ArticlePDF Available

Clinical Implications of Accurate Subtyping of Pituitary Adenomas: Perspectives from the Treating Physician

Authors:
  • The Princess Margaret Hospital/University Health Network
  • Case Western Reserve University and University Health Network Toronto

Abstract and Figures

Pituitary adenomas comprise a heterogenous group of adenohypophyseal tumours with distinct clinicopathological features across both the clinically functioning and silent groups. Although, predicting a clinically aggressive course remains challenging, accurate subtyping of pituitary adenomas offers valuable prognostic information that together with other clinical and radiological information serves as a platform for tailored treatment and follow-up. For instance, silent subtype 3 pituitary adenomas, silent corticotroph adenomas, acidophil stem cell adenomas, Crooke cell adenomas, and sparsely granulated somatotroph adenomas show more invasive growth. This review has been formulated as a set of practical questions that address the distinct clinical behaviour of a selected group of pituitary adenoma subtypes.
Content may be subject to copyright.
doi: 10.5146/tjpath.2015.01311
Review
4
Receved : 09.06.2015 Accepted : 10.06.2015
Correspondence: Özgür METE
200 Elizabeth Street, 11th Floor, Department of Pathology,
University Health Network, TORONTO, ON, CANADA
E-mail: ozgur.mete2@uhn.ca Phone: +1 416 340 3004
(Turk Patoloji Derg 2015, 31(Suppl):4-17)
ABSTRACT
Ptutary adenomas comprse a heterogenous group of adenohypophyseal tumours wth dstnct clncopathologcal features across both the
clncally functonng and slent groups. Although, predctng a clncally aggressve course remans challengng, accurate subtypng of ptutary
adenomas oers valuable prognostc nformaton that together wth other clncal and radologcal nformaton serves as a platform for talored
treatment and follow-up. For nstance, slent subtype 3 ptutary adenomas, slent cortcotroph adenomas, acdophl stem cell adenomas, Crooke
cell adenomas, and sparsely granulated somatotroph adenomas show more nvasve growth. s revew has been formulated as a set of practcal
questons that address the dstnct clncal behavour of a selected group of ptutary adenoma subtypes.
Key Words: Ptutary adenoma, Acromegaly, Hyperthyrodsm, Cushng syndrome
Clinical Implications of Accurate Subtyping of Pituitary
Adenomas: Perspectives from the Treating Physician
Karen GOMEZ-HERNANDEZ1, Shereen EZZAT1, Sylva L. ASA2, Özgür METE2
Department of 1Medicine and 2Pathology, University Health Network, University of Toronto, TORONTO, ONTARIO, CANADA
INTRODUCTION
As members of the multidisciplinary endocrine oncology
team providing care for patients with pituitary disease we
have oen been intrigued by the relative scarcity of studies
in the eld that describe the clinical relevance of accurate
pituitary adenoma subtyping. Certainly, in other elds of
endocrine oncology such as thyroid cancer, the description
of clinicopathological features has emerged as pivotal
elements in disease risk stratication and management.
Although ancillary tests that distinguish aggressive
pituitary adenomas from pituitary carcinomas are still
unavailable, the accurate subtyping of pituitary adenomas
in association with selected biomarkers is still considered
the best predictor and prognosticator (1-5). Modern
approaches to the classication of pituitary adenomas use
a panel approach by integrating adenohypophyseal cell-
lineage specic transcription factors (Pit-1, Tpit, SF-1, and
ER), monoclonal antibodies against adenohypophyseal
hormones (Growth hormone: GH, Prolactin: PRL, beta-
thyroid stimulating hormone: beta-TSH, beta-follicle
stimulating hormone: beta-FSH, beta-luteinizing hormone:
beta-LH, Adrenocorticotropic hormone: ACTH, and
alpha-subunit), low molecular weight keratin (CAM5.2
or cytokeratin 18), and Ki-67 (MIB-1) (1-3,5,6). p53
immunohistochemistry is also a part of this panel in some
practices (5). is approach identies tumours that are more
frequently associated with invasive growth (Hardys’ grade
III/IV and Knosp’s grade III/IV), higher recurrent rates, and
a distinct response to therapy (Table 1). Importantly, this
morphologic categorization into aggressive subtypes has
been suggested to be complementary and in some instances
perhaps even superior to the designation of atypical
pituitary adenomas, which are invasive adenomas showing
p53 positivity and/or a MIB-1 labeling index >3%. In this
brief review that has been formulated as a set of questions
we will address the distinct behaviour of a selected group
of pituitary adenoma subtypes in selected clinical settings.
A. CLINICALLY NONFUNCTIONING PITUITARY
ADENOMAS
Question for the Pathologist: Which type of adenoma is
it?
Clinical Relevance: Certain clinically non-functioning
pituitary adenomas are characterized by more aggressive
behaviour.
Although all pituitary adenoma subtypes can potentially
present as clinically non-functioning, based on recent
surgical series, gonadotroph adenomas are the most
frequent (7). Amongst the less common pituitary adenomas
that may present as clinically silent tumours, silent
corticotroph adenomas and silent subtype 3 adenomas have
more aggressive clinical behaviour in terms of size, invasive
growth, and recurrence rates (7,8). ese tumour subtypes
also present at an earlier mean age than gonadotroph
adenomas (7,8). Null cell adenomas have also been
recognized as more invasive than gonadotroph adenomas
5
Turkish Journal of Pathology
GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
Table I: Subtyping of pituitary adenomas
Adenoma subtype Distinct clinical features Hormones, transcription factors, and CAM5.2
Pit-1 family tumours
PRL-producing adenomas
Sparsely granulated lactotroph adenoma
Densely granulated lactotroph adenoma
Acidophil stem cell adenoma
GH-producing adenomas
Densely granulated somatotroph adenoma
Intermediate granulated somatotroph adenoma
Sparsely granulated somatotroph adenoma
Mammosomatotroph adenoma
Mixed somatotroph and lactotroph adenoma
TSH-producing adenomas
yrotroph adenoma
Monomorphous plurihormonal adenoma
Silent subtype 3 adenoma
Common subtype, responsive to dopamine agonists
Likely more resistant to dopamine agonists
Likely more resistant to dopamine agonists
Likely to respond to somatostatin analogues
Behave like densely granulated somatotroph adenomas
More resistant to somatostatin analogues
Aggressive tumours; central hyperthyroidism
Present in younger individuals when compared to
gonadotroph adenomas; invasive and recurrent
Pit-1, ER, PRL (golgi pattern)
Pit-1, ER, PRL (diuse)
Pit-1, ER, PRL (diuse), GH (variable), CAM5.2 (few brous bodies)
Pit-1, GH (diuse), α-SU, CAM5.2 (perinuclear)
Pit-1, GH (diuse), α-SU, CAM5.2 (brous bodies <70%))
Pit-1, GH (weak), CAM5.2 (brous bodies >70%)
Pit-1, ER, GH, PRL, α-SU
Pit-1, ER, GH, PRL, α-SU
Pit-1, GATA-2, β-TSH, and α-SU
Pit-1, ER (variable), α-SU (variable), and GH/PRL/β-TSH (variable)
Tpit family tumours
Densely granulated corticotroph adenoma
Sparsely granulated corticotroph adenoma
Crooke cell adenoma
If excised completely cure is possible
Aggressive tumours
Typically present with normal or mildly elevated
ACTH; Aggressive tumours
Tpit, ACTH (strong, diuse), CAM5.2 (strong, diuse)
Tpit, ACTH (weak, variable), CAM5.2 (strong, diuse)
Tpit, ACTH (juxtanuclear and peripheral), CAM5.2 (ring-like)
SF-1 family tumours
Hormone-negative gonadotroph adenoma
Hormone-positive gonadotroph adenoma
Most common clinically non-functioning adenoma;
hormone status is of no clinical signicance
SF-1, ER, GATA-2, α-SU, β-FSH and/or β-LH, CAM5.2 (-/+)
SF-1, ER, GATA-2, CAM5.2 (-/+)
Polymorphous Plurihormonal adenoma
Plurihormonal adenoma Extremely rare Multiple
Transcription factor and hormone negative
adenoma
Null cell adenoma
Tumours that have been shown to grow rapidly
preoperatively will continue to show rapid growth
of residual disease; more invasive than gonadotroph
adenomas.
Negative for all adenohypophyseal transcription factors and
hormones; CAM5.2 (-/+).
Pit-1: Pituitary transcription factor-1, ER: Estrogen Receptor, SF-1: Steroidogenic factor-1, GH: Growth hormone, PRL: Prolactin, β-TSH: Beta-thyroid stimulating hormone, ACTH:
Adrenocorticotrophic hormone, α-SU: alpha-subunit, β-FSH: Beta-follicle stimulating hormone, β-LH: Beta-luteinizing hormone.
6
Turkish Journal of Pathology GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
with mass eects (13), invasion (7), and a high frequency
of pituitary apoplexy (acute hemorrhagic necrosis) (12,
14, 15). In a study that directly compared gonadotroph
adenomas (dened based on immunoreactivity for beta-
FSH, beta-LH or alpha-subunit) to SCA, it was shown that
SCA have higher and earlier recurrences (13). Also, when
compared to other clinically silent adenomas (mainly null
cell adenomas and gonadotrophs adenomas), patients
younger than 30 years of age with silent corticotroph
adenomas more oen have multiple recurrences (>2) and
late recurrences (more than ve years aer initial resection)
(15). It has been suggested that interactions between tumour
cells and extracellular matrix may be one of the mechanisms
leading to distinct behaviour by these pituitary adenomas;
it may be that osteopontin plays a role in the invasiveness
of SCAs, whereas MMP-1 is more frequently expressed in
gonadotroph adenomas (16).
e morphologic distinction of corticotroph origin in a
non-functioning pituitary adenoma is not sucient to
complete a diagnosis, since corticotroph adenomas are a
heterogeneous group of neoplasms. Densely granulated
corticotroph adenomas typically present with a basophilic
cytoplasm correlating with diuse strong PAS positivity
and immunoreactivity for ACTH (Figure 2B); in contrast,
sparsely granulated corticotroph adenomas display focal
PAS staining and weak positivity for ACTH (1-3,17,18).
