SlideShare a Scribd company logo
1 of 14
Download to read offline
The new engl and jour nal of medicine
n engl j med 382;10  nejm.org  March 5, 2020 937
Review Article
From the Pituitary Center, Department
of Medicine, Cedars-Sinai Medical Center,
Los Angeles. Address reprint requests
to Dr. Melmed at Cedars-Sinai Medical
Center, 8700 Beverly Blvd., Rm. 2015, Los
Angeles, CA 90048.
N Engl J Med 2020;382:937-50.
DOI: 10.1056/NEJMra1810772
Copyright © 2020 Massachusetts Medical Society.
P
ituitary adenomas account for approximately 15% of intracra-
nial tumors.1
Management of these benign tumors requires diagnosis of the
specific intrasellar disease and comprehensive, multidisciplinary treatment
of local mass effects and peripheral endocrinopathies.2
Since tumors can produce
different hormones, their consequences and management vary widely.
Pathogenesis
Differentiated, hormone-expressing pituitary cell lineages may give rise to adeno-
mas, often coupled with autonomous hormone hypersecretion. Distinct hyper­
secretory syndromes depend on the cell of origin: corticotropin-secreting corti-
cotroph adenomas result in Cushing’s disease, growth hormone–secreting
somatotroph adenomas result in acromegaly, prolactin-secreting lactotroph adeno-
mas result in hyperprolactinemia, and thyrotropin-secreting thyrotroph adenomas
result in hyperthyroidism. Gonadotroph adenomas, which are typically nonsecreting,
lead to hypogonadism and often manifest incidentally as a sellar mass3-5
(Fig. 1).
Permissive hypothalamic hormone and paracrine proliferative signals lead to a
dysregulated pituitary cell cycle, with aneuploidy, chromosomal copy-number
variation, and cellular senescence restraining malignant transformation.6-8
Although
mutations in GNAS (the gene for the stimulatory alpha subunit of a guanine nucleo-
tide–binding protein [G-protein] that stimulates adenylate cyclase) and USP8 (the
gene for ubiquitin carboxyl terminal hydrolase 8, a ubiquitin-specific protease)
occur in a subgroup of nonfamilial growth hormone–secreting tumors and corti-
cotropin-secreting tumors, respectively,9
genetic evaluation of sporadic adenomas
is rarely helpful for management.
The prevalence of pituitary adenomas has increased to 115 cases per 100,000
population over the past several decades, probably as a result of enhanced aware-
ness and improved diagnostic imaging and hormone assays.10
The relative preva-
lence of prolactinomas (54 cases per 100,000 population) and nonfunctioning
adenomas (42 per 100,000) may reflect a reporting bias with respect to surgical
versus nonsurgical series, since most prolactinomas are treated medically and are
not captured in reports of surgical diagnoses.
Classification
Microadenomas are less than 10 mm in diameter. Regardless of the cell origin,
macroadenomas (≥10 mm) may impinge on critical parasellar vascular and neural
structures, with resultant visual-field defects, including bitemporal hemianopia
and decreased acuity, and headaches.5
Immunocytochemical evaluation of differ-
entiated, cell-specific pituitary transcription factors and hormones,11
as well as
clear biochemical, imaging, and clinical phenotypes, define tumor characteristics
Dan L. Longo, M.D., Editor
Pituitary-Tumor Endocrinopathies
Shlomo Melmed, M.D.​​
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10 nejm.org March 5, 2020938
The new engl and jour nal of medicine
and endocrine syndromes, allowing for indi-
vidualized treatment5,12
(Table 1).
Evaluation of a pituitary mass should include
magnetic resonance imaging (MRI) and visual-
field examination for accurate tumor localiza-
tion and assessment of local compressive mass
effects. Hormone hypersecretion should be as-
sessed to distinguish nonsecretory from secretory
tumors13
(Table 2), and pituitary-reserve function
should be tested.14
Approximately 30% of surgically resected
adenomas have persistent or progressive post-
operative growth for up to four decades or even
longer, with local invasion and an increased
percentage of cells positive for Ki-67.11,15
In one
study, more than 40% of 50 aggressive pituitary
adenomas showed cavernous sinus invasion.16
Tumors particularly prone to invasive growth
and recurrence include those arising from
sparsely granulated somatotrophs, silent corti-
cotrophs, corticotroph Crooke’s cells (CK20-
positive nonneoplastic cells with a prominent
Figure 1. Pathogenesis of Pituitary Tumors.
Pituitary adenomas arise from a differentiated hormone-expressing cell or from a null cell. Clinical phenotype is determined by the cell
of origin and the presence or absence of autonomous, specific hormone hypersecretion. The prevalence data are estimates. IGF-1 de-
notes insulin-like growth factor receptor 1, SF1 steroidogenic factor 1, and Tpit T-box factor, pituitary.
Normal pituitary Microadenoma Macroadenoma Aggressive adenoma
SOMATOTROPH
LACTOTROPH CORTICOTROPH THYROTROPH GONADOTROPH ANY
Growth hormoneSecreted hormone
Cell type
Prolactin Corticotropin Thyrotropin Luteinizing hormone or
follicle-stimulating hormone
or subunits
None
8–15Relative prevalence (%) 30–60 2–6 <1 <1 14–55
POU1F1Transcription factors POU1F1
Estrogen receptor alpha
Tpit POU1F1
GATA2
SF1
GATA2
Variable
IGF-1Target hormone Prolactin Cortisol T4 or T3 Estrogen or
testosterone
None
Acromegaly
Gigantism
Clinical phenotype Hypogonadism
Infertility
Galactorrhea
Cushing’s disease
Hypercortisolemia
Hyperthyroidism Hypergonadism Pituitary failure
Densely granulated
Sparsely granulated
Mammosomatotroph
Mixed somatolactotroph
Acidophil stem cell
Adenoma subtypes Densely granulated
Sparsely granulated
Acidophil stem cell
Densely granulated
Sparsely granulated
Crooke’s cell
Null cell
Silent gonadotroph
Silent corticotroph
Silent somatotroph
Pituitary gland
Carotid artery
Cavernous sinus
Optic
chiasm Cranial
nerves
III
IV
VI
V1
V2
Aneuploidy
Copy-number alterations
Germline or somatic mutations
Cell-cycle dysregulation
Paracrine stromal events
Epigenetic changes
Unknown driver changes
Proliferative
restraint
Senescence
Sphenoid sinus
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020 939
Pituitary-Tumor Endocrinopathies
Table 1. Individualized Approach to Pituitary Adenoma Management.*
Characteristic Finding or Outcome
Patient
Age
<30 yr More aggressive growth and recurrence, florid hormone phenotype (growth hormone)
>60 yr Less aggressive features, smaller tumor (growth hormone), larger tumor (prolactin)
Sex
Female More microadenomas (prolactin and corticotropin)
Male More macroadenomas, higher hormone levels (prolactin)
Tumor
MRI findings
Size Larger tumors are more resistant to treatment
Invasiveness Invasive tumors are more likely to recur
Intensity on T2-weighted
images
Intensity determines somatostatin receptor ligand responsiveness (growth hormone)
Histologic features
Dense granules Indolent course, responsive to somatostatin receptor ligand (growth hormone)
Sparse granules Florid course, resistant to somatostatin receptor ligand (growth hormone)
Receptor expression
Positive for SST2 Likely to respond to octreotide or lanreotide (growth hormone)
Positive for SST5 Likely to respond to pasireotide (corticotropin and growth hormone)
Positive for D2 Likely to respond to dopamine agonist (prolactin)
Negative for SST2, SST5,
and D2
Likely to be treatment-resistant
Ki-67 cell-cycle marker Recurrence less likely if <3%; recurrence more likely if ≥3%
GSP mutation Likely to respond to somatostatin receptor ligand (growth hormone)
Treatment
Transsphenoidal surgical
resection
Advantages: rapid hormone and symptom remission, one-time cost, potential for cure,
decompression of vital parasellar structures, tumor debulking may enhance adju-
vant therapy
Disadvantages: persistent tumor remnant, postoperative hypopituitarism requiring life-
long replacement therapy, some patients are not candidates for surgery
Risks: diabetes insipidus, electrolyte abnormalities, neurologic deficits, CSF rhinorrhea,
death from anesthetic agent (rarely); other risks include resection performed by low-
volume surgeons, as well as coexisting cardiac disease, cerebrovascular disease, or
diabetes
Radiation therapy Advantages: permanent results, one-time cost, long-term treatment not required, no
drug-related adverse events
Disadvantages: very slow efficacy onset, medical therapy required until effects are
­evident
Risks: hypopituitarism, visual disturbances, stroke; rarely, CNS damage, brain tumor
Pituitary-directed medical
therapy
Advantages: hormone control and symptom relief (prolactin, growth hormone, cortico-
tropin, thyrotropin), no hypopituitarism, tumor-mass control
Disadvantages: cure not permanent, long-term sustained treatment required, ongoing
cost, injection adherence required
Risks regarding somatostatin receptor ligand: gallstones or sludge, diarrhea, nausea,
hyperglycemia, sinus bradycardia, alopecia, injection-site pain, headache
Risks regarding D2 agonist: depression, nausea, vasospasm
*	Hormones in parentheses are those secreted by the adenoma. CNS denotes central nervous system, CSF cerebrospinal
fluid, and SST2 and SST5 somatostatin receptor subtype 2 and subtype 5, respectively.
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020940
The new engl and jour nal of medicine
cytoplasmic hyaline ring that displaces the nor-
mal basophilic granules of corticotropic cells),
and lactotrophs in middle-aged or older men.17
Pituitary carcinomas are exceedingly rare. They
account for less than 0.5% of pituitary tumors
and respond inconsistently to temozolomide.5
Management
Comprehensive management of a pituitary ade-
noma includes transsphenoidal surgical resec-
tion, irradiation, and medical therapy, each with
advantages and disadvantages that are specific
to the type of adenoma (Table 1). Serial or com-
bined approaches may be required.
Surgery is generally indicated for masses that
are 10 mm or more in diameter and those that
have extrasellar extension or central compressive
features, as well as for persistent tumor growth,
especially if vision is compromised or threat-
ened.18
Resection may alleviate compression of
vital structures and reverse compromised pitu-
itary hormone secretion. Predictors of remission
include experience of the surgeon, relatively low
levels of secreted hormone (if the level is elevated
at all), and small tumors. Postoperatively, hypo-
pituitarism may develop, as may diabetes insipi-
dus and cerebrospinal fluid leaks. Approximately
10% of patients have a recurrence over a period
of 10 years after surgery. Persistent tumor post-
operatively may reflect incomplete resection, in-
accessible cavernous sinus tumor tissue, or dural
nesting of hormone-secreting tumor cells.
Radiation therapy, administered by means of
conventional external-beam or proton-beam tech-
niques, or stereotactic radiosurgery requires local
expertise and is generally reserved for tumors
that are resistant to medical treatment or are not
controlled by surgery. Tumor growth is usually
arrested over a period of several years, and ade-
noma-derived hormone hypersecretion may per-
sist during the initial years.19
In most patients,
pituitary failure develops within 10 years after
radiation therapy, and lifelong hormone replace-
ment is required. Deterioration in vision and new
cranial-nerve palsies are rarely observed.19
Mortal-
ity from cerebrovascular causes is increased by a
factor of 4.42 (95% confidence interval [CI], 2.71 to
7.22) after conventional pituitary irradiation, with
16 observed deaths versus 3.6 expected deaths.20
Genetic Syndromes
Pituitary adenomas may occur in association
with several very rare genetic syndromes. Multi-
ple endocrine neoplasia type 1 is associated with
pituitary adenomas as well as parathyroid and
pancreatic-islet tumors and, less commonly, carci-
noid, thyroid, and adrenal tumors. The McCune–
Albright syndrome is characterized by polyostotic
fibrous dysplasia and cutaneous pigmentation,
with sexual precocity, hyperthyroidism, hypercor-
Table 2. Testing to Diagnose Hormone-Secreting Pituitary Tumors.*
Tumor or Disease Test Results Requiring Further Evaluation
Prolactinoma Serum prolactin level measurement Prolactin level elevated
Acromegaly Serum IGF-1 level Age-adjusted IGF-1 level elevated
Oral glucose-tolerance test (75 g of glucose)
with growth hormone measured at 0, 30,
and 60 min
Growth hormone level 0.4 μg/liter with the use of an ultra-
sensitive assay
Cushing’s disease 24-hr urinary free cortisol measurement Elevated urinary free cortisol level on at least two tests
Midnight salivary cortisol measurement Elevated free salivary cortisol level
Plasma cortisol measurement at 8 a.m., after
administration of dexamethasone (1 mg)
at 11 p.m.
Failure to suppress cortisol level to 1.8 μg/dl
Serum corticotropin measurement Low corticotropin level suggests adrenal adenoma; very high
level may indicate ectopic corticotropin source
Thyrotropin-secreting
tumor
Serum thyrotropin measurement, free T4
­measurement
Normal or increased free T4 level with measurable thyrotropin
may suggest thyrotropin-secreting tumor
*	IGF-1 denotes insulin-like growth factor 1.
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020 941
Pituitary-Tumor Endocrinopathies
tisolism, hyperprolactinemia, and acromegaly.
Rare cases of familial pituitary adenomas have
been reported in families with a predisposition
for somatotroph tumors in childhood or young
adulthood, and about 25% of these tumors have
been linked to germline mutations in AIP (the
gene for aryl hydrocarbon receptor–interacting
protein),21
which are not commonly encountered
with sporadic adenomas. The Carney complex
includes pituitary adenomas with benign cardiac
myxomas, schwannomas, thyroid adenomas, and
pigmented skin spots.22
Nonsecreting Adenomas
Although several lesions may present as nonse-
creting sellar masses23
(Fig. 2), most are non­
secreting adenomas of gonadotroph lineage.
These adenomas express differentiated hormone
products and cell-specific transcription factors,
but since the respective peripheral-blood hor-
mone levels are not elevated, the adenomas are
not associated with systemic syndrome pheno-
types.24
True null-cell adenomas express no
hormone gene product.
Figure 2. Approach to Patients Presenting with a Nonsecreting Pituitary Mass.
The main diagnostic consideration is to distinguish a nonsecreting or secreting adenoma from a nonadenomatous lesion. The treat-
ment approach is determined by the specific diagnosis. The dashed line indicates an option that may be considered under some cir­
cumstances.
Pituitary Mass
Nonsecreting Adenoma
Features
Management
according
to specific
hypersecretory
syndrome
Management
as for non-
adenomatous
parasellar
lesion
Cysts
Rathke
Craniopharyngioma
Arachnoid, epidermoid,
pineal
Neoplasms
Meningioma
Chordoma
Lymphoma
Pituicytoma
Germinoma
Sarcoma
Hemangiopericytoma
Mucoepidermoid carcinoma
Xanthogranuloma
Lipoma
Vascular
Apoplexy
Aneurysm
Angioma or hematoma
Inflammatory
Hypophysitis
Amyloidosis, sarcoidosis
Granulomatous polyangiitis
Infections
Bacterial
Syphilis, tuberculosis
Metastases
Breast, lung, prostate, sinus
Lymphoma, plasmacytoma
Miscellaneous
Ectopic pituitary
Fibrous dysplasia
Microadenoma (10 mm) Macroadenoma (≥10 mm)
Screening for hormone hypersecretion
and assessing pituitary function
MRI differential diagnosis
Observation, if no compressive
mass effects
Transsphenoidal surgery
Follow-up with MRI at 1, 2, and 5 yr
Reassessment if symptomatic
MRI annually to monitor for mass
persistence or recurrence
Pituitary-reserve testing every 6 mo for 2 yr
Hormone replacement as required
Incidental MRI finding
Neurologic
Visual-field deficits
Extraocular muscle palsy
Headache
Endocrine
Amenorrhea
Decreased libido
Infertility
Diabetes insipidus
Apoplexy
Pituitary hormonal deficiencies
Immunostaining
Gonadotropin subunits
Proopiomelanocortin
or corticotropin
Growth hormone
None
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020942
The new engl and jour nal of medicine
Clinical Features and Diagnosis
Nonsecreting adenomas may be unrecognized
for years and are usually diagnosed because of
local mass effects or hypogonadism or are de-
tected incidentally. Chiasmal compression causes
gradual, progressive deficits in vision, and about
two thirds of patients have decreased gonadotro-
pin levels and hypogonadism.25
In a study involv-
ing 385 consecutive patients with nonsecreting
adenomas, 289 had macroadenomas and 66 had
giant adenomas (4 cm in diameter), with most
of the patients presenting with headache or dis-
turbances in vision.26
Approximately 10% of pa-
tients with nonsecreting adenomas present with
pituitary apoplexy (characterized by the abrupt
onset of retroorbital pain, altered consciousness,
Figure 3. Approach to Patients Presenting with Hyperprolactinemia.
In patients with hyperprolactinemia, the clinical challenge is to rule out a secondary, nonpituitary cause before embarking on comprehen-
sive pituitary imaging and testing. The stalk effect is compression of the pituitary stalk due to a nonsecreting sellar mass that interrupts
delivery of inhibitory dopamine from the hypothalamus to the prolactin-secreting cells of the pituitary.
HyperprolactinemiaFeatures Rule out secondary causes
Consider nonsecreting mass
resulting in the stalk effect
Pregnancy
Drugs
Neuroleptics
D2 receptor blockers
Estrogens
Opiates
Antidepressants
Parasellar mass
Hypothalamic irradiation
Chest-wall injury
Renal failure
Assess pituitary-
reserve function
Test visual fields
Pituitary MRI
Dopamine agonist treatment
Dose adjustment
Unacceptable side effects
Prolactin level not reduced
No tumor shrinkage
Prolactin level normalized
Sexual function restored
Tumor shrinkage
Prolactin level reduced
Sexual function restored
Tumor persists with no local
compressive effects
Decrease dose
Consider surgery if hyper-
prolactinemia or mass
persists
Maximize dose Continue medication Continue medication
Surgery or radiotherapy
required in rare cases
Reconsider diagnosis
of nonsecreting adenoma
with stalk effect
Surgery or radiotherapy
required in rare cases
If prolactin level rises,
repeat MRI
If prolactin level rises,
repeat MRI
Compressive
effects
Microprolactinoma Macroprolactinoma
Amenorrhea,
oligomenorrhea,
infertility
Impotence,
oligospermia,
decreased libido
Galactorrhea
Osteoporosis
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020 943
Pituitary-Tumor Endocrinopathies
ophthalmoplegia, and ultimately, loss of vision
from pituitary hemorrhage or infarction).
Very rarely, circulating gonadotropin levels
are elevated, resulting in ovarian hyperstimula-
tion or increased testicular volumes. Paradoxi-
cally, however, high gonadotropin levels more
commonly down-regulate the gonadal axis.
Clinically silent tumors expressing nonsecret-
ed corticotropin or growth hormone account for
up to 20% of nonsecreting tumors. They are
usually resected after a nonsecreting macroade-
noma has been diagnosed, with the diagnosis
subsequently confirmed by histologic assess-
ment with appropriate immunostaining. These
silent tumors grow aggressively, with no appar-
ent features of hypercortisolism or acromegaly,
although their morphologic features may be in-
distinguishable from those of secretory adeno-
mas. A systematic review of 14 studies with a
total of 297 patients who underwent surgical
treatment for silent corticotroph adenomas
showed that 31% of the adenomas recurred dur-
ing a follow-up period of more than 5 years.17
Treatment
Complete resection of a nonsecreting macro­
adenoma is achieved in approximately 65% of
patients, with visual function restored in up to
80% of the patients and hypopituitarism, when
present, reversed in approximately 50%.26,27
Pre-
operative tumor invasiveness largely determines
the risk of postoperative persistent or recurrent
tumor requiring adjuvant irradiation and reopera-
tion. Radiosurgery was associated with high
rates of tumor control in a multicenter analysis
involving 512 patients followed for a median of
36 months, and pituitary failure developed in
