ArticlePDF Available

SF3B1 and EIF1AX mutations occur in primary leptomeningeal melanocytic neoplasms; Yet another similarity to uveal melanomas

Authors:

Abstract and Figures

Like uveal melanomas, primary leptomeningeal melanocytic neoplasms (LMNs) frequently carry GNAQ and GNA11 mutations. However, it is currently unknown whether these LMNs harbor mutations in BAP1, SF3B1 and/or EIF1AX like uveal melanomas as well. In this study, we used Sanger sequencing for the detection of mutations in SF3B1 (hotspots in exon 14 and 15) and EIF1AX (exon 1 and 2 and flanking intronic regions) in a series of 24 primary LMNs. Additionally, BAP1 immunohistochemistry was used as a surrogate marker for the detection of inactivating mutations in the BAP1 gene. Mutations in either SF3B1 or EIF1AX were identified in 8 out of 24 primary LMNs (33 %). The presence of these mutations was mutually exclusive and occurred in primary LMNs of different malignancy grades (melanocytomas, intermediate-grade melanocytic tumors, melanomas). Complete absence of nuclear BAP1 staining as is typically seen in BAP1-mutated tumors was not observed. Our finding that an SF3B1 or EIF1AX mutation is present in a substantial subset of primary LMNs underscores that these tumors genetically resemble uveal melanoma and are different from cutaneous melanoma at the genetic level. This information may not only aid in the differential diagnosis of primary versus metastatic melanocytic tumor in/around the central nervous system, but also in the identification of more promising therapeutic approaches targeting the molecular pathways involved in the oncogenesis of LMNs. As none of the primary LMNs in our series showed complete loss of nuclear BAP1 protein, it is unlikely that BAP1 mutations are frequent in these tumors but the role of this gene warrants further investigation.
Content may be subject to copyright.
R E S E A R C H Open Access
SF3B1 and EIF1AX mutations occur in
primary leptomeningeal melanocytic
neoplasms; yet another similarity to
uveal melanomas
Heidi V. N. Küsters-Vandevelde
1*
, David Creytens
2
, Adriana C. H. van Engen-van Grunsven
3
, Marcel Jeunink
1
,
Veronique Winnepenninckx
4
, Patricia J. T. A. Groenen
3
, Benno Küsters
3,4
,PieterWesseling
1,3,5
, Willeke A. M. Blokx
3
and Clemens F. M. Prinsen
1
Abstract
Introduction: Like uveal melanomas, primary leptomeningeal melanocytic neoplasms (LMNs) frequently carry GNAQ and
GNA11 mutations. However, it is currently unknown whether these LMNs harbor mutations in BAP1,SF3B1 and/or EIF1AX
like uveal melanomas as well. In this study, we used Sanger sequencing for the detection of mutations in SF3B1 (hotspots
in exon 14 and 15) and EIF1AX (exon 1 and 2 and flanking intronic regions) in a series of 24 primary LMNs. Additionally,
BAP1 immunohistochemistry was used as a surrogate marker for the detection of inactivating mutations in the BAP1 gene.
Results: Mutations in either SF3B1 or EIF1AX were identified in 8 out of 24 primary LMNs (33 %). The presence of these
mutations was mutually exclusive and occurred in primary LMNs of different malignancy grades (melanocytomas,
intermediate-grade melanocytic tumors, melanomas). Complete absence of nuclear BAP1 staining as is typically seen in
BAP1-mutated tumors was not observed.
Conclusions: Our finding that an SF3B1 or EIF1AX mutation is present in a substantial subset of primary LMNs underscores
that these tumors genetically resemble uveal melanoma and are different from cutaneous melanoma at the genetic level.
This information may not only aid in the differential diagnosis of primary versus metastatic melanocytic tumor in/around
the central nervous system, but also in the identification of more promising therapeutic approaches targeting the
molecular pathways involved in the oncogenesis of LMNs. As none of the primary LMNs in our series showed complete
loss of nuclear BAP1 protein, it is unlikely that BAP1 mutations are frequent in these tumors but the role of this gene
warrants further investigation.
Keywords: Leptomeningeal melanocytic neoplasms, Melanocytoma, Melanoma, Central nervous system, Neurocutaneous
melanocytosis, BAP1,SF3B1,EIF1AX
Introduction
Primary leptomeningeal melanocytic neoplasms (LMNs)
are infrequent tumors, forming a spectrum ranging
from benign or low-grade malignant melanocytomas
to frankly malignant melanomas [1]. These tumors
share molecular features with uveal melanomas (UMs).
In contrast to cutaneous melanomas (CMs), both
LMNs and UMs frequently carry mutations in the G
protein encoding genes GNAQ and GNA11,whereas
mutations in BRAF and in the TERT promoter are in-
frequent [28]. This situation reflects the heteroge-
neous molecular background of different groups of
melanoma and has important implications for tar-
geted therapy.
In the past years, inactivating mutations in the tumor
suppressor gene BAP1 (BRCA-associated protein 1) were
shown to be implicated in UM [9, 10]. The BAP1 gene is
located on chromosome 3p21.1 and encodes a nuclear
* Correspondence: h.kusters@cwz.nl
1
Department of Pathology, Canisius Wilhelmina Hospital, P.O. Box
90156500GS, Nijmegen, The Netherlands
Full list of author information is available at the end of the article
© 2016 Küsters-Vandevelde et al. 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. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5
DOI 10.1186/s40478-016-0272-0
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
ubiquitinase involved in epigenetic modulation of chroma-
tin [11]. Somatic BAP1 mutations are predominantly
present in UMs with monosomy 3 (~85 %), the latter being
a strong predictor for metastatic disease [9, 10, 12]. In this
setting BAP1 functions as a tumor suppressor gene, with
loss of one copy of chromosome 3 and mutation in the
other BAP1 allele representing the two hits causing inacti-
vation of this gene. Indeed, in UMs with disomy 3 (and a
good prognosis), mutations in BAP1 are rare [6, 9, 10]. A
small proportion of patients with UM (~23%)har-
bor a germline mutation in BAP1 [13]. These patients
suffer from the BAP1 hereditary cancer syndrome and
have an increased risk of developing cutaneous melanocy-
tic tumors as well as a spectrum of non-melanocytic neo-
plasms including mesothelioma, renal cell carcinoma,
meningeoma, and adenocarcinoma of the lung [14, 15].
Very recently, it was suggested that primary leptomenin-
geal melanoma is part of this cancer predisposition syn-
drome as well [16].
Furthermore, recurrent hotspot mutations in the
SF3B1 gene (mainly at codon 625) and mutations of
the EIF1AX gene (spread over exon 1 and 2) were
recently reported in UMs, especially in tumors with
disomy 3 (up to 30 and 50 % of disomy 3 tumors,
respectively) [1719]. These mutations in UMs ap-
peared to be largely mutually exclusively with BAP1
mutations, while in CMs these mutations were found
to be very infrequent (~1 %) [20].
It is currently unknown whether somatic mutations in
BAP1,SF3B1 and EIF1AX also characterize primary
LMNs. Using Sanger sequencing, we searched for muta-
tions in hotspot regions of SF3B1 (exon 14 and 15) and
exon 1 and 2 and flanking intronic regions of EIF1AX in
a series of 24 primary LMNs. Additionally, we per-
formed immunohistochemistry for the detection of
BAP1 protein loss as a surrogate marker for identifica-
tion of inactivating BAP1 mutations.
Materials and methods
Patients and histopathology
Formalin fixed and paraffin embedded (FFPE) tissue
samples of twenty-four primary LMNs were obtained
through the Dutch nationwide histopathology and cyto-
pathology data network and archive (PALGA) [21]. Hist-
ology was revised by HK; WHO 2007 criteria were used
for classification into melanocytoma, intermediate-grade
melanocytic tumor and melanoma [1]. Information on
mutation status of GNAQ,GNA11,BRAF,NRAS and
chromosome 3 status of cases #17, #10, #1318, and
#2224 has been published previously [2, 22, 23]. The
study was approved by the local ethics committees of
the Canisius Wilhelmina Hospital (ref.nr. LTC/TT/013
2015) and Radboud University (CMO, ref.nr/Dossier-
nummer: 20151692).
DNA extraction
Representative regions of about five 4-μm-thick FFPE sec-
tions with an estimated tumor cell percentage of at least
70 % were manually dissected and used for DNA extraction.
After heating in ATL buffer, the tissue sections were incu-
bated in proteinase K for one hour, followed by subsequent
purification of the DNA according to the manufacturer
(QIAamp DNA Mini Kit, QIAGEN GmbH, Germany).
After DNA purification, possible melanin contamination
was removed by using an inhibitor removal kit (OneStep
PCR Inhibitor Removal Kit, Zymo Research). The DNA
sample concentration was assessed spectrophotometrically
(Varian Cary 50 spectrophotometer, Agilent Technologies).
The integrity of the isolated DNA was tested by means of a
multiplex ladder PCR [24].
