaroli F, Scheithauer BW, Laeng RH, et al. Lipomatous meningioma: a clinicopathologic high-grade rhabdoid meningiomas. Neuro Oncol. 2017;19:535–545.
study of 18 cases with special reference to the issue of metaplasia. Am J Surg Pathol. 2001;25: 89. Giannini C, Fratkin JD, Wyatt-Ashmead J, Aleff PC. Rhabdoid-like meningioma with inclusions
769–775. consisting of accumulations of complex interdigitating cell processes rather than intermediate
55. Krisht KM, Altay T, Couldwell WT. Myxoid meningioma: a rare metaplastic meningioma variant filaments. Acta Neuropathol. 2014;127:937–939.
in a patient presenting with intratumoral hemorrhage. J Neurosurg. 2012;116:861–865. 90. Roncaroli F, Riccioni L, Cerati M, et al. Oncocytic meningioma. Am J Surg Pathol. 1997;21:375–382.
56. Siraj F, Ansari MK, Sharma KC, Singh A. Chordoid meningioma: a diagnostic dilemma. J Cancer 91. Gallina P, Buccoliero AM, Mariotti F, et al. Oncocytic meningiomas: cases with benign
Res Ther. 2015;11:663. histopathological features and a favorable clinical course. J Neurosurg. 2006;105:736–738.
57. Kane AJ, Sughrue ME, Rutkowski MJ, et al. Anatomic location is a risk factor for atypical and 92. Alexander RT, McLendon RE, Cummings TJ. Meningioma with eosinophilic granular inclusions.
malignant meningiomas. Cancer. 2011;117:1272–1278. Clin Neuropathol. 2004;23:292–297.
58. Willis J, Smith C, Ironside JW, et al. The accuracy of meningioma grading: a 10-year retrospective 93. Lach B, Kanaan I. Granular cell meningioma. A case report. Folia Neuropathol. 2007;45:19–22.
audit. Neuropathol Appl Neurobiol. 2005;31:141–149. 94. Haberler C, Jarius C, Lang S, et al. Fibrous meningeal tumours with extensive non-calcifying
59. Olar A, Wani KM, Sulman EP, et al. Mitotic Index is an Independent Predictor of Recurrence-Free collagenous whorls and glial fibrillary acidic protein expression: the whorling-sclerosing variant
Survival in Meningioma. Brain Pathol. 2015;25:266–275. of meningioma. Neuropathol Appl Neurobiol. 2002;28:42–47.
60. Pimentel J, Fernandes A, Pinto AE, et al. Clear cell meningioma variant and clinical aggressiveness. 95. Hope JK, Armstrong DA, Babyn PS, et al. Primary meningeal tumors in children: correlation
Clin Neuropathol. 1998;17:141–146. of clinical and CT findings with histologic type and prognosis. AJNR Am J Neuroradiol.
61. Zorludemir S, Scheithauer BW, Hirose T, et al. Clear cell meningioma. A clinicopathologic study 1992;13:1353–1364.
of a potentially aggressive variant of meningioma. Am J Surg Pathol. 1995;19:493–505. 96. Kim NR, Im SH, Chung CK, et al. Sclerosing meningioma: immunohistochemical analysis of
62. Im SH, Chung CK, Cho BK, et al. Sclerosing meningioma: clinicopathological study of four five cases. Neuropathol Appl Neurobiol. 2004;30:126–135.
cases. J Neurooncol. 2004;68:169–175. 97. Berho M, Suster S. Mucinous meningioma. Report of an unusual variant of meningioma that
63. Dhall SS, Tumialan LM, Brat DJ, Barrow DL. Spinal intradural clear cell meningioma following may mimic metastatic mucin-producing carcinoma. Am J Surg Pathol. 1994;18:100–106.
resection of a suprasellar clear cell meningioma. Case report and recommendations for 98. Liverman C, Mafra M, Chuang SS, et al. A clinicopathologic study of 11 rosette-forming
management. J Neurosurg. 2005;103:559–563. meningiomas: a rare and potentially confusing pattern. Acta Neuropathol. 2015;130:311–313.
64. Cassereau J, Lavigne C, Michalak-Provost S, et al. An intraventricular clear cell meningioma 99. Couce ME, Perry A, Webb P, et al. Fibrous meningioma with tyrosine-rich crystals. Ultrastruct
revealed by an inflammatory syndrome in a male adult: a case report. Clin Neurol Neurosurg. Pathol. 1999;23:341–345.
2008;110:743–746. 100. Miyajima Y, Oka H, Utsuki S, et al. Granulofilamentous meningioma. Brain Tumor Pathol.
65. Smith MJ, O’Sullivan J, Bhaskar SS, et al. Loss-of-function mutations in SMARCE1 cause an 2013;30:57–60.
inherited disorder of multiple spinal meningiomas. Nat Genet. 2013;45:295–298. 101. Goldman JE, Horoupian DS, Johnson AB. Granulofilamentous inclusions in a meningioma.
