ArticlePDF Available

The Genomic Landscape of Sporadic Prolactinomas

Authors:

Abstract and Figures

Somatic GNAS and USP8 mutations have been implicated in sporadic somatotrophinomas and corticotrophinomas, respectively. However, no genes are known to be recurrently mutated in sporadic prolactinomas. The prevalence of copy number variants (CNV), which is emerging as a mechanism of tumorigenesis in sporadic pituitary adenomas in general, is also unclear in prolactinomas. To characterize the genetic events underpinning sporadic prolactinomas, we performed whole exome sequencing of paired tumor and germline DNA from 12 prolactinoma patients. We observed recurrent large-scale CNV, most commonly in the form of copy number gains. We also identified sequence variants of interest in 15 genes. This included the DRD2, PRL, TMEM67, and MLH3 genes with plausible links to prolactinoma formation. Of the 15 genes of interest, CNV was seen at the gene locus in the corresponding tumor in 10 cases, and pituitary expression of eight genes was in the top 10% of tissues. However, none of our shortlisted somatic variants appeared to be classical driver mutations as no variant was found in more than one tumor. Future directions of research include mechanistic studies to investigate how CNV may contribute to prolactinoma formation, larger studies of relevant prolactinoma subsets according to clinical characteristics, and additional genetic investigations for aberrations not captured by whole exome sequencing.
Content may be subject to copyright.
The Genomic Landscape of Sporadic Prolactinomas
Sunita M. C. De Sousa
1,2,3
&Paul P. S. Wang
4
&Stephen Santoreneos
5
&Angeline Shen
6,7
&Christopher J. Yates
6,7
&
Milena Babic
2
&Leila Eshraghi
2,4,8
&Jinghua Feng
4,8
&Barbara Koszyca
9
&Samuel Roberts-Thomson
10
&
Andreas W. Schreiber
4,8,11
&David J. Torpy
1,3
&Hamish S. Scott
2,3,4,8
#Springer Science+Business Media, LLC, part of Springer Nature 2019
Abstract
Somatic GNAS and USP8 mutations have been implicated in sporadic somatotrophinomas and corticotrophinomas, respectively.
However, no genes are known to be recurrently mutated in sporadic prolactinomas. The prevalence of copy number variants
(CNV), which is emerging as a mechanism of tumorigenesis in sporadic pituitary adenomas in general, is also unclear in
prolactinomas. To characterize the genetic events underpinning sporadic prolactinomas, we performed whole exome sequencing
of paired tumor and germline DNA from 12 prolactinoma patients. We observed recurrent large-scale CNV, most commonly in
the form of copy number gains. We also identified sequence variants of interest in 15 genes. This included the DRD2,PRL,
TMEM67,andMLH3 genes with plausible links to prolactinoma formation. Of the 15 genes of interest, CNV was seen at the gene
locus in the corresponding tumor in 10 cases, and pituitary expression of eight genes was in the top 10% of tissues. However,
none of our shortlisted somatic variants appeared to be classical driver mutations as no variant was found in more than one tumor.
Future directions of research include mechanistic studies to investigate how CNV may contribute to prolactinoma formation,
larger studies of relevant prolactinoma subsets according to clinical characteristics, and additional genetic investigations for
aberrations not captured by whole exome sequencing.
Keywords Prolactinoma .Pituitaryadenoma .Wholeexome sequencing .Copynumbervariation .Lossof heterozygosity .Driver
mutation
Introduction
The genetic basis of sporadic prolactinomas is currently un-
known. This is in contrast to well-described somatic events in
other pituitary tumors, namely: GNAS mutations in
somatotrophinomas and occasional non-functioning pituitary
adenomas [13]; USP8 [3,4] and rarely NR3C1 [3,6]muta-
tions in corticotrophinomas; and CTNNB1 (encoding β-
catenin) and BRAF mutations in the vast majority of
adamantinomatous and papillary craniopharyngiomas, respec-
tively [5]. Some of these somatic events recapitulate multisys-
tem disorders, including McCune-Albright syndrome due to
*Sunita M. C. De Sousa
Sunita.DeSousa@sa.gov.au
1
Endocrine and Metabolic Unit, Royal Adelaide Hospital,
Adelaide, Australia
2
Department of Genetics and Molecular Pathology, Centre for Cancer
Biology, an SA Pathology and University of South Australia
Alliance, Adelaide, Australia
3
School of Medicine, University of Adelaide, Adelaide, Australia
4
ACRF Cancer Genomics Facility, Centre for Cancer Biology, an SA
Pathology and University of South Australia Alliance,
Adelaide, Australia
5
Department of Neurosurgery, Royal Adelaide Hospital,
Adelaide, Australia
6
Department of Diabetes and Endocrinology, Royal Melbourne
Hospital, Melbourne, Australia
7
Department of Medicine, University of Melbourne,
Melbourne, Australia
8
School of Pharmacy and Medical Sciences, University of South
Australia, Adelaide, Australia
9
Department of Anatomical Pathology, Royal Adelaide Hospital,
Adelaide, Australia
10
Department of Anatomical Pathology, Royal Melbourne Hospital,
Melbourne, Australia
11
School of Biological Sciences, University of Adelaide,
Adelaide, Australia
Endocrine Pathology
https://doi.org/10.1007/s12022-019-09587-0
GNAS somatic mosaicism [6], and a newly described
syndromic disorder including pediatric Cushings disease
due to a germline heterozygous USP8 mutation [7].
Gene-specific assessment of sporadic prolactinomas has
shown nil or only rare somatic variants in biologically plausi-
ble genes. This includes genes where germline variants cause
familial pituitary tumor syndromes (FPTS), such as MEN1 [8]
and AIP [9], and genes implicated in sporadic pituitary ade-
nomas, including GNAS [1], USP8 [4], and TP53 [10,11]. A
recurrent germline gain-of-function PRLR mutation has been
identified in prolactinoma patients, but no such variants have
been found in the somatic setting [12]. A somatic HRAS var-
iant has been identified in an aggressive prolactinoma, but this
association was not borne out in an extension study including
72 prolactinomas [13].
A limitation of single gene studies is the reliance on
existing knowledge to select candidate genes. Orphangenes
of hitherto unknown function could be contributory to
prolactinomas akin to other genetic discoveries, such as the
roles of GPR101 in X-linked acrogigantism [14]andARMC5
in bilateral macronodular adrenal hyperplasia [15]. Whole ex-
ome or genome sequencing offers an unbiased approach to
novel gene discovery. Only three pangenomic studies of
prolactinomas have been performed to date, all employing
whole exome sequencing (WES). Wang et al. [16] focused
on point variants conferring bromocriptine resistance in a co-
hort of 12 prolactinomas and identified 11 candidate genes
between initial and follow-up [17] studies. Bi et al. [18]inves-
tigated 41 pituitary macroadenomas, including three
prolactinomas. Six genes were mutated in more than one tu-
mor, but none of these were prolactinomas. Song et al. [3]
examined 125 pituitary adenomas, including 20
prolactinomas. Two genes were considered to be potential
tumorigenesis genes but only one prolactinoma harbored a
variant in these genes. Overall, these studies did not find re-
current sequence variants amongst the prolactinomas that
could constitute driver mutations. There is, however, emerg-
ing evidence of recurrent copy number variants (CNVs) in
sporadic pituitary adenomas, including the small number of
prolactinomas thus far studied [2,3,18,19].
The aim of the present study was to perform WES in a pure
prolactinoma cohort to identify recurrent somatic genetic
events. We hypothesized that, like other pituitary tumors, so-
matic driver mutations and/or CNVs might also underlie the
development of prolactinomas.
Materials and Methods
Patients
Twelve patients with clinically evident prolactinomas that had
been surgically resected were recruited from two tertiary
referral pituitary centers in Australia. Clinical data were col-
lated using medical records.
DNA Extraction
Patients provided fresh blood samples for germline DNA ex-
traction. Operative tumor specimens were retrieved for somat-
ic DNA extraction. Tumor specimens had either been stored as
fresh frozen (n= 6) or formalin-fixed paraffin-embedded
(FFPE; n= 6) tissue. Duration of tumor storage ranged from
7 months to 8 years. DNA was extracted using commercially
available kits (Qiagen and Bioline) according to manufacturer
protocols. FFPE samples were deparaffinized and additional
DNA repair steps were performed using uracil-N-glycosylase
to enzymatically remove formalin-induced cytosine deamina-
tion artifacts.
Whole Exome Sequencing
WES of germline and tumor DNA samples was performed
using the Roche NimbleGen SeqCap EZ MedExome v3.0
target enrichment kit, and the Illumina NextSeq 500 sequenc-
ing platform. The average of mean depth of coverage amongst
all samples was 129x, and 97% oftarget bases were covered
20x.
Filtration of Sequence Variants
Bioinformatic analysis was performed in the ACRF Cancer
Genomics Facility of the Centre for Cancer Biology, SA
Pathology (Adelaide, Australia). BWA-MEM was used to
align short reads to GRCh37/hg19 (version b37+decoy).
Small variants (typically < 50 bp) were called using Genome
Analysis Toolkit (GATK) HaplotypeCaller package version
3.4. Raw WES data were initially filtered for variants that
were: high quality (by GATK internal filters); very rare (<
0.2% population); potentially functional (by snpEFF impact,
branching/binding predictions, GERP, or CADD); and not in
regions of segmental duplication.
Germline variants were considered further if they had a
GATK genotype quality (GQ) score > 50 and depth of cover-
age > 30x, and were not situated in a low complexity region.
Drawing on existing literature, we searched for germline var-
iants in known FPTS genes: AIP,CDH23,CDKN1B,
DICER1,GPR101,MAX,MEN1,PRKAR1A,SDHA,SDHB,
SDHC,andSDHD [14,2023].
Artifact was observed in tumor DNA results due to reasons
including presumed normal tissue admixture and DNA degra-
dation in FFPE specimens. Raw data from tumor DNA were
thus re-analyzed by a dedicated in-house somatic variant call-
ing pipeline to identify variants present in tumor DNA and
absent in germline DNA. To increase the reliability of somatic
variant calls, this pipeline integrates four variant callers that
Endocr Pathol
detect insertions/deletions (indels) and single-nucleotide
variants (SNVs): Shimmer (v e5bafb4), Seurat (v 2.6),
Strelka2 (v 2.9.0), and VarScan2 (v 2.4.0); and three callers
that detect SNVs only: MuTect (v 1.1.4), SomaticSniper (v
1.0.5), and Virmid (v 1.1.1). Only somatic indels and SNVs
that were detected by at least two or five variant callers, re-
spectively, were considered to be candidate somatic sequence
variants.
