Skip to main content
Erschienen in: neurogenetics 2/2016

Open Access 01.04.2016 | Review Article

A heritable form of SMARCE1-related meningiomas with important implications for follow-up and family screening

verfasst von: E. H. Gerkes, J. M. Fock, W. F. A. den Dunnen, M. J. van Belzen, C. A. van der Lans, E. W. Hoving, I. E. Fakkert, M. J. Smith, D. G. Evans, M. J. W. Olderode-Berends

Erschienen in: Neurogenetics | Ausgabe 2/2016

Abstract

Childhood meningiomas are rare. Recently, a new hereditary tumor predisposition syndrome has been discovered, resulting in an increased risk for spinal and intracranial clear cell meningiomas (CCMs) in young patients. Heterozygous loss-of-function germline mutations in the SMARCE1 gene are causative, giving rise to an autosomal dominant inheritance pattern. We report on an extended family with a pediatric CCM patient and an adult CCM patient and several asymptomatic relatives carrying a germline SMARCE1 mutation, and discuss difficulties in genetic counseling for this heritable condition. Because of the few reported cases so far, the lifetime risk of developing meningiomas for SMARCE1 mutation carriers is unclear and the complete tumor spectrum is unknown. There is no surveillance guideline for asymptomatic carriers nor a long-term follow-up recommendation for SMARCE1-related CCM patients as yet. Until more information is available about the penetrance and tumor spectrum of the condition, we propose the following screening advice for asymptomatic SMARCE1 mutation carriers: neurological examination and MRI of the brain and spine, yearly from diagnosis until the age of 18 and once every 3 years thereafter, or in between if there are clinical symptoms. This advice can also be used for long-term patient follow-up. More data is needed to optimize this proposed screening advice.

Introduction

Meningiomas mostly arise in middle-aged people. Occurrence in childhood is rare [1]. Meningiomas account for a small subset (1–4 %) of all pediatric brain tumors [2]. A meningioma in childhood can be the first presenting symptom of neurofibromatosis type 2 (OMIM 101000), caused by mutations in the NF2 gene [35]. Our knowledge of the etiology of meningiomas not caused by neurofibromatosis type 2 is still limited [6]. Some meningiomas are caused by germline mutations in the SMARCB1 gene, but here the risk for single meningiomas without the occurrence of schwannomas is rare [7, 8]. Meningiomas may also occur due to germline mutations in SUFU [9, 10].
One subtype of meningiomas, the clear cell subtype, arises more frequently in young people compared to more common subtypes of meningiomas [11]. Clear cell meningiomas (CCM) are a subtype with a specific histology and in situ behavior. The tumors are more aggressive with a tendency to recur and metastasize within the CNS compared to nonclear cell meningiomas. In the World Health Organization classification of “tumors of the CNS,” clear cell meningiomas are defined as grade 2 because of their aggressiveness [12]. Early detection and treatment are therefore of paramount importance for this tumor type.
Recently, marked steps in the etiologic understanding of clear cell meningiomas were taken. In 2013 and 2014, Smith et al. reported on heterozygous germline mutations in the SMARCE1 gene in 16 patients from 11 unrelated families with spinal and intracranial CCM [11, 13, 14]. The patients were mostly children, adolescents, or young adults. The first mutations were detected after whole-exome sequencing and further cases were proven by Sanger sequencing. In the examined tumors, loss of the SMARCE1 protein was shown by immunohistochemical analysis. Tumor DNA showed loss of heterozygosity (LOH) of the wild-type allele or a second inactivating mutation as a second hit in some tumors, implying a tumor suppressor function of the SMARCE1 gene. These findings prove the existence of a hereditary tumor predisposition syndrome with an increased risk for spinal and intracranial CCMs (OMIM 607174). Genetic testing and counseling in afflicted families have now become possible by finding the causative gene but poses new questions and difficulties because of the sparse knowledge so far.
Here we present a family with a pediatric CCM patient and an adult CCM patient and several asymptomatic relatives carrying a germline SMARCE1 mutation. We propose a screening advice for asymptomatic carriers in the family and for long-term patient follow-up.

