You are on page 1of 31

12 Diseases of the Hypothalamus and the Pituitary Gland 171

Table 12.1 Genes that might be mutated in patients with Kallmann syndrome or IHH
Acronym Name Location Gene ID MIM Function
KAL1 Kallmann syndrome 1 sequence Xp22.32 3730 308700 Possible function in neural cell adhesion
(anosmin 1) and axonal migration
FGFR1 Fibroblast growth factor 8p11.2–p11.1 2260 136350 Binds both acidic and basic fibroblast
receptor 1 growth factors
FGF8 Fibroblast growth factor 8 10q24 2253 600483 Member of the fibroblast growth factor
(FGF) family involved in
organogenesis
PROK2 Prokineticin 2 3p13 60675 607002 Chemoattractant for neuronal precursor
cells in the olfactory bulb
PROKR2 Prokineticin receptor 2 20p12.3 128674 607123 G protein-coupled receptor for
prokineticins
CHD7 Chromodomain helicase DNA 8q12.2 55636 608765, Expressed in undifferentiated neuroepi-
binding protein 7 608892 thelium and in mesenchyme of neural
crest origin
KISS1 KiSS-1 metastasis-suppressor 1q32 3814 603286 Ligand of GPR54: stimulation of GnRH
(METASTIN) secretion
GPR54 G protein-coupled receptor 54 19p13.3 84634 604161 Receptor for Kiss-1: stimulation of GnRH
secretion
TAC3 tachykinin 3 (neurokinin B) 12q13–q21 6866 162330 Influences GnRH secretion
TACR3 tachykinin 3 (neurokinin B) 4q25 6870 162332 Receptor for tachykinin 3
receptor
GNRH1 gonadotropin-releasing 8q21–p11.2 2796 152760 Ligand of GnRH receptor
hormone 1
GNRHR Gonadotropin-releasing 4q21.2 2798 138850 Receptor for the gonadotropin-releasing
hormone receptor hormone

in the signal transduction of the fibroblast growth fac- not known in about 50% of cases, especially sporadic,
tor receptor 1 (FGFR1) (Dodè and Hardelin 2004). suggesting that a plethora of genes involved in olfactory
Indeed, in families with the autosomal dominant form bulb formation and GnRH neuron migration exist. Of
of the Kallmann syndrome, linkage analysis led to the note, while anosmia is invariably present in KAL1 muta-
identification of heterozygous mutations of the FGFR1 tions, sense of smell might be only reduced or even nor-
gene (Dodè et al. 2003), found in about 15% all cases mal in the case of all other genes. On the other hand,
(familial and sporadic). Mutations or deletions of the anosmia can be present as isolated symptom in the pedi-
KAL1 gene and of the FGFR1 (also known as KAL2) gree of subjects with Kallmann syndrome. Mutations of
gene result in the same phenotype, suggesting an inter- the FGFR1, PROK2 and PROKR2 show variable expres-
action between the two factors. The relevant ligand of sion, with symptoms ranging from isolated anosmia, IHH
FGFR1 is FGF8 and missense mutations of the FGF8 or delayed puberty even within the same family (De Roux
gene were found in patients with IHH with variable 2005; Pitteloud et al. 2005). This can be explained as
olfactory phenotype (Falardeau et al. 2008). variable phenotypic expression of a uniform gene defect.
Other genes mutated in Kallmann syndrome and The second pathogenetic mechanism of IHH is a
therefore involved in GnRH neuron migration are defect in activation of the GnRH secreting neurons
those encoding for prokineticin 2 (PROK2) and its resulting in lack of secretion or apulsatile secretion of
receptor (PROK2R) (responsible for about 10% of the gonadotropins. Known causes are mutations of the
disease) (Dodè et al. 2006) and the CHD7 gene (found KISS1 gene, the GPR54-(KISS1R-)gene, the TAC3
in 6% of sporadic cases) (Kim et al. 2008). Mutations (tachykinin 3 or neurokinin B) gene, the TACR3
of the CHD7 gene are usually found in the CHARGE (tachykinin 3 or neurokinin B receptor) gene, and - in
syndrome, a multisystem autosomal dominant disor- case of GnRH resistance - the GNRHR gene (see Table
der including anosmia and hypogonadism among the 12.1 and Sects. 2.2.2 and 2.2.3). GNRHR mutations are
symptoms. the most frequent ones (Bédécarrats and Kaiser 2007),
Notwithstanding this expanding number of genes cause subtypes of IHH varying from complete to par-
mutated in the Kallmann syndrome, the genetic defect is tial resistance to GnRH and account for less than 50%
172 H. M. Behre et al.

of familial IHH cases and a small fraction of sporadic The patients frequently present with uni- or bilateral
cases. The exact prevalence of TAC3 and TAC3R cryptorchidism, or orchidopexy has previously been
mutations is not known, yet (Topaloglu et al. 2009). performed. The scrotum may be hypoplastic and
Very recently, mutations of GnRH (gonadotropin- hypopigmented. Other signs of hypogonadism include
releasing hormone 1) have been shown to cause iso- underdevelopment of the penis and prostate, absent or
lated hypogonadotropic hypogonadism (Bouligand sparse pubic, axillary and body hair, lack of beard
et al. 2009, Chan et al. 2009). growth, enuchoid body proportions and a female dis-
tribution pattern of adipose tissue (Fig. 12.2).
Gynecomastia is a rare finding. Untreated men with
IHH and Kallmann syndrome are infertile because of
12.1.3 Clinical Picture aspermia or azoospermia. Without endocrine substitu-
tion these patients have no or reduced sexual activities.
Absent or incomplete pubertal development with Osteoporosis may occur as a secondary complication
severe hypogonadism is the clinical hallmark of both of longstanding hypogonadism.
IHH and Kallmann syndrome. The testis has a mean The variability of the clinical picture with regard to
volume of about 3 ml (normal for adult men: ≥12 ml). clinical severity and age of onset should be noted.

Fig. 12.2 Initial presentation of a 33-year-old patient with infantile penis and scrotum, and markedly subnormal testicular
Kallmann syndrome before (remarkably late) initiation of treat- volume. Voice mutation had not occurred yet in this patient.
ment with testosterone and subsequent treatment with hCG/ Gonadotropin and testosterone levels were subnormal, but LH
hMG. Infertility was the primary complaint. Typical presenta- and FSH concentrations increased appropriately in response to a
tion with signs of hypogonadism already present before puberty: GnRH bolus. Hematocrit was severely reduced, the prostate was
(a) arm span greater than height (eunuchoid body proportions), small, ejaculate analysis revealed azoospermia. Bone density as
female distribution pattern of adipose tissue, no beard growth; measured by quantitative computed tomography was reduced to
(b) straight frontal hair line; and (c) straight pubic hair line, 24% of the age-adjusted normal value
12 Diseases of the Hypothalamus and the Pituitary Gland 173

Besides the fully expressed disease, minor forms with be performed. For a period of 7 days 5 μg GnRH are
partial pubertal development or onset of GnRH defi- applied subcutaneously every 90–120 min using a por-
ciency in adulthood have been described (Seminara table minipump. If the gonadotropin response to a
et al. 1998). Notably, FGF8 mutations have been doc- GnRH bolus has normalized after such pretreatment, a
umented in the rare, adult onset form of IHH (Falardeau hypothalamic source of the hypogonadism is indicated.
et al. 2008). In contrast, a primary pituitary problem must be sus-
Anosmia, absent in IHH, represents the second pected if the gonadotrophs remain functionally resistant
obligatory symptom of Kallmann syndrome. The to a GnRH bolus. In this case mutations of the GNRHR
inability to perceive olfactory stimuli results from gene may be present. Another important differential
aplasia or hypoplasia of the olfactory bulbs and diagnosis is that between IHH/Kallmann syndrome and
tracts. The insensitivity relates only to aromatic olfac- constitutional pubertal delay. GnRH bolus testing after
tants; mucosal irritants such as ammonia evoke a nor- 36 h of pulsatile GnRH pretreatment with a minipump
mal reaction. may be helpful in this situation (see Sect. 12.5.4). A
In addition to hypogonadism and anosmia, other small subgroup of patients with IHH and Kallmann
anomalies may be associated with Kallmann syn- syndrome may also show low-normal FSH values in the
drome (Dodé and Hardelin 2004). 5–10% of patients presence of decreased LH levels (Spratt et al. 1987).
show impaired hearing or oral anomalies such as cleft Typically, patients with congenital hypogonadotro-
lip or high arched palate. Synkinesis of the extremities pic hypogonadism have significantly increased serum
or unilateral renal aplasia may occur in patients with levels of anti-Muellerian hormone, which can be low-
the X-linked disease. Numerous other malformations ered by hCG or testosterone therapy, and stimulated by
and functional problems have been described in single FSH (Young et al. 2003; Sinisi et al. 2008).
case reports, and may represent expression of the phe- Proving anosmia in Kallmann syndrome is usually
notypic variability of the syndrome. a straightforward procedure. The patient can be asked
to close his eyes and identify an aromatic substance
such as perfumed soap or coffee. More sophisticated
testing with a standardized set of olfactants may some-
12.1.4 Diagnosis times be necessary. Mucosal irritants (e.g., ammonia)
and gustatory stimuli are perceived normally in patients
If IHH or Kallmann syndrome is suspected, the most with Kallmann syndrome.
important endocrine parameters to be checked are the To the extent that a patient is able to produce a
basal serum levels of LH, FSH, testosterone, and semen sample in view of his testosterone deficiency,
estradiol. Measurement of prolactin, TSH, ACTH, low volume with azoospermia or only isolated sperm
IGF-1, growth and thyroid hormones, and cortisol are to be expected.
allows for evaluation of the other hypothalamic-pitu- Imaging procedures should include testicular and
itary axes. In unclear cases a GnRH test (see Sect. renal ultrasound (the latter to detect renal agenesis)
7.2), performed both before and after a period of pul- and magnetic resonance imaging of the hypotha-
satile GnRH stimulation, are mandatory. lamic-pituitary region. It is of utmost importance not to
Typically patients with Kallmann syndrome and overlook any intracranial mass which may present with
IHH have markedly subnormal basal levels of LH, clinical symptoms indistinguishable from IHH or
FSH and testosterone (hypogonadotropic hypogonad- Kallmann syndrome. Measurement of bone density
ism). Responsiveness of the gonadotropins to GnRH should be included in the routine diagnostic workup
stimulation is poor or altogether absent. This should (see Chap. 6).
not be misinterpreted as a sign of a primary abnormal- As Kallmann syndrome and IHH are a genetic disor-
ity at the pituitary level. Obviously the gonadotrophs der, it is important to take a meticulous family history.
only respond to a GnRH stimulus in a physiological The patient should be specifically questioned about rel-
fashion after a certain period of “priming”. atives with absent or delayed puberty, hypogonadism,
To differentiate between real pituitary pathology and infertility, or anosmia. Depending on the presence of
the reversible unresponsiveness due to the lack of pre- anosmia and the mode of inheritance, molecular genetic
ceding exposure to GnRH, the GnRH pump test may analysis may be indicated. In patients desiring children,
174 H. M. Behre et al.

genetic counselling should be offered before initiation human menopausal gonadotropin (hMG, correspond-
of therapy. The recurrence risk is 50% in autosomal ing to FSH) can be considered (Burgues and Calderon
dominant Kallmann syndrome or IHH. 1997; European Metrodin HP Group 1998; Liu et al.
2002; Bouloux et al. 2002; Warne et al. 2008). In the
male hCG and FSH must be injected intramuscularly
or subcutaneously two and three times per week,
12.1.5 Therapy respectively (Table 12.2). Recombinant hCG or LH are
also available, but are not (yet) licensed for treatment
Treatment in newly diagnosed patients with IHH or of male hypogonadotropic hypogonadism.
Kallmann syndrome is initiated by applying testoster- Treatment is continued either until sperm appear in
one for several months (see Chap. 21). This leads to the ejaculate or a pregnancy has been induced (Fig. 12.3).
rapid virilization, a higher level of general physical There is a good chance of achieving fertility in men
well-being and activity, and increased sexual drive. with IHH and Kallmann syndrome, and sperm produc-
Most patients welcome these dramatic physical and tion can be induced in nearly all patients (Fig. 12.4)
psychological changes. Once the initial goal of rapid (Liu et al. 1988, 2002; Schopohl et al. 1991; Büchter
virilization has been achieved, treatment is redirected et al. 1998; European Metrodin HP Group 1998; Barrio
at stimulating gametogenesis up to the point that et al. 1999; Bouloux et al. 2002; Warne et al. 2008).
mature spermatozoa are produced. Medication is This may necessitate treatment courses with GnRH or
changed from testosterone to either GnRH or gonado- gonadotropins that extend over 2 and more years (Fig.
tropins. This therapeutic approach is beneficial both 12.3). Cryptorchidism or markedly subnormal testicular
for patients who desire fertility soon, as well as for volume is not a contraindication to therapy. Nearly all
those who want to have children later on. In the latter such patients respond to treatment with a substantial
group spermatogenesis, once driven to full maturation, increase in testicular volume (Büchter et al. 1998; Liu
can probably be restimulated more effectively when et al. 2002; Warne et al. 2008) (Fig. 12.5).
fertility is actually desired (Büchter et al. 1998). Gonadotropins and GnRH appear to be similarly
The pulsatile application of GnRH by means of a efficient in stimulating spermatogenesis (Büchter et al.
portable minipump most closely simulates normal 1998). The therapist continues to be surprised about
physiology (Table 12.2). The pump delivers a small the low sperm concentrations with which patients
bolus of GnRH (usually 5–20 μg/pulse) every 120 min. under gonadotropin therapy induce pregnancies. The
Application is via a subcutaneous needle that has to be prerequisite for this is, of course, that female reproduc-
changed on a regular basis. The GnRH pulses induce tive functions be optimized, and the female partner
the secretion of gonadotropins from the pituitary. In should be strongly encouraged in this direction. When
turn, the gonadotropins stimulate gonadal steroid pro- this is the case, recourse to techniques of artificial
duction and gamete maturation. Often pulsatile GnRH reproduction is rarely needed.
therapy is not or only weakly effective in patients with Both GnRH and hCG/FSH therapy are costly and
mutations of the GnRH receptor (Caron et al. 1999b). can only be justified when fertility is actually desired
Alternatively, and presently more frequent, treat- or, in the initial phase of treatment, when spermato-
ment with human chorionic gonadotropin (hCG, cor- genesis is to be stimulated up to the point of sperm
responding to LH activity) and recombinant FSH or production. Once either goal has been achieved, ther-

Table 12.2 Therapeutic options for stimulation of spermatogenesis in patients with IHH or Kallmann syndrome (only those drugs
are listed that are licensed for the respective indication in Germany)
Drug Trade name Application Dose
Pulsatile GnRH Lutrelef< R> Subcutaneous, external minipump 5–20 μg per pulse every 120 min
(Zyklomat< R > pulse)
Alternatively to pulsatile GnRH
Human chorionic gonadotropin (hCG) Predalon< R > Intramuscular 1,000–2,500 IU
Brevactid< R > 2 times per week
in combination with recombinant FSH Gonal F< R > Subcutaneous 150–225 IU
Puregon< R> 2–3 times per week
12 Diseases of the Hypothalamus and the Pituitary Gland 175

IHH/KalS-GnRH
100.0
IHH/KalS-hCG/hMG
HYP-hCG/hMG
Sperm concentration (million/ml)
lower limit of normal
10.0

1.0

0.1

Azoo
0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48
Duration of therapy (months)

Fig. 12.3 Sperm concentrations in infertile patients with hypogo- spermatogenesis was achieved more rapidly in “HYP” patients
nadotropic hypogonadism (IHH/KalS = isolated hypogonadotro- with greater baseline testicular volume. Taken that the reproduc-
pic hypogonadism and Kallmann syndrome; HYP = hypogonadism tive functions in the female partner are optimal, pregnancies occur
due to pituitary dysfunction). The diagram shows sperm concen- with sperm concentrations of less than 3 million/ml. It may be
trations over the course of hCG/hMG or pulsatile GnRH treatment necessary to continue treatment over prolonged periods of time.
up to the point when the female partner conceived. Stimulation of Therapy should not be terminated prematurely

a b

Fig. 12.4 Kallmann syndrome: testicular histology (a) before and (b) during hCG/hMG therapy with presence of elongated sper-
matids (Provided by Prof. C.A. Paulsen†, MD, University of Washington, Seattle)

apy is switched to hCG alone, which maintains tes- fully sufficient to compensate for the androgen defi-
tosterone production and stimulates spermatogenesis ciency. In most patients substitution using testoster-
for a certain period (Depenbusch et al. 2002). In order one preparations (alternatively with GnRH or hCG/
to avoid renewed stimulation at a future timepoint, FSH when fertility is acutely desired) has to be life-
cryopreservation of the ejaculate should be consid- long to preserve secondary sexual characteristics and
ered (see Chap. 24). Thereafter substitution with tes- androgen-dependent functions and to prevent
tosterone is continued, which is cheaper, and which is osteoporosis.
176 H. M. Behre et al.