BAP1 immunohistochemistry
Immunohistochemistry was performed on 4-μm-thick
FFPE sections using an automated immunostainer
(Benchmark XT, Ventana Medical Systems, Tucson, AZ,
USA) according to the manufacturers instructions. Sec-
tions were immunostained with a primary monoclonal
antibody against BAP1 (clone C4, 1:100 dilution, Santa
Cruz Biotechnology, Dallas, TX, USA) using 3,3-diami-
nobenzidine (DAB) as chromogen. Selected cases that
showed a staining result that was difficult to interpret
including strongly pigmented tumors (patients #18,
#1216, #2224) were manually stained (clone C4, 1:50
dilution, Immunologic, Duiven, the Netherlands) accord-
ing to the manufacturers instructions. The VECTOR
NovaRED Peroxidase (HRP) Substrate Kit was used for
visualization (Vector Laboratories, USA, Catalogue
Number SK-4800). Nuclei of endothelial and lympho-
cytic cells in the slides served as positive internal control
for BAP1 protein expression. The staining results were
scored independently by three pathologists (HK, DC,
WB). The percentage of positive tumor cell nuclei was
scored only in areas with positive internal controls.
Mutation analysis
Sanger sequencing was used for analysis of mutations in
hotspot areas of SF3B1 including codon 625 and codon 700
as well as for mutations in exon 1 and 2 and flanking in-
tronic regions of EIF1AX. Primers are listed in Table 1. Nor-
mal tissue to exclude germline mutations was not available.
All primers contained a M13 forward or M13 reverse
consensus sequence for sequencing. PCR amplification
was performed in a total volume of 25 μl, containing
20 ng DNA, PCR Reaction Buffer with 20 mM MgCl2
(Roche), 200 μM of each deoxynucleotide triphosphate,
10 pmol of each primer and 2 units of FastStart Taq
DNA polymerase (Roche). DNA amplification was
performed in a Biometra T-Professional thermocycler
(Westburg). The PCR was started with 5 min. at 95 °C
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 2 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
and followed with 40 cycles of denaturation 30 s at 95 °C,
annealing at 60 °C for 45 s and extension at 72 °C for 30 s,
followed by a final extension at 72 °C for 10 min. and
cooling down for 5 min. at 22 °C. All PCR products were
purified with ExoSAP-IT (Affymetrix). Two μl of the PCR
product was used for the sequence reaction on an ABI
PRISM 3500XL DNA analyzer (Applied Biosystems). Both
strands were sequenced using the M13 primers.
Information on BAP1 mutation status was available for
three patients (#20, #23, #24) (obtained by Sanger sequen-
cing as previously described) while the chromosome 3 sta-
tus was in part obtained from a previous MLPA study [22,
25]. For all cases, the mutation status of GNAQ and
GNA11 (codons 209 and 183), BRAF (codon 600) and
NRAS (codons 12, 13 and 61) was available, part of this in-
formation has been published previously [2, 22, 23].
Results
Patients and histology
Main patient characteristics are summarized in Table 2.
The study group consisted of thirteen melanocytomas,
seven intermediate-grade melanocytic tumors and four
primary leptomeningeal melanomas. A subset of the tu-
mors was strongly pigmented.
All patients were adults, the age at operation ranging
from 2769 years. None of the patients had clinical evi-
dence of primary melanoma outside the CNS. One pa-
tient was known with a large congenital melanocytic
nevus in the buttocks in the context of neurocutaneous
melanocytosis (patient #21). Two of the patients in this
series developed liver metastases, both after an initial histo-
logical diagnosis of intermediate-grade melanocytic tumor
(patients #19 and #20). One melanoma patient developed
distant metastases to bones and lungs (patient #23).
BAP1 immunohistochemistry
BAP1 immunohistochemistry was available in 20 out of
24 cases. For three cases there was no FFPE material left
for immunohistochemistry (patients #911), while in
one patient the tumor showed intense pigmentation,
preventing reliable interpretation of staining results (pa-
tient #6). Only nuclear staining was considered as posi-
tive, although cytoplasmic staining was found in a
substantial subset of tumors as well. In two melanocyto-
mas (patients #1 and #4), a mosaic pattern with alternat-
ing positive and negative (areas of ) nuclear staining was
present, the vast majority (about 80 %) of these nuclei
being positive (Fig. 1a, b). All other cases showed mod-
erate to strong nuclear staining in 90 % or more of nu-
clei. Complete absence of nuclear BAP1 staining as is
typically seen in BAP1-mutated tumors was not ob-
served (Figs. 1, 2 and 3).
Mutation analysis
We detected a total of three mutations in SF3B1 (13 %),
including two hotspot mutations at codon 625 (R625C
and R625H) and one mutation affecting codon 634
(V634I) (Table 2). Mutations in codon 700 of SF3B1 were
not detected. In the cosmic database the V634I mutation
in SF3B1 (c.1900G > A (p.(Val634Ile))) has not been re-
ported in cancer before (http://cancer.sanger.ac.uk/cos
mic). This mutation was present in a GNAQ-mutated mel-
anocytoma of a 27-year-old male patient who developed
leptomeningeal seeding shortly after initial presentation
(patient #4) (Fig. 1a, b, e).
One case with mutation in codon 625 of SF3B1
(c.1873C > T (p.(Arg625Cys))) concerned an intermediate-
grade melanocytic tumor showing aggressive behavior
with leptomeningeal seeding (patient #18). A concomitant
GNA11 mutation was present in this tumor while immu-
nohistochemistry showed intact nuclear BAP1 expression
(Fig. 1c, d, f).
The other mutation in codon 625 of SF3B1
(c.1874G > A (p.(Arg625His))) was present in a CNS
melanoma of a 31-year-old woman (patient #21). Since
birth this patient was known with a large congenital mela-
nocytic nevus on the buttocks for which multiple excisions
had been performed, confirming melanocytic nevus with-
out malignant transformation. At the age of 29, she devel-
oped an intracranial tumor in the left frontal region with
diffuse leptomeningeal extension. Biopsy showed melan-
oma in a background of diffuse melanocytosis, consistent
with the diagnosis of primary melanoma of the CNS occur-
ring in the context of neurocutaneous melanocytosis. This
was supported by the presence of an NRAS mutation in
boththeCNSmelanomaandinamorerecentbiopsyof
the melanocytic nevus (c.182A > G (p.(Gln61Arg))). In con-
trast, the SF3B1 mutation (c.1874G > A (p.(Arg625His)))
was present in the CNS melanoma but not in the melano-
cytic nevus. Immunohistochemically, intact BAP1 protein
expression was found in the tumor cell nuclei of this
melanoma (Fig. 2).