Regardless of PAS and/or ACTH positivity, both types
of corticotroph adenoma are diusely positive for Tpit
(1,3,5,17,18). Silent densely and sparsely granulated
(9) and to rapidly grow aer surgery if they had shown a
rapid pattern of growth preoperatively (10).
Null Cell Adenomas
e appropriate classication of an adenohypophyseal
tumour into the null cell pituitary adenoma category
requires negativity of cell-type specic dierentiation
using adenohypophyseal hormones and pituitary
transcription factors. e reason this type of adenoma
was overrepresented in older surgical series is that without
the use of pituitary transcription factors, a signicant
proportion of gonadotroph adenomas, which were negative
for beta-FSH and beta-LH, were mistakenly subtyped as
“null cell adenomas. In fact, the use of SF-1 and ER has
improved the detection of gonadotroph dierentiation in
hormone-negative pituitary adenomas (Figure 1A,B). e
distinction of null cell adenoma is of clinical relevance.
Firstly, true null cell adenomas that grow rapidly before
surgery will continue to show rapid growth of residual or
recurrent disease postoperatively (10), a feature that dictates
a close surveillance strategy. Secondly, null cell adenomas
also seem to show more cavernous sinus invasion than
gonadotroph adenomas (7) which as expected predicts
residual disease (10).
Silent Corticotroph Adenomas
Clinically, silent corticotroph adenomas are characterized by
the lack of clinical signs or symptoms of Cushing’s syndrome
and normal cortisol and ACTH levels (11,12). ese tumors
oen present as macroadenomas (Figure 2A) associated
Figure 1: SF-1 immunohistochemistry helps to distinguish gonadotroph dierentiation in hormone negative adenomas. A) is
photomicrograph illustrates a pituitary adenoma that was negative for all adenohypophyseal hormones. B) Positivity for SF-1 conrms
gonadotroph cell dierentiation in this tumor.
A B
7
Turkish Journal of Pathology
GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
corticotroph adenomas are classied as type 1 (Figure 2A)
and type 2 SCAs, respectively. It has been shown that type 2
SCAs have a higher expression prole of factors regulating
tumor cell invasion/ migration and proliferation, such
as MMP-1, β1-integrin, and FGFR4, compared with type
I SCAs16. Corticotroph adenomas with Crooke’s hyaline
change, also known as “Crooke cell adenomas, have also
been associated with aggressive behaviour (19, 20).
Silent Subtype 3 Pituitary Adenomas
Silent subtype 3 pituitary adenomas are monomorphous
plurihormonal Pit-1 lineage adenomas that may be
clinically silent (3, 5). However, the term “silent” is a
misnomer, since these tumours can cause acromegaly,
hyperprolactinemia (albeit generally due to stalk eect)
or hyperthyroidism (8,21-23). ese tumours tend to be
invasive macroadenomas or giant adenomas (Figure 3A)
and they present at an earlier mean age than gonadotroph
adenonomas(7, 8, 22, 23). Also, recurrence and tumour-
free status may be as low as a third of treated patients (8).
Not surprisingly, radiotherapy is required in a substantial
number of cases (8) and it appears to achieve control of
disease (22). Somatostatin receptor expression has been
shown in some of these tumours and tumour stability on
long acting octreotide has been previously reported in two
individuals with residual disease (22).
Pathologists should distinguish these neoplasms by
demonstrating variable positivity for one or more Pit-
1-lineage hormones (GH, PRL, and beta-TSH) together
with diuse Pit-1 nuclear reactivity (Figure 3B-D) (3, 5).
It is important to distinguish true tumor cell hormone
expression from scattered positivity that represents
entrapped nontumorous adenohypophyseal cells.
Ultrastructural examination of silent subtype 3 adenomas
reveals characteristic nuclear inclusions known as
“spheridia” (3, 5, 22).
B. ADENOMAS CAUSING PROLACTIN EXCESS
Question for the Pathologist: Is it a sparsely granulated
lactotroph adenoma (most common subtype) or a less
frequent and potentially more aggressive subtype?
Clinical Relevance: Sparsely granulated lactotroph
adenomas usually respond to dopamine agonist therapy. In
patients with acidophil stem cell adenomas, the diagnosis
of GH excess may be missed due to a clinical picture that is
dominated by symptoms related to prolactin excess.
Most “prolactinomas” are sparsely granulated lactotroph
adenomas; densely granulated lactotroph adenomas and
acidophil stem cell adenomas are rare (3). While acidophil
stem cell adenomas are typically associated with prolactin
excess, concomitant GH-excess may occur causing
acromegaly or gigantism (24-26). Given the predominance
of hyperprolactinemia related symptoms, the diagnosis of
GH-excess may be missed if the GH axis is not appropriately
evaluated; this has been described as “fugitive acromegaly”.
Hyperprolactinemia in the setting of acidophil stem
cell adenomas is dierent from that observed in sparsely
A B
Figure 2: Silent corticotroph adenoma. A) Magnetic resonance image showing invasive growth of a silent subtype 2 adenoma; most
silent corticotroph adenomas are invasive macroadenomas (arrow) with a predilection of cavernous sinus invasion, B) Diuse ACTH
expression in a type 1 silent corticotroph adenoma (densely granulated corticotroph adenoma lacking any biochemical or clinical
evidence of excess ACTH) is illustrated.
8
Turkish Journal of Pathology GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
that are correlated with lack of therapeutic responsiveness
have not provided a detailed morphologic characterization
of the prolactin-producing adenomas and have instead
classied them according to size, i.e., macroprolactinomas
vs microprolactinomas (31,32). In our own experience and
as illustrated by the previous report of two pediatric cases
(33), acidophil stem cell adenomas tend to be resistant
to dopamine agonist therapy both in terms of prolactin
reduction and tumour shrinkage. is clinical observation
is supported by in vitro studies showing bromocriptine
resistance in cells of acidophil stem cell adenomas (34).
granulated lactotroph adenomas in that is not as
proportional to tumour size, i.e., larger acidophil stem
cell adenomas produce less prolactin than similar size
sparsely granulated lactotroph adenomas. Another distinct
characteristic of acidophil stem cell adenomas is that they
are usually invasive macroadenomas (27,29).
Although the vast majority of “prolactinomas” show a
good response to dopamine agonist therapy (30); there
are instances in which a reduction in tumour size and
prolactin concentrations is dicult to achieve. Studies that
have aimed at characterizing the relevant clinical features
Figure 3: Magnetic resonance image and histopathology of a silent subtype 3 adenoma. A) e magnetic resonance image shows a
macroadenoma with suprasellar extension and invasion into the le cavernous sinus; this patient had presented with hypopituitarism
and visual eld disturbances. B) Silent subtype 3 adenomas are diusely positive for Pit-1. C-D) Variable positivity for GH, PRL, and
beta-TSH render the diagnosis of these tumours (GH: C; PRL: D).
A
C
B
D
9
Turkish Journal of Pathology
GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
It has also been our experience that densely granulated
lactotroph adenomas are relatively rare; they are usually
larger and more invasive than sparsely granulated lactotroph
adenomas.
From a pathological perspective, paranuclear Golgi-type
PRL staining distinguishes sparsely granulated lactotroph
adenomas from acidophil stem cell adenomas and densely
granulated lactotroph adenomas, which oen display
diuse cytoplasmic PRL expression (3,5). Oncocytic change
with dilated mitochondria is one of the characteristics of
acidophil stem cell adenomas in addition to scattered brous
bodies and concomitant focal GH expression (3,5, 35).
When compared to controls, MEN-1 patients more
frequently harbour plurihormonal pituitary adenomas as
well as multiple adenomas (36). Plurihormonality in this
setting most oen includes GH and PRL expression. On
the other hand, when multiple synchronous adenomas
are present they are frequently a combination of PRL- and
ACTH-producing adenomas. PRL-producing tumours in
MEN-1 also tend to be larger (37) and more prone to be
resistant to dopamine agonists (37, 38).
C. ADENOMAS CAUSING GROWTH HORMONE
EXCESS
C1. Question for the Pathologist: Is the somatotroph
adenoma sparsely granulated or densely granulated?
Clinical Relevance: Dierential therapeutic responsiveness
to somatostatin analogues and clinical behaviour.
Pure (isolated) GH-producing pituitary adenomas are
histologically classied into densely granulated and sparsely
granulated somatotroph adenomas. Densely granulated
somatotroph adenomas are the most frequent subtype and
they generally respond better to somatostatin analogues
(39-42). Part of the explanation for the dierential response
is that sparsely granulated somatotroph adenomas tend
to be larger and more invasive (42-45) (Figure 4A) while
also showing relatively lower SSTR2 expression (46,47).
Also, albeit discordant reports in the literature (41,47-49),
it appears that a higher proportion of densely granulated
somatotroph adenomas have high intracellular cAMP levels
due to activation of the protein kinase-A pathway making
them excellent targets for cAMP suppression via SSTR,
whereas the mechanisms underlying sparsely granulated
somatotroph adenomas appear to involve the STAT
signaling pathway2,3. Interestingly, aected individuals
with familial isolated somatotroph adenomas (some of
which harbour germline mutations in the aryl hydrocarbon
receptor interacting gene) tend to have invasive adenomas
with less response to somatostatin analogues (50,51).
Not surprisingly, the proportion of sparsely granulated
somatotroph adenoma is overrepresented in this syndrome
(50,51). It also appears that sparsely granulated somatotroph
adenomas are overrepresented in the subgroup of
sporadic somatotroph adenomas associated with low aryl
hydrocarbon receptor interacting protein expression and
poor response to somatostatin analogue therapy (52).
Pathologists distinguish these two neoplasms by assessing
the staining characteristics of low molecular weight keratin
(CAM5.2 or CK18), GH, and alpha-subunit (Figure 4B-D).