50 The genetic basis of the mostly sporadic, but occa-


45
sionally familial Prader-(Labhart-)Willi syndrome is

y
tit
complex and cannot be described in detail in this chap-

en
40

id
ter. Probably the phenotype is not caused by the loss or
after 6–12 months of therapy
Total testicular volume (ml)

of
functional disturbance of a single gene but of a gene

e
35

lin
cluster in the region 15q11– q13 located on the proxi-
30
mal long arm of chromosome 15. This chromosomal
25 region carries a genomic imprint. Genomic imprint-
ing can modify the expression of genes according to
20
their parental origin (Morison and Reeve 1998).
15 Among various candidate genes the role of the SNRPN
10 IHH/KalS-GnRH (small nuclear ribonucleoprotein polypeptide N) is the
IHH/KalS-hCG/hMG most established in the pathophysiology of the Prader-
5 HYP-hCG/hMG (Labhart)Willi syndrome. This gene is essential for
0 parental imprinting of the proximal long arm of chro-
0 5 10 15 20 25 30 35 40 45 50 mosome 15 (Dittrich et al. 1996).
Total testicular volume before therapy (ml) In 75% of Prader-(Labhart-)Willi syndrome patients,
Fig. 12.5 Testicular volume measured by ultrasonography in a deletion in the chromosomal region 15q11–13 is
patients with hypogonadotropic hypogonadism (IHH/KalS = iso- detectable. It is always the paternally inherited chro-
lated hypogonadotropic hypogonadism and Kallmann syndrome; mosome 15 which carries the deletion. Rarely are these
HYP = hypogonadism due to pituitary dysfunction) before treat- deletions detectable by conventional chromosome
ment and during a 6–12 month course of hCG/hMG or pulsatile
GnRH. Note the significantly larger baseline testicular volume in analysis, usually sub-microscopic microdeletions are
“HYP” patients, who, for example, in a case of pituitary tumor, involved. In addition, other structural chromosomal
develop hypogonadism after puberty. However, among “isolated anomalies such as translocations or marker chromo-
hypogonadotropic hypogonadism/KalS” patients a marked somes may be observed in patients with PWS. In
increase in testicular size can also be achieved
another 25% of the patients, maternal uniparental
disomy of 15q11–13 is the mechanism underlying
Spontaneous remission of hypogonadism was Prader-(Labhart-)Willi syndrome. Here both copies of
observed in about 10% of IHH or Kallmann patients the critical chromosomal region are contributed by the
after cessation of testosterone substitution (Raivio mother. Under normal circumstances, they should be
et al. 2007). inherited in a biparental fashion. As in deletion cases,
the active paternal allele of the Prader-(Labhart-)Willi
syndrome gene cluster is lacking. A few patients with
12.2 Prader-(Labhart-)Willi Syndrome the syndrome have “imprinting mutations” which
interfere with the process of parental imprinting of
critical chromosomal regions during gametogenesis
12.2.1 Etiology and Pathogenesis (Ohta et al. 1999).

Roughly one in 10,000 individuals has Prader-


Labhart-Willi syndrome (Prader et al. 1956), today 12.2.2 Clinical Picture and Diagnosis
usually referred to as Prader-Willi syndrome (PWS).
Because of the complex disturbance of their physi-
Upon suspicion of a PWS, diagnosis is made with the
cal and mental development (Cassidy 1997),
aid of a diagnostic score system (Holm et al. 1993). The
patients with PWS are most often seen by pediatri-
score system was revised in 2001 in order to define
cians. Today they are cared for by multiprofessional
indications for molecular-genetic diagnosis in the vari-
teams (Eiholzer and Whitman 2004). As the dis-
ous age groups (Gunay-Aygun et al. 2001). Molecular-
ease must be considered in the differential diag-
genetic diagnosis is required for definitive diagnosis.
nosis of delayed and incomplete puberty, it should
Newborns and infants with Prader-(Labhart-)Willi syn-
be familiar to the andrologist.
drome often come to clinical attention because of
12 Diseases of the Hypothalamus and the Pituitary Gland 177

generalized and profound muscular hypotonia. and highly sensitive molecular test is based on analyz-
Motor development is delayed, and feeding frequently ing the methylation pattern in the “critical” gene region
poses a major problem in the first year of life because of (Gillesen-Kaesbach et al. 1995). Three to 5 ml of EDTA
reduced fluid intake. While the children fail to thrive blood are required. However, this test does not differ-
in infancy, as toddlers they tend to become obese, entiate between deletion, uniparental disomy and an
often to a remarkable extent; leading to considerable imprint mutation. For further analysis, additional spe-
morbidity (Partsch et al. 2000). Hyperphagia persisting cial tests and blood samples of the parents are needed.
into adulthood may be difficult to control. In up to 25%
of the patients type 2 diabetes mellitus develops as a
sequel of obesity (Butler et al. 2002). Men with Prader-
(Labhart-)Willi syndrome have an average final adult
12.2.3 Therapy
height of 161.6 ± 8.1 cm (Wollmann et al. 1998). Even
in relation to the short stature, hands and feet appear No causal treatment of Prader-(Labhart-)Willi syn-
small, and the hands tend to be narrow. General hypo- drome is available (Goldstone et al. 2008). Testosterone
pigmentation can be observed in every third patient. or hCG can be given to compensate the endocrine
Aberrant craniofacial features in Prader-(Labhart-)Willi hypogonadism (see Chap. 21; Eiholzer et al. 2007).
syndrome include almond-shaped palpebral fissures Whether or not such hormonal treatment is initiated
and a narrow forehead, but usually the face does not has to be decided on an individual basis, taking into
appear very dysmorphic. Middle-grade mental retar- account the psychosocial situation of the patient. Fears
dation is present in most, but not all patients with that behavioral problems typical of PWS patients will
Prader-(Labhart-)Willi syndrome. The mental handicap worsen by inducing puberty appear unfounded
may be more pronounced in some cases. Various behav- (Eiholzer et al. 2007).
ior disorders are seen in childhood as well as adulthood Growth hormone therapy has been available for
(Cassidy 1997). several years in Germany. It not only normalizes adult
The genitalia are hypoplastic in the vast majority height (Angula et al. 2007; Lindgren and Lindberg
of males with Prader-(Labhart-)Willi syndrome. 2008), but also has a number of positive effects on
Children typically have a micropenis, uni- or bilateral PWS patients (Burman et al. 2001).
maldescended testes, and above all, a hypoplastic scro-
tum (Crino et al. 2003). Pubertal development is
delayed and incomplete. While the general state of
12.2.4 Bardet-Biedl and Laurence-Moon
virilization is poor, pubic hair may be normally devel-
oped. In the pubertal age boys with Prader-(Labhart-) Syndromes
Willi syndrome display a specific form of combined
secondary (low LH and testosterone) and primary Along with the Prader-(Labhart-)Willi syndrome, the
hypogonadism (low inhibin B and high FSH) (Eiholzer Bardet-Biedl and Laurence-Moon syndromes were
et al. 2006). In addition, Leydig cell function may be previously incorrectly considered as disorders with pri-
impaired, as indicated by a subnormal response of tes- mary hypothalamic hypogonadism. The Bardet-Biedl
tosterone to injected hCG. Testicular biopsies show and Laurence-Moon syndromes are very rare familial
atrophy of the seminiferous tubules and usually sper- disorders with predominantly autosomal recessive
matogonia are absent. It is presumed that all men with inheritance. It is not conclusive whether the Lawrence-
Prader-(Labhart-)Willi syndrome are infertile (Vogels Moon syndrome represents an independent entity. The
et al. 2008). Offspring of fathers with PWS have not Bardet-Biedl syndrome (BBS) is heterogenous. At
been described in the literature, but this issue has not present 12 BBS genes are known (Stoetzel et al. 2007).
been systematically studied. However, mutations in these 12 genes explain only
Upon clinical suspicion of PWS, triggered by about 70% of BBS cases. The mode of inheritance may
hypogonadism, slight mental retardation or deficiency, be complicated by digenic inheritance (Burghes et al.
excessive food intake (hyperphagia, compulsive eat- 2001; Katsanis 2004). Genes responsible for the Bardet-
ing), central adiposity and typical behavioral anomalies Biedl syndrome are mapped to chromosomes 2, 3, 4, 7,
(Gunay-Aygun et al. 2001), diagnosis should always be 8, 9, 11, 12, 14, 15 16, and 17 (OMIM 209900). The
confirmed by molecular-genetic analysis. A reliable Bardet-Biedl syndrome is characterized by obesity,
178 H. M. Behre et al.

progressive retinal dystrophy, renal anomalies and syndrome is best defined: it is characterized by absent
mental retardation as well as hexadactyly. Individuals or incomplete pubertal development with low levels of
with Laurence-Moon syndrome display progressive LH, FSH, testosterone and estradiol, signs of spinocer-
neurological problems, including spastic paraplegia ebellar ataxia and an atypical chorioretinal dystrophy
and ataxia (Green et al. 1989; Beales et al. 1999). (Rump et al. 1997). The increase of gonadotropins fol-
Hypogonadism and hypogenitalism occur as fac- lowing GnRH administration is subnormal, even after
ultative features in both conditions. It appears that, GnRH pretreatment. This makes pituitary involvement
in contrast to previous thinking, the hypogonadism probable. There are similarities between the Boucher-
does not have a hypothalamic or pituitary basis. With Neuhäuser syndrome and the Gordon-Holmes syn-
few exceptions adult male patients have normal LH drome (Holmes cerebellar ataxia, hypogonadotropic
and FSH serum levels, and FSH may actually be hypogonadism, (spino)-cerebellar ataxia, nystagmus,
increased. GnRH testing shows a regular gonadotropin possibly mental retardation [De Michele et al. 1993])
response (Soliman et al. 1996). Furthermore, the serum and the Oliver-McFarlane syndrome (hypogonadotro-
testosterone concentration is usually within the normal pic hypogonadism, growth retardation, retinal degen-
range. Neither ejaculate parameters nor testicular his- eration, trichomegaly of eyelashes and sparse hair
tology have been systematically studied in the Bardet- [Sampson et al. 1989]). The Gordon-Holmes syndrome
Biedl and Laurence-Moon syndromes. Some case is characterized by hypogonadotrophic hypogonadism
reports suggest that spermatogenesis may be compro- which cannot be treated by pulsatile GnRH (Quinton
mised to a variable degree. However, single cases of et al. 1999; Seminara et al. 2002).
paternity of males with Bardet-Biedl syndrome have
been described (Beales et al. 1999).
12.4 Congenital Adrenal Hypoplasia
with Hypogonadotropic
12.3 Cerebellar Ataxia Hypogonadism
and Hypogonadism
The cytomegalic form of congenital adrenal hypopla-
Clinically and etiologically cerebellar ataxias constitute sia is a rare, X-chromosomal recessive disease with a
a highly heterogenous group of disorders whose clas- prevalence of approximately 1:12,500 (Kelch et al.
sification depends on recent genetic findings (Baraitser 1984). It is usually associated with hypogonadotropic
1997; Koeppen 1998). Most cerebellar ataxias are hypogonadism (Kletter et al. 1991) The LH and FSH
genetically determined. Several dozen hereditary con- deficiency is not present during the first years of life,
ditions featuring cerebellar ataxia are known. The so- but develops later (Bassett et al. 1999; Kaiserman et al.
called pure forms become symptomatic only through 1998; Peter et al. 1998; Takahashi et al. 1997). Whether
cerebellar dysfunction. Another group of disorders the hypogonadism is caused by a pituitary or hypotha-
combines this feature with extracerebellar signs and lamic dysfunction cannot be answered conclusively.
symptoms. One of these associations is that between In some patients a hypothalamic dysfunction was
cerebellar ataxia and hypogonadism (Baraitser 1997). detected (Kletter et al. 1991; Partsch and Sippell 1989);
Within this category ataxias with hypo- or hyper- treatment with pulsatile GnRH was without success in
gonadotropic hypogonadism are to be distinguished. others (Caron et al. 1999a; Habiby et al. 1996; Kletter
The term cerebellar ataxia with hypergonadotropic et al. 1991). In addition, a gonadal defect has been sug-
hypogonadism has been applied to several different gested (Caron et al. 1999a). Therefore, it is recom-
conditions such as the Marinesco-Sjögren syndrome, mended that first the level of dysfunction should be
ataxia teleangiactasia (the Louis-Bar syndrome) and determined in each patient, and the therapeutic strat-
even untypical cases of Klinefelter syndrome. It is egy should only be chosen subsequently (see Sects.
beyond the scope of this book to discuss this poorly 12.1, 12.7 and Chap. 15). The patients need lifelong
characterized group of patients. substitution with gluco- and mineralocorticoids. Their
Similarly, cerebellar ataxia with hypogonadotro- dose must be increased in stressful situations and par-
pic hypogonadism does not represent a single entity. ticularly prior to surgical procedures. Withdrawal tri-
The autosomal recessive Boucher-Neuhäuser als are absolutely contraindicated.
12 Diseases of the Hypothalamus and the Pituitary Gland 179

In the majority of the patients the disease is caused data by Largo and Prader have been confirmed in a large-
by mutations of the DAX-1 gene (Peter et al. 1998) scale cross-sectional German study (Willers et al. 1996).
which is located on the short arm of the X chromo-
some in the region Xp21.3–21.2. The disease may Delayed puberty (pubertas tarda) is present in a
occur in the form of a “microdeletion syndrome” boy by definition if pubertal development (genital
(Budarf and Emanuel 1997) with Duchenne muscular development, testicular growth) has not started
dystrophy and glycerokinase deficiency. The hypogo- by the age of 14 years (see Fig. 12.6).
nadal pathogenesis has not been conclusively explained
(Lalli and Sassone-Corsi 2003; Okuhara et al. 2008).
Genetic heterogeneity is assumed (Muscatelli et al. 12.5.2 Etiology and Pathogenesis
1994; Zanaria et al. 1994). The clinical presentation
of Constitutional Delay of Puberty
can be quite variable within one family (Merke et al.
1999). It should be noted that adrenal insufficiency
Constitutional delay of puberty (CDP) is by far
may have its onset even after the neonatal period.
the most common cause of delayed puberty
(pubertas tarda). In males the prevalence may be
as high as 1:40.
12.5 Constitutional Delay
of Development
Activation of the GnRH pulse generator with the sub-
sequent increase of hormone production and secretion
12.5.1 Normal Onset of Puberty by endocrine organs (pituitary and testes) initiates
and Definition of Delayed pubertal development. The increasingly pulsatile
Puberty GnRH secretion is delayed in boys with CDP. To date
it is not clear which neural, endocrine, or metabolic
Normal male puberty starts with the growth of the scro- signal causes maturation of hypothalamic centers and
tum and a change in color and texture of the scrotal skin the onset of puberty. Similarly, it is not clear why the
(stage G2 at 11.2 ± 3 years, mean ± twice the standard activation of this mechanism is delayed in some chil-
deviation) (Fig. 12.6). A unilateral increase of testicular dren. Recent studies have shown that in most cases of
volume to ≥3 ml (11.8 ± 1.8 years) is the most reliable CDP an autosomal dominant pattern of inheritance
sign for the onset of puberty (Largo and Prader 1983). exists (Sedlmeyer et al. 2002). Single genetic factors
Thereafter pubertal hair growth, penis growth, growth with an autosomal dominant effect are assumed whose
spurt, beard growth and voice mutation follow. Because penetrance is modified by other genetic and environ-
of the wide variability in the age of onset of puberty, a mental influences (Sedlmeyer et al. 2002; Wehkalampi
healthy boy aged between 13 and 14 years could equally et al. 2008a). In families with several afflicted
well show all, as well as no signs of puberty (Fig. 12.6) members a significant association of CDP with a
(Largo and Prader 1983; Biro et al. 1995). Recently, the locus in the pericentromere region of chromosome 2

Start of genital development G2


Unilateral testicular volume ≥ 3ml
Start of pubic hair development P2
Fig. 12.6 Schematic diagram Start of penile growth G3
of normal pubertal develop-
Peak height velocity
ment. Means ± twice the
standard deviation are Adult penis G5
presented. The broken line Adult pubic hair P5
represents the upper age limit
End of pubertal testicular growth
for normal onset of puberty.
Pubertal development
commencing thereafter is 8 9 10 11 12 13 14 15 16 17 18
defined as pubertas tarda Age (years)
180 H. M. Behre et al.