A point mutation in EIF1AX was detected in five of
the primary LMNs in this series (21 %), each leading to
Table 1 Primers used for mutational analysis
Gene Exon Forward (Fw)
Reverse (Rv)
Primer sequence 5-3
SF3B1 14 Fw TGATTATGGAAAGAAATGGTTGAAG
Rv AGGGCAATAAAGAAGGAATGC
SF3B1 15 Fw TGCAGTTTGGCTGAATAGTTG
Rv GGCCAAAGCACTGATGGT
EIF1AX 1 Fw CCCCTCGGAGCAGCAG
Rv CCTGGGTGACCTGCAATCTAC
EIF1AX 2 Fw GCCTTAATTTCATTTTATTTCATACTGTTT
Rv AGGATGTTATTTTAAAAAGCGTAATTT
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 3 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Table 2 Patient characteristics and results of immunohistochemistry and mutation analyses
Patient Sex Age Diagnosis Location BAP1
immunostaining
Chrom 3 SF3B1 exon 14
(codon 625)
SF3B1 exon 15
(codon 700)
EIF1AX exon 1 EIF1AX exon 2 Follow-up
1 F 27 MC Right CPA + (80 %) disomy wt wt Intronc.1
4C > T
wt local recurrence
2
a
M 41 MC C0-C3 + disomy wt wt c.9G > C
(p.(Lys3Asn))
wt local recurrence and LM seeding 3 years
after diagnosis
3
a
M 47 MC Extramedullary,
intradural
+ disomy wt wt wt na local recurrence and LM seeding shortly after
initial presentation
4
a
M 27 MC Tentorium
cerebelli
+ (80 %) na c.1900G > A
(p.(Val634Ile))
wt wt na LM seeding shortly after initial presentation
5 M 38 MC C56 + na wt na na na tumor spread in neck and vertebra; deceased
6
a
M 41 MC Th6 na na wt wt na na local recurrence; no distant metastasis
7
a
V 57 MC Th11 + disomy wt wt wt wt unknown
8
b
F 62 MC C0-C1 + na wt wt wt wt unknown
9 F 23 MC Fossa posterior na na wt wt wt wt local recurrence after 2 years
10
a
M55MC C36 na na wt wt wt na local recurrence 8 years after diagnosis
11
a
M na MC Spinal region na na wt wt wt wt unknown
12 M 69 MC Conus
medullaris
+ na wt wt wt wt alive
13 M 37 MC Left CPA + na wt wt wt wt unknown
14
b
F 68 IMT Tentorium
cerebelli
+ disomy wt wt c.11A > G
(p.(Asn4Ser))
wt deceased (not disease related)
15
a
V 44 IMT Cauda + disomy wt wt wt wt stable (no recurrence)
16
a
M 41 IMT Intramedullary
(NOS)
+ disomy wt wt wt c.25G > C
(p.(Gly9Arg))
local recurrence and LM seeding
17
b
V 59 IMT Vermis
cerebelli
+ na wt wt wt wt unknown
18
b
F 30 IMT Th1011 + disomy c.1873C > T
(p.(Arg625Cys))
wt wt wt leptomeningeal seeding; deceased (disease
related)
19
a
F 53 IMT Th9 + na wt wt wt c.28A > G
(p.(Lys10Glu))
liver metastasis shortly after diagnosis
20
a,e
F 48 IMT Cervical spinal
region
+ disomy wt wt wt wt local recurrence after 3 years; distant
metastasis to liver and pancreas 1 year later;
deceased 5 years after initial presentation
21
c
F 31 MM Frontal left + disomy c.1874G > A
(p.(Arg625His))
d
wt wt neurocutaneous melanocytosis patient; the
SF3B1 mutation was only present in the CNS
melanoma and not in the congenital
melanocytic nevus of the skin
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 4 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Table 2 Patient characteristics and results of immunohistochemistry and mutation analyses (Continued)
22
a
M 62 MM Th79 + na na na c.9G > C
(p.(Lys3Asn))
na unknown
23
a,e
F 59 MM S2 + Monosomy
3
wt wt wt wt distant metastases after 2 years (bone, lungs);
liver metastasis unknown
24
a,e
M 55 MM L1-L2 + na wt wt wt wt leptomeningeal seeding 1 year after initial
presentation; no distant metastases; deceased
Information on GNAQ/GNA11 mutation status and chromosome 3 status of cases 17, 10, 1318, and 2224 has been published previously [2,22,23]
Ffemale, Mmale, MC melanocytoma, IMT intermediate-grade melanocytic tumor, MM melanoma, LM leptomeningeal, na not available (BAP1 immunohistochemistry of cases #911) or not assessable, CPA
cerebello-pontine angle
+ positive nuclear staining in 90 % or more of nuclei
a
GNAQ
Q209
or
b
GNA11
Q209
or
c
NRAS
Q61
mutation present
d
the SF3B1 mutation was not present in the congenital melanocytic nevus of this neurocutaneous melanocytosis patient
e
cases for which all 17 exons of the BAP1 gene were tested with Sanger sequencing and no mutations were detected
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 5 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
an amino acid substitution (at codons 3, 4, 9 or 10).
These mutations occurred in tumors diagnosed as mela-
nocytomas, intermediate-grade tumors, and melanomas
(Fig. 3). In addition, one point mutation was detected in
the Kozak consensus sequence of exon 1 (c.14C > T)
which may have an influence on the start of translation
(patient #1). EIF1AX mutations were mutually exclusive
with SF3B1 mutations and generally co-occurred with
GNAQ or GNA11 mutations (Table 2).
Discussion
In this study we investigated whether primary LMNs
share genetic alterations with UMs in addition to GNAQ
and GNA11 mutations. Recently, a role for BAP1 in
primary melanoma of the CNS was suggested based on
the identification of a BAP1 germline mutation in a pa-
tient with primary CNS melanoma with monosomy 3
and a family history of UM and meningioma [16]. In our
study, we chose for BAP1 immunohistochemistry as a
surrogate marker for the identification of an underlying
inactivating BAP1 mutation as it offers an economical
and faster alternative to sequence analysis of all 17
exons of BAP1 [12, 26, 27]. Typically, complete loss
of nuclear BAP1 expression is found in BAP1-mu-
tated UMs [12, 2628]. None of the samples in our
series showed such complete loss of nuclear BAP1
staining, suggesting absence of underlying BAP1 mu-
tations. In three cases (including the single patient
Fig. 1 Examples of SF3B1 mutation in a primary leptomeningeal melanocytoma and intermediate-grade melanocytic tumor. a. Hematoxylin-and-
Eosin (H&E) staining of the melanocytoma of patient #4 showing round to oval, slightly pleomorphic, vesicular nuclei, often with a prominent
nucleolus, and with dispersed melanin pigment in the cytoplasm of the tumor cells. b. BAP1 immunostaining of this melanocytoma revealed
heterogeneous staining of the tumor cell nuclei, the majority of these nuclei being positive. The arrows indicate nuclear staining in endothelial
cells. Note that the tumor cells also show variable cytoplasmic staining. c. H&E staining of the intermediate-grade melanocytic tumor of patient
#18 showing a proliferation of spindle cells with invasion in glial tissue (at the right). d. BAP1 immunostaining of the tumor depicted in (c) reveals
positive nuclei in more than 90 % of tumor cells. The arrows indicate nuclear staining in endothelial cells. e. Forward sequence tracing surrounding
codon 634 of SF3B1 showing the c.1900G > A (p.(Val634Ile)) mutation detected in the tumor depicted in (a)and(b). f. Forward sequence tracing
surrounding codon 625 of SF3B1 showing the c.1873C >T (p.(Arg625Cys)) mutation present in the tumor depicted in (c) and (d)
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 6 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
with monosomy 3 in the LMN and the patient with
liver metastases but disomy 3 in the LMN), BAP1
mutation status was available through Sanger sequen-
cing analyses and confirmed absence of mutations.
However, our study has some limitations. In some
cases, BAP1 mutations may still have been missed as
BAP1 immunohistochemistry was reported to have a
sensitivity of ~ 88 % [12]. Also, very rarely, heteroge-
neous (mosaic) BAP1 nuclear immunostaining has
been described in UM cases with a BAP1 mutation
showing loss of nuclear staining in only 20 % of nu-
clei [12]. In our series, two cases showed nuclear
BAP1 staining in about 80 % of tumor cells and an
underlying BAP1 mutation can thus not completely
beruledout.Furthermore,asBAP1 is mainly
implicated in metastatic UM with monosomy 3, there
might be a selection bias in our patient group as it
mainly concerns (relatively) low-grade tumors with di-
somy for chromosome 3. A larger number of primary
LMNs with monosomy 3 should thus be investigated
to further explore the role of BAP1 in these neo-
plasms. This is also important for therapeutic reasons
as epigenetic modulators such as histone deacetylase
(HDAC) inhibitors were shown to reverse the bio-
chemical effects of BAP1 mutations in UM cells by
inducing growth arrest and differentiation. Clinical
trials are now evaluating HDAC inhibitors as a thera-
peutic option in UM patients [13, 29].
Recurrent mutations in the SF3B1 gene have been de-
tected in several types of cancer such as UMs, breast
Fig. 2 SF3B1 mutation in primary leptomeningeal melanoma in a patient with neurocutaneous melanocytosis. a. H&E staining of the primary
LMN diagnosed as melanoma; the arrows indicate mitotic figures (patient #21). b. H&E staining of the congenital melanocytic nevus of the
buttocks; in the right lower corner higher magnification of the superficial part of the nevus with a proliferation of bland nevoid cells. c. BAP1
immunostaining of the melanoma depicted in (a) with nuclear staining in more than 90 % of tumor cells. d. Sequence tracing surrounding
codon 61 of NRAS showing a c.182A >G (p.(Gln61Arg)) mutation present in both the CNS melanoma as well as in the congenital melanocytic
nevus (reverse sequence/antisense strand). e. Forward sequence tracing surrounding codon 625 of SF3B1 showing a c.1874G > A (p.(Arg625His))
mutation in the CNS melanoma. f. The SF3B1 mutation depicted in (e) is absent in the congenital melanocytic nevus of this patient
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 7 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
and pancreatic carcinoma, and hematological diseases
like CLL and MDS [18, 3033]. These mutations affect
hotspot codons, the hotspot being associated with can-
cer type. For example, codon 700 mutations are fre-
quently present in CLL and MDS, while in UMs codon
625 is much more frequently involved [18, 34]. Espe-
cially in low-grade UMs with disomy for chromosome 3,
heterozygous mutations in SF3B1 are present in 10 to
30 % of UM [6, 1719]. The fact that in UMs these
SF3B1 mutations are almost mutually exclusive with
BAP1 mutations suggests different pathways in the
oncogenesis and/or malignant progression of these neo-
plasms. SF3B1 encodes subunit 1 of splicing factor 3b,
which is a component of the spliceosome that
participates in splicing of pre-mRNA. It was shown that
SF3B1 mutations are associated with differential alterna-
tive splicing of several protein encoding genes in UMs
[19]. Moreover, SF3B1 mutant cell lines were found to
be sensitive to the SF3b complex inhibitor spliceostatin
A, suggesting a new therapeutic target in tumors carry-
ing this mutation [32]. Up to now, it was unknown
whether SF3B1 mutations also occur in primary LMNs.
