You are on page 1of 57

CHAPTER 15

The Adrenal Cortex


PAUL M. STEWART • JOHN D. C. NEWELL-PRICE

The Adrenal Cortex—Historical Milestones, 490 The control of adrenocortical function by a pituitary
Anatomy and Development, 490 factor was demonstrated in the 1920s, and this led to the
isolation of sheep adrenocorticotropic hormone (ACTH) by
Adrenal Steroids and Steroidogenesis, 492 Li, Evans, and Simpson in 1943.7 Such a concept was sup-
Corticosteroid Hormone Action, 499 ported through clinical studies, notably by Harvey Cushing
Classification and Pathophysiology of Cushing in 1932, who associated his original clinical observations
Syndrome, 510 of 1912 (a “polyglandular syndrome” caused by pituitary
basophilism) with adrenal hyperactivity.8 The neural con-
Glucocorticoid Deficiency, 524
trol of pituitary ACTH secretion by corticotropin-releasing
Congenital Adrenal Hyperplasia, 533 factor (later renamed corticotropin-releasing hormone, or
Adrenal Adenomas, Incidentalomas, and Carcinomas, 544 CRH) was defined by Harris and other workers in the 1940s,
but CRH was not characterized and synthesized until 1981
in the laboratory of Wylie Vale.9 Jerome Conn described
KEY POINTS primary aldosteronism in 1955,10 and the control of adre-
nal aldosterone secretion by angiotensin II was confirmed
• This chapter discusses mechanisms and regulation of shortly afterward. Advances in radioimmunoassay, and
adrenal steroid production, function of the hypothalamic- particularly molecular biology, have facilitated an expo-
pituitary-adrenal axis, and negative regulation. nential increase in the understanding of adrenal physiol-
• The chapter goes on to describe the transactivating and ogy and pathophysiology (Table 15-1).
transrepressive actions of glucocorticoids.
• Glucocorticoid excess and Cushing syndrome, adrenal
insufficiency and Addison disease, and inherited ANATOMY AND DEVELOPMENT
disorders of the adrenal gland are also discussed.
• Optimizing corticosteroid replacement therapies is The cells forming the adrenal cortex originate from the
addressed. intermediate mesoderm. These cells derive from the uro-
• The chapter concludes with discussion of adrenal genital ridge and have a common embryologic origin with
incidentalomas, adenomas, and carcinomas. the gonad and the kidney. Early differentiation of the
adrenogonadal primordium from the urogenital ridge
requires signaling cascades and transcription factors GLI3,
SALL1, FOXD2, WT1, PBX1, and WNT4, and the regulator
of telomerase activity, ACD (Fig. 15-1). The adrenogonadal
THE ADRENAL CORTEX—HISTORICAL primordium can be seen as the medial part of the urogeni-
MILESTONES tal ridge at 4 weeks. Separation of the adrenogonadal pri-
mordium and formation of the adrenal primordium seem
The anatomy of the adrenal glands was described almost to depend on the actions of transcription factors SF1 (ster-
450 years ago by Bartholomeo Eustacius,1 and the zonation oidogenic factor 1), DAX1, WNT4, and CITED2. The ad-
of the gland and its distinction from the medulla were renocortical primordium develops at approximately 8
elucidated shortly thereafter. However, a functional role for weeks of gestation and can be differentiated into two dis-
the adrenal glands was not accurately defined until the tinct layers, the inner fetal zone (FZ) and the outer defini-
pioneering work of Thomas Addison, who described the tive zone (DZ). At approximately 9 weeks, the adrenal
clinical and autopsy findings in 11 cases of Addison disease blastema encapsulates and the adrenal medulla develops
in his classic monograph in 1855.2 Just a year later, Brown- when neural crest cells migrate into the adrenal gland.11
Séquard demonstrated that the adrenal glands were “organs During the second trimester, the FZ enlarges, becomes
essential for life” by performing adrenalectomies in dogs, larger than the fetal kidney, and secretes abundant amounts
cats, and guinea pigs.3 In 1896, William Osler first admin- of dehydroepiandrosterone (DHEA) and dehydroepiandros-
istered adrenal extract to a patient with Addison disease, a terone sulfate (DHEAS). Concentrations of these hormones
feat that was repeated by others in animal and human abruptly decline postnatally, in parallel with the postnatal
studies over the next 40 years. Between 1937 and 1955, the involution of the FZ. The neocortex develops over the
adrenocorticosteroid hormones were isolated, and their subsequent years into the adult adrenal gland.
structures were defined and synthesized.4 Notable break- In fetal life and up to 12 months of age, two distinct
throughs included the discovery of cortisone and clinical zones are evident, an inner prominent FZ and an outer DZ
evaluation of its anti-inflammatory effect in patients with that differentiates into the adult adrenal gland. After birth,
rheumatoid arthritis5 and the isolation of aldosterone.6 the FZ regresses and the DZ, which contains an inner zona

490
CHAPTER 15 The Adrenal Cortex 491

TABLE 15-1
History of the Adrenal Cortex: Important Milestones
Year Event
1563 Eustachius describes the adrenals (published by Lancisi in 1714).
1849 Thomas Addison, while searching for the cause of pernicious anemia, stumbles on a bronzed appearance associated with the
adrenal glands—melasma suprarenale.
1855 Thomas Addison describes the clinical features and autopsy findings in 11 cases of diseases of the suprarenal capsules, at least 6
of which were tuberculous in origin.
1856 In adrenalectomy experiments, Brown-Séquard demonstrates that the adrenal glands are essential for life.
1896 William Osler prepares an oral glycerin extract derived from pig adrenals and demonstrates that it has clinical benefit in patients
with Addison disease.
1905 Bulloch and Sequeira describe patients with congenital adrenal hyperplasia.
1929 Liquid extracts of cortical tissue are used to keep adrenalectomized cats alive indefinitely (Swingle and Pfiffner); subsequently, this
extract was used successfully to treat a patient with Addison disease (Rowntree and Greene).
1932 Harvey Cushing associates the polyglandular syndrome of pituitary basophilism, which he first described in 1912, with hyperactivity
of the pituitary-adrenal glands.
1936 The concept of stress and its effect on pituitary-adrenal function are described by Selye.
1937-1952 Isolation and structural characterization of adrenocortical hormones are reported by Kendall and Reichstein.
1943 Li and colleagues isolate pure adrenocorticotropic hormone from sheep pituitary.
1950 Hench, Kendall, and Reichstein share the Nobel Prize in Medicine for describing the anti-inflammatory effects of cortisone in
patients with rheumatoid arthritis.
1953 Isolation and analysis of the structure of aldosterone are reported by Simpson and Tait.
1956 Conn describes primary aldosteronism.
1981 Characterization and synthesis of corticotropin-releasing hormone are reported by Vale.
1980-present The molecular era: cloning and functional characterization of steroid receptors, steroidogenic enzymes, and adrenal transcription
factors are reported, and the molecular basis for human adrenal diseases is defined.

Prenatal life Birth Postnatal life


Urogenital Adreno- Adrenal Fetal adrenal Adrenal Adrenal Adrenal
ridge gonadal primordium from after
primordium 2-3 yrs adrenache

Medulla ZG ZG ZG
DZ DZ
ZF ZF ZF
FZ FZ ZR ZR
Islets

4 Weeks 8 Weeks 9 Weeks 24-28 Weeks 6 Months 2-3 Years 6-8 Years

Hedgehog SF1, DAX1, NGFIB, POMC-peptides, POMC-peptides,


signaling, CITED2, growth factors, midkine, neural feedback
GLI3, SALL1, WNT4 SPARC, neural feedback
FOXD2, WT1,
PBX1, WNT4, ACD

Figure 15-1 Schematic diagram of the development of the human adrenal cortex during prenatal and postnatal life showing transcription factors that are active at each stage
(see text for details). DZ, definitive zone; FZ, fetal zone; POMC, pro-opiomelanocortin; SPARC, secreted protein, acidic, cysteine-rich (osteonectin); ZF, zona fasciculata; ZG, zona
glomerulosa; ZR, zona reticularis.

fasciculata (ZF) and an outer zona glomerulosa (ZG), pro- variable rate. Mineralocorticoids and glucocorticoids show
liferates.12,13 The innermost zone, the zona reticularis (ZR), a less age-specific variation.
is evident after 2 years of life. The differentiation of the The adult adrenal gland is a pyramidal structure, approx-
adrenal cortex into distinct zones has important functional imately 4 g in weight, 2 cm wide, 5 cm long, and 1 cm
consequences and is thought to depend on the temporal thick, that lies immediately above the kidney on its pos-
expression of transcription factors including Pref-1/ZOG, teromedial surface. Beneath the capsule, the ZG makes up
inner zone antigen, and SF1.14,15 In preadrenarchal children approximately 15% of the cortex (depending on sodium
focal reticular zone islets can be found, but the ZG and ZF intake) (Fig. 15-2). Cells are clustered in spherical nests and
are clearly differentiated.16 The occurrence of these ZR islets are small, with smaller nuclei in comparison with cells in
is consistent with the observation that DHEA and DHEAS other zones. The ZF makes up 75% of the cortex; cells are
concentrations gradually begin to rise from about 3 years large and lipid-laden and form radial cords within the
of age.17 At adrenarche, the inner zone (ZR) thickens, fibrovascular radial network. The innermost ZR is sharply
corresponding with increased production of DHEA and demarcated from both the ZF and the adrenal medulla.
DHEAS. Concurrently, changes in zone-specific enzyme Cells there are irregular with little lipid content. The main-
expression patterns, such as decreased 3β-hydroxysteroid tenance of normal adrenal size appears to involve a pro-
dehydrogenase type 2 (HSD3B2) and increased cytochrome genitor cell population lying between the ZG and ZF; cell
b5 and sulfotransferase (SULT2A1) in the ZR, lead to in- migration and differentiation occur within the fasciculata
creased flux toward DHEA. Clinically, adrenarche becomes and senescence occurs within the ZR, but the factors regu-
apparent at 6 to 8 years of age. Adrenal androgen produc- lating this important aspect of adrenal regeneration are
tion peaks in the third decade and then declines at a unknown. ACTH administration results in glomerulosa
492 SECTION IV Adrenal Cortex and Endocrine Hypertension

Capsule TABLE 15-2


IUPAC and Trivial Names of Natural and Synthetic Steroids
Trivial Name IUPAC Name
Aldosterone 4-Pregnen-11β,21-diol-3,18,20-trione
Zona glomerulosa Androstenedione 4-Androsten-3,17-dione
Cortisol 4-Pregnen-11β,17α,21-triol-3,20-dione
Cortisone 4-Pregnen-17α,21-diol-3,11,20-trione
Dehydroepiandrosterone 5-Androsten-3β-ol-17-one
Deoxycorticosterone 4-Pregnen-21-ol-3,20-dione
Dexamethasone 1,4-Pregnadien-9α-fluoro-16α-methyl-
11β,17α,21-triol-3,20-dione
Dihydrotestosterone 5α-Androstan-17β-ol-3-one
Estradiol 1,3,5(10)-Estratrien-3,17β-diol
Fludrocortisone 4-Pregnen-9α-fluoro-11β,17α,21-triol-3,20-
dione
17-Hydroxyprogesterone 4-Pregnen-17α-ol-3,20-dione
Zona fasciculata Methylprednisolone 1,4-Pregnadien-6α-methyl-11β,17α,21-triol-
3,20-dione
Prednisolone 1,4-Pregnadien-11β,17α,21-triol-3,20-dione
Cortex
Prednisone 1,4-Pregnadien-17α,21-diol-3,11,20-trione
Pregnenolone 5-Pregnen-3β-ol-20-one
Progesterone 4-Pregnen-3,20-dione
Testosterone 4-Androsten-17β-ol-3-one
Triamcinolone 1,4-Pregnadien-9α-fluoro-11β,16α,17α,21-
tetrol-3,20-dione
IUPAC, International Union of Pure and Applied Chemistry.

ocorticoids (aldosterone, deoxycorticosterone [DOC]), and


sex steroids (mainly androgens). All steroid hormones are
Zona reticularis derived from the cyclopentanoperhydrophenanthrene
structure, that is, three cyclohexane rings and a single
cyclopentane ring (Fig. 15-3). Steroid nomenclature is
defined in two ways: by trivial names (e.g., cortisol, aldos-
terone) or by the chemical structure as defined by the
International Union of Pure and Applied Chemistry
(IUPAC).18 The IUPAC classification is inappropriate for
clinical use but does provide an invaluable insight into
steroid structure. The basic structure, trivial name, and
Medulla IUPAC name of some common steroids are given in Figure
15-3 and Table 15-2. Estrogens have 18 carbon atoms (C18
steroids) and androgens have 19 carbon atoms (C19),
whereas glucocorticoids and progestogens are C21-steroid
derivatives.
Figure 15-2 Schematic diagram of the structure of the human adrenal cortex, Cholesterol is the precursor for adrenal steroidogenesis.
depicting the outer zona glomerulosa and inner zona fasciculata and zona It is provided principally from the circulation, in the form
reticularis. of low-density lipoprotein (LDL) cholesterol.19 Uptake is by
specific cell-surface LDL receptors present on adrenal
cells adopting a fasciculata phenotype, and in turn, the tissue20; LDL is then internalized via receptor-mediated
innermost fasciculata cells adopt a reticularis phenotype endocytosis,21 the resulting vesicles fuse with lysozymes,
that is reversible on withdrawal of ACTH. and free cholesterol is produced after hydrolysis. However,
The vasculature of the adrenal cortex is complex. Arte- it is clear that this cannot be the sole source of adrenal
rial supply is conveyed by up to 12 small arteries from the cholesterol, because patients with abetalipoproteinemia
aorta and the inferior phrenic, renal, and intercostal arter- who have undetectable circulating LDL and patients with
ies. These arteries branch to form a subcapsular arteriolar defective LDL receptors in the setting of familial hypercho-
plexus from which radial capillaries penetrate deeper into lesterolemia still have normal basal adrenal steroidogene-
the cortex. In the ZR, a dense sinusoidal plexus is created, sis. Cholesterol can be generated de novo within the
which empties into a central vein. The right adrenal vein adrenal cortex from acetyl coenzyme A (CoA). In addition,
is short, draining directly into the inferior vena cava, there is evidence that the adrenal gland can utilize high-
whereas the longer left adrenal vein usually drains into the density lipoprotein (HDL) cholesterol after uptake through
left renal vein. the putative HDL receptor, SR-B1.22
The biochemical pathways involved in adrenal steroido-
genesis are shown in Figure 15-4. The initial hormone-
dependent, rate-limiting step is the transport of intracellular
ADRENAL STEROIDS AND cholesterol from the outer to inner mitochondrial mem-
STEROIDOGENESIS brane for conversion to pregnenolone by cytochrome P450
side-chain cleavage enzyme (P450scc). Naturally occurring
Three main types of hormones are produced by the adrenal human mutations have confirmed the importance of a
cortex—glucocorticoids (cortisol, corticosterone), mineral- 30-kDa protein, steroidogenic acute regulatory protein
CHAPTER 15 The Adrenal Cortex 493

21
20
18

12 17
11 13 16
D
19 C 15
14
1 9
2 10 8
A B
3 5 7
4 6

CH2OH CH2OH CH3 CH3

O C O C O C O C O
OH

HO HO O O O
Dehydroepiandrosterone Pregnenolone Deoxycorticosterone 17-OH-Progesterone Progesterone
5-Androsten-3 β -ol-17-one 5-Pregnen-3 β -ol-20-one 4-Pregnen-21-ol-3,20-dione 4-Pregnen-17α -ol-3,20, dione 4-Pregnen-3,20-dione

CH2OH CH2OH CH2OH

C O C O O C O
O
CH
O OH HO OH

O O O O
Androstenedione Cortisone Cortisol Aldosterone
4-Androsten-3,17-dione 4-Pregnen-17a,21-diol-3,11,20-trione 4-Pregnen-11b,17a,21-triol-3,20-dione 4-Pregnen-11b,21-diol-3,18,20-trione
Figure 15-3 The cyclopentanoperhydrophenanthrene structure of corticosteroid hormones, highlighting the structure of some endogenous steroid hormones together with
their nomenclature.

TABLE 15-3
Nomenclature for Adrenal Steroidogenic Enzymes and Their Genes
Enzyme Name Enzyme Family Gene Chromosome
P450 Cholesterol side-chain cleavage (SCC) (desmolase) Cytochrome P450 type I CYP11A1 15q23-q24
3β-Hydroxysteroid dehydrogenase (3β-HSD) Short-chain alcohol dehydrogenase reductase HSD3B2 1p13.1
(type II isozyme) superfamily
17α-Hydroxylase/17,20-lyase Cytochrome P450 type II CYP17A1 10q24.3
21-Hydroxylase Cytochrome P450 type II CYP21A2 6p21.3
11β-Hydroxylase Cytochrome P450 type I CYP11B1 8q24.3
Aldosterone synthase Cytochrome P450 type I CYP11B2 8q24.3

(StAR), in mediating this effect. StAR is induced by an cytochrome P450 enzymes such as CYP11A1 (P450scc),
increase in intracellular cyclic adenosine monophosphate 11β-hydroxylase (CYP11B1, or P450c11b1), and aldoster-
(cAMP) after binding of ACTH to its cognate receptor, pro- one synthase (CYP11B2, or P450aldo) rely on electron
viding the first important rate-limiting step in adrenal ste- transfer facilitated by adrenodoxin and adrenodoxin
roidogenesis.23 Other transporters, including the peripheral reductase.25,26 Micrososomal (type II) cytochrome P450
benzodiazepine-like receptor, may be involved.24 enzymes localized to the endoplasmic reticulum include
Steroidogenesis involves the concerted action of several the steroidogenic enzymes 17α-hydroxylase (CYP17A1, or
enzymes, including a series of cytochrome P450 enzymes, P450c17), 21-hydroxylase (CYP21A2, or P450c21), and
all of which have been cloned and characterized (Table P450 aromatase (CYP19A1, or P450aro). The functions of
15-3). Cytochrome P450 enzymes are classified into two cytochrome P450 type II enzymes crucially depend on
types according to their subcellular localization and their P450 oxidoreductase (POR), which provides electrons
specific electron shuttle system. Mitochondrial (type I) required for monooxygenase reaction catalyzed by the
494 SECTION IV Adrenal Cortex and Endocrine Hypertension

Cholesterol
Outer
StAR Mitochondrial membrane
Inner
Cholesterol

ADR/Adx
CYP11A1
POR POR b5 PAPSS2
Pregnenolone 17-OH-Pregnenolone Dehydroepi- DHEA-S
CYP17A1 CYP17A1 SULT2A1
androsterone

HSD3B2 Pregnenediol HSD3B2 Pregnenetriol HSD3B2 DHEA, 16αOH-DHEA


Androsterone, Etiocholanolone
POR POR b5
Progesterone CYP17A1 17-OH-Progesterone CYP17A1 Androstenedione

POR Pregnanediol POR Pregnanetriol Androsterone


CYP21A2 CYP21A2 17-OH-Pregnenolone HSD17B Etiocholanolone

11-Deoxycorticosterone
* 11-Deoxycortisol Testosterone

ADR/Adx THDOC ADR/Adx THS Androsterone


Etiocholanolone
CYP11B1 CYP11B2 CYP11B1 21-Deoxycortisol

Pregnanetriolone
Corticosterone Cortisol

ADR/Adx THA, THB, H6PDH THF, 5αTHF


5αTHA, 5αTHB
CYP11B2 HSD11B1 HSD11B2

18-OH-Corticosterone Cortisone

ADR/Adx 18OH-THA THE


CYP11B2

Aldosterone

THALDO
Mineralocorticoid Glucocorticoid Androgens
Figure 15-4 Adrenal steroidogenesis. After the steroidogenic acute regulatory (StAR) protein-mediated uptake of cholesterol into mitochondria within adrenocortical cells,
aldosterone, cortisol, and adrenal androgens are synthesized through the coordinated action of a series of steroidogenic enzymes in a zone-specific fashion. The mitochondrial
cytochrome P450 (CYP) type I enzymes (CYP11A1, CYP11B1, CYP11B2) requiring electron transfer via adrenodoxin reductase (ADR) and adrenodoxin (Adx) are marked with a box
labeled ADR/Adx. The microsomal CYP type II enzymes (CYP17A1, CYP21A2) receive electrons from P450 oxidoreductase (circle labeled POR). The 17,20-lyase reaction catalyzed
by CYP17A1 requires, in addition to POR, cytochrome b5, indicated by a circle labeled b5. Urinary steroid hormone metabolites are given in italics below the plasma hormones.
The asterisk (*) indicates 11-hydroxylation of 17-OH-progesterone to 21-deoxycortisol in cases of 21-hydroxylase deficiency. The adrenal conversion of androstenedione to
testosterone is catalyzed by the aldo-keto reductase AKR1C3 (HSD17B5). CYP11A1, P450 side-chain cleavage enzyme; CYP11B1, 11β-hydroxylase; CYP11B2, aldosterone syn-
thase; CYP17A1, 17α-hydroxylase; CYP21A2, 21-hydroxylase; DHEA, dehydroepiandrosterone; DHEA-S, dehydroepiandrosterone sulfate; H6PDH, hexose-6-phosphate dehydro-
genase; HSD11B1, 11β-hydroxysteroid dehydrogenase 1; HSD11B2, 11β-hydroxysteroid dehydrogenase 2; HSD17B, 17β-hydroxysteroid dehydrogenase; HSD3B2, 3β-hydroxysteroid
dehydrogenase type 2; 17-OH-progesterone, 17α-hydroxyprogesterone; PAPSS2, 3′-phosphoadenosine, 5′-phosphosulfate synthase 2; SULT2A1, sulfotransferase 2A1;
THA, tetrahydro-11-dehydrocorticosterone; THB, tetrahydro-corticosterone; THALDO, tetrahydro-aldosterone; THDOC, tetrahydro-11-deoxycorticosterone; THF, tetrahydro-cortisol;
THS, tetrahydro-11-deoxycortisol.

P450 enzyme.26,27 This category also includes hepatic P450 Mutations in the genes encoding these enzymes result
enzymes involved in drug metabolism and enzymes in human disease, so some understanding of the underly-
involved in sterol and bile acid synthesis.26,27 In addition, ing pathways and steroid precursors is required.29 After
the 17,20-lyase activity of P450 CYP17A1 is dependent on uptake of cholesterol to the mitochondrion, cholesterol is
a flavoprotein cytochrome b5, which functions as an allo- cleaved by the P450scc enzyme to form pregnenolone.30
steric facilitator of CYP17A1, and POR interaction (Fig. In the cytoplasm, pregnenolone is converted to progester-
15-5; see also Fig. 15-4).28 one by the type II isozyme 3β-HSD through a reaction
CHAPTER 15 The Adrenal Cortex 495

HOCH2- Therefore, CYP11B2 can carry out 11β-hydroxylation, 18-


Steroid hydroxylation, and 18-methyloxidation to yield the
characteristic C11-18 hemiacetyl structure of aldosterone.
NADPH Fp Fe2+ Fe3+ CH3-Steroid

Regulation of Adrenal Steroidogenesis:


CYP.O
Functional Zonation of the Adrenal Cortex
. Glucocorticoids are secreted in relatively high amounts
NADP+ Fp Fe3+ Fe2+ H2O
(cortisol, 10 to 20 mg/day) from the ZF under the control
Andrenodoxin Adrenodoxin CYP11A1,
of ACTH; mineralocorticoids are secreted in low amounts
A reductase CYP11B1, or O2
CYP11B2 (aldosterone, 100 to 150 μg/day) from the ZG under the
principal control of angiotensin II. As a class, adrenal
HOCH2-Steroid
androgens (DHEA, DHEAS, androstenedione) are the most
abundant steroids secreted from the adult adrenal gland
NADPH Fp Fe2+ CH3-Steroid (>20 mg/day). In each case, secretion is facilitated through
the expression of steroidogenic enzymes in a specific
CYP.O zonal manner. The ZG cannot synthesize cortisol because
it does not express 17α-hydroxylase. In contrast, aldoster-
. one secretion is confined to the outer ZG because of
NADP+ Fp Fe3+ H2O the restricted expression of CYP11B2. Although CYP11B1
P450 oxidoreductase CYP17A1 and CYP11B2 share 95% homology, the 5′ promoter
or or O2 sequences differ, permitting regulation of the final steps
B cytochrome b5 CYP21A2 in glucocorticoid and mineralocorticoid biosynthesis by
Figure 15-5 A, Electron shuttle system for the mitochondrial enzymes CYP11A1, ACTH and angiotensin II, respectively. In the ZR, high
CYP11B1, and CYP11B2. Adrenodoxin reductase receives electrons from reduced levels of cytochrome b5 confer 17,20-lyase activity on
nicotinamide adenine dinucleotide phosphate (NADPH) and reduces adrenodoxin, CYP17A1 and androgen production. DHEA is sulfated in
which transfers reducing equivalents to the cytochrome P450 (CYP) enzyme. The the ZR by the DHEA SULT2A1 to form DHEAS. This sulfo-
enzyme then transfers electrons, by way of oxygen, to the steroid. B, Electron shuttle nation reaction facilitated by SULT2A1 relies on the donor
system for the microsomal enzymes CYP17A1 and CYP21A2. P450 oxidoreductase, a
3′-phosphoadenosine 5′-phosphosulfate (PAPS) to transfer
flavoprotein, accepts electrons from NADPH and transfers them to the NADPH-P450
enzyme. The enzyme then transfers electrons, by way of oxygen, to the steroid. A a sulfonate group to an acceptor molecule. PAPS is synthe-
second reducing equivalent may be supplied to CYP17A1 by NADPH-P450 oxidore- sized by PAPS synthase, of which two isoenzymes exist
ductase or cytochrome b5. Fp, flavoprotein; Fp•, reduced form of flavoprotein; NADP+, (PAPSS1 and PAPSS2).38
nicotinamide adenine dinucleotide phosphate. In the fetal adrenal, steroidogenesis occurs primarily
within the inner FZ. The FZ is a characteristic feature of
higher primates, but the biologic role of fetal androgen
production remains unclear. Because of a relative lack of
3β-HSD and high SULT2A1 activity, the principal steroido-
involving dehydrogenation of the 3-hydroxyl group and genic products are DHEA and DHEAS, which are then aro-
isomerization of the double bond at C5.31 Progesterone matized by placental trophoblast to estrogens. Therefore,
is hydroxylated to 17-hydroxyprogesterone (17-OHP) the majority of maternal estrogen across pregnancy is, indi-
through the 17α-hydroxylase activity of CYP17A1. 17α- rectly, fetally derived.39
Hydroxylation is an essential prerequisite for glucocorti- Classic endocrine feedback loops are in place to control
coid synthesis, and the ZG does not express 17α-hydroxylase. the secretion of both hormones. Cortisol inhibits the secre-
CYP17A1 also possesses 17,20-lyase activity, which results tion of CRH from the hypothalamus and ACTH from the
in production of the C19 adrenal androgens DHEA and pituitary, and aldosterone-induced sodium retention inhib-
androstenedione.32 In humans, however, 17-OHP is not an its renal renin secretion.
efficient substrate for CYP17A1, and there is negligible
conversion of 17-OHP to androstenedione. Adrenal andro- Glucocorticoid Secretion: The Hypothalamic-
stenedione secretion is dependent on the conversion of Pituitary-Adrenal Axis
DHEA to androstenedione by 3β-HSD. This enzyme also
converts 17-hydroxypregnenolone to 17-OHP, but the pre- Pro-opiomelanocortin and ACTH
ferred substrate is pregnenolone. The human adrenal gland ACTH is the principal hormone stimulating adrenal gluco-
is capable of synthesis of small but significant amounts corticoid biosynthesis and secretion. ACTH has 39 amino
of testosterone, which increases in clinical conditions asso- acids but is synthesized within the anterior pituitary as
ciated with androgen excess. This conversion is facilitated part of a much larger, 241–amino acid precursor called
by the enzyme 17β-HSD type 5 (HSD17B5), also called pro-opiomelanocortin (POMC). A transcription factor, TPIT,
aldoketoreductase 1C3 (AKR1C3).33 21-Hydroxylation of appears to be essential for differentiation of POMC-
either progesterone (in the ZG) or 17-OHP (in the ZF) is expressing cells within the anterior pituitary.40 POMC is
carried out by the product of the CYP21A2 gene, cleaved in a tissue-specific fashion by prohormone conver-
21-hydroxylase, to yield DOC or 11-deoxycortisol, respec- tases to yield smaller peptide hormones. In the anterior
tively.34 The final step in cortisol biosynthesis takes place pituitary, this results in the secretion of β-lipoprotein (β-
in the mitochondria and involves the conversion of LPH) and pro-ACTH, the latter being further cleaved to an
11-deoxycortisol to cortisol by the enzyme CYP11B1 amino-terminal peptide, joining peptide, and ACTH itself
(11β-hydroxylase).35 In the ZG, 11β-hydroxylase may also (Fig. 15-6).41,42 Postsecretion cleavage of the precursor to
convert DOC to corticosterone. The enzyme CYP11B2 γ-melanocyte-stimulating hormone (pro-γ-MSH) by a serine
(aldosterone synthase) may also carry out this reaction; protease (AsP) expressed in the outer adrenal cortex is
in addition, it is required for conversion of corticoste- thought to mediate the trophic action of ACTH on the
rone to aldosterone via the intermediate 18-OH corticoste- adrenal cortex.43 The first 24 amino acids of ACTH are
rone; CYP11B1 lacks these two enzymatic activities.36,37 common to all species, and a synthetic ACTH(1-24),
496 SECTION IV Adrenal Cortex and Endocrine Hypertension

Intron A Intron B
Exon 1 Exon 2 Exon 3

Gppp 5′ UT 3′ UT AAAAAAAAAA

Signal peptide Pre-pro-opiomelanocortin

Pro-opiomelanocortin (31K)

Pro-ACTH (22K) β-LPH

Pro-γ-MSH ACTH γ-LPH β-endorphin

N-POC (1-76) Joining peptide

N-POC (1-48) γ-MSH

Figure 15-6 Synthesis and cleavage of pro-opiomelanocortin (POMC) within the human anterior pituitary gland. Prohormone convertase enzymes sequentially cleave
POMC to adrenocorticotropic hormone (ACTH). Shaded areas represent melanocyte-stimulating hormone (MSH) structural units. β-LPH, β-lipoprotein; γ-LPH, γ-lipoprotein;
N-POC, amino-terminal pro-opiomelanocortin.

