You are on page 1of 14

Endocrine Pathology (2021) 32:3–16

https://doi.org/10.1007/s12022-021-09663-4

Genomics and Epigenomics of Pituitary Tumors: What Do Pathologists


Need to Know?
Sylvia L. Asa1,2   · Ozgur Mete2,3 · Shereen Ezzat4

Accepted: 4 January 2021 / Published online: 12 January 2021


© The Author(s), under exclusive licence to Springer Science+Business Media, LLC part of Springer Nature 2021

Abstract
Molecular pathology has advanced our understanding of many tumors and offers opportunities to identify novel therapies. In
the pituitary, the field has uncovered several genetic mutations that predispose to pituitary neuroendocrine tumor (PitNET)
development, including MEN1, CDKN1B, PRKRIα, AIP, GPR101, and other more rare events; however, these genes are only
rarely mutated in sporadic PitNETs. Recurrent genetic events in sporadic PitNETs include GNAS mutations in a subset of
somatotroph tumors and ubiquitin-specific peptidase mutations (e.g., USP8, USP48) in some corticotroph tumors; to date,
neither of these has resulted in altered management, and instead, the prognosis and management of PitNETs still rely more
on cell type and subtype as well as local growth that determines surgical resectability. In contrast, craniopharyngiomas have
either CTNNB1 or BRAFV600E mutations that correlate with adamantinomatous or papillary morphology, respectively; the
latter offers the opportunity for targeted therapy. DICER1 mutations are found in patients with pituitary blastoma. Epige-
netic changes are implicated in the pathogenesis of the more common sporadic pituitary neoplasms including the majority
of PitNETs and tumors of pituicytes.

Keywords  Pituitary neuroendocrine tumor · Pituicytoma · Craniopharyngioma · Pituitary blastoma · Mutation · Epigenetics

Introduction somatotrophs, lactotrophs, mammosomatotrophs,


and thyrotrophs, and SF1-lineage gonadotrophs.
The pituitary is a complex endocrine gland. The anterior Corticotrophs produce pro-opiomelanocortin (POMC)
lobe is composed of epithelial neuroendocrine cells; the that is cleaved into several hormones, the most critical
posterior lobe is composed of pituicytes (modified glia) and being adrenocorticotropin (ACTH) that regulates adrenal
the terminals of hypothalamic neuronal hormone-secreting glucocorticoid production; other products of POMC include
cells [1]. Together, they are responsible for synthesis and melanocyte-stimulating hormone (MSH), endorphins and
secretion of peptide hormones which regulate metabolism, enkephalins that modulate pain and anxiety perception, and
growth, and reproduction. In the adenohypophysis, there are several other bioactive peptides. The PIT1 family of cells
seven known terminally differentiated epithelial cell types is complex. Somatotrophs are responsible for production
in three lineages: TPIT-lineage corticotrophs, PIT1-lineage of growth hormone (GH) that is required for normal
somatic growth in childhood and for maintenance of lean
body mass in adults. With the additional expression of
* Sylvia L. Asa
pathlady01@gmail.com estrogen receptor-alpha (ER), PIT1-lineage cells become
mammosomatotrophs that also produce prolactin (PRL), a
1
Department of Pathology, University Hospitals Cleveland hormone that is required for lactation but also plays a role
Medical Center, Case Western Reserve University, in emotional health including bonding. Both GH and PRL
Cleveland, Ohio, USA
also have been implicated in immune regulation. Mature
2
Department of Pathology, University Health Network, lactotrophs arise from mammosomatotrophs with the
Toronto, ON, Canada
additional expression of an uncharacterized GH repressor
3
Department of Laboratory Medicine and Pathobiology, and produce only PRL. PIT1-lineage cells that also express
University of Toronto, Toronto, ON, Canada
GATA2/3 develop into thyrotrophs that produce thyrotropin
4
Department of Medicine, University Health Network (thyroid-stimulating hormone, TSH). There is evidence of
and University of Toronto, Toronto, ON, Canada

13
Vol.:(0123456789)
4 Endocrine Pathology (2021) 32:3–16

fluidity of the PIT1-lineage cells that can transdifferentiate been identified in some tumor subtypes [7–8], and the
under physiological conditions. Somatotrophs predominate importance of recognizing genetic predisposition to these
during childhood and adolescence but transform into neuroendocrine tumors should not be overlooked; however,
mammosomatotrophs then lactotrophs during pregnancy the most important role of the pathologist is to accurately
and lactation, with evidence of reversal after lactation is subtype pituitary lesions to guide the management of the
terminated [2]. Animal studies have shown that somatotrophs individual patient [9]. As will become evident, there are
can reversibly transform into thyrotrophs in hypothyroidism structural features that correlate with specific mutations but
[3]. The third lineage, SF1-directed gonadotrophs, produces morphologic classification along with clinical and radiologic
both gonadotropins, follicle-stimulating hormone (FSH) and data remains the best predictor of patient prognosis and can
luteinizing hormone (LH). An interesting complexity is that predict therapeutic responsiveness [1].
TSH, FSH, and LH are all dimeric glycoprotein hormones In this review, we will summarize the molecular altera-
that have a common alpha-subunit and distinct beta-subunits tions that predispose to the development of pituitary tumors,
that confer specificity; their various cell types also rely on the sporadic genetic events that have been described, and
expression of GATA2/3 in addition to PIT1 for thyrotrophs conclude with a discussion of the epigenetic changes that
and SF1 for gonadotrophs. seem to be more critical [10] and may represent features
Tumors of these adenohypophyseal neuroendocrine that can be modulated to prevent the development of these
cells, now classified as pituitary neuroendocrine tumors increasingly common neoplasms.
(PitNETs) [4], recapitulate the normal cell types, but in
addition, they may develop distinct morphologic subtypes
[1]; these include sparsely and densely granulated variants Genetics of Inherited Pituitary Tumors
of corticotrophs, somatotrophs, and lactotrophs, as well as
tumors that are less terminally differentiated, such as the A small proportion of PitNETs occur in patients who have
so-called “poorly differentiated PIT1-lineage” and “acidophil germline genetic alterations that predispose them to PitNET
stem cell” tumors. Rare pituitary tumors have features of development (Table 1). In general, these patients present at
neuroendocrine cells but lack any lineage determination a younger age than patients with sporadic tumors [11–15].
and are classified as “null cell” tumors. PitNETs show The first example, multiple endocrine neoplasia type 1
a spectrum of clinical behaviors that vary from indolent (MEN1), was originally described by Wermer in 1954 [16].
prolactinomas with exquisite responsiveness to medical The gene responsible for this autosomal dominant disorder
therapy to aggressive infiltrative tumors that invade brain with incomplete penetrance, MEN1 at 11q13, encodes
and surrounding sinuses and persistent hormone excess with the 610 amino acid protein menin, a scaffold protein that
life-threatening complications. Very rarely they metastasize. regulates gene transcription through interactions with
In the posterior lobe, tumors derived from pituicytes, multiple partners and regulates a number of signaling
modified glia, are known as pituicytomas; they have pathways. Mutations that inactivate menin must be
several variants that reflect the normal spectrum of associated with loss of heterozygosity (LOH) of the normal
pituicytes, including oncocytic tumors known as spindle allele to give rise to neoplasia. Interestingly, menin is
cell oncocytoma, granular cell tumors, and ependymal ubiquitously expressed but tissues that most commonly
variants [5]. Tumors of the hypothalamic neurons that develop tumors are the pituitary, parathyroids, and endocrine
form the hormone-containing posterior pituitary include pancreas. Patients with MEN1 are also more susceptible to
gangliocytomas and neurocytomas, recapitulating the the development of adrenal cortical lesions [17] and breast
neurons of the magnocellular and parvocellular nuclei of the cancer [18]. Approximately two-thirds of mutation carriers
hypothalamus [6]. These tumors all express TTF1 that is a develop a PitNET. Most kindreds have had a predominance
feature of the basal hypothalamus; the neurons also produce of tumors of PIT1 lineage, including typical prolactinomas,
vasopressin and oxytocin. tumors that produce growth hormone (GH) or GH and
Tumors of Rathke’s cleft that is the developmental PRL [11, 19–21] and poorly differentiated tumors of PIT1
pr imordium of t he adenohypophysis include lineage [22], but examples of almost any adenohypophyseal
craniopharyngiomas and pituitary blastomas [1]. tumor type have been reported. Some reports have identified
The addition of molecular diagnostics has had multiple and multicentric neoplasms in the pituitary [21, 23].
significant impact on the practice of surgical pathology. A rare syndrome that mimics MEN1 clinically can
In some areas, for example some parts of neuropathology result from defects in CDKI genes. This so-called MEN4
and hematopathology, some tumor diagnoses cannot syndrome has been reported in patients with mutations
be confirmed without a molecular test. However, in in the CDKN1B/p27Kip1 gene [24,  25] or the CDKN2C/
the classification of pituitary neoplasia, this is not the p18INK4c gene [26, 27]. In addition to PitNETs, parathyroid,
case. There are indeed specific mutations that have and pancreatic endocrine proliferations, these mutations