21% of the patients.28
In a retrospective analysis
of tumor growth rates among 237 patients who
were followed for a median of 5.9 years, 36% of
the patients had a recurrence after surgery
alone, whereas 13% had a recurrence after sur-
gery and adjuvant radiotherapy.29
Guidelines13
recommend follow-up with peri-
odic pituitary MRI, vision evaluations, and pitu-
itary-function testing, and prophylactic radiother-
apy to prevent postoperative tumor recurrence or
progression has been suggested. Of 648 inciden-
tally discovered pituitary adenomas, 10% of the
229 microadenomas and 20% of the 419 macro­
adenomas increased in size during up to 8 years
of follow-up.30
Accordingly, expectant observation
is recommended for the management of stable,
nonsecreting microadenomas and small mac-
roadenomas. Preoperative or newly manifested
postoperative pituitary failure may be insidious.
In a study involving 2795 patients, pituitary fail-
ure, especially with corticotropin deficiency, was
associated with excess overall mortality (stan-
dardized mortality ratio, 4.35; 95% CI, 1.99 to
8.26), on the basis of 2.1 expected deaths versus
9 observed deaths (P0.001).31
Prolactin-Secreting Adenomas
Prolactinomas are the most common secretory
tumor, accounting for up to 60% of all pituitary
adenomas and more than 75% of pituitary ade-
nomas in women.32
Microprolactinomas, which
have a female:male ratio of 20:1, are usually
stable and slow-growing, with continued growth
after diagnosis in less than 15% of cases.33
Clinical Features
Most patients with a pituitary mass and a serum
prolactin level exceeding 150 ng per milliliter
(reference range, 20) are found to have a pro-
lactinoma (Fig. 3). A prolactin level of more than
250 ng per milliliter is usually diagnostic of a
macroprolactinoma, and the tumor mass usually
correlates with the serum prolactin level.34
Large
prolactinomas (10 mm in diameter) that are
aggressive account for less than 5% of tumors
and are characterized by very high prolactin lev-
els (1000 ng per milliliter) and a male:female
ratio of 9:1. In a study involving 45 men and 51
women with prolactinomas, the tumors in the
men were larger than those in the women (mean
[±SD] diameter, 26±2 mm vs. 10±1 mm) and
grew more aggressively, with a mean serum
prolactin level of 2789±572 ng per milliliter in
men versus 292±74 ng per milliliter in women.35
Persistently elevated prolactin levels suppress
gonadotropin, leading to amenorrhea, oligo-
menorrhea, or a short luteal phase associated
with infertility in women and low libido, impo-
tence, oligospermia, or azoospermia in men.
About 50% of women and 35% of men have
galactorrhea, and both women and men have
reduced bone density, often associated with sex
steroid hormone deficiency and an increased
risk of vertebral fractures.
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020944
The new engl and jour nal of medicine
Diagnosis
Hyperprolactinemia is caused mainly by preg-
nancy, prolactinomas, medications, chest-wall
injury, and functional or mechanical interrup-
tion of pituitary-stalk dopamine transport. Pro-
lactin levels are elevated in approximately 30%
of patients with acromegaly.36
Prolactin levels should be measured in all
patients with a sellar mass; conversely, hyper­
prolactinemia not explained by pregnancy or
exposure to neuroleptic drugs should prompt
pituitary imaging to rule out a pituitary mass. In
patients with hyperprolactinemia and a pituitary
mass, failure to achieve tumor shrinkage with
dopamine agonists may suggest compression of
the pituitary stalk due to a nonsecreting sellar
mass that interrupts delivery of inhibitory dopa-
mine from the hypothalamus, resulting in dis-
ruption of prolactin control; this is known as the
stalk effect.
Treatment
Prolactin levels should be normalized, with sex-
ual function and fertility restored, galactorrhea
halted, and the tumor mass eliminated or re-
duced, while pituitary function is retained. Low
bone density should be addressed.
Prolactinomas are ideally managed with a
dopamine agonist,33
which should lower prolac-
tin levels and shrink the tumor mass. Bromo­
criptine is rarely used, since it requires daily
dosing. One study showed that cabergoline, ad-
ministered at a dose of 0.5 to 1 mg once or twice
weekly, lowered prolactin levels in 83% of 459
women with hyperprolactinemia,37
and this agent
restores ovulatory cycles and fertility with few
side effects. In approximately 65% of patients
with macroprolactinomas, treatment normalizes
prolactin levels and reduces the tumor mass.38
Up to 15% of patients do not have a response
to maximal dopaminergic doses (i.e., prolactin
levels are not normalized, and tumor shrinkage
is 50%). Patients with normalized prolactin
levels but inadequate tumor shrinkage may re-
quire surgery or radiotherapy.
Hyperprolactinemia remits in up to 20% of
patients after dose tapering and discontinuation
of cabergoline, which can be attempted after
more than 2 years of therapy and only when the
possibility of tumor invasion has been rigor-
ously ruled out.39
Adverse effects of cabergoline, reported in up
to 50% of patients, include nausea, nasal stuffi-
ness, depression, digital vasospasm, postural
hypotension, and in rare cases, cerebrospinal
fluid leak. Mood disorders, exacerbation of psy-
chosis, and poor impulse control are additional
rare effects.40
The low doses of cabergoline used
for the treatment of prolactinomas do not ap-
pear to place patients at risk for clinically sig-
nificant cardiac valve disease, and valvulopathy
was not observed in 192 patients who were fol-
lowed for 34 months in a cross-sectional study.41
Because asymptomatic, mild tricuspid regurgita-
tion was reported in 20% of patients, those
treated with cabergoline in whom a cardiac mur-
mur develops should undergo cardiac evaluation.
In 31 surgical series involving a total of 1224
patients with microprolactinomas, transsphe-
noidal resection resulted in normal prolactin
levels in 71% of the patients, and initial cure
rates may exceed 90% when resection is per-
formed by high-volume surgeons.42
About 50%
of macroprolactinomas remit, and persistent,
postoperative hyperprolactinemia arises from
tumor remnants.42
The efficacy of treatment
with dopamine agonists and the long-term risk
of postoperative recurrence have discouraged
clinicians from choosing surgery as a primary
treatment for macroprolactinomas. Invasive,
dopamine agonist–resistant prolactinomas may
require more than one surgery, with continued
administration of high-dose cabergoline.43
Ra-
diation therapy is reserved for patients with
treatment-resistant macroprolactinomas.
During pregnancy, pituitary expansion, espe-
cially in the case of macroprolactinomas, may
threaten the visual fields.44
Prophylactic trans­
sphenoidal resection should be considered when
vision is threatened. Dopamine agonist therapy
should be discontinued when pregnancy is con-
firmed.
Acromegaly
Acromegaly, with an incidence of approximately
10 cases per 1 million persons,32,45
is caused by a
growth hormone–secreting somatotroph tumor.
High growth hormone and insulin-like growth
factor 1 (IGF-1) levels are associated with strik-
ing somatic and metabolic dysfunction. Densely
granulated somatotroph adenomas arise insidi-
ously, whereas sparsely granulated subtypes,
which develop in younger patients, are character-
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020 945
Pituitary-Tumor Endocrinopathies
ized by aggressive growth and florid disease.12
In very rare cases, extrapituitary acromegaly is
caused by neuroendocrine tumor production of
growth hormone or growth hormone–releasing
hormone.46,47
Clinical Features
Approximately 70% of patients with acromegaly
have an invasive macroadenoma at diagnosis.
Coexisting conditions include headache and in-
sidious acral and soft-tissue changes. The diag-
nosis of acromegaly can be delayed by a mean of
approximately 10 years after the onset of symp-
toms.48
Patients may first seek dental, orthope-
dic, rheumatologic, or cardiac care. In one study,
approximately 20% of 324 patients sought care
because of an altered facial appearance, enlarged
extremities, or both.49
Other features include
increased shoe or ring size, voice deepening,
the carpal tunnel syndrome, hyperhidrosis, and
frontal-skull bossing and coarse oily skin. Prog-
nathism leads to incisor separation and jaw
malocclusion. Obstructive sleep apnea and ex-
cessive snoring are hallmarks of uncontrolled
acromegaly. Arthropathy is reported in approxi-
mately 70% of patients, with polyarticular arthri-
tis, osteophytosis, dorsal kyphosis, and vertebral
fractures.48
Coexisting cardiovascular conditions
include hypertension, arrhythmias, and left ven-
tricular dysfunction, with an increased aortic-
root diameter.50
Despite biochemical control,
cardiovascular disorders may persist.51
Growth
hormone induces insulin resistance, with glu-
cose intolerance,52
and patients with acromegaly
have an increased risk of diabetes, as compared
with a control cohort from the general popula-
tion (hazard ratio, 4.0 [95% CI, 2.7 to 5.8]; rate
per 1000 persons, 12.1 cases [95% CI, 9.0 to
16.4] vs. 3.4 cases [95% CI, 2.9 to 4.1]).53
About
30% of patients have high prolactin levels, often
with galactorrhea.
Hypertrophic colonic mucosal folds and diver-
ticulae can occur in patients with acromegaly.54,55
In a case–control study, colonic polyps were
detected in 32% of 165 patients, with an esti-
mated relative risk of 6.21 (95% CI, 4.08 to
9.48).56
Studies involving more than 2000 pa-
tients showed an increased incidence of cancer,
with an overall standardized incidence ratio of
1.5 (95% CI, 1.2 to 1.8), mostly accounted for by
colorectal, kidney, and thyroid cancers.57,58
How-
ever, other studies have shown no increase in the
incidence of cancer. Nevertheless, colonoscopy
should be performed at diagnosis and subse-
quently, according to published guidelines.59
A 20-year study showed higher mortality rates
among 333 patients with acromegaly than among
4995 controls (113 deaths [34%] vs. 1334 deaths
[27%]; odds ratio 1.6; 95% CI, 1.2 to 2.2).60
Deaths associated with acromegaly are due to
cardiovascular, respiratory, and cerebrovascular
disorders, with cancer also reported as a cause
of death in more recent years. Persistently elevat-
ed growth hormone and IGF-1 levels, diabetes,
hypertension, older age, pituitary irradiation,
and inadequate treatment of adrenal insuffi-
ciency all contribute significantly to mortality.61
Gigantism, a rare condition, is due to exces-
sive secretion of growth hormone before epiphy-
seal closure. The disorder may be associated with
germline AIP mutations, the McCune–Albright
syndrome, or acidophilic stem-cell adenomas;
X-linked gigantism is characterized by Xq26.3
chromosomal microduplications, resulting in
accelerated gigantism and overexpressed tumor
GPR101 (a gene for a G-protein–coupled recep-
tor).62
Surgical resection with adjuvant growth
hormone suppression is required to maintain
remission and prevent long-term tissue exposure
to excess growth hormone and IGF-1.63
Diagnosis
IGF-1 levels that are elevated for the patient’s age
are highly specific for acromegaly and also cor-
relate with indexes of disease activity.64
The
pulsatile nature of adenoma growth hormone
secretion precludes reliance on a random mea-
surement for diagnosis. Instead, the diagnosis is
established by using an ultrasensitive assay to
document nadir growth hormone levels of more
than 0.4 μg per liter during a 75-g glucose
load.65
Growth hormone levels are log-linearly
concordant with IGF-1 levels.66
Postoperatively,
IGF-1 levels may remain elevated for months
despite control of growth hormone secretion.
Treatment
Comprehensive treatment goals for acromegaly
include ablating or controlling the pituitary
mass, suppressing growth hormone and IGF-1
hypersecretion, and preventing the development
of associated disorders while maintaining ante-
rior pituitary function.59
In 13 studies involving
1018 patients who underwent surgical resection,
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020946
The new engl and jour nal of medicine
control of growth hormone secretion and IGF-1
levels was achieved in 73% of patients with micro-
adenomas and 61% of patients with macroade-
nomas.67-69
However, patients with macroadeno-
mas that invade the cavernous sinus invariably
show persistent growth hormone hypersecretion
after surgery. In a study involving 371 patients
treated with radiosurgery, biochemical remission
was reported in 59% of the patients, with a
mean time to remission of 38 months and a
mean time to recurrence of 17 months.70
Dopamine agonists have been proposed for
patients with mild disease, and the addition of
cabergoline may normalize IGF-1 levels in some
patients with disease that is resistant to somato­
statin therapy.71
The somatostatin receptor ligands
octreotide and lanreotide bind SST2 (somatosta-
tin receptor subtype 2), inhibiting growth hor-
mone secretion.72
A meta-analysis of 90 studies
involving 4464 patients treated with somatosta-
tin receptor ligands showed overall control of
growth hormone secretion and of IGF-1 levels in
56% and 55% of the patients, respectively.73
In a
meta-analysis involving 1685 patients in 41 stud-
ies, 53% of patients had a reduction in tumor
mass.74
Efficacy may be enhanced by increasing
the dose and injection frequency.75
Soft-tissue
swelling and headache usually resolve, sleep ap-
nea abates, and left ventricular function im-
proves,48
but hypertension may persist. Tumor
SST2 expression, densely granulated tumor, and
hypointensity on T2-weighted MRI are important
markers of treatment responsiveness.12,76
Tran-
sient gastrointestinal side effects, occurring in
approximately 30% of patients, include loose
stools and nausea. Gallbladder sludge occurs in
up to 25% of patients, although cholecystitis is
very rare. Asymptomatic sinus bradycardia also
occurs. Octreotide and lanreotide do not gener-
ally disrupt glucose homeostasis, but pasireotide,
a long-acting hexapeptide somatostatin multi-
receptor ligand, results in hyperglycemia and
new-onset diabetes in about 60% of patients.77
Pegvisomant, a growth hormone receptor
antagonist that blocks peripheral growth hor-
mone action and subsequent IGF-1 production,78
is useful for patients with disease that is resis-
tant to somatostatin receptor ligands, as well as
patients with hyperglycemia, since the drug en-
hances insulin sensitivity.79
A surveillance study
showed IGF-1 control in 63% of 1288 patients.80
Side effects include elevated hepatic aminotrans-
ferase levels, injection-site inflammation, and
lipodystrophy. Growth hormone axis blockade
with growth hormone receptor–directed pegviso-
mant combined with pituitary-directed somato­
statin receptor ligand offers greater efficacy than
either drug alone.81
Treatment approaches have advantages and
disadvantages for individualizing patient care
and maximizing growth hormone and IGF-1
control (Table 1).12
Patients should be monitored
for symptoms affecting the quality of life, in-
cluding anxiety. Management of coexisting con-
ditions, especially high blood pressure, cardiac
dysfunction, and sleep apnea, as well as elevated
blood sugar levels, is important to reduce the
risk of death.
Cushing’s Disease
Corticotropin-secreting corticotroph adenomas,
which account for up to 15% of pituitary tu-
mors, with an incidence of 1.6 cases per 1 mil-
lion persons,82
are typically small (approximately
6 mm in diameter) and are 5 to 10 times as
common in women as in men. Excessive secre-
tion of corticotropin leads to adrenal hypercor-
tisolemia. Corticotroph adenomas account for
approximately 70% of cases of Cushing’s syn-
drome, with iatrogenic hypercortisolism, ectopic
corticotropin or corticotropin-releasing hormone
production, and cortisol-producing adrenal le-
sions accounting for the rest.83
Clinical Features
Typical cushingoid features include plethoric
moon facies with thin skin, purple striae, and
easy bruising. Central obesity, hypertension, glu-
cose intolerance or diabetes, and, especially in
younger women, menstrual disturbances and
osteoporosis are observed, as are proximal mus-
cle wasting and weakness, acne, hirsutism, depres-
sion, psychoses, and susceptibility to infection.84
The disorder may be indolent or clinically
florid, with a high mortality rate if inadequately
controlled. In a study involving 502 unselected
patients, the overall standardized mortality ratio
was 2.5 (95% CI, 2.1 to 2.9), with 133 observed
deaths versus 54 expected deaths; cardiovascular
disease accounted for most of the excess deaths.85
Mortality was also elevated among the patients
with biochemical remission (standardized mor-
tality ratio, 1.9 [95% CI, 1.5 to 2.3]), with 89
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020 947
Pituitary-Tumor Endocrinopathies
observed deaths versus 47 expected deaths.
Other causes of death include infections and
suicide. Rapid-onset hypercortisolism with skin
hyperpigmentation and severe myopathy sug-
gests an ectopic tumor source of corticotropin,
often associated with hypertension and hypo-
kalemic alkalosis.83
Diagnosis
Accurate diagnosis of Cushing’s disease can be
challenging.83
Approximately 40% of corticotro-
pin-secreting corticotroph tumors are not visible
on imaging, and at least 10% of persons in the
general population have small, clinically silent
microadenomas. The disorder may therefore be
overdiagnosed, especially since clinical features
of hypercortisolemia overlap with other, much
more common disorders, including obesity, hyper-
tension, glucose intolerance, and osteoporosis.
The diagnosis of pathologic hypercortisolemia
is established on the basis of clinical features of
hypercortisolism coupled with reproducible evi-
dence of failure to suppress plasma cortisol at
8 a.m. to a level below 1.8 μg per deciliter after
the administration of 1 mg of dexamethasone at
11 p.m. or elevated 24-hour urinary free cortisol
levels and midnight salivary cortisol values.84
Basal corticotropin levels are usually inappropri-
ately high, and suppressibility of corticotropin
by glucocorticoids may distinguish a pituitary
tumor from an ectopic source.
Since the results of repeated urinary free cor-
tisol and midnight salivary cortisol measure-
ments may vary, bilateral inferior petrosal sinus
sampling of corticotropin levels may be required
to confirm the diagnosis. A central-to-peripheral
corticotropin gradient that exceeds 2 before and
after the administration of corticotropin-stimu-
lating hormone definitively localizes a cortico-
troph tumor source with more than 95% sensi-
tivity.
Treatment
Selective transsphenoidal adenomectomy is rec-
ommended as initial therapy for Cushing’s dis-
ease, with remission achieved in approximately
75% of patients and recurrence seen in approxi-
mately 10%.86
Although more radical surgery
offers the potential for total adenoma resection,
complication rates are higher and pituitary dam-
age is more likely with total resection.87
Radia-
tion therapy may control the disease,88
but the
efficacy of this treatment is delayed for several
years, and approximately 30% of patients have a
relapse.
Adrenalectomy may immediately reverse hy-
percortisolism. However, lifelong replacement of
adrenal hormones is challenging. Patients who
undergo adrenalectomy are also at risk for adre-
nal crisis and Nelson’s syndrome (pituitary en-
largement and development of corticotropin-
secreting pituitary adenomas, which often have
an invasive growth pattern).
Adrenal-targeted medical therapy may offer
clinical and biochemical improvement, but most
studies of such treatment have not been rigor-
ously controlled, and the results are often incon-
sistent.89
Adrenal steroidogenesis inhibitors block
hypercortisolemia but do not target the pituitary
tumor. Ketoconazole, an antifungal imidazole,
normalizes urinary free cortisol levels in 50% of
patients.90