Sparsely granulated somatotroph adenomas typically show
focal or weak GH expression, no alpha-subunit expression,
and prominent (>70% of the tumor cells) juxtanuclear
globular reactivity for low molecular weight keratin,
corresponding to intermediate lament aggresomes known
as “brous bodies” (3,5,44). Unlike sparsely granulated
somatotroph adenomas, densely granulated somatotroph
adenomas display diuse and strong positivity for GH and
alpha-subunit, and low molecular weight keratin stains in
a perinuclear pattern (3,5). Scattered brous bodies can be
identied in some cases within the phenotype of densely
granulated somatotroph adenomas; while these neoplasms
are considered to represent an intermediate form of
somatotroph adenoma, their biologic features including
treatment response to somatostatin do not dier from
densely granulated somatotroph adenomas (44).
C2. Questions for the Pathologist: Is the adenoma not
a pure somatotroph adenoma? Is there any evidence of
underlying somatotroph hyperplasia or multifocal disease?
Clinical Relevance: Other pituitary adenomas can also
cause excess GH in addition to other hormones, mainly
prolactin. e coexistence of multicentric disease and/or
associated hyperplasia should alert the physician to the
possibility of GHRH-producing tumors, Carney Complex
or McCune Albright syndrome.
In contrast to somatotroph adenomas, mammosomatotroph
adenomas (53,54), mixed somatotroph and lactotroph
adenomas, and other plurihormonal adenomas (silent
subtype 3 adenomas, acidophil stem cell adenomas,
and GH-producing plurihormonal adenomas) may co-
secrete GH and PRL. Currently, it is unclear whether in
acromegalic patients there is a dierential response to
dopamine agonist therapy between these tumours and
pure somatotroph adenomas. It is important to mention
though that baseline prolactin levels do not seem to predict
response to dopamine agonist therapy (55).
10
Turkish Journal of Pathology GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
Mammosomatotroph adenomas most commonly cause GH
excess leading to acromegaly or gigantism while prolactin
is usually only mildly elevated (34,53,56,57). Interestingly,
mammosomatotroph hyperplasia coexisting with
adenomas is frequently found in patients with acromegaly
in the setting of Carney complex (58-60). Precisely because
of the diuse pituitary involvement in which hyperplasia
and small tumours coexist, these patients tend to have
pituitary imaging that does not clearly identify the presence
of an adenoma. e National Institute of Health group has
been successful at treating these imaging-negative Carney
complex acromegalics with somatostatin analogues (60).
e pituitaries of acromegalic patients with McCune Al-
bright Syndrome also show areas of hyperplasia (somato-
troph/mammosomatotroph) together with areas of fully
developed adenoma (somatotroph/mammosomatotroph)
(61,62). erefore, especially in young individuals with
gigantism or acromegaly, a surgical pathology report indi-
Figure 4: Sparsely granulated somatotroph adenoma. A) Magnetic resonance image showing an invasive sparsely granulated
somatotroph adenoma; typically as shown here these tumours are hyperintense in T2-weighed imaging (arrows indicate the adenoma).
B) Careful examination of the hematoxylin-eosin stained slides can highlight the presence of juxtanuclear globular brous bodies
(arrows indicate brous bodies). C) Densely granulated somatotroph adenomas show diuse alpha-subunit and perinuclear CAM5.2
expression. D) Sparsely granulated somatotroph adenomas show prominent brous bodies on CAM5.2.
A
C
B
D
11
Turkish Journal of Pathology
GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
cating the presence of coexisting hyperplasia and adenoma
should alert the clinician to the possibility of either of these
multiple endocrine neoplasia syndromes.
Outside of Carney complex and McCune Albright
Syndrome, very early childhood onset of pituitary gigantism
(<4 years old) caused by diuse mammosomatotroph
hyperplasia has been described (63,64).
Finally, in some patients a combined pituitary neoplasm
consisting of GHRH-producing gangliocytoma and a
somatotroph adenoma can be identied in the background
of somatotroph hyperplasia (65). However, the more
frequent source of GHRH leading to somatotroph and/
or mammosomatotroph hyperplasia is ectopic GHRH
produced by a neuroendocrine tumor of the lung, pancreas,
adrenal or other sites (66,67).
D. ADENOMAS CAUSING ACTH EXCESS
A detailed surgical pathology report is particularly relevant
in the setting of Cushing disease as it provides very useful
prognostic information. It may also alert the clinician as
to the possibility of an incorrect diagnosis, i.e. pseudo-
Cushing’s syndrome.
D1. Questions for the Pathologist: Was an adenoma
identied? Did the nontumorous corticotrophs show
Crookes hyaline change?
Clinical Relevance: Lack of identication of a corticotroph
adenoma in the surgical pathology specimen is associated
with non-remission aer pituitary surgery. Identication of
Crookes hyaline change in the nontumorous corticotrophs
conrms pathological hypercortisolemia.
Corticotroph adenomas tend to be microadenomas that
sometimes can be quite small and dicult to localize.
Lack of corticotroph adenoma identication in a surgical
pathology specimen of a patient with Cushing disease is
more frequently observed in patients in whom remission
is not achieved (68, 69). ere are several possibilities as
to why an adenoma is not detected on surgical pathology:
(a) it was missed by the surgeon, (b) it was destroyed
during the procedure, (c) the diagnosis of Cushing disease
was incorrect, (d) Cushing’s syndrome was caused by
corticotroph hyperplasia, but not by an adenoma.
Diuse corticotroph hyperplasia can be due to ectopic
CRH production (70); therefore, a surgical pathology
specimen in which this type of hyperplasia is observed
should prompt the clinician to initiate investigations to
localize the responsible tumour. However, both diuse and
nodular forms of corticotroph hyperplasia may also be a
rare aetiology of pituitary Cushing disease (71, 72).
e distinction between hyperplasia and adenoma is per-
formed by assessing characteristics of the reticulin network
surrounding microacinar units of the adenohypophysis.
While pituitary adenoma shows loss of reticulin network,
hyperplasia presents with an intact but expanded reticulin
framework. Glucocorticoid excess results in Crooke’s hya-
line change of the non-tumorous corticotrophs (Figure 5
A,B), reecting the negative inhibition of excess glucocor-
ticoids on non-tumorous pituitary corticotrophs54,55. While
this can easily be identied on haematoxylin and eosin-
stained slides, PAS along with ACTH and low molecular
weight keratin can be used to highlight this cellular altera-
tion (Figure 5 A,B). Of note, Crooke’s hyaline change is typ-
ically absent in diuse corticotroph hyperplasia (due to lack
of normal corticotrophs), in pseudo-Cushing syndrome, as
well as in the non-tumorous corticotrophs of patients with
silent corticotroph adenomas. If the specimen fails to show
a corticotroph adenoma or diuse corticotroph hyperpla-
sia, the presence or absence of Crooke’s hyaline change can
provide important insights for treating physicians for the
diagnosis of a pseudo-Cushing state (3).
D2. Question for the Pathologist: Which type of adenoma
is it?
Clinical Relevance: Distinct clinical features
Tumours that produce ACTH can be classied as clinically
non-functioning (silent subtype 1 and silent subtype 2
corticotroph adenomas reviewed above) or as clinically
functioning (sparsely granulated corticotroph adenomas,
densely granulated corticotroph adenomas, and Crooke
cell adenomas). Amongst these adenomas causing
Cushing disease, Crooke cell adenomas (Figure 6 A,B) are
considered an aggressive histologic variant (19,20). ese
tumours are usually invasive macroadenomas with a high
recurrence rate that can be as high as 60% aer a mean
follow up of 6.7 years (20). In our experience, sparsely
granulated corticotroph adenomas are also associated with
more aggressive behaviour than their densely granulated
counterpart.
E. ADENOMAS CAUSING TSH EXCESS
Question for the Pathologist: Is the adenoma a thyrotroph
adenoma?
Clinical Relevance: Not all pituitary adenomas causing
TSH excess are thyrotroph adenomas “TSHomas”; however,
most adenomas leading to excess TSH are as a group
aggressive tumours.
Most physicians link central hyperthyroidism to a “TSHoma.
In fact, silent subtype III adenomas (monomorphous
12
Turkish Journal of Pathology GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
Figure 5: Crooke’s hyaline change of the nontumorus corticotrophs. A) While this cellular alteration can easily be identied on
haematoxylin and eosin-stained slides (arrows). B) PAS along with ACTH and low molecular weight keratin is used to highlight this
nding; the translocation of ACTH containing granules to the cell membrane and juxtanuclear region is accompanied by a ring like
(arrows) low molecular weight keratin expression.
A B
Figure 6: Crooke cell adenoma. A) ese rare tumors are oen invasive pituitary adenomas (arrows) and B) display a predominant
Crooke’s hyaline change.
A B
plurihormonal Pit-1 lineage adenomas), unusual
plurihormonal adenomas, and thyrotroph adenomas can
all result in TSH excess. TSH expressing adenomas are
characteristically invasive brotic macroadenomas that
present with mass eect and central hyperthyroidism (73-
76). Not surprisingly, residual/recurrent disease is frequent;
of additional note, recurrences do not seem to correlated
with the Ki67 labeling index that reects proliferation
(73). Fortunately, for tumours that are not curable with
surgery, somatostatin analogues have proven to be eective
in the management of hyperthyroidism and in some cases
treatment with these agents also results in tumour shrinkage
(76,77).
F. THYROTROPH HYPERPLASIA MIMICKING
A PITUITARY ADENOMA: AN EXAMPLE OF A
PREVENTABLE CLINICAL ERROR
Questions for the Clinician: Does the patient with a
diusely enlarged pituitary have baseline pituitary function
tests that rule out primary hypothyroidism?
13
Turkish Journal of Pathology
GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
Clinical Relevance: yrotroph hyperplasia resolves with
treatment of primary hypothyroidism
Unfortunately, and as previously reported (78-80), we have
seen surgically resected pituitaries containing thyrotroph
hyperplasia with or without lactotroph hyperplasia due to
longstanding primary hypothyroidism. Diuse pituitary
enlargement (with or without mass eect) associated
with mild hyperprolactinemia (due to TRH stimulation
of lactotrophs or stalk eect) may be misdiagnosed as a
prolactinoma (78,79). is underscores the relevance of
complete baseline anterior pituitary function assessment
for all patients with a suspected pituitary macroadenoma.