(2p13–2q13) was identified. It is believed that this later the pubertal growth spurt occurs in life, the less
region contains a gene that is part of the biological growth is achieved during the spurt. Patients with CDP
clock responsible for timing the beginning of puberty or CDGP are likely to present with slightly eunuchoid
(Wehkalampi et al. 2008b). body proportions owing to their prolonged prepubertal
growth phase, which impairs spinal growth (Albanese
and Stanhope 1994). This mild dysproportion often
The duration and sequence of pubertal events, once persists into adulthood.
puberty has started, is usually normal in boys with
CDP. Pubertal development finally ends with com-
plete sexual maturity and normal fertility. CDP
must therefore be seen as an extreme functional 12.5.4 Diagnosis
variant of the onset of normal pubertal develop-
ment. While CDP is not a disease, the delay in In addition to anamnesis, growth curve and complete
pubertal development may be a pressing psycho- physical examination, the radiographic determination of
logical burden for the adolescent. CDP occurs as a bone age of the left hand and wrist (including the ulnar
familial or sporadic condition (Sedlmeyer and and radial epiphyses) is the most important diagnostic
Palmert 2002; Sedlmeyer et al. 2002; Wehkalampi procedure. Bone age is determined according to Greulich
et al. 2008a). and Pyle (1959). CDP is the most likely diagnosis in the
event that clinical signs are harmonic and normal in
relation to bone age. However, the diagnosis of CDP
12.5.3 Clinical Picture can only be made after excluding all other possibili-
ties. The possible causes of CDP are manifold. Fifty dif-
The children or adolescents present for two possible ferent reasons were found in a large case study
reasons: The first reason is short stature. Height is (Sedlmeyer and Palmert 2002). Thus, it is important to
decreased compared to age-related standards. Patients exclude organic forms of hypogonadism such as
who have not yet reached pubertal age may belong to Kallmann syndrome, multiple pituitary hormone defi-
this subgroup. In these cases the diagnostic term of ciencies, hypercortisolism, syndromal diseases, malab-
constitutional delay of growth and puberty (CDGP) sorption, chronic inflammatory bowel diseases and – in
is preferable. These children are usually seen in pedi- general – severe chronic diseases. Psychosocial depriva-
atric departments rather than by andrologists. tion and malnutrition may also lead to pubertal delay.
The second reason is delay or arrest of pubertal The major diagnostic problem is the differentia-
development. Patients of pubertal age, older than 14 tion between CDP and isolated/idiopathic hypogo-
years, belong to this subgroup. In addition to their lack nadotropic hypogonadism (see Sect. 12.1). This
of virilization these patients may present with short differential diagnosis is important since different ther-
stature which adds to the psychological pressure these apeutic options may arise and counselling of patients
patients often feel. Short stature is due to a transitory is different. In both disorders basal gonadotropin
partial growth hormone deficiency which requires no levels and serum testosterone are low. The response
treatment (Krajewska-Siuda et al. 2006). CDP also of LH and FSH in a standard GnRH test is also low. If
occurs in adolescents with body heights in all centiles a marked increase of LH is seen in the GnRH test (cut-
(Butenandt et al. 2005). off levels to be defined for each laboratory and each
Bone age, which serves as a measure of the indi- assay), the onset of pubertal development is imminent
vidual’s biological age, is always retarded in CDP. In (Partsch et al. 1990).
extreme cases bone age retardation may reach 5 years. The short-term GnRH pump test is an elegant
Height and pubertal stage are normal in relation to diagnostic method as it imitates physiology. For diag-
bone age (Sedlmeyer and Palmert 2002). Overall, nostic purposes pulsatile GnRH is given for 36 h (see
development is harmonic if related to bone age. Sect. 12.1.4). A standard GnRH test (100 μg) after 36 h
However, it must be emphasized that most patients of pulsatile stimulation (“priming”) has relatively good
with CDP do not reach their genetic target height diagnostic sensitivity and specificity (see Sect. 12.1.4)
(Poyrazoglu et al. 2005; Wehkalampi et al. 2007). The (Partsch et al. 1985; Smals et al. 1994). In CDP patients
12 Diseases of the Hypothalamus and the Pituitary Gland 181

the increase of LH is higher than in isolated hypogo- endogenous puberty occurs after 3 months in most
nadotropic hypogonadism patients. The criterion for patients. Treatment can be repeated if spontaneous
CDP is an increase of over 3 IU/l LH following the puberty does not occur after the first treatment period.
standard GnRH bolus test. The induction of LH and In the event that a patient with bone age >13 years fails
FSH secretion in patients with isolated hypogonado- to show spontaneous pubertal development, it is likely
tropic hypogonadism warrants a stimulation time lon- that he has isolated hypogonadotropic hypogonadism.
ger than 36 h. As an alternative to the pulsatile GnRH As an alternative to testosterone, hCG (1,000–2,000
pump test the GnRH agonist test may be used, for IU/week i.m.) or pulsatile GnRH (see Sect. 12.1.5)
which different modifications exist (Kaspers et al. can be given for 3 months. Theoretically, these treat-
2004; Kauschansky et al. 2002; Wilson et al. 2006). ment regimens have the advantage that testicular
Despite numerous diagnostic tests, however, differ- growth and spermatogenesis are stimulated. However,
ential diagnosis between CDP and isolated hypogo- for achieving the goals of treatment in patients with
nadotropic hypogonadism can be made only after CDP, hCG and pulsatile GnRH have no advantages.
long-term follow-up in difficult cases.

12.6 Secondary GnRH Deficiency


12.5.5 Treatment
12.6.1 Etiology and Pathogenesis
In most CDP patients spontaneous pubertal develop-
ment starts before the age of 20 years. The indication
for treatment is usually not the delay of puberty itself, Any disturbance in the region of the diencephalon
but the psychological pressure caused by lack of viril- can also cause impairment of hypothalamic GnRH
ization and/or short stature. There is some evidence secretion. The localization and the extent of the
that bone density may be compromised in later life in damage determine the clinical picture; an isolated
patients with CDP. Whether the mildly eunuchoid pro- disturbance of GnRH secretion is rare.
portions in adulthood can be prevented by early treat-
ment is not known to date. If the patient shows either
clinical or hormonal signs of the beginning of pubertal Tumors in the region of the diencephalon (craniophar-
development, therapeutic interventions should not be yngiomas or meningiomas) and metastases of other
initiated. Intensive counselling and, in some cases, tumors can cause GnRH deficiency. Granulomatous
psychological help should be provided. illnesses such as sarcoidosis, histiocytosis, tuberculo-
The treatment of choice for adolescents without sis, neurolues or hemochromatosis can likewise lead
signs of puberty is the intramuscular injection of tes- to a hypothalamic dysfunction. Fractures of the skull
tosterone enanthate at a dose of 50–250 mg every 4 base, ischemic and hemorrhagic lesions in the area
weeks for 3–6 months (DeLuca et al. 2001; Houchin of the hypothalamus, as well as radiotherapy of
and Rogol 1998; see Chap. 21). This treatment leads to malignant tumors or metastases in the region of the
the development of secondary sex characteristics and nasopharynx, the central nervous system, the orbita
to a growth spurt. The acceleration of bone maturation and the skull can cause a dysfunction of the hypothala-
is proportional to the gain in height age: there is no mus and/or pituitary gland (Constine et al. 1993).
decrease in predicted final height due to the short dura- Gastrointestinal illnesses such as Crohn’s or celiac
tion of treatment of 3–6 months (Kelly et al. 2003; disease – even without distinctive gastrointestinal
Poyrazoglu et al. 2005). The effect of treatment should symptoms – can lead to pubertal delay (Cacciari et al.
be documented in a standardized growth chart. 1983). Significant malnutrition (malabsorption syn-
Alternatively, oral testosterone undecanoate at a dose dromes, tumor cachexia, anorexia nervosa, etc.) and
of 20–80 mg may be administered (Brown et al. 1995; chronic diseases (sickle cell anemia, thalassemia,
Schmidt et al. 1998). renal failure etc.) can cause a hypogonadotropic dis-
After cessation of treatment, spontaneous activation turbance (see Chap. 18). The probable cause of testic-
of the hypothalamic pulse generator and subsequent ular dysfunction in these cases is a dysfunction of
182 H. M. Behre et al.

hypothalamic GnRH secretion. A similar mecha- Endocrine deficiencies are to be expected if more than
nism may also cause pubertal delay in young athletes. 75% of the hypophyseal tissue is destroyed. A distur-
bance of testicular function caused by a lack of LH and
FSH secretion is often the first sign of an acquired
insufficiency. Subsequent decreases in other pituitary
12.6.2 Clinical Picture hormones (TSH, ACTH, GH) cause dysfunction of
the thyroid gland, the adrenals and a reduction in
The clinical picture of the hypogonadism depends on growth. In addition, dysfunction of the neurohypophy-
the time of manifestation of the disorder (see Table sis causes diabetes insipidus (panhypopituitarism).
5.1) and can be disguised by other symptoms. Any sus-
picion of a hypothalamic disturbance will emerge from
the general clinical appearance.
12.7.2 Clinical Picture

The clinical symptoms are determined by the time at


12.6.3 Diagnosis which the insufficiency is manifested (see Table 5.1).
Prepubertal pituitary insufficiency causes the
Diagnosis takes account of the anamnesis, a physical changes of body proportions characteristic of hypog-
examination, endocrine investigations including stim- onadism; however, because of growth-hormone defi-
ulation tests to differentiate between hypothalamic and ciency, the typical eunuchoid, tall stature is not seen.
pituitary lesions and imaging diagnosis (magnetic res- In addition to the other androgen deficiency symptoms,
onance imaging). the clinical picture is determined by symptoms of thy-
roid and adrenal dysfunction and possibly diabetes
insipidus. Puberty does not begin, or pubertal devel-
opment is delayed and incomplete. Hypopituitarism
12.6.4 Therapy appearing after puberty causes insufficiency of the
thyroid gland and adrenal cortex, may cause diabetes
Therapy should primarily be treatment of the cause insipidus and may lead to the typical symptoms of
of the underlying disease (see Chap. 18). If this is not postpubertal androgen deficiency (see Table 5.1).
possible, symptomatic treatment is applied by sub-
stitution of the missing hormones.

12.7.3 Diagnosis

12.7 Hypopituitarism Diagnosis is made by magnetic resonance imaging of


the sella region, examination of the visual fields and
12.7.1 Etiology and Pathogenesis determination of basal hormone levels in the serum
(LH, FSH, prolactin, TSH, ACTH, GH, thyroid hor-
mones, cortisol, IGF-1) and the absence of a rise in
pituitary hormone levels during the combined pitu-
The most frequent causes of pituitary insufficiency
itary stimulation test (100 μg GnRH, 200 μg TRH
are tumors (gonadotropin, prolactin, growth hor-
[thyroid-stimulating hormone releasing hormone],
mone, TSH, ACTH secreting or hormone-inac-
100 μg GRH [growth hormone releasing hormone],
tive pituitary adenomas) or metastases of the
100 μg CRH [corticotropin releasing hormone] intra-
pituitary and the hypophyseal stalk, as well as
venously; blood to be sampled before and 30, 60 and
post-operative states and radiotherapy of the pitu-
90 min after injection of releasing hormones). If a large
itary area. Likewise, trauma, infections, hemo-
pituitary tumor is suspected, pituitary stimulation tests
chromatosis and vascular disorders can lead to
are relatively contra-indicated because of possible
pituitary insufficiency.
complications such as acute tumor necrosis.
12 Diseases of the Hypothalamus and the Pituitary Gland 183

12.7.4 Therapy aggregation and prolactin deficiency point to muta-


tions in the PROP1 gene. A series of further transcrip-
tion factors with their associated diseases has been
If a pituitary tumor is responsible for the pituitary
identified in recent years (Mehta and Dattani 2008).
insufficiency, neurosurgical removal should usually
An important differential diagnosis of hypopituitar-
be performed, which is preferably accomplished by
ism during childhood is septo-optic dysplasia (Willnow
the trans-sphenoidal route with protection of the
et al. 1996). This is a highly heterogenous disease with
healthy pituitary tissue. After surgery, regeneration of
a variable phenotype consisting of optic nerve anoma-
the remaining pituitary tissue is possible. Some tumors
lies, agenesis of the septum pellucidum and pituitary
can be primarily irradiated. Prolactinomas (both
hormone deficiency. The prevalence is 6.3–10.9 in
micro- as well as macroprolactinomas) are treated pri-
100,000. Hypogonadotrophic hypogonadism is not
marily by medical drugs (see Sect. 12.9).
obligatory, but always occurs in septo-optical dyspla-
Substitution therapy of the absent hormones (corti-
sia along with other pituitary deficiencies. The genes
sol, thyroid hormones, growth hormone, testosterone,
identified as causal to date are HESX1, SOX2 and
possibly ADH) is required until restoration of pituitary
SOX3 (Kelberman and Dattani 2008).
function, if neurosurgical removal is not possible or if
A familial form of X chromosomal recessive pan-
the pituitary is completely destroyed. Infertility is
hypopituitarism is associated with variable degrees of
treated by hCG/FSH injections (see Sect. 12.1.5 and
mental retardation (Lagerström-Fermér et al. 1997). A
Figs. 12.3, 12.4). Compared to gonadotropin therapy
genetic amplification caused by interstitial duplication
for IHH, induction of pregnancy in partners of patients
in the Xq26.1-q27.3 region containing the SOX3 gene
with hypopituitarism is more successful, especially if
is assumed to be causal (Solomon et al. 2004).
the pituitary insufficiency only occurs after completion
of puberty. Once spermatogenesis has been induced
physiologically, it is easier to reinduce it using gonad-
otropins than to stimulate the testis for the first time. 12.8 Isolated LH or FSH Deficiency

