Sie sind auf Seite 1von 6

Experimental and Molecular Pathology 99 (2015) 354359

Contents lists available at ScienceDirect

Experimental and Molecular Pathology

journal homepage: www.elsevier.com/locate/yexmp

Review

Malignant progression to anaplastic meningioma: Neuropathology,


molecular pathology, and experimental models
Patrick J. Cimino
Department of Pathology, Division of Neuropathology, University of Washington, Box 359791, 325 9th Avenue, Seattle, WA 98104-2499, United States

a r t i c l e i n f o a b s t r a c t

Article history: Meningioma is a common adult intracranial tumor, and while several cases are considered benign, a subset is
Received 16 August 2015 malignant with biologically aggressive behavior and is refractory to current treatment strategies of combined
Accepted 17 August 2015 surgery and radiotherapy. Anaplastic meningiomas are quite aggressive and correspond to a World Health
Available online 22 August 2015
Organization (WHO) Grade III tumor. This highly aggressive phenotype mandates the need for more efcacious
therapies. Designing rational therapies for treatment will have its foundation in the biologic understanding of
Keywords:
Meningioma
involved genes and molecular pathways in these types of tumors. Anaplastic meningiomas (WHO Grade III)
Anaplastic meningioma can arise from malignant transformation of lower grade (WHO Grade I/II) tumors, however there is an incom-
Malignant progression plete understanding of specic genetic drivers of malignant transformation in these tumors. Here, the current
Neuropathology understanding of anaplastic meningiomas is reviewed in the context of human neuropathologic specimens
and small animal models.
2015 Elsevier Inc. All rights reserved.

Contents

1. Anaplastic meningioma introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 354


1.1. Clinical characteristics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 354
1.2. Histopathology and immunohistochemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
2. Genes and chromosomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
2.1. Genetics of lower grade meningioma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
2.2. Genomic changes associated with malignancy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356
3. Small animal models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 357
4. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
Grant numbers/source of support . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
Disclosure/conict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358

1. Anaplastic meningioma introduction grade WHO Grade I; Highest grade WHO Grade III) (McGovern
et al., 2010; Perry et al., 2007; Sun et al., 2015). Anaplastic meningioma
1.1. Clinical characteristics (WHO Grade III) is a malignant tumor with variable reports of median
overall survival, with some series ndings ranging from 1.5 to 3.5 years
Meningioma is the most common primary intracranial tumor in the (Champeaux et al., 2015; Moliterno et al., 2015; Perry et al., 1999). Ana-
adult population, and is believed to arise from the arachnoidal cap cells plastic meningiomas can present either as primary tumors or those aris-
of the leptomeninges (Perry et al., 2007; Wiemels et al., 2010). Many ing from malignant transformation of lower grade (WHO Grade I/II)
meningiomas are considered benign and amenable to surgical resection, meningiomas. Treatment of primary and recurrent anaplastic meningio-
however a subset demonstrates aggressive biologic behavior and is ma consists primarily of gross total or near-total surgical resection
challenging to manage, based on characteristics such as tumor location combined with various types of external beam radiotherapies (Sun
and increasing World Health Organization Grade (WHO) (Lowest et al., 2015; Walcott et al., 2013). The age of b60 years and the use
of radiotherapy has been associated with longer overall survival in
anaplastic meningioma (Durand et al., 2009). The utility of various
E-mail address: pjjc@uw.edu. targeted chemotherapies, such as somatostatin analogues, for

http://dx.doi.org/10.1016/j.yexmp.2015.08.007
0014-4800/ 2015 Elsevier Inc. All rights reserved.
P.J. Cimino / Experimental and Molecular Pathology 99 (2015) 354359 355