In our series, in three out of 24 cases (13 %) an SF3B1
mutation affecting codon 625 or 634 was detected, this
is at the lower end of the range of the frequency of
SF3B1 mutations reported in UMs (1030 %) and in
contrast to the very low frequency reported for CMs
(~1 %) [6, 1720]. As far as could be assessed, all three
Fig. 3 Examples of EIF1AX mutation in a primary leptomeningeal melanocytoma and intermediate-grade melanocytic tumor. a. H&E staining of
the melanocytoma of patient #2, consisting of epithelioid cells with moderate to strong melanin pigmentation. b. BAP1 immunostaining of the
melanocytoma depicted in A revealing positive staining of nuclei in more than 90 % of tumor cells. The arrows indicate nuclear staining in
endothelial cells. Note that cytoplasmic staining is also present. c. H&E staining of the intermediate-grade melanocytic tumor of patient #19 showing
a nested proliferation of epithelioid cells; the arrow indicates a mitotic figure. d. BAP1 immunostaining of the intermediate-grade melanocytic tumor
depicted in C revealing positive staining of nuclei in more than 90 % of tumor cells. e. Forward sequence tracing surrounding codon 3 of EIF1AX
showing a c.9G >C (p.(Lys3Asn)) mutation in the melanocytoma depicted in (a) and (b). f. Forward sequence tracing surrounding codon 10 of EIF1AX
showing a c.28A > G (p.(Lys10Glu)) mutation in the intermediate-grade melanocytic tumor depicted in (c) and (d)
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 8 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
LMNs showing an SF3B1 mutation were tumors with
disomy for chromosome 3. Furthermore, like in UMs, in
two cases the SF3B1 mutation co-occurred with a
GNAQ or GNA11 mutation, while in a third case (with
neurocutaneous melanocytosis) an NRAS mutation was
present. The GNAQ,GNA11 and NRAS genes are now
thought to play a role in the initiation of tumorigenesis,
while mutation in SF3B1 (or BAP1) would then occur in
a later phase of the oncogenic process [18]. The co-
occurrence of an NRAS and an SF3B1 mutation in the
CNS melanoma of the neurocutaneous melanocytosis
patient in our study is interesting in this respect as the
SF3B1 mutation was absent in the congenital melanocy-
tic nevus of this patient. Patients with neurocutaneous
melanocytosis have a large and/or multiple congenital
melanocytic nevi of the skin in association with a pri-
mary LMN [1]. Instead of GNAQ or GNA11 mutations,
these patients frequently demonstrate identical NRAS
mutations in both the congenital melanocytic nevus and
in the LMN [35, 36]. This is thought to be the result of
an NRAS-mutated clone of melanocyte precursors mi-
grating to skin and CNS early in embryogenesis [37].
The observation that the SF3B1 mutation was not
present in the melanocytic nevus of this patient suggests
that it indeed plays a role later on in tumorigenesis. In
addition, all three mutations in SF3B1 in our study were
found in tumors which clinically showed aggressive
behavior. However, the number of patients and the
follow-up in our study are too limited to allow for firm
conclusions about a prognostic role of SF3B1 in this set-
ting. Of note, in one of the three patients the SF3B1
mutation (c.1900G > A (p.(Val634Ile))) was present in a
neoplasm diagnosed as melanocytoma, suggesting that
SF3B1 mutations are not necessarily associated with
worrisome histology.
Several recent studies have reported mutations in
EIF1AX in different cancer types, including melanoma
and thyroid and ovarian cancer [17, 38, 39]. In UMs,
heterozygous mutations in exon 1 and 2 of EIF1AX have
reported to occur especially in tumors with disomy for
chromosome 3 (up to 48 % of these tumors) [17]. Muta-
tions may occur in different loci of these exons and lead
to amino acid substitutions or short deletions. Rarely
splice site mutations have been reported [6, 10, 17].
EIF1AX encodes the eukaryotic translation initiation fac-
tor 1A (eIF1A), which is involved in initiation phase of
translation of eukaryotic cells by stabilizing the forma-
tion of the functional ribosome around the AUG start
codon. The exact role of EIF1AX mutations in tumori-
genesis is currently not well understood but it has been
suggested that mutations in EIF1AX could diminish the
rate of bulk translation [17, 40]. In our series of primary
LMNs we found a relatively high frequency of EIF1AX
missense mutations (21 %) which is in the range
reported for UM (1948 %) [6, 17]. In contrast, EIF1AX
mutations are very rare in CM (5/231, ~2 %) [20]. As in
UMs, as far as could be assessed in our series, these mu-
tations occurred in primary LMNs with disomy for
chromosome 3 and were mutually exclusive with SF3B1
mutations, but co-occurred with GNAQ or GNA11 mu-
tations. In addition (and like SF3B1 mutations), the
EIF1AX mutations in our cases occurred both in mela-
nocytomas as well as melanomas, suggesting that they
are not necessarily associated with worrisome histology,
but the prognostic implications of these mutations re-
main to be elucidated. Finally, the EIF1AX and SF3B1
mutations in LMNs occurred in hotspot regions of these
genes, and in UMs such mutations were shown to be
somatic in origin [17, 18]. However, as non-neoplastic
tissue of the patients with LMNs was not available for
further testing, strictly speaking we cannot rule out the
possibility that in some of these cases it concerned a
germline mutation.
Conclusions
We report for the first time that a substantial subset of
LMNs carries a mutation in SF3B1 or EIF1AX. Like in
UMs, SF3B1 or EIF1AX mutations are mutually exclu-
sive and generally co-occur with either a GNAQ or
GNA11 mutation (SF3B1 occasionally with NRAS muta-
tion), suggesting that SF3B1 and EIF1AX mutations
occur later on in the tumorigenesis. Our findings may
offer novel therapeutic strategies for especially patients
with SF3B1-mutated LMNs. The role of BAP1 in the
pathogenesis of primary LMNs is less clear, immunohis-
tochemistry suggests that BAP1 mutation is infrequent
in tumors with disomy for chromosome 3. However,
more detailed analysis is needed for further elucidation
of the role of BAP1 in the oncogenesis of primary
LMNs. Demonstration of BAP1 mutations in primary
LMNs would potentially have therapeutic relevance as
well, as epigenetic modulators are now being evalu-
ated in patients with BAP1-mutant UMs. All in all,
this study thus underscores the genetic resemblance
of primary LMNs and UMs and provides some add-
itional clues for the diagnosis of and therapeutic
interference with these tumors.
Abbreviations
CNS: Central nervous system; CM: Cutaneous melanoma; FFPE: Formalin
fixed and paraffin embedded; HDAC: Histone deacetylase inhibitors;
LM: Leptomeningeal; LMN: Leptomeningeal melanocytic neoplasms;
UM: Uveal melanoma.
Competing interests
The authors declare that they have no competing interest.
Authorscontribution
HK, CP, WB, PW, BK, and PG contributed substantially to conception and
design of the study. All authors contributed substantially to acquisition of
data, and analysis and interpretation of data. HK wrote and edited the
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 9 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
manuscript. All authors contributed in drafting the manuscript and revising it
for intellectual content. MJ carried out the molecular genetic analyses. CP
supervised the molecular genetic analyses. DC, VW, WB, BK, and HK
supervised and scored the BAP1 immunohistochemistry. HK revised
histology. All authors read and approved the final version of the manuscript.
Acknowledgments
We thank Pieter Vandevelde for technical assistance with the lay-out of the
figures.
Author details
1
Department of Pathology, Canisius Wilhelmina Hospital, P.O. Box
90156500GS, Nijmegen, The Netherlands.
2
Department of Pathology, Ghent
University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
3
Department of
Pathology, Radboud University Medical Center, P.O. Box 91016500HB,
Nijmegen, The Netherlands.
4
Department of Pathology, Maastricht University
Medical Center, P.O. Box 58006202AZ, Maastricht, The Netherlands.
5
Department of Pathology, VU University Medical Center, P.O. Box
70571007MB, Amsterdam, The Netherlands.
Received: 6 December 2015 Accepted: 4 January 2016
References
1. Brat DJ, Perry A. Melanocytic lesions. In: Louis DN, Ohgaki H, Wiestler OD,
Cavenee WK, editors. WHO classification of tumours of the central nervous
system. Lyon: IARC; 2007. p. 1813.
2. Kusters-Vandevelde HV, Klaasen A, Kusters B, Groenen PJ, van Engen-van
Grunsven IA, van Dijk MR, et al. Activating mutations of the GNAQ gene: a
frequent event in primary melanocytic neoplasms of the central nervous
system. Acta Neuropathol. 2010;119:31723. doi:10.1007/s00401-009-0611-3.
3. Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, OBrien JM,
et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue
naevi. Nature. 2009;457:599602. doi:10.1038/nature07586.