Isolated LH or FSH deficiency are exceptionally rare


12.7.5 Hypopituitarism in Heritable forms of IHH in which only one of the two gonadotro-
pins is lacking. In isolated LH deficiency (Pasqualini
Disorders of Pituitary
syndrome [Pasqualini and Burr 1950], “fertile
Development eunuchs”) there is a striking discordance between the
clinical picture of hypogonadism, already evident pre-
Congenital hypopituitarism is rare, but can be caused pubertally, and almost normal testicular volume.
by mutations of transcription factor genes. These fac- Testicular histology shows qualitatively preserved,
tors are important for pituitary development. A gene quantitatively reduced spermatogenesis and atrophy
defect of the transcription factor POU1FI (formerly of the Leydig cells. Endocrine investigations show
Pit-1) leads to pituitary deficiency of growth hormone, decreased LH and normal FSH serum levels. A
prolactin and TSH, but not to hypogonadism. The gene homozygous mutation of the GNRHR gene was dem-
defect of the transcription factor PROP1 causes an onstrated in one case (Pitteloud et al. 2001).
additional deficiency of LH and FSH. PROP1 muta- Administration of hCG (LH activity) led to normaliza-
tions are the most frequent cause of congenital panhy- tion of testosterone serum levels and to quantitatively
popituitarism (Mehta and Dattani 2008). The normal spermatogenesis. Interestingly, following ces-
inheritance is autosomal recessive. The clinical appear- sation of hCG therapy, the patient demonstrated rever-
ance is variable. Short stature and hypothyroidism are sal of hypogonadism with normal serum testosterone
predominant during the first years of life. Most of the levels.
patients experience spontaneous puberty; gonadotro- Inactivating mutations of the LH-β subunit gene are
pin deficiency develops later in life (Deladoey et al. rare and, only four patients have been identified so far
1999; Flück et al. 1998). Hypogonadism is a constant (Lofrano-Porto et al. 2008 and references therein).
symptom in adults (Rosenbloom et al. 1999). Familial Men with LHB mutations (homozygous or compound
184 H. M. Behre et al.

heterozygous) present with hypogonadism, delayed PRF


puberty and small testicular volume. Serum FSH is
elevated while LH levels range from undetectable to c PIF
elevated, depending on whether the mutation disrupts
the immunogenicity of the molecule. Testicular histol- e
ogy shows an arrest of spermatogenesis and absence of
d
Leydig cells. Long-term hCG treatment results in nor- Hypothalamus
mal testicular volume, virilization and induction of
spermatogenesis.
Isolated FSH deficiency due to mutations of the
FSHB gene is rare (only three cases described so far)
(Lofrano-Porto et al. 2008 and references therein). All
cases presented with azoospermia but two men had b

normal puberty with normal to low normal testoster-


one and high LH levels, while the third man had low
Pituitary gland
testosterone and absent puberty.
Occasionally isolated FSH deficiency was demon- a
strated in the presence of a normal FSHB gene
sequence. These men have decreased spermatogenesis,
while LH activity and thus androgenization is normal Fig. 12.7 Schematic diagram of possible causes of hyperpro-
lactinemia: (a) prolactin-secreting tumor; (b) inhibition of trans-
(Mantovani et al. 2003). The cause of FSH deficiency port of the prolactin-inhibiting factor (PIF, dopamine), e.g., by a
in these patients is unknown. suprasellar pituitary tumor; (c) destruction of the PIF-producing
hypothalamic nuclei, e.g., by a tumor; (d) pharmacological inhi-
bition of the secretion or biological action of PIF; (e) increased
secretion of hypothalamic prolactin-releasing factor (PRF)

12.9 Hyperprolactinemia
Classification of prolactinomas should not consider only
the size of the adenomas but also their rate of growth, as
12.9.1 Etiology and Pathogenesis macroprolactinomas also once began as small, rapidly
growing adenomas. The basal level of prolactin is cor-
The peptide hormone prolactin, which is produced in related, within broad limits, with the size of the adenoma.
the anterior lobe of the pituitary gland, has no known Malignant prolactinomas are extremely rare (Mancini
physiological function in the adult male. The causes of et al. 2008). Familial prolactinomas have also been
hyperprolactinemia in man are multiple (Fig. 12.7). described (Berezin and Karasik 1995; Melmed 2008),
Small increases of prolactin levels can be caused by but in most cases prolactinomas occur sporadically.
physical or psychological “stress”. In certain patients, Prolactin secretion by the pituitary is regulated by the
even the atmosphere of the consulting room or the suppressive effect of dopamine secreted by the hypo-
announcement of taking blood can cause an increase in thalamus (Fig. 12.7). Lesions of the hypothalamus or the
prolactin levels. hypophyseal stalk can, therefore, induce excessive pro-
lactin secretion (disconnection hyperprolactinemia)
(Melmed 2008). Pituitary tumors not secreting pro-
lactin, craniopharyngiomas and other processes in the
Prolactin-secreting pituitary adenomas (prolacti-
sella region can also cause hyperprolactinemia. Primary
nomas) are the most frequent cause of hyperpro-
hypothalamic disturbances can probably cause an
lactinemia. Prolactinomas can be divided into
increased stimulation of the pituitary by increased
microprolactinomas (with a diameter <10 mm) and
levels of prolactin releasing factors (Fig. 12.7), as is
macroprolactinomas (Fig. 12.9). The latter are
assumed for TRH, vasopressin and vasoactive intestinal
large, rapidly proliferating tumors that exceed the
polypeptides (VIP) (Mancini et al. 2008). In most cases
margins of the sella and can lead to the destruction of
of disconnection hyperprolactinemia serum prolactin
surrounding structures, particularly the optic nerves.
levels do not exceed 2,000 mU/l; higher levels are
12 Diseases of the Hypothalamus and the Pituitary Gland 185

usually found only in macroprolactinomas (Karavitaki


et al. 2006).
Drug-induced hyperprolactinemia is caused by
the dopamine-antagonistic effects of the substances
administered (e.g., butyrophenones, phenothiazines,
imipramine, metoclopramide), their interference with
dopamine synthesis (e.g., α-methyldopa), the depletion
of dopamine stores (e.g., reserpine) or the direct stimu-
lation of prolactin synthesis and secretion (e.g.,
H2-blockers, estrogens). Additional important causes
for hyperprolactinemia are chronic renal failure and
hypothyroidism (Schlechte 2003).
Hyperprolactinemia can cause disturbances of male
reproductive functions via various mechanisms. At the
hypothalamic level, hyperprolactinemia can impair
pulsatile GnRH release. As a consequence of sup-
pressed LH and FSH levels, secondary gonadal insuf-
ficiency develops with low testosterone levels and Fig. 12.8 Magnetic resonance image of a newly diagnosed
suppressed spermatogenesis. Hyperprolactinemia does macroprolactinoma
not cause simultaneous gonadotropin deficiency in all
cases: sellar and suprasellar processes of different
kinds can cause both hyperprolactinemia (Fig. 12.7) hormone levels (LH, FSH, TSH, ACTH, GH, thyroid
and a simultaneous blockade of GnRH flux to the hormones, cortisol, IGF-1; as described in Sect. 12.7.3)
gonadotropic cells of the pituitary. are measured and, in certain cases, a combined pitu-
The second, and clinically more important, mecha- itary function test (GnRH, TRH, GRH, CRH) may be
nism of impaired testicular function due to hyperpro- performed.
lactinemia is the displacement and destruction of In stress-induced hyperprolactinemia the basal pro-
gonadotropic cells of the pituitary by macroprolac- lactin levels generally do not exceed twice the upper
tinomas, resulting in suppression of LH and FSH levels normal limit of 500 mU/l. Very high values (>5,000 mU/l)
(see Sect. 12.7, Fig. 12.8). A direct inhibitory effect of are typical for a macroprolactinoma; serum levels, how-
prolactin on the testis has not yet been demonstrated. ever, can be variable. In each case of repeatedly mea-
sured, significantly increased prolactin levels magnetic
resonance imaging of the hypothalamus/pituitary
12.9.2 Clinical Picture region should be performed to check for a prolactinoma
or other tumors which might cause hyperprolactinemia.
Hyperprolactinemia in men is manifested clinically by Likewise, a visual field examination is performed if
signs of androgen deficiency and infertility. Primarily, suprasellar expansion of the tumor is suspected.
some of the patients complain of disturbances of In general, dynamic tests of prolactin secretion are
libido and erectile function, and may rarely develop not suited for differential diagnosis of hyperprolactine-
gynecomastia and galactorrhea. Some men with mia (Schlechte 2003; Casanueva et al. 2006).
documented hyperprolactinemia are free of symptoms
and complaints. Processes in the sella region can cause
additional symptoms according to location and size,
e.g., headaches or visual field defects. 12.9.4 Therapy

There is no need for treatment of stress-induced hyper-


12.9.3 Diagnosis prolactinemia. For therapy of prolactinomas primarily
the dopamine agonists are used. Long-range experi-
Diagnosis is based on increased prolactin serum lev- ence exists for bromocriptine (Pravidel®, Kirim® and
els. For evaluation of hypophyseal function, the basal others) (Mancini et al. 2008), which has been applied
186 H. M. Behre et al.

for over 25 years. Because of the hypotensive effect of doses increased cardiac valve disease was also reported
bromocriptine, therapy should begin with low doses for prolactinoma patients (Colao et al. 2008). In view of
given in the late evening just before the patient retires these ambiguous connections it is presently recom-
(1.25 mg/day). The dose is raised slowly and is distrib- mended that patients with prolactinomas undergo
uted over the course of the day (morning – noon – eve- echocardiography before and during long-range therapy
ning), until prolactin levels become normal (Fig. 12.9). with cabergoline (Kars et al. 2008; Sherlock et al.
The decrease of prolactin will be accompanied by a 2009).
diminution in size of the prolactinoma (Bevan et al. In the event of side-effects with bromocriptine or
1992). At the beginning of therapy, side-effects can cabergoline, alternatively quinagolide (Norprolac®),
appear as decreased blood pressure, fatigue, headaches lisuride (Dopergin®) or metergoline (Liserdol®) are
or gastrointestinal disturbances; occasional long-term available.
side-effects include headaches, dry nasal mucosa, and For patients with prolactinomas on long-term treat-
digital vasospasms upon exposure to cold (Mancini ment with dopamine agonists who no longer show
et al. 2008). tumor tissue in magnetic resonance imaging with-
Because of high efficacy and fewer side-effects than drawal trials can be attempted after 2–3 years (Calao
bromocriptine, today the long-lasting, dopamine ago- et al. 2003; Gillam et al. 2006; Casanueva et al. 2006).
nist cabergoline (Dostinex® and others) can be used After cessation of therapy, serum prolactin should be
(De Rosa et al. 1998; Cannavo et al. 1999; Gillam et al. monitored regularly especially during the first year
2006). Usually therapy is initiated with 0.25–0.5 mg (Gillam et al. 2006; Schlechte 2007). Permanent nor-
once or twice weekly and is increased monthly until malization of prolactin without renewed tumor growth
prolactin levels normalize (Casanueva et al. 2006). is often variously observed depending on studies and
Recently cardiac valve disease was reported as a poten- initial findings, may, however, be expected in 20–30%
tial side-effect in Parkinson patients taking high doses of treated patients with macroprolactinomas (Gillam
of cabergoline (>3 mg daily) (Zanettini et al. 2007). et al. 2006; Schlechte 2007).
These effects were not seen for the usual dose of Today, trans-sphenoidal excision of prolactino-
0.25–3 mg/week given patients with prolactinoma (Kars mas is rarely indicated because of the effective drug
et al. 2008; Herring et al. 2009); however, at higher treatments available, i.e., in less than 10% of patients,

22000

20000 Bromocriptine dose 20

18000 Serum prolactin 18

16000 16

14000 14
Bromocriptine (mg)

12000 12
Prolactin (mU/l)

10000 10

8000 8

6000 6
Fig. 12.9 Graphic display of
prolactin serum levels during 4000 4
dopamine agonist therapy 2000 2
(Pravidel®) in a patient with a
macroprolactinoma. After
initial suppression of prolactin 600 1
levels by high doses of
300
Pravidel® it is possible to
reduce the dose slowly, while 0 0
prolactin levels remain 0 4 8 12 16 20 24 28 32 36 40 44 48
suppressed Duration of therapy (weeks)
12 Diseases of the Hypothalamus and the Pituitary Gland 187

and only when pharmacological treatment is ineffec- References


tive (Casanueva et al. 2006; Gillam et al. 2006; Mancini
et al. 2008). The primary indication for neurosurgery Albanese A, Stanhope R (1994) Pathogenetic mechanisms and
or neuroradiotherapy is hyperprolactinemia caused by management priorities in constitutional delay. In: Savage MO,
other tumors (Fig. 12.7). Bourguignon JP, Grossman AB (eds) Frontiers in paediatric
neuroendocrinology. Blackwell, Oxford, pp 33–37
Under effective therapy with dopamine agonists,
Angulo MA, Castro-Magna M, Lamerson M, Arguello R,
normal LH pulsatility reappears and testosterone serum Accacha S, Khan A (2007) Final adult height in children
levels soon rise. Persisting androgen deficiency must with Prader-Willi syndrome with and without growth hor-
be treated with adequate androgen substitution (see mone treatment. Am J Med Genet 143A:1456–1461
Baraitser M (ed) (1997) Cerebellar ataxia. In: The genetics of
Chap. 21). At the beginning of dopamine agonist ther-
neurological disorders, 3rd edn. Oxford University Press,
apy, however, a “wait and see” approach can be taken Oxford, pp 146–165
according to the degree of androgen deficiency, or Barrio R, de Luis D, Alonso M, Lamas A, Moreno JC (1999)
transdermal or oral androgen therapy can be initiated in Induction of puberty with human chorionic gonadotropin
and follicle-stimulating hormone in adolescent males with
order to interfere with pituitary function as little as pos-
hypogonadotropic hypogonadism. Fertil Steril 71:244–248
sible. If hypogonadism persists despite therapy, or if Bassett JH, O’Halloran DJ, Williams GR, Beardwell CG,
neurosurgery or neuroradiotherapy has been performed, Shalet SM, Thakker RV (1999) Novel DAX-1 mutations in
resulting in a loss of pituitary function, adequate testos- X-linked adrenal hypoplasia congenita and hypogonadotro-
pic hypogonadism. Clin Endocrinol 50:69–75
terone substitution therapy is necessary (see Chap. 21)
Beales PL, Elcioglu N, Woolf AS, Parker D, Flinter FA (1999)
in addition to substitution with thyroid hormones and New criteria for improved diagnosis of Bardet-Biedl syn-
cortisol (see Sect. 12.7). In patients with persistent drome: results of a population survey. J Med Genet 36:
gonadotropin suppression seeking paternity, hCG plus 437–446
Bédécarrats GY, Kaiser UB (2007) Mutations in the human
FSH therapy can be initiated (see Sect. 12.1)
gonadotropin-releasing hormone receptor: insights into
receptor biology and function. Semin Reprod Med 25:
368–378
Berezin M, Karasik A (1995) Familial prolactinoma. Clin
12.10 Gonadotropin-Secreting Tumors Endocrinol 42:483–486
Bevan JS, Webster J, Burke C, Scanlon MF (1992) Dopamine
agonists and pituitary tumor shrinkage. Endocr Rev 13:
Histologically verified gonadotropin-secreting pitu- 220–240
itary tumors are relatively frequent (approximately Biro FM, Lucky AW, Huster GA, Morrison JA (1995) Pubertal
staging in boys. J Pediatr 127:100–102
25% of all pituitary tumors) (Saeger et al. 2007). Only
Bouligand J, Ghervan C, Tello JA, Brailly-Tabard S, Salenave S,
in isolated cases do these tumors secrete intact LH and/ Chanson P, Lombès M, Millar RP, Guiochon-Mantel A,
or FSH and thus cause precocious puberty or supra- Young J (2009) Isolated familial hypogonadotropic hypogo-
physiological serum testosterone or macroorchia nadism and a GNRH1 mutation. N Engl J Med
360:2742–2748
(Chaidaran and Klibanski 2002). In most cases these
Bouloux P, Warne DW, Loumaye E, FSH Study Group in Men’s
tumors become clinically evident by significant tumor Infertility (2002) Efficacy and safety of recombinant human
growth, resulting in neurological symptoms and visual follicle-stimulating hormone in men with isolated hypogo-
field defects (Jaffe 2006). nadotropic hypogonadism. Fertil Steril 77:270–273
Brown DC, Butler GE, Kelnar CJ, Wu FC (1995) A double blind,
Standard therapy for gonadotropin-secreting mac-
placebo controlled study of the effects of low dose
roadenomas (diameter ≥ 1 cm) is trans-sphenoidal sur- testosterone undecanoate on the growth of small for age,
gery (Jaffe 2006). Because of the generally slow prepubertal boys. Arch Dis Child 73:131–135
growth of microadenomas (diameter < 1 cm), observa- Büchter D, Behre HM, Kliesch S, Nieschlag E (1998) Pulsatile
GnRH or human chorionic gonadotropin/human menopausal
tion accompanied by regular endocrinological moni-
gonadotropin as effective treatment for men with hypogo-
toring and MRI seems justified in the absence of nadotropic hypogonadism: a review of 42 cases. Eur J
clinical symptoms (Jaffe 2006). No effective drug ther- Endocrinol 139:298–303
apy of gonadotropin-secreting pituitary tumors has yet Budarf ML, Emanuel BS (1997) Progress in the autosomal seg-
mental aneusomy syndromes (SASs): single or multi-locus
been established; radiotherapy is only indicated in spe-
disorders? Hum Molec Genet 6:1657–1665
cial cases such as residual or recurrent tumors after Burghes AH, Vaessin HE, de La Chapelle A (2001) The land
trans-sphenoidal surgery (Chaidarun and Klibanski between Mendelian and multifactorial inheritance. Science
2002; Melmed 2008). 293: 2213–2214
188 H. M. Behre et al.