meningioma management are actively under investigation (Chamberlain, specic diagnostic marker for meningiomas of all grades, and appears
2012; Kaley et al., 2015; Norden et al., 2015). Poor prognostic indicators of superior to EMA and PR, especially in cases of anaplastic meningioma
anaplastic meningioma include subtotal resection (increasing Simpson (Menke et al., 2015).
Grade), posterior fossa or skull base location, tumors that arise from
lower WHO Grade I/II tumors (compared to De Novo occurrence), 2. Genes and chromosomes
increasing mitotic activity, and loss of chromosomal region 9p21
(region includes the cyclin-dependent kinase inhibitor genes CDKN2A 2.1. Genetics of lower grade meningioma
[encodes p16INK4A and p14 ARF ] and CDKN2B [encodes p15INK4B ])
(Bostrom et al., 2001; Moliterno et al., 2015; Olar et al., 2015; The most common cytogenetic changes in meningioma, regardless
Perry et al., 2002). of grade, include alterations of chromosome 22 (monosomy or 22q par-
tial loss), which occur in approximately half of all sporadic cases
1.2. Histopathology and immunohistochemistry (Dumanski et al., 1987; Dumanski et al., 1990; Ruttledge et al., 1994;
Seizinger et al., 1987; Zang, 2001). The well-studied tumor suppressor
Meningiomas are currently diagnosed and classied according to gene NF2, resides on chromosome 22q, and is altered itself in about
specic histological criteria, as set forth by the WHO in 2007 (Table 1) half of all cases of sporadic meningioma (Harada et al., 1996;
(Perry et al., 2007). Diagnostic features of anaplastic meningioma pres- Ruttledge et al., 1994). There are cases where 22q loss is observed in
ent in routine hematoxylin and eosin-stained slides include either cyto- the absence of NF2 gene mutations, postulating that there is at least
logic anaplasia (carcinoma-, sarcoma-, or melanoma-like histology) or one yet undiscovered gene in this chromosomal region responsible for
an increased mitotic rate of at least 20 mitotic gures per 10 consecutive meningioma tumorigenesis (Ng et al., 1995). The importance of the
high powered microscopic elds (Table 1). Rare cases of anaplastic me- NF2 gene mutations in meningioma is highlighted by the fact that ap-
ningioma are reported to have areas of metaplastic adenocarcinoma- proximately half of individuals with the tumor predisposition syndrome
like histomorphology (Patil et al., 2011). In challenging diagnostic Neurobromatosis Type 2 (NF2), resultant from germline mutations in
cases, immunohistochemical staining can help to support a diagnosis the NF2 gene, develop meningiomas (Asthagiri et al., 2009; Goutagny
of anaplastic meningioma. Like meningiomas of all grades, there is et al., 2012; Sanson et al., 1993). The NF2 gene encodes for a member
patchy positive immunoreactivity for Epithelial Membrane Antigen of the 4.1 protein superfamily, merlin, that acts as a linker protein to me-
(EMA), as well as some immunopositivity for Prostaglandin D Synthase diate interactions between the cell membrane and cytoskeleton
and limited keratins (AE1/AE3, CK7, CK8, CK14, CK18, CK19) in anaplas- (MacCollin et al., 1993; Trofatter et al., 1993). Two additional 4.1 super-
tic meningioma (Fig. 1) (Liu et al., 2004; Miettinen and Paetau, 2002; family members, DAL-1/4.1B and 4.1R, are also altered in a subset of
Rajaram et al., 2004; Yamashima et al., 1997). Less specic antigens de- lower grade meningiomas (Gutmann et al., 2000; Perry et al., 2000;
tected immunohistochemically in meningiomas can include vimentin, Robb et al., 2003). The protein Tumor Suppressor in Lung Cancer 1
Epidermal Growth Factor Receptor (EGFR), S100, and Sox10 (Jitawi (TLSC1), which can interact with DAL-1/4.1B, is also altered early in
et al., 1988; Liu et al., 2004; Ng et al., 2015; Wernicke et al., 2010). Com- some meningiomas, and loss of immunoreactivity increases in frequen-
pared to lower grade meningiomas, Progesterone Receptor (PR) nuclear cy with increasing WHO grade (Busam et al., 2011; Surace et al., 2004).
immunoreactivity tends to be lost in anaplastic meningioma, as illus- Recent large scale whole genome and exome sequencing efforts of
trated in the case example in Fig. 1 (Bouillot et al., 1994; Brandis et al., WHO Grade I and II meningiomas have identied recurrent non-NF2
1993; Hilbig and Barbosa-Coutinho, 1998; Hsu et al., 1997; Nagashima gene mutations, including Tumor Necrosis Factor Receptor-Associated
et al., 1995; Perry et al., 2000). As opposed to PR, there are other pro- Factor 7 (TRAF7), Smoothened (SMO), Kruppel-Like Factor 4 (KLF4), and
teins that appear to have increased expression in anaplastic meningio- v-Akt Murine Thymoma Viral Oncogene Homolog 1 (AKT1) (Brastianos
ma when compared to lower grade tumors, such as the apoptosis et al., 2013; Clark et al., 2013; Reuss et al., 2013). Mutations in these
inhibitor TRAIL-R4, the immune modulator PD-L1 (CD274), DNA Topo- four genes in meningioma appear mutually exclusive to NF2 alterations.
isomerase II alpha, and Fatty Acid Synthase (Du et al., 2015; Haase et al., The combination of gene mutations in TRAF7 and KLF4 (p.K409Q) seem
2010; Koschny et al., 2015; Liu et al., 2004; Roessler et al., 2002). So- to characterize secretory meningioma (WHO Grade I) (Reuss et al.,
matostatin receptor 2a (SSTR2) is emerging as a highly sensitive and 2013). While both NF2 and non-NF2 gene mutations are seen in WHO

Table 1
Meningioma classication according to current 2007 World Health Organization (WHO) diagnostic criteria.