It is the clinician and not the pathologist who should make
the diagnosis of hypothyroidism. Radiology can be helpful
as well, since hyperplastic pituitaries show symmetrical
enlargement with no localized gadolinium enhancement
that distinguishes non-tumorous from adenomatous
adenohypophysis. With appropriate treatment of the
primary hypothyroidism, thyrotroph and lactotroph
hyperplasia regress, requiring months to up to two years,
but ultimately magnetic resonance imaging of the pituitary
returns to normal (81, 82).
G. PITUITARY CARCINOMA
Questions for the Pathologist and the Clinician: What
denes a pituitary carcinoma? Where does an atypical
adenoma stand in this spectrum? What are the potential
treatments for pituitary carcinoma?
ere are several factors that support the belief that pituitary
carcinomas arise mainly from transformed adenomas,
namely, the initial presentation of pituitary carcinomas
as aggressive adenomas, the generally long time required
for the progression to carcinoma development, and the
progressive accumulation of genetic abnormalities (83).
However, it is important to emphasize that the diagnosis of
pituitary carcinoma is not based on morphologic criteria,
instead it is established by the identication of metastatic
disease (Figure 7), which may be cerebrospinal or systemic
(84).
e categorization of an adenoma as “atypical” does not
predict malignant behaviour. Furthermore, pituitary
adenomas with invasive growth do not necessarily exhibit
a high proliferation rate as determined by the Ki67 labeling
index. erefore, an approach that integrates accurate
subtyping of the pituitary adenoma, biomarkers such as Ki67
and p53, intraoperative and radiologic ndings of invasion,
and response to therapy is warranted. Trouillas et al. (85)
have shown that pituitary adenomas that are classied based
on clinicopathologic features as “invasive and proliferative
have a probability of tumour persistence that is 25 times
higher than “non-invasive and non-proliferative” tumours.
Furthermore, the probability of recurrence was 12 times
higher in the “invasive and proliferative group.
Unfortunately, eective therapies for pituitary carcinomas
and recurrent invasive macroadenomas resistant to
conventional modalities are still lacking. However,
temozolomide, an O6 and N7 guanine-alkylating agent
approved for the treatment of glioblastomas and anaplastic
astrocyomas (86), has emerged as a therapeutic option
in those settings. It is unclear which individuals are most
likely to benet from the use of temozolomide but given its
mechanism of action, it would seem logical to assume that
patients with a faulty DNA repair enzymes would be better
candidates for treatment. Nevertheless, studies evaluating
O6-alkylguanine DNA alkyltransferase (MGMT) expression
have yielded inconsistent results. Perhaps, rather than the
qualitative observation of low or high MGMT expression,
a more quantitative approach will provide more reliable
information; in this regard, a recent study has shown
that MGMT staining below 50% is associated with a high
likelihood of treatment response (87).
Figure 7: Pituitary carcinoma. e magnetic resonance image
shows a metastatic focus in the cerebellopontine angle identied
5 years aer pituitary adenoma diagnosis.
14
Turkish Journal of Pathology GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
CONCLUSION
Accurate subtyping of pituitary adenomas allows for
diagnostic conrmation of clinically suspected disease
conditions. Furthermore, it allows for the identication of
patients that may be at higher risk of recurrent disease and
in some circumstances may dictate therapeutic responses.
Pathologists play an essential role in the multidisciplinary
endocrine oncology team by accurately classifying pituitary
tumours as standard practice.
CONFLICT OF INTEREST
e authors have declared no conict of interest.
REFERENCES
1. Al-Brahim NY, Asa SL. My approach to pathology of the pituitary
gland. J Clin Pathol. 2006;59:1245-53.
2. Asa SL, Ezzat S. e pathogenesis of pituitary tumors. Annu Rev
Pathol. 2009;4:97-126.
3. Asa SL. Tumors of the pituitary gland. Washington, DC:Armed
Forces Institute of Pathology. 2011
4. Mete O, Ezzat S, Asa SL. Biomarkers of aggressive pituitary
adenomas. J Mol Endocrinol. 2012;49:R69-78.
5. Mete O, Asa SL. Clinicopathological correlations in pituitary
adenomas. Brain Pathol. 2012;22:443-53.
6. Nose V, Ezzat S, Horvath E, Kovacs K, Laws ER, Lloyd R, Lopes
MB, Asa SL. Protocol for the examination of specimens from
patients with primary pituitary tumors. Arch Pathol Lab Med.
2011;135:640-6.
7. Yamada S, Ohyama K, Taguchi M, Takeshita A, Morita K, Takano
K, Sano T. A study of the correlation between morphological
ndings and biological activities in clinically nonfunctioning
pituitary adenomas. Neurosurgery. 2007;61:580-4; discussion
584-5.
8. Erickson D, Scheithauer B, Atkinson J, Horvath E, Kovacs K,
Lloyd RV, Young WF Jr. Silent subtype 3 pituitary adenoma: A
clinicopathologic analysis of the Mayo Clinic experience. Clin
Endocrinol (Oxf). 2009;71:92-9.
9. Nishioka H, Inoshita N, Mete O, Asa S, Hayashi K, Takeshita A,
Fukuhara N, Yamaguchi-Okada M, Takeuchi Y, Yamada S. e
crucial role of transcription factors in the accurate diagnosis of
nonfunctioning pituitary adenomas. Submitted.
10. Balogun JA, Monsalves E, Juraschka K, Parvez K, Kucharczyk W,
Mete O, Gentili F, Zadeh G. Null cell adenomas of the pituitary
gland: An institutional review of their clinical imaging and
behavioral characteristics. Endocr Pathol. 2015;26:63-70.
11. Horvath E, Kovacs K, Killinger DW, Smyth HS, Platts ME, Singer
W. Silent corticotropic adenomas of the human pituitary gland:
A histologic, immunocytologic, and ultrastructural study. Am J
Pathol. 1980;98:617-38.
12. Scheithauer BW, Jaap AJ, Horvath E, Kovacs K, Lloyd RV, Meyer
FB, Laws ER Jr., Young WF Jr. Clinically silent corticotroph
tumors of the pituitary gland. Neurosurgery. 2000;47:723-9;
discussion 729-30.
13. Cooper O, Ben-Shlomo A, Bonert V, Bannykh S, Mirocha J,
Melmed S. Silent corticogonadotroph adenomas: Clinical and
cellular characteristics and long-term outcomes. Horm Cancer.
2010;1:80-92.
14. Alahmadi H, Lee D, Wilson JR, Hayhurst C, Mete O, Gentili F, Asa
SL, Zadeh G. Clinical features of silent corticotroph adenomas.
Acta Neurochir (Wien). 2012;154:1493-8.
15. Cho HY, Cho SW, Kim SW, Shin CS, Park KS, Kim SY. Silent
corticotroph adenomas have unique recurrence characteristics
compared with other nonfunctioning pituitary adenomas. Clin
Endocrinol (Oxf). 2010;72:648-53.
16. Mete O, Hayhurst C, Alahmadi H, Monsalves E, Gucer H,
Gentili F, Ezzat S, Asa SL, Zadeh G. e role of mediators of cell
invasiveness, motility, and migration in the pathogenesis of silent
corticotroph adenomas. Endocr Pathol. 2013;24:191-8.
17. Asa SL. Practical pituitary pathology: What does the pathologist
need to know? Arch Pathol Lab Med. 2008;132:1231-40.
18. Mete O, Asa SL. erapeutic implications of accurate classication
of pituitary adenomas.Semin Diagn Pathol. 2013;30:158-64.
19. Felix IA, Horvath E, Kovacs K. Massive Crookes hyalinization in
corticotroph cell adenomas of the human pituitary. A histological,
immunocytological, and electron microscopic study of three
cases. Acta Neurochir (Wien). 1981;58:235-43.
20. George DH, Scheithauer BW, Kovacs K, Horvath E, Young WF Jr,
Lloyd RV, Meyer FB. Crooke’s cell adenoma of the pituitary: An
aggressive variant of corticotroph adenoma. Am J Surg Pathol.
2003;27:1330-6.
21. Horvath E, Kovacs K, Smyth HS, Killinger DW, Scheithauer
BW, Randall R, Laws ER Jr, Singer W. A novel type of pituitary
adenoma: Morphological features and clinical correlations. J Clin
Endocrinol Metab. 1988;66:1111-8.
22. Horvath E, Kovacs K, Smyth HS, Cusimano M, Singer W. Silent
adenoma subtype 3 of the pituitary--immunohistochemical and
ultrastructural classication: A review of 29 cases. Ultrastruct
Pathol. 2005;29:511-24.
23. Mete O, Gomez-Hernazdez K, Ezzat S, Asa S. Pituitary Silent
Subtype III Adenomas are not Always Silent and Represent Pit-
1 Lineage Adenomas. Modern Pathology. 2015;28(S2):138A.
(Abstract no: 540)
24. Cheng S, Al-Agha R, Araujo PB, Serri O, S LA, Ezzat S. Metabolic
glucose status and pituitary pathology portend therapeutic
outcomes in acromegaly. PLoS One. 2013;8:e73543.
25. Andreeva M, Georgiev T, Bosadjieva E, Karagiosov L, Karagiosov
K, Tomova A, Matrosov P, Kirilov G, Diankov L, Christova T.
A clinicomorphological study of pituitary tumors associated
with the clinical syndrome of acromegaly. Exp Clin Endocrinol.
1991;98:223-7.
26. Maheshwari HG, Prezant TR, Herman-Bonert V, Shahinian H,
Kovacs K, Melmed S. Long-acting peptidomimergic control of
gigantism caused by pituitary acidophilic stem cell adenoma. J
Clin Endocrinol Metab. 2000;85:3409-16.
27. Horvath E, Kovacs K, Singer W, Smyth HS, Killinger DW, Erzin C,
Weiss MH. Acidophil stem cell adenoma of the human pituitary:
Clinicopathologic analysis of 15 cases. Cancer. 1981;47:761-71.
15
Turkish Journal of Pathology
GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
28. Lim JS, Ku CR, Lee MK, Kim TS, Kim SH, Lee EJ. A case of
fugitive acromegaly, initially presented as invasive prolactinoma.
Endocrine. 2010;38:1-5.
29. Mete O, Gomez-Hernandez K, Ezzat S, Asa S. Clinicopathologic
Features of Pituitary Acidophil Stem Cell Adenomas. Modern
Pathology. 2015;28(S2):138A. (Abstract no: 539).
