Meningioma Pathology, Genetics, and Biology: Abstract

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Journal of Neuropathology and Experimental Neurology Vol. 63, No.

4
Copyright q 2004 by the American Association of Neuropathologists April, 2004
pp. 275 286

Meningioma Pathology, Genetics, and Biology

KATRIN LAMSZUS, MD

Abstract. Over the past 5 to 10 years, important advances were made in the understanding of meningioma biology. Progress
in molecular genetics probably represents the most important accomplishment in the comprehensive knowledge of meningioma
pathogenesis. Several genes could be identified as targets for mutation or inactivation. Additional chromosomal regions were
found to be commonly deleted or amplified, suggesting the presence of further tumor suppressor genes or proto-oncogenes,
respectively, in these regions. Histopathologically, the most important innovation is represented by the revised WHO classi-
fication in the year 2000. Meningioma grading criteria in the new classification scheme are more precise and objective, and
should thus improve consistency in predicting tumor recurrence and aggressive behavior. This review focuses mainly on the
advances in molecular biology that were achieved in recent years. It summarizes the most important aspects of meningioma
classification as the basis to place biological observations into a correlative context, and, further, includes mechanisms of
angiogenesis and edema formation as well as the role of hormone receptors in meningiomas.

Key Words: Angiogenesis; Classification; Edema; Meningioma; Neurofibromatosis; Progesterone.

INTRODUCTION attention, although, more recently, progress has some-


what stagnated as receptor expression profiles are now
Meningiomas have long been a subject of intense ge-
relatively well characterized and therapeutic efforts tar-
netic and biological interest. They were among the first
geting these receptors have so far been largely disap-
solid neoplasms studied using cytogenetic techniques.
pointing. Another field that has attracted interest, espe-
Frequent monosomy of chromosome 22 in meningiomas
cially since the advancement of imaging techniques in
was reported as early as 1972 (1). Cytogenetically, me-
the 1990s, are mechanisms of edema formation. These
ningioma has meanwhile become one of the best-studied
appear to be closely related to tumor angioarchitecture,
neoplasms in humans. Over the past decade, advanced
and a variety of growth factors have since been analyzed
techniques such as automated sequencing, microsatellite
for their contribution to edema formation and meningi-
analysis, fluorescence in situ hybridization (FISH), or
oma angiogenesis.
comparative genomic hybridization (CGH) have further
The purpose of this review is to outline (i) important
facilitated rapid identification of alterations at the molec-
aspects of meningioma classification and grading, (ii)
ular genetic level.
common gene and chromosome alterations that form the
For the first time, the revised WHO classification of
basis for a tumor progression model, (iii) the current sta-
2000 includes genetic findings in addition to pathological
tus in hormone receptor research, and (iv) mechanisms
and immunohistochemical features. For glial tumors, ge-
of edema formation and angiogenesis.
netic alterations meanwhile have even become clinically
relevant, insofar as deletions on chromosome arms 1p CLINICOPATHOLOGICAL ASPECTS
and 19q have been found associated with responsiveness
Incidence and Etiology
to chemotherapy and therapeutic outcome of oligoden-
droglial tumors. For meningiomas, none of the typical Meningiomas are composed of neoplastic meningothe-
genetic aberrations have yet attained clinical relevance. lial (arachnoidal cap) cells. They constitute approximate-
However, the comprehensive analysis of these aberrations ly 20% of all primary intracranial tumors, with an ap-
in relation to histological grade has led to a model in proximate annual incidence of 6 per 100,000 (2). The
which early alterations that are presumably involved in peak incidence is between the sixth and seventh decades
meningioma formation could be distinguished from later of life. Particularly among middle-aged patients, menin-
alterations that are associated with tumor progression (2). giomas are significantly more frequent in woman than in
In addition to genetic alterations, other biological fea- men with a greater than 2:1 ratio. The majority of me-
tures of meningiomas have also been investigated. Since ningiomas are attached to the dura mater and arise within
the late 1970s, hormone receptors have received great the intracranial cavity, the spinal canal, or, rarely, the or-
bit. Some meningiomas, especially those of the sphenoid
wing, may arise primarily as intraosseous tumors. Ap-
From Department of Neurosurgery, University Hospital Hamburg- proximately 40% of meningioma patients suffer from sei-
Eppendorf, Hamburg, Germany. zures; other clinical symptoms depend upon the location
Correspondence to: Katrin Lamszus, MD, Laboratory for Brain Tu-
mor Biology, Department of Neurosurgery, University Hospital Ham-
of the tumor.
burg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany. E-mail: Several endogenous and exogenous factors predispose
lamszus@uke.uni-hamburg.de to meningioma development (2). The majority of patients

275

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
276 LAMSZUS

who suffer from neurofibromatosis type 2 (NF2) develop criteria correlate with higher recurrence rates (30%–
meningiomas. In addition, families with an increased sus- 40%). In addition to tumors that fulfill the criteria for
ceptibility to meningiomas but without NF2 have been atypia, chordoid and clear cell meningiomas are also as-
reported. The clearest exogenous association exists for sociated with a high rate of recurrence and aggressive
ionizing radiation, after which meningiomas develop with behavior and are therefore also classified as WHO grade
an average latency period of several decades. In addition, II.
sex hormones have been implicated, mainly due to the Anaplastic (malignant) meningiomas, WHO grade III
overrepresentation of females among meningioma pa- (MIII) are rare (1%–3% of meningiomas) (2). They dis-
tients. play histological features of frank malignancy far in ex-
cess of the abnormalities present in atypical meningio-
Classification and Prognosis mas. Such features comprise either obviously malignant
The most important factors that determine the likeli- cytology (e.g. resemblance to sarcoma, carcinoma, or
hood of meningioma recurrence are extent of tumor re- melanoma) or high mitotic indices ($20 mitoses/10 high-
section and histological grade. Extent of resection is still power fields). The rare variants of papillary or rhabdoid
classified according to a scheme proposed by Simpson in meningioma are also classified as WHO grade III due to
1957, ranging from grade 1 (complete resection) to grade their highly aggressive behavior. Malignant meningiomas
5 (decompression only) (3). The histopathological me- have recurrence rates of 50% to 80%, and are usually
ningioma classification received special attention by the fatal within less than 2 years following diagnosis.
working group that devised the 2000 WHO classification, Notably, neither brain invasion nor proliferation indi-
and several substantial changes were introduced (4). ces (e.g. MIB-1 labeling) have been incorporated into the
These changes are mainly based on a series of studies current WHO grading criteria. Brain invasive meningio-
from the Mayo Clinic in which histopathological param- mas have a higher chance of recurrence and behave like
eters were correlated with clinical prognostic parameters atypical meningiomas; therefore, some authors prefer to
(5, 6). Whereas the previous WHO classification that assign them to WHO grade II (5, 7). However, brain in-
dates back to 1993 had ill-defined borders between be- vasion is considered a feature more pertinent to staging
nign, atypical, and anaplastic meningiomas, grading cri- than to malignancy grade, and molecular genetic inves-
teria are now much more stringent and objective. tigations have failed to reveal alterations characteristic of
The majority of meningiomas (80%–90%) are benign high-grade meningiomas in MI with brain invasion (4).
and classified as WHO grade I (MI) (2, 5). A variety of In addition, proliferation indices were not included into
histopathological subtypes fall into this category, includ- the grading criteria; due to high interinstitutional and in-
ing meningothelial, fibrous, and transitional meningiomas terindividual variations, reliable cut-off levels for differ-
as the most common variants. Less common subtypes are ent grades cannot be defined. Nevertheless, brain inva-
psammomatous, angiomatous, microcystic, secretory, sion as well as proliferation indices provide valuable
lymphoplasmacyte-rich, and metaplastic meningioma. MI additional prognostic information. Therefore, the WHO
carry a relatively low risk of recurrence (7%–20%) and working group recommended adding phrases such as
of aggressive behavior. Occasional mitoses and pleomor- ‘‘with brain invasion’’ or ‘‘with high proliferative activ-
phic nuclei are tolerable features within MI. However, the ity’’ to the diagnosis if appropriate (4).
location in which these tumors arise has a critical impact Despite the incorporation of genetic and immunohis-
on prognosis. Whereas tumors of the convexity are usu- tochemical findings, meningioma classification is still
ally cured by surgical resection, skull base tumors often based entirely on conventional histological criteria. Im-
have an unfavorable outcome. In particular, meningiomas munohistochemistry mainly has a role in differential di-
that arise in the petroclival region or involve the cavern- agnosis, for example, when distinguishing meningioma
ous sinus or orbit often display slow but relentless from hemangiopericytoma or other mesenchymal tumors.
growth, leading to widespread invasion and destruction Genetic analyses have also made important contributions
of bony structures. These tumors may either remain his- to differential diagnostics, for example, when recognizing
tologically benign over many years or they may eventu- hemangiopericytoma as an entity distinct from meningi-
ally progress towards a higher grade. oma (8); however, they currently have no role in routine
Between 5% and 15% of meningiomas are classified meningioma diagnosis. The present WHO classification,
as atypical, corresponding to WHO grade II (MII) (2, 5). with its more stringent criteria, should provide an im-
Diagnostic criteria for atypical meningiomas are either proved framework for genetic studies and foster their
increased mitotic activity (defined as $4 mitoses/10 high- comparability, which, hopefully, will lead to even more
power fields of 0.16 mm2) or 3 or more of the following refined combined histologic-genetic models.
features: increased cellularity, small cells with high nu- Many of the genetic studies that are reviewed in the
cleus to cytoplasm ratio, prominent nucleoli, uninterrupt- next sections were performed on the basis of the 1993
ed patternless or sheet-like growth, and necroses. These WHO classification. It should be kept in mind that the

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
MENINGIOMAS 277

reported findings may require some adjustment according


to the current classification.