Side effects include nausea, headache,
low testosterone levels, reversible elevated liver
enzyme levels, and in rare cases, hepatotoxicity.
Metyrapone controls urinary free cortisol levels
in about 50% of patients, and accumulated ste-
roid precursors may cause acne, hirsutism, hyper-
tension, and hypokalemia. Mitotane, an adreno-
lytic agent, is used mainly for adrenal carcinoma.
Mifepristone, a glucocorticoid receptor antago-
nist, is approved for the treatment of hypergly-
cemia associated with Cushing’s syndrome in
patients in whom surgery has failed or who are
not surgical candidates. Since mifepristone blocks
the action of cortisol, corticotropin and urinary
free cortisol levels are increased.91
Adrenal fail-
ure, hypokalemia, and excessive vaginal bleeding
may limit the use of this agent.
Pituitary-targeted drugs include high-dose
cabergoline (up to 1 mg daily), which controls
hypercortisolism in up to 30% of patients, al-
though therapeutic efficacy is often not main-
tained in the long term. Pasireotide blocks ade-
noma-derived corticotropin secretion, normalizes
urinary free cortisol levels in approximately 40%
of patients with mild disease, and improves clini-
cal features. Hyperglycemia develops in most
patients.92,93
Thyrotropin-Secreting Tumors
Accounting for approximately 1% of adenomas,
thyrotropin-secreting tumors lead to elevated or
inappropriately suppressed thyrotropin levels
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020948
The new engl and jour nal of medicine
with normal or elevated thyroid hormone levels.94
Resection and adjuvant treatment are required
for adequate biochemical outcomes.
Dr. Melmed reports receiving grant support, advisory board
fees, and fees for serving as a course mentor, paid to his insti-
tution, from Pfizer, consulting fees from Ionis, Midatech, and
Chiasma, grant support from Ono, advisory board fees from
Strongbridge, and advisory fees and travel support from Ipsen.
No other potential conflict of interest relevant to this article
was reported.
Disclosure forms provided by the author are available with the
full text of this article at NEJM.org.
References
1.	 Gittleman H, Ostrom QT, Farah PD,
et al. Descriptive epidemiology of pitu-
itary tumors in the United States, 2004-
2009. J Neurosurg 2014;​121:​527-35.
2.	 Casanueva FF, Barkan AL, Buchfelder
M, et al. Criteria for the definition of Pitu-
itary Tumor Centers of Excellence (PTCOE):
a Pituitary Society statement. Pituitary 2017;​
20:​489-98.
3.	 Melmed S. Pathogenesis of pituitary
tumors. Nat Rev Endocrinol 2011;​7:​257-66.
4.	 Molitch ME. Diagnosis and treatment
of pituitary adenomas: a review. JAMA
2017;​317:​516-24.
5.	 Syro LV, Rotondo F, Ramirez A, et al.
Progress in the diagnosis and classifica-
tion of pituitary adenomas. Front Endo-
crinol (Lausanne) 2015;​6:​97.
6.	 Chesnokova V, Zonis S, Kovacs K, et al.
p21(Cip1) Restrains pituitary tumor growth.
Proc Natl Acad Sci U S A 2008;​105:​17498-
503.
7.	 Hage M, Viengchareun S, Brunet E,
et al. Genomic alterations and complex sub-
clonal architecture in sporadic GH-secret-
ing pituitary adenomas. J Clin Endocrinol
Metab 2018;​103:​1929-39.
8.	 Bi WL, Larsen AG, Dunn IF. Genomic
alterations in sporadic pituitary tumors.
Curr Neurol Neurosci Rep 2018;​18:​4.
9.	 Reincke M, Sbiera S, Hayakawa A, et al.
Mutations in the deubiquitinase gene
USP8 cause Cushing’s disease. Nat Genet
2015;​47:​31-8.
10.	 Agustsson TT, Baldvinsdottir T, Jonas­
son JG, et al. The epidemiology of pitu-
itary adenomas in Iceland, 1955-2012:
a nationwide population-based study. Eur J
Endocrinol 2015;​173:​655-64.
11.	Lopes MBS. The 2017 World Health
Organization classification of tumors of the
pituitary gland: a summary. Acta Neuro-
pathol 2017;​134:​521-35.
12.	Cuevas-Ramos D, Carmichael JD,
Cooper O, et al. A structural and func-
tional acromegaly classification. J Clin
Endocrinol Metab 2015;​100:​122-31.
13.	Freda PU, Beckers AM, Katznelson L,
et al. Pituitary incidentaloma: an Endo-
crine Society clinical practice guideline.
J Clin Endocrinol Metab 2011;​96:​894-904.
14.	Fleseriu M, Hashim IA, Karavitaki N,
et al. Hormonal replacement in hypopitu-
itarism in adults: an Endocrine Society
clinical practice guideline. J Clin Endocri-
nol Metab 2016;​101:​3888-921.
15.	Raverot G, Burman P, McCormack A,
et al. European Society of Endocrinology
clinical practice guidelines for the man-
agement of aggressive pituitary tumours
and carcinomas. Eur J Endocrinol 2018;​
178:​G1-G24.
16.	 Del Basso De Caro M, Solari D, Pagliuca
F, et al. Atypical pituitary adenomas: clin-
ical characteristics and role of ki-67 and
p53 in prognostic and therapeutic evalua-
tion: a series of 50 patients. Neurosurg
Rev 2017;​40:​105-14.
17.	 Fountas A, Lavrentaki A, Subramanian
A, Toulis KA, Nirantharakumar K, Kara-
vitaki N. Recurrence in silent cortico-
troph adenomas after primary treatment:
a systematic review and meta-analysis.
J Clin Endocrinol Metab 2018 December
21 (Epub ahead of print).
18.	Lucas JW, Bodach ME, Tumialan LM,
et al. Congress of Neurological Surgeons
systematic review and evidence-based
guideline on primary management of
patients with nonfunctioning pituitary
adenomas. Neurosurgery 2016;​79(4):​E533-
E535.
19.	 Lee CC, Vance ML, Xu Z, et al. Stereo-
tactic radiosurgery for acromegaly. J Clin
Endocrinol Metab 2014;​99:​1273-81.
20.	Ayuk J, Clayton RN, Holder G, Shep-
pard MC, Stewart PM, Bates AS. Growth
hormone and pituitary radiotherapy, but
not serum insulin-like growth factor-I
concentrations, predict excess mortality
in patients with acromegaly. J Clin Endo-
crinol Metab 2004;​89:​1613-7.
21.	 Vierimaa O, Georgitsi M, Lehtonen R,
et al. Pituitary adenoma predisposition
caused by germline mutations in the AIP
gene. Science 2006;​312:​1228-30.
22.	Correa R, Salpea P, Stratakis CA. Car-
ney complex: an update. Eur J Endocrinol
2015;​173:​M85-M97.
23.	Famini P, Maya MM, Melmed S. Pitu-
itary magnetic resonance imaging for sel-
lar and parasellar masses: ten-year expe-
rience in 2598 patients. J Clin Endocrinol
Metab 2011;​96:​1633-41.
24.	Drummond J, Roncaroli F, Grossman
AB, Korbonits M. Clinical and pathologi-
cal aspects of silent pituitary adenomas.
J Clin Endocrinol Metab 2019;​104:​2473-89.
25.	 Pappy AL II, Savinkina A, Bicknese C,
Neill S, Oyesiku NM, Ioachimescu AG.
Predictive modeling for pituitary adeno-
mas: single center experience in 501 con-
secutive patients. Pituitary 2019;​22:​520-31.
26.	Chen L, White WL, Spetzler RF, Xu B.
A prospective study of nonfunctioning
pituitary adenomas: presentation, manage-
ment, and clinical outcome. J Neurooncol
2011;​102:​129-38.
27.	Paluzzi A, Fernandez-Miranda JC,
Tonya Stefko S, Challinor S, Snyderman
CH, Gardner PA. Endoscopic endonasal
approach for pituitary adenomas: a series
of 555 patients. Pituitary 2014;​17:​307-19.
28.	Sheehan JP, Starke RM, Mathieu D,
et al. Gamma Knife radiosurgery for the
management of nonfunctioning pituitary
adenomas: a multicenter study. J Neuro-
surg 2013;​119:​446-56.
29.	 Tampourlou M, Ntali G, Ahmed S, et al.
Outcome of nonfunctioning pituitary ad-
enomas that regrow after primary treat-
ment: a study from two large UK centers.
J Clin Endocrinol Metab 2017;​102:​1889-97.
30.	Huang W, Molitch ME. Management
of nonfunctioning pituitary adenomas
(NFAs): observation. Pituitary 2018;​21:​
162-7.
31.	Olsson DS, Nilsson AG, Bryngelsson
IL, Trimpou P, Johannsson G, Andersson E.
Excess mortality in women and young
adults with nonfunctioning pituitary
­adenoma: a Swedish nationwide study.
J Clin Endocrinol Metab 2015;​100:​2651-8.
32.	Fernandez A, Karavitaki N, Wass JA.
Prevalence of pituitary adenomas: a com-
munity-based, cross-sectional study in Ban-
bury (Oxfordshire, UK). Clin Endocrinol
(Oxf) 2010;​72:​377-82.
33.	Melmed S, Casanueva FF, Hoffman
AR, et al. Diagnosis and treatment of
­hyperprolactinemia: an Endocrine Society
clinical practice guideline. J Clin Endocri-
nol Metab 2011;​96:​273-88.
34.	Burke WT, Penn DL, Castlen JP, et al.
Prolactinomas and nonfunctioning ade-
nomas: preoperative diagnosis of tumor
type using serum prolactin and tumor size.
J Neurosurg 2019 June 14 (Epub ahead of
print).
35.	Delgrange E, Trouillas J, Maiter D,
Donckier J, Tourniaire J. Sex-related dif-
ference in the growth of prolactinomas:
a clinical and proliferation marker study.
J Clin Endocrinol Metab 1997;​82:​2102-7.
36.	 Bernard V, Young J, Chanson P, Binart
N. New insights in prolactin: pathological
implications. Nat Rev Endocrinol 2015;​
11:​265-75.
37.	 Webster J, Piscitelli G, Polli A, Ferrari
CI, Ismail I, Scanlon MF. A comparison
of cabergoline and bromocriptine in the
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020 949
Pituitary-Tumor Endocrinopathies
treatment of hyperprolactinemic amenor-
rhea. N Engl J Med 1994;​331:​904-9.
38.	Biller BM, Molitch ME, Vance ML, et al.
Treatment of prolactin-secreting macro­
adenomas with the once-weekly dopa-
mine agonist cabergoline. J Clin Endocri-
nol Metab 1996;​81:​2338-43.
39.	Colao A, Di Sarno A, Cappabianca P,
Di Somma C, Pivonello R, Lombardi G.
Withdrawal of long-term cabergoline ther-
apy for tumoral and nontumoral hyper­
prolactinemia. N Engl J Med 2003;​349:​
2023-33.
40.	Ioachimescu AG, Fleseriu M, Hoff-
man AR, Vaughan Iii TB, Katznelson L.
Psychological effects of dopamine ago-
nist treatment in patients with hyperpro-
lactinemia and prolactin-secreting adeno-
mas. Eur J Endocrinol 2019;​180:​31-40.
41.	Stiles CE, Tetteh-Wayoe ET, Bestwick
J, Steeds RP, Drake WM. A meta-analysis
of the prevalence of cardiac valvulopathy
in hyperprolactinemic patients treated
with cabergoline. J Clin Endocrinol Metab
2018;​104:​523-38.
42.	Tampourlou M, Trifanescu R, Paluzzi
A, Ahmed SK, Karavitaki N. Therapy of
endocrine disease: surgery in micropro-
lactinomas: effectiveness and risks based
on contemporary literature. Eur J Endo-
crinol 2016;​175:​R89-R96.
43.	Liu W, Zahr RS, McCartney S, Cetas
JS, Dogan A, Fleseriu M. Clinical out-
comes in male patients with lactotroph
adenomas who required pituitary surgery:
a retrospective single center study. Pitu-
itary 2018;​21:​454-62.
44.	Bronstein MD. Prolactinomas and
pregnancy. Pituitary 2005;​8:​31-8.
45.	Burton T, Le Nestour E, Neary M,
Ludlam WH. Incidence and prevalence
of acromegaly in a large US health plan
database. Pituitary 2016;​19:​262-7.
46.	Thorner MO, Perryman RL, Cronin
MJ, et al. Somatotroph hyperplasia: suc-
cessful treatment of acromegaly by re-
moval of a pancreatic islet tumor secret-
ing a growth hormone-releasing factor.
J Clin Invest 1982;​70:​965-77.
47.	 Melmed S, Ezrin C, Kovacs K, Good-
man RS, Frohman LA. Acromegaly due to
secretion of growth hormone by an ectop­
ic pancreatic islet-cell tumor. N Engl J Med
1985;​312:​9-17.
48.	 Colao A, Grasso LFS, Giustina A, et al.
Acromegaly. Nat Rev Dis Primers 2019;​5:​
20.
49.	Reid TJ, Post KD, Bruce JN, Nabi
Kanibir M, Reyes-Vidal CM, Freda PU.
Features at diagnosis of 324 patients with
acromegaly did not change from 1981 to
2006: acromegaly remains under-recog-
nized and under-diagnosed. Clin Endo-
crinol (Oxf) 2010;​72:​203-8.
50.	Casini AF, Neto LV, Fontes R, França
RF, Xavier SS, Gadelha MR. Aortic root
ectasia in patients with acromegaly: expe-
rience at a single center. Clin Endocrinol
(Oxf) 2011;​75:​495-500.
51.	Annamalai AK, Webb A, Kandasamy
N, et al. A comprehensive study of clini-
cal, biochemical, radiological, vascular,
cardiac, and sleep parameters in an un-
selected cohort of patients with acromeg-
aly undergoing presurgical somatostatin
receptor ligand therapy. J Clin Endocrinol
Metab 2013;​98:​1040-50.
52.	Alexopoulou O, Bex M, Kamenicky P,
Mvoula AB, Chanson P, Maiter D. Preva-
lence and risk factors of impaired glucose
tolerance and diabetes mellitus at diagno-
sis of acromegaly: a study in 148 patients.
Pituitary 2014;​17:​81-9.
53.	Dal J, Feldt-Rasmussen U, Andersen
M, et al. Acromegaly incidence, preva-
lence, complications and long-term prog-
nosis: a nationwide cohort study. Eur J
Endocrinol 2016;​175:​181-90.
54.	 Wassenaar MJ, Cazemier M, Biermasz
NR, et al. Acromegaly is associated with
an increased prevalence of colonic diver-
ticula: a case-control study. J Clin Endo-
crinol Metab 2010;​95:​2073-9.
55.	Resmini E, Tagliafico A, Bacigalupo
L, et al. Computed tomography colonog-
raphy in acromegaly. J Clin Endocrinol
Metab 2009;​94:​218-22.
56.	Gonzalez B, Vargas G, Mendoza V,
Nava M, Rojas M, Mercado M. The preva-
lence of colonic polyps in patients with
acromegaly: a case-control, nested in a
cohort colonoscopic study. Endocr Pract
2017;​23:​594-9.
57.	 Terzolo M, Reimondo G, Berchialla P,
et al. Acromegaly is associated with in-
creased cancer risk: a survey in Italy. En-
docr Relat Cancer 2017;​24:​495-504.
58.	 Dal J, Leisner MZ, Hermansen K, et al.
Cancer incidence in patients with acro-
megaly: a cohort study and meta-analysis
of the literature. J Clin Endocrinol Metab
2018;​103:​2182-8.
59.	 Melmed S, Bronstein MD, Chanson P,
et al. A consensus statement on acromeg-
aly therapeutic outcomes. Nat Rev Endo-
crinol 2018;​14:​552-61.
60.	 Ritvonen E, Löyttyniemi E, Jaatinen P,
et al. Mortality in acromegaly: a 20-year
follow-up study. Endocr Relat Cancer
2016;​23:​469-80.
61.	 Sherlock M, Reulen RC, Aragon-Alonso
A, et al. A paradigm shift in the monitor-
ing of patients with acromegaly: last
available growth hormone may overesti-
mate risk. J Clin Endocrinol Metab 2014;​
99:​478-85.
62.	Trivellin G, Daly AF, Faucz FR, et al.
Gigantism and acromegaly due to Xq26
microduplications and GPR101 mutation.
N Engl J Med 2014;​371:​2363-74.
63.	 Goldenberg N, Racine MS, Thomas P,
Degnan B, Chandler W, Barkan A. Treat-
ment of pituitary gigantism with the
growth hormone receptor antagonist
pegvisomant. J Clin Endocrinol Metab
2008;​93:​2953-6.
64.	Clemmons DR, Van Wyk JJ, Ridgway
EC, Kliman B, Kjellberg RN, Underwood
LE. Evaluation of acromegaly by radioim-
munoassay of somatomedin-C. N Engl J
Med 1979;​301:​1138-42.
65.	Katznelson L, Laws ER Jr, Melmed S,
et al. Acromegaly: an Endocrine Society
clinical practice guideline. J Clin Endocri-
nol Metab 2014;​99:​3933-51.
66.	Faje AT, Barkan AL. Basal, but not
pulsatile, growth hormone secretion de-
termines the ambient circulating levels of
insulin-like growth factor-I. J Clin Endo-
crinol Metab 2010;​95:​2486-91.
67.	 Buchfelder M, Schlaffer SM. The sur-
gical treatment of acromegaly. Pituitary
2017;​20:​76-83.
68.	Kim JH, Hur KY, Lee JH, et al. Out-
come of endoscopic transsphenoidal sur-
gery for acromegaly. World Neurosurg
2017;​104:​272-8.
69.	Babu H, Ortega A, Nuno M, et al.
Long-term endocrine outcomes following
endoscopic endonasal transsphenoidal
surgery for acromegaly and associated
prognostic factors. Neurosurgery 2017;​
81:​357-66.
70.	Ding D, Mehta GU, Patibandla MR,
et al. Stereotactic radiosurgery for acro-
megaly: an international multicenter retro-
spective cohort study. Neurosurgery 2019;​
84:​717-25.
71.	Kuhn E, Chanson P. Cabergoline in
acromegaly. Pituitary 2017;​20:​121-8.
72.	Lamberts SW, van der Lely A-J, de
Herder WW, Hofland LJ. Octreotide. N Engl
J Med 1996;​334:​246-54.
73.	Carmichael JD, Bonert VS, Nuño M, Ly
D, Melmed S. Acromegaly clinical trial
methodology impact on reported biochem-
ical efficacy rates of somatostatin receptor
ligand treatments: a meta-analysis. J Clin
Endocrinol Metab 2014;​99:​1825-33.
74.	 Giustina A, Mazziotti G, Torri V, Spi-
nello M, Floriani I, Melmed S. Meta-analy-
sis on the effects of octreotide on tumor
mass in acromegaly. PLoS One 2012;​7(5):​
e36411.
75.	Giustina A, Mazziotti G, Cannavò S,
et al. High-dose and high-frequency lan-
reotide autogel in acromegaly: a random-
ized, multicenter study. J Clin Endocrinol
Metab 2017;​102:​2454-64.
76.	Brzana J, Yedinak CG, Gultekin SH,
Delashaw JB, Fleseriu M. Growth hor-
mone granulation pattern and somatosta-
tin receptor subtype 2A correlate with post-
operative somatostatin receptor ligand
response in acromegaly: a large single
center experience. Pituitary 2013;​16:​490-8.
77.	Schmid HA, Brue T, Colao A, et al.
Effect of pasireotide on glucose- and
growth hormone-related biomarkers in
patients with inadequately controlled ac-
romegaly. Endocrine 2016;​53:​210-9.
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.
n engl j med 382;10  nejm.org  March 5, 2020950
Pituitary-Tumor Endocrinopathies
78.	 Kopchick JJ, Parkinson C, Stevens EC,
Trainer PJ. Growth hormone receptor an-
tagonists: discovery, development, and use
in patients with acromegaly. Endocr Rev
2002;​23:​623-46.
79.	 Barkan AL, Burman P, Clemmons DR,
et al. Glucose homeostasis and safety in
patients with acromegaly converted from
long-acting octreotide to pegvisomant.
J Clin Endocrinol Metab 2005;​90:​5684-91.
80.	van der Lely AJ, Biller BM, Brue T,
et al. Long-term safety of pegvisomant in
patients with acromegaly: comprehensive
review of 1288 subjects in ACROSTUDY.
J Clin Endocrinol Metab 2012;​97:​1589-97.
81.	Neggers SJ, Franck SE, de Rooij FW,
et al. Long-term efficacy and safety of
pegvisomant in combination with long-
acting somatostatin analogs in acromegaly.
J Clin Endocrinol Metab 2014;​99:​3644-
52.
82.	Ragnarsson O, Olsson DS, Chantzi-
christos D, et al. The incidence of Cush-
ing’s disease: a nationwide Swedish
study. Pituitary 2019;​22:​179-86.
83.	Loriaux DL. Diagnosis and differen-
tial diagnosis of Cushing’s syndrome.
N Engl J Med 2017;​376:​1451-9.
84.	Nieman LK. Diagnosis of Cushing’s
syndrome in the modern era. Endocrinol
Metab Clin North Am 2018;​47:​259-73.
85.	Ragnarsson O, Olsson DS, Papakok-
kinou E, et al. Overall and disease-specif-
ic mortality in patients with Cushing dis-
ease: a Swedish nationwide study. J Clin
Endocrinol Metab 2019;​104:​2375-84.
86.	Nieman LK, Biller BM, Findling JW,
et al. Treatment of Cushing’s syndrome:
an Endocrine Society clinical practice
guideline. J Clin Endocrinol Metab 2015;​
100:​2807-31.
87.	Post KD, Pain M, Zarzour H, Beder-
son J. Transsphenoidal surgery for Cush-
ing’s disease. In:​Laws ER Jr, Cohen-Gadol
AA, Schwartz TH, Sheehan JP, eds. Trans-
sphenoidal surgery:​complication avoid-
ance and management techniques. Cham,
Switzerland:​Springer International Pub-
lishing, 2017:​427-39.
88.	Mehta GU, Ding D, Patibandla MR,
et al. Stereotactic radiosurgery for Cush-
ing disease: results of an international,
multicenter study. J Clin Endocrinol
Metab 2017;​102:​4284-91.
89.	 Feelders RA, Newell-Price J, Pivonello
R, Nieman LK, Hofland LJ, Lacroix A. Ad-
vances in the medical treatment of Cush-
ing’s syndrome. Lancet Diabetes Endocri-
nol 2019;​7:​300-12.
90.	Castinetti F, Guignat L, Giraud P,
et al. Ketoconazole in Cushing’s disease:
is it worth a try? J Clin Endocrinol Metab
2014;​99:​1623-30.
91.	Fleseriu M, Biller BM, Findling JW,
Molitch ME, Schteingart DE, Gross C.
Mifepristone, a glucocorticoid receptor
antagonist, produces clinical and meta-
bolic benefits in patients with Cushing’s
syndrome. J Clin Endocrinol Metab 2012;​
97:​2039-49.
92.	Colao A, Petersenn S, Newell-Price J,
et al. A 12-month phase 3 study of pasireo­
tide in Cushing’s disease. N Engl J Med
2012;​366:​914-24.
93.	 Lacroix A, Gu F, Gallardo W, et al. Ef-
ficacy and safety of once-monthly pasi­
reotide in Cushing’s disease: a 12 month
clinical trial. Lancet Diabetes Endocrinol
2018;​6:​17-26.
94.	 Azzalin A, Appin CL, Schniederjan MJ,
et al. Comprehensive evaluation of thyro-
tropinomas: single-center 20-year experi-
ence. Pituitary 2016;​19:​183-93.
Copyright © 2020 Massachusetts Medical Society.
images in clinical medicine
The Journal welcomes consideration of new submissions for Images in Clinical
Medicine. Instructions for authors and procedures for submissions can be found
on the Journal’s website at NEJM.org. At the discretion of the editor, images that
are accepted for publication may appear in the print version of the Journal,
the electronic version, or both.
The New England Journal of Medicine
Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission.
Copyright © 2020 Massachusetts Medical Society. All rights reserved.