13
Endocrine Pathology (2021) 32:3–16 5

Table 1  Genetic susceptibility in pituitary neuroendocrine tumors


Syndrome/condition Gene(s) PitNETs Other NETs Other non-neuroendocrine
lesions

MEN1 syndrome MEN1 PIT1-lineage Parathyroid NET Adrenal cortical neoplasms


most common Pancreatic NET Breast carcinoma
All types reported Duodenal NET
Pulmonary NET
Thymic NET
MEN4 syndrome CDKN1B Parathyroid NET Lymphoma
Pancreatic NET Breast carcinoma
Small Intestinal NET
Carney complex PRKRIα Densely granulated Pigmented nodular adrenocor-
Other (unknown) somatotroph tumors tical disease
Mammosomato- Testicular tumors
troph tumors Mucosal nevi
Cardiac and other myxomas
Malignant melanotic nerve
sheath tumors
Familial isolated pituitary AIP Sparsely granu- Pancreatic NET Scant morphologic data
adenoma (isolated familial Other (unknown) lated somatotroph Duodenal (ampullary type)
somatotropinoma) tumor most common NET
All types reported
SDHx-related familial paragan- SDHx  Acidophil stem cell Pheochromocytomas and para- Gastrointestinal stromal tumors
glioma syndromes tumors gangliomas Renal cell carcinoma
Pancreatic ­NETa
Hyperparathyroidism jaw tumor CDC73 Somatotropha Parathyroid NETs Ossifying fibromas of bone
syndrome Renal cell carcinoma
Uterine tumors
DICER syndrome DICER1 Pituitary blastoma  Pleuropulmonary blas-
toma; thyroid follicular
neoplasms;Sertoli-Leydig
cell tumors; sarcomas
Lynch syndrome (HNPCC) MLH1 Multiple types Pancreatic NET Colorectal cancer
MSH2, Endometrial cancer
MSH6 Gastric cancer
PMS2 Biliary and pancreatic cancer
Urinary tract cancers
X-linked acrogigantism Xq26 Densely granulated
microduplication somatotroph tumors
GPR101 Mammosomato-
troph tumors
McCune-Albright GNAS Densely granulated Adrenal and thyroid adenomas
somatotroph tumors Polyostotic fibrous dysplasia
Mammosomato- Cafe-au-lait skin pigmentation
troph tumors Precocious puberty
a
 Single case reported

predispose to small intestinal neuroendocrine tumors, testicular tumors, spotty pigmentation due to nevi involving
lymphoma, and breast cancer [28]. Similarly, MEN1-like mucosal surfaces including the lips, cardiac and other
acromegaly and hyperparathyroidism were associated in a myxomas, and malignant melanotic nerve sheath tumors [30].
patient with a germline mutation of CDC73 [29]. The isolated familial somatotropinoma (IFS) syndrome
Carney’s complex (CNC) [30] is an autosomal dominant [33], now known as familial isolated pituitary adenoma
disorder in which about 50% of patients have germline (FIPA) syndrome [34] since other PitNETs also occur, is
mutations in the PRKAR1Aα gene that encodes the protein a familial disorder attributed in about 50% of families to
kinase A regulatory subunit 1Aα. This results in unrestrained germline mutations in the AIP gene that encodes the aryl
cAMP signaling [31,  32] (Fig.  1) that manifests in the hydrocarbon-interacting protein (AIP); this chaperone
pituitary with GH-secreting hyperplasia and neoplasia. Other protein modulates expression of the aryl hydrocarbon
features include pigmented nodular adrenocortical disease, receptor (Fig. 1), which mediates the carcinogenic effects

13
6 Endocrine Pathology (2021) 32:3–16

Fig. 1  Molecular pathogenesis of pituitary somatotroph tumors. activation. Tumors with these alterations have a densely granulated
Sporadic or germline activating mutations in GNAS result in constitu- phenotype and respond well to somatostatin analog therapy that
tive activation of the Gsα that upregulates adenylyl cyclase resulting reduces cAMP. In contrast, changes shown on the right including
in increased cAMP, causing phosphorylation of CREB and synthesis AIP mutation or silencing, GHR inactivation, or other mechanisms of
of GH as well as αSU. In patients with Carney Complex mutations reducing STAT signaling, result in sparsely granulated somatotroph
inactivating PRKR1A have the same effect by enhancing CREB tumors that have fibrous bodies (modified from (7))

of dioxins [35, 36] thereby acting as a tumor suppressor [43]. These tumors occur almost exclusively in patients with
[15, 34, 37]. The commonest tumor type is the sparsely germline mutations of the DICER1 gene [44, 45] and may
granulated somatotroph tumor, and evidence suggests that be associated with other phenomena of that syndrome that is
germline AIP mutations are identified more frequently in also known as “pleuropulmonary blastoma-familial tumor and
pediatric and young adults (<  30  years) with sparsely dysplasia syndrome” or simply DICER1 syndrome.
granulated somatotroph tumors [12].
Rare PitNETs have been described in patients with
mutations in the genes encoding the various components Genetic Alterations in Sporadic Pituitary
of the succinate dehydrogenase (SDH) complex; these Tumors
mutations primarily cause familial pheochromocytomas and
paragangliomas, gastrointestinal stromal tumors (GISTs), The majority of PitNETs are sporadic tumors; the genes
and renal carcinomas [38, 39]. Another familial syndrome implicated in familial syndromes discussed above are not
associated with occasional PitNETs is Lynch syndrome due commonly altered in these sporadic neoplasms. While
to mutation in one of the mismatch repair enzymes, MLH1, chromosomal alterations seem to vary based on tumor
MSH2, MSH 6, or PMS2, that is usually associated with functionality in PitNETs [46], there are specific mutations
colorectal carcinomas, ovarian and endometrial carcinomas, that have been identified in subsets of the more common
as well as several other tumor types [40]. tumors. The molecular alterations are summarized in Table 2.
Inherited or sporadic Xq26 microduplications and The molecular features of the unusual plurihormonal tumors
GPR101 mutation are the cause of a rare form of early and multiple PitNETs are largely unknown given their rarity.
childhood X-linked acrogigantism (X-LAG) [41]; patients A subset of somatotroph tumors has activating mutations
with this disorder have mammosomatotroph or somatotroph of GNAS. This gene is associated with several other endocrine
hyperplasia and/or mammosomatotroph PitNET [42]. tumors including functioning thyroid adenomas, adrenal
Pituitary blastomas are rare tumors that are composed cortical adenomas, and gonadal tumors. The protein product of
of adenohypophyseal neuroendocrine cells admixed with this gene, Gsα, mediates signaling from seven transmembrane
primitive Rathke’s pouch epithelium and folliculostellate cells domain G-protein coupled receptors (including the receptors

13
Endocrine Pathology (2021) 32:3–16 7

Table 2  Common sporadic Gene(s) PitNETs Clinical correlates


genetic alterations in pituitary
tumors GNAS Densely granulated somatotroph/mammosoma- Florid acromegaly
totroph tumors T2 hypointensity on MRI
Somatostatin responsiveness
USP8 Densely granulated corticotroph Florid Cushing disease
USP48 Pasireotide responsiveness
ATRX Corticotroph tumors Aggressive, recurrent, metastatic
CTNNB1 Adamantinomatous craniopharyngioma
BRAFV600E Papillary craniopharyngioma Targeted therapy

for GHRH, TSH, ACTH, and MSH) to activate cyclic and STAT-3 upregulation has been shown to correlate
AMP (Fig. 1). Familial transmission of this mutation is not with GH expression in somatotroph tumors [56] (Fig. 1),
recognized since germline mutation is embryonic lethal; supporting the hypothesis that an altered GHR signaling
however, sporadic mosaicism gives rise to McCune-Albright pathway may play a role in the pathogenesis of sparsely
syndrome, which manifests these various endocrinopathies granulated somatotroph tumors and their fibrous bodies.
as well as fibrous dysplasia of bone and cafe-au-lait spots Dopamine is the major regulator of prolactin synthesis
of the skin. In isolated sporadic somatotroph tumors, this and secretion through tonic inhibition. While most lactotroph
mutation is associated with smaller tumors that give rise to tumors respond to dopamine, it has been suggested that loss
florid acromegaly and high GH and IGF1 levels and predicts of activity of the type 2 dopamine receptor, D2R, may be
responsiveness to somatostatin analogs. Several studies have the cause of dopamine resistance in some tumors. However,
shown that this mutation is most often seen in tumors with a mutations in the D2R gene have not been identified. D2R
densely granulated somatotroph phenotype [47, 48] but not expression is dependent on nerve growth factor (NGF)
all densely granulated tumors harbor GNAS mutations and the binding to the NGF receptor p75, and some authors have
correlation is not perfect [49, 50]; nevertheless, histopathology suggested that NGF alterations may explain dopamine
remains the best predictor of therapeutic responsiveness resistance [57]. A recent study of sporadic lactotroph tumors
[51]. For this reason, molecular testing is not recommended identified large-scale somatic copy number alterations,
as a routine analysis. Transcriptome profiling of pituitary especially gains, as well as sequence variants in 15 genes
neuroendocrine tumors with GH and PRL co-expression including DRD2, PRL, TMEM67, and MLH3 that are possibly
segregated along with somatotroph tumors in a recent study involved in tumorigenesis [58]. In a study of 38 high-risk
[46]; this may be at least partially attributable to the frequency histological subtypes of PitNETs, homozygous CDKN2A
of GNAS-related alterations in mammosomatotroph tumors deletion using FISH was reported in densely granulated
which clinically and morphologically resemble densely lactotroph tumors that also exhibited increased proliferative
granulated somatotroph tumors but also express ER and PRL. activity [59]; this finding suggests a possible utility of p16
An unusual germline SSTR5 inactivating mutation immunohistochemistry in this group of PitNETs.
was reported in a patient with acromegaly [52]. Growth Thyroid hormone receptors (TRs) are important
hormone autoregulation may also be implicated in regulators of thyrotroph function and proliferation. TRα and
somatotroph pathophysiology. Mouse studies have shown TRβ, which each undergo alternative splicing to generate
that loss of GH signaling through either deletion of the 2 isoforms, are ubiquitously expressed [60]; however, the
GHR or expression of a dominant negative GH results β2 isoform is of particular importance in the pituitary. The
in somatotroph hypertrophy and hyperplasia [53]. In β-isoforms are underexpressed in clinically silent tumors,
human tumors, treatment with a dominant negative GH, but the significance of this may be more related to the fact
pegvisomant, in vitro caused the formation of fibrous bodies, that most of these tumors are composed of gonadotrophs
and a somatic mutation in the extracellular domain of the [60]. Novel missense mutations have been reported in some
GH receptor (GHR) that impairs receptor glycosylation, thyrotroph tumors and occasional clinically silent tumors
reducing GH-ligand binding, was identified in sparsely [61, 62].
granulated somatotroph tumors that feature this keratin Some corticotroph tumors, especially those with
aggresome [54]. This suggests that impaired hormone auto- biochemical functionality, have mutations of USP8, a gene
feedback may be implicated in the pathogenesis of these encoding a member of the ubiquitin protease family that
tumors and subcellular structures [54]. This mutation has not is thought to alter EGFR signaling but more recently has
been identified in other studies but the inhibitory effect of been suggested to affect POMC and ACTH degradation
a GH agonist confirmed GH autoregulation in somatotroph [63]. This mutation has been reported mainly in small
tumors that were not characterized for tumor subtype [55] densely granulated corticotroph tumors [64–67] and is