Synacthen, is available commercially for clinical testing of to be of crucial importance in appetite control and energy
the hypothalamic-pituitary-adrenal (HPA) axis and assess- homeostasis (see later discussion).55
ing adrenal glucocorticoid reserve. The hormones α-, β-,
and γ-MSH are also cleaved products from POMC, but the
increased pigmentation characteristic of Addison disease is
Corticotropin-Releasing Hormone and Arginine Vasopressin
thought to arise directly from increased ACTH concentra- POMC secretion is tightly controlled by numerous factors,
tions binding to the melanocortin-1 receptor (MC1R) notably CRH and arginine vasopressin (AVP) (Fig. 15-7).56,57
rather than from α-MSH secretion.44 Additional control is provided through an endogenous cir-
POMC is also transcribed in many extrapituitary tissues, cadian rhythm and by stress and feedback inhibition by
notably brain, liver, kidney, gonad, and placenta.41,45,46 In cortisol itself. CRH is a 41–amino acid peptide that is syn-
these normal tissues, POMC messenger RNA (mRNA) is thesized in neurons within the paraventricular nucleus of
usually shorter (800nt) than the pituitary 1200nt transcript the hypothalamus.9,58,59 Human and rat CRH are identical,
because of lack of exons 1 and 2 and the 5′ region of exon but ovine CRH differs by 7 amino acids60,61; ovine-sequence
3.47 Because the POMC-like peptide product from this CRH is slightly more potent than human-sequence CRH in
shorter transcript lacks a signal sequence needed to cross stimulating ACTH secretion and has a longer half-life, but
the endoplasmic reticulum, it is probable that it is neither both are used diagnostically.
secreted nor active in normal circumstances. However, in CRH is secreted into the hypophyseal portal blood,
ectopic ACTH syndrome, additional POMC mRNA species where it binds to specific type I CRH receptors on anterior
are described that are longer than the normal pituitary pituitary corticotrophs62 to stimulate POMC transcription
POMC species (typically 1450nt) as a result of the use of through a process that includes activation of adenylate
alternative promoters in the 5′ region of the gene.48,49 This cyclase. It is unclear whether hypothalamic CRH contrib-
may in part explain the resistance of POMC expression to utes in any way to circulating levels; CRH is also synthe-
glucocorticoid feedback in these tumors. Other factors, sized in other tissues, and it is likely that circulating CRH
including interaction with tissue-specific transcription reflects synthesis from testis, gastrointestinal tract, adrenal
factors50 and lack of POMC promoter methylation,51 medulla, and particularly the placenta,63 in which the
may explain the ectopic expression of ACTH in some increased secretion across pregnancy results in a threefold
malignant tissues. The cleavage of POMC is also tissue increase in circulating CRH levels.64 In the circulation, CRH
specific,52 and it is possible, at least in some cases of ectopic is bound to CRH-binding protein (CRH-BP); levels of
ACTH syndrome, that circulating ACTH precursors (notably CRH-BP also increase during pregnancy so that cortisol
pro-ACTH) may cross-react in current ACTH radioimmu- secretion is not markedly elevated.65
noassays.53,54 The biologic activity of POMC itself on CRH is the principal stimulus for ACTH secretion,66 but
adrenal function is thought to be negligible. AVP is able to potentiate CRH-mediated secretion.67 In this
POMC expression and processing within neurons in the case, AVP acts through the V1b receptor to activate protein
hypothalamus, specifically the generation of α-MSH that kinase C. The peak response of ACTH to CRH does not
interacts with melanocortin-4 receptors (MC4R), appears differ across the day, but it is affected by endogenous
CHAPTER 15 The Adrenal Cortex 497

Diurnal rhythm Renal arterial pressure


β-Adrenergic action
Stressors ANP
Prostaglandins
(hypoglycemia, Dopamine
hypotension, Hypothalamus
fever, trauma, –
surgery)
CRH Extracellular
ADH Liver volume
Cytokines Renal arterial
Renin pressure

Na+ (+ water)
Pituitary Kidneys retention
Angiotensinogen
K+ excretion
ACTH
Aldosterone

ECF [K+]
Adrenals

Cortisol
Angiotensin I

Metabolism Cardiovascular system Lungs


Gluconeogenesis Myocardial contractility
Glycogenolysis Cardiac output
Proteolysis Catecholamine pressor
Lipolysis effect
Angiotensin II
A Regulation of cortisol secretion B Regulation of aldosterone secretion
Figure 15-7 Normal negative feedback regulation of cortisol and aldosterone secretion. A, Hypothalamic-pituitary-adrenal axis. Adrenocorticotropic hormone (ACTH) is secreted
from the anterior pituitary under the influence of two principal secretagogues, corticotropin-releasing hormone (CRH) and arginine vasopressin; other factors, including cytokines,
also play a role. CRH secretion is regulated by an inbuilt circadian rhythm and by additional stressors operating through the hypothalamus. Secretion of CRH and ACTH is
inhibited by cortisol, highlighting the importance of negative feedback control. B, Renin-angiotensin-aldosterone system (RAAS). Renin is secreted from the juxtaglomerular
cells in the kidney dependent on renal arterial blood pressure. Renin converts angiotensinogen to angiotensin I, which is converted in the lungs by angiotensin-converting enzyme
(ACE) into angiotensin II. Angiotensin stimulates adrenal aldosterone synthesis. Extracellular fraction (ECF) of potassium has an important direct inhibitory influence on aldosterone
secretion. ADH, antidiuretic hormone (arginine vasopressin); ANP, atrial natriuretic peptide.

function of the HPA axis in that responsiveness is reduced average ACTH pulse frequency is higher in normal adult
in subjects treated with corticosteroids but increased in men compared with women (18 versus 10 pulses/24 hours,
subjects with Cushing disease. Other reported ACTH secre- respectively). The circadian ACTH rhythm appears to be
tagogues, including angiotensin II, cholecystokinin, atrial mediated principally by an increased ACTH pulse ampli-
natriuretic factor, and vasoactive peptides, probably act to tude occurring between 5 and 9 AM but also by a reduction
modulate the CRH control of ACTH secretion.68 in ACTH pulse frequency occurring between 6 PM and
The Stress Response and Immune-Endocrine Axis. The proin- midnight.78,79 Food ingestion is a further stimulus to ACTH
flammatory cytokines, notably interleukin 1 (IL-1), IL-6, secretion. An ultradian rhythm overlies the circadian and
and tumor necrosis factor-α, also increase ACTH secretion, appears to be driven by an oscillator created between the
either directly or by augmenting the effect of CRH.69,70 secretion of ACTH, the short delay in response at the
Leukemia inhibitory factor (LIF), a cytokine of the IL-6 adrenal, and the subsequent negative feedback by cortisol
family, is a further activator of the HPA axis.71 This explains at the hypothalamus and pituitary.80
the response of the HPA axis to an inflammatory stimulus Circadian rhythm is dependent on both day-night81 and
and is an important immune-endocrine interaction (see sleep-wake82 patterns and is disrupted by alternating day-
Chapter 7). Physical stresses increase ACTH and cortisol night shift work and by long-distance travel across time
secretion, again through central actions mediated via CRH zones.83 It can take up to 2 weeks for the circadian rhythm
and AVP. Cortisol secretion rises in response to fever, to reset to an altered day-night cycle.
surgery,72 burn injury,73 hypoglycemia,74 hypotension, and Negative Feedback. An important aspect of CRH and ACTH
exercise.75 In all of these cases, this increased secretion can secretion is the negative feedback control exerted by glu-
be viewed as a normal counterregulatory response to the cocorticoids themselves. Glucocorticoids inhibit POMC
insult. Acute psychological stress raises cortisol levels,76 but transcription in the anterior pituitary56 and CRH and AVP
secretion rates appear to be normal in patients with chronic mRNA synthesis and secretion in the hypothalamus.84,85
anxiety states and underlying psychotic illness. However, Annexin 1 (formerly called lipocortin 1) may also play a
depression is associated with high circulating cortisol con- critical role in effecting the negative feedback of glucocor-
centrations, and this is an important consideration in the ticoids on ACTH and CRH release.86 The negative feedback
differential diagnosis of Cushing syndrome (see later effect depends on the dose, potency, half-life, and duration
discussion). of administration of the glucocorticoid and has important
Circadian Rhythm. ACTH, and hence cortisol, is secreted in physiologic and diagnostic consequences. Suppression of
a pulsatile fashion with a circadian rhythm; levels are the HPA axis by pharmacologic corticosteroids may persist
highest on wakening and decline throughout the day, for many months after cessation of therapy, and adreno-
reaching nadir values in the evening (Fig. 15-8).77 The cortical insufficiency should be anticipated. Diagnostically,
498 SECTION IV Adrenal Cortex and Endocrine Hypertension

ACTH Normal The ACTH Receptor and ACTH Effects on the Adrenal Gland
25
ACTH binds to a G protein–coupled, melanocortin-2 recep-
20 tor (MC2R),89 of which there are approximately 3500 on
each adrenocortical cell. Melanocortin-2 receptor accessory
15 protein (MRAP) is required for correct localization and sig-
ng/L

naling of MC2R.90 Current data suggest that MRAP might


10 promote three different activities: as a chaperone assisting
correct folding of MC2R in the endoplasmic reticulum, as
5 an accessory protein essential for trafficking of MC2R to
the plasma membrane, and as a coreceptor enabling MC2R
0 to bind or to signal ACTH response.91 Downstream signal
transduction is mediated principally through the stimula-
Cortisol Normal tion of adenylate cyclase and intracellular cAMP,92 although
600 both extracellular and intracellular Ca2+ play a role.93 Other
factors synergize with or inhibit the effects of ACTH on the
500 adrenal cortex, including angiotensin II, activin, inhibin,
400 and cytokines (tumor necrosis factor-α and leptin).94 Cell-
to-cell communication via gap junctions is also important
nmol/L

300 in mediating the effects of ACTH.95


200 ACTH produces both immediate and chronic effects on
the adrenal gland; the end result is the stimulation of
100 adrenal steroidogenesis and growth. Acutely, steroidogen-
0 esis is stimulated through a StAR-mediated increase in cho-
lesterol delivery to the CYP11A1 enzyme in the inner
mitochondrial membrane.23 Chronically (within 24 to 26
ACTH Cushing Disease hours of exposure), ACTH acts to increase the synthesis of
80 all steroidogenic CYP enzymes (CYP11A1, CYP17A1,
CYP21A2, CYP11B1) in addition to adrenodoxin,96,97 the
60 effects of which are mediated at the transcriptional level.
ACTH increases synthesis of the LDL and HDL receptors
and possibly also synthesis of 3-hydroxy-3-methylglutaryl
ng/L

40
(HMG)-CoA reductase, the rate-limiting step in cholesterol
biosynthesis. ACTH increases adrenal weight by inducing
20 both hyperplasia and hypertrophy. Adrenal atrophy is a
feature of ACTH deficiency.
0
Mineralocorticoid Secretion:
Cortisol Cushing Disease The Renin-Angiotensin-Aldosterone Axis
1400 Aldosterone is secreted from the ZG under the control of
1200 three principal secretagogues: angiotensin II, potassium,
1000 and, to a lesser extent, ACTH (see Fig. 15-7). Other factors,
notably somatostatin, heparin, atrial natriuretic factor,
nmol/L

800
and dopamine, can directly inhibit aldosterone synthesis.
600 The secretion of aldosterone and its intermediary 18-
400 hydroxylated metabolites is restricted to the ZG because
200
of the zone-specific expression of CYP11B2 (aldosterone
synthase).98 Corticosterone and DOC, synthesized in both
0 the ZF and ZG, can act as mineralocorticoids, which
becomes significant in some clinical diseases, notably some
24 2 4 6 8 10 12 14 16 18 20 22 24
forms of congenital adrenal hyperplasia (CAH) and adrenal
Time (24-hour clock) tumors. Similarly, it is now established that cortisol can
Figure 15-8 Circadian and pulsatile secretion of adrenocorticotropic hormone act as a mineralocorticoid in the setting of impaired
(ACTH) and cortisol in a normal subject (top two panels) and in a patient with Cushing metabolism of cortisol to cortisone by the enzyme
disease. In a normal subject, secretion of ACTH and cortisol is highest in early morning 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2); this
and falls to a nadir at midnight. ACTH pulse frequency and pulse amplitude are is important in patients with hypertension, ectopic ACTH
increased in Cushing disease, and circadian rhythmic secretion is lost. syndrome, or renal disease. The renin-angiotensin system
is described in detail in Chapter 16.
Angiotensin II and potassium stimulate aldosterone
secretion principally by increasing the transcription of
the feedback mechanism explains ACTH hypersecretion in CYP11B2 through common intracellular signaling path-
Addison disease, as well as undetectable ACTH levels in ways. cAMP response elements in the 5′ region of the
patients with a cortisol-secreting adrenal adenoma. Feed- CYP11B2 gene are activated after an increase in intracellular
back inhibition is principally mediated via the glucocorti- Ca2+ and activation of calmodulin kinases. The potassium
coid receptor (GR); patients with glucocorticoid resistance effect is mediated through membrane depolarization and
resulting from mutations in the GR87 and mice lacking the opening of calcium channels and the angiotensin II effect
GR gene88 have ACTH and cortisol hypersecretion due to after binding of angiotensin II to the surface AT1 receptor
perceived lack of negative feedback. and activation of phospholipase C.98
CHAPTER 15 The Adrenal Cortex 499

The effect of ACTH on aldosterone secretion is modest TABLE 15-4


and differs in the acute and chronic situation (see Chapter Dissociation of Adrenal Androgen and Glucocorticoid
16). An acute bolus of ACTH will increase aldosterone Secretion: Evidence for an Adrenal-Stimulating Hormone
secretion, principally by stimulating the early pathways of
adrenal steroidogenesis (see earlier discussion), but circulat- Dexamethasone studies: Complete cortisol suppression with chronic
ing levels increase by no more than 10% to 20% above high-dose dexamethasone; DHEA falls by only 20% (greater
baseline values. ACTH has no effect on CYP11B2 gene tran- sensitivity of DHEA to acute low-dose dexamethasone
administration).
scription or enzyme activity. Chronic continual ACTH
Adrenarche: Clinically significant rise in circulating DHEA at 6 to 8
stimulation has either no effect or an inhibitory effect on years of age; cortisol production unaltered.
aldosterone production, possibly because of receptor down- Aging: Reduction in DHEA production; no change in cortisol.
regulation or suppression of angiotensin II–stimulated Anorexia nervosa and illness: Fall in DHEA, no change (or increase) in
secretion because of a mineralocorticoid effect of cortisol, cortisol.
DOC, or corticosterone. Dopamine and atrial natriuretic
DHEA, dehydroepiandrosterone.
peptide inhibit aldosterone secretion, as does heparin.
These separate lines of control—through the HPA
axis for glucocorticoid biosynthesis and via the renin-
angiotensin system for mineralocorticoid synthesis—have activity of 17α-hydroxylase. Factors that determine whether
important clinical consequences. Patients with primary the 17-hydroxylated substrates, 17-OH pregnenolone and
adrenal failure invariably have both cortisol and aldoster- 17-OHP, will undergo 21-hydroxylation to form glucocor-
one deficiency, whereas patients with ACTH deficiency ticoid or side-chain cleavage by 17α-hydroxylase to form
due to pituitary disease have glucocorticoid deficiency but DHEA and androstenedione are unresolved and seem likely
normal aldosterone concentrations because the renin- to be important in defining the activity of any putative
angiotensin system is intact. CASH (Table 15-4).

Adrenal Androgen Secretion CORTICOSTEROID HORMONE ACTION


Adrenal androgens represent an important component
(>50%) of circulating androgens in premenopausal
women.99 In men, this contribution is much smaller
Receptors and Gene Transcription
because of the testicular production of androgens, but Both cortisol and aldosterone exert their effects after uptake
adrenal androgen excess even in men may be of clinical of free hormone from the circulation and binding to
significance, notably in patients with CAH, which results intracellular receptors; these are termed, respectively, the
in a suppression of the hypothalamic-pituitary-gonadal glucocorticoid receptor (GR, encoded by NR3C1) and the min-
axis. The adult adrenal secretes approximately 4 mg/day of eralocorticoid receptor (MR, encoded by NR3C2).104-106 These
DHEA, 7 to 15 mg/day of DHEAS, 1.5 mg of androstenedi- are members of the thyroid/steroid hormone receptor
one, and 0.05 mg/day of testosterone. superfamily of transcription factors; they consist of a
DHEA is a crucial precursor of human sex steroid bio- carboxy-terminal ligand-binding domain, a central DNA-
synthesis and exerts androgenic or estrogenic activity after binding domain that interacts with specific DNA sequences
conversion by the activities of the 3β-HSD superfamily (β- on target genes, and an amino-terminal hypervariable
HSD isozymes and aromatase); these enzymes are expressed region. Although only single genes encode the GR and MR,
in peripheral target tissues, a fact that is of clinical impor- splice variants have been described in both receptor types;
tance in many diseases.100 Some studies have postulated this, together with tissue-specific post-translational modi-
direct effects of DHEA acting as a classic hormone in fication (phosphorylation, sumoylation, and ubiquitina-
peripheral tissues. Specific plasma membrane receptors tion), is thought to account for many of the diverse actions
have been identified but await full characterization.101 Con- of corticosteroids (Fig. 15-9).107,108
ventionally, desulfated DHEA is thought to be converted Glucocorticoid hormone action has been studied in
downstream to a biologically active hormone. Serum more depth than mineralocorticoid action. The binding of
DHEAS was previously thought to represent a circulating steroid to the GRα in the cytosol results in activation of
storage pool for DHEA regeneration, but it was later sug- the steroid-receptor complex through a process that
gested that conversion of DHEAS to DHEA by steroid sul- involves the dissociation of heat shock proteins (HSP90
fatase plays a minor role in adult physiology and that the and HSP70).109 Following translocation to the nucleus,
equilibrium between serum DHEA and DHEAS is mainly gene transcription is stimulated or repressed after binding
regulated by SULT2A1 activity. This implies that serum of the dimerized GR-ligand complex to a specific DNA
DHEAS may not always appropriately reflect the active sequence in the promoter regions of target genes.110 This
DHEA pool, particularly if SULT2A1 activity is impaired, as sequence, known as the glucocorticoid-response element
in the inflammatory stress response.102 (GRE), is invariably a palindromic CGTACAnnnTGTACT
DHEAS stimulates androgen secretion; DHEA (but not sequence that binds with high affinity to two loops of DNA
DHEAS because of its increased plasma half-life) and andro- within the DNA-binding domain of the GR (zinc fingers).
stenedione demonstrate a circadian rhythm similar to This stabilizes the RNA polymerase II complex, facilitating
that of cortisol.103 However, there are many discrepancies gene transcription. The GRβ variant may act as a dominant
between adrenal androgen and glucocorticoid secretion, negative regulator of GRα transactivation.107
leading to the suggestion of an additional cortical androgen- Naturally occurring mutations in the GR (as seen in
stimulating hormone (CASH). Many putative CASHs have patients with glucocorticoid resistance) and GR mutants
been proposed, including POMC derivatives such as joining generated in vitro have highlighted critical regions of the
peptide, prolactin, and insulin-like growth factor type 1 receptor that are responsible for binding and transactiva-
(IGF-1), but conclusive proof is lacking. Efficient adrenal tion,111 but numerous other factors are required (e.g.,
steroidogenesis toward androgen synthesis is crucially coactivators, corepressors112), and this may make responses
dependent on the relative activities of 3β-HSD and tissue specific. This is a rapidly evolving field and beyond
17α-hydroxylase and, in particular, on the 17,20-lyase the scope of this chapter. However, the interaction between
500 SECTION IV Adrenal Cortex and Endocrine Hypertension

Glucocorticoid receptor Mineralocorticoid receptor

1 2 3 4 5 6 7 8 9α 9β 1β 1α 2 3 4 5 6 7 8 9

GRα mRNA 9α GRβ mRNA 9β 1α αMR mRNA 1β βMR mRNA

GRα protein GRβ protein MR protein

GRα elicits GRβ functions


specific biological as a dominant
responses negative inhibitor
of GRα receptor
Figure 15-9 Schematic structure of the human genes encoding the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR). In both cases, splice variants have been
described. In the case of the GR, there is evidence that the GRβ isoform can act as a dominant negative inhibitor of GRα action. mRNA, messenger ribonucleic acid.

Cortisol NF-κB
2 Cytokines
GR

GR
1κB
Coactivator Coactivator
complex complex +
3

HSP C-fos C-jun GR
GR
HSP

Nucleus Inflammatory response


1κB
(TNF-α, IL-1β, IL-6,
1 IL-8, MCSF)

+
Cell
membrane
1κB

Figure 15-10 The anti-inflammatory action of glucocorticoids. Cortisol binds to the cytoplasmic glucocorticoid receptor (GR). Conformational changes in the receptor-ligand
complex result in dissociation from heat shock proteins (HSP70 and HSP90) and migration to the nucleus. Binding occurs to specific DNA motifs—glucocorticoid response
elements—in association with the activator protein 1 (AP1) comprising C-fos and C-jun. Glucocorticoids mediate their anti-inflammatory effects through several mechanisms:
(1) the inhibitory protein 1κB, which binds and inactivates nuclear factor-κB (NF-κB), is induced; (2) the GR-cortisol complex is able to bind NF-κB and thereby prevent initiation
of an inflammatory process; (3) GR and NF-κB compete for the limited availability of coactivators, which include cyclic adenosine monophosphate response element–binding
protein (CREB) and steroid receptor coactivator-1. IL, interleukin; MCSF, macrophage colony-stimulating factor; TNF-α, tumor necrosis factor-α.

GR and two particular transcription factors are important nuclear factor-κB (NF-κB), a ubiquitously expressed tran-
in mediating the anti-inflammatory effects of glucocorti- scription factor that activates a series of genes involved in
coids and explain the effect of glucocorticoids on genes lymphocyte development, inflammatory response, host
that do not contain obvious GREs in their promoter defense, and apoptosis (Fig. 15-10).115 In keeping with the
regions.113 Activator protein 1 (AP1) comprises Fos and Jun diverse array of actions of cortisol, many hundreds of
subunits and is a proinflammatory transcription factor glucocorticoid-responsive genes have been identified.
induced by a series of cytokines and phorbol ester. Some glucocorticoid-induced genes and repressed genes
The GR-ligand complex can bind to c-Jun and prevent are listed in Table 15-5.
interaction with the AP1 site, thereby mediating the In contrast to the diverse actions of glucocorticoids,
so-called transrepressive effects of glucocorticoids.114 Simi- mineralocorticoids have a more restricted role, principally
larly, functional antagonism exists between the GR and stimulation of epithelial sodium transport in the distal
CHAPTER 15 The Adrenal Cortex 501

TABLE 15-5 Na+


ENaC
Some of the Genes Regulated by Glucocorticoids or
Glucocorticoid Receptors
Site of Action Induced Genes Repressed Genes
Immune IκB (nuclear factor-κB Interleukins
system inhibitor)
Haptoglobin Tumor necrosis
factor-α (TNF-α) SGK K-Ras Cortisol
T-cell receptor (TCR)-ζ Interferon-γ
p21, p27, and p57 E-selectin
Lipocortin Intercellular adhesion Regulatory Structural MR
molecule-1 proteins protein 11β-HSD2
Cyclooxygenase 2
Inducible nitric oxide Cortisone
synthase (iNOS)
Metabolic PPAR-γ Tryptophan MR
hydroxylase K+ K+
Tyrosine aminotransferase Metalloprotease
Glutamine synthase
Glycogen synthase
Glucose-6-phosphatase Na+ Na+ Aldosterone
PEPCK
Leptin Figure 15-11 Mineralocorticoid hormone action. An epithelial cell in the distal
γ-Fibrinogen nephron or distal colon is depicted. The much higher concentrations of cortisol
Cholesterol 7α-hydroxylase are inactivated by the type 2 isozyme of 11β-hydroxysteroid dehydrogenase (11β-
C/EBP/β HSD2) to cortisone, permitting the endogenous ligand, aldosterone, to bind to the
Bone Androgen receptor Osteocalcin mineralocorticoid receptor (MR). Relatively few mineralocorticoid target genes have
Calcitonin receptor Collagenase been identified, but they include serum- and glucocorticoid-induced kinase (SGK),
Alkaline phosphatase subunits of the epithelial sodium channel (ENaC), and basolateral Na+/K+-adenosine
IGFBP6 triphosphatase.
Channels and ENaC-α, -β, and -γ
transporters SGK
Aquaporin 1
Endocrine Basic fibroblast growth Glucocorticoid The MR and GR share considerable homology—57% in
factor (bFGF) receptor the steroid-binding domain and 94% in the DNA-binding
Vasoactive intestinal Prolactin domain. It is perhaps not surprising, therefore, that there
peptide is promiscuity of ligand binding, with aldosterone (and the
Endothelin POMC/CRH synthetic mineralocorticoid, fludrocortisone) binding to
Retinoid X receptor PTHrP
the GR and cortisol binding to the MR. For the MR, this is
GHRH receptor Vasopressin
Natriuretic peptide
particularly impressive: in vitro, the MR has the same
receptors inherent affinity for aldosterone, corticosterone, or corti-
Growth and Surfactant proteins A, B, Fibronectin sol.105 Specificity on the MR is conferred through the “pre-
development and C α-Fetoprotein receptor” metabolism of cortisol via the enzyme HSD11B2,
Nerve growth factor which converts cortisol and corticosterone to inactive
Erythropoietin 11-keto metabolites, enabling aldosterone to bind to the
G1 cyclins MR.120,121 Mineralocorticoid hormone action was extended
Cyclin-dependent beyond this classic action in sodium-transporting epithelia
kinases
with the demonstration that aldosterone can induce
CRH, corticotropin-releasing hormone; C/EBP/β, CAAT-enhancer binding cardiac fibrosis and inflammatory changes in renal vascu-
protein-β; ENaC, epithelial sodium channel; GHRH, growth hormone– lature. The underlying signaling pathways remain to be
releasing hormone; IGFBP6, insulin-like growth factor–binding protein 6; fully clarified, but the effects are reversible with MR
PEPCK, phosphoenolpyruvate carboxykinase; POMC, pro-opiomelanocortin;
PPAR, peroxisome proliferator-activated receptor; PTHrP, parathyroid antagonists.122
hormone-related protein; SGK, serum- and glucocorticoid-induced kinase. Finally, for both glucocorticoids and mineralocorticoids,
Modified from McKay LI, Cidlowski JA. Molecular control of immune/ there is accumulating evidence for so-called nongenomic
inflammatory responses: interactions between nuclear factor-κB and effects involving hormone response obviating the genomic
steroid receptor-signalling pathways. Endocr Rev. 1999;20:435-459.
GR or MR. A series of responses have been reported to occur
within seconds or minutes after exposure to corticosteroids
and are thought to be mediated by as yet uncharacterized
membrane-coupled receptors.123,124

nephron, distal colon, and salivary glands.116 This action is Cortisol-Binding Globulin and Corticosteroid
mediated through induction of the apical sodium channel
(comprising three subunits—α, β, and γ)117 and the α1 and
Hormone Metabolism
β1 subunits of the basolateral sodium-potassium adenosine More than 90% of circulating cortisol is bound pre-
triphosphatase pump (Na+/K+-ATPase)118 through transcrip- dominantly to the α2-globulin, cortisol-binding globulin
tional regulation of serum- and glucocorticoid-induced (CBG).125 This 383–amino acid protein is synthesized in the
kinase (SGK).119 Aldosterone binds to the MR, principally liver and binds cortisol with high affinity. Affinity for syn-
in the cytosol (although there is evidence for expression of thetic corticosteroids is negligible except for prednisolone,
the unliganded MR in the nucleus), and the hormone- which has an affinity for CBG approximately half that of
receptor complex is then translocated to the nucleus cortisol. Circulating CBG concentrations are approximately
(Fig. 15-11). 700 nmol/L. Levels are increased by estrogens and in some
502 SECTION IV Adrenal Cortex and Endocrine Hypertension

CH2OH CH2OH

H C OH H C OH
O OH HO OH

O O
20β-dihydrocortisone 20β-dihydrocortisol

20b-oxoreductase 20b-oxoreductase
CH2OH CH2OH
CH2OH CH2OH

C O C O C O C O
O OH O OH HO OH HO OH
11b-hydroxysteroid
6 β -hydroxylase dehydrogenase 6 β -hydroxylase

O O
O O
OH OH
Cortisone Cortisol
6β-hydroxycortisone 6β-hydroxycortisol
5b-reductase 5 α -reductase

CH2OH CH2OH

C O C O
HO OH HO OH

O O
5β-dihydrocortisol 5α-dihydrocortisol

3 α -hydroxysteroid 3 α -hydroxysteroid
dehydrogenase dehydrogenase

CH2OH CH2OH CH2OH

C O C O C O
O OH HO OH HO OH

HO HO HO
tetrahydrocortisone 5β-tetrahydrocortisol 5α-tetrahydrocortisol
(THE) (THF) (allo-THF)
Figure 15-12 The principal pathways of cortisol metabolism. Interconversion of hormonally active cortisol to inactive cortisone is catalyzed by two isozymes of 11β-hydroxysteroid
dehydrogenase (11β-HSD), with HSD11β1 principally converting cortisone to cortisol and HSD11β2 doing the reverse. Cortisol can be hydroxylated at the C6 and C20 positions.
A ring reduction is undertaken by 5α-reductase or 5β-reductase and 3α-HSD.

patients with chronic active hepatitis; they are reduced by normal. Only this free circulating fraction is available for
glucocorticoids and in patients with cirrhosis, nephrosis, transport into tissues for biologic activity. The excretion
and hyperthyroidism. The estrogen effect can be marked, of free cortisol through the kidneys results in urinary free
with levels increasing twofold to threefold across preg- cortisol, which represents only 1% of the total cortisol
nancy, a fact that should be taken into account when secretion.
measuring plasma total cortisol in pregnancy and in The circulating half-life of cortisol varies between 70
women taking estrogens. and 120 minutes. The major steps in cortisol metabolism
Inherited abnormalities in CBG synthesis are much are depicted in Figure 15-12128 and can be summarized as
rarer than those described for thyroxine-binding globulin follows:
but include cases of elevated CBG, partial or complete • Interconversion of the 11-hydroxyl group (cortisol, Ken-
deficiency of CBG, and CBG variants with reduced affinity dall’s compound F) to the 11-oxo group (cortisone, com-
for cortisol.126,127 In each case, alterations in CBG concen- pound E) through activity of the 11β-HSD system (EC
trations change the total circulating cortisol concentra- 1.1.1.146).129,130 The metabolism of cortisol and that of
tions accordingly, but free cortisol concentrations are cortisone then follow similar pathways.
CHAPTER 15 The Adrenal Cortex 503

• Reduction of the C4-5 double bond to form dihydrocor- given rifampicin,136 and hydrocortisone replacement ther-
tisol or dihydrocortisone, followed by hydroxylation of apy may need to be increased in treated patients who
the 3-oxo group to form tetrahydrocortisol (THF) or develop hyperthyroidism or reduced in patients with
tetrahydrocortisone (THE). The reduction of the C4-5 untreated growth hormone (GH) deficiency.
double bond can be carried out by either 5β-reductase Aldosterone is also metabolized in the liver and kidneys.
or 5α-reductase, yielding, respectively, 5β-THF (THF) In the liver, it undergoes tetrahydro reduction and is
and 5α-THF (allo-THF). In normal subjects, the ratio of excreted in the urine as a 3-glucuronide tetrahydroaldos-
THF to allo-THF is 2 : 1. THF, allo-THF, and THE are terone derivative. However, glucuronide conjugation at the
rapidly conjugated with glucuronic acid and excreted in 18 position occurs directly in the kidney, as does 3α and
the urine. 5α/5β metabolism of the free steroid.137 Because of the
• Further reduction of the 20-oxo group by either 20α- or aldehyde group at the C18 position, aldosterone is not
20β-HSD to yield α- and β-cortols and cortolones from metabolized by HSD11B2.138 Hepatic aldosterone clearance
cortisol and cortisone, respectively. Reduction of the is reduced in patients with cirrhosis, ascites, or severe con-
C20 position may also occur without A-ring reduction, gestive heart failure.
giving rise to 20α- and 20β-hydroxycortisol.
• Hydroxylation at C6 to form 6β-hydroxycortisol.
• Cleavage of THF and THE to the C19 steroids, 11-
Effects of Glucocorticoids
hydroxy- or 11-oxo- androsterone, or etiocholanolone. The principal sites of action of glucocorticoids and some
• Oxidation of the C21 position or cortols and cortolones of the consequences of glucocorticoid excess are shown in
to form the extremely polar metabolites, cortolic and Figure 15-13.
cortolonic acids.
Approximately 50% of secreted cortisol appears in the
urine as THF, allo-THF, and THE; 25% as cortols/cortolones;
Carbohydrate, Protein, and Lipid Metabolism
10% as C19 steroids; and 10% as cortolic/cortolonic Glucocorticoids increase blood glucose concentrations
acids. The remaining metabolites are free unconjugated through their action on glycogen, protein, and lipid
steroids (cortisol, cortisone, and their 6β-, and 20α/20β- metabolism. In the liver, cortisol stimulates glycogen depo-
metabolites). sition by increasing glycogen synthase and inhibiting the
The principal site of cortisol metabolism has been con- glycogen-mobilizing enzyme, glycogen phosphorylase.139
sidered to be the liver, but many of the enzymes listed Hepatic glucose output increases through the activation of
have been described in mammalian kidney, notably the key enzymes involved in gluconeogenesis, principally
interconversion of cortisol to cortisone by HSD11B2. glucose-6-phosphatase and phosphoenolpyruvate kinase
Quantitatively, this is the most important pathway. Fur- (PEPCK).140,141 In peripheral tissues (e.g., muscle, fat), cor-
thermore, the bioactivity of glucocorticoids is in part tisol inhibits glucose uptake and utilization.142 In adipose
related to the hydroxyl group at C11; because cortisone tissue, lipolysis is activated, resulting in the release of free
with a C11-oxo group is an inactive steroid, expression fatty acids into the circulation. An increase in total circulat-
of 11β-HSD in peripheral tissues plays a crucial role in ing cholesterol and triglycerides is observed, but HDL-
regulating corticosteroid hormone action. Two distinct cholesterol levels fall. Glucocorticoids also have a permissive
11β-HSD isozymes have been reported: type 1, reduced effect on other hormones, including catecholamines and
nicotinamide adenine dinucleotide phosphate (NADPH)- glucagon. The result is insulin resistance and an increase
dependent oxo-reductase expressed principally in the liver, in blood glucose concentrations, at the expense of protein
which confers bioactivity on orally administered cortisone and lipid catabolism.
by converting it to cortisol,130 and a type 2, nicotinamide Glucocorticoids stimulate adipocyte differentiation,
adenine dinucleotide (NAD)-dependent dehydrogenase. It promoting adipogenesis through the transcriptional acti-
is the HSD11B2, coexpressed with the MR in the kidney, vation of key differentiation genes, including lipoprotein
colon, and salivary gland, that inactivates cortisol to lipase, glycerol-3-phosphate dehydrogenase, and leptin.143
cortisone and permits aldosterone to bind to the MR in Long-term effects of glucocorticoid excess on adipose tissue
vivo. If this enzyme-protective mechanism is impaired, are more complex, at least in humans, in whom the deposi-
cortisol is able to act as a mineralocorticoid; this explains tion of visceral or central adipose tissue is stimulated,144
some forms of endocrine hypertension (apparent miner- providing a useful discriminatory sign for the diagnosis of
alocorticoid excess, licorice ingestion) and the mineralo- Cushing syndrome. The predilection for visceral obesity
corticoid excess state that characterizes the ectopic ACTH may relate to the increased expression of the GR145 and
syndrome.129,131 HSD11B1 in omental compared with subcutaneous adipose
Hyperthyroidism results in increased cortisol metabo- tissue.146
lism and clearance, and hypothyroidism produces the
converse, principally because of an effect of thyroid hor-
mone on hepatic HSD11B1 and 5α/5β-reductases.130 IGF-1
Skin, Muscle, and Connective Tissue
increases cortisol clearance by inhibiting hepatic HSD11B1 In addition to inducing insulin resistance in muscle tissue,
(conversion of cortisone to cortisol).132 6β-Hydroxylation glucocorticoids also cause catabolic changes in muscle,
is normally a minor pathway, but cortisol itself induces skin, and connective tissue. In the skin and connective
6β-hydroxylase so that 6β-hydroxycortisol excretion is tissue, glucocorticoids inhibit epidermal cell division and
markedly increased in patients with Cushing syndrome.133 DNA synthesis and reduce synthesis and production of
Some drugs, notably rifampicin and phenytoin, increase collagen.147 In muscle, glucocorticoids cause atrophy (but
cortisol clearance through this pathway.134 Patients with not necrosis), which seems to be specific for type II (phasic)
renal disease have impaired cortisol clearance because of muscle fibers. Muscle protein synthesis is reduced.
reduced conversion of renal cortisol to cortisone.135 These
observations have clinical implications for patients with
thyroid disease, acromegaly, or renal disease and for
Bone and Calcium Metabolism
patients taking cortisol replacement therapy. Adrenal crisis Glucocorticoids inhibit osteoblast function, which is
has been reported in steroid-replaced addisonian patients thought to account for the osteopenia and osteoporosis
504 SECTION IV Adrenal Cortex and Endocrine Hypertension

Endocrine system:
LH, FSH release
TSH release
Brain/CNS: GH secretion
Depression
Psychosis

Eye:
Glaucoma

Carbohydrate/lipid metabolism:
Hepatic glycogen deposition
Peripheral insulin resistance
GI tract:
Gluconeogenesis
Free fatty acid production Peptic ulcerations
Overall diabetogenic effect

Cardiovascular/renal:
Salt and water retention
Hypertension
Adipose tissue distribution:
Promotes visceral obesity

Bone and calcium metabolism:


Bone formation
Bone mass and osteoporosis

Growth and development:


Skin/muscle/connective tissue: Linear growth
Protein catabolism/collagen breakdown
Skin thinning
Immune system:
Muscular atrophy
Anti-inflammatory action
Immunosuppression
Figure 15-13 The principal sites of action of glucocorticoids in humans, highlighting some of the consequences of glucocorticoid excess. CNS, central nervous system; FSH,
follicle-stimulating hormone; GH, growth hormone; GI, gastrointestinal; LH, luteinizing hormone; TSH, thyroid-stimulating hormone.

that characterize glucocorticoid excess.148 Up to 1% of renal calcium excretion. As a consequence, parathyroid


Western populations are taking long-term glucocorticoid secretion is usually increased. In children, glucocorticoids
therapy,149 and glucocorticoid-induced osteoporosis is suppress growth, but the increases in body mass index
becoming a prevalent health concern, affecting 50% of (BMI) are thought to offset a deleterious effect on bone
patients treated with corticosteroids for longer than 12 mineral density.152
months. However, the complication perhaps most feared
by physicians is osteonecrosis. Osteonecrosis (also termed Salt and Water Homeostasis and Blood
avascular necrosis) produces rapid and focal deterioration of
bone quality and primarily affects the femoral head, leading
Pressure Control
to pain and ultimately to collapse of the bone, often neces- Glucocorticoids increase blood pressure by a variety of
sitating hip replacement. It can affect individuals of any mechanisms involving actions on the kidney and vascula-
age and may occur with relatively low doses of glucocorti- ture.153 In vascular smooth muscle, they increase sensitivity
coids (e.g., during corticosteroid replacement therapy for to pressor agents such as catecholamines and angiotensin
adrenal failure).150 Importantly, defects may not be detect- II while reducing nitric oxide–mediated endothelial dilata-
able on conventional radiographs but are readily seen tion. Angiotensinogen synthesis is increased by glucocor-
on magnetic resonance imaging (MRI). Glucocorticoid- ticoids.154 In the kidney, depending on the activity of
induced osteocyte apoptosis has been implicated in the HSD11B2, cortisol can act on the distal nephron to cause
pathogenesis of the condition,151 and the lack of a direct sodium retention and potassium loss (mediated via the
role for an interrupted blood supply suggests that the term MR).131 Elsewhere across the nephron, glucocorticoids
osteonecrosis is preferable to avascular femoral necrosis. increase the glomerular filtration rate, proximal tubular
However, there is still no explanation for individual epithelial sodium transport, and free water clearance.155
susceptibility. This last effect involves antagonism of the action of vaso-
Glucocorticoids also induce negative calcium balance by pressin and explains the dilutional hyponatremia seen in
inhibiting intestinal calcium absorption and increasing patients with glucocorticoid deficiency.156
CHAPTER 15 The Adrenal Cortex 505

colon; they mediate the corticosteroid control of epithelial


Anti-inflammatory Actions and the Immune System
ion transport.
Glucocorticoids suppress immunologic responses, and this
action has been the stimulus to develop a series of highly
potent pharmacologic glucocorticoids to treat a variety of
Growth and Development
autoimmune and inflammatory conditions. The inhibitory Although glucocorticoids stimulate transcription of the
effects are mediated at many levels. In the peripheral gene encoding GH in vitro, glucocorticoids in excess
blood, glucocorticoids reduce lymphocyte counts acutely inhibit linear skeletal growth,152,166 probably as a result of
(T lymphocytes > B lymphocytes) by redistributing lym- catabolic effects on connective tissue, muscle, and bone
phocytes from the intravascular compartment to the and through inhibition of the effects of IGF-1. The results
spleen, lymph nodes, and bone marrow. Conversely, neu- of experiments on mice lacking the GR gene88 have em-
trophil counts increase after glucocorticoid administration. phasized the role of glucocorticoids in normal fetal devel-
Eosinophil counts rapidly fall, an effect that was histori- opment. In particular, glucocorticoids stimulate lung
cally used as a bioassay for glucocorticoids. The immuno- maturation through the synthesis of surfactant proteins
logic actions of glucocorticoids involve direct actions on (SP-A, SP-B, and SP-C),167 and mice lacking the GR die
both T and B lymphocytes, including inhibition of immu- shortly after birth due to hypoxia from lung atelectasis.
noglobulin synthesis and stimulation of lymphocyte apop- Glucocorticoids also stimulate the enzyme phenylethanol-
tosis. Inhibition of cytokine production from lymphocytes amine N-methyltransferase (PNMT), which converts nor-
is mediated through inhibition of the action of NF-κB. adrenaline to adrenaline in adrenal medulla and chromaffin
NF-κB plays a crucial and generalized role in inducing tissue. Mice lacking the GR do not develop an adrenal
cytokine gene transcription; glucocorticoids can bind medulla.88
directly to NF-κB to prevent nuclear translocation, and
they induce NF-κB inhibitor, which sequesters NF-κB in the
cytoplasm, thereby inactivating its effect.115
Endocrine Effects
Additional anti-inflammatory effects involve the inhibi- Glucocorticoids suppress the thyroid axis, probably through
tion of monocyte differentiation into macrophages and a direct action on the secretion of thyroid-stimulating
macrophage phagocytosis and cytotoxic activity. Gluco- hormone (TSH, thyrotropin). In addition, they inhibit 5′
corticoids reduce the local inflammatory response by deiodinase activity that mediates the conversion of thyrox-
preventing the actions of histamine and plasminogen acti- ine to active triiodothyronine.
vators. Prostaglandin synthesis is impaired through the Glucocorticoids also act centrally to inhibit gonado-
induction of lipocortins, which inhibit phospholipase A2 tropin-releasing hormone (GnRH) pulsatility and release
activity.157 of luteinizing hormone (LH) and follicle-stimulating hor-
mone (FSH).
Central Nervous System and Mood
Clinical observations of patients with glucocorticoid excess
Therapeutic Corticosteroids
and deficiency reveal that the brain is an important target Since the dramatic anti-inflammatory effect of cortisone
tissue for glucocorticoids, with depression, euphoria, psy- was first demonstrated in the 1950s, a series of synthetic
chosis, apathy, and lethargy being important manifesta- corticosteroids have been developed for therapeutic pur-
tions. Both GRs and MRs are expressed in discrete regions poses. These agents are used to treat a diverse variety
of the rodent brain, including hippocampus, hypothala- of human diseases, principally relying on their anti-
mus, cerebellum, and cortex.158 Glucocorticoids cause neu- inflammatory and immunologic actions (Table 15-6). The
ronal death, notably in the hippocampus159; this effect may main corticosteroids used in clinical practice, together with
underlie the interest in glucocorticoids in relation to cogni- their relative glucocorticoid and mineralocorticoid poten-
tive function, memory, and neurodegenerative diseases cies, are listed in Table 15-7.
such as Alzheimer disease.160 Local blockade of cortisol The structures of common synthetic steroids are depicted
generation by HSD11B1 has been shown to improve cogni- in Figure 15-14. The biologic activity of a corticosteroid
tive function.161 DHEA has been shown to have neuropro- depends on a 4-3-keto, 11β-hydroxy, 17α,21-trihydroxyl
tective effects in the hippocampus region.162 CYP7B, an configuration.168 Conversion of the C11 hydroxyl group to
enzyme that metabolizes DHEA to its 7α-hydroxylated a C11 keto group (i.e., cortisol to cortisone) inactivates the
metabolite, is highly expressed in brain, but expression was
decreased in dentate neurons in the hippocampus.163
TABLE 15-6
Eye Therapeutic Use of Corticosteroids

In the eye, glucocorticoids act to raise intraocular pressure Endocrine: Replacement therapy (Addison disease, pituitary disease,
through an increase in aqueous humor production and congenital adrenal hyperplasia), Graves ophthalmopathy
Skin: Dermatitis, pemphigus
deposition of matrix within the trabecular meshwork,
Hematology: Leukemia, lymphoma, hemolytic anemia, idiopathic
which inhibits aqueous drainage. Steroid-induced glau- thrombocytopenic purpura
coma appears to have a genetic predisposition, but the Gastrointestinal: Inflammatory bowel disease (ulcerative colitis, Crohn
underlying mechanisms are unknown.164 disease)
Liver: Chronic active hepatitis, transplantation, organ rejection
Renal: Nephrotic syndrome, vasculitides, transplantation, rejection
Gut Central nervous system: Cerebral edema, raised intracranial pressure
Long-term but not acute administration of glucocorticoids Respiratory: Angioedema, anaphylaxis, asthma, sarcoidosis,
increases the risk of developing peptic ulcer disease.165 Pan- tuberculosis, obstructive airway disease
Rheumatology: Systemic lupus erythematosus, polyarteritis, temporal
creatitis with fat necrosis is reported in patients with glu-
arteritis, rheumatoid arthritis
cocorticoid excess. The GR is expressed throughout the Muscle: Polymyalgia rheumatica, myasthenia gravis
gastrointestinal tract, and the MR is expressed in the distal
506 SECTION IV Adrenal Cortex and Endocrine Hypertension

TABLE 15-7 CH2OH CH2OH


Relative Biologic Potencies of Synthetic Steroids in C O C O
Bioassay Systems HO OH O OH
Anti- Hypothalamic-
inflammatory Pituitary-Adrenal Salt
Steroid Action Suppression Retention
O O
Cortisol 1 1 1
Prednisone 3 4 0.75 Cortisol Cortisone
Prednisolone 3 4 0.75
Methylprednisolone 6.2 4 0.5
Fludrocortisone 12 12 125 CH2 OH CH2 OH
Δ1 Fludrocortisone 14 — 225
Triamcinolone 5 4 0 C O C O
Dexamethasone 26 17 0 HO OH O OH

steroid. The addition of a 1,2 unsaturated bond to cortisol


results in prednisolone, which is four times more potent O O
than cortisol in classic glucocorticoid bioassays such as Prednisone Prednisone
hepatic glycogen deposition, suppression of eosinophils,
and anti-inflammatory actions. Prednisone, widely pre- CH2OH
scribed in the United States, is the cortisone equivalent CH2OH
C O
of prednisolone and relies on conversion by HSD11B1 in HO OH C O
the liver for bioactivity.169 Potency is further increased HO OH
by the addition of a 6α-methyl group to prednisolone CH 3
(methylprednisolone).
Fludrocortisone is a synthetic mineralocorticoid having O F
125-fold greater potency than cortisol in stimulating CH3 O
sodium reabsorption. This effect is achieved through the Methylprednisolone Dexamethasone
addition of a 9α-fluoro group to cortisol. Fludrocortisone
also has glucocorticoid potency (12-fold greater than
cortisol). The addition of a 16α-methyl group and 1,2 CH 2OH CH2OH
saturated bond to fludrocortisone results in dexametha-
C O C O
sone, a highly potent glucocorticoid (25-fold greater
HO OH HO OH
potency than cortisol) that has negligible mineralocorti- OH
coid activity.168,170
F F
Administration O O

Widely used synthetic glucocorticoids in respiratory and Triamcinolone Fludrocortisone


nasal aerosol sprays are betamethasone, beclomethasone,
and fluticasone. Betamethasone has the same structure as SCH 2F CH 2 OH
dexamethasone but with a 16β-methyl group. Beclometha-
sone has the same structure as betamethasone apart from C O C O
the replacement of the 9α-fluoro group with a 9α-chloro HO OH HO OH
group. Fluticasone has the same structure as dexametha- CH3 CH3
sone with an additional 6α-fluoro group and a 5-fluoromethyl
group replacing the hydroxymethyl group. F Cl
Corticosteroids are given orally, parenterally, and by O O
F
numerous topical routes (e.g., eyes, skin, nose, inhalation,
rectal suppositories).170 Unlike hydrocortisone, which has Fluticasone Beclomethasone
a high affinity for CBG, most synthetic steroids have low
affinity for this binding protein and circulate as free steroid Figure 15-14 Structures of the natural glucocorticoid cortisol, some of the more
(approximately 30%) or bound to albumin (approximately commonly prescribed synthetic glucocorticoids, and the mineralocorticoid fludrocorti-
70%). Circulating half-lives vary depending on individual sone. Triamcinolone is identical to dexamethasone except that a 16α-hydroxyl group
variability and underlying disease, particularly renal and is substituted for the 16α-methyl group. Betamethasone, another widely used gluco-
hepatic impairment. Cortisone acetate should not be used corticoid, has a 16β-methyl group. Beclomethasone is derived from betamethasone
by replacement of the 9α-fluoro group with a chloro group. Fluticasone is identical
parenterally because it requires metabolism by the liver to to dexamethasone except that an additional 6α-fluoro group has been added, and the
active cortisol. hydroxymethyl group at position 21 has been exchanged by a 5-fluoromethyl group.
It is beyond the scope of this chapter to describe which
steroid should be given and by which route for the non-
endocrine conditions listed in Table 15-6. Acute and long- particularly in patients with respiratory or rheumatologic
term corticosteroid therapy in patients with hypoadrenalism disease, to such an extent that up to 1% of the population
or CAH is discussed in later sections. is now prescribed long-term corticosteroid therapy.149
Because of their established euphoric effect, corticosteroids
often make patients feel better but without any objective
Long-Term Therapy improvements in underlying disease parameters. In view
In addition to the undoubted benefit that corticosteroids of the long-term harm of chronic glucocorticoid excess,171
provide, there is an increasing incidence of overuse, decisions regarding treatment should be evidence based
CHAPTER 15 The Adrenal Cortex 507

and subject to regular review based on efficacy and side lone, dexamethasone) sources. Iatrogenic Cushing syn-
effects. The endocrinologic consequences of chronic gluco- drome is common,170,175 occurring to some degree in most
corticoid excess, notably suppression of the HPA axis, are patients taking long-term corticosteroid therapy. Endoge-
an important aspect of modern clinical practice and are nous causes of Cushing syndrome are rare and result in loss
described later (see “Primary and Central Hypoadrenal- of the normal feedback mechanism of the HPA axis and
ism”). Endocrinologists need to be aware of the effects of the normal circadian rhythm of cortisol secretion.
long-term therapy and of steroid withdrawal. Selective glu- The incidence of Cushing disease is estimated to be 2 to
cocorticoid receptor agonists (SEGRAs) are being developed 3 cases per 1 million population per year. The incidence of
with the aim of dissociating the transrepressive, anti- ectopic ACTH syndrome parallels that of bronchogenic
inflammatory actions of glucocorticoids from the transac- carcinoma, and although 0.5% of lung cancer patients
tivating effects that, by and large, mediate deleterious side have ectopic ACTH syndrome, rapid progression of the
effects.172 underlying disease often precludes an early diagnosis.
Cushing disease and adrenal adenomas are four times more
common in women, whereas ectopic ACTH syndrome is
Adrenocortical Diseases more common in men.
Adrenocortical diseases are relatively rare. Their impor-
tance lies in their high rates of morbidity and mortality if
untreated, coupled with the relative ease of diagnosis and
Clinical Features of Cushing Syndrome
the availability of effective therapy. The diseases are most The classic features of Cushing syndrome—centripetal
readily classified on the basis of hormone excess or defi- obesity, moon face, hirsutism, and plethora—have been
ciency (Table 15-8). well known since Cushing’s initial descriptions in 1912
and 1932 (Figs. 15-15 to 15-17). However, this gross clinical
Glucocorticoid Excess picture is not always present, and a high index of suspicion
is required in many cases. Once the normal physiologic
Cushing Syndrome effects of glucocorticoids are appreciated (see Fig. 15-13),
In 1912, Harvey Cushing first described a 23-year-old the clinical features of glucocorticoid excess are easier to
female with obesity, hirsutism, and amenorrhea, and 20 define. They are summarized in Table 15-9 together with
years later he postulated that this “polyglandular syn- the most discriminatory features that will assist in distin-
drome” was due to a primary pituitary abnormality causing guishing Cushing syndrome from simple obesity.176,177
adrenal hyperplasia.8 Adrenal tumors were shown to cause Obesity. Weight gain and obesity are the most common
the syndrome in some cases,173 but ectopic ACTH produc- signs of Cushing syndrome. At least in adults, this weight
tion was not characterized until much later, in 1962.174 The gain is invariably centripetal in nature.144,178 In fact, gener-
term Cushing syndrome is used to describe all causes, whereas alized obesity is more common in the general population
Cushing disease is reserved for pituitary-dependent Cushing
syndrome.
Cushing syndrome comprises the symptoms and signs
associated with prolonged exposure to inappropriately
elevated levels of free plasma glucocorticoids. The use of
the term glucocorticoid in the definition covers excess from
both endogenous (cortisol) and exogenous (e.g., predniso-

TABLE 15-8
Adrenocortical Diseases
Glucocorticoid Excess
Cushing syndrome
Pseudo-Cushing syndromes
Glucocorticoid Resistance

Glucocorticoid Deficiency
Primary hypoadrenalism
Secondary hypoadrenalism
Postchronic corticosteroid replacement therapy
Congenital Adrenal Hyperplasia
Deficiencies of 21-hydroxylase, 3β-HSD, 17α-hydroxylase,
11β-hydroxylase, P450 oxidoreductase, P450 side chain cleavage,
and StAR
Mineralocorticoid Excess

Mineralocorticoid Deficiency
Defects in aldosterone synthesis
Defects in aldosterone action
Hyporeninemic hypoaldosteronism
Adrenal Incidentalomas, Adenomas, and Carcinomas
Figure 15-15 Minnie G., Cushing’s index patient, at age 23 years. (From Cushing
HSD, hydroxysteroid dehydrogenase; StAR, steroidogenic acute regulatory H. The basophil adenomas of the pituitary body and their clinical manifestations
(protein). [pituitary basophilism]. Bull Johns Hopkins Hosp. 1932;50:137-195.)
508 SECTION IV Adrenal Cortex and Endocrine Hypertension

Figure 15-16 Clinical features of Cushing syndrome. A, Centripetal and some generalized obesity and dorsal kyphosis in a 30-year-old woman with Cushing disease. B, Same
patient as in A, showing moon facies, plethora, hirsutism, and enlarged supraclavicular fat pads. C, Facial rounding, hirsutism, and acne in a 14-year-old girl with Cushing
disease. D, Central and generalized obesity and moon facies in a 14-year-old boy with Cushing disease. E and F, Typical centripetal obesity with livid abdominal striae seen in
a 41-year-old woman (E) and a 40-year-old man (F) with Cushing syndrome. G, Striae in a 24-year-old patient with congenital adrenal hyperplasia treated with excessive doses
of dexamethasone as replacement therapy. H, Typical bruising and thin skin of a patient with Cushing syndrome. In this case, the bruising occurred without obvious injury.

than it is in patients with Cushing syndrome. One excep- the psychiatric state. Overall quality of life is significantly
tion is seen in pediatric patients, in whom glucocorticoid reduced in patients with Cushing syndrome, particularly
excess may result in generalized obesity. In addition to affecting physical health and functioning. Quality-of-life
centripetal obesity, patients develop fat depots over the scores improve after treatment but do not return to
thoracocervical spine (buffalo hump), in the supraclavicu- normal.184
lar region, and over the cheeks and temporal regions, Bone. In childhood, the most common presentation is poor
giving rise to the rounded, moon-like facies. The epidural linear growth and weight gain150; as discussed earlier, glu-
space, another site of abnormal fat deposition, may lead to cocorticoids have profound effects on growth and develop-
neurologic deficits. ment.166 Many patients with long-standing Cushing
Reproductive Organs. Gonadal dysfunction is common, with syndrome have lost height because of osteoporotic verte-
menstrual irregularity in females and loss of libido in both bral collapse. This can be assessed by measuring the
sexes. Hirsutism is frequently found in female patients, as patient’s sitting height or comparing the height with arm
is acne. The most common form of hirsutism is vellus span; in normal subjects, height and arm span should be
hypertrichosis on the face; this type should be distin- equal. Pathologic fractures, occurring spontaneously or
guished from the darker, terminal differentiated hirsutism after minor trauma, are not uncommon. Rib fractures, in
that may occur because of ACTH-mediated adrenal andro- contrast to those of the vertebrae, are often painless. The
gen excess. Hypogonadotropic hypogonadism occurs radiographic appearance is typical, with exuberant callus
because of a direct inhibitory effect of cortisol on GnRH formation at the site of the healing fracture. In addition,
pulsatility and LH/FSH secretion, and it is reversible on osteonecrosis of the femoral and humeral heads is a recog-
correction of the hypercortisolism.179,180 nized feature of endogenous Cushing syndrome (see Fig.
Psychiatric Features. Psychiatric abnormalities occur in 15-17). Hypercalciuria may lead to renal calculi, but hyper-
approximately 50% of patients with Cushing syndrome, calcemia is not a feature.
regardless of cause.181,182 Agitated depression and lethargy Skin. Hypercortisolism results in thinning of the skin and
are among the most common problems, but paranoia and separation and exposure of the subcutaneous vascular
overt psychosis are also well recognized. Memory and cog- tissue. On examination, wrinkling of the skin on the
nitive function may also be affected, and increased irrita- dorsum of the hand may be seen, resulting in a “cigarette
bility may be an early feature. Insomnia is common, and paper” appearance (Liddle sign). Minimal trauma may
both rapid eye movement and delta-wave sleep patterns result in bruising, which frequently resembles the appear-
are reduced.183 Lowering of plasma cortisol by medical or ance of senile purpura. The plethoric appearance of the
surgical therapy usually results in a rapid improvement in patient with Cushing syndrome is secondary to the
CHAPTER 15 The Adrenal Cortex 509

Figure 15-17 Bone abnormalities in Cushing disease. A, Aseptic necrosis of the right humeral head in a 43-year-old woman with Cushing disease of about 8 months’ dura-
tion. B, Aseptic necrosis of the right femoral head in a 24-year-old woman with Cushing disease of about 4 12 years’ duration. The arrows indicate the crescent subchondral
radiolucency, best seen in this lateral view. C, Diffuse osteoporosis, vertebral collapse, and subchondral sclerosis in the same patient shown in A. D, Rib fracture in a 38-year-
old man with Cushing disease. (A through C from Phillips KA, Nance EP Jr, Rodriguez RM, et al. Avascular necrosis of bone: a manifestation of Cushing’s disease. South Med
J. 1986;79:825-829.)

thinning of the skin185 combined with loss of facial subcu- Cushing syndrome involves the proximal muscles of the
taneous fat and is not caused by true polycythemia. Acne lower limbs and the shoulder girdle.186 Complaints of
and papular lesions may occur over the face, chest, weakness, such as inability to climb stairs or get up from a
and back. deep chair, are relatively uncommon, but testing for proxi-
The typical, almost pathognomonic, red-purple livid mal myopathy by asking the patient to rise from a crouch-
striae greater than 1 cm in width are most frequently found ing position often reveals the problem.
on the abdomen but may also be present on the upper Cardiovascular Features. Hypertension is another prominent
thighs, breasts, and arms. They are very common in feature, occurring in up to 75% of cases. Even though epi-
younger patients and less so in those older than 50 years demiologic data show a strong association between high
of age. They must be differentiated from the paler, less blood pressure and obesity, hypertension is much more
pigmented striae that occur as a result of pregnancy (striae common in patients with Cushing syndrome than in those
gravidarum) or in association with rapid weight loss. with simple obesity.153 This, together with the established
Skin pigmentation is rare in Cushing disease but metabolic consequences of the disease (diabetes, hyperlip-
common in the ectopic ACTH syndrome. It arises because idemia), is thought to explain the increased cardiovascular
of overstimulation of melanocyte receptors by ACTH and mortality rate in untreated cases.187-189 Cardiovascular
possibly POMC-derived peptides. events are also more common in patients with presumed
Muscle. Myopathy and bruising are two of the most dis- iatrogenic Cushing syndrome resulting from prescribed
criminatory features of the syndrome.176 The myopathy of corticosteroids.171 In addition, thromboembolic events
510 SECTION IV Adrenal Cortex and Endocrine Hypertension

TABLE 15-9 TABLE 15-10


Prevalence of Symptoms and Signs in Cushing Syndrome and Classification of Causes of Cushing Syndrome
Discriminant Index Compared With Prevalence of Features in
Patients With Simple Obesity ACTH-Dependent Causes
Cushing disease (pituitary-dependent)
Findings % of Patients Discriminant Index
Ectopic ACTH syndrome
Symptoms Ectopic CRH syndrome
Weight gain 91 Macronodular adrenal hyperplasia
Menstrual irregularity 84 1.6 Iatrogenic (treatment with 1-24 ACTH)
Hirsutism 81 2.8 ACTH-Independent Causes
Psychiatric dysfunction 62
Backache 43 Adrenal adenoma and carcinoma
Muscle weakness 29 8.0 Primary pigmented nodular adrenal hyperplasia and Carney syndrome.
Fractures 19 McCune-Albright syndrome
Loss of scalp hair 13 Aberrant receptor expression (gastric inhibitory polypeptide,
interleukin-1β)
Signs Iatrogenic (e.g., pharmacologic doses of prednisolone, dexamethasone)
Obesity 97 Other Causes of Hypercortisolism
Truncal 46 1.6
Generalized 55 0.8 Alcoholism
Plethora 94 3.0 Depression
Moon facies 88 Obesity
Hypertension 74 4.4 Pregnancy
Bruising 62 10.3
ACTH, adrenocorticotropic hormone; CRH, corticotropin-releasing hormone.
Red-purple striae 56 2.5
Muscle weakness 56
Ankle edema 50
Pigmentation 4
This can be diagnosed by documenting an increase in the
Other Findings
ratio of urinary cortisol to cortisone metabolites. In addi-
Hypertension 74 tion, hepatic 5α-reductase activity is inhibited, resulting in
Diabetes 50 a greater excretion of 5α-cortisol metabolites.195
Overt 13
The functions of the pituitary-thyroid axis and the
Impaired glucose tolerance test 37
Osteoporosis 50 pituitary-gonadal axis are suppressed in patients with
Renal calculi 15 Cushing syndrome because of a direct effect of cortisol on
TSH and gonadotropin secretion.196,197 Cortisol causes a
Data from Ross EJ, Linch DC. Cushing’s syndrome-killing disease: reversible form of hypogonadotropic hypogonadism but
discriminatory value of signs and symptoms aiding early diagnosis. Lancet.
1982;2:646-649.
also directly inhibits Leydig cell function. GH secretion is
reduced, possibly mediated through an increase in somato-
statinergic tone.
Eye. Ocular effects include raised intraocular pressure198 and
may be more common in Cushing patients, but this devel- exophthalmos199 (in up to one third of patients in Cush-
opment appears to be limited to those with ACTH- ing’s original series), the latter occurring because of
dependent disease.190 increased retro-orbital fat deposition. Cataracts, a well-
Infections. Infections are more common in patients with recognized complication of corticosteroid therapy, seem to
Cushing syndrome.191,192 In many instances, infections are be uncommon,200 except as a complication of diabetes. In
asymptomatic and occur because the normal inflammatory our experience, chemosis is a sensitive and underreported
response is suppressed. Reactivation of tuberculosis has feature of Cushing syndrome.
been reported193 and has even been the presenting feature
in some cases. Fungal infections of the skin (notably tinea
versicolor) and nails may occur, as may opportunistic CLASSIFICATION AND
fungal infections. Bowel perforation is more common in
patients with extreme hypercortisolism, and the hypercor- PATHOPHYSIOLOGY OF
tisolism may mask the usual symptoms and signs of the CUSHING SYNDROME
condition. Wound infections are more common and con-
tribute to poor wound healing. Cushing syndrome is most readily classified into ACTH-
Metabolic and Endocrine Features. Glucose intolerance dependent and ACTH-independent causes (Table 15-10).
occurs, and overt diabetes mellitus is present in up to one
third of patients in some series. Hepatic lipoprotein syn- ACTH-Dependent Causes
thesis is stimulated, and increases in circulating cholesterol
and triglycerides may be found.194 Hypokalemic alkalosis is Cushing Disease
found in 10% to 15% of patients with Cushing disease but When iatrogenic causes are excluded, the most common
in more than 95% of patients with ectopic ACTH syn- cause of Cushing syndrome is Cushing disease, accounting
drome. Several factors may contribute to this mineralocor- for approximately 70% of cases. The adrenal glands in
ticoid excess state, including corticosterone and DOC these patients show bilateral adrenocortical hyperplasia
excess, but the principal culprit is thought to be cortisol with widening of the ZF and ZR.177
itself. Depending on the prevailing cortisol production Cushing himself raised the question as to whether this
rate, cortisol swamps HSD11B2 in the kidney and acts as a disease was a primary pituitary condition or secondary
mineralocorticoid. Hypokalemic alkalosis is more common to an abnormality in the hypothalamus, and there has
in ectopic ACTH syndrome because cortisol production been an ongoing debate on this issue ever since.201 The
rates are higher than in patients with Cushing disease.131 hypothalamic theory states that ACTH-secreting adenomas
CHAPTER 15 The Adrenal Cortex 511

TABLE 15-11
Etiology of Cushing Disease: Hypothalamic Theory Versus Pituitary Theory
Hypothalamic Theory Pituitary Theory
Neuroendocrine abnormalities202,203 Lack of cure after pituitary stalk section
Loss of circadian rhythm, sleep disturbance, other Circulating and CSF CRH levels are suppressed204
hypothalamic defects (TSH, LH/FSH secretion) Reversal of hypothalamic defects on correction of hypercortisolism
Efficacy of centrally acting drugs205,206 (bromocriptine, High surgical cure rate (recurrences resulting from regrowth of inadequately resected
cyproheptadine, sodium valproate) tumor rather than real recurrence)207,208
Recurrences after pituitary surgery Secondary hypoadrenalism after successful pituitary surgery (may be prolonged and
Ectopic CRH-secreting tumors cause Cushing associated with reduced ACTH expression in surrounding adjacent normal
disease,210 but pathologic examination shows corticotrophs)209
basophil hyperplasia, not adenomas Pituitary ACTH-secreting adenoma in almost 90% of cases are monoclonal in origin211,212

Note: Superscript numbers indicate references listed at the end of the chapter.
ACTH, adrenocorticotropic hormone; CRH, corticotropin-releasing hormone; CSF, cerebrospinal fluid; FSH, follicle-stimulating hormone; LH, luteinizing hormone;
TSH, thyroid-stimulating hormone.