WHO grade Diagnostic histomorphological characteristics

I Predominant histologic variant

Meningothelial
Fibrous
Transitional
Psammomatous
Secretory
Microcystic
Angiomatous
Lymphoplasmacyte-rich
Metaplastic
II Any grade I histologic variant Or Atypia by elevated mitoses Or 3 atypical features, including: Or Predominant histologic variant
with brain invasion (4 to 19 mitoses per 10 high powered elds)
Hypercellularity Clear cell
Sheet-like growth Chordoid
Macro-nucleoli
Small cell change
Spontaneous necrosis
III Frank anaplasia Or Anaplasia by elevated mitoses Or Predominant histologic variant
(Sarcoma-, carcinoma-, or (20 mitoses per 10 high powered elds)
Rhabdoid
melanoma-like histology)
Papillary
356 P.J. Cimino / Experimental and Molecular Pathology 99 (2015) 354359

Fig. 1. Case example of malignant progression to anaplastic meningioma from a benign meningioma. The patient is initially presented with an intracranial benign transitional type
meningioma (up to 1 mitosis per 10 high powered elds). The tumor recurred as an atypical meningioma with increased mitotic activity (up to 15 mitoses per 10 high powered elds)
and several atypical features (necrosis was not evaluable due to prior external beam radiotherapy treatment). Three years following initial presentation and multiple treatments, the tumor
recurred as an anaplastic meningioma with up to 23 mitoses per 10 high powered elds (arrows). Typical for the course of malignant transformation, the recurrent tumors retained patchy
cytoplasmic immunoreactivity for EMA and lost nuclear reactivity of PR. (Abbreviations: WHO World Health Organization, H&E Hematoxylin and Eosin, EMA Epithelial Membrane
Antigen, PR Progesterone Receptor.)

Grade I and Grade II meningiomas, atypical meningiomas (WHO Grade subset of anaplastic meningiomas lack NF2 gene mutations, and it is
II) are relatively enriched for NF2 mutations (and Chromosome 22 loss), possible that there are yet unidentied genetic drivers that predispose
suggesting clinical relevance to this genomic divide (Clark et al., 2013; for, or drive, malignant progression.
Reuss et al., 2013). Although there is limited targeted sequencing data
available, TRAF7, SMO, or KLF4, gene mutations have not yet been re- 2.2. Genomic changes associated with malignancy
ported in anaplastic meningioma in the current literature. Larger data
sets will be needed to determine the true mutational frequency of Recurrent chromosomal abnormalities accumulate and characterize
these genes in anaplastic meningioma. In an initial report, only a single meningiomas with increasing WHO grade (Al-Mefty et al., 2004; Perry
case of anaplastic meningioma (out of three total cases) demonstrated and Brat, 2010). As discussed above, chromosome 22 abnormalities
an AKT1 (p.E17K) gene mutation (Brastianos et al., 2013). However, a are early and frequent occurrences in WHO Grade I meningiomas,
larger targeted sequencing study of 86 anaplastic meningiomas failed which themselves tend not to demonstrate additional cytogenetic ab-
to detect AKT1 gene mutations in any case (Sahm et al., 2013). In con- normalities. In addition to chromosome 22 abnormalities, recurrent
trast to the relative lack of mutations in these four non-NF2 genes, NF2 gains (1q, 9q, 12q, 15q, 17q, and 20q) and losses (1p, 6q, 10, 14q, and
gene mutations (and 22q loss) are frequently found in anaplastic me- 18q) are variably found in WHO Grade II atypical meningiomas (Perry
ningioma, indicating that NF2 may have some role in at least predispos- and Brat, 2010). WHO Grade III anaplastic meningiomas demonstrate
ing meningiomas for malignant progression relative to TRAF7, AKT1, further chromosomal instability with additional chromosomal losses
KLF4, and SMO (Al-Mefty et al., 2004; Nunes et al., 2005; Perry et al., (1p, 6q, 9p21, 10, 14q, and 18q) and 17q23 amplication (region con-
2001; Rajaram et al., 2004; Reuss et al., 2013). However, a signicant taining the Ribosomal Protein S6 Kinase Polypeptide 1 [RPS6KB1] gene)
P.J. Cimino / Experimental and Molecular Pathology 99 (2015) 354359 357

(Perry and Brat, 2010). Of these chromosomal abnormalities, loss of

Kalamarides et al. (2002),


Kalamarides et al. (2008),
Kalamarides et al. (2011)
Kalamarides et al. (2011)
Kalamarides et al. (2011)

Kalamarides et al. (2002)