These candidate somatic variants were shortlisted to a final
list of somatic variants of interest that were absent in popula-
tion genomic databases (dbSNP, 1KG, UK10K, gnomAD,
ExAC, and ESP) with evidence of being highly damaging
(high snpEFF impact). Pituitary expression of these final
genes of interest was determined using the Genotype-Tissue
Expression (GTEx) project database (https://gtexportal.org),
comprising 53 non-diseased tissue sites including 183 pitui-
tary samples.
All germline variants in known FPTS genes and somatic
variants of interest were verified by inspection of raw se-
quencing data in Integrated Genomics Viewer (IGV).
Identification of Somatic Copy Number Variants
Raw WES data were interrogated for copy number variation
(CNV) via in-house scripts, with calculation of copy number
using a normalized read depth of coverage against control
samples and correlation with minor allele frequency.
Coverage plots of sequence read depth and minor allele fre-
quency was manually inspected to identify chromosomal and
arm level copy number gains and losses as well as copy num-
ber neutral loss of heterozygosity (LOH).
Statistical Analysis
IBM SPSS Statistics 25.0 was used for statistical analysis. The
Mann-Whitney Utest was used to assess differences in the
median numbers of candidate somatic variants and chromo-
somes affected by CNV or copy neutral LOH per tumor ac-
cording to relevant categorical clinical characteristics. P
values < 0.05 were considered statistically significant.
Results
Clinical Characteristics
The study cohort consisted of six women and six men aged
1665 years at prolactinoma diagnosis. The tumors
hypersecreted prolactin alone. Apart from one patient who
presented with pituitary apoplexy, all patients were treated
with dopamine agonists (DA) preoperatively. In all cases, sur-
gical resection was by the trans-sphenoidal route and histopa-
thology confirmed pituitary adenomas with positive
immunostaining for prolactin. Postoperative tumor remnants
or recurrences were observed in ten patients, all of whom had
macroadenomas or giant adenomas at the baseline scan. The
remaining two patients had microadenomas that were resected
because of DA intolerance, with gross total resection achieved
and no evidence of tumor recurrence to date. No patients had
received other medical therapies or radiotherapy at study en-
rollment. Other clinical characteristics of the patient cohort are
given in Table 1.
Pathological Characteristics
Pathological characteristics are described in Table 2. Most
tumors were densely granulated lactotroph adenomas. Ki-67
index was only available in a minority of tumors. Few or no
mitoses were observed in the remaining tumors, arguing
against a significant degree of proliferation [24]. Histological
invasion was found in only three tumors. Despite DA pretreat-
ment in all but one patient, fibrosis was only observed in 4/11
DA-treated tumors (Fig. 1).
Germline Sequence Variants
The only known FPTS gene with germline sequence variants
after filtration was CDH23, with missense variants observed
in Patient 6 (c.1103G>A (p.Arg368His), population preva-
lence in gnomAD 0.06%, CADD 26.2, GERP 4.5) and
Patient 11 (c.4510G>T (p.Ala1504Ser), gnomAD 0.01%,
CADD 25, GERP 5.06; and c.4907C>T (p.Ala1636Val),
gnomAD 0.07%, CADD 22.5, GERP 5.75).
Somatic Sequence Variants
Filtration of WES data revealed 138 candidate somatic vari-
ants, none of which were found in more than one tumor. Only
one gene (PHTF1) was mutated in more than one tumor.
Another two genes (NBEAL2,TMEM67) were each mutated
twice in the same tumor from Patient 1. Of the 135 different
genes containing candidate somatic variants, there was no
overlap with genes implicated in FPTS or sporadic pituitary
adenomas (i.e., AIP,CDH23,CDKN1B,DICER1,GNAS,
GPR101,MAX,MEN1,NR3C1,PRKAR1A,PRLR,SDHA,
SDHB,SDHC,SDHD,TP53,USP8).
Each tumor harbored multiple candidate somatic variants
(median 9.5 per tumor, range 323). There was no significant
difference in the median number of variants according to gen-
der (male 7.5 vs female 15.0, P= 0.107), indication for sur-
gery (DA intolerance 9.5 vs other indications 14.0, P=0.624),
tumor consistency (no cystic component 9.0 vs cystic compo-
nent 15.0, P= 0.114), or extent of resection (no remnant 9.5 vs
remnant 14.0, P=0.780).
The 138 candidate somatic variants were shortlisted to 15
variants of interest (Table 3;Fig.2)thatwereabsentin
Endocr Pathol
population genomic databases and highly damaging (n=14),
or situated in a gene with another candidate somatic variant in
another tumor (n=1, PHTF1). The shortlist included non-
sense or frameshift variants in three genes (DRD2,PRL,
TMEM67) with known associations with the pituitary gland
and the MLH3 gene which is a tumorigenesis gene in other
tissues. Using the GTEx database, we observed that the pitu-
itary was in the top 10% of expressing tissues for 8/15
shortlisted genes of interest. We next used the STRING data-
base (https://string-db.org) of known and predicted protein-
protein interactions to look for interactions between the 15
genes of interest. The only interaction was the known link
between PRL encoding prolactin and DRD2 encoding the
D2 dopamine receptor (D2R), which are co-expressed in mul-
tiple species and co-mentioned in medical literature.
No patient had a germline variant in the same gene con-
taining a somatic variant of interest in their corresponding
tumor. Conversely, no candidate somatic variants were found
in CDH23 in the two patients with germline CDH23 variants.
Somatic Copy Number Variants
All but one tumor contained chromosomal or arm level CNVs
and/or copy neutral LOH (median 10.5 chromosomes affected
per tumor, range 021). There was no significant difference in
the median number of chromosomes affected according to
gender (male 10.0 vs female 12.0, P= 0.377), surgical indica-
tion (DA intolerance 8.0 vs other indications 12.0, P=0.514),
tumor consistency (no cystic component 10.0 vs cystic
component 17.0, P= 0.266), or extent of resection (no rem-
nant 10.0 vs remnant 10.0, P= 0.926). CNV most commonly
manifested as whole or partial chromosomal gain (median 10
chromosomes per tumor, range 020), and occasionally as
whole or partial chromosomal loss (median 0 chromosomes
per tumor, range 02). Copy neutral LOH was also seen (me-
dian 0.5 chromosomes per tumor, range 06).
Recurrent and single cases of chromosomal gain, loss, and
copy neutral LOH are shown in Table 4. Examples of CNV
calling are depicted in Fig. 3. The most frequent chromosomes
affected were Chr 8, 9, and 14 followed by Chr 3, 7, 12 and 20
for gains, and 1 and 15 for copy neutral LOH. No chromo-
somes showed recurrent losses.
Each tumor was assessed for regional overlap between its
CNV results and any observed variant of interest. Copy num-
ber gain waspresentin the corresponding tumor at the locus of
10/15 genes of interest and corresponding monosomy was
observed for 1/15 genes.
Discussion
The major somatic event in our cohort of 12 patients with
prolactinomas was large-scale CNV, most commonly in the
form of copy number gains. We also observed sequence var-
iants of interest in 15 genes, including genes of putative inter-
est in prolactinoma tumorigenesis. Although we found that
pituitary expression is in the top 10% of tissues for over half
of our genes of interest, these somatic variants do not appear to
Table 1 Clinical characteristics at time of diagnosis
Patient Age (yr),
gender
Tumor
maximum
diameter (mm)
Hardys
score
Tumo r
consistency
PRL
(xULN)
Surgical
indication
Postoperative
remnant
#
Tumo r
recurrence
*
140F 16
^
3
^
Solid
^
33
^
DA resistance Yes N/A
2 16 F 8 1 Cystic 9 DA intolerance No N
3 56 M 8 3 Solid 10 DA intolerance No N
4 42 F 11 3 Solid 5 DA intolerance Yes N/A
5 28 F 60 3 Solid 278 DA resistance Yes N/A
6 53 M 18 3 Solid 145 DA intolerance Yes N/A
7 32 M 26 3 Solid 20 DA resistance Yes N/A
8 64 M 52 3 Mixed 576 Apoplexy at Dx Yes N/A
9 65 M 37 3 Solid 67 DA resistance Yes N/A
10 32 F 16^ 2^ Solid
^
28^ DA intolerance No Y
11 40 M 41 3 Solid 215 DA resistance Yes N/A
12 61 F 46 3 Mixed 72 DA resistance Yes N/A
DA, dopamine agonist; Dx,diagnosis;F,female;M, male; N/A, not applicable; N/S, not stated in report and images not available for review; PRL,
prolactin; xULN, absolute level divided by upper limit of normal; yr,year
#
Based on postoperative imaging and serum prolactin results
*
Only applies to tumors that were completely resected
^
Preoperative results used as results at initial diagnosis unavailable
Endocr Pathol
be classical driver mutations as none were found in more than
one tumor. We also found rare missense germline variants in
the recently recognized FPTS gene, CDH23. However, somat-
ic second-hits were not found in the corresponding tumors and
CDH23 is a notably large gene with 69 exons, which may
increase the propensity for variants of uncertain significance.
Our results recapitulate the findings of the few systematic
genomic studies of prolactinomas that have been performed
to date (Table 5), whereby CNV is common and recurrently
mutated genes are rare.
We observed several recurrent copy number gains and copy
neutral LOH. This is in keeping with the pituitary
macroadenoma WES study by Bi et al. [18], and their
follow-up study of 114 pituitary adenomas including 14
prolactinomas [2], that indicated two patterns of CNV: a high-
ly disrupted group mostly consisting of functional adenomas
(including prolactinomas) or atypical null cell adenomas with
CNV involving a mean of 39% of the genome; and a less
disrupted group mostly consisting of non-functioning adeno-
mas with CNVinvolving a mean of 0.5% of the genome. Song
et al. [3] also found a high degree of CNV in their mixed
cohort of pituitary adenomas, with almost one-third of pitui-
tary adenomas showing CNVinvolving > 80% of the genome.
Our patientstumors demonstrated recurrent gains in Chr 7, 9,
and 14 similar to Bi et al. [18] and in Chr 1, 3, 7, 16, and 20
similar to Song et al. [3]. In contradistinction to these former
studies, we observed additional recurrent gains in Chr 57, 10,
12, 1719, 21, 22, and X. We also found no recurrent chro-
mosomal losses, whereas Bi et al. [18] found losses to be
particularly common in Chr 1p and 11 in hormonally active
adenomas. While we found recurrent copy neutral LOH in
Chr 1, 4, and 15, Bi et al. [18] only found Chr 11q LOH.