66. Smith MJ, Ahn S, Lee JI, et al. SMARCE1 mutation screening in classification of clear cell Cancer. 1980;46:156–161.
meningiomas. Histopathology. 2017;70:814–820. 102. Adlakha A, Rao K, Adlakha H, et al. Meningioma metastatic to the lung. Mayo Clin Proc.
67. Yang Y, Li D, Cao XY, et al. Clinical Features, Treatment, and Prognostic Factors of Chordoid 1999;74:1129–1133.
Meningioma: Radiological and Pathological Features in 60 Cases of Chordoid Meningioma. 103. Surov A, Gottschling S, Bolz J, et al. Distant metastases in meningioma: an underestimated
World Neurosurg. 2016;93:198–207. problem. J Neurooncol. 2013;112:323–327.

297
Practical Surgical Neuropathology
104. Takei H, Powell SZ. Tumor-to-tumor metastasis to the central nervous system. Neuropathology. 137. Ciccarelli E, Razzore P, Gaia D, et al. Hyperprolactinaemia and prolactine binding in benign
2009;29:303–308. intracranial tumours. J Neurosurg Sci. 2001;45:70–74.
105. Mawrin C, Schulz S, Hellwig-Patyk A, et al. Expression and function of somatostatin receptors 138. McCutcheon IE, Flyvbjerg A, Hill H, et al. Antitumor activity of the growth hormone
in peripheral nerve sheath tumors. J Neuropathol Exp Neurol. 2005;64:1080–1088. receptor antagonist pegvisomant against human meningiomas in nude mice. J Neurosurg.
106. Menke JR, Raleigh DR, Gown AM, et al. Somatostatin receptor 2a is a more sensitive diagnostic 2001;94:487–492.
marker of meningioma than epithelial membrane antigen. Acta Neuropathol. 2015;130:441–443. 139. Chen J, Chen G. Expression of androgen receptor in meningiomas. J Tongji Med Univ.
107. Ng J, Celebre A, Munoz DG, et al. Sox10 is superior to S100 in the diagnosis of meningioma. Appl 2001;21:140–142.
Immunohistochem Mol Morphol. 2015;23:215–219. 140. Oikonomou E, Machado AL, Buchfelder M, Adams EF. Meningiomas expressing and responding
108. Perry A, Lusis EA, Gutmann DH. Meningothelial hyperplasia: a detailed clinicopathologic, to cholecystokinin (CCK). J Neurooncol. 2005;73:199–204.
immunohistochemical and genetic study of 11 cases. Brain Pathol. 2005;15:109–115. 141. Wang CJ, Lin PC, Howng SL. Expression of thyroid hormone receptors in intracranial menin-
109. Bertoni F, Unni KK, Dahlin DC, et al. Calcifying pseudoneoplasms of the neural axis. J Neurosurg. giomas. Kaohsiung J Med Sci. 2003;19:334–338.
1990;72:42–48. 142. Yoshida T, Hirato J, Sasaki A, et al. Intranuclear inclusions of meningioma associated with
110. Qian J, Rubio A, Powers JM, et al. Fibro-osseous lesions of the central nervous system: report abnormal cytoskeletal protein expression. Brain Tumor Pathol. 1999;16:86–91.
of four cases and literature review. Am J Surg Pathol. 1999;23:1270–1275. 143. Al-Sarraj S, King A, Martin AJ, et al. Ultrastructural examination is essential for diagnosis of
111. Alshareef M, Vargas J, Welsh CT, Kalhorn SP. Calcifying Pseudoneoplasm of the Cervicomedullary papillary meningioma. Histopathology. 2001;38:318–324.
Junction: Case Report and a Literature Review. World Neurosurg. 2016;85(364):e11–e18. 144. Kepes JJ. The fine structure of hyaline inclusions (pseudopsammoma bodies) in meningiomas.
112. Smith DM, Berry AD 3rd. Unusual fibro-osseous lesion of the spinal cord with positive stain- J Neuropathol Exp Neurol. 1975;34:282–294.
ing for glial fibrillary acidic protein and radiological progression: a case report. Hum Pathol. 145. Smith MJ. Germline and somatic mutations in meningiomas. Cancer Genet. 2015;208:107–114.
1994;25:835–838. 146. Zang KD. Meningioma: a cytogenetic model of a complex benign human tumor, including data
113. Ribeiro da Cunha P, Peliz AJ, Barbosa M. Tophaceous gout of the lumbar spine mimicking a on 394 karyotyped cases. Cytogenet Cell Genet. 2001;93:207–220.
spinal meningioma. Eur Spine J. 2016. 147. Ketter R, Rahnenfuhrer J, Henn W, et al. Correspondence of tumor localization with tumor
114. Reis GF, Perry A. A 67-year-old man with a lumbar spine lesion. Brain Pathol. 2014;24: recurrence and cytogenetic progression in meningiomas. Neurosurgery. 2008;62:61–69, discussion
547–548. 69–70.