13
8 Endocrine Pathology (2021) 32:3–16

thought to be predictive of pasireotide response [66] but and mutation of TP53 has been reported in only a very few
has not yet reached clinical relevance. Pathogenic germline documented cases [80;83–87]. While there may be some
USP8 variants have also been recently defined in pediatric prognostic value of p53 staining [80], this is controversial
Cushing disease [68]. Somatic USP48 [69] and BRAFV600E and there is no consistent correlation between p53 indices
mutations [69] have also been identified in a few USP8- and tumor invasion or recurrence [88].
wild-type corticotroph tumors. BRAF mutations were shown Craniopharyngiomas represent the one tumor type
to enhance POMC transcription in functional studies [69]. with a clear-cut molecular basis [89]. These tumors have
ATRX mutations with LOH and loss of immunoreactivity two distinct morphologic variants. Adamantinomatous
have been reported in aggressive and metastatic corticotroph craniopharyngiomas resemble the dental ameloblastic
tumors [70-71]. PTEN has also been suggested to play a organ and adamantinoma; the majority of these lesions
role in metastatic spread of these tumors [71]. While this harbor mutations of the CTNNB1 gene that encodes beta-
clarifies the basis of these more aggressive tumors, there is catenin [89–92] resulting in aberrant nuclear localization
no predictive value for therapeutic guidance. of immunohistochemical reactivity for β-catenin (Fig. 2a);
Germline mutation of the GCR​ gene encoding the however, nuclear staining tends to be only focal, most
glucocorticoid receptor was identified in a small number of often found in morular structures. In contrast, papillary
patients with pituitary Cushing’s disease [72] and somatic craniopharyngiomas that are less common and rare in children
mutations in GCR​were found in some sporadic tumors [73], are composed of more regular squamous epithelium that
suggesting that diminished glucocorticoid inhibition may harbors BRAFV600E mutations in nearly all cases [89, 93], a
play a role in the development of some of these tumors. mutation that can be detected using the VE1 antibody (Fig. 2b)
Apart from these small subsets of tumors, the majority of [94]. The identification of BRAFV600E mutation has paved the
sporadic PitNETs lack recurrent mutations, and molecular way for medical therapy with targeted BRAF inhibition for
studies have shown only variable genomic instability with recurrent tumors that cannot be resected surgically [95–97].
copy number variations [58, 74–76]. There is currently no evidence of germline susceptibility
Mutations in codon 12 of H-RAS have been reported in to the development of pituicyte-derived posterior lobe tumors
only a few PitNETs including an aggressive prolactinoma including conventional pituicytomas, spindle cell oncocytomas,
and a few rare metastatic tumors [77, 78]. granular cell tumors, and sellar ependymomas. Although
While RB-deficient mice develop pituitary tumors of a single case of pituicytoma has been linked to pathogenic
the corticotroph lineage, mutations in this gene are not BRAFV600E, NF1, and TSC1 variants [98], these tumors usually
implicated in human tumors. A single report suggested lack BRAF and IDH1 mutations that are more frequent in
that loss of RB expression may play a role in metastatic low-grade astrocytomas [5]. Somatic copy number alterations
progression [79]. The role of the TP53 oncogene is also involving 1p, 5p, 14q, and 22q have been identified in a
unclear; p53 immunoreactivity was initially reported pituicytoma [99]. The limited data on molecular alterations
in carcinomas [80-81], but this is also the case in non- suggested somatic alterations leading to the activation of the
metastatic PitNETs where it became a standard feature to MAPK pathway (e.g., HRAS, FAT1, SND1) in conventional
report [82]. This is now known to be wild-type expression and oncocytic pituicytomas [98, 100].

Fig. 2  Molecular pathogenesis of craniopharyngioma. Adamanti- tein on immunohistochemistry. Papillary craniopharyngiomas (right)


nomatous craniopharyngioma (left) is characterized by mutations of harbor BRAFV600E mutations that can be identified with the VE1
β-catenin that can be identified by nuclear translocation of the pro- antibody

13
Endocrine Pathology (2021) 32:3–16 9

Epigenetic Alterations Underlying Pituitary a dominant-negative EGF-R [104]. All four receptors
Neoplasia are expressed by human PitNETs and are upregulated in
invasive and recurrent tumors [105] with the exception of
Epigenetic dysregulation is more frequent than structural ErbB4 [106], and despite the fact that there is no evidence of
genetic alterations in sporadic pituitary neoplasms (Fig. 3) amplification or mutation of EGF-R family members in these
[10]. neoplasms, pharmacotherapeutic targeting of ErbB2 has been
proposed for treatment [107]. Indeed, the only rationale for
Dysregulated Growth Factor Signaling this would be to target the putative augmented signaling of
EGF-R in corticotroph tumors with USP8 mutations.
A number of growth factors play important roles in Fibroblast growth factors (FGFs) and their receptors
pituitary cell growth and hormone secretion, and there (FGFRs) regulate development, growth, and angiogenesis
is evidence of altered expression of these growth factors in many organ systems. There are at least 23 FGFs. The
and/or their receptors in PitNETs. Transforming growth original FGFs include acidic FGF1 and basic FGF2; the
factor-α (TGF-α) is expressed in PitNETs as well as in latter was originally isolated from the bovine pituitary and
normal adenohypophyseal cells [101]; it may mediate the was subsequently shown to be produced by folliculo-stellate
proliferative effects of estrogen [102] and causes tumor cells [108, 109]. FGFs are critical for pituitary development
development in mice [103]. Epidermal growth factor (EGF) [110–112] and blockade of FGF signaling using a dominant
is expressed in adenohypophyseal cells and all types of negative receptor in mice abrogates pituitary formation
functional and nonfunctional PitNETs. EGF stimulates [113]. FGF2 is expressed by human and rodent PitNETs
PRL and ACTH synthesis and secretion. The EGF-receptor [114-116] and elevated circulating FGFs are found in
(EGF-R) family that mediates signaling of TFG-α and EGF patients with PitNETs [114,  117]. FGFs signal through
includes four members, EGF-R (ErbB1, HER1), p185 four receptors, FGFR1 through FGFR4, each of which has
Her2/neu (ErbB2 or HER2), ErbB3 (HER3), and ErbB4 multiple isoforms. Mutation in FGFR1 has been associated
(HER4). EGF-R appears to be critical for somatotroph with combined pituitary hormone deficiency [118] and two
development as shown by transgenic mice overexpressing FGFRs have been shown to be altered pituitary tumors [119].