Case report

A 10-year-old boy was referred to our centre because of recent onset of hearing loss and tinnitus of the right ear. He complained about blurry vision. His medical history was unremarkable apart from treatment with methylphenidate because of ADHD. Physical examination of the ear, nose and throat showed no abnormalities apart from an abnormal Weber test to the left, and an asymmetric reaction of facial nerve. The audiogram showed a sensorineural hearing loss of the right ear, with a downsloping audiogram and complete loss of higher tones indicating damage to the acoustic nerve. The MRI scan of the brain showed a large extrinsic tumor in the right cerebello-pontine angle with severe compression and displacement of the brainstem (Fig. 1). The tumor could be removed in two successive surgical sessions. In the first surgery, the tumor mass could be taken out almost completely except for a very adherent remnant on the vertebral artery and a second separate tumor on the other side. In spite of the close involvement of the lower cranial nerves, all these nerves could be saved anatomically and functionally as monitored intraoperatively. Pathological examination of the tumor showed a clear cell type meningioma, WHO grade II (Fig 2). Hereafter, a second surgery with the aim of radical resection of the remnants was undertaken with good results (Simpson classification I). Post-operatively, the patient experienced swallowing difficulties due to multiple cranial nerve apraxia. In due time, he recovered well and, after 4 months, he was able to speak, eat and drink normally while some atrophy of the right part of the tongue remained. There has been no local recurrence of tumor during 1-year follow-up.
Sanger sequencing of the SMARCE1 gene in DNA from blood showed a 1-bp deletion causing a frameshift in exon 9: c.814delA, p.Arg272Glyfs*5. This particular mutation was not reported before, but the nature of the mutation makes it very likely pathogenic. Analysis of tumor DNA showed homozygosity of this mutation indicating loss-of-heterozygosity (LOH) at the mutation locus (Fig 3). Mutation analysis of the NF2 gene was normal.

Family history

The parents of the patient were tested for the SMARCE1 mutation after genetic counseling. The father was shown to be a carrier. An MRI brain and spine showed no tumors. As shown in the pedigree, the paternal grandmother had a spinal tumor removed at age 36. The initial pathology report mentioned an ependymoma, but revision showed a clear cell meningioma grade II. Recent follow-up with MRI showed no abnormalities at age 70. The brother and sister of the index case and a paternal aunt proved to be asymptomatic carriers after presymptomatic testing. All were referred for neuroaxis screening and MRI of the brain and spine in follow-up. A recent MRI brain and spine showed no abnormalities in the aunt. MRI brain of the brother at age five because of temporary diplopia showed no abnormalities. The further MRI results of the brother and sister are pending. The family pedigree is shown in Fig. 4.

Discussion

This case report shows further evidence for the role of SMARCE1 mutations in the etiology of clear cell meningiomas. We address here the following questions: why to screen, who to screen, what to screen and how to screen in afflicted families and discuss the difficulties in determining what is the best practice.

Why to screen?

Early detection of meningiomas is of highest importance to prevent damage of surrounding tissue and dissemination. It also enhances the chance of total radical resection. For childhood and adolescent meningiomas, the extent of the initial surgical resection is the strongest independent prognostic factor [15]. Radical neurosurgical resection of very large tumors in eloquent areas may be more hazardous to the patient with a greater chance of (transient) post-operative morbidity, as illustrated in our case. Clear cell meningiomas tend to behave more aggressively and have a tendency to recur and metastasize, making it even more paramount to discover this type as early as possible [12]. Unfortunately, meningiomas frequently cause symptoms late, only after they have grown slowly to a large size. The mass effect of the lesion on the environment finally creates symptoms, like in our patient. This may interfere with early detection. Screening in high-risk groups could therefore be beneficial.