More Related Content

What's hot

Chapter 26 chemoprevention of cancer
Chapter 26 chemoprevention of cancerChapter 26 chemoprevention of cancer
Chapter 26 chemoprevention of cancerNilesh Kucha
 
Lymphoma overview
Lymphoma overviewLymphoma overview
Lymphoma overviewderosaMSKCC
 
Acute Myeloid Leukemia
Acute Myeloid LeukemiaAcute Myeloid Leukemia
Acute Myeloid LeukemiavigneshS354
 
A 2020 Renal Cancer update
 A 2020 Renal Cancer update   A 2020 Renal Cancer update
A 2020 Renal Cancer update malcolmbrigden
 
High grade gliomas 8 august 2016
High grade gliomas 8 august 2016High grade gliomas 8 august 2016
High grade gliomas 8 august 2016Gaurav Kumar
 
Chapter 5 hereditary cancer syndrome next generation
Chapter 5 hereditary cancer syndrome next generationChapter 5 hereditary cancer syndrome next generation
Chapter 5 hereditary cancer syndrome next generationNilesh Kucha
 
myeloid malignancy overview
myeloid malignancy overviewmyeloid malignancy overview
myeloid malignancy overviewderosaMSKCC
 
Intresting case roundsmiad
Intresting case roundsmiadIntresting case roundsmiad
Intresting case roundsmiadMiadAlsulami
 
Carcinoma medular de tiroides
Carcinoma medular de tiroidesCarcinoma medular de tiroides
Carcinoma medular de tiroidesmangellozano
 
Neuro-oncology for the Radiation Oncologist - Part 2 - October 2015
Neuro-oncology for the Radiation Oncologist - Part 2 - October 2015Neuro-oncology for the Radiation Oncologist - Part 2 - October 2015
Neuro-oncology for the Radiation Oncologist - Part 2 - October 2015Emil Lou, M.D., Ph.D, FACP
 
ER, PR and HER2 discordance between 1ry and recurrent breast cancer
ER, PR and HER2 discordance between 1ry and recurrent breast cancerER, PR and HER2 discordance between 1ry and recurrent breast cancer
ER, PR and HER2 discordance between 1ry and recurrent breast cancermohamed alhefny
 
Medullary carcinoma of thyroid genene m. bekele, md, face
Medullary carcinoma of thyroid   genene m. bekele, md, faceMedullary carcinoma of thyroid   genene m. bekele, md, face
Medullary carcinoma of thyroid genene m. bekele, md, faceGofasefer
 
Hodgkin lymphoma db.pptx
Hodgkin lymphoma db.pptxHodgkin lymphoma db.pptx
Hodgkin lymphoma db.pptxDipalee Bagal
 
Chemotherapy 101
Chemotherapy 101Chemotherapy 101
Chemotherapy 101derosaMSKCC
 
Hodgkins & Non-Hodgkins Lymphomas
Hodgkins & Non-Hodgkins LymphomasHodgkins & Non-Hodgkins Lymphomas
Hodgkins & Non-Hodgkins LymphomasDinoosh De Livera
 
Improving Treatment Outcomes of Patients With Waldenstrom Macroglobulinemia
Improving Treatment Outcomes of Patients With Waldenstrom MacroglobulinemiaImproving Treatment Outcomes of Patients With Waldenstrom Macroglobulinemia
Improving Treatment Outcomes of Patients With Waldenstrom Macroglobulinemiai3 Health
 
Medicine 5th year, 5th lecture/part one (Dr. Abdulla Sharief)
Medicine 5th year, 5th lecture/part one (Dr. Abdulla Sharief)Medicine 5th year, 5th lecture/part one (Dr. Abdulla Sharief)
Medicine 5th year, 5th lecture/part one (Dr. Abdulla Sharief)College of Medicine, Sulaymaniyah
 

What's hot (20)

Chapter 26 chemoprevention of cancer
Chapter 26 chemoprevention of cancerChapter 26 chemoprevention of cancer
Chapter 26 chemoprevention of cancer
 
Lymphoma overview
Lymphoma overviewLymphoma overview
Lymphoma overview
 
Pineoblastoma
PineoblastomaPineoblastoma
Pineoblastoma
 
Acute Myeloid Leukemia
Acute Myeloid LeukemiaAcute Myeloid Leukemia
Acute Myeloid Leukemia
 
A 2020 Renal Cancer update
 A 2020 Renal Cancer update   A 2020 Renal Cancer update
A 2020 Renal Cancer update
 
High grade gliomas 8 august 2016
High grade gliomas 8 august 2016High grade gliomas 8 august 2016
High grade gliomas 8 august 2016
 
Chapter 5 hereditary cancer syndrome next generation
Chapter 5 hereditary cancer syndrome next generationChapter 5 hereditary cancer syndrome next generation
Chapter 5 hereditary cancer syndrome next generation
 
Nuclear imaging
Nuclear imagingNuclear imaging
Nuclear imaging
 
myeloid malignancy overview
myeloid malignancy overviewmyeloid malignancy overview
myeloid malignancy overview
 
Intresting case roundsmiad
Intresting case roundsmiadIntresting case roundsmiad
Intresting case roundsmiad
 
Carcinoma medular de tiroides
Carcinoma medular de tiroidesCarcinoma medular de tiroides
Carcinoma medular de tiroides
 
Neuro-oncology for the Radiation Oncologist - Part 2 - October 2015
Neuro-oncology for the Radiation Oncologist - Part 2 - October 2015Neuro-oncology for the Radiation Oncologist - Part 2 - October 2015
Neuro-oncology for the Radiation Oncologist - Part 2 - October 2015
 
ER, PR and HER2 discordance between 1ry and recurrent breast cancer
ER, PR and HER2 discordance between 1ry and recurrent breast cancerER, PR and HER2 discordance between 1ry and recurrent breast cancer
ER, PR and HER2 discordance between 1ry and recurrent breast cancer
 
Medullary carcinoma of thyroid genene m. bekele, md, face
Medullary carcinoma of thyroid   genene m. bekele, md, faceMedullary carcinoma of thyroid   genene m. bekele, md, face
Medullary carcinoma of thyroid genene m. bekele, md, face
 
Hodgkin lymphoma db.pptx
Hodgkin lymphoma db.pptxHodgkin lymphoma db.pptx
Hodgkin lymphoma db.pptx
 
Chemotherapy 101
Chemotherapy 101Chemotherapy 101
Chemotherapy 101
 
Hodgkins & Non-Hodgkins Lymphomas
Hodgkins & Non-Hodgkins LymphomasHodgkins & Non-Hodgkins Lymphomas
Hodgkins & Non-Hodgkins Lymphomas
 