GENETICS
NF2 Gene
Mutations in the NF2 tumor suppressor gene, located
in the chromosomal region 22q12.2, represent the most
frequent gene alteration in meningiomas. Early cytoge-
netic studies had already found monosomy of chromo-
some 22 in up to 70% of meningiomas (for review see
Collins et al [9]). This observation correlated well with
subsequent molecular genetic studies that identified loss
of heterozygosity (LOH) at polymorphic markers on 22q
as the most common molecular genetic alteration, present
Fig. 1. Hypothetic model of genomic alterations associated
in 40% to 70% of all meningiomas (10, 11). Mutations with the formation of benign meningiomas and progression to-
in the NF2 gene were detected in up to 60% of sporadic wards atypia and anaplasia. Only those alterations that were
meningiomas and are typically associated with LOH 22q found in more than 30% of tumors of a specific grade in the
(12–14). majority of studies (see text) are listed as contributing to the
Most NF2 mutations have a truncating effect, indicat- development of a particular grade (arrow). Alterations that in-
crease in frequency by at least another 20% (averaged over
ing that inactivation of the NF2 gene product merlin several studies) in tumors of the next higher histological grade,
(schwannomin) is functionally important for meningioma despite already contributing to the previous grade, are listed
pathogenesis. Absent or strongly reduced immunoreactiv- again.
ity for merlin was found in the majority of meningiomas
(15–17), and was strongly associated with LOH 22q (14). involved in the formation of most benign meningiomas
Merlin belongs to the 4.1 family of structural proteins (Fig. 1).
that link the cytoskeleton to proteins of the cytoplasmic In some meningiomas no NF2 mutation or LOH 22q
membrane (18). The mechanism by which merlin exerts could be detected despite a lack of merlin. An alternative
a tumor suppressive activity is still poorly understood. mechanism that involves merlin degradation by the pro-
The disruption of the signaling cascade that leads to cy- tease m-calpain was proposed to account for this phenom-
toskeletal reorganization is considered to be critical to enon (21). Different studies demonstrated activation of
tumor formation. Overexpression of merlin in both NF2- m-calpain in more than 50% of meningiomas, although
negative and NF2-positive human meningioma cells sig- no association between activation of m-calpain and merlin
nificantly inhibited their proliferation in vitro, which pro- status could be established (14, 17). Instead, concordance
vides further evidence for a role of merlin as a negative of LOH 22q and merlin loss suggested that other mech-
regulator of tumor growth (19). anisms, such as homozygous deletions or methylation as
Molecular genetic as well as protein studies showed well as undetected NF2 mutations, may account for the
that the frequency of NF2/merlin alterations differs loss of merlin (14).
among the 3 most common benign meningioma variants.
Whereas fibroblastic and transitional meningiomas harbor Meningiomas in NF2 Patients
NF2 mutations in approximately 70% to 80% of cases, Most patients who suffer from NF2 develop meningi-
meningothelial meningiomas carry mutations in only omas. NF2-associated meningiomas differ from sporadic
25% (13). A close correlation between the fibroblastic ones in several respects: (i) they usually arise several de-
variant and LOH 22q was also found (20). Correspond- cades earlier in life, (ii) they are frequently multiple, and
ingly, reduced merlin expression was observed in the ma- (iii) they belong more often to the fibroblastic variant
jority of fibroblastic and transitional meningiomas, but (22). NF2-associated meningiomas exhibit deletions on
rarely in meningothelial tumors (15, 17). The low fre- chromosome arm 22q in almost 100% of cases, a fre-
quency of NF2 alterations in the meningothelial variant quency that is significantly higher than in sporadic me-
suggests that the genetic origin of these tumors is largely ningiomas (7, 23). Additional genetic alterations such as
independent of NF2 gene alterations. Moreover, the sim- deletions on 1p, 6q, 9p, 10q, 14q, and 18q occur at sim-
ilar frequency of NF2 mutations in atypical and anaplas- ilar frequency as in sporadic cases (23).
tic as well as in benign fibroblastic and transitional me- Most investigations found that the frequency of atyp-
ningiomas suggests that NF2 mutations are not involved ical or malignant meningiomas was not increased among
in progression to higher-grade meningiomas. Rather, NF2 NF2-associated tumors (23–25). The MIB-1 labeling in-
mutations appear to represent an early alteration that is dex in NF-2-associated meningiomas appeared to be

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
278 LAMSZUS

slightly higher in 1 study (25), but a more detailed anal- the NF2 locus in some tumors (20). Furthermore, a recent
ysis of a larger number of samples revealed no difference case report described monosomy for chromosome 22 but
compared with sporadic meningiomas (23). In a recent lack of NF2 mutation in multiple meningiomas from a
study, NF2-associated meningiomas together with pedi- single patient (33).
atric meningiomas were found to contain a higher per- Several candidate tumor suppressor genes on 22q were
centage of WHO grade II and grade III tumors than spo- cloned in recent years, and some were screened for mu-
radic cases (7). However, the feature of brain tations and/or altered expression levels in meningiomas.
invasiveness in an otherwise benign tumor was consid- Three of these genes map to the 22q12.2 region in rela-
ered equivalent to WHO grade II. The experience in our tively close proximity to the NF2 gene, namely ADTB1
institution is that NF2 patients rarely develop malignant (b-adaptin, BAM22), RRP22, and GAR22. The ADTB1
meningiomas and that mortality in these patients is usu- gene was cloned based on a 140-kb homozygous deletion
ally due to other causes. in a sporadic meningioma. Expression analysis showed
that 12% of sporadic meningiomas lacked ADTB1 tran-
Multiple and Recurrent Meningiomas scripts. However, no mutations could be identified in 110
Multiple meningiomas are independently arising and sporadic meningiomas, suggesting epigenetic mecha-
spatially separate tumors. They are observed in 1% to nisms of gene inactivation (34). Similarly, no RRP22 or
8% of meningioma patients. Multiple meningiomas are GAR22 mutations were detected in 12 meningiomas, of
particularly common in NF2 patients and also in rare which half displayed LOH in the 22q12-q22 region, al-
non-NF2 families with a hereditary meningioma predis- though none exhibited NF2 mutations (35).
position. Several studies investigated whether multiple Several other candidate genes map outside the 22q12.2
meningiomas represent metastatic meningeal seeding of region. The MN1 gene is located at the 22q12.1 segment.
1 tumor or de novo formation of separate tumors. Iden- MN1 was found to be disrupted by a translocation in a
tical NF2 mutations and patterns of X-chromosome in- meningioma (36), however, subsequent studies demon-
activation revealed that the majority of multiple as well strated that it acts as an oncogenic transcription coacti-
as recurrent meningiomas are of clonal origin, so that vator rather than a tumor suppressor.
multiplicity most likely represents subarachnoid spread The hSNF5/INI1 (SMARCB1) gene, which maps to
(26–29). Alternatively, NF2 mosaicism could underlie 22q11.23, is frequently mutated in atypical teratoid/rhab-
doid tumors. In 1 study, an identical missense mutation
some cases. NF2 mutations and clonal origin were more
in the hSNF5/INI1 gene was identified in 4 of 126 me-
frequently found in patients with a larger number of tu-
ningiomas analyzed (37). Three of these 4 tumors also
mors than in patients with only 2 tumors, which is com-
contained NF2 mutations, suggesting that loss of hSNF5/
patible with either mosaicism or with an increased ability
INI1 function does not represent an alternative mecha-
of liquorigenic seeding caused by a mutant NF2 gene.
nism to NF2 inactivation in the pathogenesis of menin-
A recent study focused specifically on differences be-
giomas, but that silencing of hSNF5/INI1 may co-operate
tween multiple meningiomas in sporadic versus familial
with impairment of NF2 function.
non-NF2 cases (30). All NF2 mutations that were de-
Expression of the CLTCL1/CLH-22 gene, which maps
tected occurred in tumors from patients with no affected
to 22q11.21, was found to be absent in 80% (37 of 46)
relatives, suggesting that the NF2 inactivation is frequent-
meningiomas analyzed, suggesting that this gene may
ly involved in multiple sporadic meningiomas but is rare also be relevant to tumor development (38). Among tu-
in multiple meningioma kindreds. In agreement with this mors with absent gene expression were cases with and
observation, linkage analysis of a family with multiple without chromosome 22 deletions. However, mutations
meningiomas showed no segregation with the NF2 locus of CLTCL1/CLH-22 have not been reported and the
(31). In another multiple meningioma kindred, immuno- mechanism of its loss of expression is unclear.
reactivity for merlin was detected, implying that the NF2 The LARGE gene maps distally to the NF2 region
gene was not inactivated (32). Interestingly, most non- (22q12.3), and represents another interesting candidate
NF2 meningioma family members develop meningothe- gene (39). However, it is one of the largest human genes,
lial meningiomas, which is in line with the observation and mutation or expression analyses have not been pub-
that this variant seems to arise independently of NF2 in- lished.
activation.
DAL-1 Gene and Other Alterations on Chromosome 18
Other Genes on Chromosome 22 The DAL-1 protein shares significant homology with
Several studies suggested that other tumor suppressor merlin and also belongs to the 4.1 family of membrane-
genes may lie outside the NF2 region on the long arm of associated proteins. It has tumor suppressor properties
chromosome 22. The frequency of LOH 22 exceeds that and maps to chromosomal region 18p11.3. Recent im-
of NF2 mutations in meningiomas, and deletion mapping munohistochemical studies showed that DAL-1 expres-
revealed interstitial deletions on 22q that did not include sion is lost in 76% of sporadic meningiomas, a frequency