Who to screen?

Patients that have been treated for a meningioma will remain in follow-up for several years but are discharged thereafter because the risk of recurrence will decline. However, patients with a history of a SMARCE1-related meningioma will probably remain at a higher risk for further CCMs, particularly if female. This makes them a possible high-risk group for which a long-term follow-up schedule might be beneficial. It is currently unknown whether the risk for CCMs remains increased during the rest of the lifetime or decreases substantially after a certain age, as in NF2 where the risk for new tumors declines with older age [16]. We believe that long-term screening is advisable until we have more knowledge on this.
Carrier family members could constitute a second high-risk group for CCMs. Presymptomatic testing of family members after genetic counseling can be beneficial because it allows for justified screening and early tumor detection in carriers. However, genetic counseling is complicated by the fact that the penetrance of the disease and the lifetime risk for CCMs are unknown at the moment. Moreover, no established guideline is available providing a screening advice. Penetrance of the disease could be overestimated based on current literature, as only a few families have been described so far with probably a detection bias towards families with more than one patient. Several asymptomatic carriers of different ages and gender were detected in the family reported here. Incomplete penetrance for meningiomas in SMARCE1 mutation- positive families was shown before by Smith et al. They found three asymptomatic male carriers, aged 17, 71, and middle age, coming from three different families with more than one meningioma patient [13, 14].

What to screen?

There is no certainty about the complete tumor spectrum of this tumor syndrome as of yet. We know from other autosomal dominant hereditary cancer predisposition syndromes that tumors can arise in different and distinct tissue types, for example, in Lynch syndrome and Von Hippel Lindau syndrome [17, 18]. In those diseases, the problem is bi-allelic shutdown of a tumor suppressor gene within the tumor tissue. It apparently depends on the nature of the exact tumor suppressor gene, which types of tissues will be at risk for tumor formation. The evidence of the few cases of SMARCE1-related CCMs that have been published so far, together with the evidence of the patient reported here, shows that the SMARCE1 gene also acts as a tumor suppressor gene [13]. In the clear cell meningiomas, there is a second hit causing inactivation of the wild-type allele [14]. So far, no other tumor types than spinal and intracranial CCMs have been described in SMARCE1-positive patients. Strategic gathering of patient and family data can help determine if we need to be on the lookout for other types of tumors in carriers, and further knowledge is needed to better understand why loss of SMARCE1 expression specifically leads to CCMs. For now, we propose to screen for spinal and intracranial CCMs only. Raffalli-Ebezant et al. reported on a carrier female who appeared to have multiple, asymptomatic spinal lesions in keeping with intradural meningiomas [11]. This suggests that multiple tumors can be present and screening of the whole brain and spine is advisable.
All mutations in the SMARCE1 gene found so far in CCM patients are loss-of-function mutations, including frameshift and nonsense mutations, an inversion and two large deletions [11, 13, 14, 19]. Missense mutations in the SMARCE1 gene cause a clinically very different syndrome called Coffin-Siris syndrome (OMIM 135900) with congenital mental retardation and dysmorphisms as main features [20, 21]. The so far known CCM patients with a loss-of-function SMARCE1 mutation have no clinical signs of Coffin-Siris syndrome, and screening for developmental delay or dysmorphisms is therefore not necessary.
It is currently unclear if missense mutations in the SMARCE1 gene causing Coffin-Siris syndrome predispose to CCMs later in life. This combination has actually been described very recently for a related gene causing Coffin-Siris syndrome, the SMARCB1 gene. A patient with Coffin-Siris syndrome phenotype and a constitutional missense SMARCB1 gene mutation developed schwannomatosis [22].

How to screen?