4. Murali R, Wiesner T, Rosenblum MK, Bastian BC. GNAQ and GNA11
mutations in melanocytomas of the central nervous system. Acta
Neuropathol. 2012;123:4579. doi:10.1007/s00401-012-0948-x.
5. Gessi M, van de Nes J, Griewank K, Barresi V, Buckland ME, Kirfel J, et al.
Absence of TERT promoter mutations in primary melanocytic tumours of
the central nervous system. Neuropathol Appl Neurobiol. 2014;40:7947.
doi:10.1111/nan.12138.
6. Dono M, Angelini G, Cecconi M, Amaro A, Esposito AI, Mirisola V, et al.
Mutation frequencies of GNAQ, GNA11, BAP1, SF3B1, EIF1AX and TERT in
uveal melanoma: detection of an activating mutation in the TERT gene
promoter in a single case of uveal melanoma. Br J Cancer. 2014;110:105865.
doi:10.1038/bjc.2013.804.
7. Van Raamsdonk CD, Griewank KG, Crosby MB, Garrido MC, Vemula S,
Wiesner T, et al. Mutations in GNA11 in Uveal Melanoma. N Engl J Med.
2010;363:21919. doi:10.1059/NEJMoa1000584.
8. Griewank KG, Murali R, Puig-Butille JA, Schilling B, Livingstone E, Potrony M,
et al. TERT promoter mutation status as an independent prognostic factor in
cutaneous melanoma. J Natl Cancer Inst. 2014;106:doi:10.1093/jnci/dju246.
9. Harbour JW, Onken MD, Roberson ED, Duan S, Cao L, Worley LA, et al.
Frequent mutation of BAP1 in metastasizing uveal melanomas. Science.
2010;330:14103. doi:10.1126/science.1194472.
10. Ewens KG, Kanetsky PA, Richards-Yutz J, Purrazzella J, Shields CL, Ganguly T,
et al. Chromosome 3 status combined with BAP1 and EIF1AX mutation
profiles are associated with metastasis in uveal melanoma. Invest
Ophthalmol Vis Sci. 2014;55:51607. doi:10.1167/iovs.14-14550.
11. Scheuermann JC, de Ayala Alonso AG, Oktaba K, Ly-Hartig N, McGinty RK,
Fraterman S, et al. Histone H2A deubiquitinase activity of the Polycomb
repressive complex PR-DUB. Nature. 2010;465:2437. doi:10.1038/nature08966.
12. Koopmans AE, Verdijk RM, Brouwer RW, van den Bosch TP, van den Berg
MM, Vaarwater J, et al. Clinical significance of immunohistochemistry for
detection of BAP1 mutations in uveal melanoma. Mod Pathol. 2014;27:
132130. doi:10.1038/modpathol.2014.43.
13. Field MG, Harbour JW. Recent developments in prognostic and predictive
testing in uveal melanoma. Curr Opin Ophthalmol. 2014;25:2349. doi:10.
1097/ICU.0000000000000051.
14. Abdel-Rahman MH, Pilarski R, Cebulla CM, Massengill JB, Christopher BN,
Boru G, et al. Germline BAP1 mutation predisposes to uveal melanoma,
lung adenocarcinoma, meningioma, and other cancers. J Med Genet. 2011;
48:8569. doi:10.1136/jmedgenet-2011-100156.
15. Carbone M, Ferris LK, Baumann F, Napolitano A, Lum CA, Flores EG, et al.
BAP1 cancer syndrome: malignant mesothelioma, uveal and cutaneous
melanoma, and MBAITs. J Transl Med. 2012;10:179. doi:10.1186/1479-5876-10-179.
16. de la Fouchardiere A, Cabaret O, Petre J, Aydin S, Leroy A, de Potter P,
et al. Primary leptomeningeal melanoma is part of the BAP1-related cancer
syndrome. Acta Neuropathol. 2015;129:9213. doi:10.1007/s00401-015-1423-2.
17. Martin M, Masshofer L, Temming P, Rahmann S, Metz C, Bornfeld N, et al.
Exome sequencing identifies recurrent somatic mutations in EIF1AX and
SF3B1 in uveal melanoma with disomy 3. Nat Genet. 2013;45:9336.
doi:10.1038/ng.2674.
18. Harbour JW, Roberson ED, Anbunathan H, Onken MD, Worley LA, Bowcock
AM. Recurrent mutations at codon 625 of the splicing factor SF3B1 in uveal
melanoma. Nat Genet. 2013;45:1335. doi:10.1038/ng.2523.
19. Furney SJ, Pedersen M, Gentien D, Dumont AG, Rapinat A, Desjardins L, et
al. SF3B1 mutations are associated with alternative splicing in uveal
melanoma. Cancer Discov. 2013;3:11229. doi:10.1158/2159-8290.cd-13-0330.
20. Kong Y, Krauthammer M, Halaban R. Rare SF3B1 R625 mutations in
cutaneous melanoma. Melanoma Res. 2014;24:3324. doi:10.1097/cmr.
0000000000000071.
21. Casparie M, Tiebosch AT, Burger G, Blauwgeers H, van de Pol A, van Krieken
JH, et al. Pathology databanking and biobanking in The Netherlands, a
central role for PALGA, the nationwide histopathology and cytopathology
data network and archive. Cell Oncol. 2007;29:1924.
22. Kusters-Vandevelde HV, van Engen-van Grunsven IA, Coupland SE, Lake SL,
Rijntjes J, Pfundt R, et al. Mutations in g protein encoding genes and
chromosomal alterations in primary leptomeningeal melanocytic neoplasms.
Pathol Oncol Res. 2015;21:43947. doi:10.1007/s12253-014-9841-3.
23. Kusters-Vandevelde HV, van Engen-van Grunsven IA, Kusters B, van Dijk MR,
Groenen PJ, Wesseling P, et al. Improved discrimination of melanotic
schwannoma from melanocytic lesions by combined morphological
and GNAQ mutational analysis. Acta Neuropathol. 2010;120:75564.
doi:10.1007/s00401-010-0749-z.
24. van Dongen JJ, Langerak AW, Bruggemann M, Evans PA, Hummel M,
Lavender FL, et al. Design and standardization of PCR primers and protocols
for detection of clonal immunoglobulin and T-cell receptor gene
recombinations in suspect lymphoproliferations: report of the BIOMED-2
Concerted Action BMH4-CT98-3936. Leukemia. 2003;17:2257317.
doi:10.1038/sj.leu.2403202.
25. van Engen-van Grunsven AC, Baar MP, Pfundt R, Rijntjes J, Kusters-
Vandevelde HV, Delbecq AL, et al. Whole-genome copy-number analysis
identifies new leads for chromosomal aberrations involved in the
oncogenesis and metastastic behavior of uveal melanomas. Melanoma Res.
2015;25:2009. doi:10.1097/CMR.0000000000000152.
26. Kalirai H, Dodson A, Faqir S, Damato BE, Coupland SE. Lack of BAP1 protein
expression in uveal melanoma is associated with increased metastatic risk
and has utility in routine prognostic testing. Br J Cancer. 2014;111:137380.
doi:10.1038/bjc.2014.417.
27. Shah AA, Bourne TD, Murali R. BAP1 protein loss by immunohistochemistry:
a potentially useful tool for prognostic prediction in patients with uveal
melanoma. Pathology. 2013;45:6516. doi:10.1097/pat.0000000000000002.
28. van Essen TH, van Pelt SI, Versluis M, Bronkhorst IH, van Duinen SG,
Marinkovic M, et al. Prognostic parameters in uveal melanoma and their
association with BAP1 expression. Br J Ophthalmol. 2014;98:173843.
doi:10.1136/bjophthalmol-2014-305047.
29. Landreville S, Agapova OA, Matatall KA, Kneass ZT, Onken MD, Lee RS, et al.
Histone deacetylase inhibitors induce growth arrest and differentiation
in uveal melanoma. Clin Cancer Res. 2012;18:40816. doi:10.1158/1078-
0432.ccr-11-0946.
30. Yoshida K, Ogawa S. Splicing factor mutations and cancer. Wiley Interdiscip
Rev RNA. 2014;5:44559. doi:10.1002/wrna.1222.
31. Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL,
et al. Pancreatic cancer genomes reveal aberrations in axon guidance
pathway genes. Nature. 2012;491:399405. doi:10.1038/nature11547.
32. Maguire SL, Leonidou A, Wai P, Marchio C, Ng CK, Sapino A, et al. SF3B1
mutations constitute a novel therapeutic target in breast cancer. J Pathol.
2015;235:57180. doi:10.1002/path.4483.
33. Wang L, Lawrence MS, Wan Y, Stojanov P, Sougnez C, Stevenson K, et al.
SF3B1 and other novel cancer genes in chronic lymphocytic leukemia.
N Engl J Med. 2011;365:2497506. doi:10.1056/NEJMoa1109016.