115. Sangoi AR, Dulai MS, Beck AH, et al. Distinguishing chordoid meningiomas from their histologic 148. Al-Mefty O, Kadri PA, Pravdenkova S, et al. Malignant progression in meningioma: documentation
mimics: an immunohistochemical evaluation. Am J Surg Pathol. 2009;33:669–681. of a series and analysis of cytogenetic findings. J Neurosurg. 2004;101:210–218.
116. Luong-Player A, Liu H, Wang HL, Lin F. Immunohistochemical reevaluation of carbonic anhydrase 149. Cai DX, Banerjee R, Scheithauer BW, et al. Chromosome 1p and 14q FISH analysis in clini-
IX (CA IX) expression in tumors and normal tissues. Am J Clin Pathol. 2014;141:219–225. copathologic subsets of meningioma: diagnostic and prognostic implications. J Neuropathol
117. Prayson RA, Chamberlain WA, Angelov L. Clear cell meningioma: a clinicopathologic study of 18 Exp Neurol. 2001;60:628–636.
tumors and examination of the use of CD10, CA9, and RCC antibodies to distinguish between 150. Wellenreuther R, Waha A, Vogel Y, et al. Quantitative analysis of neurofibromatosis type 2 gene
clear cell meningioma and metastatic clear cell renal cell carcinoma. Appl Immunohistochem transcripts in meningiomas supports the concept of distinct molecular variants. Lab Invest.
Mol Morphol. 2010;18:422–428. 1997;77:601–606.
118. Perry A, Fuller CE, Judkins AR, et al. INI1 expression is retained in composite rhabdoid tumors, 151. Kros J, de Greve K, van Tilborg A, et al. NF2 status of meningiomas is associated with tumour
including rhabdoid meningiomas. Mod Pathol. 2005;18:951–958. localization and histology. J Pathol. 2001;194:367–372.
119. Rajaram V, Brat DJ, Perry A. Anaplastic meningioma versus meningeal hemangiopericytoma: 152. Clark VE, Erson-Omay EZ, Serin A, et al. Genomic analysis of non-NF2 meningiomas reveals
immunohistochemical and genetic markers. Hum Pathol. 2004;35:1413–1418. mutations in TRAF7, KLF4, AKT1, and SMO. Science. 2013;339:1077–1080.
120. Schweizer L, Koelsche C, Sahm F, et al. Meningeal hemangiopericytoma and solitary fibrous 153. Brastianos PK, Horowitz PM, Santagata S, et al. Genomic sequencing of meningiomas identifies
tumors carry the NAB2-STAT6 fusion and can be diagnosed by nuclear expression of STAT6 oncogenic SMO and AKT1 mutations. Nat Genet. 2013;45:285–289.
protein. Acta Neuropathol. 2013;125:651–658. 154. Sahm F, Bissel J, Koelsche C, et al. AKT1E17K mutations cluster with meningothelial and
121. Fritchie KJ, Jin L, Rubin BP, et al. NAB2-STAT6 Gene Fusion in Meningeal Hemangiopericytoma transitional meningiomas and can be detected by SFRP1 immunohistochemistry. Acta Neuropathol.
and Solitary Fibrous Tumor. J Neuropathol Exp Neurol. 2016;75:263–271. 2013;126:757–762.
122. Probst-Cousin S, Villagran-Lillo R, Lahl R, et al. Secretory meningioma: clinical, histologic, and 155. Abedalthagafi M, Bi WL, Aizer AA, et al. Oncogenic PI3K mutations are as common as AKT1
immunohistochemical findings in 31 cases. Cancer. 1997;79:2003–2015. and SMO mutations in meningioma. Neuro Oncol. 2016;18:649–655.
123. Lusis EA, Chicoine MR, Perry A. High throughput screening of meningioma biomarkers using 156. Bacci C, Sestini R, Provenzano A, et al. Schwannomatosis associated with multiple meningiomas
a tissue microarray. J Neurooncol. 2005;73:219–223. due to a familial SMARCB1 mutation. Neurogenetics. 2010;11:73–80.
124. Perry A, Stafford SL, Scheithauer BW, et al. The prognostic significance of MIB-1, p53, and DNA 157. Aavikko M, Li SP, Saarinen S, et al. Loss of SUFU function in familial multiple meningioma. Am
flow cytometry in completely resected primary meningiomas. Cancer. 1998;82:2262–2269. J Hum Genet. 2012;91:520–526.