arise because of dysfunctional regulation of corticotrophs TABLE 15-12


through chronic stimulation by CRH (or AVP). Other stud- Tumors Associated With the Ectopic Adrenocorticotropic
ies provide data to support a primary pituitary defect as the Hormone Syndrome
cause of the condition (Table 15-11).
The hypothalamus may have an initiating role, but the Tumor Type Approximate Incidence (%)
overwhelming evidence is that, at presentation, the condi- Small cell lung carcinoma 50
tion is pituitary-dependent. In 85% to 90% of cases, the Non–small cell lung carcinoma 5
disease is caused by a pituitary adenoma of monoclonal Pancreatic tumors (including carcinoids) 10
origin213,214; basophil hyperplasia alone is found in 9% to Thymic tumors (including carcinoids) 5
33% of pathologic series.201 The majority of tumors are Lung carcinoids 10
Other carcinoids 2
small microadenomas (<1 cm), but larger macroadenomas
Medullary carcinoma of thyroid 5
occur in up to 10% of cases and usually signify a more Pheochromocytoma and related tumors 3
invasive tumor.215 Selective surgical removal of a causative Rare carcinomas of prostate, breast, 10
corticotroph adenoma results in remission, but on very ovary, gallbladder, colon
long-term follow-up, relapse may occur in up to 20% to
30% of micro- and macroadenoma patients.215a However, it
is possible, particularly in cases with no identifiable pitu-
itary adenoma, that Cushing disease is heterogeneous with indolent tumors, such as benign bronchial carcinoids, that
different subtypes. produce ACTH.218,219 In these cases, symptoms and signs
A key biochemical hallmark of the disease is a relative are commonly present for 18 months before clinical pre-
resistance of ACTH secretion to normal glucocorticoid sentation. Such patients present with the typical features
feedback inhibition.216 ACTH-secreting pituitary adenomas of Cushing syndrome and may be biochemically similar to
function at a higher than normal set-point for cortisol patients with Cushing disease. Therefore, once a diagnosis
feedback. The predominant finding in Cushing disease is of Cushing syndrome is established, the principal diagnos-
an increase in ACTH pulse amplitude with loss of normal tic dilemma is in the distinction of pituitary-dependent
circadian rhythm, but ACTH pulse frequency is also Cushing disease from these indolent causes of ectopic
increased in some cases (see Fig. 15-8).217 ACTH syndrome.
Tumors most commonly associated with ectopic ACTH
syndrome arise from neuroendocrine tissues, the cells of
Ectopic ACTH Syndrome which possess the ability to uptake and decarboxylate
In 15% of cases, Cushing syndrome is associated with non- amine precursors (APUD cells). Although POMC transcripts
pituitary tumors secreting ACTH—the ectopic ACTH syn- of 1200-1450nt are frequently found in small cell lung
drome.218 On clinical grounds, these tumors can be divided cancer, only 0.5% to 1% of tumors are associated with
into two entities: highly malignant tumors such as small ectopic ACTH syndrome, and the explanation for the de-
cell carcinoma of bronchus (Table 15-12) and highly pro- velopment of ectopic ACTH secretion remains unclear. In
liferative neuroendocrine carcinoma (e.g., pancreas), and contrast, POMC mRNA transcripts of the short 800nt
more indolent tumors occurring in patients with underly- length may be found in tumors not associated with ectopic
ing neuroendocrine tumors such as bronchial carcinoids. ACTH syndrome. In addition to aberrant transcriptional
In the former group, the clinical presentation is often that regulation of the POMC, interaction with tissue-specific
of a wasting syndrome with weakness and pigmentation. transcription factors or the promoter methylation status of
Circulating ACTH concentrations and cortisol secretion POMC may be involved. Once translated, POMC is cleaved
rates can be extremely high. As a result, duration of symp- in the pituitary by specific serine endoproteases to produce
toms from onset to presentation is short (<3 months); ACTH precursors; in ectopic ACTH syndrome, aberrant pe-
patients are commonly pigmented, and the metabolic ripheral processing of POMC may lead to increased con-
manifestations of glucocorticoid excess are often rapid and centrations of circulating ACTH precursors (pro-ACTH,
progressive. Weight loss, myopathy, and glucose intoler- N-POC) (see Fig. 15-6). In contrast to ACTH secretion from
ance are prominent symptoms and signs. The association pituitary adenomas, ectopic POMC/ACTH production is
of these features with hypokalemic alkalosis and peripheral not responsive to normal glucocorticoid feedback220 be-
edema should alert the clinician to the diagnosis. cause of the defective GR or GR-signaling mechanism.221
Depending on local referral practice, approximately However, this sensitivity to glucocorticoid feedback is far
20% of cases of ectopic ACTH syndrome are explained by from clearcut, which is one reason why the differential
512 SECTION IV Adrenal Cortex and Endocrine Hypertension

diagnosis of ACTH-dependent Cushing syndrome can be hirsutism is uncommon. Subclinical Cushing syndrome
challenging.218 has been reported in up to 10% of patients with adrenal
incidentalomas (see later discussion).
Ectopic Corticotropin-Releasing Hormone Syndrome
Primary Pigmented Nodular Adrenal Hyperplasia and
Ectopic production of CRH is a very rare cause of pituitary-
dependent Cushing disease. A number of cases have now
Carney Syndrome
been described in which a tumor (usually bronchial carci- About 100 cases of ACTH-independent Cushing syndrome
noid, medullary thyroid, or prostate carcinoma) has been have been reported in association with bilateral, small,
shown to secrete CRH alone or in combination with pigmented adrenal nodules. Pathologically, these nodules
ACTH.222-224 When available, pituitary histologic examina- are usually 2 to 4 mm in diameter (although they can be
tion has revealed corticotroph hyperplasia but not adenoma larger) and black or brown on cut section. Adjacent adrenal
formation. Biochemically, these patients, like those with tissue is atrophic, distinguishing this primary pigmented
ectopic ACTH syndrome, lose the normal negative gluco- nodular adrenal hyperplasia (PPNAD) from MAH. Presenta-
corticoid feedback mechanism—50% have resistance to tion is with typical features of Cushing syndrome in
high-dose dexamethasone therapy. Ectopic CRH produc- persons younger than 30 years of age and, in 50% of cases,
tion may explain the suppression of cortisol secretion after in persons younger than 15 years of age.234 Cases of PPNAD
high-dose dexamethasone that is observed in some patients have been reported without Cushing syndrome. Bilateral
with the ectopic ACTH syndrome. adrenalectomy is curative.
A familial autosomal dominant variant, called Carney
complex (Table 15-13), comprises mesenchymal tumors
Macronodular Adrenal Hyperplasia (especially atrial myxomas), spotty skin pigmentation,
In 10% to 40% of patients with Cushing disease, there is peripheral nerve tumors, and various other tumors includ-
bilateral adrenocortical hyperplasia associated with one or ing breast lesions, testicular tumors, and GH-secreting pitu-
more nodules, which may be up to several centimeters in itary tumors.235 Mutations of the gene encoding the PKA
diameter.225-228 Patients tend to be older and to have had regulatory subunit type IA (PRKAR1A) lead to abnormal
symptoms for a longer time, but they otherwise present PKA signaling and explain the phenotype in some cases.236
with the classic clinical features of Cushing syndrome. Other cases have been mapped to chromosome 2p16, but
Pathologically, the nodules are lobulated and can be mark- the underlying genetic mutation is unknown.
edly enlarged, but internodular hyperplasia is invariably
found. Macronodular adrenal hyperplasia (MAH) is thought
to result from long-standing adrenal ACTH stimulation,
McCune-Albright Syndrome
which leads to autonomous adrenal adenoma formation. In McCune-Albright syndrome, fibrous dysplasia and cuta-
Therefore, as the adrenals in a patient with Cushing disease neous pigmentation may be associated with pituitary,
become more hyperplastic, they secrete more cortisol for a thyroid, adrenal, and gonadal hyperfunction. The most
given ACTH level, which ultimately can lead to autosup- common manifestation is with sexual precocity and GH
pression. Individual clinical cases support this hypothesis, excess, but Cushing syndrome has been reported.237 The
and MAH should be regarded as an ACTH-dependent form underlying abnormality is a somatic mutation in the
of Cushing syndrome, even though ACTH levels may be α-subunit of the stimulatory G protein, which is linked to
relatively low and dexamethasone suppressibility less adenyl cyclase. The mutation results in constitutive activa-
marked than in other cases of Cushing disease.229 These tion of the G protein, mimicking constant ACTH stimula-
features can be a trap for the unwary because they may be tion at the level of the adrenal. ACTH levels are suppressed,
mistaken for primary adrenal tumors, especially as up to and adrenal adenomas may occur.
30% of patients with Cushing disease have asymmetric
adrenal hyperplasia.
Macronodular Hyperplasia
ACTH-Independent Causes Although MAH commonly occurs in patients with ACTH-
dependent Cushing syndrome, truly ACTH-independent
Cortisol-Secreting Adrenal Adenoma and Carcinoma
Excluding iatrogenic cases, adrenal adenomas are respon-
sible for about 10% to 15% of Cushing syndrome cases, TABLE 15-13
and carcinomas for less than 5%. By contrast, 65% of Clinical Features of Carney Complex
cases of Cushing syndrome in children have an adrenal
cause (15% adenomas, 50% carcinomas).227-229 Onset of Feature Prevalence (%)
clinical features is gradual in patients with adenomas, but Skin lesions 80
it is often rapid in adrenal carcinoma. Mutations of Pigmented lesions
PRKACA, which encodes the catalytic subunit of cAMP- Blue nevi
dependent protein kinase A (PKA), at the hotspot L205R Cutaneous myxomas
Cardiac myxomas 72
have been shown by several independent groups to be the Pigmented nodular adrenal hyperplasia 45
cause of approximately 50% of adrenal adenomas causing Breast lesions
Cushing syndrome.230-233 Bilateral fibroadenomas 45 (females only)
In addition to the features of hypercortisolism, patients Testicular tumors 56 (males only)
may complain of loin or abdominal pain, and a tumor may Pituitary lesions, usually growth hormone 10
be palpable. Adrenal carcinoma may secrete other steroids, secreting
such as androgens or mineralocorticoids, but this is very Neural lesions (gastric schwannomas) <5
unusual in adenomas. Therefore, in females, there may be Miscellaneous
Thyroid cancers Rare
features of virilization, with hirsutism, clitoromegaly,
Acoustic neuromas Rare
breast atrophy, deepening of the voice, temporal recession, Hepatomas Rare
and severe acne. In pure cortisol-secreting adenomas,
CHAPTER 15 The Adrenal Cortex 513

macronodular hyperplasia (AIMAH) is also recognized as a


distinct entity.238 The nodules are nonpigmented and 600
greater than 5 mm in diameter; occasionally, the adrenals 500
are massively enlarged. Most cases are explained on the
basis of aberrant receptor expression within the adrenal 400
cortex.239 Food-induced hypercortisolism due to enhanced 300
adrenal responsiveness to gastric inhibitory polypeptide
(GIP) was the first cause of AIMAH described, due to expres- 200
sion of GIP receptors within the adrenal cortex, but aber-
100
rant expression of the vasopressin V1, β-adrenergic, LH,
serotonin, and angiotensin (AT1) receptors have also been 0
linked to AIMAH. Protocols have been suggested for the
further investigation of AIMAH.239
Familial cases suggest a genetic cause for this condition
in some patients, and inactivating mutations Armadillo

Urinary cortisol/creatinine ratio


120
repeat 5 (ARMC5) have been demonstrated as a cause.240
100
Iatrogenic Cushing Syndrome 80
A careful drug history is required to exclude iatrogenic 60
Cushing syndrome. Development of the features of Cushing
syndrome depends on the dose, duration, and potency of 40
the corticosteroids used in clinical practice. ACTH is rarely 20
prescribed, but long-term administration will also result in
cushingoid features. Some features, such as an increase in 0
intraocular pressure, cataracts, benign intracranial hyper-
tension, aseptic necrosis of the femoral head, osteoporosis,
and pancreatitis, are more common in iatrogenic than 600
endogenous Cushing syndrome, whereas other features,
notably hypertension, hirsutism, and oligomenorrhea/ 500
amenorrhea, are less prevalent.
400

Special Features of Cushing Syndrome 300

Cyclic Cushing Syndrome 200


Of particular clinical interest has been a group of patients 100
with cyclic Cushing syndrome, characterized by periods of
excess cortisol production interspersed with intervals of 0
normal cortisol production (Fig. 15-18). Some of these
patients demonstrate a paradoxical rise in plasma ACTH 0 20 40 60 80 100
and cortisol when treated with dexamethasone, and occa- Day of collection
sionally a patient is benefited by dopamine agonist (bro-
Figure 15-18 Patterns of cortisol secretion in three patients with cyclic Cushing
mocriptine or cabergoline) therapy. Most patients have syndrome. In each case, the ratio of early morning urinary cortisol (in nanomoles per
been thought to have pituitary-dependent disease, and in liter) to creatinine (in millimoles per liter) are plotted against time. Variable periodicity
many of these patients, basophil adenomas have been in cortisol hypersecretion is shown. (From Atkinson AB, McCance DR, Kennedy L, et al.
removed, with long-term cure in some cases. However, Cyclical Cushing’s syndrome first diagnosed after pituitary surgery: a trap for the
cortisol secretion may show some evidence of cyclicity in unwary. Clin Endocrinol. 1992;36:297-299.)
patients with an ectopic source of ACTH and in PPNAD.241,242

Cushing Syndrome in Children


Cushing syndrome can occur at any age, but the causes puberty, glucocorticoid-mediated suppression of gonado-
differ across age groups (Fig. 15-19). In children, adrenal tropins may occur.244
causes account for 65% of all cases, and in addition to the Glucocorticoid excess influences not only the hypo-
previously mentioned features, growth arrest is almost thalamic-pituitary-gonadal axis but also the GH/IGF-1 axis,
invariable.243 The dissociation between height and weight leading to both reduction of spontaneous GH secretion
is obvious, with the height most commonly below the and pharmacologic GH response. Furthermore, direct
mean, whereas the BMI is almost always above the mean. effects of glucocorticoids on epiphyseal chrondocytes,
If the height and weight are increasing along the same probably together with disturbance of microvasculariza-
percentile line, then the diagnosis of Cushing syndrome is tion of the growth plate, result in a negative effect on
highly unlikely. Obesity in childhood Cushing syndrome growth. Poor catch-up has been reported in children after
tends to be generalized. Most patients have a delayed bone cure of Cushing disease, and evidence exists for GH inhibi-
age that is negatively correlated with height standard devi- tion by hypercortisolemia for 1 to 2 years after cure of
ation score (SDS), duration of symptoms, and age at diag- Cushing disease. GH secretion should be assessed 3 months
nosis. The observed growth failure often precedes other after treatment. If GH deficiency is demonstrated, GH in a
manifestations such as weight gain, pubertal arrest, fatigue, replacement dose of 25 μg/kg per day should be given.
depression, hypertension, and acne. Pubertal development Catch-up growth is observed in most patients and target
can be advanced in patients with virilizing tumors causing adult height is achieved. However, many patients remain
precocious pseudopuberty. However, in patients with true obese.
514 SECTION IV Adrenal Cortex and Endocrine Hypertension

a two-hit hypothesis has been put forward. Chronic liver


Cushing disease (n = 182)
14.1 yr
disease of any cause is associated with impaired cortisol
metabolism, but in alcoholic patients there is an increased
Nodular adrenal hyperplasia (n = 25) cortisol secretion rate, rather than concomitant suppres-
13.0 yr sion in the face of impaired metabolism.248 In some studies,
alcohol has directly stimulated cortisol secretion; alterna-
Ectopic ACTH syndrome (n = 11) tively, AVP levels are elevated in patients with decompen-
10.1 yr sated liver disease and may stimulate the HPA axis. With
abstinence from alcohol, the biochemical abnormalities
Adrenocortical tumors (n = 164) revert to normal within days.
4.5 yr

Adrenal hyperplasia secondary to Depression


McCune-Albright syndrome (n = 16)
1.2 yr Although the cause is unknown, it is recognized that
patients with depression may exhibit the hormonal abnor-
malities of patients with Cushing syndrome.249 These
0 5 10 15 20 30 abnormalities are reversible on correction of the psychiat-
Age (yrs) ric condition. Conversely, patients with Cushing syndrome
Figure 15-19 Etiology and age-dependency of pediatric Cushing syndrome. Note are frequently depressed, and a careful clinical and endo-
how the causes differ across age groups. (From Storr HL, Chan LF, Grossman AB, et al. crinologic assessment is required.
Paediatric Cushing’s syndrome: epidemiology, investigation and therapeutic advances.
Trends Endocrinol Metab. 2007;18:167-174.)
Obesity
Although one of the most common referrals to a clinical
endocrinologist is for exclusion of an underlying endocrine
Pregnancy
cause in a patient with obesity, the diagnosis of Cushing
Pregnancy is rare in women with Cushing syndrome syndrome in such patients should not cause difficulties.
because of associated amenorrhea due to androgen excess Patients with obesity have mildly increased cortisol secre-
or hypercortisolism. However, pregnancy has been reported tion rates, and the data suggest that this is due to activation
in approximately 100 such women, 50% of whom had of the HPA axis.250,251 However, circulating cortisol concen-
adrenal adenomas.245 A few cases of true pregnancy- trations are invariably normal, and urinary free cortisol
induced Cushing syndrome have been described, with concentrations are either normal or only slightly elevated.
regression after delivery.246 In these cases, the cause is The stimulus for the increased secretion of cortisol appears
unknown. Establishing a diagnosis and a cause can be dif- to be increased peripheral metabolism and clearance of
ficult. Clinically, striae, hypertension, and gestational dia- cortisol—principally, reduced hepatic conversion of corti-
betes are common features in normal pregnancies, yet sone to cortisol by HSD11B1 and increased conversion of
hypertension and diabetes are also the most common cortisol to 5α-reduced derivatives.251
signs of Cushing syndrome in a pregnant woman (70%
and 30% of all cases, respectively). Furthermore, biochemi- Investigation of Patients With Suspected
cally normal pregnancy is associated with a threefold
increase in plasma cortisol due to increased production of
Cushing Syndrome
cortisol and CBG. Urinary free cortisol also rises, and dexa- There are two stages in the investigation of suspected
methasone does not suppress plasma cortisol to the same Cushing syndrome: (1) Does this patient have Cushing
degree as the nonpregnant state. Left untreated, the condi- syndrome? and (2) If the answer is “yes,” what is the cause?
tion is associated with high rates of maternal and fetal Unfortunately, many investigators fail to make this distinc-
morbidity and mortality. Any adrenal or pituitary adeno- tion and ill-advisedly use tests that are relevant to the
mas should be excised. Metyrapone, which is not terato- second question in trying to answer the first question. In
genic, has been effective in many cases in controlling the particular, it is essential that radiologic investigations not
hypercortisolism. be undertaken until Cushing syndrome has been con-
firmed biochemically. The starting point should be to
investigate patients in whom there is a high clinical index
Other Syndromes of Hypercortisolemia of suspicion for the diagnosis of Cushing syndrome, focus-
Other states occur when there are clinical and biochemi- ing on the features that are most discriminating for the
cal features of Cushing syndrome, but when hypercorti- condition (see Table 15-9). Widespread indiscriminate bio-
solemia is secondary to other factors it is often referred to chemical screening in obese, hypertensive, and diabetic
as pseudo-Cushing syndrome. Resolution of the underlying populations is not recommended. Very few laboratories
cause results in disappearance of the cushingoid state. have developed methods for the measurement of free
Several causes are described. serum cortisol.252 Because more than 90% of serum cortisol
is protein bound, the results of the conventional assay are
affected by drugs and by conditions that alter CBG levels.
Alcohol Estrogen therapy or pregnancy may elevate CBG and total
In the original description of alcohol-related pseudo- serum cortisol, and estrogens should be stopped for 6
Cushing syndrome, urinary and plasma cortisol levels were weeks prior to assessment by tests using serum cortisol. The
elevated and failed to suppress with dexamethasone. major tests are listed in Table 15-14.177,249,253,254
Plasma ACTH has been found to be normal or suppressed.
The condition is rare but should be suspected in a patient
with an ongoing history of heavy alcohol intake and bio- Question 1: Does This Patient Have Cushing Syndrome?
chemical or clinical evidence of chronic liver disease.247 Circadian Rhythm of Plasma Cortisol. In normal subjects,
The pathogenesis of this condition remains unknown, but plasma cortisol levels are at their highest early in the
CHAPTER 15 The Adrenal Cortex 515

TABLE 15-14 and dexamethasone suppression testing. Patients should


Tests Used in the Diagnosis and Differential Diagnosis of
make two or three complete consecutive collections to
Cushing Syndrome
account for patient error in collecting samples and for
episodic cortisol secretion, notably from adrenal adeno-
Diagnosis—Does the Patient Have Cushing Syndrome? mas. Simultaneous creatinine excretion (which differs by
Late night salivary cortisol/circadian rhythm of plasma cortisol no more than 10% from day to day) may be used to ensure
Urinary free cortisol excretion* adequacy of collection. Urinary free cortisol is a useful
Low-dose dexamethasone suppression test* screening test, although it is accepted that the value can
Differential Diagnosis—What Is the Cause of the Cushing Syndrome? be normal in up to 8% to 15% of patients with Cushing
syndrome.253,254,259 Conversely, moderately elevated results
Plasma ACTH should always be verified by further testing before a diag-
Plasma potassium, bicarbonate
High-dose dexamethasone suppression test
nosis of Cushing syndrome is made. In addition, variations
Corticotropin-releasing hormone in levels of up to 50% have been observed in patients with
Inferior petrosal sinus sampling proven Cushing disease, reinforcing the need for multiple
CT, MRI scanning of pituitary, adrenals collections.260
Scintigraphy Measurement of the cortisol-to-creatinine ratio in the
Tumor markers first urine specimen passed on waking obviates the need
for a timed collection and has been used as a screening test,
*Valuable outpatient screening tests (see text discussion).
ACTH, adrenocorticotropic hormone; CT, computed tomography; MRI, particularly when cyclic Cushing syndrome is suspected.261
magnetic resonance imaging. Urine aliquots may be sent to the local endocrinology
laboratory, with cortisol-to-creatinine ratios greater than
25 nmol/mmol on repeated measurement being indicative
of hypercortisolism.
morning and reach a nadir (<50 nmol/L [<2 μg/dL] in a Low-Dose Overnight Dexamethasone Suppression Tests. In
nonstressed subject) at about midnight.255 This circadian normal subjects, the administration of a supraphysiologic
rhythm is lost in patients with Cushing syndrome; in the dose of glucocorticoid results in suppression of ACTH and
majority, the 9 AM plasma cortisol is normal but nocturnal cortisol secretion. In Cushing syndrome of whatever cause,
levels are raised. Random morning plasma cortisol levels there is a failure of this suppression when low doses of the
are therefore of little value in making the diagnosis, whereas synthetic glucocorticoid dexamethasone are given.216
a midnight cortisol level greater than 200 nmol/L (>7.5 μg/ The overnight test is a useful outpatient screening
dL) indicates Cushing syndrome. However, various factors test,249,253,262 in which 1 mg of dexamethasone is given at
such as stress of venipuncture, intercurrent illness, and 11 PM. A normal response is a plasma cortisol level of less
admission to hospital may lead to false-positive results. than 50 nmol/L (<1.8 μg/dL) between 8 and 9 AM the fol-
Conversely, if a serum cortisol value is less than 50 nmol/L lowing morning. The outpatient overnight test has high
at midnight, Cushing syndrome is excluded at that time. sensitivity (95%) but lower specificity, and further investi-
Ideally, patients should be hospitalized for 24 to 48 hours gation is often required.263,264
before the midnight cortisol level is measured, but some In the 48-hour low-dose dexamethasone test, plasma
centers have reported discriminant results for midnight cortisol is measured at 9 AM on day 0 and again 48 hours
levels measured in outpatients. Nevertheless, this is a cum- later, after administration of dexamethasone 0.5 mg every
bersome test and has been largely supplanted by measure- 6 hours for 48 hours. Using a postdexamethasone plasma
ment of salivary cortisol (see next). cortisol concentration of less than 50 nmol/L (<1.8 μg/dL)
Salivary Cortisol. CBG is absent from saliva, and the use of as the cutoff point, this test is reported to have a 97% to
salivary cortisol measurements offers a sensible alternative 100% true-positive rate and a false-positive rate of less than
in that the test does not require hospitalization. The diag- 1%.249,263
nostic accuracy of a single midnight salivary cortisol level Certain drugs (e.g., phenytoin, rifampicin) may increase
has been established in several studies. The cutoff points the metabolic clearance rate of dexamethasone, leading to
that define disease vary depending on the assay used. false-positive results. Simultaneous measurement of plasma
In one study, a cortisol value greater than 2.0 ng/mL dexamethasone may be useful in such cases and will also
(5.5 nmol/L) had a 100% sensitivity and a 96% specificity detect whether patients failed to take the drug, but the
for diagnosis of Cushing syndrome.253,256,257 It is important values of plasma dexamethasone that predict suppression
to note, however, that late-night salivary cortisol tends to of serum cortisol are not fully defined.264
increase with age and cardiovascular comorbid conditions Other Causes of Hypercortisolemia: Pseudo-Cushing or True
such as hypertension and diabetes, and thus the discrimi- Cushing Syndrome? In patients with depression, urinary free
nating power diminishes in the elderly population.258 cortisol concentrations may be elevated and may overlap
Urinary Free Cortisol Excretion. For many years, the diagnosis with those seen in patients with true Cushing syndrome.
of Cushing syndrome was based on the measurement Compared with patients with Cushing disease, depressed
of urinary metabolites of cortisol (24-hour urinary 17- patients have greater suppressibility after dexamethasone
hydroxycorticosteroid or 17-oxogenic steroid excretion, and reduced response to CRH, but neither test is diagnos-
depending on the method used). However, the sensitivity tic.249,265 The dexamethasone-suppressed CRH test has been
and specificity of these methods are poor, and most centers proposed as a tool to discriminate between true Cushing
have replaced these assays with the more sensitive mea- syndrome and other states but has been shown to have no
surement of urinary free cortisol. This is an integrated advantage over the standard low-dose dexamethasone sup-
measure of plasma free cortisol: as cortisol secretion pression test. In normal subjects and in patients with
increases, the binding capacity of CBG is exceeded, result- endogenous depression, insulin-induced hypoglycemia
ing in a disproportionate rise in urinary free cortisol. results in a rise in ACTH and cortisol levels, a response that
Normal values depend on the assay used and tend to be usually is not seen in patients with Cushing syndrome, but
lower when analyzed by liquid chromatography and this test has largely been abandoned for this purpose.249
tandem mass spectrometry. Although in widespread use, Diagnostic Guidelines. The Endocrine Society, in collabora-
urinary free cortisol is less sensitive that salivary cortisol tion with the European Society for Endocrinology, has
516 SECTION IV Adrenal Cortex and Endocrine Hypertension

Cushing syndrome suspected Question 2: What Is the Cause of Cushing Syndrome


(consider endocrinologist consultation) in This Patient?
Having confirmed Cushing syndrome clinically and bio-
chemically, the clinician’s next step is to determine the
Exclude exogenous glucocorticoid exposure cause (Fig. 15-21).
Morning Plasma ACTH. Ideally, ACTH should be measured
with the use of a modern, two-site immunoradiometric
Perform one of the following tests assay. Such a test differentiates ACTH-dependent from
ACTH-independent causes. The samples should be taken
in ice cold tubes and immediately separated ahead of
storage at −40° C ahead of analysis to prevent inadvertent
24-h UFC Overnight Late night salivary degradation. In Cushing disease, 50% of patients have a 9
(≥2 tests) 1-mg DST cortisol (≥2 tests) AM ACTH level within the normal reference range (2 to
11 pmol/L [9 to 52 pg/mL]); in the remainder, it is mod-
Consider caveats for each test (see text)
Use 48-h, 2-mg DST in certain populations (see text)
estly elevated. Occasionally, due to episodic secretion,
levels may be very low, and thus measurement of at least
two values is recommended to avoid misclassification of
mild Cushing disease as ACTH-independent. ACTH levels
in the ectopic ACTH syndrome are high (usually >20 pmol/L
ANY ABNORMAL Normal [>90 pg/mL]); nevertheless, overlap values are seen in
RESULT (CS unlikely) Cushing disease in 30% of cases.267 Therefore, this test
cannot be used to differentiate the two conditions (Fig.
15-22). The measurement of ACTH precursors (pro-ACTH,
Exclude physiologic causes of hypercortisolism POMC) has been suggested, but it is not routinely available
and has not been proved to detect an ectopic source
of ACTH.
Consult endocrinologist In patients with adrenal tumors, plasma ACTH is invari-
ably undetectable (<1 pmol/L). The presence of plasma
ACTH levels that are low-normal or intermittently detect-
Perform 1 or 2 other studies shown above able, which may occur in MAH, is problematic. The danger
Suggest repeating the abnormal study is that in some patients the asymmetry of the nodular
Suggest Dex-CRH or midnight serum cortisol in hyperplasia may lead to a diagnosis of adrenal adenoma,
certain populations (see text) the plasma ACTH is ignored, and an inappropriate adrenal-
ectomy is performed. Conversely, in some patients with
this syndrome, an autonomous adrenal tumor develops,
and unilateral adrenalectomy is required despite the detect-
able ACTH.
Discrepant Plasma Potassium. Hypokalemic alkalosis is present in more
Normal
(Suggest additional ABNORMAL
(CS unlikely) than 95% of patients with the ectopic ACTH syndrome but
evaluation)
in fewer than 10% of those with Cushing disease. The cause
of this mineralocorticoid excess state is now established.
Patients with the ectopic syndrome usually have higher
Cushing syndrome
rates of cortisol secretion. Cortisol saturates the renal-
protective HSD11B2 enzyme, resulting in cortisol-induced
Figure 15-20 Algorithm for testing patients with suspected Cushing syndrome (CS)
mineralocorticoid hypertension (see Chapter 16).131 In
according to the 2008 Endocrine Society clinical practice guideline. All statements
are recommendations except for those prefaced by the word Suggest. Diagnostic addition, these patients have higher levels of the ACTH-
criteria that point to Cushing syndrome are a urinary free cortisol (UFC) value greater dependent mineralocorticoid, DOC.
than the normal range for the assay, a serum cortisol level greater than 1.8 μg/dL High-Dose Dexamethasone Suppression Test. The rationale for
(>50 nmol/L) after administration of 1 mg dexamethasone (1-mg DST), and a late- the high-dose dexamethasone suppression test is that in
night salivary cortisol concentration greater than 145 ng/dL (>4 nmol/L). CRH, Cushing disease the negative feedback control of ACTH is
corticotropin-releasing hormone; Dex, dexamethasone; DST, dexamethasone suppres- reset to a higher level than normal. Therefore, cortisol
sion test. (From Nieman LK, Biller BM, Findling JW, et al. The diagnosis of Cushing’s levels do not suppress with low-dose dexamethasone but
syndrome: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. do so after high doses. The original test introduced by
2008;93:1526-1540.)
Liddle was based on giving 2 mg dexamethasone every 6
hours for 48 hours and demonstrating a fall of greater than
50% in urinary 17-hydroxycorticosteroids.216 In the modern
test, the plasma or urinary free cortisol (or both) is mea-
issued evidence-based guidelines for the diagnosis of sured at 0 and +48 hours, and a greater than 50% suppres-
Cushing syndrome.266 Recommendations are to proceed sion of plasma cortisol from the basal value has been used
initially with one of four highly sensitive screening to define a positive response. In all cases, the response is
tests: urinary free cortisol, late-night salivary cortisol, long graded and is dependent on the original cortisol secretion
overnight dexamethasone, or the 2-mg/48-hour dexameth- rate: greater suppression is often observed in patients
asone screening test. Abnormality detected by of any of with lower basal cortisol values. In women with ACTH-
these tests in a patient with clinically suspected Cushing dependent Cushing syndrome, in whom the a priori likeli-
syndrome should be confirmed with one of the additional hood of Cushing disease is 90%, the sensitivity of this test
tests; if both test results are abnormal, patients should then for the Cushing disease is 80%, lower than the pretest prob-
undergo testing for the cause of the Cushing syndrome ability. Because of this there is little logic to the continued
(Fig. 15-20). use of this test when inferior petrosal sinus sampling (IPSS)
CHAPTER 15 The Adrenal Cortex 517

Cushing syndrome
clinically/biochemically confirmed

Differential diagnosis

ACTH

Suppressed Detectable

CT of adrenals ACTH-dependent
(consider macronodular hyperplasia) disease

• 50% cortisol suppression following high-dose


dexamethasone suppression test (2 mg 6 hourly for 48 hours)
• >50% increased serum cortisol post CRH stimulation test
• Normal potassium and bicarbonate
• Positive MRI scan of pituitary

Then proceed to If tests inconclusive (i.e., failure of any above criteria)


transsphenoidal hypophysectomy

Inferior petrosal sinus sampling with CRH


Positive ACTH gradient
Cushing disease
Transsphenoidal hypophysectomy

No ACTH gradient
Ectopic ACTH syndrome
Appropriate radiology ± venous sampling

Figure 15-21 The tests to uncover the cause of Cushing syndrome are debatable and differ in any given center depending on many factors, including familiarity and turnaround
time of hormone assays and local expertise in techniques such as inferior petrosal sinus sampling. Depicted here is an algorithm in use within many endocrine units based
on the reported sensitivity and specificity of each endocrine test. ACTH, adrenocorticotropic hormone; CRH, corticotropin-releasing hormone; CT, computed tomography;
MRI, magnetic resonance imaging.

is available (see later). Moreover, if the low-dose dexa- ectopic ACTH syndrome, but false-positive results have
methasone suppression test has been used in the diagnosis been reported. In distinguishing pituitary-dependent Cush-
of Cushing syndrome and a greater than 50% fall in corti- ing disease from the ectopic ACTH syndrome, the response
sol is observed, there is no added value in the high-dose of ACTH and cortisol to CRH has a specificity and a sensi-
dexamethasone suppression test.263 tivity of approximately 90%. However, a positive response
Corticotropin-Releasing Hormone Test. CRH is a 41–amino defined as an ACTH increase of 100% or a cortisol rise of
acid peptide that was identified by Vale in 1981 from ovine 50% over baseline values effectively eliminates a diagnosis
hypothalami. The ovine sequence differs by seven amino of ectopic ACTH syndrome, which is the real benefit of this
acid residues from that of the human hormone but is test. Up to 10% of patients with Cushing disease do not
slightly more effective in stimulating the release of ACTH respond to CRH.
in humans.268 The test involves the intravenous injection Inferior Petrosal Sinus Sampling and Selective Venous Catheter-
of either ovine or human sequence CRH in a dose of 1 μg/ ization. The most robust test to distinguish Cushing disease
kg body weight or a single dose of 100 μg (Fig. 15-23). The from the ectopic ACTH syndrome is IPSS.177 Because blood
test can be performed in the morning or afternoon. After from each half of the pituitary drains into the ipsilateral
basal sampling, CRH is administered and blood samples for inferior petrosal sinus, catheterization and venous sam-
ACTH and cortisol are then taken every 15 minutes for 1 pling of both sinuses simultaneously can distinguish a
to 2 hours.261,263,269,270 pituitary from an ectopic source of ACTH (Fig. 15-24).271,272
In normal subjects, CRH produces a rise in ACTH and In virtually all patients with the ectopic ACTH syndrome,
cortisol of 15% to 20%. This response is exaggerated in the ratio of the ACTH concentration in the inferior petrosal
Cushing disease, in which typically an ACTH increase sinus and that in simultaneously drawn peripheral venous
greater than 50% and a cortisol rise greater than 20% over blood is less than 1.4 : 1. In contrast, the ratio is elevated
baseline values are seen. Responses are seldom seen in the to greater than 2.0 in Cushing disease. However, because
518 SECTION IV Adrenal Cortex and Endocrine Hypertension

sponse or the absence of a macroadenoma on pituitary MRI


Cushing Adrenal Ectopic scanning).
disease tumor ACTH
Rarely, selective catheterization of vascular beds may be
required to identify the source of ectopic ACTH secretion
4000-12,000 (e.g., from a small pulmonary carcinoid or thymic tumor).
2000-4000 Tumors causing ectopic ACTH syndrome may also produce
peptide hormones other than ACTH or its precursors.
1000-2000

900
Imaging
CT/MRI Scanning of Pituitary and Adrenal Glands. High-
Plasma immunoreactive ACTH (pg/mL)

700 resolution, thin-section, contrast-enhanced computed to-


mography (CT) or MRI imaging has revolutionized the
500 investigation of Cushing syndrome.271,272 However, it is es-
sential that the results of any imaging technique be inter-
preted alongside the biochemical results if mistakes are to
300
be avoided. When the adrenals are imaged, asymmetric
nodular hyperplasia may lead to a false diagnosis of adrenal
250 adenoma. In the general population there is an approxi-
mately 10% rate of small abnormalities on pituitary MRI,
so-called pituitary incidentalomas, which may mimic a
200 corticotrope adenoma to the unwary, thus emphasizing the
need for careful biochemical assessment.276
Pituitary MRI is the investigation of choice once the
150
biochemical tests have suggested Cushing disease, with a
sensitivity of 60% and specificity of 87%. About 90% of
100 ACTH-secreting pituitary tumors are microadenomas (i.e.,
<10 mm in diameter). The classic features of a pituitary
Normal microadenoma are a hypodense lesion after contrast
50 range enhancement, which may be associated with deviation of
8-10 AM
the pituitary stalk, and a convex upper surface of the pitu-
0
itary gland (Fig. 15-25). CT scanning is no longer indicated
for investigation of Cushing disease.
Figure 15-22 Plasma adrenocorticotropic hormone (ACTH) concentrations in For adrenal imaging, CT offers better spatial resolu-
patients with Cushing disease, Cushing syndrome associated with adrenocortical tion277 (Fig. 15-26) and is the procedure of choice, but MRI
tumors, or the ectopic ACTH syndrome. To convert values to picomoles per liter, may provide diagnostic information in patients with sus-
multiply by 0.2202. (From Besser GM, Edwards CRW. Cushing’s syndrome. Clin Endo- pected adrenal carcinoma. Once again, so-called adrenal
crinol Metab. 1972;1:451-490.) incidentalomas are present in up to 5% of normal subjects,
so adrenal imaging should not be performed unless bio-
chemical investigation has suggested a primary adrenal
of the problem of intermittent ACTH secretion, it is useful cause (i.e., undetectable ACTH concentrations). Adrenal
to take measurements before and at intervals (e.g., 2, 5, and carcinomas are usually large and are often associated with
15 minutes) after intravenous injection of 100 μg synthetic metastatic spread at presentation (Fig. 15-27).
ovine or human CRH.273,274 Using this approach, an ACTH In patients with occult ectopic ACTH syndrome, high-
petrosal sinus/peripheral ratio greater than 3.0 after CRH definition CT/MRI scanning of thorax, abdomen, and
administration has a sensitivity of 95% and specificity of pelvis with images obtained every 0.1-0.5 cm may be
nearly 100% for diagnosing Cushing disease.274 When CRH required to detect small ACTH-secreting carcinoid tumors
is not available some centers use desmopressin as a secre- (Fig. 15-28).
tagogue, but central to peripheral gradients have been Scintigraphy Studies. Scintigraphy is of value in certain
observed in some patients with ectopic ACTH when using patients with primary adrenal disease. The most com-
this peptide. When a negative gradient is found and veno- monly used agent is 131I-labeled 6β-iodomethyl-19-
grams confirm correct catheter placement, measurement of norcholesterol,278 a marker of adrenocortical cholesterol
prolactin in the samples and correction of the ACTH values uptake. In patients with adrenal adenomas, the isotope is
can reduce the false-negative rate in Cushing disease.275 taken up by the adenoma but not by the contralateral
IPSS may also be of value in lateralizing a pituitary suppressed adrenal gland. Adrenal scintigraphy is useful
tumor in a patient in whom imaging techniques have in patients with suspected adrenocortical macronodular
failed to demonstrate a microadenoma; however, some hyperplasia, although it is not in widespread use; CT
centers have found this procedure to be of little value in scanning may misleadingly suggest unilateral disease,
predicting tumor location. Because many tumors are whereas isotope scanning identifies the bilateral adrenal
central and drain into both sinuses, current evidence sug- involvement.
gests that it is unwise to base the surgical procedure on the Many neuroendocrine tumors giving rise to the ectopic
results of IPSS studies alone. ACTH syndrome express somatostatin receptors and can be
IPSS is technically demanding; it has been associated imaged by administering radiolabeled analogues of soma-
with complications (referred aural pain, thrombosis) and tostatin (most commonly 111In-labeled octreotide). This
should be performed only in an experienced tertiary refer- technique can detect tumors as small as a few millimeters
ral center. In some centers, a clinical diagnostic algorithm in diameter and should be considered for patients with
is used (see Fig. 15-21), and IPSS is performed if the dif- ACTH-dependent Cushing syndrome in whom pituitary
ferential diagnosis remains in doubt (i.e., lack of adequate disease has been excluded, although discovery of a lesion
suppression after high-dose dexamethasone or CRH re- that has not been seen on axial imaging is rare.279
CHAPTER 15 The Adrenal Cortex 519

200 120
190 Cushing disease 110 Ectopic ACTH (n = 6)
180 (n = 6)
100 Controls (n = 10)
170
Controls (n = 10) 90
160
150 80
140 70

Plasma IR-ACTH (pg/mL)


130
60
120
110 50
100 40
90 30
80
20
70
60 10
50 0
40
30 55
20 50
10 45
0
40
55 35
50
30
Plasma cortisol (mg/dL)
45
40 25
35 20
30 15
25
10
20
15 5
10 0
5
−15 0 15 30 60 90 120 150 180
0
Time (min)
−15 0 15 30 60 90 120 150 180
Time (min)
Figure 15-23 Comparison of cortisol and adrenocorticotropic hormone (ACTH) responses to an intravenous injection of ovine corticotropin-releasing hormone (1 μg/kg) in
normal subjects, patients with Cushing disease, and patients with the ectopic ACTH syndrome. IR, immunoreactive. (From Chrousos GP, Schulte HM, Oldfield EH, et al. The
corticotropin-releasing factor stimulation test: an aid in the evaluation of patients with Cushing’s syndrome. N Engl J Med. 1984;310:622-626.)