Discrepancies between the different cohorts are at least partly
explained by the heterogenous mixes of different pituitary
adenoma subtypes in previous studies. Our pure prolactinoma
cohort should be considered separately to these previous stud-
ies because of the potential for specific DA treatment effects in
11 of our 12 patients who were treated with a DA preopera-
tively. DA resistance may have led to the observed high rate of
CNV or vice versa.
Table 2 Pathological characteristics of patient cohort at time of diagnosis
Patient Positive hormone
IHC
Granulation pattern Mitoses Ki-67 index Histological invasion Fibrosis
1 PRL Undetermined Scant U/A No No
2 PRL Densely granulated < 1/10hpf U/A No No
3 PRL Densely granulated Nil U/A No Yes
4 PRL Undetermined Scant U/A No No
5 PRL, LH Densely granulated Scant U/A Yessphenoid, nasopharynx Yes
6 PRL Densely granulated < 1/10hpf U/A Yesdura No
7 PRL, TSH, LH, FSH Undetermined Scant U/A No No
8 PRL Sparsely granulated Nil U/A Yessphenoid No
9 PRL Densely granulated Nil <1% N Yes
10 PRL Densely granulated < 1/10hpf 3% No No
11 PRL Densely granulated Nil < 1% No Yes
12 PRL Sparsely granulated < 1/10hpf U/A* No No
FSH, follicle-stimulating hormone; IHC, immunohistochemistry; LH, luteinizing hormone; PRL, prolactin; TSH, thyroid-stimulating hormone; U/A,
unavailable
*Topoisomerase index 5%
Fig. 1 Hematoxylin & eosin
appearance of prolactinomas at
medium power in a case
demonstrating no fibrosis (a
Patient 12) and another
demonstrating marked fibrosis (b
Patient 11)
Endocr Pathol
By contrast to the high burden of CNV, we found relatively
few sequence variants per tumor and a lack of recurrent se-
quence variants between tumors. This argues against a major
role of driver mutations in the pathogenesis of prolactinomas.
This differs from the experience of studying
somatotrophinomas, corticotrophinomas, and
craniopharyngiomas, but mimics findings in other pituitary
tumor subtypes. Paired tumor-normal WES studies of seven
patients with non-functioning pituitary adenomas in 2013 [25]
and of four patients with TSHomas in 2016 [26]alsofoundno
recurrent variants that could be considered driver mutations. A
low number of somatic mutations have been observed in pre-
vious studies of pituitary adenomas compared with other neo-
plasms [3,18]. However, direct comparison of absolute mu-
tation numbers between studies is limited by differing
methods of variant filtration. Within studies, we and others
[3] have found no association between prolactinoma clinical
characteristics and number of sequence variants.
Some variants within individual tumors were of interest
due to their location in genes with a plausible connection to
prolactinoma formation. This includes the truncating DRD2
variant found in a patient with a 40-year history of
prolactinoma that showed DA escape over the last 3 years
despite increasing doses of cabergoline, necessitating surgery
and radiotherapy. We speculate that this variant, found in 19%
of tumor DNA, may reflect a subclone that is driving the
patients recent DA resistance. Indeed, D2R expression is typ-
ically high in prolactinomas, and downregulation has been
hypothesized as a mechanism of DA resistance [27].
Compared with DA-responsive prolactinomas, resistant tu-
mors demonstrate decreased D2R density, overall reduction
in D2R mRNA production, and altered expression of D2R
mRNA isoforms with lower expression of the more efficient
short isoform [28]. In female mouse models, D2R deficiency
induces lactotroph hyperplasia [29]. However, we did not find
DRD2 variants of interest in our other five patients with DA
Table 3 Somatic sequence variants of interest
Gene, ID
a
Chr locus Pituitary rank
amongst all
tissue expression
b
CNV at locus
c
Pt: variant VAF in tumor DNA
ANKS3, ENSG00000168096 16: 4780135 3 Trisomy Pt 1: c.15delC
(p.Ser5fs)
33%
C19orf25, ENSG00000119559 19: 1475427 3 Trisomy Pt 7: c.216dupG
(p.Ile73fs)
12%
C9orf163, ENSG00000196366 9: 139379389 2 Trisomy Pt 1: c.491delC
(p.Pro164fs)
15%
CAST, ENSG00000153113 5: 96077063 35 Tetrasomy (2:2) Pt 5: c.888+1G>T 30%
DCAF10, ENSG00000122741 9: 37860080 31 Tetrasomy (2:2) Pt 12: c.1202_1203delCT
(p.Pro401fs)
26%
DRD2, ENSG00000149295 11: 113283323 1 Monosomy Pt 1: c.1093C>T
(p.Gln365*)
19%
KLRD1, ENSG00000134539 12: 10460684 4 Trisomy Pt 1: c.7+1G>C 21%
LDB2, ENSG00000169744 4: 16597359 34 Nil Pt 9: c.3G>A
(p.Met1?)
37%
MLH3, ENSG00000119684 14: 75514552 12 Trisomy Pt 8: c.1806delA
(p.Lys602fs)
27%
NBEAL2, ENSG00000160796 3: 47036629 25 Tetrasomy (2:2) Pt 1:
p.Phe470fs, c.1407_1408delCT
41%
PHTF1, ENSG00000116793 1: 114242872 5 Nil Pt 4: n.468-7insA
Pt 7: n.468-7delA
Pt 4: 33%
Pt 7: 16%
PRL, ENSG00000172179 6: 22290411 1 Trisomy Pt 4: c.483dupA
(p.Val162fs)
13%
SKIDA1, ENSG00000180592 10: 21805663 10 Nil Pt 5: c.1088delC
(p.Pro363fs)
16%
SPTBN2, ENSG00000173898 11: 66453356 13 Nil Pt 4: c.7159A>T
(p.Lys2387*)
35%
TMEM67, ENSG00000164953 8: 94797512 2 Trisomy Pt 1: c.1194C>A
(p.Tyr398*)
40%
Chr, chromosomal; CNV, copy number variation; Pt, Patient number corresponding to tumor in which variant detected; VAF, variant allele frequency
a
HGNC gene symbol, gene ID by snpEFF
b
Pituitary rank after sorting all 53 non diseased tissue types by median TPM(transcripts per million) in GTEx
c
Corresponding CNVat the gene locus in the same tumor
Endocr Pathol
resistance. Wang et al. [16] also reported an absence of DRD2
sequence variants, although the sensitivity of this study was
reduced by its overall low depth of coverage with only 10 x
coverage in 80% of the exome. Another tumor in our cohort
harbored a frameshift variant in PRL, which is well known to
be highly expressed in lactotrophs [27]. Autocrine signaling
between prolactin and the abundant prolactin receptors on
lactotrophs has been postulated as the explanation for the sex-
ual dimorphism in lactotroph hyperplasia in D2R knockout
mice [29]. By this theory, male mice lacking the D2R do not
reach the prolactin threshold required for the feed-forward
loop to activate and trigger lactotroph hyperplasia [29]. We
also found isolated somatic variants in TMEM67 where
biallelic inactivating variants have been implicated in hypopi-
tuitarism [30], and in MLH3 which is a mismatch repair gene
with a possible role in Lynch syndrome [31]. Although a
Lynch syndrome registry study found an increased risk of
pituitary adenomas [32], there is currently no evidence of a
specific role for MLH3 in pituitary tumorigenesis.
The remaining variants of interest were located in genes
with no currently known associations with the pituitary
gland. Comparison with the previously published genomic
studies including prolactinomas showed little overlap:
Song et al. [3] found one ANKS3 frameshift variant and
two SKIDA1 variants; and Bi et al. [18] found a KLRD1
missense variant. None of these variants were seen in the
prolactinoma subsets of these studies. In addition, none of
our cases fulfilled Knudsons two-hit model of tumor sup-
pressor genes as no patients had germline variants in the 15
genes harboring somatic variants of interest and the two
patients with germline CDH23 variants had no candidate
somatic variants in CDH23. CNV might have arguably
been the second-hit in some of these tumors as 11/15
(73%) of variants of interest were in regions of CNV in a
given tumor. Trisomy and tetrasomy could be especially
relevant as increased mutant dosage can amplify a domi-
nant negative effect by a sequence variant, thereby contrib-
uting to tumorigenesis. On the other hand, the
maximum VAF was 41% among the 15 variants of interest
despite the frequent coexistence of CNV. Furthermore,
most prolactinomas in our study harbored multiple CNV
and the CNVs were large; thus, it is unlikely that any single
gene in these regions of CNV can explain the pathogenesis
of prolactinomas.
Fig. 2 Examples of somatic point variants as visualized in Integrated Genomics Viewer. aCAST missense variant in Patient 5: c.888+1G>T. bC9orf163
deletion in Patient 1: c.491delC (p.Pro164fs). cPRL insertion in Patient 4: c.483dupA (p.Val162fs)
Table 4 Somatic CNV analysis
summary showing chromosomes
with whole or partial gains, losses
and copy neutral LOH in either
multiple or single tumors
Copy neutral LOH Gain Loss Mixed CNV
and copy neutral
LOH
Recurrent Chr 1, 4, 15 Chr 1, 3, 510, 12, 14,
1622, X
nil nil
Single Chr 5, 6, 10, 11, 16,
20
Chr 2, 4, 11, 13, 15 Chr 11, 13, 15, 18,
X
Chr 1, 4, 11
Chr, chromosome; CNV, copy number variants; LOH, loss of heterozygosity
Endocr Pathol
A key limitation of our study is that WES does not
detect deep intronic and intergenic variants, balanced
translocations, fusion genes, or epigenetic changes.
Integrative genomic analyses of both DNA and RNA
[33] as well as the emerging technology of long-read se-
quencing with real-time analysis of nucleotide binding [34]
may elucidate some of these possibilities. Furthermore,
half of our tumor samples were FFPE, although we
employed an optimized DNA extraction protocol to limit
artifactual results because of this. Another limitation of our
study is that the identification of CNV and copy neutral
LOH was based on broad patterns in VAF and ploidy
based on depth of coverage. We were thus only able to
categorize CNV and copy neutral LOH at the arm or
chromosomal level. Smaller CNVs may have been missed,
although other data support the predominance of large-
scale CNV, as observed in our tumors, over smaller
CNVs [18]. The relatively low allele frequencies of our
variants of interest are also noteworthy. This may seem-
ingly contradict the known monoclonal origin of pituitary
adenomas [27]. However, the < 50% VAFs seen in our
tumor DNA results may reflect CNV, which was a com-
mon finding in our tumors, and/or normal tissue admix-
ture, particularly as pituitary adenomas are rarely resected
en bloc and interspersed normal pituitary tissue is a com-
mon microscopic finding. VAFs < 50% may alternatively
represent the presence of multiple tumor clones. The
possibility of this may be assessed in future studies through
spatial transcriptomics whereby sequencing results are
overlaid with tissue sections to compare the transcriptomes
of different tumor regions [35]. Finally, the small size of
this pilot study limited our ability to identify clinical pre-
dictors of the number of somatic variants and the number
of chromosomes affected by CNV or copy neutral LOH.