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 10 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
34. Malcovati L, Papaemmanuil E, Bowen DT, Boultwood J, Della Porta MG,
Pascutto C, et al. Clinical significance of SF3B1 mutations in myelodysplastic
syndromes and myelodysplastic/myeloproliferative neoplasms. Blood. 2011;
118:623946. doi:10.1182/blood-2011-09-377275.
35. Pedersen M, Kusters-Vandevelde HV, Viros A, Groenen PJ, Sanchez-Laorden
B, Gilhuis JH, et al. Primary melanoma of the CNS in children is driven by
congenital expression of oncogenic NRAS in melanocytes. Cancer Discov.
2013;3:45869. doi:10.1158/2159-8290.CD-12-0464.
36. Kinsler VA, Thomas AC, Ishida M, Bulstrode NW, Loughlin S, Hing S, et al.
Multiple congenital melanocytic nevi and neurocutaneous melanosis are
caused by postzygotic mutations in codon 61 of NRAS. J Invest Dermatol.
2013;133:222936. doi:10.1038/jid.2013.70.
37. Kusters-Vandevelde HV, Kusters B, van Engen-van Grunsven AC, Groenen PJ,
Wesseling P, Blokx WA. Primary melanocytic tumors of the central nervous
system: a review with focus on molecular aspects. Brain Pathol. 2015;25:
20926. doi:10.1111/bpa.12241.
38. Hunter SM, Anglesio MS, Ryland GL, Sharma R, Chiew YE, Rowley SM, et al.
Molecular profiling of low grade serous ovarian tumours identifies novel
candidate driver genes. Oncotarget. 2015;6:3766377. doi:10.18632/
oncotarget.5438.
39. Kunstman JW, Juhlin CC, Goh G, Brown TC, Stenman A, Healy JM, et al.
Characterization of the mutational landscape of anaplastic thyroid
cancer via whole-exome sequencing. Hum Mol Genet. 2015;24:231829.
doi:10.1093/hmg/ddu749.
40. Chaudhuri J, Si K, Maitra U. Function of eukaryotic translation initiation
factor 1A (eIF1A) (formerly called eIF-4C) in initiation of protein synthesis.
J Biol Chem. 1997;272:788391.
We accept pre-submission inquiries
Our selector tool helps you to find the most relevant journal
We provide round the clock customer support
Convenient online submission
Thorough peer review
Inclusion in PubMed and all major indexing services
Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
Küsters-Vandevelde et al. Acta Neuropathologica Communications (2016) 4:5 Page 11 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... [1][2][3][4][5][6][7][8] Secondary events found in this setting are mainly the inactivation of BAP1, and alterations of SF3B1 and EIF1AX, or inactivation of PRKAR1A. 3,[9][10][11] They are thought to contribute toward malignant progression. [11][12][13] In the present study, we report 4 cases of blue melanocytic neoplasms lacking GNAQ, GNA11, CYSLTR2, PLCB4 mutations, or PRKCA gene fusions but harboring a GRM1 gene fusion, which could represent a new oncogenic driver. ...
Article
Activating mutations in GNAQ, GNA11, CYSLTR2, and PLCB4 genes are regarded as the main oncogenic drivers of blue nevi (BN) and blue malignant melanocytic tumors. Here we report four cases of blue melanocytic neoplasms devoid of these mutations but harboring GRM1 gene fusions. In this short series, there was no gender predominance (sex ratio = 1). The mean age at diagnosis was 40 years (range: 12-72). Tumors were located on the face (n=2), forearm (n=1) and dorsum of the foot (n=1). Clinically, a plaque-like pre-existing blue nevus was found in 2 cases, including a deep location; another case presented as an Ota nevus. Two cases were diagnosed as melanoma ex-blue nevus, one as an atypical BN, and one as a plaque-like BN. Microscopic examination revealed a dermal proliferation of dendritic melanocytes in a sclerotic stroma. A dermal cellular nodule with atypia and mitotic activity was observed in three cases. Genetic investigation by whole-exome RNA sequencing revealed MYO10::GRM1 (n=2) and ZEB2::GRM1 (n=1) fusions. A GRM1-rearrangement was identified by FISH in the remaining case. SF3B1 co-mutations were present in the two melanomas, which both had a MYO10::GRM1 fusion. Array-CGH was feasible for three cases and displayed multiple copy number alterations (CNAs) in the two melanomas and limited CNAs in the atypical BN, all genomic profiles being compatible with those of classical blue lesions. GRM1 was overexpressed in all cases compared to a control group of blue lesions with other typical mutations. Both melanomas rapidly developed visceral metastases following diagnosis, with a fatal outcome in one case and tumor progression under palliative care in the other case. This data suggests that GRM1 gene fusions could represent an additional rare oncogenic driver in the setting of blue nevi, mutually exclusive of classical canonical mutations, especially in plaque-type or Ota subtypes.
... However, UM tumors with an EIF1AX mutation only express the mutant allele, suggesting that EIF1AX may also function in a recessive manner [64]. Mutations in this gene have also been found in thyroid and ovarian cancers, as well as the rare neoplasm primary leptomeningeal melanocytic neoplasms (LMNs) [111,112]. ...
Article
Full-text available
Simple Summary Uveal melanoma (UM) is a rare eye cancer with a high mortality rate due to metastases, leading to death in up to 50% of patients within 10 years from UM diagnosis. Moreover, patients show a median survival of 6 to 12 months after metastasis diagnosis. UM and cutaneous melanoma (CM) have the same melanocytic origin; however, they are very different in terms of molecular alterations and biological behavior. In this review, we will discuss the complex genetic and non-coding RNA-based epigenetic landscapes underlying the transformation, progression, and dissemination of UM. This knowledge will pave the way for the future identification of new biomarkers of the pathology and therapeutic targets. Abstract Uveal melanoma (UM) is the most common intraocular malignant tumor and the most frequent melanoma not affecting the skin. While the rate of UM occurrence is relatively low, about 50% of patients develop metastasis, primarily to the liver, with lethal outcome despite medical treatment. Notwithstanding that UM etiopathogenesis is still under investigation, a set of known mutations and chromosomal aberrations are associated with its pathogenesis and have a relevant prognostic value. The most frequently mutated genes are BAP1, EIF1AX, GNA11, GNAQ, and SF3B1, with mutually exclusive mutations occurring in GNAQ and GNA11, and almost mutually exclusive ones in BAP1 and SF3B1, and BAP1 and EIF1AX. Among chromosomal aberrations, monosomy of chromosome 3 is the most frequent, followed by gain of chromosome 8q, and full or partial loss of chromosomes 1 and 6. In addition, epigenetic mechanisms regulated by non-coding RNAs (ncRNA), namely microRNAs and long non-coding RNAs, have also been investigated. Several papers investigating the role of ncRNAs in UM have reported that their dysregulated expression affects cancer-related processes in both in vitro and in vivo models. This review will summarize current findings about genetic mutations, chromosomal aberrations, and ncRNA dysregulation establishing UM biology.
... The passenger anomalies found in the progression of blue naevi towards malignancy are also similar to those found in uveal and leptomeningeal melanomas and frequently involve BAP1, SF3B1 and EI1FAX. 3,20,21 Recently, inactivating mutations in PRKAR1A, the genetic hallmark of pigmented epithelioid melanocytomas, have also been described in the blue setting. [22][23][24] ...
Article
Blue naevi (BN) form a wide group of benign dermal melanocytic proliferations. They are genetically distinct from common and Spitz naevi with frequent hotspot mutations occurring in Gαq genes. Clinically, BN display a female predominance, elective sites of emergence and a great variety of subtypes related to specific regions of the skin linked to early embryological genetic events. Histologically, most BN are located in the dermis with small, bland, spindled and dendritic pigmented melanocytes within a fibrous background. Variation in tumour volume, fibrosis, and melanin pigment load can be broad. A growth in size and cellularity can occur within a subset of tumours as they acquire the morphological features of cellular blue naevi, with a biphasic architecture associating a dendritic blue naevus morphology near the surface, and deep vertical cellular expansions of medium-sized, bland melanocytes often reaching the subcutis. Sclerosing and myxoid variants can be observed either as individual or combined modifications that can add complexity to an otherwise straightforward diagnosis. Malignant progression of a cellular blue naevus is exceptional with an intermediate stage named atypical cellular blue naevus. Malignant blue melanomas are fast growing, large, pigmented tumours with most often obvious features of malignancy. However, they are difficult to separate from other malignant dermal melanocytic proliferations. Herein, we will extensively detail and illustrate the clinical, histological and genetic features of the vast spectrum of blue naevi and related entities in the skin.
... Additionally, molecular analysis of PLCB4 and CYS-LTR2 [59][60][61] and methylation profiling are especially useful in discriminating these lesions from other pigmented CNS tumors [1,59]. The presence of SF3B1-, EIF1AX-, or BAP1-mutation or complex copy number variations indicate aggressive behavior consistent with meningeal melanoma [62][63][64]. Especially in children, meningeal melanocytosis and melanomatosis are characterized by NRAS-and occasionally BRAF mutations [65][66][67]. ...