30. Molitch ME. Pharmacologic resistance in prolactinoma patients.
Pituitary. 2005;8:43-52.
31. Vroonen L, Jarain-Rea ML, Petrossians P, Tamagno G, Chanson
P, Vilar L, Borson-Chazot F, Naves LA, Brue T, Gatta B, Delemer
B, Ciccarelli E, Beck-Peccoz P, Caron P, Daly AF, Beckers A.
Prolactinomas resistant to standard doses of cabergoline: A
multicenter study of 92 patients. Eur J Endocrinol. 2012;167:651-
62.
32. Brue T, Pellegrini I, Priou A, Morange I, Jaquet P. Prolactinomas
and resistance to dopamine agonists. Horm Res. 1992;38:84-9.
33. Huang C, Ezzat S, Asa SL, Hamilton J. Dopaminergic resistant
prolactinomas in the peripubertal population. J Pediatr
Endocrinol Metab. 2006;19:951-3.
34. Asa SL, Kovacs K, Horvath E, Singer W, Smyth HS. Hormone
secretion in vitro by plurihormonal pituitary adenomas of the
acidophil cell line. J Clin Endocrinol Metab. 1992;75:68-75.
35. Horvath E, Kovacs K, Singer W, Ezrin C, Kerenyi NA. Acidophil
stem cell adenoma of the human pituitary. Arch Pathol Lab Med.
1977;101:594-9.
36. Trouillas J, Labat-Moleur F, Sturm N, Kujas M, Heymann MF,
Figarella-Branger D, Patey M, Mazucca M, Decullier E, Verges B,
Chabre O, Calender A, Groupe d’etudes des Tumeurs E. Pituitary
tumors and hyperplasia in multiple endocrine neoplasia type 1
syndrome (MEN1): A case-control study in a series of 77 patients
versus 2509 non-MEN1 patients. Am J Surg Pathol. 2008;32:534-
43.
37. Verges B, Boureille F, Goudet P, Murat A, Beckers A, Sassolas
G, Cougard P, Chambe B, Montvernay C, Calender A. Pituitary
disease in MEN type 1 (MEN1): Data from the France-Belgium
MEN1 multicenter study. J Clin Endocrinol Metab. 2002;87:457-
65.
38. Salenave S, Ancelle D, Bahougne T, Raverot G, Kamenicky
P, Bouligand J, Guiochon-Mantel A, Linglart A, Souchon PF,
Nicolino M, Young J, Borson-Chazot F, Delemer B, Chanson
P. Macroprolactinomas in children and adolescents: Factors
associated with the response to treatment in 77 patients. J Clin
Endocrinol Metab. 2015;100:1177-86.
39. Ezzat S, Kontogeorgos G, Redelmeier DA, Horvath E, Harris AG,
Kovacs K. In vivo responsiveness of morphological variants of
growth hormone-producing pituitary adenomas to octreotide.
Eur J Endocrinol. 1995;133:686-90
40. Bhayana S, Booth GL, Asa SL, Kovacs K, Ezzat S. e implication
of somatotroph adenoma phenotype to somatostatin analog
responsiveness in acromegaly. J Clin Endocrinol Metab.
2005;90:6290-5.
41. Bakhtiar Y, Hirano H, Arita K, Yunoue S, Fujio S, Tominaga
A, Sakoguchi T, Sugiyama K, Kurisu K, Yasufuku-Takano J,
Takano K. Relationship between cytokeratin staining patterns
and clinico-pathological features in somatotropinomae. J Clin
Endocrinol Metab. 2005;90:6290-5.
42. Fougner SL, Casar-Borota O, Heck A, Berg JP, Bollerslev J.
Adenoma granulation pattern correlates with clinical variables
and eect of somatostatin analogue treatment in a large series
of patients with acromegaly. Clin Endocrinol (Oxf ). 2012;76:96-
102.
43. Mazal PR, Czech T, Sedivy R, Aichholzer M, Wanschitz J, Klupp
N, Budka H. Prognostic relevance of intracytoplasmic cytokeratin
pattern, hormone expression prole, and cell proliferation in
pituitary adenomas of akromegalic patients. Clin Neuropathol.
2001;20:163-71.
44. Obari A, Sano T, Ohyama K, Kudo E, Qian ZR, Yoneda A, Rayhan
N, Mustazur Rahman M, Yamada S. Clinicopathological features
of growth hormone-producing pituitary adenomas: Dierence
among various types dened by cytokeratin distribution pattern
including a transitional form. Endocr Pathol. 2008;19:82-91.
45. Larkin S, Reddy R, Karavitaki N, Cudlip S, Wass J, Ansorge O.
Granulation pattern, but not GSP or GHR mutation, is associated
with clinical characteristics in somatostatin-naive patients with
somatotroph adenomas. Eur J Endocrinol. 2013;168:491-9.
46. Kato M, Inoshita N, Sugiyama T, Tani Y, Shichiri M, Sano T,
Yamada S, Hirata Y. Dierential expression of genes related to
drug responsiveness between sparsely and densely granulated
somatotroph adenomas. Endocr J. 2012;59:221-8.
47. Mayr B, Buslei R, eodoropoulou M, Stalla GK, Buchfelder M,
Scho C. Molecular and functional properties of densely and
sparsely granulated GH-producing pituitary adenomas. Eur J
Endocrinol. 2013;169:391-400.
48. Spada A, Arosio M, Bochicchio D, Bazzoni N, Vallar L, Bassetti
M, Faglia G. Clinical, biochemical, and morphological correlates
in patients bearing growth hormone-secreting pituitary tumors
with or without constitutively active adenylyl cyclase. J Clin
Endocrinol Metab. 1990;71:1421-6.
49. Freda PU, Chung WK, Matsuoka N, Walsh JE, Kanibir MN,
Kleinman G, Wang Y, Bruce JN, Post KD. Analysis of GNAS
mutations in 60 growth hormone secreting pituitary tumors:
Correlation with clinical and pathological characteristics and
surgical outcome based on highly sensitive GH and IGF-I criteria
for remission. Pituitary. 2007;10:275-82.
50. Leontiou CA, Gueorguiev M, van der Spuy J, Quinton R, Lolli
F, Hassan S, Chahal HS, Igreja SC, Jordan S, Rowe J, Stolbrink
M, Christian HC, Wray J, Bishop-Bailey D, Berney DM, Wass
JA, Popovic V, Ribeiro-Oliveira A, Jr., Gadelha MR, Monson JP,
Akker SA, Davis JR, Clayton RN, Yoshimoto K, Iwata T, Matsuno
A, Eguchi K, Musat M, Flanagan D, Peters G, Bolger GB, Chapple
JP, Frohman LA, Grossman AB, Korbonits M. e role of the
aryl hydrocarbon receptor-interacting protein gene in familial
and sporadic pituitary adenomas.J Clin Endocrinol Metab.
2008;93:2390-401.
16
Turkish Journal of Pathology GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
51. Daly AF, Tichomirowa MA, Petrossians P, Heliovaara E, Jarain-
Rea ML, Barlier A, Naves LA, Ebeling T, Karhu A, Raappana
A, Cazabat L, De Menis E, Montanana CF, Raverot G, Weil RJ,
Sane T, Maiter D, Neggers S, Yaneva M, Tabarin A, Verrua E,
Eloranta E, Murat A, Vierimaa O, Salmela PI, Emy P, Toledo
RA, Sabate MI, Villa C, Popelier M, Salvatori R, Jennings J,
Longas AF, Labarta Aizpun JI, Georgitsi M, Paschke R, Ronchi
C, Valimaki M, Saloranta C, De Herder W, Cozzi R, Guitelman
M, Magri F, Lagonigro MS, Halaby G, Corman V, Hagelstein MT,
Vanbellinghen JF, Barra GB, Gimenez-Roqueplo AP, Cameron
FJ, Borson-Chazot F, Holdaway I, Toledo SP, Stalla GK, Spada A,
Zacharieva S, Bertherat J, Brue T, Bours V, Chanson P, Aaltonen
LA, Beckers A. Clinical characteristics and therapeutic responses
in patients with germ-line AIP mutations and pituitary adenomas:
An international collaborative study. J Clin Endocrinol Metab.
2010;95:E373-83.
52. Denes J, Kasuki L, Trivellin G, Colli LM, Takiya CM, Stiles CE,
Barry S, de Castro M, Gadelha MR, Korbonits M. Regulation
of aryl hydrocarbon receptor interacting protein (AIP) protein
expression by MiR-34a in sporadic somatotropinomas. PLoS
One. 2015;10:e0117107.
53. Horvath E, Kovacs K, Killinger DW, Smyth HS, Weiss MH, Ezrin
C. Mammosomatotroph cell adenoma of the human pituitary: A
morphologic entity. Virchows Arch A Pathol Anat Histopathol.
1983;398:277-89.
54. Robert F, Pelletier G, Serri O, Hardy J. Mixed growth hormone
and prolactin-secreting human pituitary adenomas: A pathologic,
immunocytochemical, ultrastructural, and immunoelectron
microscopic study. Hum Pathol. 1988;19:1327-34.
55. Sherlock M, Fernandez-Rodriguez E, Alonso AA, Reulen RC,
Ayuk J, Clayton RN, Holder G, Sheppard MC, Bates A, Stewart
PM. Medical therapy in patients with acromegaly: Predictors of
response and comparison of ecacy of dopamine agonists and
somatostatin analogues. J Clin Endocrinol Metab. 2009;94:1255-
63.
56. Felix IA, Horvath E, Kovacs K, Smyth HS, Killinger DW, Vale
J. Mammosomatotroph adenoma of the pituitary associated
with gigantism and hyperprolactinemia. A morphological study
including immunoelectron microscopy. Acta Neuropathol.
1986;71:76-82.
57. Dubuis JM, Deal CL, Drews RT, Goodyer CG, Lagace G, Asa
SL, Van Vliet G, Collu R. Mammosomatotroph adenoma
causing gigantism in an 8-year old boy: A possible pathogenetic
mechanism. Clin Endocrinol (Oxf). 1995;42:539-49.