Kalamarides et al. (2008)
9p21 (region includes genes CDKN2A [encodes p16INK4A and p14ARF]
and CDKN2B [encodes p15INK4B]) occurs in about 70% of anaplastic

Peyre et al. (2013)

Peyre et al. (2013)

Peyre et al. (2013)


meningiomas and is an important poor prognosticator that portends
signicantly reduced overall survival (Bostrom et al., 2001; Perry

Reference
et al., 2002). The mechanism of chromosomal instability in meningi-
oma progression is unclear, however may in part be contributed by
Checkpoint Kinase 2 (CHK2), which is involved in DNA damage re-
sponse pathways (Antoni et al., 2007). The encoding CHEK2 gene is
located on 22q, near NF2, and is alternatively spliced or deleted
(often co-deleted with NF2) in some meningiomas (Yang et al.,

Genetically engineered mouse (GEM) models related to meningioma progression. Abbreviations: Ad-Cre, Adenovirus-Cre leptomeningeal injections at postnatal day 2 (P2); PGDS, Prostaglandin D Synthase.
2012). In vitro studies using human meningioma cell lines conrm

Skull base, cerebral convexities, spine

Skull base, cerebral convexities, spine


Skull base, cerebral convexities, spine
a functional role for CHK2 in centrosome maintenance and chromo-
somal stability (Yang et al., 2012).
In addition to CDKN2A/B, relatively few gene level alterations have

Cerebral convexities, spine

Cerebral convexities, spine


been strongly associated with malignant progression of meningioma,
and include Telomerase Reverse Transcriptase (TERT) and N-myc
Downstream-Regulated Gene family member 2 (NDRG2). Telomerase ex-

Tumor location
pression and activity predicts poor clinical behavior, and increases in
atypical and anaplastic meningiomas (DeMasters et al., 1997; Langford

Skull base
Skull base
Skull base
et al., 1997; Simon et al., 2000). The mechanism of increased telomerase
expression is associated with activating mutations in the promoter of
the TERT gene (Goutagny et al., 2014). Activating TERT promoter muta-
tions are detected in increasing frequency in high grade meningiomas
and low grade meningiomas that subsequently undergo malignant pro-
gression (Goutagny et al., 2014). Integrative genomics, coupled with
immunohistochemical studies, identied NDRG2 as a tumor suppressor

14% Grade II, and 11% Grade III)


50% (composed of 75% Grade I,

72% (composed of 66% Grade I,


31% Grade II, and 3% Grade III)
that is frequently downregulated in anaplastic meningioma (Lusis et al.,
2005). NDRG2 is also found on chromosome 14q, which is recurrently
lost in anaplastic meningioma. Downregulation of NDRG2 is strongly as-
sociated with hypermethylation of the NDRG2 gene promoter, which is
present in approximately 70% of anaplastic meningioma. Hypermethy- Tumor frequency
lation of gene promoters other than NDRG2 occur frequently in atypical

1348%
and anaplastic meningioma, indicating that this mechanism of gene reg-
38%
43%
50%

12%
37%
ulation may play an important role in meningioma progression (Liu
et al., 2005; Murnyak et al., 2015). An additional gene with potential
contribution to malignant progression is the non-coding RNA (ncRNA)
gene, Maternally Expressed Gene 3 (MEG3), which is found on chromo-
somal region 14q32 that is recurrently deleted in anaplastic meningio-
Grade I: Meningothelial, brous, and transitional

Grade I: Meningothelial, brous, and transitional


Grade I: Meningothelial, brous, and transitional

Grade I: Meningothelial, brous, and transitional


ma (Zhang et al., 2010). A pilot study identied several meningiomas
Grade I: Meningothelial, not otherwise specied

to have decreased MEG3 expression (associated with promoter hyper-


Grade II: Brain invasion or increased mitoses

Grade II: Brain invasion or increased mitoses


methylation), as well as MEG3 allelic loss to occur frequently in higher
grade meningiomas (Zhang et al., 2010). Overall, the complexity of ge-
Grade I: Meningothelial and brous

Grade I: Meningothelial and brous

nomic alterations (cytogenetic, genetic, and epigenetic) already demon-


Grade III: Sarcoma-like histology

Grade III: Sarcoma-like histology


strated during the malignant progression of meningioma, dictates the
need for integrative genomic techniques to identify and characterize ad-
ditional pathogenic genes.

