2996 PART 12 Endocrinology and Metabolism
impaired glucose tolerance). Active basic and clinical research
using new approaches and combination therapy may change the
treatment of this disease or other autoimmune conditions that share
similar pathways.
■ IMMUNE CHECKPOINT INHIBITOR–INDUCED
ENDOCRINE AUTOIMMUNITY
Therapies that block immune checkpoints, such as programmed
cell death protein 1 (PD-1), its ligand (PD-L1), or CTLA-4, are beneficial immunotherapies for many advanced-stage cancers. These
immune checkpoint inhibitors (ICIs) block negative immune regulation, thereby allowing for an immune response directed against tumor
cells. However, immune-related adverse events also occur, especially
autoimmunity directed toward self-tissues. ICI-induced T1D, thyroid
disease, hypophysitis, and adrenal insufficiency have all been reported
with these therapies. Hypothyroidism occurs in ~8% and T1D in 1%
of those receiving monoclonal antibodies directed against PD-1 or
PD-L1, and hypophysitis and adrenal insufficiency occur in <1% of
treated patients. These autoimmune side effects can develop during
or after therapy, mostly within a few weeks to months following the
start of therapy. ICI-induced T1D has a very rapid onset, presents with
diabetic ketoacidosis, is permanent, and requires lifelong exogenous
insulin therapy for treatment. There is a genetic association with
HLA-DR4 and islet autoantibodies in ~40–50% of patients at diagnosis.
The pathogenesis is immune mediated as T lymphocyte infiltration
has been documented in the pancreatic islets of an ICI-T1D patient.
Determining the mechanisms of autoimmune disease development
following ICI therapies and developing biomarkers to stratify risk for
autoimmune side effects prior to therapy are active areas of research.
■ IPEX
Immune dysregulation, polyendocrinopathy, enteropathy, and X-linked
disease (IPEX; OMIM 304790) is a rare X-linked recessive disorder. The disease onset is in infancy and is characterized by severe
enteropathy, T1D, and skin disease, as well as variable association
with several other autoimmune disorders. Many infants die within
the first days of life, but the course is variable, with some children surviving for 12–15 years. Early onset of T1D, often at birth,
is highly suggestive of the diagnosis because nearly 80% of IPEX
patients develop T1D. Although treatment of the individual disorders
can temporarily improve the situation, treatment of the underlying
immune deficiency is required and includes immunosuppressive
therapy generally followed by hematopoietic stem cell transplantation.
Transplantation is the only life-saving form of therapy and can be fully
curative by normalizing the imbalanced immune system found in this
disorder.
IPEX is caused by mutations in the FOXP3 gene, which is also
mutated in the Scurfy mouse, an animal model that shares much of
the same phenotype of IPEX patients. The FOXP3 transcription factor
is expressed in regulatory T cells designated CD4+CD25+FOXP3+
(Treg). Lack of this factor causes a profound deficiency of this Treg
population and results in rampant autoimmunity due to the lack of
peripheral tolerance normally provided by these cells. Certain mutations may lead to varying forms of expression of the full syndrome,
and there are rare cases where the FOXP3 gene is intact but other genes
involved in this pathway (e.g., CD25, IL-2Rα) may be causative. Future
therapy with autologous CD4+ T cells transfected with a functioning
FOXP3 gene may offer a better long-term outcome than has been seen
in those treated with stem cell transplantation.
■ THYMIC TUMORS
Thymomas and thymic hyperplasia are associated with several autoimmune diseases, with the most common being myasthenia gravis
(44%) and red cell aplasia (20%). Graves’ disease, T1D, and Addison’s
disease may also be associated with thymic tumors. Patients with
myasthenia gravis and thymoma may have unique anti–acetylcholine
receptor autoantibodies. Most thymomas lack AIRE expression within
the thymoma, and this could be a potential factor in the development
of autoimmunity. In support of this concept, thymoma is the one
other disease with “frequent” development of anticytokine antibodies
and mucocutaneous candidiasis in adults. The majority of tumors are
malignant, and temporary remissions of the autoimmune condition
can occur with resection of the tumor.