58. Pack SD, Kirschner LS, Pak E, Zhuang Z, Carney JA, Stratakis CA.
Genetic and histologic studies of somatomammotropic pituitary
tumors in patients with the “complex of spotty skin pigmentation,
myxomas, endocrine overactivity and schwannomas” (Carney
complex). J Clin Endocrinol Metab. 2000;85:3860-5.
59. Kurtkaya-Yapicier O, Scheithauer BW, Carney JA, Kovacs
K, Horvath E, Stratakis CA, Vidal S, Vella A, Young WF, Jr.,
Atkinson JL, Lloyd RV, Kontogeorgos G: Pituitary adenoma in
Carney complex: An immunohistochemical, ultrastructural,
and immunoelectron microscopic study. Ultrastruct Pathol.
2002;26:345-53.
60. Boikos SA, Stratakis CA. Pituitary pathology in patients with
Carney Complex: Growth-hormone producing hyperplasia or
tumors and their association with other abnormalities. Pituitary.
2006;9:203-9.
61. Kovacs K, Horvath E, orner MO, Rogol AD.
Mammosomatotroph hyperplasia associated with acromegaly
and hyperprolactinemia in a patient with the McCune-Albright
syndrome. A histologic, immunocytologic and ultrastructural
study of the surgically-removed adenohypophysis. Virchows
Arch A Pathol Anat Histopathol. 1984;403:77-86.
62. Vortmeyer AO, Glasker S, Mehta GU, Abu-Asab MS, Smith JH,
Zhuang Z, Collins MT, Oldeld EH. Somatic GNAS mutation
causes widespread and diuse pituitary disease in acromegalic
patients with McCune-Albright syndrome. J Clin Endocrinol
Metab. 2012;97:2404-13.
63. Moran A, Asa SL, Kovacs K, Horvath E, Singer W, Sagman U,
Reubi JC, Wilson CB, Larson R, Pescovitz OH. Gigantism due
to pituitary mammosomatotroph hyperplasia. N Engl J Med.
1990;323:322-7.
64. Glasker S, Vortmeyer AO, Laerty AR, Hofman PL, Li J, Weil RJ,
Zhuang Z, Oldeld EH. Hereditary pituitary hyperplasia with
infantile gigantism. J Clin Endocrinol Metab. 2011;96:E2078-87.
65. Puchner MJ, Ludecke DK, Saeger W, Riedel M, Asa SL.
Gangliocytomas of the sellar region--a review. Exp Clin
Endocrinol Diabetes. 1995;103:129-49.
66. Sano T, Asa SL, Kovacs K. Growth hormone-releasing hormone-
producing tumors: Clinical, biochemical, and morphological
manifestations. Endocr Rev. 1988;9:357-73.
67. Garby L, Caron P, Claustrat F, Chanson P, Tabarin A, Rohmer V,
Arnault G, Bonnet F, Chabre O, Christin-Maitre S, du-Boullay
H, Murat A, Nakib I, Sadoul JL, Sassolas G, Claustrat B, Raverot
G, Borson-Chazot F, Group GTE. Clinical characteristics and
outcome of acromegaly induced by ectopic secretion of growth
hormone-releasing hormone (GHRH): A French nationwide
series of 21 cases. J Clin Endocrinol Metab. 2012;97:2093-104.
68. Bochicchio D, Losa M, Buchfelder M. Factors inuencing the
immediate and late outcome of Cushing’s disease treated by
transsphenoidal surgery: A retrospective study by the European
Cushing’s Disease Survey Group. J Clin Endocrinol Metab.
1995;80:3114-20.
69. Chee GH, Mathias DB, James RA, Kendall-Taylor P.
Transsphenoidal pituitary surgery in Cushing’s disease: Can we
predict outcome? Clin Endocrinol (Oxf). 2001;54:617-26.
70. Carey RM, Varma SK, Drake CR, Jr., orner MO, Kovacs K,
Rivier J, Vale W. Ectopic secretion of corticotropin-releasing
factor as a cause of Cushings syndrome. A clinical, morphologic,
and biochemical study. N Engl J Med. 1984;311:13-20.
71. McNicol AM. Patterns of corticotropic cells in the adult human
pituitary in Cushing’s disease. Diagn Histopathol. 1981;4:335-41.
72. Kovacs K, Horvath E, Coire C, Cusimano M, Smyth H,
Scheithauer BW, Lloyd RV. Pituitary corticotroph hyperplasia
preceding adenoma in a patient with Nelsons syndrome. Clin
Neuropathol. 2006;25:74-80.
17
Turkish Journal of Pathology
GOMEZHERNANDEZ K et al: Pituitary Adenomas
Vol. 31, Suppl, 2015; Page 4-17
73. Kirkman MA, Jaunmuktane Z, Brandner S, Khan AA, Powell M,
Baldeweg SE. Active and silent thyroid-stimulating hormone-
expressing pituitary adenomas: Presenting symptoms, treatment,
outcomes, and recurrence. World Neurosurg. 2014;82:1224-31.
74. Clarke MJ, Erickson D, Castro MR, Atkinson JL. yroid-
stimulating hormone pituitary adenomas. J Neurosurg.
2008;109:17-22.
75. Brucker-Davis F, Oldeld EH, Skarulis MC, Doppman JL,
Weintraub BD. yrotropin-secreting pituitary tumors:
Diagnostic criteria, thyroid hormone sensitivity, and treatment
outcome in 25 patients followed at the National Institutes of
Health.J Clin Endocrinol Metab. 1999;84:476-86.
76. Socin HV, Chanson P, Delemer B, Tabarin A, Rohmer V, Mockel
J, Stevenaert A, Beckers A. e changing spectrum of TSH-
secreting pituitary adenomas: Diagnosis and management in 43
patients. Eur J Endocrinol. 2003;148:433-42.
77. Beck-Peccoz P, Persani L, Mannavola D, Campi I. Pituitary
tumours: TSH-secreting adenomas. Best Pract Res Clin
Endocrinol Metab. 2009;23:597-606.
78. Khalil A, Kovacs K, Sima AA, Burrow GN, Horvath E. Pituitary
thyrotroph hyperplasia mimicking prolactin-secreting adenoma.
J Endocrinol Invest. 1984;7:399-404.
79. Pioro EP, Scheithauer BW, Laws ER Jr, Randall RV, Kovacs KT,
Horvath E. Combined thyrotroph and lactotroph cell hyperplasia
simulating prolactin-secreting pituitary adenoma in long-
standing primary hypothyroidism. Surg Neurol. 1988;29:218-26.
80. Alkhani AM, Cusimano M, Kovacs K, Bilbao JM, Horvath
E, Singer W. Cytology of pituitary thyrotroph hyperplasia in
protracted primary hypothyroidism. Pituitary. 1999;1:291-5.
81. Passeri E, Tufano A, Locatelli M, Lania AG, Ambrosi B, Corbetta
S. Large pituitary hyperplasia in severe primary hypothyroidism.
J Clin Endocrinol Metab. 2011;96:22-3.
82. Johnston PC, Ellis PK, Hunter SJ. yrotroph hyperplasia.
Postgrad Med J. 2014;90:56-7.
83. Kaltsas GA, Nomikos P, Kontogeorgos G, Buchfelder M,
Grossman AB. Clinical review: Diagnosis and management of
pituitary carcinomas. J Clin Endocrinol Metab. 2005;90:3089-99.
84. DeLellis RA. Pathology and genetics of tumours of endocrine
organs. Lyon:IARC Press.2004.
85. Trouillas J, Roy P, Sturm N, Dantony E, Cortet-Rudelli C, Viennet
G, Bonneville JF, Assaker R, Auger C, Brue T, Cornelius A,
Dufour H, Jouanneau E, Francois P, Galland F, Mougel F, Chapuis
F, Villeneuve L, Maurage CA, Figarella-Branger D, Raverot G,
members of H, Barlier A, Bernier M, Bonnet F, Borson-Chazot
F, Brassier G, Caulet-Maugendre S, Chabre O, Chanson P, Cottier
JF, Delemer B, Delgrange E, Di Tommaso L, Eimer S, Gaillard
S, Jan M, Girard JJ, Lapras V, Loiseau H, Passagia JG, Patey M,
Penfornis A, Poirier JY, Perrin G, Tabarin A. A new prognostic
clinicopathological classication of pituitary adenomas: A
multicentric case-control study of 410 patients with 8 years post-
operative follow-up. Acta Neuropathol. 2013;126:123-35.
86. FDA. Temozolomide FDA label. 2011
87. Bengtsson D, Schroder HD, Andersen M, Maiter D, Berinder K,
Feldt Rasmussen U, Rasmussen AK, Johannsson G, Hoybye C,
van der Lely AJ, Petersson M, Ragnarsson O, Burman P. Long-
term outcome and MGMT as a predictive marker in 24 patients
with atypical pituitary adenomas and pituitary carcinomas
given treatment with temozolomide. J Clin Endocrinol Metab.
2015;100:1689-98.
... Los adenomas silentes tipo 3 son monomorfos y plurihormonales, tienden a ser macroadenomas invasivos o gigantes y se presentan a edades más tempranas, el tiempo de recurrencia o libre de tumor puede ser baja, en aquellos con receptores para somatostatina el uso de octreotide ha mostrado estabilidad tumoral 4,7 . ...
... Esto puede indicar una opción de tratamiento más agresivo para este tipo de tumores corticotropos frecuentes en nuestro grupo, ya que según lo reportado en la literatura donde tienen tendencia a una mayor invasión al seno cavernoso y mayores tasas de progresión [20][21][22] , considerando incluso el manejo médico coadyuvante dependiendo del tipo de receptores que presente el tumor 18,23,24 . Dada la relevancia en nuestra población de los adenomas corticotróficos silentes, es necesario destacar que éstos a su vez se subdividen en 3 subtipos, donde los tipo 1 tienen una estructura idéntica a los adenomas corticotrópico funcionantes, tipo 2 que son similares a los funcionantes pero con gránulos escasos y ultraestructura indiferenciada y son clínicamente más agresivos, tienen un riesgo mayor de apoplejía pituitaria, una mayor tasa de recurrencia y mayor invasividad; por otra parte, los adenomas silentes tipo 3 son más raros, plurihormonales (con demostración positiva para una o más líneas hormonales), con atipia citológica variable y un Ki-67 de hasta el 9% 4, 11 , por lo cual la radioterapia es requerida en un número importante de casos 4,7,25 . ...