Until more information is available about the penetrance and complete tumor spectrum of the condition, we propose the following screening advice for asymptomatic SMARCE1 mutation carriers: neurological examination and MRI of the brain and spine, yearly from diagnosis until the age of 18 and once every 3 years thereafter, or in between if there are clinical symptoms.
For asymptomatic carrier children/adolescents, more frequent neurological screening is advised than for adults, because they are less well equipped to detect and mention any neurological problems and because the risk to develop tumors in SMARCE1 carriers seems to be larger at younger age, especially for boys [11, 13, 14, 19]. Furthermore, childhood meningiomas in general tend to have a more aggressive biological behavior and a worse prognosis than the same tumors in adults, justifying a more aggressive screening approach in children to detect the tumors in an early state [1, 23, 24]. A disadvantage of starting screening at a young age is the fact that MRI of the neuraxis in young or noncooperative patients requires sedation or general anesthesia because of the long duration of the imaging. The use of melatonin as a low-risk sedation substitute could help prevent this in a subset of patients. The morbidity and mortality risk of sedation or anesthesia in children is small if done in a well-equipped centre with experience.
All symptomatic males with a SMARCE1 mutation described so far developed meningiomas in childhood (age range 2–10 years), while the symptomatic carrier females developed tumors somewhat later in adolescence or early adulthood (age range 14–30s) [11, 13, 14, 19]. This was again shown in our family. These findings suggest that penetrance of the disease might be age- and gender-dependent. Those carrier males who do develop a tumor seem to develop it at early age and females seem to be at higher risk after onset of their fertile period. The increased penetrance for males for meningiomas in childhood is also found both in sporadic meningiomas and NF2 [5] with the reverse being true in adulthood. Smith et al. hypothesize a possible hormonal influence on penetrance, but there is no scientific proof on this as yet [10, 13]. We believe that this ample amount of evidence for gender- and age-specific penetrance is not enough to adjust a screening advice depending on age or gender at the present time. If the evidence becomes stronger, less surveillance for men after a certain age could possibly be considered.
For patients after meningioma treatment, frequency and duration of the follow-up surveillance depend on the location of the tumor, the WHO grade of the tumor, and the extent of the tumor resection according to the Dutch national guideline Intracranial Meningioma [25]. The above mentioned screening advice could be used for long-term follow-up thereafter for SMARCE1-related CCM patients, because of their enlarged risk of additional primary tumors.

Future directives

Hopefully, substantiated adjustment to this proposed screening advice can be made after structural gathering of more medical and genetic data of patients and families. This will help to better estimate the age- and gender-dependant penetrance of the disease and gain more certainty about the complete tumor spectrum for carriers of a SMARCE1 mutation. The addition of this report about a larger family to the current literature on the subject hopefully is a step in the right direction, raising awareness of the condition and adding relevant data to the knowledge.

Acknowledgments

We would like to thank the family for their cooperation.