Burgues S, Calderon MD (1997) Subcutaneous self-administra- function after radiation for brain tumors. N Engl J Med 328:
tion of highly purified follicle stimulating hormone and 87–94
human chorionic gonadotrophin for the treatment of male Crino A, Schiaffini R, Ciampalini P, Spera S, Beccaria L,
hypogonadotrophic hypogonadism. Spanish Collaborative Benzi F, Bosio L, Corrias A, Gargantini L, Salvatoni A,
Group on Male Hypogonadotropic Hypogonadism. Hum Tonini G, Trifiro G, Livieri C, Genetic obesity study group
Reprod 12:980–986 of Italian Society of Pediatric Endocrinology and Diabetology
Burman P, Ritzen EM, Lindgren C (2001) Endocrine dysfunc- (SIEDP) (2003) Hypogonadism and pubertal development
tion in Prader-Willi syndrome: a review with special refer- in Prader-Willi-syndrome. Eur J Pediatr 162:327–333
ence to GH. Endocr Rev 22:787–799 De Luca F, Argente J, Cavallo L, Crowne E, Delemarre-Van de
Butenandt O, Bechtold S, Meidert A (2005) Final height in Waal HA, De Sanctis C, Di Maio S, Norjavaara E, Oostdijk
patients with constitutional delay of growth and develop- W, Severi F, Tonini G, Trifiro G, Voorhoeve PG, Wu F,
ment from tall statured families. J Pediatr Endocrinol Metab International Workshop on Management of Puberty for
18:165–169 Optimum Auxological Results (2001) Management of
Butler JV, Whittington JE, Holland AJ, Boer H, Clark D, Webb puberty in constitutional delay of growth and puberty. J
T (2002) Prevalence of, and risk factors for, physical ill- Pediatr Endocrinol Metab 14(2):953–957
health in people with Prader-Willi syndrome: a population- De Michele G, Filla A, Striano S, Rimoldi M, Campanella G
based study. Dev Med Child Neurol 44:248–255 (1993) Heterogenous findings in four cases of cerebellar
Cacciari E, Salardi S, Lazzari R (1983) Short stature and celiac ataxia associated with hypogonadism (Holmes’ type ataxia).
disease: a relationship to consider even in patients with no Clin Neurol Neurosurg 95:23–28
gastrointestinal tract symptoms. J Pediatr 103:708–711 De Rosa M, Colao A, Di Sarno A, Ferone D, Landi ML,
Cannavo S, Curto L, Squadrito S, Almoto B, Vieni A, Trimarchi Zarrilli S, Paesano L, Merola B, Lombardi G (1998)
F (1999) Cabergoline: a first-choice treatment in patients Cabergoline treatment rapidly improves gonadal function in
with previously untreated prolactin-secreting pituitary ade- hyperprolactinemic males: a comparison with bromocrip-
noma. J Endocrinol Invest 22:354–359 tine. Eur J Endocrinol 138:286–293
Caron P, Imbeaud S, Bennet A, Plantavid M, Camerino G, De Roux N (2005) Isolated gonadotropic deficiency with and
Rochiccioli P (1999a) Combined hypothalamic-pituitary- without anosmia: a developmental defect or a neuroendo-
gonadal defect in a hypogonadic man with a novel mutation crine regulation abnormality of the gonadotropic axis. Horm
in the DAX-1 gene. J Clin Endocrinol Metab 84: Res 64(2):48–55
3563–3569 Deladoey J, Flück C, Büyükgebiz A, Kuhlmann BV, Eblé A,
Caron P, Chauvin S, Christin-Maitre S, Bennet A, Lahlou N, Hindmarsh PC, Wu W, Mullis PE (1999) “Hot spot” in the
Counis R, Bouchard P, Kottler M-L (1999b) Resistance of PROP1 gene responsible for combined pituitary hormone
hypogonadic patients with mutated GnRH receptor genes to deficiency. J Clin Endocrinol Metab 84:1645–1650
pulsatile GnRH administration. J Clin Endocrinol Metab Depenbusch M, von Eckardstein S, Simoni M, Nieschlag E
84:990–996 (2002) Maintenance of spermatogenesis in hypogonadotro-
Casanueva FF, Molitch ME, Schlechte JA, Abs R, Bonert V, pic hypogondal men with hCG alone. Eur J Endocrinol
Bronstein MD, Brue T, Cappabianca P, Colao A, Fahlbusch R, 147:617–624
Fideleff H, Hadani M, Kelly P, Kleinberg D, Laws E, Marek Dittrich B, Buiting K, Korn B, Rickard S, Buxton J, Saitoh S,
J, Scanlon M, Sobrinho LG, Wass JA, Giustina A (2006) Nicholls RD, Poustka A, Winterpacht A, Zabel B, Horsthemke
Guidelines of the Pituitary Society for the diagnosis and B (1996) Imprint switching on human chromosome 15 may
management of prolactinomas. Clin Endocrinol 65: involve alternative transcripts of the SNRPN gene. Nat
265–273 Genet 14:163–170
Cassidy SB (1997) Prader-Willi syndrome. J Med Genet 34: Dodé C, Hardelin JP (2004) Kallmann syndrome: fibroblast growth
917–923 factor signaling insufficiency? J Mol Med 82:725–734
Chaidarun SS, Klibanski A (2002) Gonadotropinomas. Semin Dodé C, Levilliers J, Dupont JM, De Paepe A, Le Dû N, Soussi-
Reprod Med 20:339–348 Yanicostas N, Coimbra RS, Delmaghani S, Compain-Nouaille
Chan YM, de Guillebon A, Lang-Muritano M, Plummer L, S, Baverel F, Pêcheux C, Le Tessier D, Cruaud C, Delpech M,
Cerrato F, Tsiaras S, Gaspert A, Lavoie HB, Wu CH, Crowley Speleman F, Vermeulen S, Amalfitano A, Bachelot Y,
WF Jr, Amory JK, Pitteloud N, Seminara SB (2009) GNRH1 Bouchard P, Cabrol S, Carel JC, Delemarre-van de Waal H,
mutations in patients with idiopathic hypogonadotropic Goulet-Salmon B, Kottler ML, Richard O, Sanchez-Franco F,
hypogonadism. Proc Natl Acad Sci USA. 106:11703–11708 Saura R, Young J, Petit C, Hardelin JP (2003) Loss-of-
Colao A, Di Sarno A, Cappabianca P, Di Somma C, Pivonello R, function mutations in FGFR1 cause autosomal dominant
Lombardi G (2003) Withdrawal of long-term cabergoline Kallmann syndrome. Nat Genet 33:463–465
therapy for tumoral and nontumoral hyperprolactinemia. Dodé C, Teixeira L, Levilliers J, Fouveaut C, Bouchard P,
N Engl J Med 349:2023–2033 Kottler ML, Lespinasse J, Lienhardt-Roussie A, Mathieu M,
Colao A, Galderisi M, Di Sarno A, Pardo M, Gaccione M, Moerman A, Morgan G, Murat A, Toublanc JE, Wolczynski S,
D’Andrea M, Guerra E, Pivonello R, Lerro G, Lombardi G Delpech M, Petit C, Young J, Hardelin JP (2006) Kallmann
(2008) Increased prevalence of tricuspid regurgitation in syndrome: mutations in the genes encoding prokineticin-2
patients with prolactinomas chronically treated with caber- and prokineticin receptor-2. PLoS Genet 2:e175
goline. J Clin Endocrinol Metab 93:3777–3784 Eiholzer U, Whitman BY (2004) A comprehensive team
Constine LS, Woolf PD, Cann D, Mick G, McCormick K, approach to the management of patients with Prader-Willi
Raubertas RF, Rubin P (1993) Hypothalamic-pituitary dys- syndrome. J Pediatr Endocrinol Metab 17:1153–1175
12 Diseases of the Hypothalamus and the Pituitary Gland 189

Eiholzer U, l’Allemand D, Rousson V, Schlumpf M, Gasser T, Houchin LD, Rogol AD (1998) Androgen replacement in chil-
Girard J, Grüters A, Simoni M (2006) Hypothalamic and dren with constitutional delay of puberty: the case for
gonadal components of hypogonadism in boys with Prader- aggressive therapy. Bailliere Clin Endoc 12:427–440
Labhart-Willi syndrome. J Clin Endocrinol Metab 91: Jaffe CA (2006) Clinically non-functioning pituitary adenoma.
892–898 Pituitary 9:317–321
Eiholzer U, Grieser J, Schlumpf M, l’Allemand D (2007) Kaisermann KB, Nakamoto JM, Geffner ME, McCabe ER
Clinical effects of treatment for hypogonadism in male ado- (1998) Minipuberty of infancy and adolescent pubertal func-
lescents with Prader-Labhart-Willi syndrome. Horm Res tion in adrenal hypoplasia congenita. J Pediatr 133:300–302
68:178–184 Kallmann FJ, Schoenfeld WA, Barrera SE (1944) The genetic
European Metrodin HP Study Group (1998) Efficacy and safety aspects of primary eunuchoidism. Amer J Ment Defic
of highly purified urinary follicle-stimulating hormone with 48:203–236
human chorionic gonadotropin for treating men with iso- Karavitaki N, Thanabalasingham G, Shore HC, Trifanescu R,
lated hypogonadotropic hypogonadism. Fertil Steril 70: Ansorge O, Meston N, Turner HE, Wass JA (2006) Do the
256–262 limits of serum prolactin in disconnection hyperprolactinae-
Falardeau J, Chung WC, Beenken A, Raivio T, Plummer L, mia need re-definition? A study of 226 patients with histo-
Sidis Y, Jacobson-Dickman EE, Eliseenkova AV, Ma J, logically verified non-functioning pituitary macroadenoma.
Dwyer A, Quinton R, Na S, Hall JE, Huot C, Alois N, Pearce Clin Endocrinol 65:524–529
SH, Cole LW, Hughes V, Mohammadi M, Tsai P, Pitteloud N Kars M, Pereira AM, Bax JJ, Romijn JA (2008) Cabergoline and
(2008) Decreased FGF8 signaling causes deficiency of cardiac valve disease in prolactinoma patients: additional
gonadotropin-releasing hormone in humans and mice. J Clin studies during long-term treatment are required. Eur J
Invest 118:2822–2831 Endocrinol 159:363–367
Flück C, Deladoey J, Rutishauser K, Eblé A, Marti U, Wu W, Kaspers S, Richter-Unruh A, Schmittmann-Ohters K, Hauffa BP
Mullis PE (1998) Phenotypic variability in familial com- (2004) Buserelin testing in 70 adolescents with delayed
bined pituitary hormone deficiency caused by a PROP1 gene puberty. Horm Res 62(2):80(P2–282)
mutation resulting in the substitution of Arg → Cys at codon Katsanis N (2004) The oligogenic properties of Bardet-Biedl
120 (R120C). J Clin Endocrinol Metab 83:3727–3734 syndrome. Hum Mol Genet 13 Spec No 1:R65–71
Gillam MP, Molitch ME, Lombardi G, Colao A (2006) Advances Kauschansky A, Dickerman Z, Phillip N, Weintrob N, Strich D
in the treatment of prolactinomas. Endocr Rev 27:485–534 (2002) Use of GnRH agonist and human chorionic gonado-
Gillessen-Kaesbach G, Gross S, Kaya-Westerloh S, Passarge E, trophin tests for differentiating constitutional delayed
Horsthemke B (1995) DNA methylation based testing of puberty from gonadotrophin deficiency in boys. Clin
450 patients suspected of having Prader-Willi syndrome. Endocrinol 56:603–607
J Med Genet 32:88–92 Kelberman D, Dattani MT (2008) Septo-optic dysplasia - novel
Goldstone AP, Holland AJ, Hauffa BP, Hokken-Koelega AC, insights into the aetiology. Horm Res 69:257–265
Tauber M, on behalf of speakers and contributors at the Kelch RP, Virdis R, Rappaport R, Greig F, Levine LS, New MI
Second Expert Meeting of the Comprehensive Care of (1984) Congenital adrenal hypoplasia. Pediatr Adolesc
Patients with PWS (2008) Recommendations for the diagno- Endocrinol 13:156–161
sis and management of Prader-Willi syndrome. J Clin Kelly BP, Paterson WF, Donaldson MD (2003) Final height out-
Eindorinol Metab 93:4183–4197 come and value of height prediction in boys with constitu-
Green JS, Parfrey PS, Harnett JD, Farid NR, Cramer BC, tional delay in growth and adolescence treated with
Johnson G, Heath O, McManamon PJ, O’Leary E, Pryse- intramuscular testosterone 125 mg per month for 3 months.
Phillips W (1989) The cardinal manifestations of Bardet- Clin Endocrinol 58:267–272
Biedl syndrome, a form of Laurence-Moon-Biedl syndrome. Kim HG, Kurth I, Lan F, Meliciani I, Wenzel W, Eom SH,
N Engl J Med 321:1002–1009 Kang GB, Rosenberger G, Tekin M, Ozata M, Bick DP,
Greulich WW, Pyle SI (1959) Radiographic atlas of skeletal Sherins RJ, Walker SL, Shi Y, Gusella JF, Layman LC (2008)
development of the hand and wrist, 2nd edn. Stanford Mutations in CHD7, Encoding a chromatin-remodeling pro-
University Press, Stanford, CA tein, cause idiopathic hypogonadotropic hypogonadism and
Gunay-Aygun M, Schwartz S, Hegger S, O’Riordan MA, Kallmann syndrome. Am J Hum Genet 83:511–519
Cassidy S (2001) The changing purpose of Prader-Willi syn- Kletter GB, Gorski JL, Kelch RP (1991) Congenital adrenal
drome clinical diagnostic criteria and proposed revised crite- hypoplasia and isolated gonadotropin deficiency. Trends
ria. Pediatrics 108:e92 Endocrinol Metab 2:123–128
Habiby RL, Boepple P, Nachtigall L, Sluss PM, Crowley WF, Koeppen AH (1998) The hereditary ataxias. J Neuropathol Exp
Jameson JL (1996) Adrenal hypoplasia congenita with Neurol 57:531–543
hypogonadotropic hypogonadism: evidence that DAX-1 Krajewska-Siuda E, Malecka-Tendera E, Krajewski-Siuda A
mutations lead to combined hypothalamic and pituitary defects (2006) Are short boys with constitutional delay of growth
in gonadotropin production. J Clin Invest 98:1055–1062 and puberty candidates for rGH therapy according to FDA
Herring N, Szmigielski C, Becher H, Karavitaki N, Wass JA recommendations? Horm Res 65:192–196
(2009) Valvular heart disease and the use of cabergoline for Lagerström-Fermér M, Sundvall M, Johnsen E, Warne GL,
the treatment of prolactinoma. Clin Endocrinol 70:104–108 Forrest SM, Zajac JD, Rickards A, Ravine D, Landegren U,
Holm VA, Cassidy SB, Butler MG, Hanchett JM, Greenswag Pettersson U (1997) X-linked recessive panhypopituitarism
LR, Whitman BY, Greenberg F (1993) Prader-Willi syn- associated with a regional duplication in Xq25-q26. Am J
drome consensus diagnostic criteria. Pediatrics 91:398–402 Hum Genet 60:910–916
190 H. M. Behre et al.