Article
Full-text available
Resumen: Los adenomas pituitarios son lesiones poco comunes, que pueden surgir de cualquier tipo celular de la hipófisis anterior. Algunos ade-nomas pueden causar manifestaciones clínicas asociadas a la secreción excesiva de sus productos hormonales, mientras que los adenomas ¨no fun-cionales¨ o ¨silentes¨ no lo hacen, lo cual puede llevar a un diagnóstico tardío o encontrarse como hallazgo incidental en un estudio radiológico ce-rebral.
... However, growth hormone (GH) mRNA and protein expression was stimulated by transfecting the Pit-1 gene into AtT-20 cells [181]. Thyrotropic adenomas express Pit-1 in a similar manner to other acidophilic neuroendocrine tumors [182][183][184][185]. ...
Article
Pituitary adenoma (PA) is the third most common primary intracranial tumor in terms of overall disease incidence. Although they are benign tumors, they can have a variety of clinical symptoms, but are mostly asymptomatic, which often leads to diagnosis at an advanced stage when surgical intervention is ineffective. Earlier identification of PA could reduce morbidity and allow better clinical management of the affected patients. Non-coding RNAs (ncRNAs) do not generally code for proteins, but can modulate biological processes at the post-transcriptional level through a variety of molecular mechanisms. An increased number of ncRNA expression profiles have been found in PAs. Therefore, understanding the expression patterns of different ncRNAs could be a promising method for developing non-invasive biomarkers. This review summarizes the expression patterns of dysregulated ncRNAs (microRNAs, long non-coding RNAs, and circular RNAs) involved in PA, which could one day serve as innovative biomarkers or therapeutic targets for the treatment of this neoplasia. We also discuss the potential molecular pathways by which the dysregulated ncRNAs could cause PA and affect its progression.
... CAM5.2 is a low molecular weight cytokeratin antibody clone that seems to react against CK18 65 . Based on the granulation pattern under CAM5.2 immunostaining, corticotropinomas can be classified into two subtypes 65,66 . Most tumors are of the densely granulated subtype and contain abundant secretory granules that appear as a diffuse and intense ACTH staining. ...
Article
Full-text available
Cushing's disease (CD) is the most common cause of endogenous hypercortisolemia. The clinical management of this condition is complex and entails multiple therapeutic strategies, treatment of chronic comorbidities, and lifelong surveillance for recurrences and complications. The identification of robust, practical, and reliable markers of disease behavior and prognosis could potentially allow for a tailored and cost-efficient management of each patient, as well as for a reduction of the medical procedure associated stress. For this purpose, multiple clinical, biochemical, imaging, histopathological, molecular, and genetic features have been evaluated over the years. Only a handful of them, however, have been sufficiently validated for their application in the routine care of patients with CD. This review summarizes the current status of the established and potential biomarkers of CD, bases for their use, proposed and/or established utility, as well as advantages and barriers for their implementation in the clinic. (REV INVEST CLIN. 2022;74(5):244-57)
... Therefore, the 2022 WHO Classification does not replace the key anatomic site-based information to be included in pathological diagnoses for these organs. For instance, pituitary NETs are subtyped based on cell lineage, cell type, and related characteristics combining the routine use of transcription factors and hormones as well as other biomarkers including keratins, and Ki67, which is regarded as a continuous variable [54,64,[92][93][94][95]. ...
Article
Full-text available
In this review, we detail the changes and the relevant features that are applied to neuroendocrine neoplasms (NENs) in the 2022 WHO Classification of Endocrine and Neuroendocrine Tumors. Using a question-and-answer approach, we discuss the consolidation of the nomenclature that distinguishes neuronal paragangliomas from epithelial neoplasms, which are divided into well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs). The criteria for these distinctions based on differentiation are outlined. NETs are generally (but not always) graded as G1, G2, and G3 based on proliferation, whereas NECs are by definition high grade; the importance of Ki67 as a tool for classification and grading is emphasized. The clinical relevance of proper classification is explained, and the importance of hormonal function is examined, including eutopic and ectopic hormone production. The tools available to pathologists for accurate classification include the conventional biomarkers of neuroendocrine lineage and differentiation, INSM1, synaptophysin, chromogranins, and somatostatin receptors (SSTRs), but also include transcription factors that can identify the site of origin of a metastatic lesion of unknown primary site, as well as hormones, enzymes, and keratins that play a role in functional and structural correlation. The recognition of highly proliferative, well-differentiated NETs has resulted in the need for biomarkers that can distinguish these G3 NETs from NECs, including stains to determine expression of SSTRs and those that can indicate the unique molecular pathogenetic alterations that underlie the distinction, for example, global loss of RB and aberrant p53 in pancreatic NECs compared with loss of ATRX, DAXX, and menin in pancreatic NETs. Other differential diagnoses are discussed with recommendations for biomarkers that can assist in correct classification, including the distinctions between epithelial and non-epithelial NENs that have allowed reclassification of epithelial NETs in the spine, in the duodenum, and in the middle ear; the first two may be composite tumors with neuronal and glial elements, and as this feature is integral to the duodenal lesion, it is now classified as composite gangliocytoma/neuroma and neuroendocrine tumor (CoGNET). The many other aspects of differential diagnosis are detailed with recommendations for biomarkers that can distinguish NENs from non-neuroendocrine lesions that can mimic their morphology. The concepts of mixed neuroendocrine and non-neuroendocrine (MiNEN) and amphicrine tumors are clarified with information about how to approach such lesions in routine practice. Theranostic biomarkers that assist patient management are reviewed. Given the significant proportion of NENs that are associated with germline mutations that predispose to this disease, we explain the role of the pathologist in identifying precursor lesions and applying molecular immunohistochemistry to guide genetic testing.
... Densely granulated corticotroph adenomas with large and abundant secretory granules on electron microscopy are usually functioning corticotroph adenomas with hypercortisolism and Cushing's symptoms. In contrast, sparsely granulated corticotroph adenomas with small and few secretory granules are silent corticotroph adenomas lacking hypercortisolism and Cushing's symptoms (7). Tumors that have a Ki-67 positive rate >3% were classified as atypical adenomas according to the previous WHO classification (8). ...
Article
Full-text available
Pasireotide, which has a high affinity for somatostatin receptor (SSTR) 5, has attracted attention as a new treatment for refractory Cushing's disease. The patient was a 28-year-old man. He had refractory Cushing's disease and underwent multiple surgeries, radiotherapy, and medication therapy. An examination of the adenoma by immunohistochemistry revealed a low SSTR5 expression. An USP8 mutation was not detected by reverse transcription polymerase chain reaction. Although we administered pasireotide, it was ineffective. While a further investigation is necessary, the analysis of SSTR5 expression may support the prediction of the efficiency of pasireotide for Cushing's disease. We report this case as a useful reference when considering whether or not to use pasireotide for refractory corticotroph adenomas.
Chapter
Overview Pituitary neoplasms represent a phenotypically and pathologically diverse family of tumors. Symptoms may derive from abnormal hormone production, compression of adjacent nervous system structures, and in rare cases, metastases. New cell lineage classification based on transcription factors brings new insights into pathogenesis and treatment. Transsphenoidal surgery represents a command well‐tolerated treatment for most pituitary adenomas; however, advances in radiation and chemotherapy have increased treatment options enhancing treatment efficacy and eligibility.
Article
Nonfunctioning pituitary neuroendocrine tumors (NF-PitNETs) are tumors that are not associated with clinical evidence of hormonal hypersecretion. According to the World Health Organization (WHO), there are some subtypes of PitNETs that exhibit more aggressive behavior than others. Among the types of potentially aggressive PitNETs, three are nonfunctional: silent sparsely granulated somatotropinomas, silent corticotropinomas, and poorly differentiated PIT-1 lineage tumors. Several biological markers have been investigated in NF-PitNETs. However, there is no single biomarker able to independently predict aggressive behavior in NF-PitNETs. Thus, a more complex and multidisciplinary proposal of a comprehensive definition of aggressive NF-PitNETs is necessary. Here, we suggest a combined and more complete criterion for the NF-PitNETs classification. We propose that aggressiveness is due to a multifactorial combination, and we emphasize the need to include new emerging markers that are involved in the aggressiveness of NF-PitNETs and the need to identify.
Article
In the Turkish saddle area, there is a wide variety of pathological processes, the vast majority of which present as tumors of various origins (up to 90%). For a clear morphological verification of the diagnosis, it is proposed to use a diagnostic algorithm that includes the stages of differential diagnosis of normal adenohypophysis and neurohypophysis with tumors in the anterior and posterior lobes of the pituitary gland, non-pituitary origin neoplasms, as well as with non-tumor pathological processes (inflammation, cystic masses, and hyperplasia). For morphological diagnosis, histochemical and immunohistochemical methods are recommended using various staining techniques (silver impregnation, periodic acid Schiff reaction) of tissue specimens and antibody panels (pituitary hormones, low-molecular cytokeratins, pituitary transcription factors, neuroendocrine markers, etc.).
Chapter
Although growth hormone (GH)–producing pituitary neuroendocrine tumors are by far the most common neoplastic cause of acromegaly and gigantism, other neoplastic causes exist, including hypothalamic tumors, which secrete eutopic GH-releasing hormone (GHRH) as well as a number of other tumor types throughout the body, which may secrete GHRH or GH in an ectopic fashion. This chapter is devoted to these less common but equally important entities and focuses on their clinical, radiological, biochemical, and pathological characteristics, in addition to diagnostic challenges that may be encountered when dealing with these interesting neoplasms.
Article
Full-text available
In an effort to improve the diagnosis of pituitary tumors, we propose a synoptic approach to pituitary pathology reporting that will provide clear information to endocrinologists, neurosurgeons, neuropathologists, and surgical pathologists to advance the diagnosis and classification of pituitary adenomas.