An independent validation set of another group of
prolactinomas using the same platforms employed in this
study would have been ideal to further explore these pu-
tative clinicopathological correlations and our identified
genes of interest; however, surgery is rarely performed
for prolactinomas and thus, further tumors were not avail-
able for investigation.
Larger studies involving sufficient numbers of different
prolactinoma subsets (e.g., cystic prolactinomas or young
onset prolactinomas in males) with use of fresh frozen
tumor samples may better elucidate the genetic drivers
of tumorigenesis. Our findings of suspicious albeit isolat-
ed somatic variants in strong candidate genes such as
DRD2 may be a function of the heterogenous patient
and tumor case mix in the prolactinoma studies to date.
We kept our inclusion criteria at a minimum in order to
capture sufficient numbers of prolactinomas, which other-
wise tend to be medically managed with DAs. Routine
biobanking of pituitary adenomas will help facilitate fu-
ture studies, although resected tumor tissue is often
Fig. 3 Examples of tumor DNA calls of normal diploid status,
chromosomal gain, chromosomal loss, and copy neutral loss of
heterozygosity (LOH) based on somatic heterozygous variant allele fre-
quency (VAF) (top panels) and ploidy estimates using depth of coverage
(bottom panels). aDisomic baseline in Chr 2 in Patient 6 represented by
the usual 0.5 heterozygous VAF and ploidy count of 2. bChr 3 trisomy
(2:1) in Patient 10 represented by separation of heterozygous VAF into
VAFs of approximately 0.4 and 0.6 and increased ploidy count at 3. cChr
9 tetrasomy (2:2) in Patient 10 represented by usual 0.5 heterozygous
VAF but increased ploidy count at 4. dChr X monosomy in Patient 1
(female) represented by separation of heterozygous VAF and decreased
ploidy count at 1. eChr 4 copy neutral LOH in Patient 1 represented by
separation of heterozygous VAF but normal ploidy count of 2
Endocr Pathol
piecemeal, in small quantity and potentially damaged by
intraoperative cauterisation.
In conclusion, this systematic genomic study of all coding
genes in a pure prolactinoma cohort demonstrated variants in
genes of biologically plausible interest within individual tu-
mors, without overlap between prolactinomas in this study or
with the few other published pangenomic studies [2,3,16,
17]. We instead found a high degree of CNV, corroborating
other preliminary studies of sporadic prolactinomas [19]and
larger studies of mixed pituitary adenoma subtypes [2,3,18].
Further research is required to determine how CNV may con-
tribute to prolactinoma formation and ways in which this
could be therapeutically targeted.
Funding SMCD is supported by an A.R. Clarkson Scholarship from the
Royal Adelaide Hospital. This study was produced with support from a
Royal Adelaide Hospital Health Services Charitable Gifts Board grant,
and with the financial and other support of the Cancer Council SAsBeat
Cancer Project on behalf of its donors and the State Government of South
Australia through the Department of Health.
Compliance with Ethical Standards
Conflict of Interest The authors declare that they have no conflict of
interest.
Ethical Approval All procedures performed in studies involving human
participants were in accordance with the ethical standards of institutional
research committees (Melbourne Health: HREC/16/MH/132; Royal
Table 5 Pangenomic studies of paired tumor and germline DNA from prolactinoma patients. All studies employed whole exome sequencing
Study Cohort Filter for GOI GOI Recurrent CNV
Wang 2014
[16],
Gao 2015
[17]
DA responsive vs resistant PRLoma
(n=12)
Variants differing between
responsive
vs resistant PRLoma
C1orf170
DPCR1
DSPP
KRTAP103
MUC4
MX2
POTEF
PRB3*
PRDM2*
PRG4
RP1L1
N/T
Song 2016
[3]
Pituitary adenoma (n=120,incl20
PRLoma)
Recurrently mutated in multiple
PA
GRB10*
IARS
KIF5A*
SP100
TRIP12
Gains:
Chr 1p13.2, 1q31.3, 3p22.3, 7q21.11,
16q12.2, 20p13,
20q13.33
Losses:
Chr 1p36.31, 3p21.31, 9q34.11, 11q13.2,
11p15.5, 16p13.3
Bi 2017 [18] Pituitary macroadenomas (n=42,incl
3 PRLoma)
Recurrently mutated in multiple
PA
ATAD3B
BHLHE22
KDM2B
OR5M1
TTN*
VPS13B
Gains:
Chr 7, 9q, 14q
Losses:
Chr1,2,4,10,11,15q,18
Copy neutral LOH:
Chr 11q
Present
study
PRLoma
(n=12)
Absent in population databases
and strong
in silico prediction for
pathogenicity, or
recurrently mutated in
multiple PRLoma
ANKS3
C19orf25
C9orf163
CAST
DCAF10
DRD2*
KLRD1
LDB2
MLH3
NBEAL2
PHTF1
PRL*
SKIDA1
SPTBN2
TMEM67*
Gains:
Chr1,3,510, 12, 14, 1622, X
Losses:
nil
Copy neutral LOH:
Chr1,4,15
Chr, chromosome; CNV, copy number variants; DA, dopamine agonist; GOI, genes of interest; incl, including; LOH, loss of heterozygosity; n,number of
cases that underwent whole exome sequencing; N/T, not tested; PA, pituitary adenoma; PRLoma, prolactinoma
*particular genes of interest
Endocr Pathol
Adelaide Hospital: SSA/18/CALHN/445) and with the National Health
and Medical Research Council guidelines.
Informed Consent Informed consent was obtained from all individual
participants included in the study.
References
1. Tordjman K, Stern N, Ouaknine G, Yossiphov Y, Razon N,
Nordenskjold M, Friedman E (1993) Activating mutations of the
Gs alpha-gene in nonfunctioning pituitary tumors. J Clin
Endocrinol Metab 77 (3):765769. https://doi.org/10.1210/jcem.
77.3.8396579
2. Bi WL, Greenwald NF, Ramkissoon SH, Abedalthagafi M, Coy
SM,LigonKL,MeiY,MacConaill L, Ducar M, Min L,
Santagata S, Kaiser UB, Beroukhim R, Laws ER, Jr., Dunn IF
(2017) Clinical Identification of Oncogenic Drivers and Copy-
Number Alterations in Pituitary Tumors. Endocrinology 158 (7):
22842291. https://doi.org/10.1210/en.2016-1967
3. Song ZJ, Reitman ZJ, Ma ZY, Chen JH, Zhang QL, Shou XF,
Huang CX, Wang YF, Li SQ, Mao Y, Zhou LF, Lian BF, Yan H,
Shi YY, Zhao Y (2016) The genome-wide mutational landscape of
pituitary adenomas. Cell Res 26 (11):12551259. https://doi.org/
10.1038/cr.2016.114
4. Reincke M, Sbiera S, Hayakawa A, Theodoropoulou M, Osswald
A, Beuschlein F (2015) Mutations in the deubiquitinase gene USP8
cause Cushing's disease. Nat Genet 47 (1):3138. https://doi.org/
10.1038/ng.3166
5. Brastianos PK, Taylor-Weiner A, Manley PE, Jones RT, Dias-
Santagata D, Thorner AR, Lawrence MS, Rodriguez FJ, Bernardo
LA, Schubert L, Sunkavalli A, Shillingford N, Calicchio ML,
Lidov HG, Taha H, Martinez-Lage M, Santi M, Storm PB, Lee
JY, Palmer JN, Adappa ND, Scott RM, Dunn IF, Laws ER, Jr.,
Stewart C, Ligon KL, Hoang MP, Van Hummelen P, Hahn WC,
Louis DN, Resnick AC, Kieran MW, Getz G, Santagata S (2014)
Exome sequencing identifies BRAF mutations in papillary
craniopharyngiomas. Nat Genet 46 (2):161165. https://doi.org/
10.1038/ng.2868
6. Bi WL, Larsen AG, Dunn IF (2018) Genomic Alterations in
Sporadic Pituitary Tumors. Curr Neurol Neurosci Rep 18 (1):4.
https://doi.org/10.1007/s11910-018-0811-0
7. Cohen M, Persky R, Stegemann R, Hernandez-Ramirez LC, Zeltser
D, Lodish MB, Chen A, Keil MF, Tatsi C, Faucz F, Buchner D,
Stratakis CA, Tiosano D (2019) Germline USP8 mutation associ-
ated with pediatric Cushing disease and other clinical features: a
new syndrome. J Clin Endocrinol Metab. https://doi.org/10.1210/
jc.2019-00697
8. Poncin J, Stevenaert A, Beckers A (1999) Somatic MEN1 gene
mutation does not contribute significantly to sporadic pituitary tu-
morigenesis. Eur J Endocrinol 140 (6):573576
9. Raitila A, Georgitsi M, Karhu A, Tuppurainen K, Makinen MJ,
Birkenkamp-Demtroder K, Salmenkivi K, Orntoft TF, Arola J,
Launonen V, Vahteristo P, Aaltonen LA (2007) No evidence of
somatic aryl hydrocarbon receptor interacting protein mutations in
sporadic endocrine neoplasia. Endocr Relat Cancer 14 (3):901906.
https://doi.org/10.1677/erc-07-0025
10. Tanizaki Y, Jin L, Scheithauer BW, Kovacs K, Roncaroli F, Lloyd
RV (2007) P53 gene mutations in pituitary carcinomas. Endocr
Pathol 18 (4):217222. https://doi.org/10.1007/s12022-007-9006-y
11. Yagnik G, Jahangiri A, Chen R, Wagner JR, Aghi MK (2017) Role
of a p53 polymorphism in the development of nonfunctional pitu-
itary adenomas. Mol Cell Endocrinol 446:8190. https://doi.org/10.