■ ANTI-INSULIN RECEPTOR ANTIBODIES
This is a very rare disorder where severe insulin resistance (type B) is
caused by the presence of anti-insulin receptor antibodies. It is associated with acanthosis nigricans, which can also be associated with other
forms of less severe insulin resistance. About one-third of patients
have an associated autoimmune illness such as systemic lupus erythematosus or Sjögren’s syndrome. Therefore, the presence of antinuclear
antibodies, elevated erythrocyte sedimentation rate, hyperglobulinemia, leukopenia, and hypocomplementemia may accompany the presentation. The presence of anti-insulin receptor autoantibodies leads
to marked insulin resistance, requiring >100,000 units of insulin to be
given daily with only partial control of hyperglycemia. Patients can also
have severe hypoglycemia due to partial activation of the insulin receptor by the antibody. The course of the disease is variable, and several
patients have had spontaneous remissions. A therapeutic approach that
targets B lymphocytes, including rituximab, cyclophosphamide, and
pulse steroids, has been validated in follow-on case reports to induce
remission of the disease.
■ INSULIN AUTOIMMUNE SYNDROME
(HIRATA’S SYNDROME)
The insulin autoimmune syndrome, associated with Graves’ disease and
methimazole therapy (or other sulfhydryl-containing medications), is
of particular interest due to a remarkably strong association with a specific HLA haplotype. Such patients with elevated titers of anti-insulin
antibodies frequently present with hypoglycemia. In Japan, the disease
is restricted to HLA-DR4-positive individuals with DRB1*
04:06, while
Caucasian patients predominantly have DRB1*
04:03 (which is related
to DRB1*
04:06). In Hirata’s syndrome, the anti-insulin antibodies are
often polyclonal. Discontinuation of the medication generally leads
to resolution of the syndrome over time. There are very rare cases of
insulin autoimmune syndrome not associated with sulfhydryl-containing
medications that result in profound, life-threatening hypoglycemia.
Treatment involves treating the underlying condition that causes antiinsulin antibodies, such as a B lymphocyte lymphoma (tend to have
monoclonal insulin antibodies) or systemic lupus erythematosus. As
hypoglycemia is profound when elevated titers of high affinity insulin
antibodies bind secreted insulin and then release it into circulation,
treatment that begins with high-dose glucocorticoids and rituximab to
target B lymphocytes has been shown to be effective.
■ POEMS SYNDROME
POEMS (polyneuropathy, organomegaly, endocrinopathy, M-protein,
and skin changes; also known as Crow-Fukase syndrome; OMIM
192240) patients usually present with a progressive sensorimotor
polyneuropathy, diabetes mellitus (50%), primary gonadal failure
(70%), and a plasma cell dyscrasia with sclerotic bony lesions. Associated findings can be hepatosplenomegaly, lymphadenopathy, and
hyperpigmentation. Patients often present in the fifth to sixth decade
of life and have a median survival after diagnosis of <3 years. The
syndrome is assumed to be secondary to circulating immunoglobulins, but patients have excess vascular endothelial growth factor as
well as elevated levels of other inflammatory cytokines such as IL1-β,
IL-6, and tumor necrosis factor α. Patients have been treated with
thalidomide, and more recently lenalidomide, leading to a decrease in
vascular endothelial growth factor. Hyperglycemia responds to small,
subcutaneous doses of insulin. The hypogonadism is due to primary
gonadal disease with elevated plasma levels of follicle-stimulating hormone and luteinizing hormone. Temporary resolution of the features
of POEMS, including normalization of blood glucose, may occur after
radiotherapy for localized plasma cell lesions of bone or after chemotherapy, lenalidomide and dexamethasone, or autologous stem cell
transplantation.
2997 Sex Development CHAPTER 390
■ OTHER DISORDERS
Other diseases can exhibit polyendocrine deficiencies, including
Kearns-Sayre syndrome, DIDMOAD syndrome (diabetes insipidus,
diabetes mellitus, progressive bilateral optic atrophy, and sensorineural
deafness; also termed Wolfram’s syndrome), Down’s syndrome or trisomy 21 (OMIM 190685), Turner’s syndrome (monosomy X, 45,X0),
and congenital rubella.
Kearns-Sayre syndrome (OMIM 530000) is a rare mitochondrial
DNA disorder characterized by myopathic abnormalities leading to
ophthalmoplegia and progressive weakness in association with several endocrine abnormalities, including hypoparathyroidism, primary
gonadal failure, diabetes mellitus, and hypopituitarism. Crystalline
mitochondrial inclusions are found in muscle biopsy specimens, and
such inclusions have also been observed in the cerebellum. Antiparathyroid antibodies have not been described; however, antibodies to the
anterior pituitary gland and striated muscle have been identified, and
the disease may have autoimmune components. These mitochondrial
DNA mutations occur sporadically and do not appear to be associated
with a familial syndrome.