3. Small animal models


Grade I: Fibrous
Histology/grade

Initial mouse models of all grades of meningioma largely relied on


xenografts (orthotopic and heterotopic) of human meningioma-
derived cells into immunocompromised mice. The malignant human
meningioma cell lines IOMM-Lee and CH-157MN have most often
been used (recently reviewed in Kalamarides et al. (2010)). These trans-
plantation models do have some desirable features for studying menin-
giomas including recapitulating human histopathology, the ability to
Ad-Cre; Nf2ox2/ox2; Ink4ab+/

Ad-Cre; Nf2ox2/ox2; Ink4ab/


PGDS-Cre; Nf2ox2/; Tp53ox/
PGDS-Cre; Nf2ox2/; Ink4a/

Ad-Cre; Nf2ox2/ox2; Ink4a/

label cells and image them with bioluminescence, and demonstrating


Ad-Cre; Nf2ox2/ox2; Tp53+/

reproducible take rate and growth characteristics (Ragel et al.,


2008). However, there are challenges to interpreting xenograft nd-
ox2/

Ad-Cre; Nf2ox2/ox2

ings due to conditions that do not reect human tumors, including


altered tumor microenvironment in immunocompromised host mice,
PGDS-Cre; Nf2

tumor cells undergoing undetermined genetic and cellular selection relat-


ed to in vitro cell culture prior to transplantation, and tumors growing in
Table 2

GEM

abnormal inltrative growth patterns. To overcome these obstacles, re-


cent focus has turned to genetically engineered mouse (GEM) models of
358 P.J. Cimino / Experimental and Molecular Pathology 99 (2015) 354359