Article
Full-text available
Clinically nonfunctioning pituitary adenomas (NFAs) may be hormonally inactive tumors of differentiated cells, mainly not only gonadotroph adenomas (GAs) but also silent corticotroph adenomas (SCAs) and other differentiated silent adenomas. Recently, the use of transcription factors has been recommended to confirm cytodiffererentiation of these neoplasms. Our objective was to assess the clinical significance of the new classification system using transcription factors. Five hundred sixteen consecutive NFAs were studied retrospectively. They were initially classified based on hormone immunohistochemistry as follows: 119 hormone-negative adenomas (23.1 %), 300 GAs (58.1 %), 51 SCAs (9.9 %), and 46 other silent adenomas. The 119 hormone-negative adenomas were further evaluated for expression of transcription factors including steroidogenic factor-1 (SF-1), estrogen receptor-α (ERα), pituitary-specific transcription factor 1 (Pit-1), and t-box transcription factor (Tpit). One hundred thirteen of 119 (95 %) hormone-negative adenomas showed mutually exclusive lineage-specific differentiation as gonadotrophs (SF-1 positive), corticotrophs (Tpit positive), or somatotrophs/mammosomatotrophs/lactotrophs/thyrotrophs (Pit-1 positive) in 79 cases (66.4 %), 32 cases (26.9 %), and 2 cases, respectively. The 32 ACTH-negative and Tpit-positive adenomas had higher pro-opiomelanocortin mRNA expression levels compared with GAs (P = 0.0001) on quantitative real-time PCR. They showed a female preponderance (P < 0.0001) and were more frequently giant adenomas (P = 0.0028) associated with marked cavernous sinus invasion (P < 0.0001) compared with GAs. These clinical features were identical to those of the 51 ACTH-positive SCAs. Our results justify the complementary role of transcription factors in the precise classification of NFAs that can more accurately characterize biological behavior. Our data suggest that more than one quarter of hormone-negative adenomas are SCAs that share distinct clinicopathological features with ACTH-expressing SCAs.
Article
Full-text available
Patients with germline AIP mutations or low AIP protein expression have large, invasive somatotroph adenomas and poor response to somatostatin analogues (SSA). To study the mechanism of low AIP protein expression 31 sporadic somatotropinomas with low (n = 13) or high (n = 18) AIP protein expression were analyzed for expression of AIP messenger RNA (mRNA) and 11 microRNAs (miRNAs) predicted to bind the 3'UTR of AIP. Luciferase reporter assays of wild-type and deletion constructs of AIP-3'UTR were used to study the effect of the selected miRNAs in GH3 cells. Endogenous AIP protein and mRNA levels were measured after miRNA over- and underexpression in HEK293 and GH3 cells. No significant difference was observed in AIP mRNA expression between tumors with low or high AIP protein expression suggesting post-transcriptional regulation. miR-34a was highly expressed in low AIP protein samples compared high AIP protein adenomas and miR-34a levels were inversely correlated with response to SSA therapy. miR-34a inhibited the luciferase-AIP-3'UTR construct, suggesting that miR-34a binds to AIP-3'UTR. Deletion mutants of the 3 different predicted binding sites in AIP-3'UTR identified the c.*6-30 site to be involved in miR-34a's activity. miR-34a overexpression in HEK293 and GH3 cells resulted in inhibition of endogenous AIP protein expression. Low AIP protein expression is associated with high miR-34a expression. miR-34a can down-regulate AIP-protein but not RNA expression in vitro. miR-34a is a negative regulator of AIP-protein expression and could be responsible for the low AIP expression observed in somatotropinomas with an invasive phenotype and resistance to SSA.
Article
Full-text available
Context/objective: Locally aggressive pituitary tumors (LAPT) and pituitary carcinomas respond poorly to conventional therapy and cytotoxic drugs. Temozolomide (TMZ) is an oral alkylating drug with good tolerability, approved for treatment of malignant gliomas. The experience of its use in pituitary tumors is limited. Design and setting: We report on 24 patients with aggressive pituitary tumors (16 LAPTs, 8 carcinomas) treated with TMZ for a median of 6 months (range 1-23). Follow-up ranged from 4 to 91 months with a median of 32.5 months. 19/24 tumors were hormone secreting (PRL 9, ACTH 4, GH 4, GH/PRL 2). Ki-67 was 2-50% in LAPTs, and 5-80% in carcinomas. Main outcome: Response to TMZ and the association with tumor expression of O6-methylguanine DNA methyltransferase (MGMT), MLH1, MSH2, and MSH6, examined by immunohistochemistry. Results: Complete tumor regression occurred in two carcinomas and persisted at follow-up after 48 and 91 months, respectively. Partial regress of tumor mass ranging from 35% to 80% occurred in 5 LAPTs and 2 carcinomas. Another patient with LAPT had a 71% decrease in prolactin levels without change in tumor volume. Three LAPTs could not be evaluated. Median MGMT staining was 9% (5-20%) in responders vs 93% (50-100%) in nonresponders. Loss of MSH2 and MSH 6 was observed in a single patient who had a rapid development of resistance to TMZ. Conclusions: This study shows that TMZ is a valuable treatment option for patients with uncontrolled pituitary tumors. The data suggest that tumoral MGMT staining below 50% is associated with a high likelihood of treatment response.
Article
Full-text available
Background: Pituitary adenomas are rare in children and adolescents. The response of macroprolactinomas to dopamine agonists (DA) in this age group has been less extensively studied than in adults. Objective: We retrospectively analyzed data on a large cohort of young patients with macroprolactinomas. Patients and methods: Patients aged younger than 20 years at macroprolactinoma diagnosis and seen in three tertiary referral centers between 1983 and 2013 were studied by analyzing their clinical and genetic (AIP and MEN1) characteristics. Hormonal and tumoral responses to DA were analyzed, and the patients' status at their last visit, after a mean (±SD) follow-up of 8.2 ± 5.8 years, was assessed. Results: The cohort comprised 77 patients (26 males, 51 females). Mean age at diagnosis was 16.1 ± 2.5 years (range, 4.5-20 y). In both sexes, the most frequent revealing symptom was a pubertal disorder (49%), followed by visual problems (24%) and growth retardation (24%). Basal prolactin (PRL) levels and maximal tumor diameter were significantly higher in boys than in girls (7168 ng/mL, 202-40 168 vs 1433 ng/mL, 115-20 000, P = .002; and 33 ± 14 mm, 15-64 vs 19 ± 9 mm; 10-50, P < .001, respectively). PRL levels normalized in 74% of the patients treated with DA. A mutation of AIP or MEN1 was found in 14% of the patients. Factors associated with resistance to DA were young age, higher PRL levels, larger volume, and the presence of a MEN1 (but not an AIP) mutation. Conclusion: Macroprolactinomas are rare below the age of 20 years, mainly occurring in girls and during adolescence. Like adults, young patients are very sensitive to DA, which should therefore be considered the first-line treatment. DA resistance is associated with a higher PRL level and larger tumor size, both parameters being closely linked together. About 14% of these young patients have an AIP or MEN1 mutation, this latter being an independent predictor of DA resistance.
Article
The aim of the study was to establish if the null cell adenoma (NCA) forms a distinct subgroup with unique clinicopathological characteristics within the nonfunctioning pituitary adenoma group particularly in relation to the silent gonadotroph adenomas (SGAs). We identified 31 patients with the pathological diagnosis of NCA verified by routine histology and immunohistochemistry with distinct differentiation from SGAs by an established negative testing for SF-1 at the Toronto Western Hospital between December 2004 and August 2010. We reviewed their demographic data, clinical features, magnetic resonance imaging, and the histologic variables: MIB-1, FGFR4, and P27. We compared these to 63 SGAs identified within the same period. All the NCAs were macroadenomas with diameter ranging from 15-57 mm and tumor volumes between 1.95-53.5 mm(3). Preoperative cavernous sinus tumor growth was able to predict the presence of a residual after surgery (p = 0.023). Furthermore, preoperative cavernous sinus extension (p = 0.002) and negative P27 expression (p = 0.035) were able to independently predict the subsequent growth of the postoperative tumor residual. Comparing the NCA to SGA, we found that MIB-1 was higher in NCA (mean ± SD = 3.43 ± 2.76 %) compared to SGAs (mean ± SD = 2.49 ± 1.41 %) (p = 0.044). The preoperative and postoperative tumor volume doubling times (TVDTs) displayed a negative correlation in the SGA (r = -0.855, p = 0.002) while in the NCA, a positive correlation was evident (r = 0.718, p = 0.029). Our study suggests that the NCAs are a distinct group with differing behavioral characteristics from the SGAs. It also appears that the finding of cavernous sinus extension on preoperative imaging and a negative P27 expression on immunohistochemistry in NCAs may be valuable tools in predicting residual tumor growth which may impact on postoperative care.
Article
Objective Thyroid-stimulating hormone-(TSH-)expressing pituitary adenomas are a rare but important entity with a spectrum of clinical manifestations. There is currently no data to indicate whether a difference exists in the natural progression of active and silent TSH-expressing pituitary adenomas (defined by the presence or absence of clinical hyperthyroidism, respectively). Here we report our experience (including presenting symptoms, treatment and outcome) with managing both groups over eleven years – the largest single-centre data published to date. Methods We reviewed retrospectively all patients with histopathologically-proven TSH-expressing pituitary adenomas presenting to our centre between 2002 and 2012 inclusive. Data reviewed included clinical presentation, biochemical status, tumour size, management, histopathological results and long-term post-operative outcomes. Results 32 patients (16 males) were identified from a total of 902 operations for pituitary adenomas performed between 2002 and 2012. Mean follow-up was 6.7 years. A quarter (25%) of patients were clinically hyperthyroid at presentation. Visual disturbance was the commonest presenting complaint in 34%. All patients underwent transsphenoidal surgery. Thirty one percent of patients had a recurrence. The clinically active and silent TSH-expressing pituitary adenomas behaved in a similar manner with respect to recurrence rates. Conclusions TSH-expressing pituitary adenomas present with a wide clinical spectrum. Visual disturbance is common. Despite radiological evidence of clearance following surgery, and extended follow-up, they may still recur whether clinically ‘active’ or ‘silent’. Our data supports the need for close long-term follow-up of these patients.