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
MENINGIOMAS 279

similar to that detected for loss of merlin (40, 41). Lack 13% to 26% of MI, in 40% to 76% of MII, and 70% to
of DAL-1 protein was only slightly, and not significantly, 100% of MIII. Moreover, 1p deletions were found to be
more frequent in anaplastic meningiomas (87%) than in associated with tumor progression in several individual
benign and atypical meningiomas (70%–76%), suggest- cases of recurrent meningiomas (42, 44–50). These find-
ing that it represents an early event in meningioma tu- ings suggest that loss of genomic information from 1p is
morigenesis (41). These immunohistochemical findings relevant to meningioma progression rather than tumor
were largely reflected at the mRNA expression level. Al- formation (Fig. 1).
though LOH at 18p11.3 was detected in 71% of inves- Several genes on 1p were screened for alterations in
tigated cases (40), the mechanism of DAL-1 inactivation meningiomas, including CDKN2C (p18 INK4c ), p73,
is still unresolved. Mutations were not detected, but RAD45L, and ALPL. In 3 different studies of more than
screening had not included the full gene (40). Given the 100 meningiomas only 1 mutation in the CDKN2C gene,
frequent LOH at the DAL-1 gene location, homozygous which is located at 1p32, and 1 homozygous deletion
deletions are unlikely, so that epigenetic alterations re- were found (49, 51, 52). No loss of CDKN2C transcripts
main a more likely possibility. and no aberrant methylation were detected, suggesting
Combined loss of DAL-1 and merlin was detected in that the CDKN2C gene is rarely altered in meningiomas
58% of investigated cases (41), suggesting that both are (49, 51). The RAD54L gene also maps to 1p32. No
not part of a single growth regulatory pathway in which RAD54L mutations were detected in a series of 29 me-
inactivation of either member causes the same effect. A ningiomas (53), however, a silent polymorphism at nu-
tendency for more frequent combined DAL-1 and merlin cleotide 2,290 turned out to be associated with a higher
loss was observed in anaplastic meningiomas (70%) frequency of meningioma development (54).
compared to atypical (60%) and benign (50%) ones, sug- The ALPL gene at 1p36.1-p34 encodes for alkaline
gesting that although both alterations are considered early phosphatase. Loss of alkaline phosphatase activity was
changes, simultaneous loss of both proteins may provide reported to be strongly associated with loss of 1p in me-
a selective growth advantage (41). ningiomas, leading to the assumption that ALPL might
Cytogenetic and CGH analyses have failed to recog- have tumor suppressor function (55, 56). However, struc-
nize 18p11.3 as a region of frequent chromosomal losses. tural ALPL alterations have not been documented, and
Microsatellite analysis revealed that deletions in this re- functional evidence for tumor suppressor properties of
gion are centered around the DAL-1 gene locus and are alkaline phosphatase is lacking.
too small to be detected by karyotyping or CGH (41). In Another candidate meningioma suppressor gene is
contrast, CGH analysis frequently identified losses of ge- TP73, which maps to 1p36.32. However, only 1 mutation
netic material from the long arm of chromosome 18. was found in more than 50 meningiomas analyzed (57,
These losses were associated with increasing histological 58), which argues against a significant role of TP73 in-
grade and therefore appear to be associated with menin- activation in meningioma progression. Expression of
gioma progression (Fig. 1). Losses on 18q were detected TP73 was found to increase with tumor grade, suggesting
in 67% of MIII and 40% of MII, but only 13% of MI that TP73 might have a dominant oncogenic function
(42, 43). Büschges et al investigated 37 meningiomas for rather than a classic tumor suppressor function (58).
mutation and expression of 4 tumor suppressor genes lo- Taken together, the analyses of individual genes on 1p
cated at 18q21, namely MADH2, MADH4, APM-1, and do not support a significant meningioma suppressor func-
DCC (43). However, only 1 missense mutation in the tion of any of the genes investigated. Deletion mapping
APM-1 gene was found in an atypical meningioma. No studies defined at least 2 different commonly deleted re-
other mutations were identified and transcripts for all 4 gions on 1p, suggesting that more than 1 tumor suppres-
genes were detectable in all tumors. These findings do sor gene on 1p might be involved in meningioma pro-
not support a significant role for MADH2, MADH4, gression. One of these regions was mapped to 1p32 (52,
APM-1, and DCC alterations in meningioma pathogene- 54, 59, 60). However, sequence information that became
sis. Based on CGH analysis, which identified a single available through the human genome project indicates
meningioma in which loss was restricted to 18q22-qter that mapping data that were previously interpreted mainly
(42), and on studies of non-CNS tumors with losses on on the basis of recombination events require revision in
18q, it was suggested that a putative meningioma pro- several instances. For example, Sulman et al had mapped
gression-associated gene may be located distally to the the minimally deleted region at 1p32 between microsat-
18q21 region (43). ellite markers D1S2713 distally and D1S2134 proximally
(59). According to current databases, however, D1S2713
Chromosome 1 maps to 1p34.1, and D1S2134 maps to 1p33 (http://
Deletions on 1p are the second most frequent chro- genome.cse.ucsc.edu/cgi-bin/, and http://www.ncbi.nlm.
mosomal abnormality in meningiomas. The frequency of nih.gov/mapview/maps.cgi). Similarly, Leraud et al had
1p deletions increases with tumor grade and occurs in identified a commonly deleted region between D1S234