Wolfram’s syndrome (OMIM 222300, chromosome 4; OMIM
598500, mitochondrial) is a rare autosomal recessive disease that is
also called DIDMOAD. Neurologic and psychiatric disturbances are
prominent in most patients and can cause severe disability. The disease
is caused by defects in the Wolfram syndrome 1 (WFS1) gene, which
encodes a 100-kDa transmembrane protein that has been localized to
the endoplasmic reticulum and is found in neuronal and neuroendocrine tissue. Its expression induces ion channel activity with a resultant
increase in intracellular calcium and may play an important role in
intracellular calcium homeostasis. Wolfram’s syndrome appears to be
a slowly progressive neurodegenerative process, and there is nonautoimmune selective destruction of the pancreatic beta cells. Diabetes
mellitus with an onset in childhood is usually the first manifestation.
Diabetes mellitus and optic atrophy are present in all reported cases,
but expression of the other features is variable. Treatments targeting
endoplasmic reticulum dysfunction are being tested and may be a
bridge until gene therapy can be developed to treat the most severely
affected cases.
Down’s syndrome, or trisomy 21 (OMIM 190685), is associated with
the development of T1D, thyroiditis, and celiac disease. Patients with
Turner’s syndrome also appear to be at increased risk for the development of thyroid disease and celiac disease. It is recommended to
screen patients with trisomy 21 and Turner’s syndrome for associated
autoimmune diseases on a regular basis.
■ GLOBAL CONSIDERATIONS
Identification of these syndromes requires access to central laboratories with the ability to detect unique autoantibodies and to sequence
the specific genes that may underlie these disorders. Early recognition
of the clinical features of these disorders and timely referral and/or
consultation with tertiary care centers to confirm the diagnosis and
initiate therapy are important to improving outcomes. The AIRE
recessive gene mutations found in APS-1 were originally described in
high frequency in several populations including Finns, Iranian Jews,
Sardinians, Norwegians, and Irish. Although individuals from many
other countries have now been found to have these mutations and the
newly identified dominant AIRE gene mutations, understanding the
frequency in the background population may raise the clinician’s level
of suspicion for these rare disorders. Hirata’s syndrome was originally
reported in Japanese populations but also may be found in other populations, as noted.
■ FURTHER READING
Anderson MS, Su MA: AIRE expands: New roles in immune tolerance and beyond. Nat Rev Immunol 16:247, 2016.
Husebye ES et al: Autoimmune polyendocrine syndromes. N Engl J
Med 378:1132, 2018.
Postow MA et al: Immune-related adverse events associated with
immune checkpoint blockade. N Engl J Med 378:158, 2018.
Section 2 Sex- and Gender-Based
Medicine
390 Sex Development
Courtney Finlayson, J. Larry Jameson,
John C. Achermann
Sex development begins in utero but continues into young adulthood
with the achievement of sexual maturity and reproductive capability.
The major determinants of sex development can be divided into three
components: chromosomal sex, gonadal sex (sex determination), and
phenotypic sex (sex differentiation) (Fig. 390-1). Variations at each
of these stages can result in differences (or disorders) of sex development (DSDs) (Table 390-1). In the newborn period, ~1 in 5000 babies
undergo investigation because of atypical or ambiguous genitalia.
Urgent assessment is indicated, because some causes such as congenital adrenal hyperplasia (CAH) can be associated with life-threatening
adrenal crises. An experienced multidisciplinary team is important for
counseling, planning appropriate investigations, discussing long-term
well-being, supporting parents, and providing clear communication
about the diagnosis and management options. DSDs can also present at
other ages and to a range of health professionals (Table 390-2). Subtler
forms of gonadal dysfunction (e.g., Klinefelter syndrome [KS], Turner
syndrome [TS]) often are diagnosed later in life by internists. Because
DSDs are associated with a variety of psychological, reproductive, and
potential medical consequences, an open dialogue must be established
between the patient and health care providers to ensure continuity
and attention to these issues across the life span. Gender variance and
gender dysphoria are more common among some individuals with
DSD than in the general population. Thus, attention to and comfort
discussing gender identity is important. Support groups also have a
valuable role to play for many patients and families.