Fig. 3  Epigenetic alterations in pituitary neuroendocrine tumors. or PTAG, and dysregulate growth factors, releasing cells into
Nuclear transcription is regulated by menin, DNMTs, Ikaros, and proliferation. Changes in miRNAs also affect expression levels
HMGAs. Alterations in these proteins, by mutation and LOH of of important cell cycle regulators, growth factors, and adhesion
menin in MEN1 or by dysregulation in the other factors in sporadic molecules that restrain growth and enhance invasion, which is also
tumors, change chromatin accessibility to decrease expression of enhanced by a polymorphism in the MMP-1 promoter
tumor suppressors such as CDKIs, Rb, p53, GADD45γ, MEG3,

13
10 Endocrine Pathology (2021) 32:3–16

FGFR2-IIIb is a receptor isoform that is expressed in double knockout mice [145]. No mutations of these genes
the normal pituitary but is subject to promoter methylation have been reported in sporadic tumors but protein levels
and down-regulation in pituitary tumors [119]. FGFR2-IIIb of p27 are reduced in functioning corticotroph tumors
inhibits pituitary tumor cell proliferation [120] and silences [146,  147]; interestingly, this is not the case in silent
expression of the cancer/testis-melanoma-associated corticotroph tumors [128], suggesting that glucocorticoid
antigens (MAGE-A) [121]. In contrast to the expression excess mediates protein stability [148]. Expression of
of FGFRs 1, 2, and 3 in the normal gland, FGFR4 is not ­p16ink4A is also silenced by promoter methylation [149, 150].
normally expressed but is identified in PitNETs [119]. Similarly, GADD45γ (growth arrest and DNA damage-
Epigenetic changes causing alternative transcription inducible 45γ) and MEG3 (a human homolog of the mouse
initiation [122, 123] result in expression of a truncated ptd- maternally imprinted Gtl2 gene) are downregulated in
FGFR4 that displaces N-cadherin from the cell membrane, PitNETs by CpG island promoter methylation [151–153].
disrupting cell adhesion [124,  125] and expression of Another gene involved in the DNA damage repair response,
FGFR4 is associated with aggressive behavior [126–128]. CHEK2, is upregulated in corticotroph tumors and
This mechanism of altering cell adhesion is also suggested in suppressed in somatotroph tumors [154]. Pituitary tumor
reports that PSA-NCAM expression enhances tumor growth apoptosis gene (PTAG), on chromosome 22, was identified
and invasiveness [129], since PSA-NCAM would also as a methylation-sensitive anti-apoptotic gene expressed in
disrupt the tri-protein FGFR4.N-cadherin-NCAM complex. PitNETs [155]..
The FGFR4 gene has a polymorphism, FGFR4-G488R,
which alters signaling of this receptor; the FGFR4-R388 Epigenetic Regulators Implicated in Pituitary
isoform has been associated with more aggressive behavior Tumors
in a number of epithelial malignancies. In the pituitary, it
enhances cell growth and impairs normal hormone regulation The striking epigenetic changes resulting in silencing of tumor
in corticotrophs and somatotrophs [130, 131], providing a suppressors in pituitary tumors have led to studies of epigenetic
possible genetic basis for the development of larger tumors regulators in these tumors. Indeed, menin, the protein product
with less hormonal activity, that is, sparsely granulated of the MEN1 gene, is itself a nuclear scaffold protein that
variants of corticotroph and somatotroph tumors. It may also regulates gene transcription by chromatin remodeling.
modulate response to somatostatin analog therapy [132]. Other factors that regulate gene expression include DNA
Another polymorphism that has been implicated in methyltransferase (DNMT) enzymes [10]. Studies have shown
invasiveness of PitNETs is in the MMP-1 promoter where that DNMT1 and DNMT3a are not altered but DNMT3b is
insertion of a guanine residue increases the risk of tumor overexpressed in pituitary tumor cells, and it is implicated in
invasion [133]. the silencing of pRB1, p21, and p27 [10, 156].
In addition to DNA methylation, chromatin remodeling
Dysregulated Tumor Suppressors is mediated, in part, through post-translational histone
modifications that control access of DNA to transcription
Epigenetic silencing of several tumor suppressor genes has factors. One of the factors that is involved in chromatin
been documented in PitNETs [8-10]. remodeling in the pituitary is Ikaros, originally described
The retinoblastoma (RB1) tumor suppressor gene on in lymphoid cells [157]. Ikaros in both the wild-type
chromosome 13q has long been a target of investigation, and dominant-negative (dn) isoforms are expressed in
since RB1-deficient mice develop pituitary intermediate hypothalamic and pituitary neuroendocrine cells. Ikaros is
lobe tumors [134]. RB1 mutations, including in the promoter required for normal development of the pituitary TPIT lineage
region, are not identified in human PitNETs [135–137], but [158] as well as hypothalamic GHRH neurons that are required
instead, RB1 promoter methylation causing gene silencing for pituitary somatotroph growth and function [159]. It also
has been described [136] as well as LOH at 13q in tumors may regulate PIT1-lineage cell functional differentiation by
with unmethylated RB1 [138]. The ARF tumor suppressor modulating GH and PRL hormone gene expression [160]. A
may modulate RB1-mediated tumorigenesis [139], and the dominant negative isoform of Ikaros is expressed in pituitary
inhibitor of differentiation Id2 also interacts with RB1 [140] tumors [123] where it activates the Bcl-XL promoter [161],
and vascular endothelial growth factor (VEGF) [140] but no enhancing pituitary cell survival and evasion of apoptosis.
mutations have been described in these genes in PitNETs. Ikaros also can regulate cholesterol uptake through the low-
Cyclins promote mitosis through cyclin-dependent density lipoprotein receptor (LDL-R) and sterol-regulatory
kinases (CDKs) that are inhibited by CDK inhibitors element binding protein 2 (SREBP2) to provide lipid required
(CDKIs) [141]. Knockout of cyclins and CDKIs in mice for synthesis of secretory granule membranes [162].
consistently result in pituitary tumors; these include Chromatin remodeling is also regulated by the high
­p27kip1–null mice [142–144], ­p18ink4c–null mice [145] and mobility group (HMG) proteins containing AT-hook

13
Endocrine Pathology (2021) 32:3–16 11

domains (HMGA). Mice overexpressing HMGA1 or Epigenetic changes are implicated in the pathogenesis of
HMGA2 develop pituitary tumors of the PIT1 lineage the more common sporadic pituitary neoplasms including
[163, 164], thought to be through silencing of pRB and the majority of PitNETs and tumors of pituicytes.
induction of E2F1 [165] and upregulation of cyclin B2[166].
HMGA1 [166-167] and HMGA2 [168] have been found
to be amplified and overexpressed in a small number of Funding  This research did not receive any specific grant from funding
agencies in the public, commercial, or not-for-profit sectors.
prolactinomas.
Non-coding RNAs are implicated in the epigenetics of
neoplasia. MicroRNAs (miRs) are endogenous small RNAs
Compliance with Ethical Standards 
that play important roles in the regulation of expression of Conflict of Interest  The authors declare that they have no conflict of
genes that control cell proliferation, differentiation, and interest.
apoptosis. For example, the altered expression of HMGA2
in primary tumors without amplification of this gene may
be due to altered microRNA expression, particularly of
let-7 [169]. An increasing number of aberrantly expressed References
miRNAs is currently being discovered in human pituitary
tumor studies [10]. 1. Asa SL, Perry A. Tumors of the Pituitary Gland. Atlas of Tumor
Gene expression can also modulate the invasive growth nd Nontumor Pathology, Series 5, Fascicle 1. Arlington VA: ARP
Press, 2020.
in PitNETs [170]. PITX2, a homeobox gene product, is 2. Asa SL, Penz G, Kovacs K, Ezrin C. Prolactin cells in the human
significantly overexpressed in non-functional PitNETs with pituitary. A quantitative immunocytochemical analysis. Arch
cavernous sinus invasion [171]. Similarly, protein expression Pathol Lab Med 1982; 106:360–363.
studies on invasiveness of silent corticotroph tumors 3. Horvath E, Lloyd RV, Kovacs K. Propylthiouracyl-induced
hypothyroidism results in reversible transdifferentiation of
identified different patterns than in other non-functional somatotrophs into thyroidectomy cells. A morphologic study
PitNETs [172]. These findings underscore the importance of the rat pituitary including immunoelectron microscopy. Lab
of the regulation of cell-to-extracellular matrix interactions Invest 1990; 63:511–520.
that are likely related to epigenetic dysregulation. 4. Asa SL, Casar-Borota O, Chanson P et  al. From pituitary
adenoma to pituitary neuroendocrine tumor (PitNET): an
Epigenetic alterations including distinct miRNA profiling International Pituitary Pathology Club proposal. Endocr Relat
were noted in oncocytic pituicytomas with recurrent disease Cancer 2017; 24(4):C5-C8.
[173]. In addition, transcriptome profiling of oncocytic 5. Mete O, Lopes MB, Asa SL. Spindle cell oncocytomas and
pituicytomas underscored the role of cell cycle and its granular cell tumors of the pituitary are variants of pituicytoma.
Am J Surg Pathol 2013; 37(11):1694-1699.
regulators as well as metabolic alterations [173]. 6. Asa SL, Mete O. Hypothalamic Endocrine Tumors: An Update.
J Clin Med 2019; 8(10).
7. Asa SL, Ezzat S. The pathogenesis of pituitary tumors. Annu Rev
Conclusions Pathol 2009; 4:97-126.
8. Ezzat S, Asa SL. Mechanisms of disease: The pathogenesis
of pituitary tumors. Nat Clin Pract Endocrinol Metab 2006;
Molecular pathology has led to advances in the 2(4):220-230.
understanding of many tumors and offers opportunities 9. Gomez-Hernandez K, Ezzat S, Asa SL, Mete O. Clinical
to identify novel therapies. In the pituitary, the field has Implications of Accurate Subtyping of Pituitary Adenomas:
Perspectives from the Treating Physician. Turk Patoloji Derg
uncovered a number of genetic mutations that predispose 2015; 31 Suppl 1:4-17.
to PitNET development, including MEN1, CDKN1B, 10. Ezzat S, Cheng S, Asa SL. Epigenetics of pituitary tumors:
PRKRIα, AIP, and other rarer events, but these genes are Pathogenetic and therapeutic implications. Mol Cell Endocrinol
rarely mutated in sporadic tumors. Recurrent genetic 2018; 469:70-76.
11. Trouillas J, Labat-Moleur F, Sturm N et al. Pituitary tumors and
events in sporadic PitNETs include GNAS mutations in a hyperplasia in multiple endocrine neoplasia type 1 syndrome
subset of somatotroph tumors as well as USP8 and USP48 (MEN1): a case-control study in a series of 77 patients
mutations in some corticotroph tumors; to date, neither of versus 2509 non-MEN1 patients. Am J Surg Pathol 2008;
these has resulted in altered management, and instead, the 32(4):534-543.
12. Beckers A, Aaltonen LA, Daly AF, Karhu A. Familial
prognosis and management of PitNETs relies more on cell isolated pituitary adenomas (FIPA) and the pituitary adenoma
type and subtype as well as local growth that determines predisposition due to mutations in the aryl hydrocarbon receptor
surgical resectability. In contrast, craniopharyngiomas have interacting protein (AIP) gene. Endocr Rev 2013; 34(2):239-277.
either CTNNB1 or BRAFV600E mutations that correlate with 13. Daly AF, Tichomirowa MA, Petrossians P et  al. Clinical
characteristics and therapeutic responses in patients with
adamantinomatous or papillary morphology, respectively; germ-line AIP mutations and pituitary adenomas: an
the latter offers the opportunity for targeted therapy. DICER1 international collaborative study. J Clin Endocrinol Metab 2010;
mutations are found in patients with pituitary blastoma. 95(11):E373-E383.