Compliance with ethical standards

Informed consent was obtained from all individual participants included in the study.
For this type of study, formal consent is not required.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Thuijs NB, Uitdehaag BM, Van Ouwerkerk WJ, van der Valk P, Vandertop WP, Peerdeman SM (2012) Pediatric meningiomas in The Netherlands 1974–2010: a descriptive epidemiological case study. Childs Nerv Syst 28:1009–1015CrossRefPubMedPubMedCentral Thuijs NB, Uitdehaag BM, Van Ouwerkerk WJ, van der Valk P, Vandertop WP, Peerdeman SM (2012) Pediatric meningiomas in The Netherlands 1974–2010: a descriptive epidemiological case study. Childs Nerv Syst 28:1009–1015CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Caroli E, Russillo M, Ferrante L (2006) Intracranial meningiomas in children: report of 27 new cases and critical analysis of 440 cases reported in the literature. J Child Neurol 21:31–36CrossRefPubMed Caroli E, Russillo M, Ferrante L (2006) Intracranial meningiomas in children: report of 27 new cases and critical analysis of 440 cases reported in the literature. J Child Neurol 21:31–36CrossRefPubMed
4.
Zurück zum Zitat Perry A, Giannini C, Raghavan R, Scheithauer BW, Banerjee R, Margraf L, Bowers DC, Lytle RA, Newsham IF, Gutmann DH (2001) Aggressive phenotypic and genotypic features in pediatric and NF2-associated meningiomas: a clinicopathologic study of 53 cases. J Neuropathol Exp Neurol 60:994–1003CrossRefPubMed Perry A, Giannini C, Raghavan R, Scheithauer BW, Banerjee R, Margraf L, Bowers DC, Lytle RA, Newsham IF, Gutmann DH (2001) Aggressive phenotypic and genotypic features in pediatric and NF2-associated meningiomas: a clinicopathologic study of 53 cases. J Neuropathol Exp Neurol 60:994–1003CrossRefPubMed
5.
Zurück zum Zitat Smith MJ, Higgs JE, Bowers NL, Halliday D, Paterson J, Gillespie J, Huson SM, Freeman SR, Lloyd S, Rutherford SA, King AT, Wallace AJ, Ramsden RT, Evans DG (2011) Cranial meningiomas in 411 neurofibromatosis type 2 (NF2) patients with proven gene mutations: clear positional effect of mutations, but absence of female severity effect on age at onset. J Med Genet 48:261–265CrossRefPubMed Smith MJ, Higgs JE, Bowers NL, Halliday D, Paterson J, Gillespie J, Huson SM, Freeman SR, Lloyd S, Rutherford SA, King AT, Wallace AJ, Ramsden RT, Evans DG (2011) Cranial meningiomas in 411 neurofibromatosis type 2 (NF2) patients with proven gene mutations: clear positional effect of mutations, but absence of female severity effect on age at onset. J Med Genet 48:261–265CrossRefPubMed
6.
Zurück zum Zitat Plotkin SR, Blakeley JO, Evans DG, Hanemann CO, Hulsebos TJ, Hunter-Schaedle K, Kalpana GV, Korf B, Messiaen L, Papi L, Ratner N, Sherman LS, Smith MJ, Stemmer-Rachamimov AO, Vitte J, Giovannini M (2013) Update from the 2011 international schwannomatosis workshop: from genetics to diagnostic criteria. Am J Med Genet A 161A:405–416CrossRefPubMed Plotkin SR, Blakeley JO, Evans DG, Hanemann CO, Hulsebos TJ, Hunter-Schaedle K, Kalpana GV, Korf B, Messiaen L, Papi L, Ratner N, Sherman LS, Smith MJ, Stemmer-Rachamimov AO, Vitte J, Giovannini M (2013) Update from the 2011 international schwannomatosis workshop: from genetics to diagnostic criteria. Am J Med Genet A 161A:405–416CrossRefPubMed
7.