Improving Treatment Outcomes of Patients With Waldenstrom Macroglobulinemia
Improving Treatment Outcomes of Patients With Waldenstrom MacroglobulinemiaImproving Treatment Outcomes of Patients With Waldenstrom Macroglobulinemia
Improving Treatment Outcomes of Patients With Waldenstrom Macroglobulinemia
 
Medicine 5th year, 5th lecture/part one (Dr. Abdulla Sharief)
Medicine 5th year, 5th lecture/part one (Dr. Abdulla Sharief)Medicine 5th year, 5th lecture/part one (Dr. Abdulla Sharief)
Medicine 5th year, 5th lecture/part one (Dr. Abdulla Sharief)
 
Neuroblastoma
NeuroblastomaNeuroblastoma
Neuroblastoma
 

Similar to Endocrinopatias hipofisiarias

Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...Sheldon Stein
 
RT in Benign diseases.pptx
RT in Benign diseases.pptxRT in Benign diseases.pptx
RT in Benign diseases.pptxAtulGupta369
 
Bohomolets Oncology Lecture year 5
Bohomolets Oncology Lecture year 5Bohomolets Oncology Lecture year 5
Bohomolets Oncology Lecture year 5Dr. Rubz
 
Metastatic diseases of nervous system
Metastatic diseases of nervous systemMetastatic diseases of nervous system
Metastatic diseases of nervous systemNeurologyKota
 
Tumors of the endocrine system
Tumors of the endocrine systemTumors of the endocrine system
Tumors of the endocrine systemDr./ Ihab Samy
 
Approach to pituitary tumours
Approach to pituitary tumoursApproach to pituitary tumours
Approach to pituitary tumoursVivek Rana
 
Approach to pituitary_tumours
Approach to pituitary_tumoursApproach to pituitary_tumours
Approach to pituitary_tumoursMohit Aggarwal
 
Stereotactic Radiosurgery and Radiotherapy of Pituitary Adenomas Clinical Whi...
Stereotactic Radiosurgery and Radiotherapy of Pituitary Adenomas Clinical Whi...Stereotactic Radiosurgery and Radiotherapy of Pituitary Adenomas Clinical Whi...
Stereotactic Radiosurgery and Radiotherapy of Pituitary Adenomas Clinical Whi...Brainlab
 
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...CrimsonPublishersAICS
 
O.O.Bogomolets National Medical University's Achivement
O.O.Bogomolets National Medical University's AchivementO.O.Bogomolets National Medical University's Achivement
O.O.Bogomolets National Medical University's AchivementMevar Nirav
 
1. cancer care.pdf medical surgical nursing 1
1. cancer care.pdf medical surgical nursing 11. cancer care.pdf medical surgical nursing 1
1. cancer care.pdf medical surgical nursing 1akoeljames8543
 
multipleendocrineneoplasiamensyndromes-180316184612.pdf
multipleendocrineneoplasiamensyndromes-180316184612.pdfmultipleendocrineneoplasiamensyndromes-180316184612.pdf
multipleendocrineneoplasiamensyndromes-180316184612.pdfAsmauBelko
 
Multiple Myeloma
Multiple MyelomaMultiple Myeloma
Multiple MyelomaSadia Sadiq
 
Terminal illness care
Terminal illness careTerminal illness care
Terminal illness careMohammad Asif
 

Similar to Endocrinopatias hipofisiarias (20)

Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
 
low grade glioma.pptx
low grade glioma.pptxlow grade glioma.pptx
low grade glioma.pptx
 
RT in Benign diseases.pptx
RT in Benign diseases.pptxRT in Benign diseases.pptx
RT in Benign diseases.pptx
 
Neuroblastoma and nephroblastoma
Neuroblastoma and nephroblastoma Neuroblastoma and nephroblastoma
Neuroblastoma and nephroblastoma
 
Bohomolets Oncology Lecture year 5
Bohomolets Oncology Lecture year 5Bohomolets Oncology Lecture year 5
Bohomolets Oncology Lecture year 5
 
Metastatic diseases of nervous system
Metastatic diseases of nervous systemMetastatic diseases of nervous system
Metastatic diseases of nervous system
 
Tumors of the endocrine system
Tumors of the endocrine systemTumors of the endocrine system
Tumors of the endocrine system
 
Approach to pituitary tumours
Approach to pituitary tumoursApproach to pituitary tumours
Approach to pituitary tumours
 
Approach to pituitary_tumours
Approach to pituitary_tumoursApproach to pituitary_tumours
Approach to pituitary_tumours
 
Stereotactic Radiosurgery and Radiotherapy of Pituitary Adenomas Clinical Whi...
Stereotactic Radiosurgery and Radiotherapy of Pituitary Adenomas Clinical Whi...Stereotactic Radiosurgery and Radiotherapy of Pituitary Adenomas Clinical Whi...
Stereotactic Radiosurgery and Radiotherapy of Pituitary Adenomas Clinical Whi...
 
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
 
O.O.Bogomolets National Medical University's Achivement
O.O.Bogomolets National Medical University's AchivementO.O.Bogomolets National Medical University's Achivement
O.O.Bogomolets National Medical University's Achivement
 
1. cancer care.pdf medical surgical nursing 1
1. cancer care.pdf medical surgical nursing 11. cancer care.pdf medical surgical nursing 1
1. cancer care.pdf medical surgical nursing 1
 
multipleendocrineneoplasiamensyndromes-180316184612.pdf
multipleendocrineneoplasiamensyndromes-180316184612.pdfmultipleendocrineneoplasiamensyndromes-180316184612.pdf
multipleendocrineneoplasiamensyndromes-180316184612.pdf
 
Neuroblastoma
Neuroblastoma Neuroblastoma
Neuroblastoma
 
Multiple Myeloma
Multiple MyelomaMultiple Myeloma
Multiple Myeloma
 
Tumor Immunology
Tumor ImmunologyTumor Immunology
Tumor Immunology
 
Testicular tumours
Testicular tumoursTesticular tumours
Testicular tumours
 
Tumor targeting
Tumor targetingTumor targeting
Tumor targeting
 
Terminal illness care
Terminal illness careTerminal illness care
Terminal illness care
 

Recently uploaded

Isotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on IoIsotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on IoSérgio Sacani
 
Nanoparticles synthesis and characterization​ ​
Nanoparticles synthesis and characterization​  ​Nanoparticles synthesis and characterization​  ​
Nanoparticles synthesis and characterization​ ​kaibalyasahoo82800
 
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Sérgio Sacani
 
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |aasikanpl
 
Work, Energy and Power for class 10 ICSE Physics
Work, Energy and Power for class 10 ICSE PhysicsWork, Energy and Power for class 10 ICSE Physics
Work, Energy and Power for class 10 ICSE Physicsvishikhakeshava1
 
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxAnalytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxSwapnil Therkar
 
Recombinant DNA technology (Immunological screening)
Recombinant DNA technology (Immunological screening)Recombinant DNA technology (Immunological screening)
Recombinant DNA technology (Immunological screening)PraveenaKalaiselvan1
 
Cultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxCultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxpradhanghanshyam7136
 
Biological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfBiological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfmuntazimhurra
 
Boyles law module in the grade 10 science
Boyles law module in the grade 10 scienceBoyles law module in the grade 10 science
Boyles law module in the grade 10 sciencefloriejanemacaya1
 
Scheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxScheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxyaramohamed343013
 
G9 Science Q4- Week 1-2 Projectile Motion.ppt
G9 Science Q4- Week 1-2 Projectile Motion.pptG9 Science Q4- Week 1-2 Projectile Motion.ppt
G9 Science Q4- Week 1-2 Projectile Motion.pptMAESTRELLAMesa2
 
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...Sérgio Sacani
 
VIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C PVIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C PPRINCE C P
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real timeSatoshi NAKAHIRA
 
Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )aarthirajkumar25
 
zoogeography of pakistan.pptx fauna of Pakistan
zoogeography of pakistan.pptx fauna of Pakistanzoogeography of pakistan.pptx fauna of Pakistan
zoogeography of pakistan.pptx fauna of Pakistanzohaibmir069
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfSwapnil Therkar
 
A relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfA relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfnehabiju2046
 

Recently uploaded (20)

Isotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on IoIsotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on Io
 
Nanoparticles synthesis and characterization​ ​
Nanoparticles synthesis and characterization​  ​Nanoparticles synthesis and characterization​  ​
Nanoparticles synthesis and characterization​ ​
 
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
 
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
 
Work, Energy and Power for class 10 ICSE Physics
Work, Energy and Power for class 10 ICSE PhysicsWork, Energy and Power for class 10 ICSE Physics
Work, Energy and Power for class 10 ICSE Physics
 
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxAnalytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
 
Recombinant DNA technology (Immunological screening)
Recombinant DNA technology (Immunological screening)Recombinant DNA technology (Immunological screening)
Recombinant DNA technology (Immunological screening)
 
Cultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxCultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptx
 
Biological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfBiological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdf
 
Boyles law module in the grade 10 science
Boyles law module in the grade 10 scienceBoyles law module in the grade 10 science
Boyles law module in the grade 10 science
 
Scheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxScheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docx
 
G9 Science Q4- Week 1-2 Projectile Motion.ppt
G9 Science Q4- Week 1-2 Projectile Motion.pptG9 Science Q4- Week 1-2 Projectile Motion.ppt
G9 Science Q4- Week 1-2 Projectile Motion.ppt
 
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
 
VIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C PVIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C P
 
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real time
 
Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )
 
zoogeography of pakistan.pptx fauna of Pakistan
zoogeography of pakistan.pptx fauna of Pakistanzoogeography of pakistan.pptx fauna of Pakistan
zoogeography of pakistan.pptx fauna of Pakistan
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
 
A relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfA relative description on Sonoporation.pdf
A relative description on Sonoporation.pdf
 