125. Roser F, Samii M, Ostertag H, Bellinzona M. The Ki-67 proliferation antigen in meningiomas. 158. Kijima C, Miyashita T, Suzuki M, et al. Two cases of nevoid basal cell carcinoma syndrome
Experience in 600 cases. Acta Neurochir (Wien). 2004;146:37–44, discussion 44. associated with meningioma caused by a PTCH1 or SUFU germline mutation. Fam Cancer.
126. Nakasu S, Li DH, Okabe H, et al. Significance of MIB-1 staining indices in meningiomas: 2012;11:565–570.
comparison of two counting methods. Am J Surg Pathol. 2001;25:472–478. 159. Sahm F, Schrimpf D, Olar A, et al. TERT Promoter Mutations and Risk of Recurrence in Meningioma.
127. Custer BS, Koepsell TD, Mueller BA. The association between breast carcinoma and meningioma J Natl Cancer Inst. 2016;108.
in women. Cancer. 2002;94:1626–1635. 160. Furtjes G, Kochling M, Peetz-Dienhart S, et al. hTERT promoter methylation in meningiomas
128. Lieu AS, Hwang SL, Howng SL. Intracranial meningioma and breast cancer. J Clin Neurosci. and central nervous hemangiopericytomas. J Neurooncol. 2016;130:79–87.
2003;10:553–556. 161. Goutagny S, Nault JC, Mallet M, et al. High incidence of activating TERT promoter mutations
129. Bickerstaff ER, Small JM, Guest IA. The relapsing course of certain meningiomas in relation to in meningiomas undergoing malignant progression. Brain Pathol. 2014;24:184–189.
pregnancy and menstruation. J Neurol Neurosurg Psychiatry. 1958;21:89–91. 162. Bostrom J, Meyer-Puttlitz B, Wolter M, et al. Alterations of the tumor suppressor genes CDKN2A
130. Smith JS, Quinones-Hinojosa A, Harmon-Smith M, et al. Sex steroid and growth factor profile (p16(INK4a)), p14(ARF), CDKN2B (p15(INK4b)), and CDKN2C (p18(INK4c)) in atypical and
of a meningioma associated with pregnancy. Can J Neurol Sci. 2005;32:122–127. anaplastic meningiomas. Am J Pathol. 2001;159:661–669.
131. Lusis EA, Scheithauer BW, Yachnis AT, et al. Meningiomas in pregnancy: a clinicopathologic 163. Rogers L, Barani I, Chamberlain M, et al. Meningiomas: knowledge base, treatment outcomes,
study of 17 cases. Neurosurgery. 2012;71:951–961. and uncertainties. A RANO review. J Neurosurg. 2015;122:4–23.
132. Laviv Y, Ohla V, Kasper EM. Unique features of pregnancy-related meningiomas: lessons learned 164. Han SJ, Reis G, Kohanbash G, et al. Expression and prognostic impact of immune modulatory
from 148 reported cases and theoretical implications of a prolactin modulated pathogenesis. molecule PD-L1 in meningioma. J Neurooncol. 2016;130:543–552.
Neurosurg Rev. 2016. 165. Kaley TJ, Wen P, Schiff D, et al. Phase II trial of sunitinib for recurrent and progressive atypical
133. Perry A, Gutmann DH, Reifenberger G. Molecular pathogenesis of meningiomas. J Neurooncol. and anaplastic meningioma. Neuro Oncol. 2015;17:116–121.
2004;70:183–202. 166. Graillon T, Defilles C, Mohamed A, et al. Combined treatment by octreotide and everolimus:
134. Wolfsberger S, Doostkam S, Boecher-Schwarz HG, et al. Progesterone-receptor index in octreotide enhances inhibitory effect of everolimus in aggressive meningiomas. J Neurooncol.
meningiomas: correlation with clinico-pathological parameters and review of the literature. 2015;124:33–43.
Neurosurg Rev. 2004;27:238–245. 167. Moazzam AA, Wagle N, Zada G. Recent developments in chemotherapy for meningiomas: a
135. Roser F, Nakamura M, Bellinzona M, et al. The prognostic value of progesterone receptor status review. Neurosurg Focus. 2013;35:E18.
in meningiomas. J Clin Pathol. 2004;57:1033–1037. 168. Norden AD, Ligon KL, Hammond SN, et al. Phase II study of monthly pasireotide LAR (SOM230C)
136. Ji Y, Rankin C, Grunberg S, et al. Double-Blind Phase III Randomized Trial of the Antiprogestin for recurrent or progressive meningioma. Neurology. 2015;84:280–286.
Agent Mifepristone in the Treatment of Unresectable Meningioma: SWOG S9005. J Clin Oncol. 169. Bi WL, Wu WW, Santagata S, et al. Checkpoint inhibition in meningiomas. Immunotherapy.
2015;33:4093–4098. 2016;8:721–731.

298

Das könnte Ihnen auch gefallen