Care of individuals with DSDs has evolved from primarily focusing
on medical and surgical intervention to “genitalia” to a more holistic
approach involving medical, surgical, and psychosocial care, acknowledging that the best way to care for individuals with DSD is not always
clear and should be individualized. This includes many controversies,
particularly concerning whether genitoplasty or prophylactic gonadectomy in selected conditions should be performed for infants and young
children prior to the age of consent. Accepted nomenclature is also
controversial. Previous terms such as intersex and hermaphrodite were
Testis-determining
genes
Gonadal steroids
& peptides
(T, DHT, AMH/MIS)
Gonadal steroids
(E2)
Ovary-determining
genes
XX
Chromosomal Sex
Gonadal Sex
Phenotypic Sex
XY
FIGURE 390-1 Sex development can be divided into three major components:
chromosomal sex, gonadal sex, and phenotypic sex. AMH, anti-müllerian hormone
also known as Müllerian-inhibiting substance, MIS; DHT, dihydrotestosterone;
T, testosterone.
2998 PART 12 Endocrinology and Metabolism
changed by the 2006 Consensus Statement to disorder of sex development
and ovotesticular DSD, but these terms are not universally accepted.
SEX DEVELOPMENT
Chromosomal sex, defined by a karyotype, describes the X and/or Y
chromosome complement (46,XY; 46,XX) established at the time of
fertilization. The presence of a normal Y chromosome determines
TABLE 390-1 Classification of Disorders of Sex Development (DSDs)a
SEX CHROMOSOME DSD 46,XY DSD (SEE TABLE 390-3) 46,XX DSD (SEE TABLE 390-4)
47,XXY (Klinefelter syndrome and
variants)
45,X (Turner syndrome and
variants)
45,X/46,XY mosaicism (mixed
gonadal dysgenesis)
46,XX/46,XY (chimerism/
mosaicism)
Disorders of gonadal (testis) development
Complete or partial gonadal dysgenesis (e.g., SRY, SOX9, SF1, WT1,
DMRT1, DHH, GATA4, ZFPM2, MAP3K1, ESR2, ZNRF3, SOX8, DHX37)
Impaired fetal Leydig cell function (e.g., SF1/NR5A1, CXorf6/
MAMLD1, HHAT, SAMD9)
Ovotesticular DSD
Testis regression
Disorders in androgen synthesis or action
Disorders of androgen biosynthesis
LH receptor (LHCGR)
Smith-Lemli-Opitz syndrome (DHCR7)
Steroidogenic acute regulatory (StAR) protein
Cholesterol side-chain cleavage (CYP11A1)
3β-Hydroxysteroid dehydrogenase II (HSD3B2)
17α-Hydroxylase/17,20-lyase (CYP17A1)
P450 oxidoreductase (POR)
Cytochrome b5 (CYB5A)
17β-Hydroxysteroid dehydrogenase III (HSD17B3)
5α-Reductase II (SRD5A2)
Aldo-keto reductase 1C2 (AKR1C2)
Disorders of androgen action
Androgen insensitivity syndrome
Drugs and environmental modulators
Other
Syndromic associations of male genital development
Associated with fetal growth restriction
Persistent müllerian duct syndrome
Vanishing testis syndrome
Isolated hypospadias
Congenital hypogonadotropic hypogonadism
Cryptorchidism
Environmental influences
Disorders of gonadal (ovary) development
Gonadal dysgenesis
Ovotesticular DSD
Testicular DSD (e.g., SRY+, dup SOX9, RSPO1, SF1/NR5A1,
NR2F2, WT1)
Androgen excess
Fetal
3β-Hydroxysteroid dehydrogenase II (HSD3b2)
21-Hydroxylase (CYP21A2)
P450 oxidoreductase (POR)
11β-Hydroxylase (CYP11B1)
Fetoplacental
Aromatase deficiency (CYP19)
Oxidoreductase deficiency (POR)
Maternal
Maternal virilizing tumors (e.g., luteomas)
Androgenic drugs
Other
Syndromic associations (e.g., cloacal anomalies)
Müllerian agenesis/hypoplasia (e.g., MRKH)
Uterine abnormalities (e.g., MODY5)
Vaginal atresia (e.g., McKusick-Kaufman)
Labial adhesions
a
Some experts and patient advocacy groups prefer to define DSD as differences of sex development rather than disorders of sex development.
Abbreviations: LH, luteinizing hormone; MODY, maturity-onset diabetes of the young; MRKH, Mayer-Rokitansky-Küster-Hauser syndrome.
Source: Reproduced with permission from IA Hughes et al: Consensus statement on management of intersex disorders. J Pediatr Urol 2:148, 2006.