Cavernous Inferior Treatment of Cushing Syndrome


sinus petrosal sinus
Adrenal Causes
Unilateral adrenal adenomas should be removed by adre-
nalectomy; the cure rate is 100%, with serum cortisol
values that are less than 50 nmol/L at 9 AM, or even lower
in many modern assays.280 With the increasing experience
of laparoscopic adrenalectomy in most tertiary centers, this
procedure has now become the surgical treatment of choice
for unilateral tumors, offering reduced surgical morbidity
and postoperative hospital stay compared with traditional
open approaches.281 After surgery, it may take many months
or even years for the contralateral suppressed adrenal to
Jugular recover. Glucocorticoid replacement regimens vary, but
vein many centers use low doses (15-20 mg) of hydrocortisone,
and withdrawal regimens differ. One practical approach is
to measure the morning plasma cortisol having omitted
the dose of hydrocortisone in the morning at 3-month
Confluent intervals. Patients with serum cortisol values less than
pituitary veins 200 nmol/L (7.0 μg/dL) should continue glucocorticoid
replacement, whereas in patients with values above
Figure 15-24 Anatomy of the venous drainage of the pituitary gland through the
500 nmol/L (18.3 μg/dL) this replacement can be stopped.
inferior petrosal venous sinuses. (From Oldfield EH, Chrousos GP, Schulte HM, et al.
Preoperative lateralization of ACTH-secreting pituitary microadenomas by bilateral In patients with values between 200 and 500 nmol/L,
and simultaneous inferior petrosal sinus sampling. N Engl J Med. 1985;312: ACTH(1-24) testing can be used to assess whether there is
100-103.) sufficient response to stress, although insulin tolerance
520 SECTION IV Adrenal Cortex and Endocrine Hypertension

A B
Figure 15-25 A, Magnetic resonance imaging (MRI) scan of pituitary demonstrates the typical appearance of a pituitary microadenoma. A hypodense lesion is seen in the
right side of the gland (arrow), with deviation of the pituitary stalk away from the lesion. After a biochemical diagnosis of Cushing disease, this patient was cured by trans-
sphenoidal hypophysectomy. B, MRI scan of the pituitary gland demonstrates a large macroadenoma (arrow) in a patient with Cushing disease. In contrast to smaller tumors,
large macroadenomas are invariably invasive and recur after surgery.

A B

C D
Figure 15-26 A, Adrenal computed tomographic (CT) scan demonstrates bilateral adrenal hyperplasia (arrows) in a patient with Cushing disease. B, CT scan of a typical solitary
left adrenal adenoma (arrow) causing Cushing syndrome. C, Cushing syndrome caused by massive macronodular hyperplasia. Adrenal glands are replaced by multiple nodules
(arrows). The combined weight of the adrenal glands was more than 100 g. D, Cushing syndrome caused by surgically proven primary pigmented nodular adrenal disease in a
21-year-old patient. Notice the multiple small nodules with relatively atrophic internodular adrenocortical tissue involving the medial limb of the right adrenal gland (arrow).
(C and D, From Findling JW, Doppman JL. Biochemical and radiologic diagnosis of Cushing’s syndrome. Endocrinol Metab Clin North Am. 1994;23:511-537.)

testing may be used in some centers. In the interim, all usual practice to try to remove the primary tumor, even
patients should carry a steroid alert card and increase though metastases may be present, so as to enhance the
their dose of replacement therapy in the event of an inter- response to the adrenolytic agent o,p′-DDD283 (mitotane).
current illness. Radiotherapy to the tumor bed and to some metastases,
Adrenal carcinomas have had a very poor prognosis, and such as those in the spine, may be of limited value.
most patients have died within 2 years of diagnosis.282 It is However, in recent years significant progress has been
CHAPTER 15 The Adrenal Cortex 521

A B
Figure 15-27 Computed tomographic scan of a patient with rapidly progressing Cushing syndrome caused by an adrenal carcinoma. An irregular right adrenal mass is shown
in A, and a large liver metastasis is seen in B.

A B

C D
Figure 15-28 Imaging of the thorax in a patient with the ectopic adrenocorticotropic hormone (ACTH) syndrome. A, Plain chest radiograph demonstrates a suspicious lesion
behind the left heart border (arrow). B and C, Axial and sagittal computed tomographic images demonstrate a bronchial carcinoid tumor (arrow) abutting the diaphragm.
D, Three-dimensional reconstruction illustrates adherence of the tumor to the diaphragm (arrow), which was confirmed at surgery. (From Newell-Prince J, Trainer P, Besser M,
et al. The diagnosis and differential diagnosis of Cushing’s syndrome and pseudo-Cushing’s states. Endocr Rev. 1998;19:647-672.)

made by implementing collaborative multicenter studies. Pituitary-Dependent Cushing Syndrome


Phase III trials of mitotane therapy achieving therapeutic
plasma mitotane levels have shown significant benefit284; The treatment of Cushing disease has been significantly
drug combinations include etoposide, doxorubicine, and enhanced with transsphenoidal surgery conducted by an
cisplatin plus mitotane or streptozotocin plus mitotane. experienced surgeon.286 Before the capability of selective
Several targeted therapies, including IGF-1 inhibitors, suni- removal of a pituitary microadenoma was developed,
tinib, and sorafenib, may be of value in cases of mitotane the treatment of choice was bilateral adrenalectomy,
failure. The 10-year survival rate for patients with T1 N0 which had an appreciable mortality rate even in the best
M0 disease is about 80% but is significantly impaired with centers (up to 4%) and a significant morbidity rate. The
increased tumor mass, positive lymph nodes, and distant major risk was the subsequent development of Nelson syn-
metastases, reaching less than 20% for patients with T1-4 drome (postadrenalectomy hyperpigmentation with a
N0-1 M1.285 locally aggressive pituitary tumor) (Fig. 15-29), which was
522 SECTION IV Adrenal Cortex and Endocrine Hypertension

D E
Figure 15-29 A young woman with Cushing disease, photographed initially beside her identical twin sister (A). In this case, treatment with bilateral adrenalectomy was
undertaken. Several years later, the patient presented with Nelson syndrome and a right third cranial nerve palsy (B and C) related to cavernous sinus infiltration from a locally
invasive corticotropinoma (D). Hypophysectomy and radiotherapy were performed with reversal of the third cranial nerve palsy (E). Note the advancing skin pigmentation of
Nelson syndrome.

attributed to loss of any negative feedback after adrenalec- but may be performed if pituitary surgery has failed or the
tomy but is more likely to be due to an aggressive pituitary condition has recurred.
tumor from the outset.287 In an attempt to avoid this com- The surgical outcome for transsphenoidal hypophysec-
plication, pituitary irradiation was often carried out at tomy varies from center to center and with surgical exper-
the time of bilateral adrenalectomy.288 In addition, these tise.210 Because of the hazards of untreated Cushing disease
patients required lifelong replacement therapy with hydro- and the potential complications of surgery, the endocri-
cortisone and fludrocortisone. Currently, bilateral adrenal- nologist should refer patients only to a recognized surgical
ectomy is rarely indicated for patients with Cushing disease specialist at a center in which outcome data have been
CHAPTER 15 The Adrenal Cortex 523

A B C Plasma cortisol response following transsphenoidal


microsurgery for Cushing disease
0.8
0.7 Transsphenoidal

0900 Plasma cortisol µmol/L


microadenectomy
0.6 I.H.T.

0.5
ACTH-ve ACTH ACTH-ve 0.4
0.3
ACTH +++ ACTH-ve ACTH +
0.2
0.1
Cortisol +++ Cortisol ND Cortisol +

+++ Secretion above normal 0 5 10 15 20 25


+ Secretion is detectable Weeks
-ve Secretion suppressed Figure 15-31 Gradual recovery of function of the hypothalamic-pituitary-adrenal
ACTH secreting tumor axis in a patient after removal of a pituitary adrenocorticotropic hormone-secreting
Figure 15-30 Selective removal of a microadenoma and its effect on the microadenoma. Morning (9 AM) plasma cortisol levels were measured. The insulin
hypothalamic-pituitary-adrenal axis. A, Before treatment. B, After total removal of hypoglycemia test (I.H.T.) eventually demonstrated the return of a normal stress
adenoma. C, After incomplete excision. Because the surrounding normal pituitary response.
corticotrophs are suppressed in a patient with an adrenocorticotropic hormone
(ACTH)-secreting pituitary adenoma, successful removal of the tumor results in
ACTH, and hence adrenocortical, deficiency, with an undetectable (<50 nmol/L
[2 μg/dL]) plasma cortisol level. A postoperative plasma cortisol level higher than
bilateral adrenalectomy, and patients with established
50 nmol/L (2 μg/dL) implies that the patient is not cured. (Courtesy of Professor Peter
Trainer.) Nelson syndrome.
The management of recurrent Cushing disease involves
a consideration of repeat surgery, gamma knife radiosur-
gery, and medical therapies.286,289
established. In optimal centers, remission rates are 70% to
90% for microadenomas and 50% for macroadenomas.202
Rates for postoperative hypopituitarism and permanent
Ectopic ACTH Syndrome
diabetes insipidus depend on how aggressive the surgeon Treatment of the ectopic ACTH syndrome depends on the
was in removing pituitary tissue. The ideal outcome is a cause. If the tumor can be found and has not spread, then
cured patient with intact pituitary function, but this result its removal can lead to cure (e.g., bronchial carcinoid,
may not be possible for a patient with Cushing disease in thymoma). However, the prognosis for small cell lung
whom a pituitary adenoma was not identified preopera- cancer associated with the ectopic ACTH syndrome is poor.
tively or during the operation itself. The cortisol excess and associated hypokalemic alkalosis
In centers that lack facilities for frequent monitoring of and diabetes mellitus can be ameliorated by medical
cortisol levels, perioperative and postoperative hydrocorti- therapy. Treatment of the small cell tumor itself will also,
sone cover is advised; this can be reduced to maintenance at least initially, produce improvement. Sometimes, if the
replacement doses usually within 3 to 7 days. On days 2 ectopic source of ACTH cannot be found, it may be neces-
to 5 postoperatively, a 9 AM plasma cortisol level should be sary to perform bilateral adrenalectomy and then monitor
measured with the patient having omitted hydrocortisone the patient carefully (sometimes for several years) before
for 24 hours. After selective removal of a microadenoma, the primary tumor becomes apparent.
the surrounding corticotrophs are usually suppressed (Fig.
15-30). As a result, plasma cortisol levels are less than
30 nmol/L (<1 μg/dL) postoperatively, and ongoing gluco-
Medical Treatment of Cushing Syndrome
corticoid replacement therapy is required. The HPA axis Several drugs have been used in the treatment of Cushing
usually (but not invariably) exhibits gradual recovery, and syndrome.286 Metyrapone inhibits 11β-hydroxylase and
glucocorticoid therapy is needed until this occurs (Fig. has been the most commonly given agent, often with a
15-31). A nonsuppressed plasma cortisol postoperatively goal of lowering cortisol concentrations before definitive
suggests that the patient is not in remission even though therapy or while awaiting benefit from pituitary irradia-
cortisol secretion may have fallen to normal or subnormal tion. The daily dose must be determined by measurements
values.203,204 The recurrence rate in patients with an estab- of plasma or urinary free cortisol. The aim should be to
lished remission after pituitary surgery is 2%, but this achieve a mean plasma cortisol concentration of about
value is higher in children (up to 40%).205,206 A detailed 300 nmol/L (11 μg/dL) during the day or a normal urinary
assessment of residual pituitary function is required in free cortisol level. The drug is usually given in doses
each patient, and close follow-up of such individuals is ranging from 250 mg twice daily to 1.5 g every 6 hours,
warranted. with lower doses for adrenal adenoma and higher for
In the past, pituitary irradiation was often used in the ectopic ACTH. Nausea is a side effect that can be helped (if
treatment of Cushing disease. However, because of the it is not caused by adrenal insufficiency) by giving the drug
improvements in pituitary surgery, far fewer patients are so with milk.208
treated. In children, pituitary irradiation appears to be Ketoconazole is an imidazole that has been widely used
more effective.207 Radiotherapy is not recommended as a as an antifungal agent but causes abnormal liver function
primary treatment but is reserved for patients not respond- tests in about 15% of patients. Ketoconazole blocks a
ing to pituitary microsurgery, those who have undergone variety of steroidogenic cytochrome P450–dependent
524 SECTION IV Adrenal Cortex and Endocrine Hypertension

enzymes and thus lowers plasma cortisol levels. For effec-


Glucocorticoid Resistance
tive control of Cushing syndrome, 400 to 1600 mg daily
has been required.212,290 Ketoconazole frequently causes an A small number of patients have been described as having
increase in hepatic enzyme values, and if this level remains increased cortisol secretion but without the stigmata of
less than three times the upper limit of normal the drug Cushing syndrome.87,297 These patients are resistant to sup-
may be continued, but hepatic failure has been described pression of cortisol with low-dose dexamethasone but
and the drug has been withdrawn from the market for the respond to high doses. ACTH levels are elevated and
treatment of fungal infections. lead to increased adrenal production of androgens and
Mitotane (o,p′-DDD) is an adrenolytic drug that is taken DOC. Therefore, these patients may present with the fea-
up by both normal and malignant adrenal tissue, causing tures of androgen or mineralocorticoid excess, or both.
adrenal atrophy and necrosis.283 Because of its toxicity, it Treatment with a dose of dexamethasone (usually >3 mg/
has been used mainly in the management of adrenal car- day) adequate to suppress ACTH results in a fall in adrenal
cinoma. Doses of up to 5 g/day are required to control androgens and often returns plasma potassium and blood
glucocorticoid excess, although evidence that the drug pressure to normal levels. Many of these patients have
causes tumor shrinkage or improves long-term survival is been found to have point mutations in the steroid-
lacking. This agent also produces mineralocorticoid defi- binding domain of the GR, with consequent reduction of
ciency, and concomitant glucocorticoid and mineralocor- glucocorticoid-binding affinity, but this finding is not
ticoid replacement therapy may be required. Side effects invariable. A useful clinical discriminatory test to differen-
are common and include fatigue, skin rashes, neurotoxic- tiate this condition from Cushing syndrome is to measure
ity, and gastrointestinal disturbance. It has also been used bone mineral density: it is preserved in patients with glu-
at lower doses in Cushing disease.291 cocorticoid resistance or even increased in females because
Somatostatin analogues such as octreotide and lanreo- of the androgen excess. In addition, the circadian rhythm
tide are generally ineffective in Cushing disease. However, for ACTH and cortisol is preserved in patients with gluco-
the multireceptor somatostatin analogue, pasireotide, corticoid resistance.
which demonstrates high-affinity binding to somatostatin
receptor subtypes 1, 2, 3, and 5, normalizes urinary free
cortisol in 17% of patients with Cushing disease, with GLUCOCORTICOID DEFICIENCY
hyperglycemia being a common side effect.292 The GR
antagonist mifepristone has been shown to improve diabe-
tes in patients with Cushing syndrome, but biochemical
Primary and Central Hypoadrenalism
monitoring is not possible when using this drug.293 Primary hypoadrenalism refers to glucocorticoid deficiency
occurring in the setting of adrenal disease, whereas central
hypoadrenalism arises because of deficiency of ACTH
Prognosis of Cushing Syndrome (Table 15-15). A major distinction between these forms of
Studies performed before the introduction of effective hypoadrenalism is that mineralocorticoid deficiency invari-
therapy revealed that 50% of patients with untreated ably accompanies primary hypoadrenalism, but this does
Cushing syndrome died within 5 years, principally from not occur in central hypoadrenalism: here only ACTH is
vascular disease.187 Even with modern management, an deficient, and the renin-angiotensin-aldosterone (RAA)
increased prevalence of cardiovascular risk factors persists axis is intact. A further important cause of adrenal insuf-
for many years after an apparent remission.188,189 Paradoxi- ficiency in which there may be dissociation of glucocorti-
cally, on correction of the hypercortisolism, patients coid and mineralocorticoid secretion is CAH.
often feel worse. Skin desquamation, steroid-withdrawal
arthropathy, profound lethargy, and mood changes may Primary Hypoadrenalism
occur and can take several weeks or months to resolve.294
In our experience, these features, together with postural Addison Disease
hypotension, are particularly severe in patients in remis- Thomas Addison described the condition now known as
sion who are also rendered vasopressin deficient. They can primary hypoadrenalism in his classic monograph published
usually be ameliorated by a transient increase in glucocor- in 1855.2 Addison disease is a rare condition with an esti-
ticoid replacement therapy. Patients are invariably GH mated incidence in the developed world of 0.8 cases per
deficient, and GH replacement therapy may produce clini- 100,000 and a prevalence of 4 to 11 cases per 100,000
cal benefit. population. Nevertheless, it is associated with significant
Features of Cushing syndrome disappear over a period morbidity and mortality rates, but once the diagnosis is
of 2 to 12 months after treatment. Hypertension and dia- made it can be easily treated.298,299 Causes of Addison
betes mellitus improve, but as with other secondary causes, disease are listed in Table 15-15.
they may not resolve completely. The osteopenia of Autoimmune Adrenalitis. In the Western world, autoimmune
Cushing syndrome improves rapidly during the first 2 years adrenalitis accounts for more than 70% of all cases of
after treatment but resolves more slowly thereafter.295 Ver- primary hypoadrenalism.300 Pathologically, the adrenal
tebral fractures and osteonecrosis are irreversible, and per- glands are atrophic with loss of most of the cortical
manent deformity results. Visceral obesity and myopathy cells, but the medulla is usually intact. In 75% of cases,
are both reversible features. Reproductive and sexual func- adrenal autoantibodies can be detected.301 Fifty percent
tion return to normal within 6 months, provided that of patients with this form of Addison disease have an
anterior pituitary function was not compromised. Long- associated autoimmune disease (Table 15-16), thyroid
term health-related quality of life in adults significantly disease being the most common. Conversely, only 1%
improves after treatment, but quality-of-life scores do not to 2% of patients with more common autoimmune dis-
return to normal levels.184 Similar observations have been eases such as insulin-dependent diabetes mellitus or thyro-
made in pediatric patients, in whom significant improve- toxicosis have antiadrenal autoantibodies and develop
ment was noted before and after treatment but residual adrenal disease, although the figure is higher in patients
impairment of health-related quality of life persisted 1 year with autoimmune hypoparathyroidism (16%). These auto-
after cure.296 immune polyglandular syndromes (APSs) have been
CHAPTER 15 The Adrenal Cortex 525

TABLE 15-15 TABLE 15-16


Etiology of Adrenocortical Insufficiency (Excluding Congenital Incidence of Other Endocrine and Autoimmune Diseases in
Adrenal Hyperplasia) Patients With Autoimmune Adrenal Insufficiency
Primary Causes: Addison Disease Disease Incidence (%)
Autoimmune Thyroid disease
Sporadic Hypothyroidism 8
Autoimmune polyendocrine syndrome type I (Addison disease, Nontoxic goiter 7
chronic mucocutaneous candidiasis, hypoparathyroidism, dental Thyrotoxicosis 7
enamel hypoplasia, alopecia, primary gonadal failure—see Gonadal failure
Chapter 40) Ovarian 20
Autoimmune polyendocrine syndrome type II (Schmidt syndrome) Testicular 2
(Addison disease, primary hypothyroidism, primary Insulin-dependent diabetes mellitus 11
hypogonadism, insulin-dependent diabetes, pernicious anemia, Hypoparathyroidism 10
vitiligo—Chapter 40) Pernicious anemia 5
Infections None 53
Tuberculosis
Fungal infections
Cytomegalovirus
HIV Infections. Worldwide, infectious diseases are the most
Metastatic tumor common cause of primary adrenal insufficiency. These dis-
Infiltrations eases include tuberculosis, fungal infections (histoplasmo-
Amyloid
Hemochromatosis
sis, cryptococcosis), and cytomegalovirus infection. Adrenal
Intra-adrenal hemorrhage (Waterhouse-Friderichsen syndrome) after failure may also occur in the acquired immunodeficiency
meningococcal septicemia syndrome (AIDS).302
Adrenoleukodystrophies Tuberculous Addison disease results from hematogenous
Congenital adrenal hypoplasia spread of the infection from elsewhere in the body; extra-
DAX1 (NR0B1) mutations adrenal disease is usually evident. The adrenals are initially
SF1 mutations enlarged, with extensive epithelioid granulomas and case-
ACTH resistance syndromes ation, and both the cortex and the medulla are affected.
MC2R gene mutations Fibrosis ensues, and the adrenals become normal in size or
MRAP gene mutations
AAAS (ALADIN) gene mutations (triple-A syndrome)
smaller, with calcification evident in 50% of cases.
Bilateral adrenalectomy The adrenals are frequently involved in patients with
AIDS302,303; adrenalitis may occur after infection with cyto-
Secondary Causes: Central Hypoadrenalism megalovirus or atypical mycobacteria, and Kaposi sarcoma
Exogenous glucocorticoid therapy may result in adrenal replacement. Onset is often insidious,
Hypopituitarism but if tested, more than 10% of patients with AIDS will
Selective removal of ACTH-secreting pituitary adenoma demonstrate a subnormal cortisol response to a short Syn-
Pituitary tumors and pituitary surgery, craniopharyngiomas acthen test. Adrenal insufficiency may be precipitated
Pituitary apoplexy
Granulomatous disease (tuberculosis, sarcoid, eosinophilic granuloma)
through the concomitant administration of appropriate
Secondary tumor deposits (breast, bronchus) anti-infectives such as ketoconazole (which inhibits corti-
Postpartum pituitary infarction (Sheehan syndrome) sol synthesis) or rifampicin (which increases cortisol
Pituitary irradiation (effect usually delayed for several years) metabolism). Rarely, patients with AIDS and features of
Isolated ACTH deficiency adrenal insufficiency are found to have elevated circulating
Idiopathic ACTH and cortisol concentrations that fail to suppress nor-
Lymphocytic hypophysitis mally after low-dose dexamethasone administration. This
TPIT (TBX19) gene mutations is thought to reflect an acquired form of glucocorticoid
PCSK1 gene mutation (POMC processing defect) resistance resulting from reduced GR affinity, but the
POMC gene mutations
Multiple pituitary hormone deficiencies
underlying cause remains unknown.304
HESX1 gene mutations
LHX4 gene mutations Acquired Primary Adrenal Insufficiency
SOX3 gene mutations
PROP1 gene mutations With the exception of tuberculosis and autoimmune
adrenal failure, other causes of Addison disease are rare (see
ACTH, adrenocorticotropic hormone; HIV, human immunodeficiency virus;
POMC, pro-opiomelanocortin.
Table 15-15). Adrenal metastases (most commonly from
primary tumors in the lung or breast) are often found at
postmortem examinations, but they uncommonly cause
adrenal insufficiency,305 perhaps because more than 90% of
the adrenal cortex must be compromised before symptoms
classified into two distinct variants.301 APS type I, or auto- and signs become apparent. Necrosis of the adrenals due
immune polyendocrinopathy-candidiasis-ectodermal dys- to intra-adrenal hemorrhage should be considered in any
plasia (APECED), is a rare autosomal recessive condition severely sick patient, particularly a patient with underlying
comprising Addison disease, chronic mucocutaneous can- infection, trauma, or coagulopathy.306 Intra-adrenal bleed-
didiasis, and hypoparathyroidism. The more common APS ing may be found in patients with severe septicemia of
type II comprises Addison disease, autoimmune thyroid any cause, particularly in children, in whom a common
disease, diabetes mellitus, and hypogonadism. Here, auto- cause is infection with Pseudomonas aeruginosa. When
antibodies to 21-hydroxylase are usually present and are caused by meningococci, the association with adrenal
predictive for the development of adrenal destruction.301 insufficiency is known as the Waterhouse-Friderichsen syn-
Polyglandular autoimmune syndromes are discussed in drome. Adrenal replacement may also occur with amyloi-
greater detail in Chapter 40. dosis and hemochromatosis.
526 SECTION IV Adrenal Cortex and Endocrine Hypertension

child with normal activity of the RAA system is highly sug-


Inherited Primary Adrenal Insufficiency
gestive of FGD. The diagnosis can be confirmed by demon-
Adrenal hypoplasia congenita (AHC) is an X-linked disorder strating low cortisol in combination with increased ACTH
comprising congenital adrenal insufficiency and combined concentrations and normal plasma renin and aldosterone
primary and central hypogonadotropic hypogonadism. measurements.91 The type 1 variant accounts for approxi-
The condition is caused by mutations in the DAX1 (NROB1) mately 25% of all cases and is explained by inactivating
gene, a member of a nuclear receptor family that is mutations in the ACTH-binding receptor, MC2R.322-324 The
expressed in the adrenal cortex, gonads, and hypothala- FGD type 2 variant is caused by mutations in the MRAP
mus.307,308 Depending on the molecular defect, the clinical gene, which is thought to mediate intracellular trafficking
presentation can be highly variable. Severe cases often of the MC2R, has been reported in some families.90 However,
manifest with mineralocorticoid deficiency and gradually 50% of patients with FGD do not have mutations in either
develop glucocorticoid deficiency. Hypogonadism is com- MC2R or MRAP; other loci are being defined.
bined with primary testicular abnormalities and low A variant called the triple A syndrome or Allgrove syndrome
gonadotropin levels. However, the so-called minipuberty refers to the triad of adrenal insufficiency due to ACTH
of infancy can be normal.309,310 Patients presenting with resistance, achalasia, and alacrima. It is caused by muta-
late-onset adrenal failure have also been described.311 tions in the AAAS gene, which encodes ALADIN, a
Mutations in another transcription factor, SF1, may also tryptophan/aspartate WD-repeat–containing protein of the
result in adrenal insufficiency due to lack of development nuclear pore complex.325,326 The exact function of ALADIN
of a functional adrenal cortex. The transcriptional regula- is unknown, but its interaction with other proteins of the
tion of many P450 steroidogenic enzymes is dependent on nuclear pore complex suggest that it is part of a structural
SF1.15 When it was first described, SF1 mutation was associ- scaffold.
ated with complete sex reversal causing 46,XY disorder of Several syndromic disorders are associated with adrenal
sex development (DSD).312 However, novel clinical pheno- insufficiency for which the underlying molecular genetic
types in SF1-deficient patients are now emerging; they defect remains to be elucidated.11
range from isolated adrenal failure313 to isolated gonadal
failure314 and ovarian insufficiency.315 AHC may also Secondary Hypoadrenalism
occur in association with glycerol kinase deficiency and
muscular dystrophy caused by gene deletion, including the Inherited Central Hypoadrenalism
DAX1 gene.316 Central hypoadrenalism may be defined as hypocorti-
Adrenoleukodystrophy has a prevalence rate of 1 : 20,000 solemia secondary to a deficiency in ACTH. The prevalence
and is a cause of adrenal insufficiency in association with of central hypoadrenalism is 125 to 280 per million,1,326a
demyelination within the nervous system; demyelination which is likely to be an underestimate considering the use
results from a failure of β-oxidation of fatty acids within of therapeutic corticosteroids in the general population. It
peroxisomes owing to reduced activity of very long chain occurs in up to one third of patients with pituitary
acyl-CoA synthetase.317 Increased accumulation of very disease.3,326b ACTH deficiency is an important diagnosis to
long chain fatty acids (VLCFAs) occurs in many tissues, and make; in subjects with tumoral and posttraumatic hypopi-
serum assays can be used diagnostically. Only males have tuitarism, central hypoadrenalism is associated with
the fully expressed condition, and carrier females are increased mortality rates.4-6,326c-326e The causes of central
usually normal. Several forms are recognized: a childhood hypoadrenalism are outlined in Table 15-15; the most
cerebral form (30% to 40% of cases), adult adrenomy- common reason is ACTH suppression by exogenous gluco-
eloneuropathy (40%), and Addison disease (7%). The corticoid treatment.7,178
childhood-onset form manifests at 5 to 10 years of age with When caused by pituitary disease, other pituitary hor-
progression eventually to a blind, mute, and severely mones are often deficient, so the patient presents with
spastic tetraplegic state. Adrenal insufficiency is usually partial or complete hypopituitarism. The clinical features
present but does not appear to correlate with the neuro- of hypopituitarism make this a relatively easy diagnosis. By
logic deficit. Nevertheless, this is the most common form contrast, isolated ACTH deficiency is rare, and the diagno-
of adrenal insufficiency in a child younger than 7 years of sis is difficult to make. It may occur in patients with lym-
age.318 Adrenomyeloneuropathy, by contrast, manifests phocytic hypophysitis. Mutations in the TBX19 gene, the
later in life with the gradual development of spastic paresis product of which (TPIT) regulates POMC expression, have
and peripheral neuropathy. Both the childhood and the been reported in a few cases of isolated ACTH deficiency
adult condition result from mutations in the ABCD1 gene occurring in neonatal life.327 A rare but fascinating cause
on chromosome Xq28, which encodes for an ABC peroxi- relates to a defect in the normal post-translational process-
somal membrane protein involved in the import of VLCFA ing of POMC to ACTH by the prohormone convertase
into the peroxisome.319 So far, more than 400 mutations enzymes (PC1 and PC2).328 Such patients may have more
have been reported in the ABCD1 gene with no relation- generalized defects in peptide processing (e.g., cleavage of
ship between genotype and phenotype.320,321 Treatment proinsulin to insulin) giving rise to diabetes mellitus.
options are few. Monounsaturated fatty acids, which block Some patients have mutations in the POMC gene that
the synthesis of the saturated VLCFA, have been used; a interrupt the synthesis of ACTH and cause ACTH defi-
combination of erucic acid and oleic acid (Lorenzo’s oil) ciency. Elucidation of the phenotype of these patients has
has led to normal levels of VLCFA. Treatment does not alter uncovered a novel role for POMC peptides in regulating
the rate of neurologic deterioration but may prevent new appetite and hair color: in addition to adrenal insufficiency,
neurologic damage in asymptomatic cases.321 Bone marrow POMC mutations result in severe obesity and red hair pig-
transplantation is a further possibility. mentation.329 A central role for α-MSH in regulating food
Familial glucocorticoid deficiency (FGD), or inherited unre- intake via the hypothalamic MC4R has been established,55
sponsiveness to ACTH, is a rare autosomal recessive cause and in recombinant mice lacking the POMC gene, the
of hypoadrenalism that usually manifests in childhood. obese phenotype can be reversed by giving an α-MSH
Most patients present with neonatal hypoglycemia or later agonist peripherally.330
in life with increasing pigmentation, and they often have Other rare inborn causes of secondary insufficiency are
enhanced growth velocity. Primary adrenal failure in a the result of mutations in genes involved in pituitary
CHAPTER 15 The Adrenal Cortex 527