1016/j.mce.2017.02.017
12. Gorvin CM, Newey PJ, Rogers A, Stokes V, Neville MJ, Lines KE,
Ntali G, Lees P, Morrison PJ, Singhellakis PN, Malandrinou FC,
Karavitaki N, Grossman AB, Karpe F, Thakker RV (2018)
Association of prolactin receptor (PRLR) variants with
prolactinomas. Hum Mol Genet. https://doi.org/10.1093/hmg/
ddy396
13. Cai WY, Alexander JM, Hedley-Whyte ET, Scheithauer BW,
Jameson JL, Zervas NT, Klibanski A (1994) ras mutations in hu-
man prolactinomas and pituitary carcinomas. J Clin Endocrinol
Metab 78 (1):8993. https://doi.org/10.1210/jcem.78.1.8288721
14. Trivellin G, Daly AF, Faucz FR, Yuan B, Rostomyan L, Larco DO,
Schernthaner-Reiter MH, Szarek E, Leal LF, Caberg J-H,
Castermans E, Villa C, Dimopoulos A, Chittiboina P, Xekouki P,
Shah N, Metzger D, Lysy P, Ferrante E, Strebkova N, Mazerkina N,
Zatelli M, Lodish M, Horvath A, de Alexandre R, Manning A,
Levy I, Keil M, Sierra Mde L, Palmeira L, Coppieters W,
Georges M, Naves L, Jamar M, Bours V, Wu T, Choong C,
Bertherat J, Chanson P, Kamenický P, Farrell W, Barlier A,
Quezado M, Bjelobaba I, Stojilkovic S, Wess J, Costanzi S, Liu P,
Lupski J, Beckers A, Stratakis C (2014) Gigantism andacromegaly
due to Xq26 microduplications and GPR101 mutation. N Engl J
Med 371 (25):23632374
15. Assié G, Libé R, Espiard S, Rizk-Rabin M, Guimier A, Luscap W,
Barreau O, Lefèvre L, Sibony M, Guignat L, Rodriguez S,
Perlemoine K, René-Corail F, Letourneur F, Trabulsi B, Poussier
A, Chabbert-Buffet N, Borson-Chazot F, Groussin L, Bertagna X,
Stratakis C, Ragazzon B, Bertherat J (2013) ARMC5 mutations in
macronodular adrenal hyperplasia with Cushing's syndrome. N
Engl J Med 369 (22):21052114
16. Wang F, Gao H, Li C, Bai J, Lu R, Cao L, Wu Y, Hong L, Wu Y,
Lan X, Zhang Y (2014) Low levels of PRB3 mRNA are associated
with dopamine-agonist resistance and tumor recurrence in
prolactinomas. J Neurooncol 116 (1):8388. https://doi.org/10.
1007/s11060-013-1276-2
17. Gao H, Wang F, Lan X, Li C, Feng J, Bai J, Cao L, Gui S, Hong L,
Zhang Y (2015) Lower PRDM2 expression is associated with
dopamine-agonist resistance and tumor recurrence in
prolactinomas. BMC Cancer 15:272. https://doi.org/10.1186/
s12885-015-1267-0
18. Bi WL, Horowitz P, Greenwald NF, Abedalthagafi M, Agarwalla
PK, Gibson WJ, Mei Y, Schumacher SE, Ben-David U, Chevalier
A, Carter S, Tiao G, Brastianos PK, Ligon AH, Ducar M,
MacConaill L, Laws ER, Jr., Santagata S, Beroukhim R, Dunn IF
(2017) Landscape of Genomic Alterations in Pituitary Adenomas.
Clin Cancer Res 23 (7):18411851. https://doi.org/10.1158/1078-
0432.ccr-16-0790
19. Wierinckx A, Delgrange E, Bertolino P, François P, Chanson P,
Jouanneau E, Lachuer J, Trouillas J, Raverot G (2018) Sex-
Related Differences in Lactotroph Tumor Aggressiveness Are
Associated With a Specific Gene-Expression Signature and
Genome Instability. Front Endocrinol (Lausanne) 9:706706.
https://doi.org/10.3389/fendo.2018.00706
20. De Sousa SM, McCabe MJ, Wu K, Roscioli T, Gayevskiy V, Brook
K, Rawlings L, Scott HS, Thompson TJ, Earls P, Gill AJ, Cowley
MJ, Dinger ME, McCormack AI (2017) Germline variants in fa-
milial pituitary tumour syndrome genes are common in young pa-
tients and families with additional endocrine tumours. Eur J
Endocrinol 176 (5):635644. https://doi.org/10.1530/eje-16-0944
21. Zhang Q, Peng C, Song J,Zhang Y, Chen J,SongZ, Shou X, Ma Z,
Peng H, Jian X, He W, Ye Z, Li Z, Wang Y, Ye H, Zhang Z, Shen M,
Tang F, Chen H, Shi Z, Chen C, Chen Z, Shen Y, Wang Y, Lu S,
Zhang J, Li Y, Li S, Mao Y, Zhou L, Yan H, Shi Y, Huang C, Zhao
Y (2017) Germline Mutations in CDH23, Encoding Cadherin-
Related 23, Are Associated with Both Familial and Sporadic
Pituitary Adenomas. Am J Hum Genet 100 (5):817823. https://
doi.org/10.1016/j.ajhg.2017.03.011
Endocr Pathol
22. Rostomyan L, Daly AF, Petrossians P, Nachev E, Lila AR, Lecoq
AL,LecumberriB,TrivellinG,SalvatoriR,MoraitisAG,
Holdaway I, Kranenburg-van Klaveren DJ, Chiara Zatelli M,
Palacios N, Nozieres C, Zacharin M, Ebeling T, Ojaniemi M,
Rozhinskaya L, Verrua E, Jaffrain-Rea ML, Filipponi S,
Gusakova D, Pronin V, Bertherat J, Belaya Z, Ilovayskaya I,
Sahnoun-Fathallah M, Sievers C, Stalla GK, Castermans E,
Caberg JH, Sorkina E, Auriemma RS, Mittal S, Kareva M, Lysy
PA, Emy P, De Menis E, Choong CS, Mantovani G, Bours V, De
Herder W, Brue T, Barlier A, Neggers SJ,Zacharieva S, Chanson P,
Shah NS, Stratakis CA, Naves LA, Beckers A (2015) Clinical and
genetic characterization of pituitary gigantism: an international col-
laborative study in 208 patients. Endocr Relat Cancer 22 (5):745
757. https://doi.org/10.1530/erc-15-0320
23. Pepe S, Korbonits M, Iacovazzo D (2019) Germline and mosaic
mutations causing pituitary tumours: genetic and molecular aspects.
J Endocrinol 240 (2):R21-r45. https://doi.org/10.1530/joe-18-0446
24. De Sousa SMC, McCormack AI (2019) Aggressive Pituitary
Tumors and Pituitary Carcinomas. In: Feingold KR, Anawalt B,
Boyce A et al. (eds) Endotext. MDText.com, Inc., South
Dartmouth (MA),
25. Newey PJ, Nesbit MA, Rimmer AJ, Head RA, Gorvin CM, Attar
M, Gregory L, Wass JA, Buck D, Karavitaki N, Grossman AB,
McVean G, Ansorge O, Thakker RV (2013) Whole-exome se-
quencing studies of nonfunctioning pituitary adenomas. J Clin
Endocrinol Metab 98 (4):E796E800. https://doi.org/10.1210/jc.
2012-4028
26. Sapkota S, Horiguchi K, Tosaka M, Yamada S, Yamada M (2017)
Whole-Exome Sequencing Study of Thyrotropin-Secreting
Pituitary Adenomas. J Clin Endocrinol Metab 102 (2):566575.
https://doi.org/10.1210/jc.2016-2261
27. Evans CO, Moreno CS, Zhan X, McCabe MT, Vertino PM,
Desiderio DM, Oyesiku NM (2008) Molecular pathogenesis of
human prolactinomas identified by gene expression profiling, RT-
qPCR, and proteomic analyses. Pituitary 11 (3):231245. https://
doi.org/10.1007/s11102-007-0082-2
28. Wu ZB, Zheng WM, Su ZP, Chen Y, Wu JS, Wang CD, Lin C, Zeng
YJ, Zhuge QC (2010) Expression of D2RmRNA isoforms and
ERmRNA isoforms in prolactinomas: correlation with the response
to bromocriptine and with tumor biological behavior. J Neurooncol
99 (1):2532. https://doi.org/10.1007/s11060-009-0107-y
29. Kelly MA, Rubinstein M, Asa SL, Zhang G, Saez C, Bunzow JR,
Allen RG, Hnasko R, Ben-Jonathan N, Grandy DK, Low MJ
(1997) Pituitary lactotroph hyperplasia and chronic
hyperprolactinemia in dopamine D2 receptor-deficient mice.
Neuron 19 (1):103113
30. Brancati F, Camerota L, Colao E, Vega-Warner V, Zhao X, Zhang
R, Bottillo I (2018) Biallelic variants in the ciliary gene TMEM67
cause RHYNS syndrome. Eur J Hum Genet 26 (9):12661271.
https://doi.org/10.1038/s41431-018-0183-6
31. Wu Y, Berends MJ, Sijmons RH, Mensink RG, Verlind E, Kooi
KA, van der Sluis T, Kempinga C, van dDer Zee AG, Hollema H,
Buys CH, Kleibeuker JH, Hofstra RM (2001) A role for MLH3 in
hereditary nonpolyposis colorectal cancer. Nat Genet 29 (2):137
138. https://doi.org/10.1038/ng1001-137
32. Bengtsson D, Joost P, Aravidis C, Askmalm Stenmark M, Backman
AS, Melin B,von Salome J, Zagoras T, Gebre-Medhin S, Burman P
(2017) Corticotroph Pituitary Carcinoma in a Patient With Lynch
Syndrome (LS) and Pituitary Tumors in a Nationwide LS Cohort. J
Clin Endocrinol Metab 102 (11):39283932. https://doi.org/10.
1210/jc.2017-01401
33. Branford S, Wang P, Yeung DT, Thomson D, Purins A, Wadham C,
Shahrin NH, Marum JE, Nataren N, Parker WT, Geoghegan J, Feng
J, Shanmuganathan N, Mueller MC, Dietz C, Stangl D, Donaldson
Z, Altamura H, Georgievski J, Braley J, Brown A, Hahn C, Walker
I, Kim SH, Choi SY, Park SH, Kim DW, White DL, Yong ASM,
Ross DM, Scott HS, Schreiber AW, Hughes TP (2018) Integrative
genomic analysis reveals cancer-associated mutations at diagnosis
of CML in patients with high-risk disease. Blood 132 (9):948961.
https://doi.org/10.1182/blood-2018-02-832253
34. De Sousa SM, Hardy TS, Scott HS, Torpy DJ (2018) Genetic
Testing in Endocrinology. Clin Biochem Rev 39 (1):1728
35. Thrane K, Eriksson H, Maaskola J, Hansson J, Lundeberg J (2018)
Spatially Resolved Transcriptomics Enables Dissection of Genetic
Heterogeneity in StageIII Cutaneous Malignant Melanoma. Cancer
Res 78 (20):59705979. https://doi.org/10.1158/0008-5472.can-
18-0747
PublishersNote Springer Nature remains neutral with regard to jurisdic-
tional claims in published maps and institutional affiliations.