meningioma, which offer the advantages of in vivo spatial and temporal Grant numbers/source of support
control of gene manipulation.
GEM models of meningioma based on Cre-loxP-mediated inacti- This work was supported with departmental funds provided by the
vation the Nf2 gene have been successfully employed and have pro- University of Washington Department of Pathology.
vided some insight into the biology of initiation and progression of
meningioma (models summarized in Table 2). Conditional inactiva-
Disclosure/conict of interest
tion of the Nf2 gene (Nf2ox2/ox2 mice), either with Cre recombinase
under control of the endogenous Prostaglandin D Synthase (PGDS)
The author declares that there are no conicts of interest.
promoter or delivered exogenously via postnatal adenoviral injec-
tion into the leptomeninges, is sufcient to initiate and maintain
low grade (WHO Grade I) meningioma with variable penetrance References
(Kalamarides et al., 2002; Kalamarides et al., 2011; Kalamarides
Al-Mefty, O., et al., 2004. Malignant progression in meningioma: documentation of a
et al., 2008). Consistent with the observation that TP53 gene muta-
series and analysis of cytogenetic ndings. J. Neurosurg. 101, 210218.
tions are exceedingly rare in human meningioma, the addition of Antoni, L., et al., 2007. CHK2 kinase: cancer susceptibility and cancer therapy two sides
Tp53 hemizygosity to Nf2 inactivation in mice does not increase of the same coin? Nat. Rev. Cancer 7, 925936.
frequency of tumor formation or lead to malignant progression Asthagiri, A.R., et al., 2009. Neurobromatosis type 2. Lancet 373, 19741986.
Bostrom, J., et al., 2001. Alterations of the tumor suppressor genes CDKN2A (p16(INK4a)),
of meningioma (Joachim et al., 2001; Kalamarides et al., 2002; p14(ARF), CDKN2B (p15(INK4b)), and CDKN2C (p18(INK4c)) in atypical and ana-
Kalamarides et al., 2011). Concomitant Ink4a/p16Ink4a nullizygosity plastic meningiomas. Am. J. Pathol. 159, 661669.
in adenovirus injected mice signicantly increases the frequency of Bouillot, P., et al., 1994. Quantitative imaging of estrogen and progesterone receptors,
estrogen-regulated protein, and growth fraction: immunocytochemical assays in 52
meningioma formation, however it does not lead malignant pro- meningiomas. Correlation with clinical and morphological data. J. Neurosurg. 81,
gression (Kalamarides et al., 2008). However, loss of one or both 765773.
copies of the entire Cdkn2ab/Ink4ab gene locus (encoding the three Brandis, A., et al., 1993. Immunohistochemical detection of female sex hormone receptors
in meningiomas: correlation with clinical and histological features. Neurosurgery 33,
related genes p16Ink4a, p15Ink4b, and p19ARF) does lead to malignant 212217 (discussion 217-8).
progression in a small subset of meningiomas as demonstrated by Brastianos, P.K., et al., 2013. Genomic sequencing of meningiomas identies oncogenic
the formation of Grade II and III meningiomas (Peyre et al., 2013). SMO and AKT1 mutations. Nat. Genet. 45, 285289.
Busam, R.D., et al., 2011. Structural basis of tumor suppressor in lung cancer 1 (TSLC1)
Additionally, homozygous loss of the Cdkn2ab/Ink4ab gene further
binding to differentially expressed in adenocarcinoma of the lung (DAL-1/4.1B).
increases the frequency of tumor formation (Peyre et al., 2013). J. Biol. Chem. 286, 45114516.
With a majority (66 to 75%) of the meningiomas in these mice oc- Chamberlain, M.C., 2012. The role of chemotherapy and targeted therapy in the treatment
of intracranial meningioma. Curr. Opin. Oncol. 24, 666671.
curring as benign (Grade I) tumors, it appears that the loss of the
Champeaux, C., et al., 2015. World Health Organization grade III meningiomas. A retro-
Cdkn2ab/Ink4ab gene added to bi-allelic Nf2 loss, is insufcient to spective study for outcome and prognostic factors assessment. Br. J. Neurosurg. 1-6.
promote malignant progression in most tumors. Clark, V.E., et al., 2013. Genomic analysis of non-NF2 meningiomas reveals mutations in
When cultured, Grade I and II tumor-derived cells from Nf2ox2/ox2; TRAF7, KLF4, AKT1, and SMO. Science 339, 10771080.
DeMasters, B.K., et al., 1997. Differential telomerase expression in human primary intra-
Ink4ab/; and Ad-Cre mice demonstrate various mouse chromosomal cranial tumors. Am. J. Clin. Pathol. 107, 548554.
abnormalities, including gains (15, 15q, 19q), losses (4q, 7, 12, 14q), Du, Z., et al., 2015. Increased expression of the immune modulatory molecule PD-L1
and structural rearrangements (16 and 19) (Peyre et al., 2013). This (CD274) in anaplastic meningioma. Oncotarget 6, 47044716.