development, such as HESX1,331 LHX4,332 SOX3,333 and 5 mg of prednisone daily.335 This report has formed the
PROP1.334 These defects result in congenital hypopituita- basis of the practice of assessing HPA axis function in
rism with multiple pituitary hormone deficiencies: ACTH patients who have been on more than 5 mg of predniso-
deficiency may not be present at the time of diagnosis, it lone or equivalent for more than 3 months. Recently, lower
but develops progressively over time. doses of glucocorticoid have been shown to suppress cor-
Secondary hypoadrenalism is also observed in patients tisol production; in one study, more than 60% of subjects
with Cushing disease after successful and selective removal on a glucocorticoid dose equivalent of less than 5 mg of
of the ACTH-secreting pituitary adenoma. The function of prednisolone per day had a subnormal ACTH or cortisol
adjacent normal pituitary corticotrophs is suppressed and response to CRF.335a In patients on long-term tapering of
may remain so for many months after curative surgery.203-205 prednisolone who were considered for steroid withdrawal
once they had reached a dose of 7 mg/day, 48% had adrenal
insufficiency defined by basal cortisol (<100 nmol/L) or
ACTH Suppression by Exogenous Glucocorticoids
response to 250 μg synthetic ACTH (<550 nmol/L); a
The ability of exogenously administered corticosteroids to longer duration of prednisolone therapy (13.7 years) was
cause adrenal atrophy has been appreciated since their seen in the hypoadrenal group defined by a low baseline
discovery in the 1940s. HPA axis suppression by exogenous cortisol but not in the group defined by cortisol response
glucocorticoid is not trivial and has been described with to 250 μg synthetic ACTH (6.1 years).335b Dynamic testing
intra-articular, topical, ocular, rectal, and inhaled, as well of adrenal function, such as the Synacthen stimulation test
as systemic, therapy.334a,334b,337 (SST) and CRF tests, suggests that cortisol production recov-
There is considerable interindividual variability in ers after withdrawal of long-term glucocorticoids; in a
response to glucocorticoids; there are no absolute cutoff meta-analysis of patients receiving glucocorticoid therapy,
values for the type of steroid taken, dose, route of admin- 46% to 100% had insufficient cortisol response 1 day after
istration, duration of treatment, or time since steroid with- withdrawal, which improved to 26% to 49% in patients at
drawal that predict adrenal suppression. However, there are assessment 1 week later.36 Up to 10% of patients still have
some generic issues that can guide diagnosis and therapy. biochemical evidence of hypoadrenalism between 6 and 20
Relative steroid potencies in their affinity/transactivation months after withdrawal of glucocorticoids.335c
of the GR have been described based on suppression of All patients receiving long-term therapy with cortico-
corticosterone production, in vitro binding to the glucocor- steroids should be treated in a similar fashion to patients
ticoid (GR), and by functional changes in GC target tissues with chronic ACTH deficiency; they should carry steroid
by individual steroids334c; these suggest that oral doses of cards and be offered steroid alert bracelets or necklaces. In
20 mg hydrocortisone, 5 mg of prednisolone, and 0.75 mg the event of an intercurrent stress (e.g., infection, surgery),
of dexamethasone are bioequivalent. Dexamethasone has supplemental steroid cover should be given. If the patient
a longer half-life and higher affinity for the GR than hydro- is unable to take drugs orally, parenteral therapy is required.
cortisone and exerts a more sustained suppressive effect on During recovery from suppression and without replace-
the HPA axis. Similarly, it is unclear how the potency of oral ment therapy, patients may experience symptoms of glu-
hydrocortisone compares to that of glucocorticoids taken cocorticoid deficiency, including anorexia, nausea, weight
by other routes. Budesonide has more potent action at the loss, arthralgia, lethargy, skin desquamation, and postural
GR than dexamethasone or prednisolone,334d but its effect dizziness (see the later discussions of adrenal insuffi-
on adrenal suppression is dependent on systemic absorp- ciency).336 To avoid these symptoms, steroids should be
tion of the inhaled compound. Comparison of inhaled cautiously withdrawn over a period of months.175 Assum-
fluticasone with inhaled steroids such as budesonide or ing that the underlying disease permits steroid reduction,
beclomethasone indicated that fluticasone was more fre- doses should be reduced from pharmacologic levels to
quently associated with suppression of the HPA axis and physiologic levels (equivalent to 7.5 mg/day prednisolone)
that adults using over 1000 μg of inhaled fluticasone for over a few weeks. Thereafter, doses should be reduced by
over a year were at risk of adrenal suppression.175 1 mg/day of prednisolone every 2 to 4 weeks depending
Concomitant therapies can augment potency and on patient well-being. An alternative approach is to switch
adrenal suppression. The coadministration of inhaled fluti- the patient to hydrocortisone 20 mg/day and reduce the
casone with one of the many medications that suppress its daily dose by 2.5 mg/day every week to a level of 10 mg/
clearance by inhibition of CYP3A4 is associated with day. After 2 to 3 months on reduced doses, endogenous
adrenal suppression (see Table 15-2).334f Co-prescriptions of function of the HPA axis can be assessed by a corticotropin
agents that do not affect glucocorticoid clearance but have (ACTH-Synacthen) stimulation test or an insulin-induced
affinity for the GR are also associated with adrenal suppres- hypoglycemia test. A “pass” response to these tests indi-
sion, the most notorious example being progesterone cates adequate function of the HPA axis, and corticosteroid
derivatives such as medroxyprogesterone acetate given in therapy can be safely withdrawn. In those patients who are
high dose to oncology patients.172 taking physiologic doses of prednisolone (less than 5 to
In terms of dose and duration adrenal atrophy and sub- 7.5 mg/day) or equivalent corticosteroid, an SST given 12
sequent deficiency should be anticipated in any subject to 24 hours after omitted steroid therapy will provide an
who has taken more than the equivalent of 30 mg hydro- immediate answer as to whether sudden or gradual with-
cortisone per day orally (>7.5 mg/day prednisolone or drawal of steroid therapy is indicated (Table 15-17).337
>0.75 mg/day dexamethasone) for longer than 3 weeks. In Iatrogenic-induced Cushing syndrome occurs in patients
addition to the magnitude of the dose of glucocorticoid, who take suppressive doses of corticosteroids for longer
the timing of administration may affect the degree of than 3 weeks.175 The rapidity of onset of clinical features
adrenal suppression. If prednisolone is given as 5 mg at depends on the administered dose but can occur within 1
night and 2.5 mg in the morning, there will be more month of therapy.
marked suppression of the HPA axis compared with 2.5 mg
at night and 5 mg in the morning, because the larger
evening dose blocks the early morning surge of ACTH.
Hypoadrenalism During Critical Illness
LaRochelle and colleagues reported that adrenal function Hypoadrenalism may also complicate critical illness, even
recovered if patients’ steroid doses could be tapered to in individuals with a previously intact HPA axis.338 This has
528 SECTION IV Adrenal Cortex and Endocrine Hypertension

TABLE 15-17
Suggested Plan for Steroid Replacement in Patients Withdrawing From Chronic Corticosteroid Therapy
Duration of Glucocorticoid Treatment
Pred Dose (mg/day) ≤3 wk* >3 wk
≥7.5 Can stop ↓ rapidly (e.g., 2.5 mg q3-4d)
THEN
5-7.5 Can stop ↓ 1 mg q2-4 wk OR Convert 5 mg pred to 20 mg HC, then ↓ 2.5 mg/wk to 10 mg/day
THEN THEN
<5 Can stop ↓ 1 mg q2-4 wk After 2-3 mo HC 10 mg/day, administer SST/ITT:
Pass → Withdraw
Fail → Continue

*Beware of frequent steroid courses (e.g., in asthma).


HC, hydrocortisone; ITT, insulin tolerance test; pred, prednisolone; SST, short Synacthen test.

been termed functional adrenal insufficiency to reflect the TABLE 15-18


notion that hypoadrenalism is transient and is not caused Clinical Features of Primary Adrenal Insufficiency
by a structural lesion. Functional adrenal insufficiency has
been difficult to define biochemically and is of uncertain Feature Frequency (%)
cause. Inability to mount an adequate and appropriate Symptoms
cortisol response to overwhelming stress or sepsis is fre- Weakness, tiredness, fatigue 100
quently encountered in intensive care units and substan- Anorexia 100
tially increases the risk of death during acute illness.339 This Gastrointestinal symptoms 92
has stimulated attempts to define functional adrenal insuf- Nausea 86
ficiency quantitatively and to treat it with supplemental Vomiting 75
corticosteroids. Although this diagnosis remains highly Constipation 33
Abdominal pain 31
contentious, if a suboptimal cortisol response is suspected,
Diarrhea 16
the current recommendations suggest (1) treatment with Salt craving 16
hydrocortisone, 200 mg/day in four divided doses or, pref- Postural dizziness 12
erably, 10 mg/hour as a continuous infusion, for patients Muscle or joint pains 13
with septic shock and (2) treatment with methylpredniso-
Signs
lone, 1 mg/kg per day, for patients with severe early acute
respiratory distress syndrome. Glucocorticoid treatment Weight loss 100
should be tapered off rather than stopped abruptly. Treat- Hyperpigmentation 94
Hypotension (<110 mm Hg systolic) 88-94
ment of critical illness–related adrenal insufficiency with Vitiligo 10-20
dexamethasone is not recommended.340 Auricular calcification 5
Laboratory Findings
Clinical Features of Adrenal Insufficiency Electrolyte disturbances 92
Patients with primary adrenal failure usually have both Hyponatremia 88
glucocorticoid and mineralocorticoid deficiency. In con- Hyperkalemia 64
trast, those with secondary adrenal insufficiency have an Hypercalcemia 6
Azotemia 55
intact RAA system. This accounts for differences in salt and Anemia 40
water balance in the two groups of patients, which in turn Eosinophilia 17
result in different clinical presentations. The most obvious
feature that differentiates primary from secondary hypo-
adrenalism is skin pigmentation (Table 15-18), which is
almost always present in cases of primary adrenal insuffi-
ciency (unless of short duration) and absent in secondary hypotension; abdominal, flank, or lower chest pain;
insufficiency. The pigmentation is seen in sun-exposed anorexia; and vomiting. The condition is difficult to diag-
areas, recent rather than old scars, axillae, nipples, palmar nose, but evidence of occult hemorrhage (rapidly falling
creases, pressure points, and mucous membranes (buccal, hemoglobin), progressive hyperkalemia, and shock should
vaginal, vulval, anal). The cause of the pigmentation has alert the clinician to the diagnosis.
long been debated but is thought to reflect increased stimu- Alternatively, the patient may present with vague fea-
lation of the MC1R by ACTH itself. In autoimmune Addison tures of chronic adrenal insufficiency—weakness, tired-
disease, there may be associated vitiligo (Fig. 15-32). ness, weight loss, nausea, intermittent vomiting, abdominal
The clinical features relate to the rate of onset and the pain, diarrhea or constipation, general malaise, muscle
severity of adrenal deficiency.298 In many cases, the disease cramps, arthralgia, and symptoms suggestive of postural
has an insidious onset and a diagnosis is made only when hypotension (see Table 15-18). Salt craving may be a
the patient presents with an acute crisis during an intercur- feature, and a low-grade fever may be present. Supine
rent illness. Acute adrenal insufficiency, termed an adrenal blood pressure is usually normal, but almost invariably
crisis or addisonian crisis, is a medical emergency manifest- there is a fall in blood pressure on standing. Adrenal andro-
ing as hypotension and acute circulatory failure (Table gen secretion is lost; this is clinically more apparent in
15-19). Anorexia may be an early feature; it progresses to women, who may complain of loss of axillary and pubic
nausea, vomiting, diarrhea, and sometimes abdominal hair and frequently have dry and itchy skin. Psychiatric
pain. Fever may be present, and hypoglycemia may occur. symptoms may occur in long-standing cases and include
Patients presenting acutely with adrenal hemorrhage have memory impairment, depression, and psychosis. Formal
CHAPTER 15 The Adrenal Cortex 529

B C

D E
Figure 15-32 Pigmentation in Addison disease. A, Hands of an 18-year-old woman with autoimmune polyendocrine syndrome and Addison disease. Pigmentation in a patient
with Addison disease before (B) and after (C) treatment with hydrocortisone and fludrocortisone. Notice the additional presence of vitiligo. D, Similar changes in a 60-year-old
man with tuberculous Addison disease before (left) and after (right) corticosteroid therapy. E, Buccal pigmentation in the same patient as in D. (B and C, Courtesy of Professor
C.R.W. Edwards.)

TABLE 15-19 quality-of-life measures indicate significant impairment in


Clinical and Laboratory Features of an Adrenal Crisis patients with primary or secondary adrenal insufficiency.341
Tiredness is often profound, and patients may be inappro-
Dehydration, hypotension, or shock out of proportion to severity of priately diagnosed with chronic fatigue syndrome or
current illness anorexia nervosa.
Nausea and vomiting with a history of weight loss and anorexia
In secondary adrenal insufficiency due to hypopituita-
Abdominal pain, so-called acute abdomen
Unexplained hypoglycemia
rism, the presentation may relate to deficiency of hor-
Unexplained fever mones other than ACTH, notably LH/FSH (infertility,
Hyponatremia, hyperkalemia, azotemia, hypercalcemia, or eosinophilia oligorrhea/amenorrhea, poor libido) and TSH (weight gain,
Hyperpigmentation or vitiligo cold intolerance). Fasting hypoglycemia occurs because of
Other autoimmune endocrine deficiencies, such as hypothyroidism or loss of the gluconeogenic effects of cortisol. It is rare in
gonadal failure adults unless there is concomitant alcohol abuse or addi-
tional GH deficiency. However, hypoglycemia is a common
presenting feature of ACTH/adrenal insufficiency in child-
hood.342 In addition, patients with ACTH deficiency present
530 SECTION IV Adrenal Cortex and Endocrine Hypertension

with malaise, weight loss, and other features of chronic short duration and does not include hydrocortisone (which
adrenal insufficiency. Rarely, the presentation is more acute would cross-react in the cortisol assay). A prolonged ACTH
in patients with pituitary apoplexy. stimulation test involving the administration of depot or
intravenous infusions of tetracosactin for 24 to 48 hours
Investigation of Hypoadrenalism differentiates primary from secondary hypoadrenalism. In
normal subjects, the plasma cortisol level at 4 hours is
Routine Biochemical Profile greater than 1000 nmol/L (36 μg/dL); beyond that time,
Among patients with established primary adrenal insuffi- there is no further increase. Patients with secondary hypo-
ciency, hyponatremia is present in about 90% and hyper- adrenalism show a delayed response and usually have a
kalemia in 65%. The blood urea concentration is usually much higher value at 24 and 48 hours than at 4 hours. In
elevated. Hyperkalemia occurs because of aldosterone defi- patients with primary hypoadrenalism, there is no response
ciency, so it is usually absent in patients with secondary at either time. However, the test is rarely required if plasma
adrenal failure. Hyponatremia may be depletional in an ACTH has been appropriately measured at baseline. In
addisonian crisis, but vasopressin levels are elevated, result- primary adrenal insufficiency, the ACTH level is dispropor-
ing in increased free water retention.343 Therefore, in sec- tionately elevated in comparison to plasma cortisol.349
ondary adrenal insufficiency, there may be a dilutional Whereas there is agreement on the investigation of
hyponatremia with normal or low blood urea. suspected primary adrenal failure, the diagnosis of cen-
Reversible abnormalities in liver transaminases fre- tral hypoadrenalism, notably in patients with existing
quently occur. Hypercalcemia occurs in 6% of all cases344 hypothalamic/pituitary disease, is contentious. Based on
and may be particularly marked in patients with coex- correlations with the response of circulating cortisol to
isting thyrotoxicosis. Free thyroxine concentrations are surgery, the insulin-induced hypoglycemia test or insulin
usually low or normal, but TSH values are frequently mod- tolerance test (ITT) was introduced more than 40 years ago
erately elevated.345 This is a direct effect of glucocorticoid as a laboratory test to assess integrity of the HPA axis, and
deficiency and reverses with replacement therapy. Persis- it should be considered the gold standard in this regard.350
tent elevation of TSH in association with positive thyroid It should not be performed in patients with ischemic heart
autoantibodies suggests concomitant autoimmune thy- disease (always check an electrocardiogram before the test),
roid disease. epilepsy, or severe hypopituitarism (i.e., 9 AM plasma cor-
tisol < 180 nmol/L [<6.5 μg/dL]). The test involves the
intravenous administration of soluble insulin in a dose of
Mineralocorticoid Status
0.1 to 0.15 U/kg body weight, with measurement of plasma
In primary hypoadrenalism, mineralocorticoid deficiency cortisol at 0, 30, 45, 60, 90, and 120 minutes. Adequate
usually occurs, manifested by elevated plasma renin hypoglycemia (blood glucose <2.2 mmol/L with signs of
activity and either low or low-normal plasma aldosterone. neuroglycopenia—sweating and tachycardia) is essential.
The investigation of ZG activity is frequently neglected In normal subjects, the peak plasma cortisol concentration
in Addison disease, compared with assessment of ZF func- exceeds 500 nmol/L (18 μg/dL). However, the cortisol
tion. In secondary adrenal insufficiency, the RAA system response to hypoglycemia can be reliably predicted by the
is intact. SST—a safer, cheaper, and quicker test.347,351
The SST relies on the principle that the cortisol response
to an exogenous bolus of ACTH is determined by the
Assessing Adequacy of Function of the HPA Axis endogenous ACTH trophic drive to the adrenal cortex;
Clinical suspicion of the diagnosis should be confirmed impaired ACTH secretion from the anterior pituitary results
with definitive diagnostic tests. Basal plasma cortisol and in an impaired cortisol response after Synacthen adminis-
urinary free cortisol levels are often in the low-normal tration. However, the ACTH test should not be used to
range and cannot be used to exclude the diagnosis. diagnose central hypoadrenalism in patients with a recent
However, a basal cortisol value greater than 400 nmol/L pituitary insult (e.g., surgery, apoplexy). Total hypophysec-
(14.5 μg/dL) invariably indicates an intact HPA axis.346 In tomy results in a failed cortisol response to ITT immedi-
practice, rather than wait for results of insensitive basal ately thereafter, but it takes 2 to 3 weeks for the adrenal
tests, all patients with suspected adrenal insufficiency cortex to readjust to the reduced level of ACTH secretion;
should have an ACTH stimulation test; in patients with in the interim, a false-positive cortisol response is seen. The
an addisonian crisis, however, treatment should be insti- SST should also be avoided in patients with a primary
gated immediately and stimulation tests conducted at a diagnosis of Cushing disease, in whom an exaggerated
later stage. cortisol response to ACTH may persist.
The ACTH stimulation test or SST involves intramuscu- In clinical practice, if the ACTH test is normal, insulin
lar or intravenous administration of 250 μg tetracosactin, hypoglycemia testing is not necessary in most cases unless
a synthetic ACTH(1-24) comprising the first 24 amino acids there is also a need to document endogenous GH reserve
of normally secreted ACTH(1-39).347 Plasma cortisol levels in a patient with pituitary disease. In our practice, an ITT
are measured at 0 and 30 minutes after ACTH administra- is performed in a patient with suspected hypopituitarism
tion, and a normal response is defined by a peak plasma if there is a subnormal response to ACTH. Some patients
cortisol level greater than 550 nmol/L (>20 μg/dL).348 This have an inadequate response to ACTH but then respond
value equates to the 5th percentile response in normal normally to hypoglycemia351; they do not require cortico-
subjects but is very much assay-dependent, with different steroid replacement therapy. This approach is open to
cortisol radioimmunoassays giving different results. Incre- debate, and even taking into account the caveats listed,
mental responses (i.e., the difference between peak and false-positive results have been reported for the SST.352
basal values) are of no value in defining a “pass” response, Although these are rare (<2%), the possibility should be
with the possible exception of diagnosing relative adrenal noted, particularly in patients with ongoing symptoms and
insufficiency in patients with critical illness. Response is signs indicative of hypoadrenalism.
unaffected by the time of day of the test, and the test can A low-dose SST giving only 1 μg ACTH has been pro-
be performed in patients who have commenced cortico- posed as a screen for adequacy of function of the HPA axis,
steroid replacement therapy, as long as this therapy is of with the suggestion that it may be more sensitive than the
CHAPTER 15 The Adrenal Cortex 531

conventional 250-μg test.353-355 Other researchers dispute Mycobacterium tuberculosis should be performed if tubercu-
this suggestion,356,357 and further validation of this test is losis is suspected. CT-guided adrenal biopsy may reveal an
required to support such a concept. underlying diagnosis in patients with suspected malignant
Two other tests have been advocated to assess adequacy deposits in the adrenal gland. Adrenoleukodystrophy can
of function of the HPA axis, but their use in modern clini- be diagnosed by measuring circulating levels of VLCFA.
cal practice should be restricted to difficult diagnostic Finally, appropriate investigations, including pituitary MRI
cases. In the overnight metyrapone test, 30 mg/kg (maxi- scans and an assessment of anterior function, are required
mum, 3 g) metyrapone is given at midnight, and plasma for patients with suspected secondary hypoadrenalism
cortisol and 11-deoxycortisol are measured at 8 AM the fol- who are not taking corticosteroid therapy.
lowing morning. In patients with an intact axis, ACTH
levels rise after the blockade of cortisol synthesis by me-
tyrapone, and a normal result is signified by a peak
Treatment of Acute Adrenal Insufficiency
11-deoxycortisol value greater than 7 μg/dL.358 The CRH Acute adrenal insufficiency is a life-threatening emergency,
stimulation test has been used to diagnose adrenal insuf- and treatment should not be delayed while waiting for
ficiency; unlike the metyrapone test, it differentiates pri- definitive proof of diagnosis (Table 15-20). However, in
mary from secondary causes. Patients with primary adrenal addition to measurement of plasma electrolytes and blood
failure have high ACTH levels that rise further after CRH glucose, appropriate samples for ACTH and cortisol should
stimulation. Patients with secondary adrenal failure have be taken before corticosteroid therapy is given. If the
low ACTH levels that fail to respond to CRH. Patients with patient is not critically ill, an acute ACTH stimulation test
hypothalamic disease show a steady rise in ACTH levels can be performed.
after CRH administration.359 In adults, intravenous hydrocortisone should be given
Testing the HPA Axis During Critical Illness. Many factors in a dose of 100 mg every 6 to 8 hours. If this is not pos-
complicate investigation of the HPA axis during critical sible, then the intramuscular route should be used. In the
illness. Cortisol levels vary broadly with disease severity, patient with shock, 1 L of normal saline should be given
making it difficult to define appropriate responses. Addi- intravenously over the first hour. Because of possible hypo-
tionally, CBG levels decrease substantially, leading to glycemia, it is normal to give 5% dextrose in saline. Sub-
increases in the ratio of free to bound serum cortisol; for sequent saline and dextrose therapy will depend on
this reason, tests that assess the whole axis (e.g., ITT) are biochemical monitoring and the patient’s condition. Clini-
not appropriate in the critical care setting. Investigations cal improvement, especially in the blood pressure, should
are therefore limited to basal cortisol levels or values mea- be seen within 4 to 6 hours if the diagnosis is correct. It is
sured after the SST. important to recognize and treat any associated condition
Recent guidance has indicated that a random cortisol (e.g., infection) that may have precipitated the acute
value of less than 400 nmol/L (<15 μg/dL) suggests corti- adrenal crisis.
costeroid insufficiency, whereas a level greater than After the first 24 hours, the dose of hydrocortisone can
900 nmol/L (>33 μg/dL) is unlikely to occur in patients be reduced, usually to 50 mg intramuscularly every 6 hours
with compromised HPA axis function. For individuals with
intermediate cortisol levels, an SST should be performed; a
cortisol increment of less than 250 nmol/L (<9 μg/dL) is an
independent prognostic marker for death in critically ill TABLE 15-20
patients.339 Treatment of Acute Adrenal Insufficiency (Adrenal Crisis)
An initial multicenter randomized trial of patients with in Adults
septic shock showed that those with an increment less than
250 nmol/L across an SST had a significant improvement Emergency Measures
in survival when given replacement corticosteroids.360 1. Establish intravenous access with a large-gauge needle.
However, in a more recent study hydrocortisone treatment 2. Draw blood for immediate serum electrolytes and glucose and
hastened reversal of shock but did not improve overall routine measurement of plasma cortisol and ACTH. Do not wait for
survival in patients with septic shock.361 The SST was laboratory results.
not useful in predicting benefit from glucocorticoids. It 3. Infuse 2-3 L of 154 mmol/L NaCl (0.9% saline) solution, or 50 g/L
is possible that differences in results between these two (5%) dextrose in 154 mmol/L NaCl (0.9% saline) solution, as
quickly as possible. Monitor for signs of fluid overload by
trials were due to differences in patient selection (e.g.,
measuring central or peripheral venous pressure and listening for
severity of sepsis) and the speed of administration of pulmonary rales. Reduce infusion rate if indicated.
glucocorticoids. 4. Inject intravenous hydrocortisone (100 mg immediately and every
In light of this uncertainty, recent recommendations 6 hr).
continue to suggest hydrocortisone treatment for septic 5. Use supportive measures as needed.
shock and methylprednisolone for patients with severe Subacute Measures After Stabilization of the Patient
early acute respiratory distress syndrome, particularly those
with poor response to fluid resuscitation and vasopressor 1. Continue intravenous 154 mmol/L NaCl (0.9% saline) solution at a
slower rate for next 24-48 hr.
agents.340 The role of glucocorticoids in the management
2. Search for and treat possible infectious precipitating causes of the
of critically ill patients with other conditions requires adrenal crisis.
further research. 3. Perform a short ACTH stimulation test to confirm the diagnosis of
Other Tests. Radioimmunoassays to detect autoantibodies adrenal insufficiency (if patient does not have known adrenal
such as those against the 21-hydroxylase antigen are now insufficiency).
available and should be analyzed in patients with primary 4. Determine the type of adrenal insufficiency and its cause, if not
adrenal failure. In autoimmune Addison disease, it is also already known.
important to look for evidence of other organ-specific auto- 5. Taper glucocorticoids to maintenance dosage over 1-3 days, if
immune disease. A CT scan may reveal enlarged or calcified precipitating or complicating illness permits.
6. Begin mineralocorticoid replacement with fludrocortisone (0.1 mg
adrenals, suggesting an infective, hemorrhagic, or malig-
by mouth daily) when saline infusion is stopped.
nant diagnosis (Fig. 15-33). Chest radiography, tuberculin
testing, and early morning urine samples cultured for ACTH, adrenocorticotropic hormone.
532 SECTION IV Adrenal Cortex and Endocrine Hypertension

A B

C D
Figure 15-33 Computed tomographic (CT) scans of patients with primary adrenal insufficiency. The affected adrenal glands are indicated by arrows. A, CT scan of a 59-year-
old man with histoplasmosis. Notice the subcapsular calcium in both glands. B, CT scan of a 59-year-old man with metastatic melanoma. C, CT scan of an 80-year-old man
with bilateral adrenal hemorrhage resulting from anticoagulation for pulmonary emboli. D, Bilateral adrenal tuberculomas in a 79-year-old man with tuberculosis affecting the
urogenital tract. (A and B, Courtesy of Dr. William D. Salmon, Jr.; C, Courtesy of Dr. Craig R. Sussman.)

and then to oral hydrocortisone, 40 mg in the morning hydrocortisone,365 highlighting the need to strive for mini-
and 20 mg at 6 PM. This dose can then be rapidly reduced mally effective but safe doses.366,367 Possibly because of the
to a more standard replacement dose of 20 mg on awaken- known action of IGF-1 to increase cortisol clearance,132 it
ing and 10 mg at 6 PM. is our experience that glucocorticoid requirements are
slightly lower in hypopituitary, GH-deficient subjects than
in patients with primary adrenal insufficiency.
Long-Term Replacement Therapy In primary adrenal failure, mineralocorticoid replace-
The aim of long-term therapy is to give replacement doses ment is usually also required in the form of fludrocortisone
of hydrocortisone to mimic the normal cortisol secretion (or 9α-fluorinated hydrocortisone), 0.05 to 0.2 mg/day.
rate (Table 15-21). In the past, this rate was thought to be The mineralocorticoid activity of this is about 125 times
approximately 25 to 30 mg/day, but stable isotope studies that of hydrocortisone. After the acute phase has passed,
have indicated lower normal cortisol production rates of 8 the adequacy of mineralocorticoid replacement should be
to 15 mg/day.362 Most patients can cope with less than assessed by measuring electrolytes, supine and erect blood
30 mg/day (usually 15 to 25 mg/day in divided doses). pressures, and plasma renin activity.368 Too little fludrocor-
Doses are usually given on awakening, with a smaller dose tisone may cause postural hypotension with elevated
at 6 PM, but some patients feel better with three-times-a- plasma renin activity, whereas too much causes the con-
day dosing. In cases of primary adrenal failure, cortisol verse. Mineralocorticoid replacement therapy is all too fre-
day curves with simultaneous ACTH measurements are quently neglected in patients with adrenal failure.
advocated to provide some insight into the adequacy of Patients on glucocorticoid replacement therapy should
replacement therapy.363 There are no good biomarkers of be advised to double their daily dose in the event of inter-
glucocorticoid adequacy in patients with central hypo- current febrile illness, accident, or mental stress such as an
adrenalism. Decisions regarding doses of replacement ther- important examination. If the patient is vomiting and
apy are largely based on crude yet important end points cannot take medication by mouth, parenteral hydrocorti-
such as weight, well-being, and blood pressure.364 Bone sone must be given urgently. For minor surgery, 50 to
mineral density is moderately reduced in a dose-dependent 100 mg hydrocortisone hemisuccinate is given with the
manner in patients treated with more than 25 mg/day of premedication. For major operations, this pretreatment is
CHAPTER 15 The Adrenal Cortex 533

TABLE 15-21 For patients with both primary and secondary adrenal
Treatment of Chronic Primary Adrenal Insufficiency in Adults
failure, beneficial effects of adrenal androgen replacement
therapy with 25 to 50 mg/day of DHEA have been reported.
Maintenance Therapy To date, reported benefit is principally confined to female
Glucocorticoid Replacement patients and includes improvement in sexual function
• Hydrocortisone 15-20 mg on awakening and 5-10 mg in early and well-being.369 However, patients with adrenal insuffi-
afternoon ciency on current steroid replacement regimens have
• Monitor clinical symptoms and morning plasma ACTH. significantly impaired health-related subjective health
status irrespective of the origin of disease or concomitant
Mineralocorticoid Replacement disease.341 Delayed-release hydrocortisone preparations,
• Fludrocortisone 0.1 (0.05-0.2) mg orally such as Plenadren, that more closely replicate normal cir-
• Liberal salt intake
• Monitor lying and standing blood pressure and pulse, edema,
cadian cortisol concentrations, have recently been licensed
serum potassium, and plasma renin activity. and approved; early clinical trials show improved quality
• Educate patient about the disease, how to manage minor illnesses of life in both primary and central hypoadrenalism com-
and major stresses, and how to inject steroid intramuscularly. pared to conventional twice- or thrice-daily hydrocortisone
• Obtain MedicAlert bracelet/necklace, Emergency Medical administration.369a
Information card.
Treatment of Minor Febrile Illness or Stress
• Increase glucocorticoid dose twofold to threefold for the few days
CONGENITAL ADRENAL HYPERPLASIA
of illness; do not change mineralocorticoid dose.
• Contact physician if illness worsens or persists for more than 3 CAH comprises a group of autosomal recessive disorders
days or if vomiting develops. caused by deficient adrenal corticosteroid biosynthe-
• No extra supplementation is needed for most uncomplicated, sis.370,371 It results from defects in one of the steroidogenic
outpatient dental procedures with local anesthesia. General enzymes involved in cortisol biosynthesis or in the electron-
anesthesia or intravenous sedation should not be used in the providing factor, POR. Congenital lipoid adrenal hyperpla-
office. sia, caused by StAR deficiency affecting mitochondrial
Emergency Treatment of Severe Stress or Trauma cholesterol uptake, is a subform of this disease complex
with the unique feature of lipid accumulation leading to
• Inject contents of prefilled dexamethasone (4-mg) syringe
intramuscularly. cell destruction. In each case, there is reduced negative
• Get to physician as quickly as possible. feedback inhibition of cortisol and, depending on the ster-
oidogenic pathway involved, alteration in adrenal miner-
Steroid Coverage for Illness or Surgery in Hospital
alocorticoid and androgen secretion (Table 15-22).
• For moderate illness, give hydrocortisone 50 mg bid PO or IV. Aldosterone synthase deficiency does not affect gluco-
Taper rapidly to maintenance dose as patient recovers. corticoid biosynthesis and does not lead to adrenal hyper-
• For severe illness, give hydrocortisone 100 mg IV q8h. Taper to plasia, but it has been historically grouped into this disease
maintenance level by decreasing by half every day. Adjust dose
complex. All forms of CAH together represent a disease
according to course of illness.
• For minor procedures under local anesthesia and most radiologic continuum, ranging from severe forms caused by complete
studies, no extra supplementation is needed. loss-of-function defects to milder forms in which the defec-
• For moderately stressful procedures such as barium enema, tive proteins have partial residual activity.
endoscopy, or arteriography, give a single 100-mg IV dose of
hydrocortisone just before the procedure.
• For major surgery, give hydrocortisone 100 mg IV just before 21-Hydroxylase Deficiency
induction of anesthesia and continue q8h for first 24 hr. Taper dose Between 90% and 95% of cases of CAH are caused by
rapidly, decreasing by half per day, to maintenance level. 21-hydroxylase deficiency.370 In Western societies, the inci-
ACTH, adrenocorticotropic hormone; bid, twice a day; IV, intravenous; PO, dence varies from 1 in 10,000 to 1 in 15,000 live births,
orally; q, every. but in isolated communities the incidence may be much
higher (e.g., 1 : 300 in Alaskan Inuit populations). Nonclas-
sic CAH is more common, with an incidence of about 1 in
500 to 1 in 1000 live births. The condition arises because
followed by the same regimen as for acute adrenal insuf- of defective conversion of 17α-OHP to 11-deoxycortisol.
ficiency (see Table 15-21). Pregnancy proceeds normally in Reduced cortisol biosynthesis results in reduced negative
patients taking replacement therapy, but daily doses of feedback drive and increased ACTH secretion; as a conse-
hydrocortisone are usually increased modestly (5 to 10 mg/ quence, adrenal androgens are produced in excess (Fig.
day) in the last trimester. Progesterone is a mineralocorti- 15-34). Seventy-five percent of patients have clinically
coid antagonist, and the rising levels across pregnancy may manifest mineralocorticoid deficiency because of failure to
necessitate an increased dose of fludrocortisone. During convert sufficient progesterone to DOC in the ZG. Clini-
labor, patients should be well hydrated with a saline drip cally, several distinct variants of 21-hydroxylase deficiency
and should receive hydrocortisone 50 mg intramuscularly have been recognized (Table 15-23).
every 6 hours until delivery. Thereafter, doses can be rapidly
tapered to prepregnancy levels.
Every patient on glucocorticoid therapy should be
Simple Virilizing Form
advised to register for a medical alert bracelet or necklace In the simple virilizing form of 21-hydroxylase deficiency,
and to carry a steroid card. Patients should receive regular the enhanced ACTH drive to adrenal androgen secretion
education regarding the requirements of stress-related glu- in utero leads to virilization of an affected female fetus.
cocorticoid dose adjustment, which should involve the Depending on the severity, clitoral enlargement, labial
patient’s partner and family as well. Parenteral prepara- fusion, and development of a urogenital sinus may occur,
tions of hydrocortisone for self-administration may be leading to sexual ambiguity at birth and even inappropri-
required for patients living far from hospitals and those ate sex assignment. Males are phenotypically normal at
planning vacations. birth and are at risk of not being diagnosed; this explains
TABLE 15-22
534