Endocr Pathol
... [10][11][12][13] Exome sequencing identified germline CDH23 and somatic PHTF1 and POU6F2 variants, which still need to be confirmed in other cohorts. 11,[13][14][15][16] Whole-genome sequencing in a selected lactotroph tumour cohort discovered a somatic variant p.Arg625His in exon 14 of the splicing factor 3b subunit 1 (SF3B1) gene in 2/21 cases as well as in an aggressive lactotroph tumour. 17,18 In this large cohort study, SF3B1 p.Arg625His was reported in 19.8% of cases. ...
... 19 However, other whole-exome sequencing studies did not report SF3B1 variants in lactotroph tumours. 11,13,15,20,21 The aim of our multicentre study was to screen the exon 14 of the SF3B1 hotspot region for driver variants and determine their prevalence in a large independent cohort of lactotroph tumours and correlation with clinicopathological data. ...
... Histologically [12][13][14][15][16] vs 0 (0-40); Mann-Whitney U test, P = .008); we did not detect any differences in p53 immunostaining (Table 1). ...
Article
Objective A somatic mutational hotspot in the SF3B1 gene was reported in lactotroph tumours. The aim of our study was to examine the prevalence of driver SF3B1 variants in a multicentre independent cohort of patients with lactotroph tumours and correlate with clinical data. Design and methods This was a retrospective, multicentre study involving 282 patients with lactotroph tumours (including 6 metastatic lactotroph tumours) from 8 European centres. We screened SF3B1 exon 14 hotspot for somatic variants using Sanger sequencing and correlated with clinicopathological data. Results We detected SF3B1 variants in seven patients with lactotroph tumours: c.1874G > A (p.Arg625His) (n = 4, 3 of which metastatic) and a previously undescribed in pituitary tumours variant c.1873C > T (p.Arg625Cys) (n = 3 aggressive pituitary tumours). In two metastatic lactotroph tumours with tissue available, the variant was detected in both primary tumour and metastasis. The overall prevalence of likely pathogenic SF3B1 variants in lactotroph tumours was 2.5%, but when we considered only metastatic cases, it reached the 50%. SF3B1 variants correlated with significantly larger tumour size; higher Ki67 proliferation index; multiple treatments, including radiotherapy and chemotherapy; increased disease-specific death; and shorter postoperative survival. Conclusions SF3B1 variants are uncommon in lactotroph tumours but may be frequent in metastatic lactotroph tumours. When present, they associate with aggressive tumour behaviour and worse clinical outcome.
... in sporadic PAs (Bi et al. 2017b, De Sousa et al. 2019. CNVs are especially frequent in functioning PAs, particularly prolactinomas, as well as PAs with high proliferative indices (Bi et al. 2017b). ...
... In an Australian WES study of prolactinomas, recurrent copy number gains and copy-neutral LOH were observed at several sites (De Sousa et al. 2019). Other NGS studies involving a mix of PA subtypes have demonstrated copy number losses, in addition to gains (Song et al. 2016, Bi et al. 2017b) and LOH (Bi et al. 2017b). ...
... This may be at least partly explained by the heterogenous mixes of different PA subtypes and clinical features. In prolactinomas, for example, DA resistance may lead to accumulated copy number variation or vice versa (De Sousa et al. 2019). ...
Article
Full-text available
'Pituitary tumours' is an umbrella term for various tumours originating from different regions of the hypothalamic-pituitary system. The vast majority of pituitary tumours are pituitary adenomas, also recently referred to as pituitary neuroendocrine tumours (PitNET). The prevalence of clinically relevant pituitary adenomas is approximately 1 in 1000; other pituitary tumours such as craniopharyngioma and pituicytoma are comparatively very rare. This review addresses the molecular and genetic aspects of pituitary adenomas. We first discuss the germline genetic variants underlying familial pituitary tumours, which account for approximately 5% of all pituitary adenoma cases. This includes variants in established pituitary adenoma/hyperplasia predisposition genes (MEN1, PRKAR1A, AIP, CDKN1B, GPR101, SDHA, SDHB, SDHC, SDHD, SDHAF2) as well as emerging genetic associations. In addition, we discuss McCune-Albright syndrome which lies between the germline and somatic pituitary tumour genes as the causative GNAS mutations are postzygotic rather than being inherited, and the condition is associated with multiglandular features due to involvement of different cell lines rather than being limited to the pituitary. By contrast, somatic GNAS mutations contribute to sporadic acromegaly. USP8 is the only other gene where somatic driver mutations have been established in sporadic pituitary tumorigenesis. However, there are now known to be a variety of other somatic genetic and molecular changes underpinning sporadic pituitary adenomas which we will review, namely: copy number variation, molecular changes in signalling and hypoxia pathways, epithelial-mesenchymal transition, DNA repair, senescence, the immune microenvironment and epigenetics.
... DA resistance in prolactinomas has been associated with reduced D2 receptor density, overall reduction in D2 receptor mRNA production and altered expression of D2 receptor mRNA isoforms with lower expression of the more efficient short isoform (Wu et al. 2010). A nonsense DRD2 variant has been found in tumour DNA from an aggressive prolactinoma with DA resistance (De Sousa et al. 2019b). However, DRD2 variants are not a consistent feature of resistant prolactinomas (Wang et al. 2014, De Sousa et al. 2019b. ...
... A nonsense DRD2 variant has been found in tumour DNA from an aggressive prolactinoma with DA resistance (De Sousa et al. 2019b). However, DRD2 variants are not a consistent feature of resistant prolactinomas (Wang et al. 2014, De Sousa et al. 2019b. ...
... As features such as larger tumour size and greater tumour invasiveness are predictive of both DA resistance and poorer surgical outcomes (Honegger et al. 2020, Wright et al. 2021, there is a need for research into novel prolactinoma treatments that overcome the limitations of both DA therapy and surgery. Understanding the molecular basis of prolactinoma tumorigenesis may reveal new medical treatment targets, although genomic studies to date have not shown druggable driver mutations (De Sousa et al. 2019b). ...
Article
Full-text available
The current treatment paradigm for prolactinomas involves dopamine agonist (DA) therapy as the first-line treatment, with surgical resection reserved for cases where there is DA failure due to resistance or intolerance. This review highlights how DA therapy can be optimised to overcome its increasingly recognised pitfalls, whilst also addressing the potential for expanding the use of surgery in the management of prolactinomas. The first part of the review discusses the limitations of DA therapy, namely: DA resistance; common DA side effects; and the rare but serious DA-induced risks of cardiac valvulopathy, impulse control disorders, psychosis, CSF rhinorrhoea and tumour fibrosis. The second part of the review explores the role of surgery in prolactinoma management with reference to its current second-line position and recent calls for surgery to be considered as an alternative first-line treatment alongside DA therapy. Randomised trials comparing medical versus surgical therapy for prolactinomas are currently underway. Pending these results, a low surgical threshold approach is herein proposed, whereby DA therapy remains the default treatment for prolactinomas unless there are specific triggers to consider surgery, including concern regarding DA side effects or risks in vulnerable patients, persistent and bothersome DA side effects, emergence of any serious risks of DA therapy, expected need for long-term DA therapy, as well as the traditional indications for surgery. This approach should optimise the use of DA therapy for those who will most benefit from it, whilst instituting surgery early in others in order to minimise the cumulative burden of prolonged DA therapy.
... The primary cohort comprised 29 patients with PAs (12 prolactinomas, 10 thyrotropinomas, 7 cyclical Cushing disease) recruited from the Royal Adelaide Hospital, Royal Melbourne Hospital, and Royal North Shore Hospital as previously described (15)(16)(17)(18). Participants were required to have a prolactinoma, thyrotropinoma, or corticotropinoma causing cyclical Cushing disease, defined by biochemical documentation of episodic cortisol excess interspersed by periods of normal or low cortisol secretion (19). ...
... These 3 PA types were selected as the original aim of the primary cohort study was to identify novel somatic driver mutations, and these patient groups have traditionally lacked identifiable driver mutations. Our somatic WES analysis of these cohorts has previously been reported, with no recurrent sequence variants that could constitute somatic driver mutations being found (15)(16)(17). No other selection criteria were applied in the primary cohort apart from availability of a tumor specimen. ...
Article
Full-text available
Context CHEK2 is a cell cycle checkpoint regulator gene with a long-established role as a clinically relevant, moderate risk breast cancer predisposition gene, with greater risk ascribed to truncating variants than missense variants. Methods We assessed 165 individuals with pituitary adenomas for CHEK2 variants. The study consisted of a primary cohort of 29 individuals who underwent germline and tumour whole exome sequencing, and a second, independent cohort of 136 individuals who had a targeted next-generation sequencing panel performed on both germline and tumour DNA (n=52) or germline DNA alone (n=84). Results We identified rare, coding, non-synonymous germline CHEK2 variants amongst 3/29 (10.3%) patients in our primary cohort and 5/165 (3.0%) patients overall, with affected patients having a range of hormone secretion types (prolactinoma, thyrotrophinoma, somatotrophinoma and non-functioning pituitary adenoma). No somatic variants were identified. Two variants were definitive null variants (c.1100delC, c.444+1G>A), classified as pathogenic. Two variants were missense variants (p.Asn186His, p.Thr476Met), classified as likely pathogenic. Even when considering the null variants only, the rate of CHEK2 variants was higher in our cohort compared to national control data (1.8% vs. 0.5%, P=0.049). Conclusions This is the first study to suggest a role for the breast cancer predisposition gene, CHEK2, in pituitary tumorigenesis, with pathogenic/likely pathogenic variants found in 3% of patients with pituitary adenomas. As pituitary adenomas are relatively common and typically lack classical autosomal dominant family histories, risk alleles – such as these variants found in CHEK2 – might be a significant contributor to pituitary adenoma risk in the general population.
... Decreased expression of dopamine receptor type 2 has been reported (9). The dopamine agonist binding is normal and dopamine receptor pathogenic variants have been rarely described in PA (8,10,11). One previous study described a pathogenic variant in a case of resistant prolactinoma (10). ...
... The dopamine agonist binding is normal and dopamine receptor pathogenic variants have been rarely described in PA (8,10,11). One previous study described a pathogenic variant in a case of resistant prolactinoma (10). In this report, we explore an SDHB pathogenic variant as a possible cause of resistance to cabergoline (CBG) in a patient with a giant prolactinoma. ...