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
280 LAMSZUS

distally and D1S2797 proximally, supposedly at 1p32, et al had mapped a critical region to 14q24.3-q32.33 be-
whereas current databases map D1S234 to 1p36.11 and tween microsatellites D14S48 and D14S23 (45). Similar-
D1S2797 to 1p33 (52). Even the order of microsatellite ly, Menon et al had identified 14q24.3-q32.3 as the small-
markers that were chosen in different studies is not al- est commonly deleted region. Also Tse et al described a
ways in agreement with current databases. Correct inter- cluster region of deletion at 14q24.3-q31, but found an-
pretation of previous mapping results would require an other region at 14q32.1-q32.2 (63), whereas Leone et al
extensive re-analysis of the data on the basis of the avail- suggested 2 different regions, one at 14q22-q24 and an-
able sequence information. Such an analysis is beyond other at 14q32 (47). In contrast, a CGH study by Weber
the capacity and scope of this review. et al, 1 tumor defined a region of common deletion at
A second common region of deletion was mapped to 14q21 (42). It has to be concluded from these studies that
no consistent commonly deleted region has yet been iden-
1pter-p34 distally to the D1S496 locus (46, 49). Accord-
tified on 14q, and no particular tumor suppressor gene
ing to current databases, this microsatellite still maps to
has evolved as prime candidate for a meningioma pro-
1p34.3 and consequently there is less confusion for this
gression gene.
region than for the more proximal region. A terminally
located region of deletion at 1p36 was also reported by Chromosome 10
Bello et al (60). This group further identified up to 3 other Deletions on chromosome 10 have received almost as
commonly deleted regions, including a frequently affect- much attention in meningiomas as alterations on 14q.
ed region at 1p34-p32 and 2 less frequently deleted re- Rempel at al initially discovered an association between
gions at 1p22 and 1p21.1-p13. allelic losses on chromosome 10 with meningioma pro-
Taken together, it appears that at least 2 common re- gression (65). Most subsequent LOH or CGH analyses
gions of deletion are present on chromosome arm 1p, detected deletions preferably on the long arm of chro-
namely one at 1p34-p32 and another at 1pter-p34. It re- mosome 10. In several large studies, frequencies of 10q
mains to be shown whether re-analysis of previous stud- deletions were in the order of 5% to 12% for MI, 29%
ies based on genomic sequence information can clarify to 40% for MII, and 40% to 58% for MIII (42, 45, 66,
some of the confusing mapping data and can narrow 67). These percentages were even higher in 2 recent stud-
down regions containing putative tumor suppressor ies by Mihaila et al in which 11 different microsatellite
genes. Naturally, the current confusion is not unique to loci on chromosome 10 in 208 meningiomas were ana-
meningiomas, since deletions on 1p are also common in lyzed (68, 69). Correlative analyses revealed an unfavor-
able prognostic significance for LOH at D10S179 (1p14)
oligodendrogliomas, neuroblastomas, and many epithelial
or D10S169 (1q26.3), which predicted higher tumor
cancers.
grade, and of D10S209 (10q26.12) and D10S169
(10q26.3), which may predict shorter survival and/or
Chromosome 14
shorter time to recurrence, respectively (68).
Cytogenetically, loss of chromosome 14 represents the No specific tumor suppressor gene on chromosome 10
third most frequently detected abnormality in meningio- has yet been shown to be inactivated in a major fraction
mas after aberrations of chromosomes 22 and 1 (61). of meningiomas. The PTEN gene at 10q23.3 was ana-
LOH and FISH analyses identified deletions on chro- lyzed in a large number of samples, however, mutations
mosome arm 14q in up to 31% of MI, 40% to 57% of were only detected in 2 different MIII and no homozy-
MII, and 55% to 100% of MIII (42, 45, 47, 50, 62, 63). gous deletions were found (66, 70, 71). The DMBT1
The strongly increased frequency of 14q deletions in tu- gene, which maps to 10q26.11-q26.12, has also been in-
mors of higher grade suggests an involvement in menin- vestigated, but no homozygous deletions were found in
gioma progression (Fig. 1). A recent study demonstrated atypical or malignant meningiomas (67).
Mapping studies defined several different commonly
that deletions on this chromosome arm were an indepen-
deleted regions on chromosome 10. Simon et al had ini-
dent adverse prognostic parameter, which when combined
tially mapped such a region to 10q24-qter (45). However,
with histological grade and patient age could identify pa-
according to current databases, the order and placement
tients at increased risk of relapse (64). A similar obser- of microsatellite loci are slightly different, so that a par-
vation was made in another study in which losses of 14q tial deletion in one of the analyzed tumors would now
in MI were predictive of tumor recurrence (50). place the minimally deleted region distally to a marker
No specific meningioma suppressor gene has yet been at 10q25.3. Peters et al observed LOH most often at mi-
identified on chromosome 14. Deletion mapping studies crosatellite loci D10S676 (10q22.1) and D10S677
have defined several commonly deleted regions on 14q; (10q23.33) (66). By CGH analysis, Weber et al obtained
however, as already described for 1p, their interpretation evidence for commonly deleted regions on both chro-
requires caution since most studies were performed be- mosome arms, namely one at 10p15 and one at 10q25-
fore completion of the Human Genome Project. Simon qter (42). The latest study by Mihaila et al defined 1

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
MENINGIOMAS 281

region on 10p between 10pter and D10S89 (p12.1), and astrocytomas in particular. However, TP53 mutations are
3 regions on 10q, namely between D10S109 (10q22.3) rare in meningiomas. Several relatively large studies re-
and D10S215 (10q23.31), between D10S187 (10q25.3) ported either no or only single meningiomas with spo-
and D10S209 (10q26.12), and between D10S169 radic TP53 mutations (49, 71, 74, 75). Mutation of TP53
(10q26.3) and 10qter. Taken together, these studies indi- frequently results in increased stability of the p53 protein,
cate that the pattern of allelic losses on chromosome 10 which, in contrast to wild-type p53, then becomes de-
is so complex that no single consistent minimal region tectable by immunohistochemistry. Other stabilization
of deletion could yet be identified. mechanisms may also lead to p53 accumulation and thus
detectability. Immunohistochemical analyses of p53 in
Chromosome 9
meningiomas have yielded contradictory results, and
Losses of genetic material on chromosome 9 were fre- there seems to be a substantial degree of overlap between
quently found in malignant meningiomas, but only rarely different malignancy grades (76). In essence, most (but
in benign or atypical ones (42). The short arm of chro- not all) studies demonstrated an association between p53
mosome 9 has attracted particular attention, as it contains accumulation and malignancy grade (75–78). The biolog-
the tumor suppressor genes CDKN2A (p16INK4a/MTS1), ical significance of this p53 accumulation in meningio-
p14ARF, and CDKN2B (p15INK4b/MTS2) at 9p21, which are mas is not clear.
inactivated at a high frequency in a variety of human More recently, the long arm of chromosome 17 has
tumors. The p16 and p15 proteins regulate cell cycle pro- also attracted interest since CGH analysis demonstrated
gression at the G1/S-phase checkpoint by inhibiting cy- high-level amplification on 17q in 42% of anaplastic me-
clin-cdk complexes, which are involved in the transcrip- ningiomas but in almost none of the non-malignant me-
tional control and phosphorylation of pRB. The p14ARF ningiomas (42). A subsequent study identified copy num-
protein blocks Mdm2-mediated degradation of p53. ber increases at 17q microsatellite markers in 61% of
Losses on 9p, as determined by combined CGH and MIII in contrast to only 21% in MII and 14% MI (79).
microsatellite analysis, were found in 38% of MIII, 18% However, only 2 MIII in this series displayed high-level
of MII, and 5% of MI (49). By FISH analysis the fre- amplification defined as a .5-fold increase in the copy
quencies of deletion at 9p21 or monosomy 9 were ;2- number of 1 allele. The amplification patterns defined
fold to 3-fold higher, most likely because FISH analysis several common regions of increased allele copy number,
on tumor sections also detects alterations that are only one of which encompassed the putative proto-oncogene
focal (72). Boström et al found homozygous deletions of
PS6K at 17q23. However, in contrast to another recent
CDKN2A, p14ARF, and CDKN2B in 46% of MIII, and 3%
study by Cai et al, which identified PS6K amplification
of MII but never in MI (49). Two of 13 MIII carried
in 3/22 anaplastic meningiomas (80), only low level copy
somatic point mutations in CDKN2A and p14ARF. In ad-
number increases were detected in 13/44 tumors (i.e. 4/
dition, 5 tumors without homozygous loss or mutation
19 MII and 9/18 MIII), despite high-level amplification
lacked detectable transcripts for at least 1 of the 3 genes
of adjacent loci in 2 MIII. This discrepancy could have
(5% of MI, 9% of MII, and 8% of MIII). Thus, the ma-
technical reasons, because Cai et al used FISH analysis,
jority of malignant meningiomas displayed alterations of
which recognizes also only focal areas with high-level
CDKN2A, p14ARF, and CDKN2B, whereas these were in-
PS6K amplification, whereas focal high-level amplifica-
frequent in benign meningiomas. Similarly, in an expres-
sion analysis by Simon et al, only ;26% of MI and MII, tions may have appeared as low-level amplification in the
but 57% of MIII lacked both p16 and p15 protein ex- real-time PCR analysis performed by Büschges et al (79).
pression. In addition, 36% of non-malignant but 71% of Taken together, these studies suggest that PS6K may
the anaplastic meningiomas lacked p14 protein (73). not be the major target gene for high-level amplification
These studies show that both the pRB and p53 pathways detected by microsatellite analyses in malignant menin-
are disrupted in the majority of MIII, and that inactivation giomas, but that its amplification in tumor cell subpop-
of cell cycle regulation at the G1/S-phase checkpoint may ulations may still be important for progression. Mapping
be critical to the malignant progression of meningiomas. results define at least a bipartite common region of am-
Interestingly, a recent study demonstrated a prognostic plification at 17q22-q23, between markers D17S790 and
relevance for CDKN2A alterations, showing that among D17S1607 as well as between D17S1160 and PS6K, sug-
patients with anaplastic meningiomas, those with gesting the possibility of more than one relevant proto-
CDKN2A deletions had a significantly shorter survival oncogene (79).
(72).
Alterations on Other Chromosomes
Chromosome 17 Numerous other chromosomal alterations have also
The tumor suppressor gene TP53, which is located on been identified in meningiomas, albeit less consistently
the short arm of chromosome 17, is one of the most fre- than those described in the previous paragraphs. Rela-
quently mutated genes in human cancer in general and in tively frequent among them are losses on 3p, 6q, X, and