TABLE 390-2 Presentation of Disorders of Sex Development (DSD) at Different Stages of Life
PRESENTATION FEATURES PROFESSIONAL EXAMPLES
Prenatal Karyotype-phenotype discordance Obstetrician; fetal medicine Many
Neonatal Atypical genitalia Obstetrician; neonatal medicine Many
Salt-losing crisis Pediatrician CAH (CYP21)
Childhood Hernia Surgeon CAIS
Androgenization Endocrinologist CAH (CYP21, CYP11B1)
Poor growth Pediatrician Turner, 45,X/46,XY
Associated features Oncologist/nephrologist Wilms’ tumor
Puberty Androgenization
Estrogenization
Endocrinologist
Endocrinologist
17β-HSD, 5α-reductase, SF1
Ovotestis
Absent puberty Endocrinologist Gonadal dysgenesis, CAH (CYP17A1), Turner
Post-puberty Amenorrhea Gynecologist CAIS
Adult Infertility Andrologist Klinefelter, 45,X/46,XY, SF1
Abbreviations: CAH, congenital adrenal hyperplasia; CAIS, complete androgen insensitivity syndrome; 17β-HSD, 17β-hydroxysteroid dehydrogenase deficiency; SF1,
steroidogenic factor 1 (NR5A1).
that testis development will occur even in the presence of multiple X
chromosomes (e.g., 47,XXY). Loss of an X chromosome impairs gonad
development (45,X or 45,X/46,XY mosaicism). Fetuses with no X chromosome (45,Y) are not viable.
Gonadal sex refers to the histologic and functional characteristics
of gonadal tissue as testis or ovary. The embryonic gonad is initially
“bipotential” and can develop (from ~42 days after conception) into
2999 Sex Development CHAPTER 390
Urogenital ridge
Granulosa cells Sertoli cells Leydig cells
WT1
SF1
SRY
SOX9
Other
genes
WNT4
RSPO1
FOXL2
46,XX
AMH Testosterone
DHT
46,XY
Bipotential gonad
Ovary Testis
Müllerian
regression
Follicle
development
Male sexual
differentiation
FIGURE 390-2 The genetic regulation of gonadal development. See text for
additional genes involved. AMH, anti-müllerian hormone (müllerian-inhibiting
substance); DHT, dihydrotestosterone; FOXL2, forkhead transcription factor L2;
RSPO1, R-spondin 1; SF1, steroidogenic factor 1 (also known as NR5A1); SOX9, SRYrelated HMG-box gene 9; SRY, sex-determining region on the Y chromosome; WNT4,
wingless-type MMTV integration site 4; WT1, Wilms’ tumor–related gene 1.
either a testis or an ovary (Fig. 390-2). Testis development is initiated
by expression of the gene SRY (sex-determining region on the Y chromosome). Disruption of SRY prevents testis development in 46,XY
individuals, whereas translocation of SRY in 46,XX individuals induces
testis development and a male phenotype. The main target of SRY
is SOX9 (SRY-related HMG-box gene 9). SOX9 is upregulated in the
developing testis but is suppressed in the ovary. Many other genes are
involved in testis development, including in Sertoli cell maturation and
Leydig cell differentiation/steroidogenesis. In addition to transcription
factors, these genes encode an array of signaling molecules and paracrine growth factors, some of which influence other organ systems. For
example, WT1 (Wilms’ tumor–related gene 1) acts early in the genetic
pathway and also regulates kidney development, whereas steroidogenic
factor 1 (SF1, NR5A1) influences both gonad and adrenal development.
Pathogenic variants causing loss of function of SF1 are found in ~10%
of XY patients with gonadal dysgenesis and impaired androgenization.
Of note, duplication of a related gene DAX1/NR0B1 impairs testis
development, revealing the exquisite sensitivity of the testis-determining
pathway to gene dosage effects.
Although ovarian development once was considered a “default”
genetic pathway, it is now clear that specific genes are expressed during
the earliest stages of ovary development. Some of these factors may
repress testis development (e.g., WNT4, R-spondin-1) (Fig. 390-2).
Once the ovary has formed, additional factors are required for normal
follicular development (e.g., follicle-stimulating hormone [FSH] receptor). Steroidogenesis in the ovary requires the development of follicles
that contain granulosa cells and theca cells surrounding the oocytes
(Chap. 392). Thus, there is relatively limited ovarian steroidogenesis
until puberty.
Germ cells also develop in a sex dimorphic manner. In the developing ovary, primordial germ cells (PGCs) proliferate and enter meiosis,
whereas they proliferate and then undergo mitotic arrest in the developing testis. PGC entry into meiosis is potentially initiated by retinoic
acid. The developing testis produces high levels of CYP26B1, an
enzyme that degrades retinoic acid, preventing PGC entry into meiosis.