13
12 Endocrine Pathology (2021) 32:3–16

14. Tahir A, Chahal HS, Korbonits M. Molecular genetics of the aip 34. Beckers A, Daly AF. The clinical, pathological, and genetic
gene in familial pituitary tumorigenesis. Prog Brain Res 2010; features of familial isolated pituitary adenomas. Eur J Endocrinol
182:229-253. 2007; 157(4):371-382.
15. Georgitsi M, De Menis E, Cannavo S et al. Aryl hydrocarbon 35. Pesatori AC, Baccarelli A, Consonni D et al. Aryl hydrocarbon
receptor interacting protein (AIP) gene mutation analysis in receptor-interacting protein and pituitary adenomas: a
children and adolescents with sporadic pituitary adenomas. Clin population-based study on subjects exposed to dioxin
Endocrinol (Oxf) 2008; 69(4):621-627. after the Seveso, Italy, accident. Eur J Endocrinol 2008;
16. 16. Wermer P. Genetic aspects of adenomatosis of endocrine 159(6):699-703.
glands. Am J Med 1954; 16:363-371. 36. Stiles CE, Korbonits M. Familial Isolated Pituitary Adenoma.
17. Thakker RV. Multiple endocrine neoplasia type 1 (MEN1) and 2000.
type 4 (MEN4). Mol Cell Endocrinol 2014; 386(1-2):2-15. 37. Vierimaa O, Georgitsi M, Lehtonen R et al. Pituitary adenoma
18. Brennan P. Breast cancer risk in MEN1 - a cancer genetics predisposition caused by germline mutations in the AIP gene.
perspective. Clin Endocrinol (Oxf) 2015; 82(3):327-229. Science 2006; 312(5777):1228-1230.
19. DeLellis RA. Multiple endocrine neoplasia syndromes revisited. 38. Gill AJ. Succinate dehydrogenase (SDH)-deficient neoplasia.
Clinical, morphologic and molecular features. Lab Invest 1995; Histopathology 2018; 72(1):106-116.
72:494–505. 39. Xekouki P, Stratakis CA. Succinate dehydrogenase (SDHx)
20. Ezzat S, Asa SL. The multiple endocrine neoplasia syndromes. mutations in pituitary tumors: could this be a new role for
In: Kovacs K, Asa SL, editors. Functional Endocrine Pathology. mitochondrial complex II and/or Krebs cycle defects? Endocr
Boston: Blackwell Science, 1998: 952-966. Relat Cancer 2012; 19(6):C33-C40.
21. Scheithauer BW, Laws ER, Jr., Kovacs K, Horvath E, Randall 40. Bengtsson D, Joost P, Aravidis C et al. Corticotroph Pituitary
RV, Carney JA. Pituitary adenomas of the multiple endocrine Carcinoma in a Patient With Lynch Syndrome (LS) and Pituitary
neoplasia type I syndrome. Semin Diagn Pathol 1987; Tumors in a Nationwide LS Cohort. J Clin Endocrinol Metab
4(3):205-211. 2017; 102(11):3928-3932.
22. Mete O, Gomez-Hernandez K, Kucharczyk W et  al. Silent 41. Trivellin G, Daly AF, Faucz FR et al. Gigantism and acromegaly
subtype 3 pituitary adenomas are not always silent and represent due to Xq26 microduplications and GPR101 mutation. N Engl J
poorly differentiated monomorphous plurihormonal Pit-1 lineage Med 2014; 371(25):2363-2374.
adenomas. Mod Pathol 2016; 29(2):131-142. 42. Moran A, Asa SL, Kovacs K et al. Gigantism due to pituitary
23. Shintani Y, Yoshimoto K, Horie H et al. Two different pituitary mammosomatotroph hyperplasia. N Engl J Med 1990;
adenomas in a patient with multiple endocrine neoplasia type 1 323:322-327.
associated with growth hormone-releasing hormone-producing 43. Scheithauer BW, Kovacs K, Horvath E et al. Pituitary blastoma.
pancreatic tumor: clinical and genetic features. Endocr J 1995; Acta Neuropathol 2008; 116(6):657-666.
42:331-340. 44. de Kock L, Sabbaghian N, Plourde F et al. Pituitary blastoma:
24. Pellegata NS, Quintanilla-Martinez L, Siggelkow H et al. Germ- a pathognomonic feature of germ-line DICER1 mutations. Acta
line mutations in p27Kip1 cause a multiple endocrine neoplasia Neuropathol 2014; 128(1):111-122.
syndrome in rats and humans. Proc Natl Acad Sci U S A 2006; 45. Sahakitrungruang T, Srichomthong C, Pornkunwilai S et al.
103(42):15558-15563. Germline and somatic DICER1 mutations in a pituitary blastoma
25. Georgitsi M, Raitila A, Karhu A et al. Germline CDKN1B/ causing infantile-onset Cushing’s disease. J Clin Endocrinol
p27Kip1 mutation in multiple endocrine neoplasia. J Clin Metab 2014; 99(8):E1487-E1492.
Endocrinol Metab 2007; 92(8):3321-3325. 46. Neou M, Villa C, Armignacco R et al. Pangenomic Classification
26. Agarwal SK, Mateo CM, Marx SJ. Rare germline mutations in of Pituitary Neuroendocrine Tumors. Cancer Cell 2020;
cyclin-dependent kinase inhibitor genes in multiple endocrine 37(1):123-134.
neoplasia type 1 and related states. J Clin Endocrinol Metab 47. Spada A, Arosio M, Bochicchio D et al. Clinical, biochemical and
2009; 94(5):1826-1834. morphological correlates in patients bearing growth hormone-
27. Georgitsi M. MEN-4 and other multiple endocrine neoplasias secreting pituitary tumors with or without constitutively active
due to cyclin-dependent kinase inhibitors (p27(Kip1) and adenylyl cyclase. J Clin Endocrinol Metab 1990; 71:1421-1426.
p18(INK4C)) mutations. Best Pract Res Clin Endocrinol Metab 48. Asa SL, Kucharczyk W, Ezzat S. Pituitary acromegaly: not one
2010; 24(3):425-437. disease. Endocr Relat Cancer 2017; 24(3):C1-C4.
28. Alrezk R, Hannah-Shmouni F, Stratakis CA. MEN4 and 49. Bakhtiar Y, Hirano H, Arita K et  al. Relationship between
CDKN1B mutations: the latest of the MEN syndromes. Endocr cytokeratin staining patterns and clinico-pathological features
Relat Cancer 2017; 24(10):T195-T208. in somatotropinomae. Eur J Endocrinol 2010; 163(4):531-539.
29. Nachtigall LB, Guarda FJ, Lines KE et  al. Clinical MEN-1 50. Larkin S, Reddy R, Karavitaki N, Cudlip S, Wass J, Ansorge O.
Among a Large Cohort of Patients With Acromegaly. J Clin Granulation pattern, but not GSP or GHR mutation, is associated
Endocrinol Metab 2020; 105(6). with clinical characteristics in somatostatin-naive patients with
30. Carney JA, Gordon H, Carpenter PC, Shenoy BV, Go VL. The somatotroph adenomas. Eur J Endocrinol 2013; 168(4):491-499.
complex of myxomas, spotty pigmentation, and endocrine 51. Ezzat S, Caspar-Bell GM, Chik CL et al. Predictive markers for
overactivity. Medicine (Baltimore) 1985; 64(4):270-283. postsurgical medical management of acromegaly: a systematic
31. Kirschner LS, Carney JA, Pack SD et al. Mutations of the gene review and consensus treatment guideline. Endocr Pract 2019;
encoding the protein kinase A type I-alpha regulatory subunit in 25(4):379-393.
patients with the Carney complex. Nat Genet 2000; 26(1):89-92. 52. Ballare E, Persani L, Lania AG et al. Mutation of somatostatin
32. Yin Z, Williams-Simons L, Parlow AF, Asa S, Kirschner receptor type 5 in an acromegalic patient resistant to
LS. Pituitary-specific knockout of the Carney complex gene somatostatin analog treatment. J Clin Endocrinol Metab 2001;
prkar1a leads to pituitary tumorigenesis. Mol Endocrinol 2008; 86(8):3809-3814.
22(2):380-387. 53. Asa SL, Coschigano KT, Bellush L, Kopchick JJ, Ezzat S.
33. Soares BS, Frohman LA. Isolated familial somatotropinoma. Evidence for growth hormone (GH) autoregulation in pituitary
Pituitary 2004; 7(2):95-101. somatotrophs in GH antagonist-transgenic mice and GH
receptor-deficient mice. Am J Pathol 2000; 156(3):1009-1015.