Article
Full-text available
Context Germline succinate dehydrogenase subunit B (SDHB) pathogenic variants are characteristic of familial paraganglioma (PGL) syndrome type 4. This syndrome frequently presents with abdominal PGL and has high tendency for locally aggressive behavior and distant metastasis. The vast majority of pituitary adenomas (PAs) are sporadic. However, PAs can be part of a number of familial tumor syndromes such as multiple endocrine neoplasia type 1 (MEN 1) or more rarely in association with pheochromocytoma and PGL (referred to as 3P syndrome). Only a limited number of PAs in association with SDHB-related PGL has been reported and the vast majority occurred subsequently or simultaneously with pheochromocytoma/PGL (collectively abbreviated as PPGL). In this report, we describe a young patient who had a giant pituitary macroprolactinoma resistant to large doses of cabergoline (CBG) and external beam radiotherapy (XRT). The patient did not have personal history of PPGL but was found to carry a germline SDHB pathogenic variant. Case report A 38-year-old woman presented with headache, visual disturbances and galactorrhea and was found to have a 34-mm macroprolactinoma. She was treated with CBG 3-4 mg per week but PA continued to grow and caused significant cranial pressure symptoms. She underwent two transsphenoidal surgeries with rapid tumor recurrence after each one. She received XRT but PA continued to grow. She was finally treated with temozolomide with excellent response. Whole exome and subsequent Sanger sequencing confirmed that she has a pathogenic monoallelic SDHB mutation (NM_003000:c.C343T, p.R115*). PA tissue showed loss of heterozygosity for the same mutation and absent SDHB immunostaining confirming the pathogenic role of this SDHB mutation. Conclusion Germline SDHB mutations can rarely cause PA in the absence of PPGL. They should be considered as a possible cause of aggressiveness and resistance to dopamine agonists in similar cases.
... Cyclical Cushing's disease was defined by biochemical documentation of episodic cortisol excess interspersed by periods of normal or low cortisol secretion (8). Clinical and genetic characteristics of some study participants have been previously published (9)(10)(11). Pathogenic germline variants in established pituitary predisposition genes (AIP, CDKN1B, MEN1, PRKAR1A, SDHA, SDHB, SDHC and SDHD) had been excluded in all individuals. ...
Article
Full-text available
Introduction Germline loss-of-function variants in PAM , encoding peptidylglycine α-amidating monooxygenase (PAM), were recently discovered to be enriched in conditions of pathological pituitary hypersecretion, specifically: somatotrophinoma, corticotrophinoma, and prolactinoma. PAM is the sole enzyme responsible for C-terminal amidation of peptides, and plays a role in the biosynthesis and regulation of multiple hormones, including proopiomelanocortin (POMC). Methods We performed exome sequencing of germline and tumour DNA from 29 individuals with functioning pituitary adenomas (12 prolactinomas, 10 thyrotrophinomas, 7 cyclical Cushing’s disease). An unfiltered analysis was undertaken of all PAM variants with population prevalence <5%. Results We identified five coding, non-synonymous PAM variants of interest amongst seven individuals (six germline, one somatic). The five variants comprised four missense variants and one truncating variant, all heterozygous. Each variant had some evidence of pathogenicity based on population prevalence, conservation scores, in silico predictions and/or prior functional studies. The yield of predicted deleterious PAM variants was thus 7/29 (24%). The variants predominated in individuals with thyrotrophinomas (4/10, 40%) and cyclical Cushing’s disease (2/7, 29%), compared to prolactinomas (1/12, 8%). Conclusion This is the second study to demonstrate a high yield of suspected loss-of-function, predominantly germline, PAM variants in individuals with pathological pituitary hypersecretion. We have extended the association with corticotrophinoma to include the specific clinical entity of cyclical Cushing’s disease and demonstrated a novel association between PAM variants and thyrotrophinoma. PAM variants might act as risk alleles for pituitary adenoma formation, with a possible genotype-phenotype relationship between truncating variants and altered temporal secretion of cortisol.
... Exome sequencing of eight thyropinomas have not revealed convincing recurrent somatic variants [11] in this study group, but exome analysis of larger group of 42 PitNETs indicated several mutations and insertions/ deletions in genes ATAD3B, BHLHE22, KDM2B, OR5M1, TTN, VPS13B that were encounter in more that one PitNET [12]. No recurrent somatic variants were found in the whole-exome sequencing of 12 prolactinomas [13]. Nemeth et al. have studied mitochondrial variants in 44 PitNET cases and found no significant association with clinical features of the tumor, however, they demonstrated that samples with the highest homoplasmic variant rate had the highest Ki-67 index [14]. ...
Article
Full-text available
Somatic genetic alterations in pituitary neuroendocrine tumors (PitNET) tissues have been identified in several studies, but detection of overlapping somatic PitNET candidate genes is rare. We sequenced and by employing multiple data analysis methods studied the exomes of 15 PitNET patients to improve discovery of novel factors involved in PitNET development. PitNET patients were recruited to the study before PitNET removal surgery. For each patient, two samples for DNA extraction were acquired: venous blood and PitNET tissue. Exome sequencing was performed using Illumina NexSeq 500 sequencer and data analyzed using two separate workflows and variant calling algorithms: GATK and Strelka2. A combination of two data analysis pipelines discovered 144 PitNET specific somatic variants (mean = 9.6, range 0–19 per PitNET) of which all were SNVs. Also, we detected previously known GNAS PitNET mutation and identified somatic variants in 11 genes, which have contained somatic variants in previous WES and WGS studies of PitNETs. Noteworthy, this is the third study detecting somatic variants in gene RYR1 in the exomes of PitNETs. In conclusion, we have identified two novel PitNET candidate genes ( AC002519 . 6 and AHNAK ) with recurrent somatic variants in our PitNET cohort and found 13 genes overlapping from previous PitNET studies that contain somatic variants. Our study demonstrated that the use of multiple sequencing data analysis pipelines can provide more accurate identification of somatic variants in PitNETs.
... 63,247,273 In contrast, it did not reveal additional driver mutations in somatotroph, thyrotroph, lactotroph, or nonfunctioning pituitary tumors. 245,[274][275][276] On the other hand, applying variable omics techniques on pituitary tumors revealed variable chromosomal alterations and epigenomic signatures that do not always coincide with the established histological diagnosis and invasive behavior. 63,247,273,277 It is possible that the formation of pituitary tumors with no known driver mutation is triggered by genetic defects hidden in the noncoding region. ...
Chapter
Pituitary tumors are the second most frequent intracranial neoplasm. They are sporadic in their origin and their pathophysiology is tightly linked to deregulated hypothalamic, peripheral hormonal as well as autocrine/paracrine signals. Pituitary tumors are monoclonal, but only two genes, GNAS and USP8, are found to be mutated more than 5% of sporadic pituitary tumors. GNAS1 is mutated in ~40% of somatotroph and USP8 in ~50% of corticotroph tumors. Overall, few driver mutations have been discovered, indicating the presence of other still obscure genetic defects that may trigger and drive tumorigenesis in the majority of pituitary tumors.
... Prolactinomas can appear as a result of germline mutation present in multiple endocrine neoplasia, familial isolated pituitary adenomas (FIPA) [64] or Carney complex [65]. However, somatic mutations, as occur in other pituitary tumours such as corticotrophinoma, have only occasionally been reported [66,67]. Nevertheless, Li et al. [68] have recently identified a hotspot somatic mutation in splicing factor 3 subunit B1 (SF3B1) in up to 19.8% of prolactinomas. ...
Article
Full-text available
Prolactinoma has the highest incidence rate among patients with functional pituitary tumours. Although mostly benign, there is a subgroup that can be aggressive. Some clinical, radiological and pathology features have been associated with a poor prognostic. Therefore, it can be considered as a group of heterogeneous tumours. The aim of this paper is to give an overview of the molecular pathways involved in the behaviour of prolactinoma in order to improve our approach and gain deeper insight into the better understanding of tumour development and its management. This is essential for identifying patients harbouring aggressive prolactinoma and to establish personalised therapeutics options.
Article
Full-text available
Background Somatic mutations in the ubiquitin-specific peptidase 8 (USP8) gene are common in corticotropinomas of children with Cushing disease (CD). We present a unique patient with a germline USP8 mutation, who presented with CD and a constellation of other findings that constitute a new genetic syndrome. Case description We describe a 16-year-old female with CD, developmental delay, dysmorphic features, ichthyosiform hyperkeratosis, chronic lung disease, chronic kidney disease, hyperglycemia, dilated cardiomyopathy with congestive heart failure (CHF), and previous history of hyperinsulinism and partial growth hormone (GH) deficiency. She was diagnosed with CD at 14 years old and underwent trans-sphenoidal surgery. Despite initial improvement, she developed recurrent CD. Methods DNA was extracted from peripheral blood and tumor DNA; whole-exome and Sanger confirmatory sequencing were performed. Immunohistochemistry was performed on the resected adenoma. Results A de novo germline heterozygous USP8 mutation (c.2155T>C, p.S719P) in the critical 14-3-3 binding motif hotspot locus of the gene was identified in both the peripheral blood and tumor DNA. Histopathologic evaluation of the resected tumor confirmed an ACTH-secreting adenoma. Conclusions Somatic USP8 mutations are common in adenomas causing CD, but to date no germline defects have been reported. We describe a patient with a de novo germline USP8 mutation with recurrent CD and multiple other medical problems. This unique patient informs us of the multitude of signaling events that may be controlled by USP8.
Chapter
Full-text available
Aggressive pituitary tumors (APT) refer to pituitary adenomas exhibiting rapid growth, resistance to conventional treatments and/or early/multiple recurrences, with abandonment of the previous term ‘atypical pituitary adenoma’. Pituitary carcinomas (PC) are defined by non-contiguous craniospinal or distant metastasis. Whilst PC is exceedingly rare, comprising only 0.1-0.2% of all pituitary neoplasms, APT may account for up to 15% of all pituitary neoplasms, depending on the definition used. Typically evolving from known pituitary macroadenomas, APT/PC is most commonly diagnosed in the fifth decade of life with corticotroph and lactotroph neoplasms predominating. Diagnosis relies on MRI, hormonal studies and histological assessment including proliferative markers and immunohistochemistry for pituitary hormones and, most recently, transcription factors. Structural and molecular mechanisms have been proposed in the pathogenesis of APT/PC, although there appears to be no contribution from known familial pituitary tumor syndrome genes such as MEN1. Treatment is multimodal, ideally delivered by an expert team with a high-volume caseload. Surgical resection may be performed with the aim of either gross total resection or tumor debulking. Radiotherapy may be administered either as fractionated external beam radiation or stereotactic radiosurgery. Standard pituitary medical therapies such as somatostatin analogues have limited efficacy in APT/PC, whereas temozolomide yields a clear survival benefit. Evidence is emerging for the use of peptide receptor radionuclide therapy, tyrosine kinase inhibitors, VEGF inhibitors, and immunotherapy. Avenues for further research in APT/PC include molecular biomarkers, nuclear imaging, establishment of an international register, and routine pituitary tumor biobanking.