Endocrinopatias hipofisiarias

  • 1. The new engl and jour nal of medicine n engl j med 382;10  nejm.org  March 5, 2020 937 Review Article From the Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles. Address reprint requests to Dr. Melmed at Cedars-Sinai Medical Center, 8700 Beverly Blvd., Rm. 2015, Los Angeles, CA 90048. N Engl J Med 2020;382:937-50. DOI: 10.1056/NEJMra1810772 Copyright © 2020 Massachusetts Medical Society. P ituitary adenomas account for approximately 15% of intracra- nial tumors.1 Management of these benign tumors requires diagnosis of the specific intrasellar disease and comprehensive, multidisciplinary treatment of local mass effects and peripheral endocrinopathies.2 Since tumors can produce different hormones, their consequences and management vary widely. Pathogenesis Differentiated, hormone-expressing pituitary cell lineages may give rise to adeno- mas, often coupled with autonomous hormone hypersecretion. Distinct hyper­ secretory syndromes depend on the cell of origin: corticotropin-secreting corti- cotroph adenomas result in Cushing’s disease, growth hormone–secreting somatotroph adenomas result in acromegaly, prolactin-secreting lactotroph adeno- mas result in hyperprolactinemia, and thyrotropin-secreting thyrotroph adenomas result in hyperthyroidism. Gonadotroph adenomas, which are typically nonsecreting, lead to hypogonadism and often manifest incidentally as a sellar mass3-5 (Fig. 1). Permissive hypothalamic hormone and paracrine proliferative signals lead to a dysregulated pituitary cell cycle, with aneuploidy, chromosomal copy-number variation, and cellular senescence restraining malignant transformation.6-8 Although mutations in GNAS (the gene for the stimulatory alpha subunit of a guanine nucleo- tide–binding protein [G-protein] that stimulates adenylate cyclase) and USP8 (the gene for ubiquitin carboxyl terminal hydrolase 8, a ubiquitin-specific protease) occur in a subgroup of nonfamilial growth hormone–secreting tumors and corti- cotropin-secreting tumors, respectively,9 genetic evaluation of sporadic adenomas is rarely helpful for management. The prevalence of pituitary adenomas has increased to 115 cases per 100,000 population over the past several decades, probably as a result of enhanced aware- ness and improved diagnostic imaging and hormone assays.10 The relative preva- lence of prolactinomas (54 cases per 100,000 population) and nonfunctioning adenomas (42 per 100,000) may reflect a reporting bias with respect to surgical versus nonsurgical series, since most prolactinomas are treated medically and are not captured in reports of surgical diagnoses. Classification Microadenomas are less than 10 mm in diameter. Regardless of the cell origin, macroadenomas (≥10 mm) may impinge on critical parasellar vascular and neural structures, with resultant visual-field defects, including bitemporal hemianopia and decreased acuity, and headaches.5 Immunocytochemical evaluation of differ- entiated, cell-specific pituitary transcription factors and hormones,11 as well as clear biochemical, imaging, and clinical phenotypes, define tumor characteristics Dan L. Longo, M.D., Editor Pituitary-Tumor Endocrinopathies Shlomo Melmed, M.D.​​ The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 2. n engl j med 382;10 nejm.org March 5, 2020938 The new engl and jour nal of medicine and endocrine syndromes, allowing for indi- vidualized treatment5,12 (Table 1). Evaluation of a pituitary mass should include magnetic resonance imaging (MRI) and visual- field examination for accurate tumor localiza- tion and assessment of local compressive mass effects. Hormone hypersecretion should be as- sessed to distinguish nonsecretory from secretory tumors13 (Table 2), and pituitary-reserve function should be tested.14 Approximately 30% of surgically resected adenomas have persistent or progressive post- operative growth for up to four decades or even longer, with local invasion and an increased percentage of cells positive for Ki-67.11,15 In one study, more than 40% of 50 aggressive pituitary adenomas showed cavernous sinus invasion.16 Tumors particularly prone to invasive growth and recurrence include those arising from sparsely granulated somatotrophs, silent corti- cotrophs, corticotroph Crooke’s cells (CK20- positive nonneoplastic cells with a prominent Figure 1. Pathogenesis of Pituitary Tumors. Pituitary adenomas arise from a differentiated hormone-expressing cell or from a null cell. Clinical phenotype is determined by the cell of origin and the presence or absence of autonomous, specific hormone hypersecretion. The prevalence data are estimates. IGF-1 de- notes insulin-like growth factor receptor 1, SF1 steroidogenic factor 1, and Tpit T-box factor, pituitary. Normal pituitary Microadenoma Macroadenoma Aggressive adenoma SOMATOTROPH LACTOTROPH CORTICOTROPH THYROTROPH GONADOTROPH ANY Growth hormoneSecreted hormone Cell type Prolactin Corticotropin Thyrotropin Luteinizing hormone or follicle-stimulating hormone or subunits None 8–15Relative prevalence (%) 30–60 2–6 <1 <1 14–55 POU1F1Transcription factors POU1F1 Estrogen receptor alpha Tpit POU1F1 GATA2 SF1 GATA2 Variable IGF-1Target hormone Prolactin Cortisol T4 or T3 Estrogen or testosterone None Acromegaly Gigantism Clinical phenotype Hypogonadism Infertility Galactorrhea Cushing’s disease Hypercortisolemia Hyperthyroidism Hypergonadism Pituitary failure Densely granulated Sparsely granulated Mammosomatotroph Mixed somatolactotroph Acidophil stem cell Adenoma subtypes Densely granulated Sparsely granulated Acidophil stem cell Densely granulated Sparsely granulated Crooke’s cell Null cell Silent gonadotroph Silent corticotroph Silent somatotroph Pituitary gland Carotid artery Cavernous sinus Optic chiasm Cranial nerves III IV VI V1 V2 Aneuploidy Copy-number alterations Germline or somatic mutations Cell-cycle dysregulation Paracrine stromal events Epigenetic changes Unknown driver changes Proliferative restraint Senescence Sphenoid sinus The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 3. n engl j med 382;10  nejm.org  March 5, 2020 939 Pituitary-Tumor Endocrinopathies Table 1. Individualized Approach to Pituitary Adenoma Management.* Characteristic Finding or Outcome Patient Age <30 yr More aggressive growth and recurrence, florid hormone phenotype (growth hormone) >60 yr Less aggressive features, smaller tumor (growth hormone), larger tumor (prolactin) Sex Female More microadenomas (prolactin and corticotropin) Male More macroadenomas, higher hormone levels (prolactin) Tumor MRI findings Size Larger tumors are more resistant to treatment Invasiveness Invasive tumors are more likely to recur Intensity on T2-weighted images Intensity determines somatostatin receptor ligand responsiveness (growth hormone) Histologic features Dense granules Indolent course, responsive to somatostatin receptor ligand (growth hormone) Sparse granules Florid course, resistant to somatostatin receptor ligand (growth hormone) Receptor expression Positive for SST2 Likely to respond to octreotide or lanreotide (growth hormone) Positive for SST5 Likely to respond to pasireotide (corticotropin and growth hormone) Positive for D2 Likely to respond to dopamine agonist (prolactin) Negative for SST2, SST5, and D2 Likely to be treatment-resistant Ki-67 cell-cycle marker Recurrence less likely if <3%; recurrence more likely if ≥3% GSP mutation Likely to respond to somatostatin receptor ligand (growth hormone) Treatment Transsphenoidal surgical resection Advantages: rapid hormone and symptom remission, one-time cost, potential for cure, decompression of vital parasellar structures, tumor debulking may enhance adju- vant therapy Disadvantages: persistent tumor remnant, postoperative hypopituitarism requiring life- long replacement therapy, some patients are not candidates for surgery Risks: diabetes insipidus, electrolyte abnormalities, neurologic deficits, CSF rhinorrhea, death from anesthetic agent (rarely); other risks include resection performed by low- volume surgeons, as well as coexisting cardiac disease, cerebrovascular disease, or diabetes Radiation therapy Advantages: permanent results, one-time cost, long-term treatment not required, no drug-related adverse events Disadvantages: very slow efficacy onset, medical therapy required until effects are ­evident Risks: hypopituitarism, visual disturbances, stroke; rarely, CNS damage, brain tumor Pituitary-directed medical therapy Advantages: hormone control and symptom relief (prolactin, growth hormone, cortico- tropin, thyrotropin), no hypopituitarism, tumor-mass control Disadvantages: cure not permanent, long-term sustained treatment required, ongoing cost, injection adherence required Risks regarding somatostatin receptor ligand: gallstones or sludge, diarrhea, nausea, hyperglycemia, sinus bradycardia, alopecia, injection-site pain, headache Risks regarding D2 agonist: depression, nausea, vasospasm * Hormones in parentheses are those secreted by the adenoma. CNS denotes central nervous system, CSF cerebrospinal fluid, and SST2 and SST5 somatostatin receptor subtype 2 and subtype 5, respectively. The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 4. n engl j med 382;10  nejm.org  March 5, 2020940 The new engl and jour nal of medicine cytoplasmic hyaline ring that displaces the nor- mal basophilic granules of corticotropic cells), and lactotrophs in middle-aged or older men.17 Pituitary carcinomas are exceedingly rare. They account for less than 0.5% of pituitary tumors and respond inconsistently to temozolomide.5 Management Comprehensive management of a pituitary ade- noma includes transsphenoidal surgical resec- tion, irradiation, and medical therapy, each with advantages and disadvantages that are specific to the type of adenoma (Table 1). Serial or com- bined approaches may be required. Surgery is generally indicated for masses that are 10 mm or more in diameter and those that have extrasellar extension or central compressive features, as well as for persistent tumor growth, especially if vision is compromised or threat- ened.18 Resection may alleviate compression of vital structures and reverse compromised pitu- itary hormone secretion. Predictors of remission include experience of the surgeon, relatively low levels of secreted hormone (if the level is elevated at all), and small tumors. Postoperatively, hypo- pituitarism may develop, as may diabetes insipi- dus and cerebrospinal fluid leaks. Approximately 10% of patients have a recurrence over a period of 10 years after surgery. Persistent tumor post- operatively may reflect incomplete resection, in- accessible cavernous sinus tumor tissue, or dural nesting of hormone-secreting tumor cells. Radiation therapy, administered by means of conventional external-beam or proton-beam tech- niques, or stereotactic radiosurgery requires local expertise and is generally reserved for tumors that are resistant to medical treatment or are not controlled by surgery. Tumor growth is usually arrested over a period of several years, and ade- noma-derived hormone hypersecretion may per- sist during the initial years.19 In most patients, pituitary failure develops within 10 years after radiation therapy, and lifelong hormone replace- ment is required. Deterioration in vision and new cranial-nerve palsies are rarely observed.19 Mortal- ity from cerebrovascular causes is increased by a factor of 4.42 (95% confidence interval [CI], 2.71 to 7.22) after conventional pituitary irradiation, with 16 observed deaths versus 3.6 expected deaths.20 Genetic Syndromes Pituitary adenomas may occur in association with several very rare genetic syndromes. Multi- ple endocrine neoplasia type 1 is associated with pituitary adenomas as well as parathyroid and pancreatic-islet tumors and, less commonly, carci- noid, thyroid, and adrenal tumors. The McCune– Albright syndrome is characterized by polyostotic fibrous dysplasia and cutaneous pigmentation, with sexual precocity, hyperthyroidism, hypercor- Table 2. Testing to Diagnose Hormone-Secreting Pituitary Tumors.* Tumor or Disease Test Results Requiring Further Evaluation Prolactinoma Serum prolactin level measurement Prolactin level elevated Acromegaly Serum IGF-1 level Age-adjusted IGF-1 level elevated Oral glucose-tolerance test (75 g of glucose) with growth hormone measured at 0, 30, and 60 min Growth hormone level 0.4 μg/liter with the use of an ultra- sensitive assay Cushing’s disease 24-hr urinary free cortisol measurement Elevated urinary free cortisol level on at least two tests Midnight salivary cortisol measurement Elevated free salivary cortisol level Plasma cortisol measurement at 8 a.m., after administration of dexamethasone (1 mg) at 11 p.m. Failure to suppress cortisol level to 1.8 μg/dl Serum corticotropin measurement Low corticotropin level suggests adrenal adenoma; very high level may indicate ectopic corticotropin source Thyrotropin-secreting tumor Serum thyrotropin measurement, free T4 ­measurement Normal or increased free T4 level with measurable thyrotropin may suggest thyrotropin-secreting tumor * IGF-1 denotes insulin-like growth factor 1. The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 5. n engl j med 382;10  nejm.org  March 5, 2020 941 Pituitary-Tumor Endocrinopathies tisolism, hyperprolactinemia, and acromegaly. Rare cases of familial pituitary adenomas have been reported in families with a predisposition for somatotroph tumors in childhood or young adulthood, and about 25% of these tumors have been linked to germline mutations in AIP (the gene for aryl hydrocarbon receptor–interacting protein),21 which are not commonly encountered with sporadic adenomas. The Carney complex includes pituitary adenomas with benign cardiac myxomas, schwannomas, thyroid adenomas, and pigmented skin spots.22 Nonsecreting Adenomas Although several lesions may present as nonse- creting sellar masses23 (Fig. 2), most are non­ secreting adenomas of gonadotroph lineage. These adenomas express differentiated hormone products and cell-specific transcription factors, but since the respective peripheral-blood hor- mone levels are not elevated, the adenomas are not associated with systemic syndrome pheno- types.24 True null-cell adenomas express no hormone gene product. Figure 2. Approach to Patients Presenting with a Nonsecreting Pituitary Mass. The main diagnostic consideration is to distinguish a nonsecreting or secreting adenoma from a nonadenomatous lesion. The treat- ment approach is determined by the specific diagnosis. The dashed line indicates an option that may be considered under some cir­ cumstances. Pituitary Mass Nonsecreting Adenoma Features Management according to specific hypersecretory syndrome Management as for non- adenomatous parasellar lesion Cysts Rathke Craniopharyngioma Arachnoid, epidermoid, pineal Neoplasms Meningioma Chordoma Lymphoma Pituicytoma Germinoma Sarcoma Hemangiopericytoma Mucoepidermoid carcinoma Xanthogranuloma Lipoma Vascular Apoplexy Aneurysm Angioma or hematoma Inflammatory Hypophysitis Amyloidosis, sarcoidosis Granulomatous polyangiitis Infections Bacterial Syphilis, tuberculosis Metastases Breast, lung, prostate, sinus Lymphoma, plasmacytoma Miscellaneous Ectopic pituitary Fibrous dysplasia Microadenoma (10 mm) Macroadenoma (≥10 mm) Screening for hormone hypersecretion and assessing pituitary function MRI differential diagnosis Observation, if no compressive mass effects Transsphenoidal surgery Follow-up with MRI at 1, 2, and 5 yr Reassessment if symptomatic MRI annually to monitor for mass persistence or recurrence Pituitary-reserve testing every 6 mo for 2 yr Hormone replacement as required Incidental MRI finding Neurologic Visual-field deficits Extraocular muscle palsy Headache Endocrine Amenorrhea Decreased libido Infertility Diabetes insipidus Apoplexy Pituitary hormonal deficiencies Immunostaining Gonadotropin subunits Proopiomelanocortin or corticotropin Growth hormone None The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 6. n engl j med 382;10  nejm.org  March 5, 2020942 The new engl and jour nal of medicine Clinical Features and Diagnosis Nonsecreting adenomas may be unrecognized for years and are usually diagnosed because of local mass effects or hypogonadism or are de- tected incidentally. Chiasmal compression causes gradual, progressive deficits in vision, and about two thirds of patients have decreased gonadotro- pin levels and hypogonadism.25 In a study involv- ing 385 consecutive patients with nonsecreting adenomas, 289 had macroadenomas and 66 had giant adenomas (4 cm in diameter), with most of the patients presenting with headache or dis- turbances in vision.26 Approximately 10% of pa- tients with nonsecreting adenomas present with pituitary apoplexy (characterized by the abrupt onset of retroorbital pain, altered consciousness, Figure 3. Approach to Patients Presenting with Hyperprolactinemia. In patients with hyperprolactinemia, the clinical challenge is to rule out a secondary, nonpituitary cause before embarking on comprehen- sive pituitary imaging and testing. The stalk effect is compression of the pituitary stalk due to a nonsecreting sellar mass that interrupts delivery of inhibitory dopamine from the hypothalamus to the prolactin-secreting cells of the pituitary. HyperprolactinemiaFeatures Rule out secondary causes Consider nonsecreting mass resulting in the stalk effect Pregnancy Drugs Neuroleptics D2 receptor blockers Estrogens Opiates Antidepressants Parasellar mass Hypothalamic irradiation Chest-wall injury Renal failure Assess pituitary- reserve function Test visual fields Pituitary MRI Dopamine agonist treatment Dose adjustment Unacceptable side effects Prolactin level not reduced No tumor shrinkage Prolactin level normalized Sexual function restored Tumor shrinkage Prolactin level reduced Sexual function restored Tumor persists with no local compressive effects Decrease dose Consider surgery if hyper- prolactinemia or mass persists Maximize dose Continue medication Continue medication Surgery or radiotherapy required in rare cases Reconsider diagnosis of nonsecreting adenoma with stalk effect Surgery or radiotherapy required in rare cases If prolactin level rises, repeat MRI If prolactin level rises, repeat MRI Compressive effects Microprolactinoma Macroprolactinoma Amenorrhea, oligomenorrhea, infertility Impotence, oligospermia, decreased libido Galactorrhea Osteoporosis The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 7. n engl j med 382;10  nejm.org  March 5, 2020 943 Pituitary-Tumor Endocrinopathies ophthalmoplegia, and ultimately, loss of vision from pituitary hemorrhage or infarction). Very rarely, circulating gonadotropin levels are elevated, resulting in ovarian hyperstimula- tion or increased testicular volumes. Paradoxi- cally, however, high gonadotropin levels more commonly down-regulate the gonadal axis. Clinically silent tumors expressing nonsecret- ed corticotropin or growth hormone account for up to 20% of nonsecreting tumors. They are usually resected after a nonsecreting macroade- noma has been diagnosed, with the diagnosis subsequently confirmed by histologic assess- ment with appropriate immunostaining. These silent tumors grow aggressively, with no appar- ent features of hypercortisolism or acromegaly, although their morphologic features may be in- distinguishable from those of secretory adeno- mas. A systematic review of 14 studies with a total of 297 patients who underwent surgical treatment for silent corticotroph adenomas showed that 31% of the adenomas recurred dur- ing a follow-up period of more than 5 years.17 Treatment Complete resection of a nonsecreting macro­ adenoma is achieved in approximately 65% of patients, with visual function restored in up to 80% of the patients and hypopituitarism, when present, reversed in approximately 50%.26,27 Pre- operative tumor invasiveness largely determines the risk of postoperative persistent or recurrent tumor requiring adjuvant irradiation and reopera- tion. Radiosurgery was associated with high rates of tumor control in a multicenter analysis involving 512 patients