Approximately 7 million germ cells are present in the fetal ovary in the
second trimester, and 1 million remain at birth. Only 400 are ovulated
during a woman’s reproductive life span (Chap. 392).
Phenotypic sex refers to the structures of the external and internal
genitalia and secondary sex characteristics. In addition to bipotential
gonads in the fetuses, they also initially possess internal and external
genitalia, which can develop along a male- or female-typical pathway,
with sex-specific development occurring as a result of hormone action
(Fig. 390-3). The developing testis releases anti-müllerian hormone
(AMH; also known as müllerian-inhibiting substance [MIS]) from
Sertoli cells and testosterone from Leydig cells. AMH acts through
specific receptors to cause regression of the müllerian structures from
60–80 days after conception. At ~60–140 days after conception, testosterone supports the maintenance of wolffian structures, including
the epididymides, vasa deferentia, and seminal vesicles. Testosterone is
the precursor for dihydrotestosterone (DHT), a potent androgen that
promotes development of the external genitalia, including the penis
and scrotum (60–100 days, and thereafter) (Fig. 390-3). The urogenital
sinus develops into the prostate and prostatic urethra in the male and
into the urethra and lower portion of the vagina in the female. The
genital tubercle becomes the glans penis in the male and the clitoris
in the female. The urogenital swellings form the scrotum or the labia
majora, and the urethral folds fuse to form the shaft of the penis and
the male urethra or the labia minora. In the female, wolffian ducts
regress and the müllerian ducts form the fallopian tubes, uterus, and
upper segment of the vagina. A female phenotype will develop in the
absence of the gonad, but estrogen is needed for maturation of the
uterus and breast at puberty.
The prenatal hormone environment is likely one of many factors
influencing aspects of gender identity and behavior. This is an area of
ongoing research and is beyond the scope of this chapter.
DISORDERS OF CHROMOSOMAL SEX
Variations in sex chromosome number and structure can present as
DSDs (e.g., 45,X/46,XY). KS (47,XXY) and TS (45,X) do not usually
present with genital ambiguity but are associated with gonadal dysfunction (Table 390-3).
■ KLINEFELTER SYNDROME (47,XXY)
Pathophysiology The classic form of KS (47,XXY) occurs after
meiotic nondisjunction of the sex chromosomes during gametogenesis
(40% during spermatogenesis, 60% during oogenesis). Other forms
of KS (including mosaic 46,XY/47,XXY [10–20%], 48,XXYY, and
48,XXXY) are less common. KS has an incidence of at least 1 in 1000
men, but ~75% of cases are not diagnosed. Of those diagnosed, historically only 10% were identified prepubertally. However, the advent
of noninvasive prenatal testing is leading to increased detection at an
earlier age.
Clinical Features KS is most commonly characterized by small
testes, infertility, gynecomastia, tall stature/increased leg length, and
hypogonadism in phenotypic males. At birth, most infants with KS
do not have clinical features, although there are higher rates of cryptorchidism and hypospadias. Most patients present in puberty with
arrested pubertal development caused by testicular insufficiency.
Others are diagnosed after puberty, based on low androgens, gynecomastia, or infertility. Testes are small and firm (median length 2.5 cm
[4 mL volume]; almost always <3.5 cm [12 mL]) and typically seem
inappropriately small for the degree of androgenization. Biopsies are
not usually necessary but typically reveal seminiferous tubule hyalinization and azoospermia. Other clinical features of KS are listed in
Table 390-3. Plasma concentrations of FSH and luteinizing hormone
(LH) are increased in most adults with 47,XXY, and plasma testosterone is decreased (50–75%), reflecting primary gonadal insufficiency.
Estradiol is often increased, resulting in gynecomastia (Chap. 391).
Patients with mosaic forms of KS have less severe clinical features, have
larger testes, and sometimes achieve spontaneous fertility.
TREATMENT
Klinefelter Syndrome
Growth, endocrine function, and bone mineralization should
be monitored, especially from adolescence. Educational and
psychological support is important for many individuals with
3000 PART 12 Endocrinology and Metabolism
Genital tubercle
Genital swelling
Urethral fold and groove
Clitoris
Labia minora
Labia majora
Vagina
B
Ovary
Fallopian
tube
Uterus
Vagina
Female Male
Female Male
Gonad
Mesonephros
Mullerian duct ..
Glans penis
Shaft of
penis
Scrotum
Penoscrotal
raphe
Wolffian duct
Urogenital
sinus
Epididymis
Testis
Vas
deferens
Seminal
vesicle
Prostate
A
FIGURE 390-3 Sex development. A. Internal urogenital tract. B. External genitalia.