Zurück zum Zitat van den Munckhof P, Christiaans I, Kenter SB, Baas F, Hulsebos TJ (2012) Germline SMARCB1 mutation predisposes to multiple meningiomas and schwannomas with preferential location of cranial meningiomas at the falx cerebri. Neurogenetics 13:1–7CrossRefPubMed van den Munckhof P, Christiaans I, Kenter SB, Baas F, Hulsebos TJ (2012) Germline SMARCB1 mutation predisposes to multiple meningiomas and schwannomas with preferential location of cranial meningiomas at the falx cerebri. Neurogenetics 13:1–7CrossRefPubMed
8.
Zurück zum Zitat Prescott TE, Smith MJ, Evans DG (2012) Comment on the article "germline SMARCB1 mutation predisposes to multiple meningiomas and schwannomas with preferential location of cranial meningiomas at the falx cerebri" by van den munckhof et al. Neurogenetics 13:103–104CrossRefPubMed Prescott TE, Smith MJ, Evans DG (2012) Comment on the article "germline SMARCB1 mutation predisposes to multiple meningiomas and schwannomas with preferential location of cranial meningiomas at the falx cerebri" by van den munckhof et al. Neurogenetics 13:103–104CrossRefPubMed
9.
Zurück zum Zitat Aavikko M, Li SP, Saarinen S, Alhopuro P, Kaasinen E, Morgunova E, Li Y, Vesanen K, Smith MJ, Evans DG, Poyhonen M, Kiuru A, Auvinen A, Aaltonen LA, Taipale J, Vahteristo P (2012) Loss of SUFU function in familial multiple meningioma. Am J Hum Genet 91:520–526CrossRefPubMedPubMedCentral Aavikko M, Li SP, Saarinen S, Alhopuro P, Kaasinen E, Morgunova E, Li Y, Vesanen K, Smith MJ, Evans DG, Poyhonen M, Kiuru A, Auvinen A, Aaltonen LA, Taipale J, Vahteristo P (2012) Loss of SUFU function in familial multiple meningioma. Am J Hum Genet 91:520–526CrossRefPubMedPubMedCentral
10.
11.
Zurück zum Zitat Raffalli-Ebezant H, Rutherford SA, Stivaros S, Kelsey A, Smith M, Evans DG, Kilday JP (2015) Pediatric intracranial clear cell meningioma associated with a germline mutation of SMARCE1: a novel case. Childs Nerv Syst 31:441–447CrossRefPubMed Raffalli-Ebezant H, Rutherford SA, Stivaros S, Kelsey A, Smith M, Evans DG, Kilday JP (2015) Pediatric intracranial clear cell meningioma associated with a germline mutation of SMARCE1: a novel case. Childs Nerv Syst 31:441–447CrossRefPubMed
12.
Zurück zum Zitat Perry A, Louis DN, Scheithauwer BW, Budka H, von Deimling A (2007) Meningiomas. In: Louis DN, Ohgaki H, Wiestler OD, Cavenee WK (eds) WHO classification of tumors of the central nervous system. Lyon, France, International Agency for Research on Cancer, pp. 164–172 Perry A, Louis DN, Scheithauwer BW, Budka H, von Deimling A (2007) Meningiomas. In: Louis DN, Ohgaki H, Wiestler OD, Cavenee WK (eds) WHO classification of tumors of the central nervous system. Lyon, France, International Agency for Research on Cancer, pp. 164–172
13.
Zurück zum Zitat Smith MJ, O’Sullivan J, Bhaskar SS, Hadfield KD, Poke G, Caird J, Sharif S, Eccles D, Fitzpatrick D, Rawluk D, du Plessis D, Newman WG, Evans DG (2013) Loss-of-function mutations in SMARCE1 cause an inherited disorder of multiple spinal meningiomas. Nat Genet 45:295–298CrossRefPubMed Smith MJ, O’Sullivan J, Bhaskar SS, Hadfield KD, Poke G, Caird J, Sharif S, Eccles D, Fitzpatrick D, Rawluk D, du Plessis D, Newman WG, Evans DG (2013) Loss-of-function mutations in SMARCE1 cause an inherited disorder of multiple spinal meningiomas. Nat Genet 45:295–298CrossRefPubMed