Congenital Adrenal Hyperplasia: Features for Each Enzyme Defect


Lipoid P450
11β- 17α- Adrenal Side-Chain Aldosterone Apparent Cortisone
Deficiency 21-Hydroxylase Hydroxylase Hydroxylase 3β-HSD Type 2 P450 Oxidoreductase Hyperplasia Cleavage Synthase Reductase
OMIM No. +201910 #202010 #202110 +201810 #201750 *600617 +118485 *124080 *138090
Gene/Protein CYP21A2 CYP11B1 CYP17A1 HSD3B2 POR StAR CYP11A1 CYP11B2 H6PDH
Alias P450c21 P450c11 P450c17 3β-HSD CPR, CYPOR P450scc P450aldo
Incidence Classic: 1 : 10,000 to 1 : 100,000 to Rare Rare Unknown Rare Rare Rare Rare
1 : 15,000 1 : 200,000
Nonclassic: 1 : 500 to
1 : 1000
DSD Classic: 46,XX 46,XX 46,XY 46,XY* 46,XX + 46,XY† 46,XY 46,XY No No
Nonclassic: No
Primary affected Adrenal Adrenal Adrenal, gonads Adrenal, gonads Adrenal, gonads, Adrenal, Adrenal, Adrenal Liver, adrenal, all
organ liver, all CYP type gonads gonads H6PDH/HSD11B1–
2–expressing expressing tissues
tissues
Glucocorticoids Classic: Reduced Reduced Reduced Reduced Reduced to normal, Reduced Reduced Normal Normal, but reduced
Nonclassic: Normal impaired stress tissue levels due
response to increased
cortisol clearance
SECTION IV Adrenal Cortex and Endocrine Hypertension

Mineralocorticoids Classic: Reduced in SW Increased, Increased Reduced often Reduced to Reduced Reduced Reduced Normal
Nonclassic: Normal mainly increased
precursors
Sex hormones Increased Increased Reduced Reduced in males, Reduced Reduced Reduced Normal Increased
increased in
females‡
Increased marker 17-OHP, 21-DOF DOC, S Pregnenolone, 17-OH, pregneno- Pregnenolone, DOC, B,
metabolites in progesterone, lone, DHEA progesterone, 18-OHB
plasma DOC, S 17-OHP
Increased marker Pregnanetriol, 17-OH THDOC, THS THDOC, THB, Pregnanetriol Pregnenediol,
metabolites in pregnenolone, pregnenediol, pregnanediol,
urine pregnanetriolone pregnanediol pregnanetriol,
17-OH
pregnanolone
PRA Classic: Increased Reduced Reduced Increased Increased Increased Increased Normal
Nonclassic: Normal to
mildly increased
Hypertension No Yes Yes No No or mild No No No No
Plasma sodium Classic: Reduced in SW Increased Increased Reduced in SW Normal Reduced Reduced Reduced Normal
Nonclassic: Normal
Plasma potassium Classic: Increased in SW Reduced Reduced Increased in SW Normal Increased Increased Increased Normal
Nonclassic: Normal
Urinary salt loss Classic: Yes No No Yes No Yes Yes Yes No
Nonclassic: No
Skeletal No No No No Yes§ No No No No
malformation

*Masculinization of the external genitalia in females at birth is rare and usually mild; signs of increased androgens usually manifest later.

DSD is observed in both sexes, and normal sex-specific sexual development is also reported.

Steroid hormone conversion by HSD3B1 in peripheral tissues.
§
In most cases published thus far; however, absence of skeletal malformations does not rule out POR deficiency.
B, corticosterone; CYP, cytochrome P450; DHEA, dehydroepiandrosterone; DOC, 11-deoxycorticosterone; 21-DOF, 21-deoxycortisol; DSD, disorder of sex development; H6PDH, hexose-6-phosphate dehydrogenase;
HSD, hydroxysteroid dehydrogenase; OMIM, Online Mendelian Inheritance in Man; 18-OHB, 18-hydroxycorticosterone; 17-OHP, 17-hydroxyprogesterone; POR, P450 oxidoreductase; PRA, plasma renin activity;
S, 11-deoxycortisol; StAR, steroidogenic acute regulatory protein; SW, salt wasting; THB, tetrahydro-corticosterone; THS, tetrahydro-11-deoxycortisol; THDOC, tetrahydro-11-deoxycorticosterone.
CHAPTER 15 The Adrenal Cortex 535

Cholesterol

StAR

Cholesterol

CYP11A1
CYP17A1 CYP17A1
Pregnenolone 17-OH Pregnenolone DHEA

HSD3B2

Progesterone 17-OH Progesterone Androstenedione

(Blocked at) (Blocked at)


CYP21A2 CYP21A2

DOC 11-Deoxycortisol

CYP11B2 CYP11B1 CYP11B1

Corticosterone Cortisol

CYP11B2

18-OH Corticosterone

CYP11B2

Aldosterone

Mineralocorticoid Glucocorticoid Androgens


Figure 15-34 Congenital adrenal hyperplasia related to 21-hydroxylase deficiency. The normal synthesis of cortisol is impaired, and adrenocorticotropic hormone (ACTH)
levels increase because of loss of normal negative feedback inhibition, resulting in an increase in adrenal steroid precursors proximal to the block. The results are cortisol
deficiency, variable mineralocorticoid deficiency, and excessive secretion of adrenal androgens. CYP, cytochrome P450; DHEA, dehydroepiandrosterone; DOC, deoxycorticoster-
one; HSD, hydroxysteroid dehydrogenase; StAR, steroidogenic acute regulatory protein.

TABLE 15-23
Forms of 21-Hydroxylase Deficiency
Phenotype Classic Salt Wasting Simple Virilizing Nonclassic
Age at diagnosis Newborn to 6 mo Female: Newborn to 2 yr Child to adult
Male: 2-4 yr
Genitalia Female: Ambiguous Female: Ambiguous Female: Virilized
Male: Normal Male: Normal Male: Normal
Incidence 1 : 20,000 1 : 60,000 1 : 1000
Hormones
Aldosterone Reduced Normal Normal
Renin Increased Normal or increased Normal
Cortisol Reduced Reduced Normal
17-OHP >5000 ng/dL 2500-5000 ng/dL 500-2500 ng/dL (ACTH stimulation)
Testosterone Increased Increased Variable, increased
Growth −2 to −3 SD −1 to −2 SD Probably normal
21-Hydroxylase activity (% of wild type) 0 1-5 20-50
Typical CYP21A2 mutations Deletions, conversions, nt656g I172N V281L
G110Δ8nt, R356W Intron 2 splice site (nt656g) P30L
I236N, V237E, M239K, Q318X

ACTH, adrenocorticotropic hormone; 17-OHP, 17-hydroxyprogesterone; SD, standard deviation.

the skewed female-to-male ratio of simple virilizing CAH Salt-Wasting Form


diagnosed in the preneonatal screening era. Such patients
may present in early childhood with signs of precocious Seventy-five percent of patients of both sexes who have the
pseudopuberty such as sexual precocity, pubic hair devel- salt-wasting form of 21-hydroxylase deficiency also have
opment, or growth acceleration due to premature andro- concomitant, clinically manifested aldosterone deficiency.
gen excess. If left untreated, this stimulates premature In addition to the described features, neonates commonly
epiphyseal closure, and final adult height is invariably present after the first 2 weeks of life with a salt-wasting
diminished.372,373 crisis and hypotension. The clinical signs and symptoms
536 SECTION IV Adrenal Cortex and Endocrine Hypertension

of salt wasting include poor feeding, vomiting, failure to recovery from glucocorticoid and mineralocorticoid defi-
thrive, lethargy, and sepsis-like symptoms. These features ciency during later life might be explained by significant
may alert the clinician to the diagnosis in a male baby, but 21-hydroxylase activity of the cytochrome P450 enzymes
the diagnosis is still delayed in many cases, and the condi- CYP2C19 and CYP3A4.383
tion carries a significant neonatal mortality rate.
Diagnostic Criteria
Nonclassic or Late-Onset 21-Hydroxylase Deficiency A diagnosis of 21-hydroxylase deficiency should be consid-
Patients with nonclassic 21-hydroxylase deficiency present ered in any newborn infant with genital ambiguity and salt
in childhood or early adulthood with premature pubarche wasting, hypotension, or hypoglycemia. Hyponatremia
or with a phenotype that may masquerade as polycystic and hypokalemia with raised plasma renin activity are
ovary syndrome (PCOS).370,374,375 Indeed, nonclassic CAH is found in salt-wasters. In later life, adrenal androgen excess
a recognized secondary cause of PCOS and appears to be (DHEAS, androstenedione) is found in patients presenting
more common than the classic variant. Recent evidence with sexual precocity or a PCOS-like phenotype. Randomly
suggests that at least 30% of adult patients have an impaired timed measurements of the plasma 17-OHP concentration
cortisol response to ACTH(1-24)376 and may be prone to are significantly increased in classic 21-hydroxylase defi-
stress-induced adrenal insufficiency. Routine assessment of ciency. Commonly, 17-OHP concentrations in patients
adrenal glucocorticoid reserve is indicated. In some series with salt-wasting CAH are higher than in non–salt-losing
from tertiary referral centers, nonclassic 21-hydroxylase patients.
deficiency accounts for up to 12% of all PCOS patients, but In nonclassic CAH, an SST is required to establish normal
more realistic prevalence rates are probably 1% to 3%.377 adrenal glucocorticoid reserve. Clinically useful nomo-
Females present with hirsutism, primary or secondary grams have been developed that compare circulating
amenorrhea, or anovulatory infertility.374 Androgenic alo- concentrations of 17-OHP before and 60 minutes after
pecia and acne may be other presenting features. exogenous ACTH administration to investigate borderline
cases and to differentiate between nonclassic CAH and
heterozygous carriers (Fig. 15-36).384 This separates patients
Heterozygote 21-Hydroxylase Deficiency with classic and nonclassic 21-hydroxylase deficiency from
Salt wasting, simple virilizing, and late-onset 21-hydroxylase heterozygote carriers and normal subjects, but there is
deficiency are all caused by homozygous or compound some overlap between values seen in heterozygotes and in
heterozygote mutations in the human 21-hydroxylase normal subjects. 17-OHP is measured basally and then 60
gene (CYP21A2). In the carrier or heterozygote state, only minutes after administration of 250 μg Synacthen. Stimu-
one allele is mutated. The clinical significance of the het- lated values are invariably grossly elevated (>35 nmol/L
erozygote state is uncertain; it does not appear to disadvan- [>11 μg/L]) in patients with classic and nonclassic forms of
tage reproductive capability but may cause signs of the disorder. Heterozygote patients usually have stimulated
hyperandrogenism in adult women.370 values between 10 and 30 nmol/L (330 and 1000 ng/dL)
(see Fig. 15-36). Stimulation tests are not always required
to make a diagnosis; for example, a basal 17-OHP concen-
Molecular Genetics
tration of less than 5 nmol/L (<150 ng/dL) in the follicular
21-Hydroxylase deficiency is inherited as an autosomal phase of the menstrual cycle effectively excludes late-onset
recessive trait, and the higher incidence of the condition 21-hydroxylase deficiency.374 CYP21A2 genotyping to
in some ethnic communities almost certainly relates to confirm the clinical and biochemical diagnosis is a useful
consanguinity. The CYP21A2 gene and its highly homolo- adjunct to hormonal measurements. Androgen excess in
gous pseudogene (CYP21A1P) are located on the short arm 21-hydroxylase deficiency is readily suppressed after gluco-
of chromosome 6 (6p21.3). Because of the genomic local- corticoid administration.
ization within the human leukocyte antigen (HLA) locus, Prenatal diagnosis of 21-hydroxylase deficiency has
a region with a high frequency of genomic recombinations, been advocated, because treatment of an affected female
most of the mutations causing 21-hydroxylase deficiency may prevent masculinization in utero.385 17-OHP can be
are generated by gene conversion events. Complete gene assayed in amniotic fluid, but the most robust approach is
deletions or conversions of the CYP21A2 gene, eight the rapid genotyping of fetal cells obtained by chorionic
pseudogene-derived point mutations, and an 8–base pair villous sampling in early gestation. In patients with known
deletion are found in more than 95% of cases. Other rare 21-hydroxylase deficiency (male or female) seeking fertil-
pseudogene-independent CYP21A2-inactivating mutations ity, determination of 17-OHP levels across an SST in the
have been reported in single families or small populations. partner before conception will uncover nonclassic or het-
Approximately 65% to 75% of CAH patients are compound erozygote cases and provide the endocrinologist/geneticist
heterozygous for the disease-causing mutations.378 with some assignment of risk before pregnancy.
The genotype-phenotype correlation in CAH due to the
21-hydroxylase deficiency is well established. The clinical
phenotype correlates with the less severely mutated allele
Treatment
and, consequently, with the residual 21-hydroxylase activ- The objectives for treatment of 21-hydroxylase deficiency
ity (Fig. 15-35).379,380 This correlation appears to be high, differ with age, but at all ages treatment and overall patient
although divergence between genotype and phenotype has management can be fraught with difficulties. In childhood,
been observed.381 The 21-hydroxylase activity measured by the overall goal is to replace glucocorticoid and mineralo-
in vitro analysis provides a possibility for estimating dis- corticoid, thereby preventing further salt-wasting crises,
ease severity, although some phenotypic variability (e.g., but also to normalize adrenal androgen secretion so that
salt wasting, age at onset) seems likely to depend on other normal growth and skeletal maturation can proceed. Accu-
interacting genes and maturation processes rather than rate replacement is essential; in excess, glucocorticoids will
CYP21A2 itself. One such factor might be the length of suppress growth, whereas inadequate replacement will
the CAG repeats in the androgen receptor modulating result initially in accelerated linear growth and ultimately
androgen action.382 Potential variations in the degree of in short stature due to premature epiphyseal closure.370
CHAPTER 15 The Adrenal Cortex 537

A
Telomere Centromere

TNXA TNXB CREBL1

CYP21A1P RP2 CYP21A2


LTA RP1 C4A C4B

B
CYP21A2
5′ 1 2 3 4 5 6 7 8 9 10 3′

E6 cluster
P453S
in2 splice

F306 +t

R356W
Q318X
V281L
I172N
∆8 bp
P30L

CYP21A1P
5′ 1 2 3 4 5 6 7 8 9 10 3′

C Gene del/conversion
∆8 bp
E6 cluster
F306 +t
Q318X
R356W
0% Intron 2 splice site

0-1% I172N

1-5% P30L
V281L
20-30% P453S
Disease severity
21-hydroxylase deficiency 30-50%
Mutation group Null A B C
CAH form Salt wasting Simple virilizing Nonclassic
Positive predictive values
Speiser et al., 1992 96% 85% 73% 63%
Krone et al., 2000 100% 90% 74% 65%
Stikkelbroeck et al., 2003 97% 96% 53% 100%
Prader genital stages
Speiser et al., 1992 II-IV (IV) III-V (IV) I-IV (III) 0-IV (0)
Wedell et al., 1994 III-IV II-V I-IV
Jääskeläinen et al., 1997 II-V 0-V II-V 0
Krone et al., 2000 III-V (IV) II-V (IV) II-V (III) 0-IV (0)
Figure 15-35 Genetics of 21-hydroxylase deficiency. A, Genomic organization of the functional CYP21A2 gene and its nonfunctional CYP21A1P pseudogene. B, Nine out of
10 common mutations are transferred by microconversions from the CYP21A1P pseudogene into the CYP21A2 gene. C, Genotype-phenotype correlation in 21-hydroxylase
deficiency is well established. Based on the in vitro enzyme activity, the CYP21A2 gene-inactivating mutations can be categorized into four major mutation groups. Although
variation has been reported for the milder mutations, the overall correlation is high regarding expression of the adrenal phenotype. Considerable variability exists for the correla-
tion with genital virilization. CAH, congenital adrenal hyperplasia.

Response is best monitored through growth velocity and growth, and skeletal maturation indicate inadequate or
bone age, with biochemical markers from blood (17-OHP, excessive glucocorticoid treatment.386
androstenedione, testosterone), urine, and saliva (17-OHP, Corrective surgery (e.g., clitoral reduction, vaginoplasty)
androstenedione, testosterone) being useful adjuncts. In is frequently required during childhood. The method of
difficult cases, a day curve study, as described for patients choice should be a one-stage complete repair using the
with primary adrenal failure but measuring the ACTH and newest techniques of vaginoplasty, clitoral, and labial
17-OHP response before and after corticosteroid replace- surgery.386
ment, may confirm overreplacement or underreplacement. In late childhood and adolescence, appropriate replace-
The optimal glucocorticoid dose fails to suppress 17-OHP ment therapy is equally important. Overtreatment may
and its metabolites and maintains sex hormone concentra- result in obesity and delayed menarche/puberty with
tions in the middle of the age- and sex-specific normal sexual infantilism, whereas underreplacement will result in
range. Ideally, the biochemical investigations will indicate sexual precocity. Compliance with regular medication is
the need for dose adjustments before physical changes, often an issue throughout adolescence.
538 SECTION IV Adrenal Cortex and Endocrine Hypertension

ng/dL Log10 replacement of adrenal insufficiency because treatment


105 5 also aims at normalization of ACTH-driven adrenal andro-
17-OHP 60 min gen excess. The optimal timing for providing the highest
dose of hydrocortisone remains an ongoing matter of
17-OH progesterone

c debate, with no data supporting either circadian replace-


ment (giving the highest dose in the morning) or reverse-
phase therapy (giving the largest dose of hydrocortisone
at night). Long-acting steroids such as prednisone, pred-
104 4 nisolone, and dexamethasone are more effective in
v this regard but should not be given before the end of
puberty in order to avoid oversuppression and reduction
in linear growth.
Stimulated

Fludrocortisone is required for patients with salt wasting


General population (although this may spontaneously improve with age).
Genetically unaffected Fludrocortisone doses during the first year of life are com-
Patients with:
Congenital adrenal monly 150 μg/m2 per day. Sodium needs to be supple-
103 3
hyperplasia mented, as milk feeds provide only maintenance sodium
h Aquired adrenal requirements. Adequate mineralocorticoid replacement
hyperplasia usually leads to hydrocortisone dose reduction. The rela-
p Cryptic 21-OH
deficiency tive dose in relation to body surface decreases throughout
u Heterozygotes for: life. Fludrocortisone doses of 100 μg/m2 per day after the
Congenital adrenal first 2 years of life are commonly sufficient. This require-
hyperplasia ment drops further with adolescence and adulthood to a
Aquired adrenal
102 2
hyperplasia daily dose of 100 to 200 μg (50 to 100 μg/m2 per day).
Cryptic 21-OH Mineralocorticoid substitution is monitored by measure-
deficiency ments of plasma renin activity (low or suppressed levels
indicating overtreatment) and blood pressure.390
Log10 1 2 3 4 5
Adrenomedullary dysplasia has been reported in the
10 102 103 104 105 CAH adrenal gland, probably because of relative glucocor-
Baseline 17-OH progesterone ng/dL ticoid deficiency, which results in epinephrine deficiency.391
Figure 15-36 Basal and stimulated plasma 17α-hydroxyprogesterone (17-OHP) Benefits of epinephrine replacement on the metabolic
concentrations in patients with 21-hydroxylase (21-OH, CYP21A2) deficiency. To response to exercise have been reported in children,392 but
convert values to nmol/L, multiply by 0.0303. The mean for each group is indicated further studies are required before routine catecholamine
by a large cross and an adjacent letter: c, patients with classic CYP21A2 deficiency; replacement therapy can be advocated. In clinical practice,
h, heterozygotes for all forms of CYP21A2 deficiency; p, general population; u, known sufficient supplementation of glucose during exercise and
unaffected persons (e.g., siblings of patients with CYP21A2 deficiency who carry illness should be guaranteed, to prevent hypoglycemic epi-
neither affected parental haplotype as determined by human leukocyte antigen sodes. Bilateral adrenalectomy is effective but should be
typing); v, patients with nonclassic (acquired and cryptic) CYP21A2 deficiency. (From regarded as a last resort393; in addition, because of the
White PC, New MI, Dupont B. Congenital adrenal hyperplasia: part 1. N Engl J Med.
1987;316:1519-1524.)
requirement for lifelong corticosteroid replacement ther-
apy, patients could also develop feedback ACTH-secreting
pituitary tumors.394 This procedure also bears a number of
risks, including surgical and anesthetic complications, and
the patient is left completely adrenal insufficient.
Although much has been written about adequate control Prenatal dexamethasone treatment is effective to avoid
in childhood, adults with CAH often provide an ongoing virilization of the external genitalia in the female fetus.
dilemma for the endocrinologist. The follow-up of such Unlike hydrocortisone, which is inactivated by placental
patients should involve multidisciplinary clinics, initially 11β-HSD, maternally administered dexamethasone can
with transition adolescence clinics to facilitate transfer cross the placenta to suppress the fetal HPA axis. One
from pediatric to adult care. Problems in adulthood relate approach is to advocate use of dexamethasone therapy as
to fertility concerns, hirsutism, and menstrual irregularity soon as pregnancy is confirmed in high-risk cases and to
in women; obesity, metabolic consequences, and impact of continue this therapy until the diagnosis is excluded in the
short stature; probable increased cardiovascular risk; sexual female fetus. If the fetus is affected, only those of female
dysfunction; and psychological problems.370,387,388 Counsel- sex require dexamethasone therapy across gestation.
ing is often required in addition to endocrine support. Therapy must be instigated at 6 to 7 weeks of gestation to
Males may develop enlargement of the testes due to be effective. The suggested dexamethasone dose is 20 to
so-called testicular adrenal rest tumors—that is, ectopic 25 μg/kg in three divided doses per day (total maximum
adrenal tissue, which may regress after glucocorticoid sup- dose, 1.5 mg/day).370 However, because only one in eight
pression. These patients need adequate endocrine therapy pregnancies treated in this way will result in an affected
rather than urologic referral with ensuing risk of removal female fetus, the use of steroid therapy in this setting has
of testis mistaken for a tumor.389 been questioned.395 The fetal sex can be determined as early
In the absence of any evidence-based data, there are no as week 6 of gestation with the use of novel molecular
prescriptive steroid regimens to treat patients with CAH at diagnostic methods that analyze free fetal DNA from
any age, and as a result, many individualized regimens are maternal blood using real-time polymerase chain reaction.
used in clinical practice. Hydrocortisone is recommended In this way, the number of unnecessarily treated cases can
for replacement therapy from the newborn period to ado- be reduced to three out of eight. Dexamethasone can lead
lescence.386 Usual starting doses of hydrocortisone in to maternal cushingoid effects in pregnancy396 and may in
childhood are 10 to 15 mg/m2 per day in three divided turn have long-term, deleterious effects on the fetus,
doses, with up to 25 mg/m2 in infancy only seldom including metabolic, psychological, and intellectual conse-
required. These doses are higher than those employed for quences. Prenatal treatment is controversial and has to be
CHAPTER 15 The Adrenal Cortex 539

regarded as experimental; patients treated should be almost absent 11β-hydroxylase enzyme activity, with only
included in ongoing multicenter studies.397 some cases of mild or nonclassic 11-hydroxylase deficiency
In adult women with hyperandrogenism and untreated reported.407,408
nonclassic CAH, there is no evidence that final height is Loss of negative cortisol feedback and enhanced ACTH-
affected. In this setting, glucocorticoid suppression in isola- mediated adrenal androgen excess occur in 11β-hydroxylase
tion rarely controls hirsutism, and additional antiandrogen deficiency (Fig. 15-37). Clinical features therefore are very
therapy is often required (e.g., cyproterone acetate, spi- similar to those reported in the simple virilizing form of
ronolactone, flutamide together with an oral estrogen con- CAH (46,XX DSD including virilization of the external
traceptive pill). However, ovulation induction rates with genitalia and sexual ambiguity); and again, milder cases
gonadotropin therapy are improved after suppression of can manifest later in childhood or even young adulthood.
nocturnal ACTH levels with 0.25 to 0.5 mg dexametha- The principal difference from 21-hydroxylase deficiency is
sone. Hypogonadotropic hypogonadism in male patients hypertension, which is thought to be secondary to the
is a consequence of increased aromatization of adrenal mineralocorticoid effect of DOC excess. However, there is
androgens, in particular androstenedione to estrone, result- a poor correlation between DOC secretion and the pres-
ing in suppression of pituitary LH and FSH secretion. The ence of hypertension, and unexplained salt wasting has
condition is reversible after optimization of glucocorticoid been reported in few patients during early life. On this
therapy. However, overreplacement in men or women may clinical background, the diagnosis can be made by measur-
also lead to hypogonadotropic hypogonadism due to ing a plasma ACTH–stimulated 11-deoxycortisol value,
glucocorticoid-mediated suppression of GnRH secretion. which will be higher than three times the 95th percentile
for an age-matched normal group. Basal concentrations of
17-OHP are commonly increased but may be normal even
Long-Term Complications and Comorbid Conditions during the first weeks of life.409
Outcome assessed by final height is not optimal in many Although established heterozygotes may not demon-
patients treated for 21-hydroxylase deficiency. In a meta- strate an increase in 11-deoxycortisol above normal values
analysis including 18 studies published between 1977 and after Synacthen stimulation410 (unlike the 17-OHP response
2001, the mean adult height of patients with classic CAH observed in heterozygote patients with 21-hydroxylase
was 10 cm (−1.4 SDS) below the population mean and −1.2 deficiency), exaggerated ACTH-stimulated responses have
SDS calculated for target height. The pubertal growth spurt been observed in patients with hirsutism411 and in patients
occurs earlier and is less pronounced than normal. An with essential hypertension,412 suggesting partial defects in
often overlooked problem is glucocorticoid overtreatment 11β-hydroxylase activity.
during the first 2 years of life; overtreatment suppresses the Treatment is with replacement glucocorticoid therapy;
infant growth spurt, which is characterized by the highest with suppression of DOC secretion, the plasma renin activ-
postnatal growth velocity. Therefore, the lowest optimal ity, which is suppressed at baseline, increases into the
dose of glucocorticoid replacement should be established normal range. In general, higher glucocorticoid doses are
as early in life as possible. needed to suppress hyperandrogenism compared with the
Increased fat mass and obesity are common among situation in 21-hydroxylase deficiency, and add-on antihy-
children and adolescents with CAH.398-400 Glucocorticoid pertensive therapy may be necessary in some cases. Anti-
dose, chronologic age, advanced bone age maturation, and hypertensive treatment should be commenced at an early
parental obesity all contribute to elevated BMI SDS.399 stage to avoid excessive glucocorticoid exposure.
Increased fat mass and higher insulin levels have been
described in women older than 30 years of age with CAH.
However, clear evidence of cardiovascular risk factors had
17α-Hydroxylase Deficiency
not been shown. Women with CAH do have a significantly Approximately 150 cases of 17α-hydroxylase deficiency
higher rate of gestational diabetes, a possible forerunner have been reported.413-415 Mutations within the CYP17A1
for the development of type 2 diabetes,401 and women with gene result in failure to synthesize cortisol (17α-hydroxylase
nonclassic CAH402 and young adult CAH patients403 have activity), adrenal androgens (17,20-lyase activity), and
reduced insulin sensitivity. Increased intima media thick- gonadal steroids (Fig. 15-38). Therefore, in contrast to
ness as a marker of atherosclerosis has been detected.403 21- and 11β-hydroxylase deficiencies, 17α-hydroxylase
Daytime systolic blood pressure in children and adoles- deficiency results in adrenal and gonadal insufficiency
cents with CAH is elevated, and the physiologic nocturnal and causes 46,XY DSD. A single enzyme is expressed in
dip in blood pressure is absent.404 Elevated systolic blood the adrenal and the gonads and possesses both 17α-
pressure correlates with the degree of overweight and hydroxylation and 17,20-lyase activities, but rare patients
obesity.405 There are no long-term outcome data on adults. with isolated deficiency in the hydroxylation of 17-OHP or
17,20-lyase deficiency have been reported.414 Loss of nega-
tive feedback results in increased secretion of steroids prox-
11β-Hydroxylase Deficiency imal to the block, and mineralocorticoid synthesis is
11β-Hydroxylase deficiency accounts for 7% of all cases of enhanced. Corticosterone has weaker glucocorticoid activ-
CAH, with an incidence of 1 in 100,000 live births.406 The ity than cortisol, but corticosterone excess generally pre-
condition arises because of mutations in the 11β-hydroxylase vents adrenal crises. Accumulation of corticosterone and
(CYP11B1) gene that result in loss of enzyme activity and a DOC results in severe hypokalemic hypertension. Sex
block in the conversion of 11-deoxycortisol to cortisol. The steroid deficiency caused by loss of 17,20-lyase activity
CYP11B1 gene is located on chromosome 8q24.3, approxi- results in 46,XY DSD; this manifests as undervirilization in
mately 40 kilobases from the highly homologous aldoster- male newborns and primary amenorrhea in 46,XX indi-
one synthase gene (CYP11B2).406 CYP11B1-inactivating viduals. There is lack of pubertal development due to
mutations have been shown to be distributed over the hypergonadotropic hypogonadism in both sexes.415
entire coding region consisting of nine exons. Although The 17α-hydroxylase enzyme is a microsomal cyto-
mutation clusters are reported in exons 2, 6, 7, and 8,378,406 chrome P450 type II enzyme that requires electron transfer
real hot spots, as seen in 21-hydroxylase deficiency, do not from NADPH via POR for catalytic activity.26 For efficient
exist. Most of the reported mutations lead to absent or catalysis of the 17,20-lyase reaction, the CYP17A1-POR
540 SECTION IV Adrenal Cortex and Endocrine Hypertension

Cholesterol

StAR

Cholesterol

CYP11A1
CYP17A1 CYP17A1
Pregnenolone 17-OH Pregnenolone DHEA

HSD3B21

Progesterone 17-OH Progesterone Androstenedione

CYP21A2 CYP21A2

DOC 11-Deoxycortisol

CYP11B2

CYP11B1 CYP11B1

Corticosterone Cortisol

CYP11B2

18-OH Corticosterone

CYP11B2

Aldosterone

Mineralocorticoids Glucocorticoids Androgens


Figure 15-37 Congenital adrenal hyperplasia related to 11β-hydroxylase deficiency. The normal synthesis of cortisol is impaired, and adrenocorticotropic hormone (ACTH)
levels increase because of the loss of normal negative feedback inhibition, which results in an increase in adrenal steroid precursors proximal to the block. The results are
cortisol deficiency, mineralocorticoid excess related to excessive deoxycorticosterone (DOC) secretion, and excessive secretion of adrenal androgens. CYP, cytochrome P450;
DHEA, dehydroepiandrosterone; HSD, hydroxysteroid dehydrogenase; StAR, steroidogenic acute regulatory protein.