KS. Androgen supplementation improves virilization, libido,
energy, hypofibrinolysis, and bone mineralization in men with
low testosterone levels but may occasionally worsen gynecomastia (Chap. 391). Gynecomastia can be treated by surgical reduction if it causes concern (Chap. 391). Fertility has been achieved by
using in vitro fertilization in men with oligospermia or with intracytoplasmic sperm injection (ICSI) after retrieval of spermatozoa
by testicular sperm extraction techniques. In specialized centers,
successful spermatozoa retrieval using this technique is possible in
>50% of men with nonmosaic KS. Results may be better in younger
men. After ICSI and embryo transfer, successful pregnancies can be
achieved in ~50% of these cases. The risk of transmitting chromosomal anomalies needs to be considered and counseling provided,
although this outcome is much less common than originally predicted. Long-term monitoring of men with KS is important given
the increased risk of breast cancer, cardiovascular disease, metabolic syndrome, osteoporosis, and autoimmune disorders. Because
most men with KS are never diagnosed, it is important that all
internists consider this diagnosis in men with these features who
might be seeking medical advice for other conditions.
■ TURNER SYNDROME
(GONADAL DYSGENESIS; 45,X)
Pathophysiology TS is caused by complete or partial loss of one
X chromosome and affects ~1 in 2500 women. Approximately onehalf of women with TS have a 45,X karyotype, ~20% have 45,X/46,XX
mosaicism, and the remainder have structural abnormalities of the X
chromosome such as X fragments, isochromosomes, rings, or Y chromosome material. The clinical features of TS result from haploinsufficiency of multiple X chromosomal genes (e.g., short stature homeobox,
SHOX), either directly or through effects on autosomal gene expression. However, imprinted genes are also proposed to be affected when
the inherited X has different parental origins.
Clinical Features TS is characterized by female external genitalia, short stature, hypergonadotropic hypogonadism, infertility, and
other phenotypic features (Table 390-3). Infants may present with
lymphedema, nuchal folds, low hairline, or left-sided cardiac defects or
later in childhood with unexplained growth failure or delayed puberty.
Although limited spontaneous pubertal development occurs in up to
30% of girls with TS (10%, 45,X; 60%, 45,X/46,XX) and up to 20%
have menarche, the vast majority of women with TS develop complete
ovarian insufficiency. Therefore, this diagnosis should be considered
in all women who present with primary or secondary amenorrhea and
elevated gonadotropin levels.
TREATMENT
Turner Syndrome
The management of girls and women with TS requires a multidisciplinary approach to address many potentially affected organ
systems according to TS practice guidelines. Individuals require
long-term monitoring by an experienced cardiologist to follow
3001 Sex Development CHAPTER 390
congenital heart defects (CHDs) (30%) (bicuspid aortic valve,
30–50%; coarctation of the aorta, 30%; aortic root dilation, 5%),
antibiotic prophylaxis for dental or surgical procedures, and serial
magnetic resonance imaging (MRI) of aortic root dimensions, as
progressive aortic root dilation is associated with increased risk
of aortic dissection. Individuals found to have congenital renal
and urinary tract malformations (30%) are at risk for urinary
tract infections, hypertension, and nephrocalcinosis. Hypertension
can occur independently of cardiac and renal malformations and
should be monitored and treated as in other patients with essential hypertension. Regular assessment of thyroid function, weight,
dentition, hearing, speech, vision, and educational issues should be
performed during childhood. Counseling about long-term growth
and fertility issues should be provided. Patient support groups are
active throughout the world and can play an invaluable role.
Short stature is common, and untreated final height rarely
exceeds 150 cm in nonmosaic 45,X TS. Recombinant growth hormone has been used in an attempt to increase growth, sometimes
with oxandrolone in older children. Girls with evidence of ovarian
insufficiency require estrogen replacement to induce breast and
uterine development, support growth, and maintain bone mineralization. Most physicians now initiate low-dose estrogen therapy
to induce puberty at an age-appropriate time (~11 years). Doses of
estrogen are increased gradually to allow development over a 2- to
4-year period. Progestins are added later to regulate withdrawal
bleeds. A very small percentage of women with TS have had spontaneous pregnancy, whereas others have achieved successful pregnancy after ovum donation and in vitro fertilization, but the risks of
cardiac complications are high, and expert counseling and management are needed. Long-term follow-up of women with TS includes
careful surveillance of sex hormone replacement and reproductive
function, bone mineralization, cardiac function and aortic root
dimensions, blood pressure, weight and glucose tolerance, hepatic
and lipid profiles, thyroid function, skin examination, and hearing.