Lalli E, Sassone-Corsi P (2003) DAX-1, an unusual orphan failed to repress steroidogenic acute regulatory protein
receptor at the crossroads of steroidogenic function and sex- (StAR) and luteinizing hormone beta-subunit gene promoter
ual differentiation. Mol Endocrinol 17:1445–1453 activity. Endocr J 55: 97–103
Largo RH, Prader A (1983) Pubertal development of Swiss boys. Partsch C-J, Sippell WG (1989) Hypothalamic hypogonadism in
Helv Paediatr Acta 38: 211–228 congenital adrenal hypoplasia. Horm Metab Res 11:
Lindgren AC, Lindberg A (2008) Growth hormone treatment 623–625
completely normalizes adult height and improves body com- Partsch C-J, Hermanussen M, Sippell WG (1985) Differentiation
position in Prader-Willi syndrome: experience from KIGS of male hypogonadotropic hypogonadism and constitutional
(Pfizer International Growth Database). Horm Res delay of puberty by pulsatile administration of gonadotro-
70:182–187 pin-releasing hormone. J Clin Endocrinol Metab 60:
Liu L, Chaudhari N, Corle D, Sherins RJ (1988) Comparison of 1196–1203
pulsatile subcutaneous gonadotropin-releasing hormone and Partsch C-J, Hümmelink R, Sippell WG (1990) Reference ranges
exogenous gonadotropins in the treatment of men with iso- for lutropin and follitropin in the luliberin test in prepubertal
lated hypogonadotropic hypogonadism. Fertil Steril and pubertal children using a monoclonal immunoradiomet-
49:302–308 ric assay. J Clin Chem Clin Biochem 28:49–52
Liu PY, Gebski VJ, Turner L, Conway AJ, Wishart SM, Partsch C-J, Lämmer C, Gillesen-Kaesbach G, Pankau R (2000)
Handelsman DJ (2002) Predicting pregnancy and spermato- Adult patients with Prader-Willi syndrome: clinical charac-
genesis by survival analysis during gonadotrophin treatment teristics, life circumstances and growth hormone secretion.
of gonadotrophin-deficient infertile men. Hum Reprod Growth Horm IGF Res 10(Suppl B):S81-S85
17:625–633 Pasqualini RQ, Burr GE (1950) Sindrome hypoandrogenico con
Lofrano-Porto A, Casulari LA, Nascimento PP, Giacomini L, gametogenesis conservada. Classification del la insufficien-
Naves LA, da Motta LD, Layman LC (2008) Effects of fol- cia testicular. Rev Asoc Med Argent 64:6–19
licle-stimulating hormone and human chorionic gonadotro- Peter M, Viemann M, Partsch C-J, Sippell WG (1998) Congenital
pin on gonadal steroidogenesis in two siblings with a adrenal hypoplasia: clinical spectrum, experience with hor-
follicle-stimulating hormone beta subunit mutation. Fertil monal diagnosis, and report on new points mutations of the
Steril 90:1169–1174 DAX-1-Gene. J Clin Endocrinol Metab 83:2666–2674
Mancini T, Casanueva FF, Giustina A. Hyperprolactinemia and Pitteloud N, Boepple PA, DeCruz S, Valkenburgh SB, Crowley
prolactinomas (2008) Endocrinol Metab Clin North Am WF Jr, Hayes FJ (2001) The fertile eunuch variant of idio-
37:67–99 pathic hypogonadotropic hypogonadism: spontaneous rever-
Mantovani G, Borgato S, Beck-Peccoz P, Romoli R, Borretta G, sal associated with a homozygous mutation in the
Persani L (2003) Isolated follicle-stimulating hormone gonadotropin-releasing hormone receptor. J Clin Endocrinol
(FSH) deficiency in a young man with normal virilization Metab 86:2470–2475
who did not have mutations in the FSHbeta gene. Fertil Pitteloud N, Acierno JS Jr, Meysing AU, Dwyer AA, Hayes FJ,
Steril 79:434–436 Crowley WF Jr (2005) Reversible Kallmann syndrome,
Mehta A, Dattani MT (2008) Developmental disorders of the delayed puberty, and isolated anosmia occurring in a single
hypothalamus and pituitary gland associated with congenital family with a mutation in the fibroblast growth factor recep-
hypopituitarism. Best Pract Res Clin Endocrinol Metab tor 1 gene. J Clin Endocrinol Metab 90:1317–1322
22:191–206 Poyrazoglu S, Günöz H, Drendeliler F, Saka N, Bundak R, Bas
Melmed S (2008) Update in pituitary disease. J Clin Endocrinol F (2005) Constitutional delay of growth and puberty: from
Metab 93:331–338 presentation to final height. J Pediatr Endocrinol Metab 18:
Merke DP, Tajima T, Baron J, Cutler GB (1999) Hypogonadotropic 171–179
hypogonadism in a female caused by an X-linked recessive Prader A, Labhart A, Willi H (1956) Ein Syndrom von Adipositas,
mutation in the DAX1 gene. N Engl J Med 340:1248–1252 Kleinwuchs, Kryptorchismus und Oligophrenie nach myoa-
Morison IM, Reeve AE (1998) A catalogue of imprinted genes tonieartigem Zustand im Neugeborenenalter. Schweiz Med
and parent-of-origin effects in humans and animals. Hum Wschr 86:1260–1261
Molec Genet 7:1599–1609 Quinton R, Barnett P, Coskeran P, Bouloux PM (1999) Gordon
Muscatelli F, Strom TM, Walker AP, Zanaria E, Récan D, Holmes spinocerebellar ataxia: a gonadotrophin deficiency
Meindl A, Bardoni B, Guioli S, Zehetner G, Rabl W, Schwarz syndrome resistant to treatment with pulsatile gonadotro-
HP, Kaplan JC, Camerino G, Meitinger T, Monaco AP phin-releasing hormone. Clin Endocrinol 51:525–529
(1994) An unusual member of the nuclear hormone receptor Raivio T, Falardeau J, Dwyer A, Quinton R, Hayes FJ,
superfamily responsible for X-linked adrenal hypoplasia Hughes VA, Cole LW, Pearce SH, Lee H, Boepple P, Crowley
congenita. Nature 372:672–676 WF Jr, Pitteloud N (2007) Reversal of idiopathic hypogo-
Ohta T, Gray TA, Rogan PK, Buiting K, Gabriel JM, Saitoh S, nadotropic hypogonadism. N Engl J Med 357:863–873
Muralidhar B, Bilienska B, Krajewska-Walasek M, Rosenbloom AL, Almonte SA, Brown MR, Fisher DA,
Driscoll DJ, Horsthemke B, Butler MG, Nicholls RD (1999) Baumbach L, Parks JS (1999) Clinical and biochemical phe-
Imprinting-mutation mechanisms in Prader-Willi syndrome. notype of familial anterior hypopituitarism from mutations
Am J Hum Genet 64:397–413 of the PROP1 gene. J Clin Endocrinol Metab 84:50–57
Okuhara K, Abe S, Kondo T, Fujita K, Koda N, Mochizuki K, Rump P, Hamel BCJ, Pinckers AJLG, van Dop PA (1997) Two
Fujieda K, Tajima T (2008) Four Japanese patients with sibs with chorioretinal dystrophy, hypogonadotrophic
adrenal hypoplasia congenita and hypogonadotropic hypog- hypogonadism, and cerebellar ataxia: Boucher-Neuhäuser
onadism caused by DAX-1 gene mutations: mutant DAX-1 syndrome. J Med Genet 34:767–771
12 Diseases of the Hypothalamus and the Pituitary Gland 191

Saeger W, Lüdecke DK, Buchfelder M, Fahlbusch R, Quabbe HJ, Forrest SM, Thomas PQ (2004) Array comparative genomic
Petersenn S (2007) Pathohistological classification of pitu- hybridisation analysis of boys with X linked hypopituitarism
itary tumors: 10 years of experience with the German identifies a 3.9 Mb duplicated critical region at Xq27 con-
Pituitary Tumor Registry. Eur J Endocrinol 156:203–216 taining SOX3. J Med Genet 41:669–678
Sampson JR, Tolmie JL, Cant JS (1989) Oliver McFarlane syn- Spratt DI, Carr DB, Merriam GR, Scully RE, Rao PN, Crowley
drome: a 25-year follow-up. Am J Med Genet 34:199–201 WF Jr (1987) The spectrum of abnormal patterns of gonado-
Schlechte JA (2003) Clinical practice. Prolactinoma. N Engl J tropin-releasing hormone secretion in men with idiopathic
Med 349:2035–2041 hypogonadotropic hypogonadism: clinical and laboratory
Schlechte JA (2007) Long-term management of prolactinoma. correlations. J Clin Endocrinol Metab 64:283–291
J Clin Endocrinol Metab 92:2861–2865 Stoetzel C, Muller J, Laurier V, Davis EE, Zaghloui NA,
Schmidt H, Knorr D, Schwarz HP (1998) Oral testosterone Vicaire S, Jacquelin C, Plewniak F, Leitch CC, Sarda P,
undecanoate for the induction of puberty in anorchid boys. Hamel C, de Ravel TJ, Lewis RA, Friederich E, Thibault C,
Arch Dis Child 78:395 Danse JM, Verloes A, Bonneau D, Katsanis N, Poch O,
Schopohl J, Mehltretter G, von Zumbusch R, Eversmann T, von Mandel JL, Dollfus H (2007) Identification of a novel BBS
Werder K (1991) Comparison of gonadotropin-releasing gene (BBS12) highlights the major role of a vertebrate-spe-
hormone and gonadotropin therapy in male patients with cific branch of chaperonin-related proteins in Bardet-Biedl
idiopathic hypothalamic hypogonadism. Fertil Steril syndrome. Am J Hum Genet 80:1–11
56:1143–1150 Takahashi T, Shoji Y, Haraguchi N, Takahashi I, Takada G (1997)
Schwanzel-Fukuda M, Bick D, Pfaff DW (1989) Luteinizing Active hypothalamic-pituitary-gonadal axis in an infant with
hormone-releasing hormone (LHRH)-expressing cells do X-linked adrenal hypoplasia congenita. J Pediatr 130: 485–488
not migrate normally in an inherited hypogonadal (Kallmann) Topaloglu AK, Reimann F, Guclu M, Yalin AS, Kotan LD, Porter
syndrome. Mol Brain Res 6:311–326 KM, Serin A, Mungan NO, Cook JR, Ozbek MN, Imamoglu
Sedlmeyer IL, Palmert MR (2002) Delayed puberty: analysis of S, Akalin NS, Yuksel B, O'Rahilly S, Semple RK (2009)
a large case series from an academic center. J Clin Endocrinol TAC3 and TACR3 mutations in familliar hypogonadotropic
Metab 87:1613–1620 hypogonadism reveal a key role for Neurokinin B in the cen-
Sedlmeyer IL, Hirschhorn JN, Palmert MR (2002) Pedigree tral control of reproduction. Nat Genet. 41:354–358
analysis of constitutional delay of growth and maturation: Vogels A, Moerman P, Frijns JP, Bogaert GA (2008) Testicular
determination of familial aggregation and inheritance pat- histology in boys with Prader-Willi syndrome: fertile or
terns. J Clin Endocrinol Metab 87:5581–5586 infertile? J Urol 180(4):1800–1804
Seminara SB, Hayes FJ, Crowley WF (1998) Gonadotropin- Warne DW, Decosterd G, Okada H, Yano Y, Koide N, Howles
releasing hormone deficiency in the human (idiopathic CM (2008) A combined analysis of data to identify predic-
hypogonadotropic hypogonadism and Kallmann’s syn- tive factors for spermatogenesis in men with hypogonado-
drome): pathophysiological and genetic considerations. tropic hypogonadism treated with recombinant human
Endocr Rev 19:521–539 follicle-stimulating hormone and human chorionic gonado-
Seminara SB, Acierna JS Jr, Abdulwahid NA, Crowley WF Jr, tropin. Fertil Steril [Epub ahead of print]
Marglin DH (2002) Hypogonadotropic hypogonadism and Wehkalampi K, Vangonen K, Laine T, Dunkel L (2007)
cerebellar ataxia: detailed phenotypic characterization of a Progressive reduction of relative height in childhood pre-
large, extended kindred. J Clin Endocrinol Metab 87: dicts adult stature below target height in boys with constitu-
1607–1612 tional delay of growth and puberty. Horm Res 68:99–104
Sherlock M, Toogood AA, Steeds R (2009) Dopamine agonist Wehkalampi K, Widen E, Laine T, Palotie A, Dunkel L (2008a)
therapy for hyperprolactinaemia and cardiac valve fibrosis; a Patterns of inheritance of constitutional delay of growth and
lot done but much more to do. Heart [Epub ahead of print] puberty in families of adolescent girls and boys referred to spe-
Sinisi AA, Esposito D, Maione L, Quinto MC, Visconti D, De cialist pediatric care. J Clin Endocrinol Metab 93:723–728
Bellis A, Bellastella A, Conzo G, Bellastella G (2008) Wehkalampi K, Widen E, Laine T, Palotie A, Dunkel L (2008b)
Seminal anti-Müllerian hormone level is a marker of sper- Association of the timing of puberty with a chromosome 2
matogenic response during long-term gonadotropin therapy locus. J Clin Endocrinol Metab [Epub ahead of print]
in male hypogonadotropic hypogonadism. Hum Reprod Willers B, Engelhardt L, Pelz L (1996) Sexual maturation in
23:1029–1034 East German boys. Acta Paediatr 85:785–788
Smals AGH, Hermus ARM, Boers GHJ, Pieters GFF, Benraad TJ, Willnow S, Kiess W, Butenandt O, Dörr HG, Enders A, Strasser-
Kloppenborg PWC (1994) Predictive value of luteinizing Vogel B, Egger J, Schwarz HP (1996) Endocrine disorders
hormone releasing hormone (LHRH) bolus testing before in septo-optic dysplasia (De Morsier syndrome) – evaluation
and after 36-hour pulsatile LHRH administration in the dif- and follow up of 18 patients. Eur J Pediatr 155:179–184
ferential diagnosis of constitutional delay of puberty and Wilson DA, Hofman PL, Miles HL, Unwin KE, McGrail CE,
male hypogonadotropic hypogonadism. J Clin Endocrinol Cutfield WS (2006) Evaluation of the buserelin stimulation
Metab 78:602–608 test in diagnosing gonadotropin deficiency in males with
Soliman AT, Rajab A, AlSalmi I, Asfour MG (1996) Empty delayed puberty. J Pediatr 148:89–94
sellae, impaired testosterone secretion, and defective hypo- Wollmann HA, Schultz U, Grauer ML, Ranke MB (1998)
thalamic-pituitary growth and gonadal axes in children with Reference values for height and weight in Prader-Willi syn-
Bardet-Biedl syndrome. Metabolism 45:1230–1234 drome based on 315 patients. Eur J Pediatr 157:634–642
Solomon NM, Ross SA, Morgan T, Belsky JL, Hol FA, Young J, Rey R, Schaison G, Chanson P (2003) Hypogonadotropic
Karnes PS, Hopwood NJ, Myers SE, Tan AS, Warne GL, hypogonadism as a model of post-natal testicular anti-Müllerian
192 H. M. Behre et al.