Dumanski, J.P., et al., 1987. Deletion mapping of a locus on human chromosome 22 in-
chromosomal instability mimics genetic abnormalities seen in increas- volved in the oncogenesis of meningioma. Proc. Natl. Acad. Sci. U. S. A. 84, 92759279.
ing grades of human meningioma. Orthotopic transplantation of these Dumanski, J.P., et al., 1990. Molecular genetic analysis of chromosome 22 in 81 cases of
Ad-Cre; Nf2ox2/ox2; and Ink4ab/ meningioma cells into the subdural meningioma. Cancer Res. 50, 58635867.
Durand, A., et al., 2009. WHO grade II and III meningiomas: a study of prognostic factors.
space of wild-type mice produces tumors of all grades, including a subset J. Neuro-Oncol. 95, 367375.
that progresses to Grade III tumors (Peyre et al., 2013). While Goutagny, S., et al., 2012. Long-term follow-up of 287 meningiomas in neurobromatosis
the mechanisms of genomic instability and progression are not type 2 patients: clinical, radiological, and molecular features. Neuro-Oncology 14,
10901096.
completely understood, these mouse models underscore the impor-
Goutagny, S., et al., 2014. High incidence of activating TERT promoter mutations in me-
tance of the synergistic effects of Nf2 and Cdkn2ab/Ink4ab, in combi- ningiomas undergoing malignant progression. Brain Pathol. 24, 184189.
nation with chromosomal instability, to induce malignant progression to Gutmann, D.H., et al., 2000. Loss of DAL-1, a protein 4.1-related tumor suppressor, is an
important early event in the pathogenesis of meningiomas. Hum. Mol. Genet. 9,
anaplastic meningioma. Furthermore, due to their genetic similarity
14951500.
to human meningioma, these small animal models are promising as pre- Haase, D., et al., 2010. Fatty acid synthase as a novel target for meningioma therapy.
clinical platforms to test emerging therapeutic strategies in malignant Neuro-Oncology 12, 844854.
meningioma. Harada, T., et al., 1996. Molecular genetic investigation of the neurobromatosis type 2
tumor suppressor gene in sporadic meningioma. J. Neurosurg. 84, 847851.
Hilbig, A., Barbosa-Coutinho, L.M., 1998. Meningiomas and hormonal receptors. Immuno-
histochemical study in typical and non-typical tumors. Arq. Neuropsiquiatr. 56,
4. Conclusions 193199.
Hsu, D.W., et al., 1997. Progesterone and estrogen receptors in meningiomas: prognostic
considerations. J. Neurosurg. 86, 113120.
Given the associated high morbidity and mortality rates, there is a Jitawi, S.A., et al., 1988. The expression of S-100 protein and neuron-specic enolase in
therapeutic imperative for anaplastic meningioma. Chromosomal insta- meningiomas. Dis. Markers 6, 109117.
Joachim, T., et al., 2001. Comparative analysis of the NF2, TP53, PTEN, KRAS, NRAS and
bility with recurrent cytogenetic alterations is often associated with ma- HRAS genes in sporadic and radiation-induced human meningiomas. Int. J. Cancer.
lignant progression of meningioma. While some genes are known to 94, 218221.
associate with anaplastic meningioma, there is still an incomplete un- Kalamarides, M., et al., 2002. Nf2 gene inactivation in arachnoidal cells is rate-limiting for
meningioma development in the mouse. Genes Dev. 16, 10601065.
derstanding of the mechanism of progression. A multipronged approach
Kalamarides, M., et al., 2010. Meningioma mouse models. J. Neuro-Oncol. 99, 325331.
combining genomic strategies with experimental models will likely lead Kalamarides, M., et al., 2011. Identication of a progenitor cell of origin capable of gener-
to further candidate genes of interest. Small animal models of meningi- ating diverse meningioma histological subtypes. Oncogene 30, 23332344.
Kalamarides, M., et al., 2008. Natural history of meningioma development in mice reveals:
oma progression will allow a preclinical platform to study these genes,
a synergy of Nf2 and p16(Ink4a) mutations. Brain Pathol. 18, 6270.
as well as rapidly test possible targeted therapies aimed at halting Kaley, T.J., et al., 2015. Phase II trial of sunitinib for recurrent and progressive atypical and
malignant progression. Furthermore, a better understanding of the anaplastic meningioma. Neuro-Oncology 17, 116121.
genes involved in malignant progression may lead to better patient Koschny, R., et al., 2015. WHO grade related expression of TRAIL-receptors and apoptosis
regulators in meningioma. Pathol. Res. Pract. 211, 109116.
risk stratication and help guide clinical decisions in the era of pre- Langford, L.A., et al., 1997. Telomerase activity in ordinary meningiomas predicts poor
cision medicine. outcome. Hum. Pathol. 28, 416420.
P.J. Cimino / Experimental and Molecular Pathology 99 (2015) 354359 359