followed for a median of 36 months, and pituitary failure developed in 21% of the patients.28 In a retrospective analysis of tumor growth rates among 237 patients who were followed for a median of 5.9 years, 36% of the patients had a recurrence after surgery alone, whereas 13% had a recurrence after sur- gery and adjuvant radiotherapy.29 Guidelines13 recommend follow-up with peri- odic pituitary MRI, vision evaluations, and pitu- itary-function testing, and prophylactic radiother- apy to prevent postoperative tumor recurrence or progression has been suggested. Of 648 inciden- tally discovered pituitary adenomas, 10% of the 229 microadenomas and 20% of the 419 macro­ adenomas increased in size during up to 8 years of follow-up.30 Accordingly, expectant observation is recommended for the management of stable, nonsecreting microadenomas and small mac- roadenomas. Preoperative or newly manifested postoperative pituitary failure may be insidious. In a study involving 2795 patients, pituitary fail- ure, especially with corticotropin deficiency, was associated with excess overall mortality (stan- dardized mortality ratio, 4.35; 95% CI, 1.99 to 8.26), on the basis of 2.1 expected deaths versus 9 observed deaths (P0.001).31 Prolactin-Secreting Adenomas Prolactinomas are the most common secretory tumor, accounting for up to 60% of all pituitary adenomas and more than 75% of pituitary ade- nomas in women.32 Microprolactinomas, which have a female:male ratio of 20:1, are usually stable and slow-growing, with continued growth after diagnosis in less than 15% of cases.33 Clinical Features Most patients with a pituitary mass and a serum prolactin level exceeding 150 ng per milliliter (reference range, 20) are found to have a pro- lactinoma (Fig. 3). A prolactin level of more than 250 ng per milliliter is usually diagnostic of a macroprolactinoma, and the tumor mass usually correlates with the serum prolactin level.34 Large prolactinomas (10 mm in diameter) that are aggressive account for less than 5% of tumors and are characterized by very high prolactin lev- els (1000 ng per milliliter) and a male:female ratio of 9:1. In a study involving 45 men and 51 women with prolactinomas, the tumors in the men were larger than those in the women (mean [±SD] diameter, 26±2 mm vs. 10±1 mm) and grew more aggressively, with a mean serum prolactin level of 2789±572 ng per milliliter in men versus 292±74 ng per milliliter in women.35 Persistently elevated prolactin levels suppress gonadotropin, leading to amenorrhea, oligo- menorrhea, or a short luteal phase associated with infertility in women and low libido, impo- tence, oligospermia, or azoospermia in men. About 50% of women and 35% of men have galactorrhea, and both women and men have reduced bone density, often associated with sex steroid hormone deficiency and an increased risk of vertebral fractures. The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 8. n engl j med 382;10  nejm.org  March 5, 2020944 The new engl and jour nal of medicine Diagnosis Hyperprolactinemia is caused mainly by preg- nancy, prolactinomas, medications, chest-wall injury, and functional or mechanical interrup- tion of pituitary-stalk dopamine transport. Pro- lactin levels are elevated in approximately 30% of patients with acromegaly.36 Prolactin levels should be measured in all patients with a sellar mass; conversely, hyper­ prolactinemia not explained by pregnancy or exposure to neuroleptic drugs should prompt pituitary imaging to rule out a pituitary mass. In patients with hyperprolactinemia and a pituitary mass, failure to achieve tumor shrinkage with dopamine agonists may suggest compression of the pituitary stalk due to a nonsecreting sellar mass that interrupts delivery of inhibitory dopa- mine from the hypothalamus, resulting in dis- ruption of prolactin control; this is known as the stalk effect. Treatment Prolactin levels should be normalized, with sex- ual function and fertility restored, galactorrhea halted, and the tumor mass eliminated or re- duced, while pituitary function is retained. Low bone density should be addressed. Prolactinomas are ideally managed with a dopamine agonist,33 which should lower prolac- tin levels and shrink the tumor mass. Bromo­ criptine is rarely used, since it requires daily dosing. One study showed that cabergoline, ad- ministered at a dose of 0.5 to 1 mg once or twice weekly, lowered prolactin levels in 83% of 459 women with hyperprolactinemia,37 and this agent restores ovulatory cycles and fertility with few side effects. In approximately 65% of patients with macroprolactinomas, treatment normalizes prolactin levels and reduces the tumor mass.38 Up to 15% of patients do not have a response to maximal dopaminergic doses (i.e., prolactin levels are not normalized, and tumor shrinkage is 50%). Patients with normalized prolactin levels but inadequate tumor shrinkage may re- quire surgery or radiotherapy. Hyperprolactinemia remits in up to 20% of patients after dose tapering and discontinuation of cabergoline, which can be attempted after more than 2 years of therapy and only when the possibility of tumor invasion has been rigor- ously ruled out.39 Adverse effects of cabergoline, reported in up to 50% of patients, include nausea, nasal stuffi- ness, depression, digital vasospasm, postural hypotension, and in rare cases, cerebrospinal fluid leak. Mood disorders, exacerbation of psy- chosis, and poor impulse control are additional rare effects.40 The low doses of cabergoline used for the treatment of prolactinomas do not ap- pear to place patients at risk for clinically sig- nificant cardiac valve disease, and valvulopathy was not observed in 192 patients who were fol- lowed for 34 months in a cross-sectional study.41 Because asymptomatic, mild tricuspid regurgita- tion was reported in 20% of patients, those treated with cabergoline in whom a cardiac mur- mur develops should undergo cardiac evaluation. In 31 surgical series involving a total of 1224 patients with microprolactinomas, transsphe- noidal resection resulted in normal prolactin levels in 71% of the patients, and initial cure rates may exceed 90% when resection is per- formed by high-volume surgeons.42 About 50% of macroprolactinomas remit, and persistent, postoperative hyperprolactinemia arises from tumor remnants.42 The efficacy of treatment with dopamine agonists and the long-term risk of postoperative recurrence have discouraged clinicians from choosing surgery as a primary treatment for macroprolactinomas. Invasive, dopamine agonist–resistant prolactinomas may require more than one surgery, with continued administration of high-dose cabergoline.43 Ra- diation therapy is reserved for patients with treatment-resistant macroprolactinomas. During pregnancy, pituitary expansion, espe- cially in the case of macroprolactinomas, may threaten the visual fields.44 Prophylactic trans­ sphenoidal resection should be considered when vision is threatened. Dopamine agonist therapy should be discontinued when pregnancy is con- firmed. Acromegaly Acromegaly, with an incidence of approximately 10 cases per 1 million persons,32,45 is caused by a growth hormone–secreting somatotroph tumor. High growth hormone and insulin-like growth factor 1 (IGF-1) levels are associated with strik- ing somatic and metabolic dysfunction. Densely granulated somatotroph adenomas arise insidi- ously, whereas sparsely granulated subtypes, which develop in younger patients, are character- The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 9. n engl j med 382;10  nejm.org  March 5, 2020 945 Pituitary-Tumor Endocrinopathies ized by aggressive growth and florid disease.12 In very rare cases, extrapituitary acromegaly is caused by neuroendocrine tumor production of growth hormone or growth hormone–releasing hormone.46,47 Clinical Features Approximately 70% of patients with acromegaly have an invasive macroadenoma at diagnosis. Coexisting conditions include headache and in- sidious acral and soft-tissue changes. The diag- nosis of acromegaly can be delayed by a mean of approximately 10 years after the onset of symp- toms.48 Patients may first seek dental, orthope- dic, rheumatologic, or cardiac care. In one study, approximately 20% of 324 patients sought care because of an altered facial appearance, enlarged extremities, or both.49 Other features include increased shoe or ring size, voice deepening, the carpal tunnel syndrome, hyperhidrosis, and frontal-skull bossing and coarse oily skin. Prog- nathism leads to incisor separation and jaw malocclusion. Obstructive sleep apnea and ex- cessive snoring are hallmarks of uncontrolled acromegaly. Arthropathy is reported in approxi- mately 70% of patients, with polyarticular arthri- tis, osteophytosis, dorsal kyphosis, and vertebral fractures.48 Coexisting cardiovascular conditions include hypertension, arrhythmias, and left ven- tricular dysfunction, with an increased aortic- root diameter.50 Despite biochemical control, cardiovascular disorders may persist.51 Growth hormone induces insulin resistance, with glu- cose intolerance,52 and patients with acromegaly have an increased risk of diabetes, as compared with a control cohort from the general popula- tion (hazard ratio, 4.0 [95% CI, 2.7 to 5.8]; rate per 1000 persons, 12.1 cases [95% CI, 9.0 to 16.4] vs. 3.4 cases [95% CI, 2.9 to 4.1]).53 About 30% of patients have high prolactin levels, often with galactorrhea. Hypertrophic colonic mucosal folds and diver- ticulae can occur in patients with acromegaly.54,55 In a case–control study, colonic polyps were detected in 32% of 165 patients, with an esti- mated relative risk of 6.21 (95% CI, 4.08 to 9.48).56 Studies involving more than 2000 pa- tients showed an increased incidence of cancer, with an overall standardized incidence ratio of 1.5 (95% CI, 1.2 to 1.8), mostly accounted for by colorectal, kidney, and thyroid cancers.57,58 How- ever, other studies have shown no increase in the incidence of cancer. Nevertheless, colonoscopy should be performed at diagnosis and subse- quently, according to published guidelines.59 A 20-year study showed higher mortality rates among 333 patients with acromegaly than among 4995 controls (113 deaths [34%] vs. 1334 deaths [27%]; odds ratio 1.6; 95% CI, 1.2 to 2.2).60 Deaths associated with acromegaly are due to cardiovascular, respiratory, and cerebrovascular disorders, with cancer also reported as a cause of death in more recent years. Persistently elevat- ed growth hormone and IGF-1 levels, diabetes, hypertension, older age, pituitary irradiation, and inadequate treatment of adrenal insuffi- ciency all contribute significantly to mortality.61 Gigantism, a rare condition, is due to exces- sive secretion of growth hormone before epiphy- seal closure. The disorder may be associated with germline AIP mutations, the McCune–Albright syndrome, or acidophilic stem-cell adenomas; X-linked gigantism is characterized by Xq26.3 chromosomal microduplications, resulting in accelerated gigantism and overexpressed tumor GPR101 (a gene for a G-protein–coupled recep- tor).62 Surgical resection with adjuvant growth hormone suppression is required to maintain remission and prevent long-term tissue exposure to excess growth hormone and IGF-1.63 Diagnosis IGF-1 levels that are elevated for the patient’s age are highly specific for acromegaly and also cor- relate with indexes of disease activity.64 The pulsatile nature of adenoma growth hormone secretion precludes reliance on a random mea- surement for diagnosis. Instead, the diagnosis is established by using an ultrasensitive assay to document nadir growth hormone levels of more than 0.4 μg per liter during a 75-g glucose load.65 Growth hormone levels are log-linearly concordant with IGF-1 levels.66 Postoperatively, IGF-1 levels may remain elevated for months despite control of growth hormone secretion. Treatment Comprehensive treatment goals for acromegaly include ablating or controlling the pituitary mass, suppressing growth hormone and IGF-1 hypersecretion, and preventing the development of associated disorders while maintaining ante- rior pituitary function.59 In 13 studies involving 1018 patients who underwent surgical resection, The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 10. n engl j med 382;10  nejm.org  March 5, 2020946 The new engl and jour nal of medicine control of growth hormone secretion and IGF-1 levels was achieved in 73% of patients with micro- adenomas and 61% of patients with macroade- nomas.67-69 However, patients with macroadeno- mas that invade the cavernous sinus invariably show persistent growth hormone hypersecretion after surgery. In a study involving 371 patients treated with radiosurgery, biochemical remission was reported in 59% of the patients, with a mean time to remission of 38 months and a mean time to recurrence of 17 months.70 Dopamine agonists have been proposed for patients with mild disease, and the addition of cabergoline may normalize IGF-1 levels in some patients with disease that is resistant to somato­ statin therapy.71 The somatostatin receptor ligands octreotide and lanreotide bind SST2 (somatosta- tin receptor subtype 2), inhibiting growth hor- mone secretion.72 A meta-analysis of 90 studies involving 4464 patients treated with somatosta- tin receptor ligands showed overall control of growth hormone secretion and of IGF-1 levels in 56% and 55% of the patients, respectively.73 In a meta-analysis involving 1685 patients in 41 stud- ies, 53% of patients had a reduction in tumor mass.74 Efficacy may be enhanced by increasing the dose and injection frequency.75 Soft-tissue swelling and headache usually resolve, sleep ap- nea abates, and left ventricular function im- proves,48 but hypertension may persist. Tumor SST2 expression, densely granulated tumor, and hypointensity on T2-weighted MRI are important markers of treatment responsiveness.12,76 Tran- sient gastrointestinal side effects, occurring in approximately 30% of patients, include loose stools and nausea. Gallbladder sludge occurs in up to 25% of patients, although cholecystitis is very rare. Asymptomatic sinus bradycardia also occurs. Octreotide and lanreotide do not gener- ally disrupt glucose homeostasis, but pasireotide, a long-acting hexapeptide somatostatin multi- receptor ligand, results in hyperglycemia and new-onset diabetes in about 60% of patients.77 Pegvisomant, a growth hormone receptor antagonist that blocks peripheral growth hor- mone action and subsequent IGF-1 production,78 is useful for patients with disease that is resis- tant to somatostatin receptor ligands, as well as patients with hyperglycemia, since the drug en- hances insulin sensitivity.79 A surveillance study showed IGF-1 control in 63% of 1288 patients.80 Side effects include elevated hepatic aminotrans- ferase levels, injection-site inflammation, and lipodystrophy. Growth hormone axis blockade with growth hormone receptor–directed pegviso- mant combined with pituitary-directed somato­ statin receptor ligand offers greater efficacy than either drug alone.81 Treatment approaches have advantages and disadvantages for individualizing patient care and maximizing growth hormone and IGF-1 control (Table 1).12 Patients should be monitored for symptoms affecting the quality of life, in- cluding anxiety. Management of coexisting con- ditions, especially high blood pressure, cardiac dysfunction, and sleep apnea, as well as elevated blood sugar levels, is important to reduce the risk of death. Cushing’s Disease Corticotropin-secreting corticotroph adenomas, which account for up to 15% of pituitary tu- mors, with an incidence of 1.6 cases per 1 mil- lion persons,82 are typically small (approximately 6 mm in diameter) and are 5 to 10 times as common in women as in men. Excessive secre- tion of corticotropin leads to adrenal hypercor- tisolemia. Corticotroph adenomas account for approximately 70% of cases of Cushing’s syn- drome, with iatrogenic hypercortisolism, ectopic corticotropin or corticotropin-releasing hormone production, and cortisol-producing adrenal le- sions accounting for the rest.83 Clinical Features Typical cushingoid features include plethoric moon facies with thin skin, purple striae, and easy bruising. Central obesity, hypertension, glu- cose intolerance or diabetes, and, especially in younger women, menstrual disturbances and osteoporosis are observed, as are proximal mus- cle wasting and weakness, acne, hirsutism, depres- sion, psychoses, and susceptibility to infection.84 The disorder may be indolent or clinically florid, with a high mortality rate if inadequately controlled. In a study involving 502 unselected patients, the overall standardized mortality ratio was 2.5 (95% CI, 2.1 to 2.9), with 133 observed deaths versus 54 expected deaths; cardiovascular disease accounted for most of the excess deaths.85 Mortality was also elevated among the patients with biochemical remission (standardized mor- tality ratio, 1.9 [95% CI, 1.5 to 2.3]), with 89 The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 11. n engl j med 382;10  nejm.org  March 5, 2020 947 Pituitary-Tumor Endocrinopathies observed deaths versus 47 expected deaths. Other causes of death include infections and suicide. Rapid-onset hypercortisolism with skin hyperpigmentation and severe myopathy sug- gests an ectopic tumor source of corticotropin, often associated with hypertension and hypo- kalemic alkalosis.83 Diagnosis Accurate diagnosis of Cushing’s disease can be challenging.83 Approximately 40% of corticotro- pin-secreting corticotroph tumors are not visible on imaging, and at least 10% of persons in the general population have small, clinically silent microadenomas. The disorder may therefore be overdiagnosed, especially since clinical features of hypercortisolemia overlap with other, much more common disorders, including obesity, hyper- tension, glucose intolerance, and osteoporosis. The diagnosis of pathologic hypercortisolemia is established on the basis of clinical features of hypercortisolism coupled with reproducible evi- dence of failure to suppress plasma cortisol at 8 a.m. to a level below 1.8 μg per deciliter after the administration of 1 mg of dexamethasone at 11 p.m. or elevated 24-hour urinary free cortisol levels and midnight salivary cortisol values.84 Basal corticotropin levels are usually inappropri- ately high, and suppressibility of corticotropin by glucocorticoids may distinguish a pituitary tumor from an ectopic source. Since the results of repeated urinary free cor- tisol and midnight salivary cortisol measure- ments may vary, bilateral inferior petrosal sinus sampling of corticotropin levels may be required to confirm the diagnosis. A central-to-peripheral corticotropin gradient that exceeds 2 before and after the administration of corticotropin-stimu- lating hormone definitively localizes a cortico- troph tumor source with more than 95% sensi- tivity. Treatment Selective transsphenoidal adenomectomy is rec- ommended as initial therapy for Cushing’s dis- ease, with remission achieved in approximately 75% of patients and recurrence seen in approxi- mately 10%.86 Although more radical surgery offers the potential for total adenoma resection, complication rates are higher and pituitary dam- age is more likely with total resection.87 Radia- tion therapy may control the disease,88 but the efficacy of this treatment is delayed for several years, and approximately 30% of patients have a relapse. Adrenalectomy may immediately reverse hy- percortisolism. However, lifelong replacement of adrenal hormones is challenging. Patients who undergo adrenalectomy are also at risk for adre- nal crisis and Nelson’s syndrome (pituitary en- largement and development of corticotropin- secreting pituitary adenomas, which often have an invasive growth pattern). Adrenal-targeted medical therapy may offer clinical and biochemical improvement, but most studies of such treatment have not been rigor- ously controlled, and the results are often incon- sistent.89 Adrenal steroidogenesis inhibitors block hypercortisolemia but do not target the pituitary tumor. Ketoconazole, an antifungal imidazole, normalizes urinary free cortisol levels in 50% of patients.90 Side effects include nausea, headache, low testosterone levels, reversible elevated liver enzyme levels, and in rare cases, hepatotoxicity. Metyrapone controls urinary free cortisol levels in about 50% of patients, and accumulated ste- roid precursors may cause acne, hirsutism, hyper- tension, and hypokalemia. Mitotane, an adreno- lytic agent, is used mainly for adrenal carcinoma. Mifepristone, a glucocorticoid receptor antago- nist, is approved for the treatment of hypergly- cemia associated with Cushing’s syndrome in patients in whom surgery has failed or who are not surgical candidates. Since mifepristone blocks the action of cortisol, corticotropin and urinary free cortisol levels are increased.91 Adrenal fail- ure, hypokalemia, and excessive vaginal bleeding may limit the use of this agent. Pituitary-targeted drugs include high-dose cabergoline (up to 1 mg daily), which controls hypercortisolism in up to 30% of patients, al- though therapeutic efficacy is often not main- tained in the long term. Pasireotide blocks ade- noma-derived corticotropin secretion, normalizes urinary free cortisol levels in approximately 40% of patients with mild disease, and improves clini- cal features. Hyperglycemia develops in most patients.92,93 Thyrotropin-Secreting Tumors Accounting for approximately 1% of adenomas, thyrotropin-secreting tumors lead to elevated or inappropriately suppressed thyrotropin levels The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 12. n engl j med 382;10  nejm.org  March 5, 2020948 The new engl and jour nal of medicine with normal or elevated thyroid hormone levels.94 Resection and adjuvant treatment are required for adequate biochemical outcomes. Dr. Melmed reports receiving grant support, advisory board fees, and fees for serving as a course mentor, paid to his insti- tution, from Pfizer, consulting fees from Ionis, Midatech, and Chiasma, grant support from Ono, advisory board fees from Strongbridge, and advisory fees and travel support from Ipsen. No other potential conflict of interest relevant to this article was reported. Disclosure forms provided by the author are available with the full text of this article at NEJM.org. References 1. Gittleman H, Ostrom QT, Farah PD, et al. Descriptive epidemiology of pitu- itary tumors in the United States, 2004- 2009. J Neurosurg 2014;​121:​527-35. 