Article
Full-text available
While 95% of pituitary adenomas arise sporadically without a known inheritable predisposing mutation, in about 5% of the cases they can arise in a familial setting, either isolated (familial isolated pituitary adenoma or FIPA) or as part of a syndrome. FIPA is caused, in 15-30% of all kindreds, by inactivating mutations in the AIP gene, encoding a co-chaperone with a vast array of interacting partners and causing most commonly growth hormone excess. While the mechanisms linking AIP with pituitary tumorigenesis have not been fully understood, they are likely to involve several pathways, including the cAMP-dependent protein kinase A pathway via defective G inhibitory protein signalling or altered interaction with phosphodiesterases. The cAMP pathway is also affected by other conditions predisposing to pituitary tumours, including X-linked acrogigantism caused by duplications of the GPR101 gene, encoding an orphan G stimulatory protein-coupled receptor. Activating mosaic mutations in the GNAS gene, coding for the Gα stimulatory protein, cause McCune-Albright syndrome, while inactivating mutations in the regulatory type 1α subunit of protein kinase A represent the most frequent genetic cause of Carney complex, a syndromic condition with multi-organ manifestations also involving the pituitary gland. In this review, we discuss the genetic and molecular aspects of isolated and syndromic familial pituitary adenomas due to germline or mosaic mutations, including those secondary to AIP and GPR101 mutations, multiple endocrine neoplasia type 1 and 4, Carney complex, McCune-Albright syndrome, DICER1 syndrome and mutations in the SDHx genes underlying the association of familial paragangliomas and phaeochromocytomas with pituitary adenomas.
Article
Full-text available
Sex-related differences have been reported in various cancers, in particular men with lactotroph tumors have a worse prognosis than women. While the underlying mechanism of this sexual dimorphism remains unclear, it has been suggested that a lower estrogen receptor alpha expression may drive the sex differences observed in aggressive and malignant lactotroph tumors that are resistant to dopamine agonists. Based on this observation, we aimed to explore the molecular importance of the estrogen pathway through a detailed analysis of the transcriptomic profile of lactotroph tumors from 20 men and 10 women. We undertook gene expression analysis of the selected lactotroph tumors following their pathological grading using the five-tiered classification. Chromosomic alterations were further determined in 13 tumors. Functional analysis showed that there were differences between tumors from men and women in gene signatures associated with cell morphology, cell growth, cell proliferation, development, and cell movement. Hundred-forty genes showed an increased or decreased expression with a minimum 2-fold change. A large subset of those genes belonged to the estrogen receptor signaling pathway, therefore confirming the potent role of this pathway in lactotroph tumor sex-associated aggressiveness. Genes belonging to the X chromosome, such as CTAG2, FGF13, and VEGF-D, were identified as appealing candidates with a sex-linked dysregulation in lactotroph tumors. Through our comparative genomic hybridization analyses (CGH), chromosomic gain, in particular chromosome 19p, was found only in tumors from men, while deletion of chromosome 11 was sex-independent, as it was found in most (5/6) of the aggressive and malignant tumors. Comparison of transcriptomic and CGH analysis revealed four genes (CRB3, FAM138F, MATK, and STAP2) located on gained regions of chromosome 19 and upregulated in lactotroph tumors from men. MATK and STAP2 are both implicated in cell growth and are reported to be associated with the estrogen signaling pathway. Our work confirms the proposed involvement of the estrogen signaling pathway in favoring the increased aggressiveness of lactotroph tumors in men. More importantly, we highlight a number of ER-related candidate genes and further identify a series of target molecules with sex-specific expression that could contribute to the aggressive behavior of lactotroph tumors in men.
Article
Full-text available
Prolactinomas are the most frequent type of pituitary tumors, which represent 10-20% of all intracranial neoplasms in humans. Prolactinomas develop in mice lacking the prolactin receptor (PRLR), which is a member of the cytokine receptor superfamily that signals via Janus kinase-2-signal transducer and activator of transcription-5 (JAK2-STAT5) or phosphoinositide 3-kinase-Akt (PI3K-Akt) pathways to mediate changes in transcription, differentiation and proliferation. To elucidate the role of the PRLR gene in human prolactinomas, we determined the PRLR sequence in 50 DNA samples (35 leucocyte, 15 tumors) from 46 prolactinoma patients (59% males, 41% females). This identified six germline PRLR variants, which comprised four rare variants (Gly57Ser, Glu376Gln, Arg453Trp and Asn492Ile) and two low-frequency variants (Ile76Val, Ile146Leu), but no somatic variants. The rare variants, Glu376Gln and Asn492Ile, which were in complete linkage disequilibrium, and are located in the PRLR intracellular domain, occurred with significantly higher frequencies (p<0.0001) in prolactinoma patients than in 60,706 individuals of the Exome Aggregation Consortium cohort and 7,045 individuals of the Oxford Biobank. In vitro analysis of the PRLR variants demonstrated that the Asn492Ile variant, but not Glu376Gln, when compared to wild-type PRLR, increased prolactin-induced pAkt signaling (>1.3-fold, p<0.02) and proliferation (1.4-fold, p<0.02), but did not affect pSTAT5 signaling. Treatment of cells with an Akt1/2 inhibitor or everolimus, which acts on the Akt pathway, reduced Asn492Ile signaling and proliferation to wild-type levels. Thus, our results identify an association between a gain-of-function PRLR variant and prolactinomas, and reveal a new aetiology and potential therapeutic approach for these neoplasms.
Article
Full-text available
Cutaneous malignant melanoma (melanoma) is characterized by a high mutational load, extensive intertumoral and intratumoral genetic heterogeneity, and complex tumor microenvironment (TME) interactions. Further insights into the mechanisms underlying melanoma are crucial for understanding tumor progression and responses to treatment. Here we adapted the technology of spatial transcriptomics (ST) to melanoma lymph node biopsies and successfully sequenced the transcriptomes of over 2,200 tissue domains. Deconvolution combined with traditional approaches for dimensional reduction of transcriptome-wide data enabled us to both visualize the transcriptional landscape within the tissue and identify gene expression profiles linked to specific histologic entities. Our unsupervised analysis revealed a complex spatial intratumoral composition of melanoma metastases that was not evident through morphologic annotation. Each biopsy showed distinct gene expression profiles and included examples of the coexistence of multiple melanoma signatures within a single tumor region as well as shared profiles for lymphoid tissue characterized according to their spatial location and gene expression profiles. The lymphoid area in close proximity to the tumor region displayed a specific expression pattern, which may reflect the TME, a key component to fully understanding tumor progression. In conclusion, using the ST technology to generate gene expression profiles reveals a detailed landscape of melanoma metastases. This should inspire researchers to integrate spatial information into analyses aiming to identify the factors underlying tumor progression and therapy outcome. Significance: Applying ST technology to gene expression profiling in melanoma lymph node metastases reveals a complex transcriptional landscape in a spatial context, which is essential for understanding the multiple components of tumor progression and therapy outcome. Cancer Res; 78(20); 5970–9. ©2018 AACR.
Article
Full-text available
The recent genomic revolution, characterised by surges in the number of available genetic tests and known genetic associations, calls for improved genetic literacy amongst medical scientists and clinicians. This has been driven by next generation sequencing, a technology allowing multiple genes to be sequenced in parallel, thereby reducing the time and financial costs associated with genetic testing in both research and clinical settings. Endocrinology is an intuitive setting in which to consider the principles of genetic testing because endocrine disorders are due to defects in circumscribed pathways, providing clues to candidate genes. This article discusses genetic testing in contemporary endocrine practice with reference to examples of endocrine genetic disorders or multisystem genetic disorders with endocrine manifestations. Monogenic disorders are prioritised as these form the bulk of endocrine genetic disorders and the associated genetic testing is readily understandable, clinically available and practice-changing. Although it remains true that genetic testing should be embarked upon only if the result will alter management, the clinical utility of genetic testing is often underestimated and there are expanding indications for genetic testing across all areas of endocrinology.
Article
Full-text available
A rare syndrome was first described in 1997 in a 17-year-old male patient presenting with Retinitis pigmentosa, HYpopituitarism, Nephronophthisis and Skeletal dysplasia (RHYNS). In the single reported familial case, two brothers were affected, arguing for X-linked or recessive mode of inheritance. Up to now, the underlying genetic basis of RHYNS syndrome remains unknown. Here we applied whole-exome sequencing in the originally described family with RHYNS to identify compound heterozygous variants in the ciliary gene TMEM67. Sanger sequencing confirmed a paternally inherited nonsense c.622A > T, p.(Arg208*) and a maternally inherited missense variant c.1289A > G, p.(Asp430Gly), which perturbs the correct splicing of exon 13. Overall, TMEM67 showed one of the widest clinical continuum observed in ciliopathies ranging from early lethality to adults with liver fibrosis. Our findings extend the spectrum of phenotypes/syndromes resulting from biallelic TMEM67 variants to now eight distinguishable clinical conditions including RHYNS syndrome.
Article
Full-text available
Purpose of review: Pituitary tumors are undergoing a transformation in histopathologic and molecular classification, coincident with the continued refinement of increasingly powerful methods of genomic annotation and discovery. We highlight novel genomic alterations identified in pituitary adenomas and craniopharyngiomas and discuss their clinical implications. Recent findings: Sporadic pituitary adenomas are associated with relatively few recurrent somatic mutations. Recurrent mutations occur largely in subsets of hormone-producing tumors, including GNAS and GPR101 in somatotroph adenomas and USP8 in corticotroph adenomas. Additionally, they manifest with a dichotomous signature of copy number alterations, ranging from almost none to widespread genome instability, while microduplication of chromosome Xq26.3, containing the GNAS gene, defines X-linked acrogigantism. Papillary craniopharyngiomas are defined by BRAF V600E mutations while β-catenin alterations characterize adamantinomatous craniopharyngiomas. Genomic annotation of pituitary tumors is defining increasing subsets of neuroendocrine adenohypophyseal tumors and craniopharyngiomas, offering rationale-based pharmacologic targets and potential biomarkers for clinical outcome.
Article
Key Points Next-generation sequencing revealed variants in cancer-associated genes at diagnosis of CML more frequently in patients with poor outcomes. All patients at BC had mutated cancer genes, including fusions, that predated BCR-ABL1 kinase domain mutations in a majority.