This service is provided by a dedicated TS clinic in some centers.
■ 45,X/46,XY MOSAICISM
The phenotype of individuals with 45,X/46,XY mosaicism (sometimes
called mixed gonadal dysgenesis) can vary considerably. Some have a
predominantly female phenotype (see TS above). Most 45,X/46,XY
individuals have a male phenotype and testes, and the diagnosis is
made incidentally after amniocentesis or during investigation of
infertility. In practice, most newborns referred for assessment have
atypical genitalia and variable somatic features. There is often marked
asymmetry, with a streak gonad and hemiuterus on one side and a
partially descended dysgenetic testis and hemiscrotum on the other
side. Many children are raised as boys, but in some children, sex designation (whether to raise the baby as male or female) must be decided
by parents and the multidisciplinary team. In these children, gender
identity may be harder to predict. There is an increased risk of germ
cell cancer (GCC), up to 35% in intraabdominal gonads, so prophylactic removal of intraabdominal gonads is usually considered. Individuals raised as males often have reconstructive surgery for hypospadias
and removal of dysgenetic or streak gonads if the gonads cannot be
brought down into the scrotum. Scrotal testes can be preserved but
require regular examination for tumor development and sonography
at the time of puberty. Biopsy for carcinoma in situ is recommended
in adolescence, and testosterone supplementation may be required to
support androgenization in puberty or if low testosterone is detected
in adulthood. As 45,X/46,XY mosaicism can be associated with other
features (e.g., cardiac, renal), individuals should be monitored according to TS guidelines. Infertility is typical, but non-azoospermia or focal
spermatogenesis has been reported, highlighting the importance of
individualized approaches to management.
■ OVOTESTICULAR DSD
Ovotesticular DSD (OTDSD) is a condition in which an individual has
both ovarian and testicular tissue, either by having both an ovary and
a testis or by having an ovotestis. Most individuals with this diagnosis
have a 46,XX karyotype (especially in individuals of African ancestry),
although 46,XX/46,XY chimerism and rarely a 46,XY karyotype is
also possible. OTDSD usually presents with atypical genitalia at birth
TABLE 390-3 Possible Associated Clinical Features of Chromosomal Disorders of Sex Development (DSDs)
GENITALIA
DISORDER COMMON CHROMOSOMAL COMPLEMENT GONAD EXTERNAL INTERNAL BREAST DEVELOPMENT
Klinefelter
syndrome
47,XXY or 46,XY/47,XXY Hyalinized testes Male Male Gynecomastia
Clinical Features
Small testes, azoospermia, decreased facial and axillary hair, decreased libido, tall stature and increased leg length, decreased penile length,
increased risk of breast tumors, thromboembolic disease, learning difficulties, anxiety, speech delay and decreased verbal IQ, obesity, diabetes
mellitus, metabolic syndrome, varicose veins, hypothyroidism, systemic lupus erythematosus, epilepsy
Turner syndrome 45,X or 45,X/46,XX Streak gonad or immature
ovary
Female Hypoplastic female Immature female
Clinical Features
Infancy: lymphedema, web neck, shield chest, low-set hairline, cardiac defects and coarctation of the aorta, urinary tract malformations, and
horseshoe kidney
Childhood: short stature, cubitus valgus, short neck, short fourth metacarpals, hypoplastic nails, micrognathia, scoliosis, otitis media and
sensorineural hearing loss, ptosis and amblyopia, multiple nevi and keloid formation, autoimmune thyroid disease, visuospatial learning
difficulties
Adulthood: absent puberty and primary amenorrhea, hypertension, obesity, dyslipidemia, impaired glucose tolerance and insulin resistance,
autoimmune thyroid disease, cardiovascular disease, aortic root dilation, osteoporosis, inflammatory bowel disease, chronic hepatic dysfunction,
increased risk of colon cancer, hearing loss
45,X/46,XY
mosaicism
45,X/46,XY Testis or streak gonad Variable Variable Usually male
Clinical Features
Short stature, increased risk of gonadal tumors, some Turner syndrome features
Ovotesticular DSD 46,XX/46,XY Testis and ovary or ovotestis Variable Variable Gynecomastia
Clinical Features
Possible increased risk of gonadal tumors
No comments:
Post a Comment
اكتب تعليق حول الموضوع