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
282 LAMSZUS

Y, as well as gains on 1q, 9q, 12q, 15q, and 20q (42, 81– necessary for unlimited cell proliferation. In contrast to
83). Very few studies have searched for specific alter- germ-line cells and most embryonic cells, telomerase is
ations on these chromosome arms. For example, some not active in most normal adult tissues. However, it is
studies showed that the CDK4 and MDM2 genes, which frequently reactivated in cancer.
are located on chromosome arm 12q, are rarely amplified Telomerase activity has been detected in only 3% to
in meningiomas (42, 49, 84, 85). Similarly, no other spe- 21% of benign meningiomas, but in 58% to 92% of MII
cific gene alterations on these less commonly altered and 100% of MIII (88–90). Langford et al described that
chromosomes could be identified in a significant number telomerase activity was strongly associated with poor
of cases. outcome in benign meningiomas, suggesting that it may
provide a prognostic marker (88). Two essential compo-
Radiation-Induced Meningiomas nents were identified in human telomerase: an RNA sub-
Ionizing radiation to the skull is a well-established risk unit termed hTR (telomerase RNA) that contains a tem-
factor for meningioma development. Most of the knowl- plate sequence for telomeric repeat synthesis, and a
edge about this association was obtained from immi- reverse transcriptase subunit hTERT (telomerase reverse
grants to Israel who had been treated with low-dose cra- transcriptase). Generally, expression of hTERT correlates
nial irradiation for tinea capitis between 1948 and 1960. best with telomerase activity, whereas hTR can be present
Usually, patients who were treated during childhood de- even when enzyme activity is repressed. Simon et al
veloped meningiomas after a latency period of 20 to 40 showed that the detection rate for hTERT expression in
years. Radiation-induced meningiomas (RIM) are often meningiomas was even higher than that for telomerase
clinically and histologically more aggressive (corre- activity and that all telomerase-positive tumors also ex-
sponding to MII or MIII), are often multiple, and have a pressed hTERT, but not vice versa (89). In addition, some
higher proliferative activity than their sporadic counter- tumors that subsequently recurred expressed hTERT but
parts (2). lacked detectable telomerase activity. These findings sug-
A few studies addressed the genetic mechanisms in- gest that hTERT might even be a more sensitive marker
volved in RIM development. Mutation analyses of alto- for an aggressive clinical course than telomerase activity.
gether more than 30 RIM showed that NF2 mutations are
relatively rare and occur in less than 25% of RIM com- BIOLOGY
pared to more than 50% in sporadic control cases (71, Angiogenesis, Edema, and Growth Factors
86). Likewise, allelic losses on 22q were only detected
in 2/7 RIM (86), and a recent CGH study found mono- Meningiomas derive their blood supply predominantly
somy for chromosome 22 in only 1/5 cases (87). Thus, from meningeal vessels originating in the external carotid
NF2 alterations appear to play a less important role in circulation. Additional supply from cerebral-pial vessels
the pathogenesis of RIM than in sporadic meningiomas. is present in approximately 60% of patients (91). Menin-
Several other genes, including some that are prone to giomas are of variable vascularity, ranging from sparsely
developing radiation-associated mutations, were also an- vascularized to highly vascular angiomatous meningio-
alyzed in RIM. Mutations in the TP53 and PTEN genes mas. They further display variable degrees of peritumoral
were confined to single cases, and no mutations were brain edema, ranging from absent to life-threatening con-
observed in the HRAS, KRAS and NRAS genes (71). Al- ditions. In contrast to gliomas, there is no obvious asso-
lelic losses on 1p were detected at a slightly higher fre- ciation between edema and histological grade in menin-
quency in RIM than in sporadic meningiomas and CGH giomas.
analysis identified losses on 7p, which are uncommon in Vascular endothelial growth factor-A (VEGF-A),
sporadic meningiomas, in 67% of RIM (87). Therefore, which has also been termed vascular permeability factor,
putative tumor suppressor genes on 1p and 7p could be is considered a key regulator of angiogenesis and edema
relevant to the development of RIM, although the avail- formation. The VEGF-A mRNA is expressed by menin-
able series are too small to draw reliable conclusions. gioma cells (92, 93). Several studies demonstrated that
VEGF-A levels in meningiomas are associated with the
Telomerase extent of peritumoral edema (92–94). Moreover, menin-
Telomeres consist of stretches of repetitive DNA se- giomas with striking VEGF-A expression usually ap-
quences that maintain chromosomal stability. Telomeric peared to receive some blood supply through cerebral-
DNA is progressively shortened with each mitosis. When pial arteries, and both VEGF-A expression as well as
telomeres reach a critical length, cells ultimately become cerebral-pial blood supply were associated with the extent
senescent. Moreover, shortened telomeres can initiate ab- of brain edema (94).
errant fusion or recombination of chromosome ends and Two small studies suggested that VEGF-A mRNA ex-
thus contribute to cancer development. Telomerase is a pression may correlate with meningioma vascularity (93,
reverse transcriptase that stabilizes telomere length and is 95). However, when determining VEGF-A protein levels

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
MENINGIOMAS 283

in a relatively large number of 69 meningiomas, no as- that the presence of progesterone receptors was a favor-
sociation with microvessel density could be confirmed able prognostic factor (103). Meningioma cell prolifera-
(96). Moreover, and in contrast to gliomas, there was no tion was found to be inhibited by the progesterone re-
association between microvessel density and histological ceptor antagonist mifepristone (RU486) in vitro,
grade. Nevertheless, VEGF-A levels were increased 10- suggesting a treatment modality. However, several small
fold in anaplastic meningiomas and 2-fold in atypical me- clinical trials that were performed reported only marginal
ningiomas compared to benign ones. VEGF-A contained responses (reviewed in Sanson and Cornu [99] and Ragel
in protein extracts of human meningiomas induced cap- and Jensen [104]), and some prospective trials are still
illary-like tube formation and migration of endothelial awaiting completion. Anti-androgens also have antipro-
cells in vitro, indicating biological activity in this context liferative effects on meningioma cells in vitro, but their
(96). Another recent study reported a correlation between clinical usefulness has not yet been investigated.
VEGF-A protein expression and recurrence of benign Meningiomas also express non-steroid hormone recep-
meningiomas (97). Taken together, these findings suggest tors, including growth hormone, somatostatin and dopa-
that VEGF-A is probably involved in vascular remodel- mine receptors. Several in vitro or experimental in vivo
ing and angiogenesis in meningiomas which does, how- studies demonstrated antiproliferative effects of the
ever, not result in a net increase in vessel number with growth hormone receptor antagonist pegvisomant, the so-
increasing histological grade. Supposing that the more matostatin agonist octreotide, and the dopamine D2 re-
malignant meningiomas have a higher oxygen and met- ceptor agonist bromocriptine on meningiomas (99, 104).
abolic demand, VEGF-A might facilitate adaptation by However, full-scale clinical trials with these substances
modulating vascular permeability. have not been performed.
Several other growth factors, including VEGF-B, pla-
centa growth factor, scatter factor/hepatocyte growth fac- OUTLOOK
tor, and fibroblast growth factor-2 have also been ana- The completion of the Human Genome Project togeth-
lyzed in meningiomas. However, no clear correlation er with the development of increasingly more sophisti-
between either of these factors and meningioma angio- cated molecular research techniques have set the stage
genesis or malignancy grade has yet been established. for future high resolution genome-wide studies. The ex-
Moreover, expression of several other growth factors and ample of the DAL-1 tumor suppressor gene, where allelic
their receptors, including epidermal growth factor, plate- deletions were too small to be picked up by conventional
let-derived growth factor, and insulin-like growth factor screening methods, underscores the need for more sen-
and others has been analyzed in meningiomas. Their de- sitive techniques. A great improvement in sensitivity is
tailed discussion is beyond the scope of this article; for achieved by matrix-CGH (array-CGH), which has an ap-
reviews see Black et al (98) and Sanson and Cornu (99). proximately 100-fold higher resolution than conventional
CGH analysis and is able to narrow chromosomal gains
Hormones or losses to 75,000 bp segments. Another expanding field
Meningiomas are more than 2-fold more frequent in is that of high-density cDNA arrays, which have so far
woman than in men (2, 99). Accelerated meningioma been applied to meningiomas in only a few studies (105,
growth has been observed during pregnancy and during 106). Gene expression profiling by microarrays revealed
the luteal phase of the menstrual cycle. Moreover, me- characteristic profiles for different meningioma grades
ningiomas seem to arise at a slightly increased rate in and can thus provide prognostic information. Patterns of
breast cancer patients. Taken together, these observations gene over- or underexpression may also lead to discovery
suggest an etiological role for sex hormones in the of unknown tumor suppressor genes or oncogenes. How-
growth of these tumors. ever, for several genes that lack expression in a major
Most hormone receptor studies in meningiomas were fraction of meningiomas no structural gene alterations
performed in the 1990s using immunohistochemistry. can be identified. Inactivation of these genes apparently
These studies showed that estrogen receptors are only does not follow the classic scheme described for tumor
expressed in approximately 10% of meningiomas and suppressor genes. Instead, epigenetic inactivation by ab-
only at very low levels (reviewed in Black et al [98], errant methylation or suppression of transcription by oth-
McCutcheon [100], and Sanson and Cornu [99]). In con- er mechanisms most likely accounts for the absence of
trast, progesterone and androgen receptors are present in gene expression. Recently, array technology was expand-
approximately two thirds of meningiomas. Both are more ed also to methylation analysis, so that high-throughput
frequently expressed in women than in men (101–103). screening may soon generate comprehensive profiles of
Progesterone receptors have attracted the greatest interest. methylated genes. The investigation to what extent ab-
Their expression is inversely associated with histological errant methylation can explain inactivation of genes rel-
grade, and a study on 70 meningioma patients showed evant to meningioma formation or progression remains