hormone secretion in humans. Mol Cell Endocrinol 211: superfamily responsible for X-linked adrenal hypoplasia
51–54 congenita. Nature 372:635–641
Zanaria E, Muscatelli F, Bardoni B, Strom TM, Guioli S, Zanettini R, Antonini A, Gatto G, Gentile R, Tesei S, Pezzoli
Weiwen G, Lalli E, Moser C, Walker AP, McCabe ERB, G (2007) Valvular heart disease and the use of dopamine
Meitinger T, Monaco AP, Sassone-Corsi P, Camerino G agonists for Parkinson’s disease. N Engl J Med 356:
(1994) An unusual member of the nuclear hormone receptor 39–46
Disorders at the Testicular Level
13
Eberhard Nieschlag, Hermann M. Behre, Peter Wieacker,
Dieter Meschede, Axel Kamischke, and Sabine Kliesch

Contents 13.7.3 Diagnosis .................................................. 208


13.7.4 Therapy ..................................................... 208
13.1 Anorchia ................................................................ 194 13.8 Specific Structural Sperm Defects ....................... 208
13.1.1 Congenital Anorchia ................................. 194 13.8.1 Globozoospermia ...................................... 208
13.1.2 Acquired Anorchia .................................... 195 13.8.2 9 + 0 Syndrome......................................... 209
13.8.3 Syndrome of Immotile Cilia ..................... 209
13.2 Polyorchidism ........................................................ 197
13.8.4 Clinical Picture ......................................... 209
13.8.5 Diagnosis .................................................. 209
13.3 Maldescended Testes............................................. 197 13.8.6 Therapy ..................................................... 209
13.3.1 Pathophysiology and Classification .......... 197 13.9 Klinefelter Syndrome ........................................... 210
13.3.2 Infertility and Risk of Malignancy ........... 198
13.3.3 Diagnosis .................................................. 199 13.9.1 Incidence and Etiology ............................. 210
13.3.4 Therapy ..................................................... 199 13.9.2 Clinical Picture ......................................... 211
13.9.3 Diagnosis .................................................. 212
13.4 Varicocele ............................................................... 200 13.9.4 Therapy ..................................................... 213
13.4.1 Pathophysiology ....................................... 200 13.10 XX Male Syndrome .............................................. 214
13.4.2 Prevalence and Influence of
Varicocele on Fertility .............................. 201 13.11 XYY Syndrome ..................................................... 216
13.4.3 Clinical Picture ......................................... 201 13.12 Noonan Syndrome................................................. 216
13.4.4 Diagnosis .................................................. 202
13.4.5 Influence of Therapy on Fertility .............. 202 13.13 Structural Chromosome
13.4.6 Meta-analysis of Studies on Treatment .... 204 Abnormalities ........................................................ 216
13.4.7 Treatment Modalities ................................ 204 13.13.1 Structural Abnormalities
13.4.8 Varicocele in Adolescence ........................ 205 of Sex Chromosomes ................................ 217
13.5 Orchitis .................................................................. 206 13.13.2 Y Chromosome Microdeletions................ 218
13.13.3 Structural Abnormalities
13.5.1 Clinical Picture and Diagnosis ................. 206 of the Autosomes ...................................... 220
13.5.2 Therapy ..................................................... 206
13.14 Oviduct Persistence............................................... 220
13.6 Germ Cell Aplasia (SCO Syndrome) .................. 206
13.15 Gonadal Dysgenesis .............................................. 221
13.6.1 Pathophysiology ....................................... 206
13.6.2 Clinical Picture and Diagnosis ................. 207 13.15.1 Definition .................................................. 221
13.6.3 Therapy ..................................................... 207 13.15.2 Clinical Picture ......................................... 221
13.15.3 Diagnosis .................................................. 221
13.7 Spermatogenic Arrest ........................................... 207 13.15.4 Therapy ..................................................... 222
13.7.1 Pathophysiology ....................................... 207 13.16 46,XY Disorders of Sexual
13.7.2 Clinical Picture ......................................... 208 Development (DSD) due to Disturbed
Testosterone Synthesis .......................................... 222
13.16.1 Definition .................................................. 222
E. Nieschlag () 13.16.2 Etiology .................................................... 222
Centre of Reproductive Medicine and Andrology 13.16.3 Clinical Picture ......................................... 222
of the University, Domagkstr. 11, D-48149 Münster, Germany 13.16.4 Diagnosis .................................................. 223
e-mail: Eberhard.Nieschlag@ukmuenster.de 13.16.5 Therapy ..................................................... 223

E. Nieschlag et al. (eds.), Andrology, 193


DOI: 10.1007/978-3-540-78355-8_13, © Springer-Verlag Berlin Heidelberg 2010
194 E. Nieschlag et al.

13.17 Mutations of Gonadotropin Receptors ............... 224 13.1 Anorchia


13.17.1 Inactivating LH Receptor Mutations:
Leydig Cell Hypoplasia ............................ 224
13.17.2 Activating LH Receptor Mutations........... 225
13.17.3 Inactivating FSH Receptor Mutations ...... 225
Unilateral or bilateral absence of testicular tis-
13.17.4 Activating FSH Receptor Mutations......... 226 sue in genetic males is called anorchia. Anorchia
has to be differentiated from partial or complete tes-
13.18 Ovotesticular Disturbances of Sexual
Development (DSD) .............................................. 226 ticular atrophy, e.g., following torsion or orchitis,
when, at least histologically, degenerated remains
13.18.1 Definition and Etiology ............................ 226
13.18.2 Clinical Picture ......................................... 226 of a male gonad can be found. Anorchia can be con-
13.18.3 Diagnosis .................................................. 226 genital or acquired.
13.18.4 Therapy ..................................................... 226
13.19 Testicular Tumors ................................................. 227
13.19.1 Incidence ................................................... 227 13.1.1 Congenital Anorchia
13.19.2 Testicular Intraepithelial
Neoplasia (TIN) ........................................ 227
13.19.3 Germ Cell Tumors .................................... 228 Bilateral congenital anorchia occurs only in one of
13.19.4 Testicular Tumors with Endocrine 20,000 males. Unilateral congenital anorchia is
Activity ..................................................... 231 about four times as frequent. Vascular and genetic dis-
References ........................................................................... 231 turbances, intrauterine infections, trauma or terato-
genic factors are discussed as causes for the loss of one
or both testes. A suspected abnormality in the sex-
determining region of the Y chromosome, the SRY
gene, could not be confirmed so far (Lobacarro et al.
1993). Nor could mutations in other genes responsible
for testicular development and descent be clearly iden-
tified to date (Vinci et al. 2004; Philibert et al. 2007).
Currently intrauterine torsion is favored as the most
probable cause.
The morphological findings in bilateral congenital
anorchia can be directly derived from the embryonic
development of the male genital system. In the 8th week
of pregnancy the testes developing from the undifferen-
tiated gonadal system start to secrete anti-Muellerian
hormone (AMH) and, only at a later stage, testosterone.
If the testicular tissue is lost before testosterone produc-
tion begins, the Muellerian ducts have regressed; how-
ever, the androgen-dependent differentiation of the
Wolffian ducts as well as the masculinization of the uro-
genital sinus and the external genitalia have not yet
started. If the testes originally present are destroyed
after they have produced testosterone for a certain
period, the androgen-dependent target organs of the
urogenital tract are more or less completely differenti-
ated towards the male phenotype (Josso et al. 2006).
Subjects with bilateral congenital anorchia, in
whom the testes had produced AMH but no testoster-
one, present with the phenotype of male pseudoher-
maphroditism including female external genitalia.
Neither gonads nor derivatives of the Muellerian ducts
13 Disorders at the Testicular Level 195

(oviducts, uterus, upper vagina) nor the Wolffian ducts expected puberty (see Chap. 21). In phenotypically
(epididymis, ductus deferens, seminal vesicles) can be female patients estrogen substitution will be started.
found. If the testes have produced testosterone during Intersexual external genitalia may be corrected by plas-
embryonal development, the external genitalia are male tic surgery. For cosmetic reasons, testicular protheses
and the Wolffian duct derivatives are developed. A small may be implanted into the scrotum. Infertility in bilateral
penis may indicate diminished androgen-dependent anorchia cannot be treated.
growth during fetal development. If left untreated,
pubertal development will not start in patients with
bilateral congenital anorchia and the typical phenotype
13.1.2 Acquired Anorchia
of eunuchoidism develops. Since a single intact testis is
functionally sufficient, disorders of sexual differentia-
tion and pubertal development do not occur in congeni- 13.1.2.1 Accidental Castration
tal unilateral anorchia although unilateral retention of
Muellerian ducts can be present (depending on timing Testes can be lost due to trauma, tumors, severe
of testicular loss). inflammation, torsion, surgical accidents (e.g., during
herniotomy or orchidopexy) or surgical removal (e.g.
because of a testosterone-dependent tumor such as
13.1.1.1 Diagnosis prostate carcinoma) and very rarely after self-mutila-
tion. The loss of one testis will be compensated for by
In patients with congenital bilateral anorchia testicu- the remaining testis if it is normal concerning fertility
lar tissue cannot be demonstrated either by morpho- and testosterone production, and therapy will not be
logical or by endocrine techniques (see Chap. 6). FSH necessary. However, some authors argue in favor of
and LH serum levels may already be elevated in chil- removing testicular tissue remaining after treatment
dren and rise to castrate levels from the age of puberty for torsion because in some unexplained manner the
onwards. In contrast, testosterone is very low. For a function of the remaining organ can be negatively
differential diagnosis cryptorchidism must be ruled affected (Arap et al. 2007). In cases of acute bilateral
out (see Sect. 13.3). The hCG-test is used for differen- testicular loss it should be remembered that sperm
tiation (see Chap. 7). While a rise in serum testosterone may still be found weeks after the acute incident and
can be measured in patients with cryptorchidism, the should be cryopreserved for paternity unless the man
values remain low even after a 7-day period of stimula- has definitely completed his family (Fig. 13.1) (see
tion in patients with bilateral anorchia (Davenport et al. Chap. 24).
1995; McEachern et al. 2004). In addition, AMH,
25
which is lacking in anorchia, should be measured since 23.1
it shows a higher sensitivity, but equal specificity com-
Sperm count (mill / ejaculate)

pared to testosterone (Lee et al. 1997). In cases with 20

suspected unilateral anorchia the absence of gonadal


tissue must be ascertained by imaging diagnostic pro- 15

cedures (sonography, computer tomography, MRT)


and if necessary by exploratory surgery/laparoscopy, 10

since non-descended and dysgenetic gonads have a


high rate of malignant degeneration. 5
2.3
< 0.1
0
10 19 41
13.1.1.2 Therapy Days after bilateral orchidectomy

Unilateral anorchia does not require therapy as long as Fig. 13.1 Sperm in the ejaculate of a 15-year-old boy following
bilateral orchidectomy due to delayed discovery of testicular tor-
testosterone production is sufficient. In phenotypically
sion. Although several ejaculations had taken place, even 10
male patients with bilateral congenital anorchia testos- days after surgery (when the patient was first seen by us), enough
terone substitution has to be implemented at the time of sperm were present to make cryopreservation worthwhile
196 E. Nieschlag et al.

The clinical symptoms of bilateral testicular loss however, lucrative positions were the possible rewards,
depend on the time when testicular function is lost. many candidates underwent the risk (Wagenseil 1993;
Before puberty, testicular loss leads to the characteris- Mitamura 1992; Wilson and Roehrborn 1999). The
tic phenotype of eunuchoidism; after puberty it will last castrate of the Chinese empire, terminated by
lead to the phenotype of postpubertal testosterone defi- revolution in 1912, Sun Yaoting, died at the age of 93
ciency (see Chap. 5, Table 5.1). in 1996.
When both testes are lost, testosterone must be Castration before puberty maintains the high voice
permanently substituted from the time of the expected of boys so that in the adult soprano and alto voices
beginning of puberty in order to induce pubertal devel- with the acoustic volume of a male result. Such high-
opment and in an adult immediately after testicular loss pitched voices were considered desirable among
to maintain the various androgen-dependent functions music-lovers. Prepubertal castrates belonged to casts
(see Chap. 21). Testicular protheses (usually silastic) of operas in the seventeenth and eighteenth century
may be implanted for psychological or cosmetic and in the Vatican choirs these voices could be heard
reasons. until the early twentieth century (Ortkemper 1993).
Roman physicians recommended castration for the
treatment of leprosy and epilepsy; in the seventeenth
13.1.2.2 Medical and Legal Castration century it was practised to cure gout and dementia.
Until the early twentieth century castration was prac-
If the testes have been removed because of prostate tised in the USA for treatment of the mentally handi-
carcinoma or as a legal prophylactic procedure in sex- capped for the reason that it made them more easily
ual delinquents, testosterone must certainly not be manageable.
substituted since the elimination of its effect is the Castration has also been reported as self-mutilation
intended therapeutic purpose in these cases. In some for religious reasons since ancient times. The early
countries castration is a legal procedure for sexual church father Origines (186–254) is one of the most
delinquency in order to avoid imprisonment. For exam- prominent examples. More recently, castration was
ple, in the Federal Republic of Germany 400 men were practised in southern Russia among members of the
castrated between 1970 and 1980 on the basis of the Scoptic sect founded in the eighteenth century. The
“law for voluntary castration” (1969, amended 1973) largest contemporary groups of castrates are the Hijras
(Wille 1991). In other countries this procedure is ille- in India. They function as professional well-wishers at
gal. In some countries medical castration (e.g., by anti- birth rites and receive considerable financial rewards.
androgens) is used instead but this is also controversial Several thousand of them exist.
ethically. In Germany, surgical castration is currently Patients with acquired anorchia could (involuntarily)
not practised nor is chemical castration applied. Instead, contribute to the question whether the shorter life
psychosocial treatment modalities are preferred. expectancy of men compared to woman (Fig. 21.1)
may be caused by testosterone or the presence of testes,
as has often been claimed. Astonishing little, however, is
13.1.2.3 Socio-cultural Castration known about this “model”. A retrospective analysis of
the life expectancy of inmates of an institution for the
Beyond medical purposes, castration has been and is mentally handicapped in the USA came to the conclu-
still performed for socio-cultural reasons. Societies sion that early castration may lead to a higher life expec-
practicing polygamy have been known to employ cas- tancy (Hamilton and Mestler 1969). However, this may
trates as overseers (eunuchs as harem guards) who be explained by the preference for castration of physi-
may then have gained political influence. For exam- cally active inmates whereas lack of mobility is the major
ple, at the Chinese imperial court castration consist- predictor of shortened life expectancy among institution-
ing of amputation of penis, testes and scrotum without alized men. In contrast, we could find no difference in
anesthesia was performed in adult men. These proce- the lifespan of intact and prepubertally castrated singers
dures were extremely painful and accompanied fre- from the sixteenth to the nineteenth century when ana-
quently by severe complications so that 25% of those lyzing their biographical data (Fig. 13.2) (Nieschlag
undergoing this “treatment” did not survive. Since, et al. 1993b). Since neither the investigation of mentally
13 Disorders at the Testicular Level 197

100 Most patients have no symptoms and the supernu-


90 merary testis may be discovered incidentally. Fertility
80 may be normal. However, malignancies and torsions
70 may occur in the third testis and require acute interven-
Age (years)

60
tion. Most are discovered in association with hernias
50
or because of maldescent. A third testis with epididymis
40
and vas may be the rare cause of persistent fertility fol-
30
lowing vasectomy (Hakami and Mosavi 1975).
20
In recent years sonography and magnetic resonance
10 P=0.65 (n.s.)
imaging have aided the diagnosis and may make surgi-
0
Castrated Intact singers cal exploration for histological confirmation superflu-
ous (Oner et al. 2005).
Fig. 13.2 Lifespan of castrated and intact singers from the six-
Therapeutic management must be decided on a
teenth to nineteenth centuries. In each group 50 singers with
similar birthdates were selected. Box and whisker plot: solid case-to-case basis. While tumors and torsion require
line = median, dotted line = average (Nieschlag et al. 1993b) surgical removal, the testis may remain in situ in other
cases. Such cases should be followed by regular inves-
tigations including sonography and tumor markers for
handicapped nor historical analysis can be considered early discovery of a tumor.
representative for the present normal population, these In the medical literature polyorchidism was first
studies can only provide hints, while the question con- documented in 1880 (Ahlfeld 1880). Historically, super-
cerning the 7-year shorter life expectancy of men as numerary testes have often been associated with extraor-
opposed to women remains unresolved. Analysis of dinary strength and sexual vigour of their bearers. Several
lifespan of 292 female opera singers who died between such cases have been reported anecdotally. Of these, the
1900 and 1995 revealed that sopranos, being more highly Venetian admiral Bartolomeo Colleoni (1400–1475)
estrogenized, live significantly longer than altos, who achieved special fame (“colleoni” in Italian means
are more highly androgenized. This was interpreted to “testes” and nowadays is spelled “coglioni”) and in 1539
mean that isosexual hormones tend to increase lifespan. Philipp Count of Hesse (1504–1567) was granted per-
Among the 505 male opera singers analyzed, basses, as mission by Martin Luther to take a second wife simul-
opposed to tenors, showed only a tendency towards lon- taneously because of an alleged third testis.
ger life expectancy (Nieschlag et al. 2003).