Article
Full-text available
Background: Meningeal melanocytomas (MM) are rare primary melanocytic tumors of the leptomeninges with an incidence of 1:10,000,000. Until now, there has been only sparse information about this tumor entity. Here, we provide a meta-analysis of all cases published in the English language since 1972. Methods: A literature review was performed using PubMed and Web of Science. All published cases were evaluated for location, sex, age, therapeutic approach, and outcome. In total, we included 201 patient cases in our meta-analysis. Results: The majority of MM was diagnosed more frequently in men between the third and fifth decade of life. Surgery is the preferred therapeutic approach, and total resection is associated with the best outcome. Patients with partial resection or tumor recurrence benefit from adjuvant radiotherapy, whereas chemo- or immunotherapies do not improve the disease course. Malignant transformation was described in 18 patients. Of these, 11 patients developed metastasis. Conclusions: We present the first retrospective meta-analysis of all MM cases published in the English language, including an evaluation of different treatment strategies allowing us to suggest a novel treatment guideline highlighting the importance of total resection for recurrence-free survival and characterizing those cases which benefit from adjuvant radiotherapy.
Article
Full-text available
Alternative splicing (AS) is an important molecular biological mechanism regulated by complex mechanisms involving a plethora of cis and trans-acting elements. Furthermore, AS is tissue specific and altered in various pathologies, including infectious, inflammatory, and neoplastic diseases. Recently developed immuno-oncological therapies include monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR) T cells targeting, among others, immune checkpoint (ICP) molecules. Despite therapeutic successes have been demonstrated, only a limited number of patients showed long-term benefit from these therapies with tumor entity-related differential response rates were observed. Interestingly, splice variants of common immunotherapeutic targets generated by AS are able to completely escape and/or reduce the efficacy of mAb- and/or CAR-based tumor immunotherapies. Therefore, the analyses of splicing patterns of targeted molecules in tumor specimens prior to therapy might help correct stratification, thereby increasing therapy success by antibody panel selection and antibody dosages. In addition, the expression of certain splicing factors has been linked with the patients’ outcome, thereby highlighting their putative prognostic potential. Outstanding questions are addressed to translate the findings into clinical application. This review article provides an overview of the role of AS in (tumor) diseases, its molecular mechanisms, clinical relevance, and therapy response.
Chapter
Epidemiology: Central nervous system (CNS) cancers affect a staggering percentage of the population across all ages accounting to 1.6% of global incidence of all cancers [1]. CNS cancer ranks tenth in terms of mortality [1]. Eastern Asia has the highest cases and fatality rate, followed by South-Central Asia and Northern America. According to GLOBOCAN, males show higher global incidence (168,346 cases; cumulative risk: 0.40) and mortality rates (138,277 deaths; cumulative risk 0.34) than females (incidence: 139,756 cases; cumulative risk 0.31; mortality: 113,052 deaths; cumulative risk 0.26). In the United States, the median age of diagnosis and death are 59 and 66, respectively and the age-adjusted rate of incidence was 7.08 per 100,000 during 2013–2017 [2]. Malignant tumors constitute 29.7% and nonmalignant tumors comprise 70.3% of all the brain and CNS cancers [3]. The frequencies of nonmalignant tumors also vary by type: nonmalignant meningioma (37.9%) and nonmalignant glioma (1.0%) [3]. Besides the incidence, it is found that primary brain malignancies are more frequent in the frontal region (24.3%) followed by the temporal (17.5%), parietal (10.4%), and occipital (2.6%) areas making up to 54.8% of the total [3].
Article
Primary central nervous system (CNS) melanoma is an extremely rare condition, with an incidence rate of 0.01 per 100,000 individuals per year. Despite its rarity, the etiology and pathogenesis of this disease are not yet fully understood. Primary CNS melanoma exhibits highly aggressive biological behavior and presents clinically in a distinct manner from other types of melanomas. It can develop at any age, predominantly affecting the meninges as the primary site, with clinical symptoms varying depending on the neoplasm’s location. Due to the lack of specificity in its presentation and the challenging nature of imaging diagnosis, distinguishing primary CNS melanoma from other CNS diseases. The combination of challenges in early detection, heightened tumor aggressiveness, and the obscured location of its origin contribute to an unfavorable prognostic outcome. Furthermore, there has been currently no consensus on a standardized treatment approach for primary CNS melanoma. Despite recent advancements in targeted therapy and immunotherapy for CNS melanoma, patients with primary CNS melanoma have limited treatment options due to their inadequate response to these therapies. Here, we provided a comprehensive summary of the epidemiology, clinical features, molecular pathological manifestations, and available diagnostic and therapeutic approaches of primary CNS melanoma. Additionally, we proposed potential therapeutic strategies for it.
Article
Full-text available
Alternative pre-mRNA splicing is a critical mechanism that generates multiple mRNA from a single gene, thereby increasing the diversity of the proteome. Recent research has highlighted the significance of specific splicing isoforms in cellular processes, particularly in regulating cell numbers. In this review, we examine the current understanding of the role of alternative splicing in controlling cancer cell growth and discuss specific splicing factors and isoforms and their molecular mechanisms in cancer progression. These isoforms have been found to intricately control signaling pathways crucial for cell cycle progression, proliferation, and apoptosis. Furthermore, studies have elucidated the characteristics and functional importance of splicing factors that influence cell numbers. Abnormal expression of oncogenic splicing isoforms and splicing factors, as well as disruptions in splicing caused by genetic mutations, have been implicated in the development and progression of tumors. Collectively, these findings provide valuable insights into the complex interplay between alternative splicing and cell proliferation, thereby suggesting the potential of alternative splicing as a therapeutic target for cancer.
Chapter
Melanoma is the third most common skin malignancy but is the one with the highest propensity to metastasize, potentially able to disseminate to any organ. Spreading to the central nervous system has been historically regarded as an ominous event due to the poor prognosis of the affected patients. Magnetic resonance is fundamental for screening and diagnosis, although histological analysis remains invaluable. Management relies on a multidisciplinary discussion between oncologists, neurosurgeons, and radiologists. Recent trials with dual checkpoint inhibitors have expanded the panorama of treatment options for these patients and are contributing to reshape our conception and approach. With a growing number of immunotherapy agents and small molecules inhibitors available, future trials will help define the optimal combinations and sequence, as well as to implement these therapies with radiosurgery. A further understanding of disease pathogenesis will promote new strategies to overcome tumor resistance and develop personalized treatment plan based on the tumoral signature and the microenvironment phenotype.
Article
Full-text available
Low grade serous ovarian tumours are a rare and under-characterised histological subtype of epithelial ovarian tumours, with little known of the molecular drivers and facilitators of tumorigenesis beyond classic oncogenic RAS/RAF mutations. With a move towards targeted therapies due to the chemoresistant nature of this subtype, it is pertinent to more fully characterise the genetic events driving this tumour type, some of which may influence response to therapy and/or development of drug resistance. We performed genome-wide high-resolution genomic copy number analysis (Affymetrix SNP6.0) and mutation hotspot screening (KRAS, BRAF, NRAS, HRAS, ERBB2 and TP53) to compare a large cohort of ovarian serous borderline tumours (SBTs, n = 57) with low grade serous carcinomas (LGSCs, n = 19). Whole exome sequencing was performed for 13 SBTs, nine LGSCs and one mixed low/high grade carcinoma. Copy number aberrations were detected in 61% (35/57) of SBTs, compared to 100% (19/19) of LGSCs. Oncogenic RAS/RAF/ERBB2 mutations were detected in 82.5% (47/57) of SBTs compared to 63% (12/19) of LGSCs, with NRAS mutations detected only in LGSC. Some copy number aberrations appeared to be enriched in LGSC, most significantly loss of 9p and homozygous deletions of the CDKN2A/2B locus. Exome sequencing identified BRAF, KRAS, NRAS, USP9X and EIF1AX as the most frequently mutated genes. We have identified markers of progression from borderline to LGSC and novel drivers of LGSC. USP9X and EIF1AX have both been linked to regulation of mTOR, suggesting that mTOR inhibitors may be a key companion treatment for targeted therapy trials of MEK and RAF inhibitors.
Article
Full-text available
Primary melanocytic tumors of the central nervous system (CNS) represent a spectrum of rare tumors. They can be benign or malignant and occur in adults as well as in children, the latter often in the context of neurocutaneous melanosis. Until recently, the genetic alterations in these tumors were largely unknown. This is in contrast with cutaneous and uveal melanomas, which are known to harbor distinct oncogenic mutations that can be used as targets for treatment with small molecule inhibitors in the advanced setting. Recently, novel insights in the molecular alterations underlying primary melanocytic tumors of the CNS were obtained, including different oncogenic mutations in tumors in adult patients (esp. GNAQ, GNA11) versus children (esp. NRAS). In this review, the focus is on molecular characteristics of primary melanocytic tumors of the CNS. We summarize what is known about their genetic alterations and discuss implications for pathogenesis and differential diagnosis with other pigmented tumors in or around the CNS. Finally, new therapeutic options with targeted therapy are discussed. This article is protected by copyright. All rights reserved.