13
Endocrine Pathology (2021) 32:3–16 13

54. Asa SL, DiGiovanni R, Jiang J et al. A growth hormone receptor 73. Karl M, von Wichert G, Kempter E et al. Nelson’s syndrome
mutation impairs growth hormone autofeedback signaling in associated with a somatic frame shift mutation in the
pituitary tumors. Cancer Res 2007; 67(15):7505-7511. glucocorticoid receptor gene. J Clin Endocrinol Metab 1996;
55. Cuny T, Zeiller C, Bidlingmaier M et al. In vitro impact of 81:124-129.
pegvisomant on growth hormone-secreting pituitary adenoma 74. Newey PJ, Nesbit MA, Rimmer AJ et  al. Whole-exome
cells. Endocr Relat Cancer 2016; 23(7):509-519. sequencing studies of nonfunctioning pituitary adenomas. J Clin
56. Zhou C, Jiao Y, Wang R, Ren SG, Wawrowsky K, Melmed Endocrinol Metab 2013; 98(4):E796-E800.
S. STAT3 upregulation in pituitary somatotroph adenomas 75. Bi WL, Horowitz P, Greenwald NF et al. Landscape of Genomic
induces growth hormone hypersecretion. J Clin Invest 2015; Alterations in Pituitary Adenomas. Clin Cancer Res 2017;
125(4):1692-1702. 23(7):1841-1851.
57. Fiorentini C, Guerra N, Facchetti M et al. Nerve growth factor 76. Hage M, Viengchareun S, Brunet E et al. Genomic Alterations
regulates dopamine D(2) receptor expression in prolactinoma and Complex Subclonal Architecture in Sporadic GH-Secreting
cell lines via p75(NGFR)-mediated activation of nuclear factor- Pituitary Adenomas. J Clin Endocrinol Metab 2018;
kappaB. Mol Endocrinol 2002; 16(2):353-366. 103(5):1929-1939.
58. De Sousa SMC, Wang PPS, Santoreneos S et al. The Genomic 77. Karga HJ, Alexander JM, Hedley-Whyte ET, Klibanski A,
Landscape of Sporadic Prolactinomas. Endocr Pathol 2019; Jameson JL. Ras mutations in human pituitary tumors. J Clin
30(4):318-328. Endocrinol Metab 1992; 74:914-919.
59. Kara M, Tokat F, Pamir MN, Danyeli AE. Frequency and Role 78. Cai WY, Alexander JM, Hedley-Whyte ET et al. Ras mutations
of CDKN2A Deletion in High-Risk Pituitary Neuroendocrine in human prolactinomas and pituitary carcinomas. J Clin
Tumors. Endocr Pathol 2020; 31(2):166-173. Endocrinol Metab 1994; 78:89-93.
60. Gittoes NJL, McCabe CJ, Verhaeg J, Sheppard MC, Franklyn 79. Hinton DR, Hahn JA, Weiss MH, Couldwell WT. Loss of Rb
JA. Thyroid hormone and estrogen receptor expression in normal expression in an ACTH-secreting pituitary carcinoma. Cancer
pituitary and nonfunctioning tumors of the anterior pituitary. J Lett 1998; 126(2):209-214.
Clin Endocrinol Metab 1997; 82:1960-1967. 80. Thapar K, Scheithauer BW, Kovacs K, Pernicone PJ, Laws
61. Ando S, Sarlis NJ, Krishnan J et al. Aberrant alternative splicing ER, Jr. p53 expression in pituitary adenomas and carcinomas:
of thyroid hormone receptor in a TSH- secreting pituitary tumor Correlation with invasiveness and tumor growth fractions.
is a mechanism for hormone resistance. Mol Endocrinol 2001; Neurosurgery 1996; 38:765-771.
15(9):1529-1538. 81. Pernicone PJ, Scheithauer B, Sebo TJ et al. Pituitary carcinoma:A
62. McCabe CJ, Gittoes NJ, Sheppard MC, Franklyn JA. Thyroid clinicopathologic study of 15 cases. Cancer 1997; 79:804-812.
receptor alpha1 and alpha2 mutations in nonfunctioning pituitary 82. DeLellis RA, Lloyd RV, Heitz PU, Eng C. Pathology and
tumors. J Clin Endocrinol Metab 1999; 84(2):649-653. Genetics of Tumours of Endocrine Organs. Lyons, France: IARC
63. Sesta A, Cassarino MF, Terreni M et  al. Ubiquitin-Specific Press, 2004.
Protease 8 Mutant Corticotrope Adenomas Present Unique 83. Tanizaki Y, Jin L, Scheithauer BW, Kovacs K, Roncaroli F, Lloyd
Secretory and Molecular Features and Shed Light on the Role of RV. P53 gene mutations in pituitary carcinomas. Endocr Pathol
Ubiquitylation on ACTH Processing. Neuroendocrinology 2020; 2007; 18(4):217-222.
110(1-2):119-129. 84. Buckley N, Bates AS, Broome JC et al. p53 protein accumulation
64. Ballmann C, Thiel A, Korah HE et  al. USP8 Mutations in in Cushings adenomas and invasive non-functional adenomas. J
Pituitary Cushing Adenomas-Targeted Analysis by Next- Clin Endocrinol Metab 1994; 79:1513-1516.
Generation Sequencing. J Endocr Soc 2018; 2(3):266-278. 85. Sumi T, Stefaneanu L, Kovacs K, Asa SL, Rindi G.
65. Reincke M, Sbiera S, Hayakawa A et  al. Mutations in the Immunohistochemical study of p53 protein in human and animal
deubiquitinase gene USP8 cause Cushing’s disease. Nat Genet pituitary tumors. Endocr Pathol 1993; 4:95-99.
2015; 47(1):31-38. 86. Levy A, Hall L, Yeundall WA, Lightman SL. p53 gene mutations
66. Hayashi K, Inoshita N, Kawaguchi K et al. The USP8 mutational in pituitary adenomas: rare events. Clin Endocrinol (Oxf) 1994;
status may predict drug susceptibility in corticotroph adenomas 41:809-814.
of Cushing’s disease. Eur J Endocrinol 2015. 87. Kawashima ST, Usui T, Sano T et al. P53 gene mutation in an
67. Ma ZY, Song ZJ, Chen JH et al. Recurrent gain-of-function USP8 atypical corticotroph adenoma with Cushing’s disease. Clin
mutations in Cushing’s disease. Cell Res 2015; 25(3):306-317. Endocrinol (Oxf) 2009; 70(4):656-657.
68. Cohen M, Persky R, Stegemann R et  al. Germline USP8 88. Amar AP, Hinton DR, Krieger MD, Weiss MH. Invasive pituitary
Mutation Associated With Pediatric Cushing Disease and Other adenomas: significance of proliferation parameters. Pituitary
Clinical Features: A New Syndrome. J Clin Endocrinol Metab 1999; 2(2):117-122.
2019; 104(10):4676-4682. 89. Goschzik T, Gessi M, Dreschmann V et al. Genomic Alterations
69. Chen J, Jian X, Deng S et al. Identification of recurrent USP48 of Adamantinomatous and Papillary Craniopharyngioma. J
and BRAF mutations in Cushing’s disease. Nat Commun 2018; Neuropathol Exp Neurol 2017; 76(2):126-134.
9(1):3171. 90. Sekine S, Shibata T, Kokubu A et al. Craniopharyngiomas of
70. Casar-Borota O, Boldt HB, Engstrom BE et al. Corticotroph adamantinomatous type harbor beta-catenin gene mutations. Am
aggressive pituitary tumours and carcinomas frequently harbour J Pathol 2002; 161(6):1997-2001.
ATRX mutations. J Clin Endocrinol Metab 2020. 91. Buslei R, Nolde M, Hofmann B et al. Common mutations of
71. Guo F, Wang G, Wang F, Xu D, Liu X. Identification of Novel beta-catenin in adamantinomatous craniopharyngiomas but
Genes Involved in the Pathogenesis of an ACTH-Secreting not in other tumours originating from the sellar region. Acta
Pituitary Carcinoma: A Case Report and Literature Review. Front Neuropathol 2005; 109(6):589-597.
Oncol 2018; 8:510. 92. Oikonomou E, Barreto DC, Soares B, De Marco L, Buchfelder
72. Karl M, Lamberts SWJ, Koper JW et  al. Cushing’s disease M, Adams EF. Beta-catenin mutations in craniopharyngiomas
preceded by generalized glucocorticoid resistance: clinical and pituitary adenomas. J Neurooncol 2005; 73(3):205-209.
consequences of a novel dominant-negative glucocorticoid 93. Brastianos PK, Taylor-Weiner A, Manley PE et  al. Exome
receptor mutation. Proc Assoc Am Physicians 1996; sequencing identifies BRAF mutations in papillary
108:296-307. craniopharyngiomas. Nat Genet 2014; 46(2):161-165.