Cholesterol

StAR

Cholesterol

CYP11A1

Pregnenolone CYP17A1 17-OH Pregnenolone CYP17A1 DHEA

HSD3B21

Progesterone CYP17A1 17-OH Progesterone CYP17A1 Androstenedione

CYP21A2 CYP21A2

DOC 11-Deoxycortisol

CYP11B2 CYP11B1 CYP11B1

Corticosterone Cortisol

CYP11B2

18-OH Corticosterone

CYP11B2

Aldosterone

Mineralocorticoids Glucocorticoids Androgens


Figure 15-38 Congenital adrenal hyperplasia related to 17α-hydroxylase deficiency. The normal synthesis of cortisol is impaired, and adrenocorticotropic hormone (ACTH)
levels increase because of loss of normal negative feedback inhibition resulting in an increase in adrenal steroid precursors proximal to the block. The result is cortisol deficiency
and mineralocorticoid excess usually related to deoxycorticosterone (DOC) excess. Because gonadal 17α-hydroxylase activity is also absent, sex steroid secretion in addition
to adrenal androgen secretion is severely impaired, resulting in hypogonadism. CYP, cytochrome P450; DHEA, dehydroepiandrosterone; HSD, hydroxysteroid dehydrogenase;
StAR, steroidogenic acute regulatory protein.
CHAPTER 15 The Adrenal Cortex 541

complex requires additional allosteric interaction with The paradox of fetal virilization but sex hormone defi-
cytochrome b5.416,417 The CYP17A1 gene consists of eight ciency in postnatal life might be mediated by a newly
exons and is located on chromosome 10q24.3. A variety of discovered “backdoor” pathway of androgen synthesis in
different mutations have been described, without evidence fetal life that relies on neither androstenedione nor testos-
of a hot spot.378,418 Relative hydroxylase/lyase activities of terone as an intermediate.426-428 Pubertal development in
CYP17A1 mutants have been shown to vary in in vitro POR deficiency appears to be dominated by the conse-
functional assays, but correlations with clinical phenotype quences of sex steroid deficiency,428 and most patients
are lacking. Patients with clinically pure 17,20-lyase defi- require sex hormone substitution. The overall incidence of
ciency have CYP17A1 mutations that selectively compro- POR deficiency has not been established. However, a rela-
mise 17,20-lyase activity.416,419,420 Mutations underlying tively large number of patients with POR deficiency were
isolated 17,20-lyase deficiency are located within the area reported within a short period after the initial description
of the CYP17A1 molecule that is thought to interact with of the molecular cause of the disease.428,429
the cofactor cytochrome b5, thereby disrupting the electron The POR gene is located on chromosome 7q11.2 and
transfer from POR to CYP17A1, specifically for the conver- consists of 15 translated exons spanning 32.9 kilobytes and
sion of 17-hydroxypregnenolone to DHEA.416,419 encoding a protein of 680 amino acids. A variety of POR-
The diagnosis is usually made at the time of puberty inactivating mutations have been reported, including mis-
when patients present with hypertension, hypokalemia, sense, frameshift, and splice site mutations. A287P is the
and hypergonadotropic hypogonadism, the last occurring most common mutation in Caucasians, whereas R457H is
because of lack of CYP17A1 expression within the gonad the most frequent founder mutation in the Japanese popu-
and impaired gonadal steroidogenesis. As a result, LH and lation. All patients carry POR mutations that are either
FSH levels are elevated. Female patients (XX) have primary partially inactivating or, in case of major loss-of-function
amenorrhea with absent sexual characteristics, whereas mutations, manifest only in the compound heterozygous
46,XY individuals present with 46,XY DSD with female state. Homozygous mutations with total loss of function
external genitalia but absent uterus and fallopian tubes. are most likely not viable—a view supported by the nonvi-
The intra-abdominal testes should be removed, and such ability of complete POR gene deletion in the murine
patients are usually reared as females. model.430
Glucocorticoid replacement reverses the DOC-induced
suppression of the renin-angiotensin system and lowers
blood pressure. Additional sex steroid replacement is re-
3β-Hydroxysteroid Dehydrogenase Deficiency
quired from puberty onward. In this rare form of CAH, the secretion of all classes of
adrenal and ovarian steroids is impaired due to mutations
within the HSD3B2 gene encoding 3β-HSD type 2.431,432
P450 Oxidoreductase Deficiency: Apparent There are two isoforms of 3β-HSD, encoded by HSD3B1
Combined 17α-Hydroxylase and and HSD3B2, respectively. The HSD3B2 gene is located on
chromosome 1p13.1 and consists of four exons. HSD3B2
21-Hydroxylase Deficiencies is expressed mainly in the adrenal and the gonad, whereas
Patients have been described with biochemical evidence of HSD3B1 is expressed in the placenta and almost ubi-
apparent combined 17α-hydroxylase and 21-hydroxylase quitously in peripheral target tissues.27,432 The enzyme
deficiencies.421 Urinary gas chromatography/mass spec- HSD3B2 catalyzes three key reactions in adrenal steroido-
trometry analysis reveals a typical pattern comprising genesis: the conversion of the Δ5 steroids pregnenolone,
increased pregnenolone and progesterone metabolites, 17-hydroxypregnenolone, and DHEA to the Δ4 steroids
slightly increased corticosterone metabolites, increased progesterone, 17-OHP, and androstenedione, respectively.
pregnanetriolone excretion, and low androgen metabolites 3β-HSDII deficiency affects all three steroid hormone
(see Fig. 15-4). Mothers pregnant with an affected child pathways (i.e., mineralocorticoids, glucocorticoids, and
present with low serum estriol and a characteristic urinary sex steroids).
steroid profile, allowing for prenatal biochemical diagno- The clinical spectrum shows a wide variety of disease
sis.422,423 The analysis of serum steroids only may lead to expression. Patients usually present in early infancy with
misdiagnosis.424 Cortisol baseline secretion may be normal, adrenal insufficiency. Loss of mineralocorticoid secretion
but most, if not all, patients show an insufficient cortisol results in salt wasting, although this is absent in 30% to
response to ACTH stimulation and therefore require gluco- 40% of cases (Fig. 15-39). As with 21-hydroxylase defi-
corticoid replacement. Impaired 17,20-lyase activity results ciency, absence of salt wasting may delay the presentation
in deficient androgen synthesis, and affected boys are often into childhood or puberty, ranging from a severe salt-
born undervirilized. Most of the affected girls are born with wasting form with or without ambiguous genitalia in
virilized genitalia. Therefore, patients can present with affected male neonates to isolated premature pubarche in
46,XY or 46,XX DSD or with appropriate development of infants and children of both sexes and a late-onset variant
the external genitalia in both sexes. After birth, virilization manifesting with hirsutism and menstrual irregularities. In
does not progress, and circulating androgen concentrations general, the functional and biochemical data are in close
are typically low. Some mothers develop signs of viriliza- agreement with the expressed phenotype in patients with
tion during midpregnancy with an affected child; this the non–salt-wasting form of HSD3B2 deficiency. However,
commonly resolves soon after birth, further indicating some variability exists, and identical mutations have been
intrauterine androgen excess.422 In addition to these fea- found in the HSD3B2 gene in both salt-wasters and non–
tures of CAH, affected children may also present with salt-wasters.431,432 The correlation between the impairment
bone malformations, including midface hypoplasia, cra- in male sexual differentiation and salt wasting is poor. The
niosynostosis, and radiohumeral synostosis, in some cases spectrum of genital development is variable in both sexes.
resembling the Antley-Bixler congenital malformation In males, because the HSD3B2 enzyme is also expressed
syndrome.425,426 The bone phenotype in affected patients within the gonad, 46,XY DSD may occur, resulting in
with POR deficiency is most likely caused by an impair- female external genitalia. However, most patients present
ment of sterol biosynthesis, specifically of POR-dependent with hypospadias, and even normal male genitalia may be
14α-lanosterol demethylase (CYP51A1). found. In females, genital development can be normal, but
542 SECTION IV Adrenal Cortex and Endocrine Hypertension

Cholesterol

StAR

Cholesterol

CYP11A1
CYP17A1 CYP17A1
Pregnenolone 17-OH Pregnenolone DHEA

HSD3B2 HSD3B2

Progesterone 17-OH Progesterone Androstenedione

CYP21A2 CYP21A2

DOC 11-Deoxycortisol

CYP11B2 CYP11B1 CYP11B1

Corticosterone Cortisol

CYP11B2

18-OH Corticosterone

CYP11B2

Aldosterone

Mineralocorticoids Glucocorticoids Androgens


Figure 15-39 Congenital adrenal hyperplasia related to 3β-hydroxysteroid dehydrogenase (3β-HSD) type 2 deficiency resulting in cortisol deficiency and variable mineralo-
corticoid deficiency. Gonadal 3β-HSD type 2 activity is also absent, resulting in 46,XY DSD and hypogonadism or primary amenorrhea in females. Virilization in females can
occur due to 3β-hydroxysteroid dehydrogenase type 1 activity. DOC, deoxycorticosterone; DHEA, dehydroepiandrosterone; DSD, disorder of sex development; StAR, steroidogenic
acute regulatory protein.

usually there is evidence of mild virilization, presumably rate. As a result, there is deficiency of all adrenal and
because of enhanced adrenal DHEA secretion, which is gonadal steroid hormones.23,437 The adrenal glands are
converted peripherally to testosterone. A late-onset form often massively enlarged and full of lipid; before the char-
has been described in patients with premature pubarche433 acterization of StAR, the condition was termed congenital
and a PCOS-like phenotype (i.e., hirsutism, oligorrhea/ lipoid adrenal hyperplasia.437 StAR deficiency severely but
amenorrhea).434 incompletely abolishes pregnenolone synthesis. Choles-
Because activity of the HSD3B1 enzyme, present in terol esters accumulate under the increased tone of ACTH
peripheral tissues, is intact, levels of circulating Δ4 steroids stimulation. Consequently, the lipid accumulation worsens
(progesterone, 17-OHP, androstenedione) may be normal the dysfunction and leads to adrenal cell destruction. Pre-
(or even increased). However, a diagnosis is established by sentation is with acute adrenal insufficiency in the neona-
demonstration of an increased ratio of Δ5 steroids (preg- tal period, and males exhibit 46,XY DSD due to absent
nenolone, 17-hydroxypregnenolone, DHEA) to Δ4 steroids gonadal steroids.
in plasma or urine. Hormonal criteria have been refined for The most severe form of this disorder manifests with
the diagnosis of HSD3B2 deficiency based on genotyping 46,XY DSD and combined adrenal insufficiency. Salt
of the HSD3B2 gene. The 17-hydroxypregnenolone con- wasting typically develops in the neonatal period or after
centrations and the ratios of 17-hydroxypregnenolone to a few weeks of life, but later onset may also occur. Females
cortisol at baseline and after ACTH stimulation are of the can show spontaneous pubertal development. A milder
highest discriminatory value in differentiating between form of StAR deficiency has also been described in nor-
patients affected by HSD3B2 deficiency and those with mally virilized 46,XY individuals who present with adrenal
milder biochemical abnormalities, who are commonly failure during early childhood.438 Treatment consists of glu-
negative for HSD3B2 mutations.435,436 Treatment is with cocorticoid and mineralocorticoid replacement and substi-
replacement glucocorticoids, fludrocortisone (if indicated), tution of sex hormones in later life.
and sex steroids from puberty onward.
P450 Side-Chain Cleavage Deficiency
StAR Deficiency: Congenital Lipoid
Deficiency of P450scc (CYP11A1) enzyme is a rare inborn
Adrenal Hyperplasia error of steroidogenesis, with only seven cases reported. It
Mutations in the gene encoding StAR results in a failure was previously thought that such mutations would not be
of transport of cholesterol from the outer to the inner viable, because the maintenance of human pregnancy
mitochondrial membrane in steroidogenic tissues. StAR- relies on placentally produced progesterone. The produc-
independent cholesterol transport occurs only at a low tion is facilitated by the fetal part of the placenta from the
CHAPTER 15 The Adrenal Cortex 543

second trimester onward. P450scc deficiency manifests Primary Defects in Aldosterone Biosynthesis: Aldosterone
clinically and biochemically with similar signs and symp-
toms as StAR deficiency, but patients do not have enlarged
Synthase Deficiency
adrenals.439-441 Depending on the impairment of CYP11A1 Before the characterization of the CYP11B2 gene, two dis-
function, a spectrum of clinical presentation ranges from eases were recognized: corticosterone methyl oxidase type I
46,XY DSD with severe adrenal insufficiency in the newborn (CMO I) deficiency and corticosterone methyl oxidase type
period to hypospadias and cryptorchidism and later mani- II (CMO II) deficiency.446 Subsequently, both variants
festation of adrenal insufficiency during childhood.442 were shown to be secondary to mutations in aldosterone
Concentrations of all steroid hormones are decreased, in synthase (CYP11B2), and they are now termed aldosterone
keeping with impaired conversion of cholesterol to preg- synthase deficiency, types I and II.447 Aldosterone synthase
nenolone. Treatment with glucocorticoid, mineralocorti- catalyzes the three terminal steps of aldosterone bio-
coid, and sex steroid replacement is required. synthesis, 11β-hydroxylation of DOC to corticosterone,
18-hydroxylation to 18-hydroxycorticosterone, and 18-
oxidation to aldosterone. Patients with type I aldosterone
Cortisone Reductase Deficiency synthase deficiency have low to normal levels of 18-
In cortisone reductase deficiency, adrenal glands become hydroxycorticosterone but undetectable levels of aldos-
hyperplastic because of ACTH stimulation due to a defect terone (or urinary tetrahydroaldosterone), whereas pa-
in cortisol metabolism rather than an inherent defect tients with the type II variant have high levels of
within the gland itself.130,443,444 Patients with this condition 18-hydroxycorticosterone and only subnormal or even
have a defect in the conversion of cortisone to cortisol, normal levels of aldosterone. This suggests blockade of
suggesting inhibition of 11-oxo-reductase activity and, by only the terminal 18-oxidation step, with some residual
implication, inhibition of HSD11B1 (see Fig. 15-12). Corti- aldosterone synthase activity remaining. The explanation
sol clearance is increased, and as a consequence, ACTH for the variable biochemical phenotype is unknown, par-
secretion is elevated to maintain normal circulating corti- ticularly now that the same mutation in aldosterone syn-
sol concentrations but at the expense of adrenal androgen thase has been uncovered in both variants. It is possible
excess. Female patients present with hirsutism, menstrual that the phenotypic variation may reflect polymorphic
irregularity, androgenic alopecia, or some combination of variants in the residual and normal product of the CYP11B1
these features. Males may present with premature pubarche. gene, 11β-hydroxylase.
Dexamethasone treatment to suppress ACTH has been Both variants are rare and are inherited as autosomal
used with some success to control the hyperandrogenism recessive traits.447 Patients usually present in neonatal life
in these cases. Urinary tetrahydrometabolites of cortisol with a salt-wasting crisis involving severe dehydration,
and cortisone show almost exclusively THE with little or vomiting, and failure to grow and thrive. Hyperkalemia,
no detectable THF or allo-THF; the ratio of THF+allo-THF metabolic acidosis, dehydration, and hyponatremia are
to THE is less than 0.05 (reference range, 0.8 to 1.3). The found. The plasma renin activity is elevated, and plasma
molecular bases for cortisone reductase deficiency are inac- aldosterone levels are low. Plasma 18-hydroxycorticosterone
tivating mutations in hexose-6-phosphate dehydrogenase levels, the ratio of plasma 18-hydroxycorticosterone to
(H6PDH).444 H6PDH, located in the endoplasmic reticu- aldosterone, and the levels of their urinary metabolites
lum, catalyzes the conversion of glucose 6-phosphate to are used to differentiate the type I and II variants. In
glucose 6-phosphogluconate, thereby generating NADPH, most infants, the disorders become less severe as the
which is crucial in conveying oxo-reductase activity on child ages; indeed, in older children, adolescents, and
HSD11B1. adults, the abnormal steroid pattern described may be
Patients with PCOS share many of the same clinical present and may persist throughout life without clinical
characteristics as those with cortisone reductase deficiency. manifestations.
Whereas there is evidence to support increased cortisol Patients with CYP11B2 deficiency typically respond
secretion rates in PCOS, perhaps indicating a defect in the well to 9α-fludrocortisone (starting dose, 150 μg/m2 per
conversion of cortisone to cortisol, a consensus with respect day in neonates and infants) and may also benefit from
to THF+allo-THF : THE ratios is still lacking.445 Association salt supplementation. Patients with failure to grow and
studies using single-nucleotide polymorphic markers in the thrive usually show a good catch-up growth. Electrolytes
HSD11B1 and H6PDH genes have largely been negative. often tend to normalize spontaneously between 3 and 4
years of age. However, untreated patients are at significant
risk for being growth retarded, although spontaneous
Mineralocorticoid Deficiency normalization of growth can occur. Adults are usually
The mineralocorticoid deficiency syndromes are listed in asymptomatic but are more susceptible to salt loss.
Table 15-24. They can be divided into congenital and Rarely, presentation is in adulthood.448 Mineralocorticoid
acquired syndromes. Mineralocorticoid deficiency may treatment in later life has to be established on an indi-
occur in some forms of CAH and with other causes of vidual basis.
adrenal insufficiency (e.g., Addison disease, CAH).
Postadrenalectomy Hypoaldosteronism
TABLE 15-24
In a patient with a unilateral aldosteronoma (Conn syn-
Causes of Mineralocorticoid Deficiency drome), the contralateral ZG is frequently suppressed.
Addison disease Without reversal of the chronic volume expansion preop-
Adrenal hypoplasia eratively, patients may develop severe hyperkalemia and
Congenital adrenal hyperplasia (21-hydroxylase and 3β-hydroxysteroid hypotension lasting several days to several weeks after
dehydrogenase deficiencies) adrenalectomy. This effect may be exacerbated by the use
Pseudohypoaldosteronism types I and II of spironolactone preoperatively. Spironolactone has a
Hyporeninemic hypoaldosteronism
long half-life and should be discontinued 2 to 3 days before
Aldosterone biosynthetic defects
Drug induced
surgery to minimize the risk of postoperative mineralocor-
ticoid deficiency.
544 SECTION IV Adrenal Cortex and Endocrine Hypertension

HSD11B2 activity and allows unmetabolized cortisol to


Defects in Aldosterone Action: Pseudohypoaldosteronism
bind to and activate MRs in a manner similar to that of
Pseudohypoaldosteronism (PHA) is a rare, inherited salt- aldosterone.453 It was found to be ineffective in patients
wasting disorder that was first described by Cheek and with multiorgan PHA-I.
Perry in 1958 as a defective renal tubular response to min- Two other variants of PHA have been described—types
eralocorticoid in infancy. Patients present in the neonatal II and III. Type II PHA, or Gordon syndrome, is in retro-
period with dehydration, hyponatremia, hypokalemia, spect a misnomer. Patients with Gordon syndrome share
metabolic acidosis, and failure to thrive despite normal some of the features of patients with PHA-I, notably hyper-
glomerular filtration and normal renal and adrenal func- kalemia and metabolic acidosis, but they exhibit salt reten-
tion.449 Renin levels and plasma aldosterone are grossly tion with mild hypertension and suppressed plasma renin
elevated. When patients fail to respond to mineralocor- activity rather than salt wasting. The condition is explained
ticoid therapy, PHA is suspected as the underlying by mutations in a serine threonine kinase family (WNK1
disorder. and WNK4) that result in increased expression of these
PHA type I can be divided into two distinct disorders proteins with activation of the thiazide-sensitive sodium
based on unique physiologic and genetic characteristics: chloride cotransporter in the cortical and medullary col-
the renal form of PHA, which is inherited as an autosomal lecting ducts.454 The condition represents the exact oppo-
dominant trait, and a generalized autosomal recessive form site of Gitelman syndrome but is not a true form of PHA.
of PHA. The autosomal dominant form is usually less Type III PHA is an acquired and usually transient form
severe; the patient’s condition often improves spontane- of mineralocorticoid resistance seen in patients with under-
ously within the first several years of life, allowing discon- lying renal diseases including obstruction and infection
tinuation of therapy. By contrast, the autosomal recessive and in patients with excessive loss of salt through the gut
form produces a multiorgan disorder, with mineralocorti- or skin. Reduced glomerular filtration rate is a hallmark of
coid resistance seen in the kidney, sweat and salivary the condition. The cause is unknown, although increased
glands, and the colonic mucosa. The condition does not transforming growth factor-β–mediated aldosterone resis-
spontaneously improve with age and is generally more tance has been suggested to be an underlying factor.
severe than the autosomal dominant form.
The underlying basis for the autosomal dominant form
of PHA is explained on the basis of inactivating mutations
Hyporeninemic Hypoaldosteronism
in the MR (hMR, NR3C2).449,450 By contrast, inactivating Angiotensin II is a key stimulus to aldosterone secretion,
mutations in the α-subunit and, to a lesser extent, in the and damage or blockade of the renin-angiotensin system
β- and γ-subunits of the epithelial sodium channel (ENaC) may result in mineralocorticoid deficiency. Various renal
account for the generalized autosomal recessive form of diseases have been associated with damage to the juxtaglo-
mineralocorticoid resistance.451,452 (In effect, this represents merular apparatus and subsequent renin deficiency. These
the opposite of Liddle syndrome—see Chapter 16.) Gener- include systemic lupus erythematosus, myeloma, amyloid,
alized loss of ENaC activity leads to renal salt wasting (as AIDS, and damage related to use of nonsteroidal anti-
seen in the renal form) in addition to recurrent respiratory inflammatory drugs, but the most common (>75% of cases)
infections and neonatal respiratory distress, cholelithiasis, is diabetic nephropathy.455,456
and polyhydramnios. The usual picture is that of an elderly patient with
PHA type I is resistant to mineralocorticoid therapy, so hyperkalemia, acidosis, and mild to moderate impairment
standard treatment involves supplementation with salt (2 of renal function. Plasma renin activity and aldosterone
to 8 g/day) in the form of sodium chloride and sodium levels are low and fail to respond to sodium depletion, erect
bicarbonate as well as cation exchange resins. This sup- posture, or furosemide administration. In contrast to
plementation usually corrects the patient’s biochemical adrenal insufficiency, patients have normal or elevated
imbalance. However, if a patient shows signs of severe blood pressure and no postural hypotension. Muscle weak-
hyperkalemia, peritoneal dialysis may be necessary. Hyper- ness and cardiac arrhythmias may also occur. Other factors
calciuria has been reported in some cases of PHA-I. The may contribute to the hyperkalemia, including the use of
recommended course of treatment for these patients potassium-sparing diuretics, potassium supplementation,
usually involves indomethacin or hydrochlorothiazide. insulin deficiency, and use of β-adrenoceptor blocking
Indomethacin is thought to act by causing a reduction in drugs or prostaglandin synthetase inhibitors, which inhibit
the glomerular filtration rate or an inhibition of the effect renin release.
of prostaglandin E2 on renal tubules. Indomethacin has Treatment of primary renin deficiency is with fludrocor-
been shown to reduce polyuria, sodium loss, and hypercal- tisone in the first instance together with dietary potassium
ciuria. Hydrochlorothiazide has been used to diminish restriction. However, these patients are not salt depleted
hyperkalemia and reduce hypercalciuria in patients with and may become hypertensive with fludrocortisone. In
PHA-I. such a scenario, the addition of a loop-acting diuretic such
In patients with the autosomal dominant or renal form as furosemide is appropriate. This will increase acid excre-
of PHA-I, the signs and symptoms of PHA decrease with tion and improve the metabolic acidosis.
age; nevertheless, these patients usually require salt supple-
mentation for the first 2 to 3 years of life. In patients with
the autosomal recessive or multiorgan type of PHA-I, resis-
tance to therapy with sodium chloride or drugs that ADRENAL ADENOMAS,
decrease serum potassium concentrations often occurs and INCIDENTALOMAS, AND CARCINOMAS
may even lead to death in infancy from hyperkalemia.
PHA-I patients with multiorgan involvement often require
very high amounts of salt in their diet (up to 45 g NaCl
Adenomas
per day). Carbenoxolone, a derivative of glycyrrhetinic Cortisol-secreting adrenal adenomas were discussed earlier,
acid in licorice, has been used with moderate success in and aldosterone-secreting adenomas (Conn syndrome) are
helping to reduce the high levels of dietary salt needed by discussed in Chapter 16. Pure virilizing benign adrenal
renal PHA-I patients. Carbenoxolone acts by inhibiting adenomas are rare, with approximately 50 cases reported
CHAPTER 15 The Adrenal Cortex 545

in the literature. Most cases occur in women; in males, the events, osteoporosis, and fatality. However, no prospective
disorder is restricted to childhood, when presentation is study has proved that the adrenal adenoma is the cause of
with sexual precocity and accelerated bone age. Such the observed complications, as these are highly prevalent
tumors have to be considered in the differential diagnosis in the population at this age. Intervention by adrenalec-
of CAH in patients presenting during childhood. In females, tomy has shown some benefit, but the studies performed
most patients present before menopause with marked hir- have been retrospective and highly selected, and hence the
sutism, deepening of the voice, and amenorrhea. Clitoro- approach to each patient needs individualization with
megaly is found in 80% of cases. Testosterone is usually most being observed in current clinical practice.
strikingly elevated, but gonadotropin levels may not be As a result, all patients with incidentally discovered
suppressed. By definition, urinary free cortisol is normal. adrenal masses should undergo appropriate endocrine
Tumors vary in size and should be treated surgically. Post- screening tests. This testing should comprise 24-hour uri-
operatively, clinical features invariably improve, and nary catecholamine collection or measurement of plasma
normal menses return.457 metanephrines, 24-hour urinary free cortisol (or a mid-
night salivary cortisol level), and overnight dexameth-
asone suppression tests. Because of the reported poor
Incidentalomas sensitivity of serum potassium measurements in detecting
Autopsy series had defined the prevalence of adrenal ade- primary aldosteronism, circulating levels of plasma renin
nomas greater than 1 cm in diameter to be between 1.5% activity and aldosterone are required in hypertensive
and 7% before the advent of high-resolution imaging pro- patients. DHEAS should be measured as a marker of adrenal
cedures such as CT and MRI; since that time, incidentally androgen secretion. Low levels may occur in patients with
discovered adrenal masses have become a common clinical suppressed ACTH concentrations due to autonomous cor-
problem. An adrenal mass is uncovered in up to 4% of tisol secretion from the adenoma.461 Some studies have
patients imaged for nonadrenal disease.458 Incidentalomas also documented high levels of 17-OHP after ACTH stimu-
are uncommon in patients younger than 30 years of age lation tests, suggesting partial defects in 21-hydroxylase in
but increase in frequency with age; they occur equally in some tumors.
males and females, most commonly in the sixth and The possibility of malignancy should be considered in
seventh decades. Clinically, two issues arise: whether the each case. In patients with a known extra-adrenal primary
lesion is functional (i.e., secreting hormones) and whether tumor, the incidence of malignancy is obviously much
it is malignant. Most incidentalomas are adrenocortical higher; for example, up to 20% of patients with lung cancer
adenomas, but occasionally they represent myelolipomas, have adrenal metastases on CT scanning. In those with no
hamartomas, or granulomatous infiltrations of the adrenal evidence of malignancy, adrenal carcinoma is rare; in one
gland and result in a characteristic CT/MRI appearance study, only 26 of 630 incidentalomas were found to be
(Fig. 15-40). Functioning tumors (pheochromocytomas adrenal carcinomas.458 Many studies contain a positive
and those secreting cortisol, aldosterone, or sex steroids) bias, and true risk of malignancy may be much lower.462 In
and carcinomas account for around 4% of all incidentalo- true incidentalomas, size appears to be predictive of malig-
mas. In addition, it is established that some incidentalomas nancy: fewer than 2% of incidentalomas smaller than 4 cm
cause abnormal hormone secretion without obvious clini- but 25% of those larger than 6 cm in diameter are malig-
cal manifestations of a hormone excess state. The best nant (Fig. 15-41).463 Smooth, homogeneous adenomas on
example is so-called subclinical Cushing syndrome, which an enhanced adrenal scan with a Hounsfield unit score
occurs in up to 20% to 30% of all cases.458-460 There is debate (a marker of radiodensity) less than 10 HU are invariably
as to the best means to biochemically define this phenom- benign; malignancy is suspected in irregular, inhomoge-
enon, but serum cortisol after dexamethasone testing has neous adenomas with a score greater than 20 HU. On this
the widest acceptance, although cutoff values vary. When background, adrenalectomy is indicated for functional
there is evidence of low-grade excess biochemical hyper- tumors and for tumors larger than 4 cm in diameter. Repeat
cortisolism there is an associated increase in the prevalence CT scanning in patients with smaller tumors can be used
of diabetes, obesity, hypertension, new cardiovascular to guide management, but development of functional

A B
Figure 15-40 A, Adrenal incidentaloma (arrow) discovered in a woman undergoing investigation for abdominal pain. B, Incidentally discovered right adrenal myelolipoma
(arrow).
546 SECTION IV Adrenal Cortex and Endocrine Hypertension

Overall, the prognosis is poor, with 5-year survival rates


100
of less than 20%. Newer chemotherapies are being
90 evaluated.285
80 Metastases
Other
70
Myelolipoma
Etiology of Adrenal Tumors
Percent

60 Ganglioneuroma Other than the factors discussed under adrenal Cushing


50 Adrenal cyst syndrome, the underlying basis for adrenal tumorigenesis
40 Pheochromocytoma is unknown. Clonal analysis suggests progression from a
Adrenal carcinoma normal to an adenomatous to a carcinomatous lesion, but
30
Adenoma
the molecular pathways involved remain obscure. Several
20 factors have been associated with malignant transforma-
10 tion, including the genes encoding p53, p57 cyclin-
dependent kinase, menin, IGF-2, MC2R, and inhibin-α.464
0
Mice lacking the inhibin-α gene develop adrenal tumors
<2-4 4.1-6 >6 Total through a process that is also gonadotropin dependent.465
A cm cm cm

2%
ACKNOWLEDGMENTS
6% Adenoma Acknowledgment is given to Professor W. Arlt, Dr. M.S.
4% 8%
Adrenal carcinoma Cooper, Dr. J.W. Tomlinson, and Dr. W. Young for assis-
Pheochromocytoma tance with parts of this chapter.
5% Adrenal cyst
52%
Ganglioneuroma
11%
Myelolipoma REFERENCES
Other 1. Eustachius B. Tabulae Anatomicae. In: Lancisi JM, ed. Amsterdam: R. &
12% G. Wetstenios; 1774.
Metastases 2. Addison T. On the Constitutional and Local Effects of Disease of the Supra-
B Renal Capsules. London: Highley; 1866.
3. Brown-Sequard CE. Recherches experimentales sur la physiologie et la
Figure 15-41 Distribution of diagnosis of adrenal incidentalomas. A, Data from pathologie des capsules surrenales. Arch Gen Med. 1856;5:385-401.
eight studies with histologically determined diagnoses (n = 103) relating to tumor 4. Medvei VC. A History of Clinical Endocrinology. Pearl River, NY: Parthe-
size. B, Distribution of 380 incidental adrenal masses by histologic diagnosis. (From non; 1993.
Mansmann G, Lau L, Balk E, et al. The clinically inapparent adrenal mass: update in 5. Hench PS, Kendall EC, Slocumb CH, et al. The effect of a hormone of
the adrenal cortex (17-hydroxy-11-dehydrocorticosterone; compound
diagnosis and management. Endocr Rev. 2004;25:309-340.)
E) and of pituitary adrenocorticotropic hormone on rheumatoid
arthritis. Mayo Clin Proc. 1949;24:181-197.
6. Simpson SA, Tait JF. Recent progress in methods of isolation, chemis-
autonomy is very rare, and patients may be discharged if try, and physiology of aldosterone. Recent Prog Horm Res. 1955;11:
183-210.
tumors are static. Laparoscopic adrenalectomy is the treat- 7. Li CH, Evans HM, Simpson ME. Adrenocorticotrophic hormone. J Biol
ment of choice, offering shorter hospital stays and reduced Chem. 1943;149:413-424.
operative complications (e.g., blood loss, morbidity) com- 8. Cushing H. The basophil adenomas of the pituitary body and their
pared with open adrenalectomy. The possible exception is clinical manifestations (pituitary basophilism). Bull Johns Hopkins
Hosp. 1932;50:137-195.
the patient with highly suggestive adrenal carcinoma, 9. Vale W, Spiess J, Rivier C, et al. Characterization of a 41-residue ovine
because breach of the tumor capsule is associated with a hypothalamic peptide that stimulates secretion of corticotropin and
poorer outcome. Adequate preparation and close endo- β-endorphin. Science. 1981;213:1394-1397.
crine supervision perioperatively and postoperatively are 10. Conn JW. Primary aldosteronism, a new clinical syndrome. J Lab Clin
Med. 1955;45:3-17.
required for functional tumors. 11. Else T, Hammer GD. Genetic analysis of adrenal absence: agenesis and
aplasia. Trends Endocrinol Metab. 2005;16:458.
12. Mesiano S, Jaffe RB. Developmental and functional biology of the
Carcinomas primate fetal adrenal cortex. Endocr Rev. 1997;18:378-403.
13. Jaffe RB, Mesiano S, Smith R, et al. The regulation and role of fetal
Primary adrenal carcinoma is very rare, with an annual adrenal development in human pregnancy. Endocr Res. 1998;24:
incidence of 1 per 1 million population. Women are more 919-926.
commonly affected than men, with a ratio of 2.5 : 1. The 14. Okamoto M, Takemori H. Differentiation and zonation of the adrenal
mean age at onset is between 40 and 50 years, although cortex. Curr Opin Endocrinol Diab. 2000;7:122-127.
15. Luo X, Ikeda Y, Parker KL. A cell-specific nuclear receptor is essential
men tend to be older at presentation. Eighty percent of for adrenal and gonadal development and sexual differentiation. Cell.
tumors are functional, most commonly secreting glucocor- 1994;77:481-490.
ticoids alone (45%), glucocorticoids and androgens (45%), 16. Havelock JC, Auchus RJ, Rainey WE. The rise in adrenal androgen
or androgens alone (10%). Fewer than 1% of all tumors biosynthesis: adrenarche. Semin Reprod Med. 2004;22:337-347.
17. Remer T, Boye KR, Hartmann MF, et al. Urinary markers of adrenarche:
secrete aldosterone. Patients present with features of the reference values in healthy subjects, aged 3-18 years. J Clin Endocrinol
hormone excess state (glucocorticoid, androgen, or both) Metab. 2005;90:2015-2021.
but abdominal pain, weight loss, anorexia, and fever occur 18. IUPAC Commission on the Nomenclature of Organic Chemistry
in 25% of cases. An abdominal mass may be palpable. (CNOC) and IUPAC-IUB Commission on Biochemical Nomenclature
(CBN). Definitive rules for the nomenclature of steroids. Pure Appl
Current treatment choices for what is often an aggressive Chem. 1972;31:285-322.
tumor are poor. Surgery offers the only chance of cure for 19. Gwynne JT, Strauss JF 3rd. The role of lipoproteins in steroidogenesis
patients with local disease, but metastatic spread is evident and cholesterol metabolism in steroidogenic glands. Endocr Rev.
in 75% of cases at presentation. Radiotherapy is ineffective, 1982;3:299-329.
20. Faust JR, Goldstein JL, Brown MS. Receptor-mediated uptake of low
as are most chemotherapeutic regimens. Mitotane in high density lipoprotein and utilization of its cholesterol for steroid
doses offers benefit in reducing tumor growth284 and in synthesis in cultured mouse adrenal cells. J Biol Chem. 1977;252:
controlling hormonal hypersecretion in 75% of cases.283 4861-4871.

You might also like