14.
Zurück zum Zitat Smith MJ, Wallace AJ, Bennett C, Hasselblatt M, Elert-Dobkowska E, Evans LT, Hickey WF, van Hoff J, Bauer D, Lee A, Hevner RF, Beetz C, du Plessis D, Kilday JP, Newman WG, Evans DG (2014) Germline SMARCE1 mutations predispose to both spinal and cranial clear cell meningiomas. J Pathol 234:436–440CrossRefPubMed Smith MJ, Wallace AJ, Bennett C, Hasselblatt M, Elert-Dobkowska E, Evans LT, Hickey WF, van Hoff J, Bauer D, Lee A, Hevner RF, Beetz C, du Plessis D, Kilday JP, Newman WG, Evans DG (2014) Germline SMARCE1 mutations predispose to both spinal and cranial clear cell meningiomas. J Pathol 234:436–440CrossRefPubMed
15.
Zurück zum Zitat Kotecha RS, Pascoe EM, Rushing EJ, Rorke-Adams LB, Zwerdling T, Gao X, Li X, Greene S, Amirjamshidi A, Kim SK, Lima MA, Hung PC, Lakhdar F, Mehta N, Liu Y, Devi BI, Sudhir BJ, Lund-Johansen M, Gjerris F, Cole CH, Gottardo NG (2011) Meningiomas in children and adolescents: a meta-analysis of individual patient data. Lancet Oncol 12:1229–1239CrossRefPubMed Kotecha RS, Pascoe EM, Rushing EJ, Rorke-Adams LB, Zwerdling T, Gao X, Li X, Greene S, Amirjamshidi A, Kim SK, Lima MA, Hung PC, Lakhdar F, Mehta N, Liu Y, Devi BI, Sudhir BJ, Lund-Johansen M, Gjerris F, Cole CH, Gottardo NG (2011) Meningiomas in children and adolescents: a meta-analysis of individual patient data. Lancet Oncol 12:1229–1239CrossRefPubMed
16.
Zurück zum Zitat Evans DG, Raymond FL, Barwell JG, Halliday D (2012) Genetic testing and screening of individuals at risk of NF2. Clin Genet 82:416–424CrossRefPubMed Evans DG, Raymond FL, Barwell JG, Halliday D (2012) Genetic testing and screening of individuals at risk of NF2. Clin Genet 82:416–424CrossRefPubMed
17.
Zurück zum Zitat Kohlmann W, Gruber SB (1993) Lynch syndrome. In: Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, Bird TD, Fong CT, Smith RJH, Stephens K (eds) GeneReviews(R). University of Washington, Seattle, Seattle (WA) Kohlmann W, Gruber SB (1993) Lynch syndrome. In: Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, Bird TD, Fong CT, Smith RJH, Stephens K (eds) GeneReviews(R). University of Washington, Seattle, Seattle (WA)
18.
Zurück zum Zitat Frantzen C, Klasson TD, Links TP, Giles RH (1993) Von Hippel-Lindau syndrome. In: Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, Bird TD, Fong CT, Smith RJH, Stephens K (eds) GeneReviews(R). University of Washington, Seattle, Seattle (WA) Frantzen C, Klasson TD, Links TP, Giles RH (1993) Von Hippel-Lindau syndrome. In: Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, Bird TD, Fong CT, Smith RJH, Stephens K (eds) GeneReviews(R). University of Washington, Seattle, Seattle (WA)
19.
Zurück zum Zitat Evans LT, Van Hoff J, Hickey WF, Smith MJ, Evans DG, Newman WG, Bauer DF (2015) SMARCE1 mutations in pediatric clear cell meningioma: case report. J Neurosurg Pediatr 16(3):296–300 Evans LT, Van Hoff J, Hickey WF, Smith MJ, Evans DG, Newman WG, Bauer DF (2015) SMARCE1 mutations in pediatric clear cell meningioma: case report. J Neurosurg Pediatr 16(3):296–300
20.