13
14 Endocrine Pathology (2021) 32:3–16

94. Fukuhara N, Iwata T, Inoshita N et al. Immunohistochemistry mesenchymal-epithelial signalling during mouse organogenesis.
or Molecular Analysis: Which Method Is Better for Subtyping Develop 2000; 127(3):483-492.
Craniopharyngioma? Endocr Pathol 2020. 113. Celli G, LaRochelle WJ, Mackem S, Sharp R, Merlino G. Soluble
95. Brastianos PK, Shankar GM, Gill CM et al. Dramatic Response of dominant-negative receptor uncovers essential roles for fibroblast
BRAF V600E Mutant Papillary Craniopharyngioma to Targeted growth factors in multi-organ induction and patterning. EMBO J
Therapy. J Natl Cancer Inst 2016; 108(2). 1998; 17(6):1642-1655.
96. Roque A, Odia Y. BRAF-V600E mutant papillary 114. Ezzat S, Smyth HS, Ramyar L, Asa SL. Heterogeneous in vivo
craniopharyngioma dramatically responds to combination BRAF and in vitro expression of basic fibroblast growth factor by human
and MEK inhibitors. CNS Oncol 2017; 6(2):95-99. pituitary adenomas. J Clin Endocrinol Metab 1995; 80:878-884.
97. Rostami E, Witt NP, Libard S, Wikstrom J, Casar-Borota O, 115. Li Y, Koga M, Kasayama S et  al. Identification and
Gudjonsson O. Recurrent papillary craniopharyngioma with characterization of high molecular weight forms of basic
BRAFV600E mutation treated with neoadjuvant-targeted therapy. fibroblast growth factor in human pituitary adenomas. J Clin
Acta Neurochir (Wien ) 2017; 159(11):2217-2221. Endocrinol Metab 1992; 75:1436-1441.
98. Viaene AN, Lee EB, Rosenbaum JN, Nasrallah IM, Nasrallah 116. Heaney AP, Horwitz GA, Wang Z, Singson R, Melmed S. Early
MP. Histologic, immunohistochemical, and molecular features involvement of estrogen-induced pituitary tumor transforming
of pituicytomas and atypical pituicytomas. Acta Neuropathol gene and fibroblast growth factor expression in prolactinoma
Commun 2019; 7(1):69. pathogenesis. Nature Medicine 1999; 5:1317-1321.
99. Phillips JJ, Misra A, Feuerstein BG, Kunwar S, Tihan T. 117. Zimering MB, Katsumata N, Sato Y et  al. Circulating basic
Pituicytoma: characterization of a unique neoplasm by FGF-1 like substance in multiple endocrine neoplasia type 1
histology, immunohistochemistry, ultrastructure, and array-based and acromegaly. Fibroblast Growth Factor Family Meeting,San
comparative genomic hybridization. Arch Pathol Lab Med 2010; Diego,CA , 40. 1991 (Abstract)
134(7):1063-1069. 118. Correa FA, Trarbach EB, Tusset C et al. FGFR1 and PROKR2
100. Miller MB, Bi WL, Ramkissoon LA et al. MAPK activation and rare variants found in patients with combined pituitary hormone
HRAS mutation identified in pituitary spindle cell oncocytoma. deficiencies. Endocr Connect 2015; 4(2):100-107.
Oncotarget 2016; 7(24):37054-37063. 119. Abbass SAA, Asa SL, Ezzat S. Altered expression of fibroblast
101. Ezzat S, Walpola IA, Ramyar L, Smyth HS, Asa SL. Membrane- growth factor receptors in human pituitary adenomas. J Clin
anchored expression of transforming growth factor-a in human Endocrinol Metab 1997; 82:1160-1166.
pituitary adenoma cells. J Clin Endocrinol Metab 1995; 120. Zhu X, Lee K, Asa SL, Ezzat S. Epigenetic silencing through
80:534-539. DNA and histone methylation of fibroblast growth factor
102. Liu SC, Sanfilippo B, Perroteau I, Derynck R, Salomon DS, receptor 2 in neoplastic pituitary cells. Am J Pathol 2007;
Kidwell WR. Expression of transforming growth factor a (TGFa) 170(5):1618-1628.
in differentiated rat mammary tumors: estrogen induction of TGFa 121. Zhu X, Asa SL, Ezzat S. Fibroblast Growth Factor 2 and Estrogen
production. Mol Endocrinol 1987; 1:683-692. Control the Balance of Histone 3 Modifications Targeting
103. McAndrew J, Paterson AJ, Asa SL, McCarthy KJ, Kudlow JE. MAGE-A3 in Pituitary Neoplasia. Clin Cancer Res 2008;
Targeting of transforming growth factor-a expression to pituitary 14(7):1984-1996.
lactotrophs in transgenic mice results in selective lactotroph 122. Yu S, Asa SL, Weigel RJ, Ezzat S. Pituitary tumor AP-2alpha
proliferation and adenomas. Endocrinology 1995; 136:4479-4488. recognizes a cryptic promoter in intron 4 of fibroblast growth
104. Roh M, Paterson AJ, Asa SL, Chin E, Kudlow JE. Stage-sensitive factor receptor 4. J Biol Chem 2003; 278(22):19597-19602.
blockade of pituitary somatomammotrope development by 123. Ezzat S, Yu S, Asa SL. Ikaros isoforms in human pituitary tumors:
targeted expression of a dominant negative epidermal growth distinct localization, histone acetylation, and activation of the 5’
factor receptor in transgenic mice. Mol Endocrinol 2001; fibroblast growth factor receptor-4 promoter. Am J Pathol 2003;
15(4):600-613. 163(3):1177-1184.
105. LeRiche V, Asa SL, Ezzat S. Epidermal growth factor and 124. Ezzat S, Zheng L, Asa SL. Pituitary tumor-derived fibroblast
its receptor (EGF-R) in human pituitary adenomas: EGF-R growth factor receptor 4 isoform disrupts neural cell-adhesion
correlates with tumor aggressiveness. J Clin Endocrinol Metab molecule/N-cadherin signaling to diminish cell adhesiveness: a
1996; 81:656-662. mechanism underlying pituitary neoplasia. Mol Endocrinol 2004;
106. Cooper O, Vlotides G, Fukuoka H, Greene MI, Melmed S. 18(10):2543-2552.
Expression and function of ErbB receptors and ligands in the 125. Ezzat S, Zheng L, Winer D, Asa SL. Targeting N-Cadherin
pituitary. Endocr Relat Cancer 2011; 18(6):R197-R211. through Fibroblast Growth Factor Receptor-4: Distinct
107. Liu X, Kano M, Araki T et al. ErbB receptor-driven prolactinomas Pathogenetic and Therapeutic Implications. Mol Endocrinol 2006;
respond to targeted lapatinib treatment in female transgenic mice. 20(11):2965-2975.
Endocrinology 2015; 156(1):71-79. 126. Qian ZR, Sano T, Asa SL et  al. Cytoplasmic expression of
108. Gospodarowicz D, Ferrara N, Schweigerer L, Neufeld G. fibroblast growth factor receptor-4 in human pituitary adenomas:
Structural characterization and biological functions of fibroblast relation to tumor type, size, proliferation, and invasiveness. J Clin
growth factor. Endocr Rev 1987; 8:95-114. Endocrinol Metab 2004; 89(4):1904-1911.
109. Ferrara N, Schweigerer L, Neufeld G, Mitchell R, Gospodarowicz 127. Morita K, Takano K, Yasufuku-Takano J et al. Expression of
D. Pituitary follicular cells produce basic fibroblast growth factor. pituitary tumour-derived, N-terminally truncated isoform of
Proc Natl Acad Sci USA 1987; 84:5773-5777. fibroblast growth factor receptor 4 (ptd-FGFR4) correlates
110. Scully KM, Rosenfeld MG. Pituitary development: with tumour invasiveness but not with G-protein alpha subunit
regulatory codes in mammalian organogenesis. Science 2002; (gsp) mutation in human GH-secreting pituitary adenomas. Clin
295(5563):2231-2235. Endocrinol (Oxf) 2008; 68(3):435-441.
111. Itoh N, Ornitz DM. Evolution of the Fgf and Fgfr gene families. 128. Mete O, Cintosun A, Pressman I, Asa SL. Epidemiology and
Trends Genet 2004; 20(11):563-569. biomarker profile of pituitary adenohypophysial tumors. Mod
112. De Moerlooze L, Spencer-Dene B, Revest J, Hajihosseini Pathol 2018; 31(6):900-909.
M, Rosewell I, Dickson C. An important role for the IIIb 129. Daniel L, Trouillas J, Renaud W et al. Polysialylated-neural cell
isoform of fibroblast growth factor receptor 2 (FGFR2) in adhesion molecule expression in rat pituitary transplantable