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
284 LAMSZUS

one of the major challenges in the field besides further 18. Gusella JF, Ramesh V, MacCollin M, Jacoby LB. Merlin: The neu-
genomic and expression analyses. rofibromatosis 2 tumor suppressor. Biochim Biophys Acta 1999;
1423:M29–M36
ACKNOWLEDGMENTS 19. Ikeda K, Saeki Y, Gonzalez-Agosti C, Ramesh V, Chiocca EA.
Inhibition of NF2-negative and NF2-positive primary human me-
I thank Prof. Guido Reifenberger and Prof. Manfred Westphal for
ningioma cell proliferation by overexpression of merlin due to vec-
critically reading the manuscript and for helpful suggestions. I wish to
tor-mediated gene transfer. J Neurosurg 1999;91:85–92
apologize for not being able to cite several articles pertinent to the
20. Ruttledge MH, Xie YG, Han FY, et al. Deletions on chromosome
subject due to limitations in space and number of references allowed.
22 in sporadic meningioma. Genes Chromosomes Cancer 1994;10:
122–30
REFERENCES
21. Kimura Y, Koga H, Araki N, et al. The involvement of calpain-
1. Zankl H, Zang KD. Cytological and cytogenetical studies on brain dependent proteolysis of the tumor suppressor NF2 (merlin) in
tumors. 4. Identification of the missing G chromosome in human schwannomas and meningiomas. Nat Med 1998;4:915–22
meningiomas as no. 22 by fluorescence technique. Humangenetik 22. McLendon RE, Tien RD. Genetic syndromes associated with tu-
1972;14:167–69 mors and/or hamartomas. In: Bigner DD, McLendon RE, Bruner
2. Louis DN, Scheithauer BW, Budka H, von Deimling A, Kepes JJ. JM, eds. Russel and Rubinstein’s pathology of tumors of the ner-
Meningiomas. Pathology and genetics of tumours of the nervous vous system. London: Arnold Publishers, 1998:371–418
system. In: Kleihues P, Cavenee WK, eds. World Health Organi- 23. Lamszus K, Vahldiek F, Mautner VF, et al. Allelic losses in neu-
zation classification of tumours. Lyon: IARC Press, 2000:176–84 rofibromatosis 2-associated meningiomas. J Neuropathol Exp
3. Simpson D. The recurrence of intracranial meningiomas after sur- Neurol 2000;59:504–12
gical treatment. J Neurol Neurosurg Psychiatry 1957;20:22–39 24. Louis DN, Ramesh V, Gusella JF. Neuropathology and molecular
4. Kleihues P, Louis DN, Scheithauer BW, et al. The WHO classifi- genetics of neurofibromatosis 2 and related tumors. Brain Pathol
cation of tumors of the nervous system. J Neuropathol Exp Neurol 1995;5:163–72
2002;61:215–25 25. Antinheimo J, Haapasalo H, Haltia M, et al. Proliferation potential
5. Perry A, Stafford SL, Scheithauer BW, Suman VJ, Lohse CM. Me- and histological features in neurofibromatosis 2-associated and spo-
ningioma grading: An analysis of histologic parameters. Am J Surg radic meningiomas. J Neurosurg 1997;87:610–14
Pathol 1997;21:1455–65 26. Larson JJ, Tew JM, Jr., Simon M, Menon AG. Evidence for clonal
6. Perry A, Scheithauer BW, Stafford SL, Lohse CM, Wollan PC. spread in the development of multiple meningiomas. J Neurosurg
‘‘Malignancy’’ in meningiomas: A clinicopathologic study of 116 1995;83:705–9
patients, with grading implications. Cancer 1999;85:2046–56 27. von Deimling A, Kraus JA, Stangl AP, et al. Evidence for subarach-
7. Perry A, Giannini C, Raghavan R, et al. Aggressive phenotypic and noid spread in the development of multiple meningiomas. Brain
genotypic features in pediatric and NF2-associated meningiomas: Pathol 1995;5:11–14
A clinicopathologic study of 53 cases. J Neuropathol Exp Neurol 28. von Deimling A, Larson J, Wellenreuther R, et al. Clonal origin of
2001;60:994–1003 recurrent meningiomas. Brain Pathol 1999;9:645–50
8. Joseph JT, Lisle DK, Jacoby LB, et al. NF2 gene analysis distin- 29. Stangl AP, Wellenreuther R, Lenartz D, et al. Clonality of multiple
guishes hemangiopericytoma from meningioma. Am J Pathol 1995;
meningiomas. J Neurosurg 1997;86:853–58
147:1450–55
30. Heinrich B, Hartmann C, Stemmer-Rachamimov AO, Louis DN,
9. Collins VP, Nordenskjold M, Dumanski JP. The molecular genetics
MacCollin M. Multiple meningiomas: Investigating the molecular
of meningiomas. Brain Pathol 1990;1:19–24
basis of sporadic and familial forms. Int J Cancer 2003;103:483–88
10. Seizinger BR, de la Monte S, Atkins L, Gusella JF, Martuza RL.
31. Pulst SM, Rouleau GA, Marineau C, Fain P, Sieb JP. Familial me-
Molecular genetic approach to human meningioma: Loss of genes
ningioma is not allelic to neurofibromatosis 2. Neurology 1993;43:
on chromosome 22. Proc Natl Acad Sci U S A 1987;84:5419–23
2096–98
11. Dumanski JP, Carlbom E, Collins VP, Nordenskjold M. Deletion
32. Maxwell M, Shih SD, Galanopoulos T, Hedley-Whyte ET, Cosgro-
mapping of a locus on human chromosome 22 involved in the
ve GR. Familial meningioma: Analysis of expression of neurofi-
oncogenesis of meningioma. Proc Natl Acad Sci U S A 1987;84:
bromatosis 2 protein Merlin. Report of two cases. J Neurosurg
9275–79
12. Ruttledge MH, Sarrazin J, Rangaratnam S, et al. Evidence for the 1998;88:562–69
complete inactivation of the NF2 gene in the majority of sporadic 33. Lomas J, Bello MJ, Alonso ME, et al. Loss of chromosome 22 and
meningiomas. Nat Genet 1994;6:180–84 absence of NF2 gene mutation in a case of multiple meningiomas.
13. Wellenreuther R, Kraus JA, Lenartz D, et al. Analysis of the neu- Hum Pathol 2002;33:375–78
rofibromatosis 2 gene reveals molecular variants of meningioma. 34. Peyrard M, Pan HQ, Kedra D, et al. Structure of the promoter and
Am J Pathol 1995;146:827–32 genomic organization of the human beta’-adaptin gene (BAM22)
14. Ueki K, Wen-Bin C, Narita Y, Asai A, Kirino T. Tight association from chromosome 22q12. Genomics 1996;36:112–17
of loss of merlin expression with loss of heterozygosity at chro- 35. Zucman-Rossi J, Legoix P, Thomas G. Identification of new mem-
mosome 22q in sporadic meningiomas. Cancer Res 1999;59: bers of the Gas2 and Ras families in the 22q12 chromosome region.
5995–98 Genomics 1996;38:247–54
15. Hitotsumatsu T, Iwaki T, Kitamoto T, et al. Expression of neurofi- 36. Lekanne Deprez RH, Riegman PH, et al. Cloning and characteriza-
bromatosis 2 protein in human brain tumors: An immunohisto- tion of MN1, a gene from chromosome 22q11, which is disrupted
chemical study. Acta Neuropathol (Berl) 1997;93:225–32 by a balanced translocation in a meningioma. Oncogene 1995;10:
16. Huynh DP, Mautner V, Baser ME, Stavrou D, Pulst SM. Immuno- 1521–28
histochemical detection of schwannomin and neurofibromin in ves- 37. Schmitz U, Mueller W, Weber M, Sevenet N, Delattre O, von
tibular schwannomas, ependymomas and meningiomas. J Neuro- Deimling A. INI1 mutations in meningiomas at a potential hotspot
pathol Exp Neurol 1997;56:382–90 in exon 9. Br J Cancer 2001;84:199–201
17. Evans JJ, Jeun SS, Lee JH, et al. Molecular alterations in the neu- 38. Kedra D, Peyrard M, Fransson I, et al. Characterization of a second
rofibromatosis type 2 gene and its protein rarely occurring in men- human clathrin heavy chain polypeptide gene (CLH-22) from chro-
ingothelial meningiomas. J Neurosurg 2001;94:111–17 mosome 22q11. Hum Mol Genet 1996;5:625–31