Zurück zum Zitat Tsurusaki Y, Okamoto N, Ohashi H, Kosho T, Imai Y, Hibi-Ko Y, Kaname T, Naritomi K, Kawame H, Wakui K, Fukushima Y, Homma T, Kato M, Hiraki Y, Yamagata T, Yano S, Mizuno S, Sakazume S, Ishii T, Nagai T, Shiina M, Ogata K, Ohta T, Niikawa N, Miyatake S, Okada I, Mizuguchi T, Doi H, Saitsu H, Miyake N, Matsumoto N (2012) Mutations affecting components of the SWI/SNF complex cause coffin-siris syndrome. Nat Genet 44:376–378CrossRefPubMed Tsurusaki Y, Okamoto N, Ohashi H, Kosho T, Imai Y, Hibi-Ko Y, Kaname T, Naritomi K, Kawame H, Wakui K, Fukushima Y, Homma T, Kato M, Hiraki Y, Yamagata T, Yano S, Mizuno S, Sakazume S, Ishii T, Nagai T, Shiina M, Ogata K, Ohta T, Niikawa N, Miyatake S, Okada I, Mizuguchi T, Doi H, Saitsu H, Miyake N, Matsumoto N (2012) Mutations affecting components of the SWI/SNF complex cause coffin-siris syndrome. Nat Genet 44:376–378CrossRefPubMed
21.
Zurück zum Zitat GW S, E A, AT V-v S, C P, BW v B, IJ v M, R S, CA v d L, M B, MM L, L V, MJ v d W, MJ V, consortium C-S, MH B, M K, A v H, JT d D, A H, J C-S, BB d V, RC H, MJ v B (2013) Coffin-siris syndrome and the BAF complex: genotype-phenotype study in 63 patients. Hum Mutat 34:1519–1528CrossRef GW S, E A, AT V-v S, C P, BW v B, IJ v M, R S, CA v d L, M B, MM L, L V, MJ v d W, MJ V, consortium C-S, MH B, M K, A v H, JT d D, A H, J C-S, BB d V, RC H, MJ v B (2013) Coffin-siris syndrome and the BAF complex: genotype-phenotype study in 63 patients. Hum Mutat 34:1519–1528CrossRef
22.
Zurück zum Zitat Gossai N, Biegel JA, Messiaen L, Berry SA, Moertel CL (2015) Report of a patient with a constitutional missense mutation in SMARCB1, coffin-siris phenotype, and schwannomatosis. Am J Med Genet A 167(12):3186–3191 Gossai N, Biegel JA, Messiaen L, Berry SA, Moertel CL (2015) Report of a patient with a constitutional missense mutation in SMARCB1, coffin-siris phenotype, and schwannomatosis. Am J Med Genet A 167(12):3186–3191
23.
Zurück zum Zitat Kotecha RS, Junckerstorff RC, Lee S, Cole CH, Gottardo NG (2011) Pediatric meningioma: current approaches and future direction. J Neuro-Oncol 104:1–10CrossRef Kotecha RS, Junckerstorff RC, Lee S, Cole CH, Gottardo NG (2011) Pediatric meningioma: current approaches and future direction. J Neuro-Oncol 104:1–10CrossRef
25.
Zurück zum Zitat Brandsma D, Gilhuijs NDB, Groen RJM, Habets EJJ, Hoenderdaal P, Jacobi-Postma AA, Mendez Romero A, Niel CGJH, Peerdeman SM, Petersen-Baltussen H, Schepers VPM, Snijders TJ, van Valkenburg H, Wesseling P, Zwinkels JMC, Hes FJ (2015) Dutch National guideline Intracranial Meningioma, Oncoline www.oncoline.nl Brandsma D, Gilhuijs NDB, Groen RJM, Habets EJJ, Hoenderdaal P, Jacobi-Postma AA, Mendez Romero A, Niel CGJH, Peerdeman SM, Petersen-Baltussen H, Schepers VPM, Snijders TJ, van Valkenburg H, Wesseling P, Zwinkels JMC, Hes FJ (2015) Dutch National guideline Intracranial Meningioma, Oncoline www.oncoline.nl
Metadaten
Titel
A heritable form of SMARCE1-related meningiomas with important implications for follow-up and family screening
verfasst von
E. H. Gerkes
J. M. Fock
W. F. A. den Dunnen
M. J. van Belzen
C. A. van der Lans
E. W. Hoving
I. E. Fakkert
M. J. Smith
D. G. Evans
M. J. W. Olderode-Berends
Publikationsdatum
01.04.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Neurogenetics / Ausgabe 2/2016
Print ISSN: 1364-6745
Elektronische ISSN: 1364-6753
DOI
https://doi.org/10.1007/s10048-015-0472-y

Weitere Artikel der Ausgabe 2/2016

neurogenetics 2/2016 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.