Liu, Y., et al., 2005. Aberrant CpG island hypermethylation prole is associated with atyp- Perry, A., et al., 1999. Malignancy in meningiomas: a clinicopathologic study of 116
ical and anaplastic meningiomas. Hum. Pathol. 36, 416425. patients, with grading implications. Cancer 85, 20462056.
Liu, Y., et al., 2004. Expression of cytokeratin by malignant meningiomas: diagnostic Peyre, M., et al., 2013. Meningioma progression in mice triggered by Nf2 and Cdkn2ab
pitfall of cytokeratin to separate malignant meningiomas from metastatic carcinoma. inactivation. Oncogene 32, 42644272.
Mod. Pathol. 17, 11291133. Ragel, B.T., et al., 2008. A comparison of the cell lines used in meningioma research. Surg.
Lusis, E.A., et al., 2005. Integrative genomic analysis identies NDRG2 as a candidate Neurol. 70, 295307 (discussion 307).
tumor suppressor gene frequently inactivated in clinically aggressive meningioma. Rajaram, V., et al., 2004. Anaplastic meningioma versus meningeal hemangiopericytoma:
Cancer Res. 65, 71217126. immunohistochemical and genetic markers. Hum. Pathol. 35, 14131418.
MacCollin, M., et al., 1993. DNA diagnosis of neurobromatosis 2. Altered coding sequence Reuss, D.E., et al., 2013. Secretory meningiomas are dened by combined KLF4 K409Q and
of the merlin tumor suppressor in an extended pedigree. JAMA 270, 23162320. TRAF7 mutations. Acta Neuropathol. 125, 351358.
McGovern, S. L., et al., 2010. A comparison of World Health Organization tumor grades at Robb, V.A., et al., 2003. Identication of a third protein 4.1 tumor suppressor, protein 4.1R,
recurrence in patients with non-skull base and skull base meningiomas. J. Neurosurg. in meningioma pathogenesis. Neurobiol. Dis. 13, 191202.
112, 92533. Roessler, K., et al., 2002. Topoisomerase II alpha as a reliable proliferation marker in me-
Menke, J.R., et al., 2015. Somatostatin receptor 2a is a more sensitive diagnostic marker of ningiomas. Neurol. Res. 24, 241243.
meningioma than epithelial membrane antigen. Acta Neuropathol. Ruttledge, M.H., et al., 1994. Deletions on chromosome 22 in sporadic meningioma. Genes
Miettinen, M., Paetau, A., 2002. Mapping of the keratin polypeptides in meningiomas Chromosomes Cancer 10, 122130.
of different types: an immunohistochemical analysis of 463 cases. Hum. Pathol. 33, Sahm, F., et al., 2013. AKT1E17K mutations cluster with meningothelial and transitional
590598. meningiomas and can be detected by SFRP1 immunohistochemistry. Acta
Moliterno, J., et al., 2015. Survival in patients treated for anaplastic meningioma. Neuropathol. 126, 757762.
J. Neurosurg. 123, 2330. Sanson, M., et al., 1993. Germline deletion in a neurobromatosis type 2 kindred inacti-
Murnyak, B., et al., 2015. Epigenetics of meningiomas. Biomed. Res. Int. 2015, 532451. vates the NF2 gene and a candidate meningioma locus. Hum. Mol. Genet. 2,
Nagashima, G., et al., 1995. Immunohistochemical detection of progesterone receptors 12151220.
and the correlation with Ki-67 labeling indices in parafn-embedded sections of Seizinger, B.R., et al., 1987. Molecular genetic approach to human meningioma: loss of
meningiomas. Neurosurgery 37, 478482 (discussion 483). genes on chromosome 22. Proc. Natl. Acad. Sci. U. S. A. 84, 54195423.
Ng, H.K., et al., 1995. Combined molecular genetic studies of chromosome 22q and the Simon, M., et al., 2000. Telomerase activity and expression of the telomerase catalytic sub-
neurobromatosis type 2 gene in central nervous system tumors. Neurosurgery 37, unit, hTERT, in meningioma progression. J. Neurosurg. 92, 832840.
764773. Sun, S.Q., et al., 2015. An evidence-based treatment algorithm for the management of
Ng, J., et al., 2015. Sox10 is superior to S100 in the diagnosis of meningioma. Appl. WHO Grade II and III meningiomas. Neurosurg. Focus. 38, E3.
Immunohistochem. Mol. Morphol. 23, 215219. Surace, E.I., et al., 2004. Loss of tumor suppressor in lung cancer-1 (TSLC1) expression in
Norden, A.D., et al., 2015. Phase II study of monthly pasireotide LAR (SOM230C) for recurrent meningioma correlates with increased malignancy grade and reduced patient surviv-
or progressive meningioma. Neurology 84, 280286. al. J. Neuropathol. Exp. Neurol. 63, 10151027.
Nunes, F., et al., 2005. Inactivation patterns of NF2 and DAL-1/4.1B (EPB41L3) in sporadic Trofatter, J.A., et al., 1993. A novel moesin-, ezrin-, radixin-like gene is a candidate for the
meningioma. Cancer Genet. Cytogenet. 162, 135139. neurobromatosis 2 tumor suppressor. Cell 75, 826.
Olar, A., et al., 2015. Mitotic index is an independent predictor of recurrence-free survival Walcott, B.P., et al., 2013. Radiation treatment for WHO Grade II and III meningiomas.
in meningioma. Brain Pathol. 25, 266275. Front. Oncol. 3, 227.
Patil, S., et al., 2011. Malignant meningiomas with epithelial (adenocarcinoma-like) Wernicke, A.G., et al., 2010. Assessment of epidermal growth factor receptor (EGFR)
metaplasia: a study of 3 cases. Neurosurgery 69, 884892. expression in human meningioma. Radiat. Oncol. 5, 46.
Perry, A., et al., 2002. A role for chromosome 9p21 deletions in the malignant progression Wiemels, J., et al., 2010. Epidemiology and etiology of meningioma. J. Neuro-Oncol. 99,
of meningiomas and the prognosis of anaplastic meningiomas. Brain Pathol. 12, 307314.
183190. Yamashima, T., et al., 1997. Prostaglandin D synthase (beta-trace) in human arachnoid
Perry, A., Brat, D.J., 2010. Practical Surgical Neuropathology : A Diagnostic Approach. and meningioma cells: roles as a cell marker or in cerebrospinal uid absorption, tu-
Churchill Livingstone/Elsevier, Philadelphia, PA. morigenesis, and calcication process. J Neurosci. 17, 23762382.
Perry, A., et al., 2000. Merlin, DAL-1, and progesterone receptor expression in clinico- Yang, H.W., et al., 2012. Alternative splicing of CHEK2 and codeletion with NF2 promote
pathologic subsets of meningioma: a correlative immunohistochemical study of chromosomal instability in meningioma. Neoplasia 14, 2028.
175 cases. J. Neuropathol. Exp. Neurol. 59, 872879. Zang, K.D., 2001. Meningioma: a cytogenetic model of a complex benign human tumor,
Perry, A., et al., 2001. Aggressive phenotypic and genotypic features in pediatric and NF2- including data on 394 karyotyped cases. Cytogenet. Cell Genet. 93, 207220.
associated meningiomas: a clinicopathologic study of 53 cases. J. Neuropathol. Exp. Zhang, X., et al., 2010. Maternally expressed gene 3, an imprinted noncoding RNA gene, is
Neurol. 60, 9941003. associated with meningioma pathogenesis and progression. Cancer Res. 70,
Perry, A., Louis, D.N., Scheithauer, B.W., Budka, H., von Deimling, A., 2007. Meningiomas. 23502358.
In: Louis, D.N., Ohgaki, H., Wiestler, O.D., Cavenee, W.K. (Eds.), WHO Classication
of Tumours of the Central Nervous System. IARC, Lyon.

Das könnte Ihnen auch gefallen