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
MENINGIOMAS 285
39. Peyrard M, Seroussi E, Sandberg-Nordqvist AC, et al. The human 56. Muller P, Henn W, Niedermayer I, et al. Deletion of chromosome
LARGE gene from 22q12.3-q13.1 is a new, distinct member of the 1p and loss of expression of alkaline phosphatase indicate progres-
glycosyltransferase gene family. Proc Natl Acad Sci U S A 1999; sion of meningiomas. Clin Cancer Res 1999;5:3569–77
96:598–603 57. Lomas J, Bello MJ, Arjona D, et al. Analysis of p73 gene in me-
40. Gutmann DH, Donahoe J, Perry A, et al. Loss of DAL-1, a protein ningiomas with deletion at 1p. Cancer Genet Cytogenet 2001;129:
4.1-related tumor suppressor, is an important early event in the 88–91
pathogenesis of meningiomas. Hum Mol Genet 2000;9:1495–1500 58. Nozaki M, Tada M, Kashiwazaki H, et al. p73 is not mutated in
41. Perry A, Cai DX, Scheithauer BW, et al. Merlin, DAL-1, and pro- meningiomas as determined with a functional yeast assay but p73 ex-
gesterone receptor expression in clinicopathologic subsets of me- pression increases with tumor grade. Brain Pathol 2001;11:296–305
ningioma: A correlative immunohistochemical study of 175 cases. 59. Sulman EP, Dumanski JP, White PS, et al. Identification of a con-
J Neuropathol Exp Neurol 2000;59:872–79 sistent region of allelic loss on 1p32 in meningiomas: Correlation
42. Weber RG, Bostrom J, Wolter M, et al. Analysis of genomic alter- with increased morbidity. Cancer Res 1998;58:3226–30
ations in benign, atypical, and anaplastic meningiomas: Toward a 60. Bello MJ, de Campos JM, Vaquero J, Kusak ME, Sarasa JL, Rey
genetic model of meningioma progression. Proc Natl Acad Sci U JA. High-resolution analysis of chromosome arm 1p alterations in
S A 1997;94:14719–24 meningioma. Cancer Genet Cytogenet 2000;120:30–36
43. Buschges R, Bostrom J, Wolter M, et al. Analysis of human me- 61. Zang KD. Meningioma: A cytogenetic model of a complex benign
ningiomas for aberrations of the MADH2, MADH4, APM-1 and human tumor, including data on 394 karyotyped cases. Cytogenet
DCC tumor suppressor genes on the long arm of chromosome 18. Cell Genet 2001;93:207–20
Int J Cancer 2001;92:551–54 62. Tse JY, Ng HK, Lau KM, Lo KW, Poon WS, Huang DP. Loss of
44. Bello MJ, de Campos JM, Kusak ME, et al. Allelic loss at 1p is heterozygosity of chromosome 14q in low- and high-grade menin-
associated with tumor progression of meningiomas. Genes Chro- giomas. Hum Pathol 1997;28:779–85
63. Menon AG, Rutter JL, von Sattel JP, et al. Frequent loss of chro-
mosomes Cancer 1994;9:296–98
mosome 14 in atypical and malignant meningioma: Identification
45. Simon M, von Deimling A, Larson JJ, et al. Allelic losses on chro-
of a putative ‘tumor progression’ locus. Oncogene 1997;14:611–16
mosomes 14, 10, and 1 in atypical and malignant meningiomas: A
64. Maillo A, Orfao A, Sayagues JM, et al. New classification scheme
genetic model of meningioma progression. Cancer Res 1995;55:
for the prognostic stratification of meningioma on the basis of chro-
4696–4701
mosome 14 abnormalities, patient age, and tumor histopathology. J
46. Bostrom J, Muhlbauer A, Reifenberger G. Deletion mapping of the
Clin Oncol 2003;21:3285–95
short arm of chromosome 1 identifies a common region of deletion
65. Rempel SA, Schwechheimer K, Davis RL, Cavenee WK, Rosen-
distal to D1S496 in human meningiomas. Acta Neuropathol (Berl)
blum ML. Loss of heterozygosity for loci on chromosome 10 is
1997;94:479–85
associated with morphologically malignant meningioma progres-
47. Leone PE, Bello MJ, de Campos JM, et al. NF2 gene mutations
sion. Cancer Res 1993;53:2386–92
and allelic status of 1p, 14q and 22q in sporadic meningiomas.
66. Peters N, Wellenreuther R, Rollbrocker B, et al. Analysis of the
Oncogene 1999;18:2231–39
PTEN gene in human meningiomas. Neuropathol Appl Neurobiol
48. Lamszus K, Kluwe L, Matschke J, Meissner H, Laas R, Westphal
1998;24:3–8
M. Allelic losses at 1p, 9q, 10q, 14q, and 22q in the progression 67. von Deimling A, Fimmers R, Schmidt MC, et al. Comprehensive
of aggressive meningiomas and undifferentiated meningeal sarco- allelotype and genetic anaysis of 466 human nervous system tu-
mas. Cancer Genet Cytogenet 1999;110:103–10 mors. J Neuropathol Exp Neurol 2000;59:544–58
49. Bostrom J, Meyer-Puttlitz B, Wolter M, et al. Alterations of the 68. Mihaila D, Jankowski M, Gutierrez JA, et al. Meningiomas: Loss
tumor suppressor genes CDKN2A (p16[INK4a]), p14(ARF), of heterozygosity on chromosome 10 and marker-specific correla-
CDKN2B (p15(INK4b), and CDKN2C (p18[INK4c]) in atypical tions with grade, recurrence, and survival. Clin Cancer Res 2003;
and anaplastic meningiomas. Am J Pathol 2001;159:661–69 9:4443–51
50. Cai DX, Banerjee R, Scheithauer BW, Lohse CM, Kleinschmidt- 69. Mihaila D, Gutierrez JA, Rosenblum ML, Newsham IF, Bogler O,
Demasters BK, Perry A. Chromosome 1p and 14q FISH analysis Rempel SA. Meningiomas: Analysis of loss of heterozygosity on
in clinicopathologic subsets of meningioma: Diagnostic and prog- chromosome 10 in tumor progression and the delineation of four
nostic implications. J Neuropathol Exp Neurol 2001;60:628–36 regions of chromosomal deletion in common with other cancers.
51. Santarius T, Kirsch M, Nikas DC, Imitola J, Black PM. Molecular Clin Cancer Res 2003;9:4435–42
analysis of alterations of the p18INK4c gene in human meningio- 70. Bostrom J, Cobbers JM, Wolter M, et al. Mutation of the PTEN
mas. Neuropathol Appl Neurobiol 2000;26:67–75 (MMAC1) tumor suppressor gene in a subset of glioblastomas but
52. Leuraud P, Marie Y, Robin E, et al. Frequent loss of 1p32 region not in meningiomas with loss of chromosome arm 10q. Cancer Res
but no mutation of the p18 tumor suppressor gene in meningiomas. 1998;58:29–33
J Neurooncol 2000;50:207–13 71. Joachim T, Ram Z, Rappaport ZH, et al. Comparative analysis of
53. Mendiola M, Bello MJ, Alonso J, et al. Search for mutations of the the NF2, TP53, PTEN, KRAS, NRAS and HRAS genes in sporadic
hRAD54 gene in sporadic meningiomas with deletion at 1p32. Mol and radiation-induced human meningiomas. Int J Cancer 2001;94:
Carcinog 1999;24:300–304 218–21
54. Leone PE, Mendiola M, Alonso J, Paz-y-Mino C, Pestana A. Im- 72. Perry A, Banerjee R, Lohse CM, Kleinschmidt-DeMasters BK,
plications of a RAD54L polymorphism (2290C/T) in human me- Scheithauer BW. A role for chromosome 9p21 deletions in the ma-
ningiomas as a risk factor and/or a genetic marker. BMC Cancer lignant progression of meningiomas and the prognosis of anaplastic
2003;3:6 meningiomas. Brain Pathol 2002;12:183–90
55. Niedermayer I, Feiden W, Henn W, Steilen-Gimbel H, Steudel WI, 73. Simon M, Park TW, Koster G, et al. Alterations of INK4a(p16-
Zang KD. Loss of alkaline phosphatase activity in meningiomas: p14ARF)/INK4b(p15) expression and telomerase activation in me-
A rapid histochemical technique indicating progression-associated ningioma progression. J Neurooncol 2001;55:149–58
deletion of a putative tumor suppressor gene on the distal part of 74. Ohgaki H, Eibl RH, Schwab M, et al. Mutations of the p53 tumor
the short arm of chromosome 1. J Neuropathol Exp Neurol 1997; suppressor gene in neoplasms of the human nervous system. Mol
56:879–86 Carcinog 1993;8:74–80