Article
Full-text available
Mutations in genes encoding proteins involved in RNA splicing have been found to occur at relatively high frequencies in several tumour types including myelodysplastic syndromes, chronic lymphocytic leukaemia, uveal melanoma, pancreatic cancer and at lower frequencies in breast cancer. To investigate whether dysfunction in RNA splicing is implicated in the pathogenesis of breast cancer, we performed a re-analysis of published exome and whole genome sequencing data. This analysis revealed that mutations in spliceosomal component genes occurred in 5.6% of unselected breast cancers, including hotspot mutations in the SF3B1 gene, which were found in 1.8% of unselected breast cancers. SF3B1 mutations were significantly associated with ER-positive disease, AKT1 mutations and distinct copy number alterations. Additional profiling of hotspot mutations in a panel of special histological subtypes of breast cancer showed that 16% and 6% of papillary and mucinous carcinomas of the breast harboured the SF3B1 K700E mutation. RNA sequencing identified differentially spliced events expressed in tumours with SF3B1 mutations including the protein coding genes TMEM14C, RPL31, DYNL111, UQCC and ABCC5 and the long non-coding RNA CRNDE. Moreover, SF3B1 mutant cell lines were found to be sensitive to the SF3b complex inhibitor spliceostatin A and treatment resulted in perturbation of the splicing signature. Albeit rare, SF3B1 mutations result in alternative splicing events, and may constitute drivers and a novel therapeutic target in a subset of breast cancers.
Article
Myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms Myelodysplastic syndromes (MDS) are a group of clonal disorders of the hematopoietic stem cell (HSC) characterized by ineffective hematopoiesis. This leads to peripheral blood cytopenias, and an excess of marrow blast cells. In some cases this can progress to acute myeloid leukemia (AML). Myelodysplastic syndromes are thought to result from the accumulation of genetic or epigenetic lesions (such as promoter hypermethylation) initially in a very immature progenitor cell. This leads to a proliferative advantage of the MDS clone over normal immature progenitors. MDS lineage-committed progenitors display both abnormal terminal differentiation and an increased susceptibility to apoptosis. These two features explain the clinical consequences of blast cell accumulation and peripheral blood cytopenias. The diagnosis of MDS is generally suggested by the presence of peripheral blood (PB) cytopenias. However, the diagnosis requires the integration of the blood manifestations with the clinical history and physical examination. Although the etiology of MDS is generally unknown, antecedent cytotoxic therapy or radiation exposure and congenital bone marrow failure syndromes can predispose to MDS. Physical examination is in general normal in MDS, while splenomegaly can be found in myelodysplastic/myeloproliferative neoplasms (MDS/MPN). Careful morphological review of bone marrow for dysplasia is central to the diagnosis of MDS and MDS/MPN, and cytogenetic analysis for prognosis. Molecular genetics and flow cytometry assays are under evaluation, and, in future, may provide information for further refinement of the prognosis. The specific features of each of these diagnostic modalities will be discussed.
Article
The BRCA1-associated Protein 1 (BAP1) gene encodes a ubiquitin hydrolase that plays a role in several key cellular processes, including transcriptional regulation, growth, DNA damage response, and chromatin dynamics [1]. Heterozygous germline BAP1 mutations were initially associated with a higher risk of mesothelioma and uveal melanoma, as well as multiple atypical cutaneous melanocytic tumors. However, recent reports have extended their clinical phenotype to others tumors such as clear cell renal cancer, lung adenocarcinoma, meningioma, paraganglioma, and multiple basal cell carcinomas [1, 4].In this case report, a 40-year-old woman, with a recent history of seizures, was diagnosed with a supratentorial brain mass. Surgical exploration revealed a heavily pigmented, 5-cm mass adhered to the leptomeninges. Pathological analysis and immunohistochemistry confirmed a malignant melanoma. Due to the absence of a cutaneous or ocular melanoma, a diagnosis of primary leptomeningeal melanoma was ...
Article
To further elucidate the genetic underpinnings of uveal melanoma (UM) and identify new markers that correlate with disease outcome, archival formalin-fixed, paraffin-embedded enucleation specimens from 25 patients with UM and a mean follow-up of 14 years were analyzed for whole-genome copy-number alterations using OncoScan analysis. Copy-number alterations of chromosomes 1, 3, 6, and 8 were also analyzed in these tumors using multiplex ligation-dependent probe-amplification, and mutations in GNAQ, GNA11, and BAP1 were searched for by Sanger sequencing. Our study confirms the previously reported GNAQ and GNA11 mutation frequencies in UMs as well as the presence of monosomy 3 as a factor strongly indicating poor prognosis. Two cases with metastatic disease, but without monosomy of chromosome 3, showed loss of a small region in the distal part of chromosome 2p. Also, UMs leading to metastatic disease had more chromosomal aberrations than those without metastases. Three UMs lacking a GNAQ or a GNA11 mutation showed a gain of chromosome 8q; one of these cases showed extensive chromothripsis. Another case (with suspect lung metastasis) showed focal chromothripsis. Our whole-genome copy-number analysis shows that focal loss of chromosome 2p may be involved in the metastatic spread of UMs without monosomy 3; metastatic UMs carry more chromosomal aberrations than those without metastases; and chromothripsis may play a role in the oncogenesis of UMs, but does not necessarily indicate a poor prognosis. The clinical and particularly diagnostic utility of these findings needs to be corroborated in a larger set of patients with UM.
Article
Anaplastic thyroid carcinoma (ATC) is a frequently lethal malignancy that is often unresponsive to available therapeutic strategies. The tumorigenesis of ATC and its relationship to the widely prevalent well-differentiated thyroid carcinomas are unclear. We have analyzed 22 cases of ATC as well as 4 established ATC cell lines using whole-exome sequencing. A total of 2,674 somatic mutations (121/sample) were detected. Ontology analysis revealed that the majority of variants aggregated in the MAPK, ErbB, and RAS signaling pathways. Mutations in genes related to malignancy not previously associated with thyroid tumorigenesis were observed, including mTOR, NF1, NF2, MLH1, MLH3, MSH5, MSH6, ERBB2, EIF1AX and USH2A; some of which were recurrent and were investigated in 24 additional ATC cases and 8 ATC cell lines. Somatic mutations in established thyroid cancer genes were detected in 14 of 22 (64%) tumors and included recurrent mutations in BRAF, TP53, and RAS-family genes (6 cases each), as well as PIK3CA (2 cases) and single cases of CDKN1B, CDKN2C, CTNNB1 and RET mutations. BRAF V600E and RAS mutations were mutually exclusive; all ATC cell lines exhibited a combination of mutations in either BRAF and TP53 or NRAS and TP53. A hypermutator phenotype in two cases with >8 times higher mutational burden than the remaining mean was identified; both cases harbored unique somatic mutations in MLH mismatch-repair genes. This first comprehensive exome-wide analysis of the mutational landscape of ATC identifies novel genes potentially associated with ATC tumorigenesis, some of which may be targets for future therapeutic intervention. © The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.
Article
Limited data is available on the genetic features of primary leptomeningeal melanocytic neoplasms (LMNs). Similarities with uveal melanoma were recently suggested as both entities harbor oncogenic mutations in GNAQ and GNA11. Whether primary LMNs share additional genetic alterations with uveal melanoma including copy number variations is unknown. Twenty primary LMNs ranging from benign and intermediate-grade melanocytomas to melanomas were tested by direct sequencing for hotspot mutations in the genes GNA11, GNAQ, BRAF, NRAS and HRAS. Furthermore, the lesions were tested for copy number variations of chromosomes frequently present in uveal melanoma (1p, 3, 6 and 8q) by multiplex ligation-dependent probe amplification (MLPA). Genome-wide analyses of copy number alterations of two leptomeningeal melanocytic neoplasms were performed using the OncoScan SNP-array. GNAQ(Q209) mutations were present in eleven LMNs, while two of 20 cases carried a GNA11(Q209) mutation. No BRAF, HRAS or NRAS hotspot mutations were detected. Monosomy 3 and gain of 8q were present in one leptomeningeal melanoma, and one intermediate-grade melanocytoma harbored a gain of chromosome 6. With MLPA, the melanocytomas did not show any further gross chromosomal variations. Our data shows that primary LMNs, like uveal melanoma, harbor oncogenic mutations in GNAQ and GNA11 but lack mutations in BRAF, NRAS and HRAS. This finding may help in the differential diagnosis between a primary LMN and a metastasis from a cutaneous melanoma to the central nervous system. Copy number variations in some aggressive LMNs resemble those present in uveal melanoma but their prognostic significance is unclear.