13
Endocrine Pathology (2021) 32:3–16 15

tumors (spontaneous mammotropic transplantable tumor in Skp2 independent of PTEN in pituitary corticotroph cells.
Wistar-Furth rats) is related to growth rate and malignancy. Brain Pathol 2002; 12(4):412-419.
Cancer Res 2000; 60(1):80-85. 149. Woloschak M, Yu A, Post KD. Frequent inactivation of the
130. Nakano-Tateno T, Tateno T, Hlaing MM et  al. FGFR4 p16 gene in human pituitary tumors by gene methylation. Mol
polymorphic variants modulate phenotypic features of Cushing Carcinog 1997; 19(4):221-224.
disease. Mol Endocrinol 2014; 28(4):525-533. 150. Frost SJ, Simpson DJ, Clayton RN, Farrell WE. Transfection
131. Tateno T, Asa SL, Zheng L, Mayr T, Ullrich A, Ezzat S. The of an inducible p16/CDKN2A construct mediates reversible
FGFR4-G388R Polymorphism Promotes Mitochondrial STAT3 growth inhibition and G1 arrest in the AtT20 pituitary tumor
Serine Phosphorylation to Facilitate Pituitary Growth Hormone cell line. Mol Endocrinol 1999; 13(11):1801-1810.
Cell Tumorigenesis. PLoS Genet 2011; 7(12):e1002400. 151. Zhang X, Sun H, Danila DC et  al. Loss of expression of
132. Ezzat S, Wang R, Pintilie M, Asa SL. FGFR4 polymorphic GADD45 gamma, a growth inhibitory gene, in human pituitary
alleles modulate mitochondrial respiration: A novel target for adenomas: implications for tumorigenesis. J Clin Endocrinol
somatostatin analog action in pituitary tumors. Oncotarget 2017; Metab 2002; 87(3):1262-1267.
8(2):3481-3494. 152. Bahar A, Bicknell JE, Simpson DJ, Clayton RN, Farrell
133. Altas M, Bayrak OF, Ayan E et al. The effect of polymorphisms WE. Loss of expression of the growth inhibitory gene
in the promoter region of the MMP-1 gene on the occurrence and GADD45gamma, in human pituitary adenomas, is associated
invasiveness of hypophyseal adenoma. Acta Neurochir (Wien ) with CpG island methylation. Oncogene 2004; 23(4):936-944.
2010; 152(9):1611-1617. 153. Zhao J, Dahle D, Zhou Y, Zhang X, Klibanski A.

134. Jacks T, Fazeli A, Schmitt EM, Bronson RT, Goodell MA, Hypermethylation of the promoter region is associated with
Weinberg RA. Effects of an Rb mutation in the mouse. Nature the loss of MEG3 gene expression in human pituitary tumors.
1992; 359:295-300. J Clin Endocrinol Metab 2005; 90(4):2179-2186.
135. Cryns VL, Alexander JM, Klibanski A, Arnold A. The 154. Metin-Armagan D, Comunoglu N, Bulut G et  al. A Novel
retinoblastoma gene in human pituitary tumors. J Clin Endocrinol Expression Profile of Cell Cycle and DNA Repair Proteins
Metab 1993; 77:644-646. in Nonfunctioning Pituitary Adenomas. Endocr Pathol 2020;
136. Woloschak M, Yu A, Xiao J, Post KD. Abundance and state of 31(1):2-13.
phosphorylation of the retinoblastoma gene product in human 155. Bahar A, Simpson DJ, Cutty SJ et  al. Isolation and
pituitary tumors. Int J Cancer 1996; 67(1):16-19. characterization of a novel pituitary tumor apoptosis gene. Mol
137. Pei L, Melmed S, Scheithauer B, Kovacs K, Benedict WF, Endocrinol 2004; 18(7):1827-1839.
Prager D. Frequent loss of heterozygosity at the retinoblastoma 156. Zhu X, Mao X, Hurren R, Schimmer AD, Ezzat S,
susceptibility gene (RB) locus in aggressive pituitary tumors: Asa SL. Deoxyribonucleic acid methyltransferase 3B
Evidence for a chromosome 13 tumor suppressor gene other than promotes epigenetic silencing through histone 3 chromatin
RB. Cancer Res 1995; 55:1613-1616. modifications in pituitary cells. J Clin Endocrinol Metab 2008;
138. Bates AS, Farrell WE, Bicknell EJ et al. Allelic deletion in 93(9):3610-3617.
pituitary adenomas reflects aggressive biological activity and 157. Georgopoulos K, Winandy S, Avitahl N. The role of the Ikaros
has potential value as a prognostic marker. J Clin Endocrinol gene in lymphocyte development and homeostasis. Annu Rev
Metab 1997; 82:818-824. Immunol 1997; 15:155-176.
139. Tsai KY, MacPherson D, Rubinson DA et al. ARF mutation 158. Ezzat S, Mader R, Yu S, Ning T, Poussier P, Asa SL. Ikaros
accelerates pituitary tumor development in Rb+/- mice. Proc integrates endocrine and immune system development. J Clin
Natl Acad Sci U S A 2002; 99(26):16865-16870. Invest 2005; 115(4):1021-1029.
140. Lasorella A, Rothschild G, Yokota Y, Russell RG, Iavarone A. 159. Ezzat S, Mader R, Fischer S, Yu S, Ackerley C, Asa SL. An
Id2 mediates tumor initiation, proliferation, and angiogenesis in essential role for the hematopoietic transcription factor Ikaros
Rb mutant mice. Mol Cell Biol 2005; 25(9):3563-3574. in hypothalamic-pituitary-mediated somatic growth. Proc Natl
141. Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin- Acad Sci U S A 2006; 103(7):2214-2219.
dependent kinases. Genes Dev 1995; 9:1149-1163. 160. Ezzat S, Yu S, Asa SL. The zinc finger Ikaros transcription
142. Nakayama K, Ishida N, Shirane M et al. Mice lacking p­ 27Kip1 factor regulates pituitary growth hormone and prolactin gene
display increased body size,multiple organ hyperplasia, retinal expression through distinct effects on chromatin accessibility.
dysplasia, and pituitary tumors. Cell 1996; 85:707-720. Mol Endocrinol 2005; 19(4):1004-1011.
143. Kiyokawa H, Kineman RD, Manova-Todorova KO et  al. 161. Ezzat S, Zhu X, Loeper S, Fischer S, Asa SL. Tumor-derived
Enhanced growth of mice lacking the cyclin-dependent kinase Ikaros 6 acetylates the Bcl-XL promoter to up-regulate a
inhibitor function of ­p27Kip1. Cell 1996; 85:721-732. survival signal in pituitary cells. Mol Endocrinol 2006;
144. Fero ML, Rivkin M, Tasch M et al. A syndrome of multiorgan 20(11):2976-2986.
hyperplasia with features of gigantism, tumorigenesis, and 162. Loeper S, Asa SL, Ezzat S. Ikaros modulates cholesterol
female sterility in ­p27Kip1-deficient mice. Cell 1996; 85:733-744. uptake: A link between tumor suppression and differentiation.
145. Franklin DS, Godfrey VL, Lee H et  al. CDK inhibitors Cancer Res 2008; 68(10):3715-3723.
p18(INK4c) and p27(Kip1) mediate two separate pathways to 163. Fedele M, Battista S, Kenyon L et al. Overexpression of the
collaboratively suppress pituitary tumorigenesis. Genes Dev HMGA2 gene in transgenic mice leads to the onset of pituitary
1998; 12(18):2899-2911. adenomas. Oncogene 2002; 21(20):3190-3198.
146. Bamberger CM, Fehn M, Bamberger AM et  al. Reduced 164. Fedele M, Pentimalli F, Baldassarre G et  al. Transgenic
expression levels of the cell-cycle inhibitor p27Kip1 in human mice overexpressing the wild-type form of the HMGA1
pituitary adenomas. Eur J Endocrinol 1999; 140(3):250-255. gene develop mixed growth hormone/prolactin cell pituitary
147. Dahia PL, Aguiar RC, Honegger J et al. Mutation and expression adenomas and natural killer cell lymphomas. Oncogene 2005;
analysis of the p27/kip1 gene in corticotrophin- secreting 24(21):3427-3435.
tumours. Oncogene 1998; 16(1):69-76. 165. Fedele M, Visone R, De M, I et al. HMGA2 induces pituitary
148. Liu W, Asa SL, Ezzat S. Vitamin D and its analog EB1089 tumorigenesis by enhancing E2F1 activity. Cancer Cell 2006;
induce p27 accumulation and diminish association of p27 with 9(6):459–471.

13
16 Endocrine Pathology (2021) 32:3–16

166. De Martino I, Visone R, Wierinckx A et al. HMGA proteins 171. Tamura R, Ohara K, Morimoto Y et al. PITX2 Expression in
up-regulate CCNB2 gene in mouse and human pituitary Non-functional Pituitary Neuroendocrine Tumor with Cavernous
adenomas. Cancer Res 2009; 69(5):1844-1850. Sinus Invasion. Endocr Pathol 2019; 30(2):81-89.
167. Evans CO, Moreno CS, Zhan X et al. Molecular pathogenesis 172. Mete O, Hayhurst C, Alahmadi H et al. The role of mediators
of human prolactinomas identified by gene expression of cell invasiveness, motility, and migration in the pathogenesis
profiling, RT-qPCR, and proteomic analyses. Pituitary 2008; of silent corticotroph adenomas. Endocr Pathol 2013;
11(3):231-245. 24(4):191-198.
168. Finelli P, Pierantoni GM, Giardino D et al. The High Mobility 173. Krokker L, Nyiro G, Reiniger L et al. Differentially Expressed
Group A2 gene is amplified and overexpressed in human miRNAs Influence Metabolic Processes in Pituitary Oncocytoma.
prolactinomas. Cancer Res 2002; 62(8):2398-2405. Neurochem Res 2019; 44(10):2360-2371.
169. Qian ZR, Asa SL, Siomi H et al. Overexpression of HMGA2
relates to reduction of the let-7 and its relationship to Publisher’s Note Springer Nature remains neutral with regard to
clinicopathological features in pituitary adenomas. Mod Pathol jurisdictional claims in published maps and institutional affiliations.
2009; 22(3):431-441.
170. Mete O, Ezzat S, Asa SL. Biomarkers of aggressive pituitary
adenomas. J Mol Endocrinol 2012; 49(2):R69-R78.

13

You might also like