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018
286 LAMSZUS

75. Wang JL, Zhang ZJ, Hartman M, et al. Detection of TP53 gene subunit, hTERT, in meningioma progression. J Neurosurg 2000;92:
mutation in human meningiomas: A study using immunohistochem- 832–40
istry, polymerase chain reaction/single-strand conformation poly- 90. Falchetti ML, Larocca LM, Pallini R. Telomerase in brain tumors.
morphism and DNA sequencing techniques on paraffin-embedded Childs Nerv Syst 2002;18:112–17
samples. Int J Cancer 1995;64:223–28 91. Bitzer M, Wockel L, Luft AR, et al. The importance of pial blood
76. Perry A, Stafford SL, Scheithauer BW, Suman VJ, Lohse CM. The supply to the development of peritumoral brain edema in menin-
prognostic significance of MIB-1, p53, and DNA flow cytometry giomas. J Neurosurg 1997;87:368–73
in completely resected primary meningiomas. Cancer 1998;82: 92. Goldman CK, Bharara S, Palmer CA, et al. Brain edema in menin-
2262–69 giomas is associated with increased vascular endothelial growth
77. Matsuno A, Nagashima T, Matsuura R, et al. Correlation between factor expression. Neurosurgery 1997;40:1269–77
MIB-1 staining index and the immunoreactivity of p53 protein in 93. Provias J, Claffey K, delAguila L, Lau N, Feldkamp M, Guha A.
recurrent and non-recurrent meningiomas. Am J Clin Pathol 1996; Meningiomas: Role of vascular endothelial growth factor/vascular
106:776–81 permeability factor in angiogenesis and peritumoral edema. Neu-
78. Karamitopoulou E, Perentes E, Tolnay M, Probst A. Prognostic rosurgery 1997;40:1016–26
significance of MIB-1, p53, and bcl-2 immunoreactivity in menin- 94. Bitzer M, Opitz H, Popp J, et al. Angiogenesis and brain oedema
giomas. Hum Pathol 1998;29:140–45 in intracranial meningiomas: Influence of vascular endothelial
79. Buschges R, Ichimura K, Weber RG, Reifenberger G, Collins VP. growth factor. Acta Neurochir (Wien) 1998;140:333–40
Allelic gain and amplification on the long arm of chromosome 17 95. Samoto K, Ikezaki K, Ono M, et al. Expression of vascular endo-
in anaplastic meningiomas. Brain Pathol 2002;12:145–53 thelial growth factor and its possible relation with neovasculariza-
80. Cai DX, James CD, Scheithauer BW, Couch FJ, Perry A. PS6K tion in human brain tumors. Cancer Res 1995;55:1189–93
amplification characterizes a small subset of anaplastic meningio- 96. Lamszus K, Lengler U, Schmidt NO, Stavrou D, Ergun S, Westphal
mas. Am J Clin Pathol 2001;115:213–18 M. Vascular endothelial growth factor, hepatocyte growth factor/
81. Lekanne-Deprez RH, Riegman PH, van Drunen E, et al. Cytoge- scatter factor, basic fibroblast growth factor, and placenta growth
netic, molecular genetic and pathological analyses in 126 menin- factor in human meningiomas and their relation to angiogenesis and
giomas. J Neuropathol Exp Neurol 1995;54:224–35 malignancy. Neurosurgery 2000;46:938–47
82. Carlson KM, Bruder C, Nordenskjold M, Dumanski JP. 1p and 3p 97. Yamasaki F, Yoshioka H, Hama S, Sugiyama K, Arita K, Kurisu
deletions in meningiomas without detectable aberrations of chro- K. Recurrence of meningiomas. Cancer 2000;89:1102–10
mosome 22 identified by comparative genomic hybridization. 98. Black P, Carroll R, Zhang J. The molecular biology of hormone
Genes Chromosomes Cancer 1997;20:419–24 and growth factor receptors in meningiomas. Acta Neurochir Suppl
83. Sayagues JM, Tabernero MD, Maillo A, et al. Incidence of numer- (Wien) 1996;65:50–53
ical chromosome aberrations in meningioma tumors as revealed by 99. Sanson M, Cornu P. Biology of meningiomas. Acta Neurochir
fluorescence in situ hybridization using 10 chromosome-specific (Wien) 2000;142:493–505
probes. Cytometry 2002;50:153–59 100. McCutcheon IE. The biology of meningiomas. J Neurooncol
84. Sato K, Schauble B, Kleihues P, Ohgaki H. Infrequent alterations 1996;29:207–16
of the p15, p16, CDK4 and cyclin D1 genes in non-astrocytic hu- 101. Carroll RS, Glowacka D, Dashner K, Black PM. Progesterone
man brain tumors. Int J Cancer 1996;66:305–8 receptor expression in meningiomas. Cancer Res 1993;53:1312–16
85. Tse JY, Ng HK, Lo KW, et al. Analysis of cell cycle regulators: 102. Carroll RS, Zhang J, Dashner K, Sar M, Wilson EM, Black PM.
p16INK4A, pRb, and CDK4 in low- and high-grade meningiomas. Androgen receptor expression in meningiomas. J Neurosurg 1995;
Hum Pathol 1998;29:1200–1207 82:453–60
86. Shoshan Y, Chernova O, Juen SS, et al. Radiation-induced menin- 103. Hsu DW, Efird JT, Hedley-Whyte ET. Progesterone and estrogen
gioma: A distinct molecular genetic pattern? J Neuropathol Exp receptors in meningiomas: Prognostic considerations. J Neurosurg
Neurol 2000;59:614–20 1997;86:113–20
87. Rajcan-Separovic E, Maguire J, Loukianova T, Nisha M, Kalousek 104. Ragel B, Jensen RL. New approaches for the treatment of refrac-
D. Loss of 1p and 7p in radiation-induced meningiomas identified tory meningiomas. Cancer Control 2003;10:148–58
by comparative genomic hybridization. Cancer Genet Cytogenet 105. Watson MA, Gutmann DH, Peterson K, et al. Molecular charac-
2003;144:6–11 terization of human meningiomas by gene expression profiling
88. Langford LA, Piatyszek MA, Xu R, Schold SC, Jr., Wright WE, using high-density oligonucleotide microarrays. Am J Pathol
Shay JW. Telomerase activity in ordinary meningiomas predicts 2002;161:665–72
poor outcome. Hum Pathol 1997;28:416–20 106. Fathallah-Shaykh HM, He B, Zhao LJ, et al. Genomic expression
89. Simon M, Park TW, Leuenroth S, Hans VH, Loning T, Schramm discovery predicts pathways and opposing functions behind phe-
J. Telomerase activity and expression of the telomerase catalytic notypes. J Biol Chem 2003;278:23830–33

J Neuropathol Exp Neurol, Vol 63, April, 2004

Downloaded from https://academic.oup.com/jnen/article-abstract/63/4/275/2916476


by guest
on 31 July 2018

You might also like