2. Casanueva FF, Barkan AL, Buchfelder M, et al. Criteria for the definition of Pitu- itary Tumor Centers of Excellence (PTCOE): a Pituitary Society statement. Pituitary 2017;​ 20:​489-98. 3. Melmed S. Pathogenesis of pituitary tumors. Nat Rev Endocrinol 2011;​7:​257-66. 4. Molitch ME. Diagnosis and treatment of pituitary adenomas: a review. JAMA 2017;​317:​516-24. 5. Syro LV, Rotondo F, Ramirez A, et al. Progress in the diagnosis and classifica- tion of pituitary adenomas. Front Endo- crinol (Lausanne) 2015;​6:​97. 6. Chesnokova V, Zonis S, Kovacs K, et al. p21(Cip1) Restrains pituitary tumor growth. Proc Natl Acad Sci U S A 2008;​105:​17498- 503. 7. Hage M, Viengchareun S, Brunet E, et al. Genomic alterations and complex sub- clonal architecture in sporadic GH-secret- ing pituitary adenomas. J Clin Endocrinol Metab 2018;​103:​1929-39. 8. Bi WL, Larsen AG, Dunn IF. Genomic alterations in sporadic pituitary tumors. Curr Neurol Neurosci Rep 2018;​18:​4. 9. Reincke M, Sbiera S, Hayakawa A, et al. Mutations in the deubiquitinase gene USP8 cause Cushing’s disease. Nat Genet 2015;​47:​31-8. 10. Agustsson TT, Baldvinsdottir T, Jonas­ son JG, et al. The epidemiology of pitu- itary adenomas in Iceland, 1955-2012: a nationwide population-based study. Eur J Endocrinol 2015;​173:​655-64. 11. Lopes MBS. The 2017 World Health Organization classification of tumors of the pituitary gland: a summary. Acta Neuro- pathol 2017;​134:​521-35. 12. Cuevas-Ramos D, Carmichael JD, Cooper O, et al. A structural and func- tional acromegaly classification. J Clin Endocrinol Metab 2015;​100:​122-31. 13. Freda PU, Beckers AM, Katznelson L, et al. Pituitary incidentaloma: an Endo- crine Society clinical practice guideline. J Clin Endocrinol Metab 2011;​96:​894-904. 14. Fleseriu M, Hashim IA, Karavitaki N, et al. Hormonal replacement in hypopitu- itarism in adults: an Endocrine Society clinical practice guideline. J Clin Endocri- nol Metab 2016;​101:​3888-921. 15. Raverot G, Burman P, McCormack A, et al. European Society of Endocrinology clinical practice guidelines for the man- agement of aggressive pituitary tumours and carcinomas. Eur J Endocrinol 2018;​ 178:​G1-G24. 16. Del Basso De Caro M, Solari D, Pagliuca F, et al. Atypical pituitary adenomas: clin- ical characteristics and role of ki-67 and p53 in prognostic and therapeutic evalua- tion: a series of 50 patients. Neurosurg Rev 2017;​40:​105-14. 17. Fountas A, Lavrentaki A, Subramanian A, Toulis KA, Nirantharakumar K, Kara- vitaki N. Recurrence in silent cortico- troph adenomas after primary treatment: a systematic review and meta-analysis. J Clin Endocrinol Metab 2018 December 21 (Epub ahead of print). 18. Lucas JW, Bodach ME, Tumialan LM, et al. Congress of Neurological Surgeons systematic review and evidence-based guideline on primary management of patients with nonfunctioning pituitary adenomas. Neurosurgery 2016;​79(4):​E533- E535. 19. Lee CC, Vance ML, Xu Z, et al. Stereo- tactic radiosurgery for acromegaly. J Clin Endocrinol Metab 2014;​99:​1273-81. 20. Ayuk J, Clayton RN, Holder G, Shep- pard MC, Stewart PM, Bates AS. Growth hormone and pituitary radiotherapy, but not serum insulin-like growth factor-I concentrations, predict excess mortality in patients with acromegaly. J Clin Endo- crinol Metab 2004;​89:​1613-7. 21. Vierimaa O, Georgitsi M, Lehtonen R, et al. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science 2006;​312:​1228-30. 22. Correa R, Salpea P, Stratakis CA. Car- ney complex: an update. Eur J Endocrinol 2015;​173:​M85-M97. 23. Famini P, Maya MM, Melmed S. Pitu- itary magnetic resonance imaging for sel- lar and parasellar masses: ten-year expe- rience in 2598 patients. J Clin Endocrinol Metab 2011;​96:​1633-41. 24. Drummond J, Roncaroli F, Grossman AB, Korbonits M. Clinical and pathologi- cal aspects of silent pituitary adenomas. J Clin Endocrinol Metab 2019;​104:​2473-89. 25. Pappy AL II, Savinkina A, Bicknese C, Neill S, Oyesiku NM, Ioachimescu AG. Predictive modeling for pituitary adeno- mas: single center experience in 501 con- secutive patients. Pituitary 2019;​22:​520-31. 26. Chen L, White WL, Spetzler RF, Xu B. A prospective study of nonfunctioning pituitary adenomas: presentation, manage- ment, and clinical outcome. J Neurooncol 2011;​102:​129-38. 27. Paluzzi A, Fernandez-Miranda JC, Tonya Stefko S, Challinor S, Snyderman CH, Gardner PA. Endoscopic endonasal approach for pituitary adenomas: a series of 555 patients. Pituitary 2014;​17:​307-19. 28. Sheehan JP, Starke RM, Mathieu D, et al. Gamma Knife radiosurgery for the management of nonfunctioning pituitary adenomas: a multicenter study. J Neuro- surg 2013;​119:​446-56. 29. Tampourlou M, Ntali G, Ahmed S, et al. Outcome of nonfunctioning pituitary ad- enomas that regrow after primary treat- ment: a study from two large UK centers. J Clin Endocrinol Metab 2017;​102:​1889-97. 30. Huang W, Molitch ME. Management of nonfunctioning pituitary adenomas (NFAs): observation. Pituitary 2018;​21:​ 162-7. 31. Olsson DS, Nilsson AG, Bryngelsson IL, Trimpou P, Johannsson G, Andersson E. Excess mortality in women and young adults with nonfunctioning pituitary ­adenoma: a Swedish nationwide study. J Clin Endocrinol Metab 2015;​100:​2651-8. 32. Fernandez A, Karavitaki N, Wass JA. Prevalence of pituitary adenomas: a com- munity-based, cross-sectional study in Ban- bury (Oxfordshire, UK). Clin Endocrinol (Oxf) 2010;​72:​377-82. 33. Melmed S, Casanueva FF, Hoffman AR, et al. Diagnosis and treatment of ­hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocri- nol Metab 2011;​96:​273-88. 34. Burke WT, Penn DL, Castlen JP, et al. Prolactinomas and nonfunctioning ade- nomas: preoperative diagnosis of tumor type using serum prolactin and tumor size. J Neurosurg 2019 June 14 (Epub ahead of print). 35. Delgrange E, Trouillas J, Maiter D, Donckier J, Tourniaire J. Sex-related dif- ference in the growth of prolactinomas: a clinical and proliferation marker study. J Clin Endocrinol Metab 1997;​82:​2102-7. 36. Bernard V, Young J, Chanson P, Binart N. New insights in prolactin: pathological implications. Nat Rev Endocrinol 2015;​ 11:​265-75. 37. Webster J, Piscitelli G, Polli A, Ferrari CI, Ismail I, Scanlon MF. A comparison of cabergoline and bromocriptine in the The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 13. n engl j med 382;10  nejm.org  March 5, 2020 949 Pituitary-Tumor Endocrinopathies treatment of hyperprolactinemic amenor- rhea. N Engl J Med 1994;​331:​904-9. 38. Biller BM, Molitch ME, Vance ML, et al. Treatment of prolactin-secreting macro­ adenomas with the once-weekly dopa- mine agonist cabergoline. J Clin Endocri- nol Metab 1996;​81:​2338-43. 39. Colao A, Di Sarno A, Cappabianca P, Di Somma C, Pivonello R, Lombardi G. Withdrawal of long-term cabergoline ther- apy for tumoral and nontumoral hyper­ prolactinemia. N Engl J Med 2003;​349:​ 2023-33. 40. Ioachimescu AG, Fleseriu M, Hoff- man AR, Vaughan Iii TB, Katznelson L. Psychological effects of dopamine ago- nist treatment in patients with hyperpro- lactinemia and prolactin-secreting adeno- mas. Eur J Endocrinol 2019;​180:​31-40. 41. Stiles CE, Tetteh-Wayoe ET, Bestwick J, Steeds RP, Drake WM. A meta-analysis of the prevalence of cardiac valvulopathy in hyperprolactinemic patients treated with cabergoline. J Clin Endocrinol Metab 2018;​104:​523-38. 42. Tampourlou M, Trifanescu R, Paluzzi A, Ahmed SK, Karavitaki N. Therapy of endocrine disease: surgery in micropro- lactinomas: effectiveness and risks based on contemporary literature. Eur J Endo- crinol 2016;​175:​R89-R96. 43. Liu W, Zahr RS, McCartney S, Cetas JS, Dogan A, Fleseriu M. Clinical out- comes in male patients with lactotroph adenomas who required pituitary surgery: a retrospective single center study. Pitu- itary 2018;​21:​454-62. 44. Bronstein MD. Prolactinomas and pregnancy. Pituitary 2005;​8:​31-8. 45. Burton T, Le Nestour E, Neary M, Ludlam WH. Incidence and prevalence of acromegaly in a large US health plan database. Pituitary 2016;​19:​262-7. 46. Thorner MO, Perryman RL, Cronin MJ, et al. Somatotroph hyperplasia: suc- cessful treatment of acromegaly by re- moval of a pancreatic islet tumor secret- ing a growth hormone-releasing factor. J Clin Invest 1982;​70:​965-77. 47. Melmed S, Ezrin C, Kovacs K, Good- man RS, Frohman LA. Acromegaly due to secretion of growth hormone by an ectop­ ic pancreatic islet-cell tumor. N Engl J Med 1985;​312:​9-17. 48. Colao A, Grasso LFS, Giustina A, et al. Acromegaly. Nat Rev Dis Primers 2019;​5:​ 20. 49. Reid TJ, Post KD, Bruce JN, Nabi Kanibir M, Reyes-Vidal CM, Freda PU. Features at diagnosis of 324 patients with acromegaly did not change from 1981 to 2006: acromegaly remains under-recog- nized and under-diagnosed. Clin Endo- crinol (Oxf) 2010;​72:​203-8. 50. Casini AF, Neto LV, Fontes R, França RF, Xavier SS, Gadelha MR. Aortic root ectasia in patients with acromegaly: expe- rience at a single center. Clin Endocrinol (Oxf) 2011;​75:​495-500. 51. Annamalai AK, Webb A, Kandasamy N, et al. A comprehensive study of clini- cal, biochemical, radiological, vascular, cardiac, and sleep parameters in an un- selected cohort of patients with acromeg- aly undergoing presurgical somatostatin receptor ligand therapy. J Clin Endocrinol Metab 2013;​98:​1040-50. 52. Alexopoulou O, Bex M, Kamenicky P, Mvoula AB, Chanson P, Maiter D. Preva- lence and risk factors of impaired glucose tolerance and diabetes mellitus at diagno- sis of acromegaly: a study in 148 patients. Pituitary 2014;​17:​81-9. 53. Dal J, Feldt-Rasmussen U, Andersen M, et al. Acromegaly incidence, preva- lence, complications and long-term prog- nosis: a nationwide cohort study. Eur J Endocrinol 2016;​175:​181-90. 54. Wassenaar MJ, Cazemier M, Biermasz NR, et al. Acromegaly is associated with an increased prevalence of colonic diver- ticula: a case-control study. J Clin Endo- crinol Metab 2010;​95:​2073-9. 55. Resmini E, Tagliafico A, Bacigalupo L, et al. Computed tomography colonog- raphy in acromegaly. J Clin Endocrinol Metab 2009;​94:​218-22. 56. Gonzalez B, Vargas G, Mendoza V, Nava M, Rojas M, Mercado M. The preva- lence of colonic polyps in patients with acromegaly: a case-control, nested in a cohort colonoscopic study. Endocr Pract 2017;​23:​594-9. 57. Terzolo M, Reimondo G, Berchialla P, et al. Acromegaly is associated with in- creased cancer risk: a survey in Italy. En- docr Relat Cancer 2017;​24:​495-504. 58. Dal J, Leisner MZ, Hermansen K, et al. Cancer incidence in patients with acro- megaly: a cohort study and meta-analysis of the literature. J Clin Endocrinol Metab 2018;​103:​2182-8. 59. Melmed S, Bronstein MD, Chanson P, et al. A consensus statement on acromeg- aly therapeutic outcomes. Nat Rev Endo- crinol 2018;​14:​552-61. 60. Ritvonen E, Löyttyniemi E, Jaatinen P, et al. Mortality in acromegaly: a 20-year follow-up study. Endocr Relat Cancer 2016;​23:​469-80. 61. Sherlock M, Reulen RC, Aragon-Alonso A, et al. A paradigm shift in the monitor- ing of patients with acromegaly: last available growth hormone may overesti- mate risk. J Clin Endocrinol Metab 2014;​ 99:​478-85. 62. Trivellin G, Daly AF, Faucz FR, et al. Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. N Engl J Med 2014;​371:​2363-74. 63. Goldenberg N, Racine MS, Thomas P, Degnan B, Chandler W, Barkan A. Treat- ment of pituitary gigantism with the growth hormone receptor antagonist pegvisomant. J Clin Endocrinol Metab 2008;​93:​2953-6. 64. Clemmons DR, Van Wyk JJ, Ridgway EC, Kliman B, Kjellberg RN, Underwood LE. Evaluation of acromegaly by radioim- munoassay of somatomedin-C. N Engl J Med 1979;​301:​1138-42. 65. Katznelson L, Laws ER Jr, Melmed S, et al. Acromegaly: an Endocrine Society clinical practice guideline. J Clin Endocri- nol Metab 2014;​99:​3933-51. 66. Faje AT, Barkan AL. Basal, but not pulsatile, growth hormone secretion de- termines the ambient circulating levels of insulin-like growth factor-I. J Clin Endo- crinol Metab 2010;​95:​2486-91. 67. Buchfelder M, Schlaffer SM. The sur- gical treatment of acromegaly. Pituitary 2017;​20:​76-83. 68. Kim JH, Hur KY, Lee JH, et al. Out- come of endoscopic transsphenoidal sur- gery for acromegaly. World Neurosurg 2017;​104:​272-8. 69. Babu H, Ortega A, Nuno M, et al. Long-term endocrine outcomes following endoscopic endonasal transsphenoidal surgery for acromegaly and associated prognostic factors. Neurosurgery 2017;​ 81:​357-66. 70. Ding D, Mehta GU, Patibandla MR, et al. Stereotactic radiosurgery for acro- megaly: an international multicenter retro- spective cohort study. Neurosurgery 2019;​ 84:​717-25. 71. Kuhn E, Chanson P. Cabergoline in acromegaly. Pituitary 2017;​20:​121-8. 72. Lamberts SW, van der Lely A-J, de Herder WW, Hofland LJ. Octreotide. N Engl J Med 1996;​334:​246-54. 73. Carmichael JD, Bonert VS, Nuño M, Ly D, Melmed S. Acromegaly clinical trial methodology impact on reported biochem- ical efficacy rates of somatostatin receptor ligand treatments: a meta-analysis. J Clin Endocrinol Metab 2014;​99:​1825-33. 74. Giustina A, Mazziotti G, Torri V, Spi- nello M, Floriani I, Melmed S. Meta-analy- sis on the effects of octreotide on tumor mass in acromegaly. PLoS One 2012;​7(5):​ e36411. 75. Giustina A, Mazziotti G, Cannavò S, et al. High-dose and high-frequency lan- reotide autogel in acromegaly: a random- ized, multicenter study. J Clin Endocrinol Metab 2017;​102:​2454-64. 76. Brzana J, Yedinak CG, Gultekin SH, Delashaw JB, Fleseriu M. Growth hor- mone granulation pattern and somatosta- tin receptor subtype 2A correlate with post- operative somatostatin receptor ligand response in acromegaly: a large single center experience. Pituitary 2013;​16:​490-8. 77. Schmid HA, Brue T, Colao A, et al. Effect of pasireotide on glucose- and growth hormone-related biomarkers in patients with inadequately controlled ac- romegaly. Endocrine 2016;​53:​210-9. The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.
  • 14. n engl j med 382;10  nejm.org  March 5, 2020950 Pituitary-Tumor Endocrinopathies 78. Kopchick JJ, Parkinson C, Stevens EC, Trainer PJ. Growth hormone receptor an- tagonists: discovery, development, and use in patients with acromegaly. Endocr Rev 2002;​23:​623-46. 79. Barkan AL, Burman P, Clemmons DR, et al. Glucose homeostasis and safety in patients with acromegaly converted from long-acting octreotide to pegvisomant. J Clin Endocrinol Metab 2005;​90:​5684-91. 80. van der Lely AJ, Biller BM, Brue T, et al. Long-term safety of pegvisomant in patients with acromegaly: comprehensive review of 1288 subjects in ACROSTUDY. J Clin Endocrinol Metab 2012;​97:​1589-97. 81. Neggers SJ, Franck SE, de Rooij FW, et al. Long-term efficacy and safety of pegvisomant in combination with long- acting somatostatin analogs in acromegaly. J Clin Endocrinol Metab 2014;​99:​3644- 52. 82. Ragnarsson O, Olsson DS, Chantzi- christos D, et al. The incidence of Cush- ing’s disease: a nationwide Swedish study. Pituitary 2019;​22:​179-86. 83. Loriaux DL. Diagnosis and differen- tial diagnosis of Cushing’s syndrome. N Engl J Med 2017;​376:​1451-9. 84. Nieman LK. Diagnosis of Cushing’s syndrome in the modern era. Endocrinol Metab Clin North Am 2018;​47:​259-73. 85. Ragnarsson O, Olsson DS, Papakok- kinou E, et al. Overall and disease-specif- ic mortality in patients with Cushing dis- ease: a Swedish nationwide study. J Clin Endocrinol Metab 2019;​104:​2375-84. 86. Nieman LK, Biller BM, Findling JW, et al. Treatment of Cushing’s syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2015;​ 100:​2807-31. 87. Post KD, Pain M, Zarzour H, Beder- son J. Transsphenoidal surgery for Cush- ing’s disease. In:​Laws ER Jr, Cohen-Gadol AA, Schwartz TH, Sheehan JP, eds. Trans- sphenoidal surgery:​complication avoid- ance and management techniques. Cham, Switzerland:​Springer International Pub- lishing, 2017:​427-39. 88. Mehta GU, Ding D, Patibandla MR, et al. Stereotactic radiosurgery for Cush- ing disease: results of an international, multicenter study. J Clin Endocrinol Metab 2017;​102:​4284-91. 89. Feelders RA, Newell-Price J, Pivonello R, Nieman LK, Hofland LJ, Lacroix A. Ad- vances in the medical treatment of Cush- ing’s syndrome. Lancet Diabetes Endocri- nol 2019;​7:​300-12. 90. Castinetti F, Guignat L, Giraud P, et al. Ketoconazole in Cushing’s disease: is it worth a try? J Clin Endocrinol Metab 2014;​99:​1623-30. 91. Fleseriu M, Biller BM, Findling JW, Molitch ME, Schteingart DE, Gross C. Mifepristone, a glucocorticoid receptor antagonist, produces clinical and meta- bolic benefits in patients with Cushing’s syndrome. J Clin Endocrinol Metab 2012;​ 97:​2039-49. 92. Colao A, Petersenn S, Newell-Price J, et al. A 12-month phase 3 study of pasireo­ tide in Cushing’s disease. N Engl J Med 2012;​366:​914-24. 93. Lacroix A, Gu F, Gallardo W, et al. Ef- ficacy and safety of once-monthly pasi­ reotide in Cushing’s disease: a 12 month clinical trial. Lancet Diabetes Endocrinol 2018;​6:​17-26. 94. Azzalin A, Appin CL, Schniederjan MJ, et al. Comprehensive evaluation of thyro- tropinomas: single-center 20-year experi- ence. Pituitary 2016;​19:​183-93. Copyright © 2020 Massachusetts Medical Society. images in clinical medicine The Journal welcomes consideration of new submissions for Images in Clinical Medicine. Instructions for authors and procedures for submissions can be found on the Journal’s website at NEJM.org. At the discretion of the editor, images that are accepted for publication may appear in the print version of the Journal, the electronic version, or both. The New England Journal of Medicine Downloaded from nejm.org at Linkoping University Library on March 4, 2020. For personal use only. No other uses without permission. Copyright © 2020 Massachusetts Medical Society. All rights reserved.