13.3 Maldescended Testes


13.2 Polyorchidism
13.3.1 Pathophysiology
Supernumerary testes are a rare finding with little
more than 100 cases reported in the literature. In most and Classification
instances a third testis is found (triorchidism), predomi-
nantly on the left side. The most likely explanation for About the end of the first trimester of early embryonic
this condition is a transverse division or doubling over of development the testes move from the region of the
the genital ridge caused by peritoneal bands in early kidneys to the inner opening of the inguinal channel.
embryonic development (4th to 6th week). The duplica- During the last 2 months of gestation the testes descend
tion may only concern the testis, but doubling of the from the position close to the inner opening of the
epididymis and vas deferens may also be found (Leung inguinal channel into the scrotum. At the time of birth,
1988; O’Sullivan et al. 1995) so that the supernumarary testicular descent is normally completed and is a char-
testis may either have its own or a joint epidydimis and acteristic sign of maturity of a newborn. Descent may,
seminal duct together with the neighboring testis (type however, come to a halt at various positions. According
A) or has no excurrent duct (type B) (Bergholz et al. to the location of the testes, the following anomalies
2007). are differentiated:
198 E. Nieschlag et al.

In most cases the etiology of testicular maldescent is


unclear, so that in 85% a so-called idiopathic maldes-
cent is diagnosed. Presumably, the etiology is multifac-
torial. Since in most cases the incompletely descended
testis has passed the inner opening of the inguinal chan-
nel, disturbances of inguinal scrotal descent are assumed.
INSL3 and its receptor LGR8, secreted by the Leydig
cells, are responsible for the transabdominal phase of
descent from the kidney region to the inguinal canal.
Testosterone is required for the inguinal-scrotal phase of
descent and development of the gubernaculum (Hutson
and Hasthorpe 2005; Virtanen and Toppari 2008). In addi-
Fig. 13.3 Thirty-two-year-old patient with bilateral inguinal tion to endocrine disturbances, defects in neurotransmitter
testis secretion from a not fully matured genitor-femoral nerve
are involved.
• Cryptorchidism: The testis lies above the inner Testicular maldescent is frequently found in patients
inguinal channel intraabdominally or retroperi- with hypopthalamic-pituitary disturbances (see Chap.
toneally and can neither be seen nor palpated. 12) or in patients with disorders of testosterone synthe-
• Inguinal testis: The testis is positioned firmly in sis or testosterone action. Next to endocrine causes,
the inguinal channel (Fig. 13.3). anatomical abnormalities could be identified as causes
• Retractile testis: The testis lies at the orifice of of maldescent as well as reduced abdominal wall ten-
the inguinal channel and can be pushed into the sion leading to lowered intraabdominal pressure (e.g.,
scrotum, but returns to the original position when prune-belly syndrome, bladder exstrophy, omphalo-
released. In other cases the testis alternates its cele, gastroschisis). Maldescent is also frequently found
position spontaneously from the scrotum into in primary testicular disorders such as Klinefelter syn-
the inguinal channel and back. This movement drome (see Sect. 13.9), Noonan syndrome (see Sect.
will also be induced by the cremasteric reflex 13.12), XX men (see Sect. 13.10) and in gonadal dys-
when exposed to cold or during coitus. genesis (see Sect. 13.15).
• Testicular ectopy: The testis lies outside the
normal route of descent, e.g., femorally or in the
groin.
13.3.2 Infertility and Risk of Malignancy

Unilateral maldescent is five times as frequent as bilat- Abnormal testicular location is associated with a dis-
eral maldescent. Retractile testes occur relatively fre- turbance of the germinal epithelium. Many men, even
quently and rarely have pathological significance. The with unilaterally maldescended testis have impaired
incidence of other anomalies of descent was summa- fertility (Lee 2005). In our infertility clinic the percent-
rized in a large systematic analysis encompassing 46 age of patients with successfully treated or still existing
studies and 704,225 patients (Sijstermans et al. 2007). maldescended testes is 8.4% and thus significantly
Male newborns born at term and/or weighing ≥2.5 kg higher than the prevalence of 0.5% in the general male
showed maldescended testes in 1–4.6%. By 1 year of population. Patients with azoospermia were repre-
age the incidence was only 1–1.5%. In premature sented by a percentage as high as 17.2% (Table 4.2).
infants and/or weighing <2.5 kg the incidence was 0.1– Fertility-reducing alterations may occur very early
9.0%, declining to 1.1–2.1% by completion of the 1st and not – as once assumed – only at the time of puberty.
year of life. In 15 year-old boys the incidence was 1.6– Morphological alterations of the testis can be demon-
2.2%. It must be noted that the 46 studies were partly strated even in the 1st years of life. Thus the maturation
performed using different criteria and definitions and of gonocytes to A-spermatogonia as first step of postna-
that no longitudinal investigations were done. In adult tal spermatogenesis is disturbed (Huff et al. 1991). The
men maldescent is still seen in 0.5% cases. longer the maldescent exists, the more pronounced the
13 Disorders at the Testicular Level 199

morphological alterations will be. In 163 men with a 13.3.4 Therapy


history of maldescended testes from our own fertility
clinic who underwent biopsy, 28% showed moderate Testicular maldescent should be treated as early as pos-
hypospermatogenesis and in two thirds a Sertoli- sible but, because of the high rate of spontaneous descent,
cell-only syndrome or spermatogenetic arrest was not before the 6th month (Ritzen et al. 2007). Treatment
diagnosed. In nine of these patients a carcinoma in situ should be completed by the end of the 1st year. For pre-
or a seminoma was found. Maldescended testes usu- mature infants the corrected age should be followed
ally do not show impaired endocrine function. (AWMF 2008). A large number of medicinal therapeutic
In addition to impaired fertility, patients with mal- regimes are recommended. The current guidelines rec-
descended testes show an increased risk of testicular ommend one for the sake of simplicity and standardiza-
tumors. The risk of malignancy remains increased tion. If the testes do not descend by the end of the first
even in testes which are brought into the scrotum by 6th months presurgical, hormonal therapy (see below)
orchidopexy as well as in the contralateral, always- should be initiated. According to the guidelines, 3 ×
scrotal, testis. However, these testes are more accessi- 400 μg/day GnRH (3x daily one aerosol application of
ble for diagnostic purposes after orchidopexy. In 2.8% 200 μg in each nostril) for 4 weeks and immediately
of 599 men with maldescended testes a testicular tumor thereafter (as a continued therapy block) hCG at a dose
or a carcinoma in situ was histologically ascertained in of 1 × 500 IU for 3 weeks should be applied (AWMF
one or both testes (Giwercman et al. 1993). The risk of 2008). This regime with GnRH and hCG induces descent
developing a testicular tumor is 4–5 times higher in with a 20% success rate (Henna et al. 2004).
men with maldescended testes than in the general male Under hCG and GnRH therapy erections may
population. It must be assumed that intrinsic congeni- occur, some pubic hair may grow, testicular volume
tal factors influence the potential for infertility and may increase and the boys may appear more aggres-
malignancy in the maldescended as well as in the con- sive. These testosterone-dependent side-effects are
tralateral testis. Successful treatment before the 13th reversible after the end of therapy. Additional admin-
year of life reduces the relative risk of malignancy to istration of FSH does not improve therapeutic results
2.2 but is 5.4 for patients treated after the age of 13 (Hoorweg-Nijman et al. 1994). This underlines the
(Petterson et al. 2007). role of Leydig cell secretory products as leading fac-
tors in testicular descent. The lower the testis is
located prior to treatment and the younger the patient
is (< or >4 years) the higher the success of hormonal
13.3.3 Diagnosis therapy will be (Pyörälä et al. 1995). hCG-induced
testicular damage observed in rats (Kaleva et al.
When investigating the patient, the testis should first be 1996) continues to be discussed controversially for
investigated with the patient standing. When the result humans (Cortes et al. 2000) but is one reason, in addi-
of this palpation remains unclear, he should be reexam- tion to the low success rates, why Scandinavian pedi-
ined in a recumbent position. Exposure to cold and atricians do not favor hormonal treatment (Ritzen
stimulation should be avoided since they may induce et al. 2007).
the cremasteric reflex and retraction of the testes. If hormonal treatment does not induce testicular
Occasionally it may even be necessary to examine the descent, surgical orchidopexy may be required, which
scrotum after a warm bath. Testicular volume measured likewise should be performed by the age of 12 months
with an orchidometer or ultrasound should be recorded. (AWMF 2008). If a hernia exists in addition or if the
Imaging sonography supports the diagnosis when the testis is in an ectopic position, surgical correction
testis is in a still higher position. In bilateral cryp- should be performed either through open surgery or by
torchidism the hCG-test is essential for differentiating laparoscopy. Success rates are 85% for abdominal tes-
from anorchia if the baseline testosterone is in the cas- tes and 95% for inguinal testes (Taran and Elder 2006).
trate range (see Sect. 13.1.1). If testosterone rises under Orchidopexy followed by GnRH analogue treatment
hCG-stimulation and confirms the existence of testicu- has been found to result in better semen parameters in
lar tissue, the testes should be searched for by imaging later adulthood (Hadziselimovic and Herzog 1997).
diagnostic procedures such as CT or MRI. Autotransplantation of high intraabdominal testes into
200 E. Nieschlag et al.

the scrotum may be attempted (Bukowski et al. 1995). guidelines for treatment. This is made abundantly clear
This operation may be indicated particularly in cases by the the guidelines simultaneously published in
of bilateral cryptorchidism; it should be performed as Germany (AWMF 2008) and by Scandinavian coun-
early in childhood as possible. However, successful tries (Ritzen et al. 2007), i.e., by countries with few
operations in adolescence and young adults have been ethnic and cultural differences. These controversies
reported. “Success” means correction of the location of are at least in part due to differing definitions and clas-
the testis and maintenance of the endocrine function, sifications (Süsterman et al. 2007), but which suffer
while fertility will rarely be achieved in bilateral cryp- above all from a lack of representative controlled and
torchidism. Biopsy of testicular tissue should not be randomized therapy studies which would have to cover
undertaken as, for unclear reasons, this often results in a time span of about 30 years. To date there has been
testicular tumors later in life (Swerdlow et al. 1997). no initiative for such tedious studies.
In adults hCG and GnRH therapy are useless. If
orchidopexy is not or cannot be performed, regular
check-ups – e.g., at annual or semi-annual intervals –
for signs of malignancy are mandatory. Imaging sono-
13.4 Varicocele
graphy is of decisive importance in the framework of
these preventive procedures to diagnose a malignancy 13.4.1 Pathophysiology
early (see Chap. 6). The sonographic picture will influ-
ence the frequency of check-ups. Even successfully
treated maldescended testes should be regularly pal- The term varicocele denotes a tortuous convoluted
pated and investigated by ultrasonography because of formation of the internal spermatic vein forming
the increased incidence of malignant degeneration. the pampiniform plexus in the scrotum.
Finally, the patient should be instructed in regular self-
palpation and should consult his physician if he encoun-
ters suspicious alterations. The varicose alteration is favored by the extended free
The general recommendation of early therapy for passage of the testicular vein in the retroperitoneum,
maldescended testes is based on findings of alterations by the lack of supporting muscle pump, by congeni-
in testicular morphology which have been documented tally weak vessel walls or by an atonic cremaster mus-
as early as the 1st year of life. Treatment shortly before cle in part accompanying the spermatic vein. For many
or during puberty as practised in earlier times could years it was assumed that the condition was caused by
not prevent testicular damage. It is now hoped but not incompetence or aplasia of the spermatic venous
proven that timely therapy will reduce the incidence valves. More recently conducted cadaver studies and
of infertility and malignancy. However, since early angiographic investigations have revealed that men
treatment has been generally practised for less than 2 without varicocele may also lack valves (Ergün et al.
decades and not enough patients have reached the stage 1996). Probably because of hemodynamically unfa-
when they want to father children, it cannot yet be vorable merging of the spermatic vein into the renal
ascertained that early treatment will indeed help to vein – the right spermatic vein leads directly into the
avoid infertility in patients with maldescended testes v. cava inferior – a varicocele is found on the left side
(Lee 2005). Only large-scale investigations of patients in about 95% of patients. When the internal spermatic
treated early according to the new therapeutic scheme vein is compressed by a neoplasm (e.g., a kidney
and who have reached reproductive age will clarify tumor) one speaks of a secondary varicocele.
this question. By just what mechanism varicocele influences
For adult infertile patients with a history of malde- fertility remains unclear. Various possibilities con-
scended testes currently no rational therapy is avail- tinue to be discussed: reduced perfusion of the affected
able; as symptomatic treatment, techniques of assisted testis because of increased venous pressure leading to
fertilization may be considered and applied (see atrophy with typical reduction of testicular volume;
Chap. 23). an increase in scrotal temperature or insufficient
Although a large number of patients are affected by removal or backflow of toxic substances of renal origin.
maldescent, to date there are no uncontroversial The hypothesis that a varicocele is nature’s attempt to
13 Disorders at the Testicular Level 201

compensate for an otherwise damaged testis is also in one partner can usually be overcome by particularly
unproven. good reproductive functions of the other. Only when
disturbances in both partners coincide do problems
arise (see Sect. 1.4). Investigations in fathers with and
without varicocele suggest that varicocele may be asso-
13.4.2 Prevalence and Influence
ciated with poor semen and hormone parameters
of Varicocele on Fertility (Nagao et al. 1986; Pasqualettto et al. 2005). Further, it
is assumed that the connections between varicocele and
The prevalence of idiopathic varicocele is high with testicular function are not static. Although initially fer-
estimates varying greatly. This is in part due to the tility parameters may be largely normal, these are said
nature and origin of the study population examined. to decline over the course of years, considerable indi-
Beyond this, the diagnosis of varicocele remains partly vidual variations notwithstanding (Chehval and Persel
subjective, all attempts to achieve objectivity notwith- 1992). These results are based on rather small patient
standing. This applies above all to first and second groups and in our own patient population we are not
degree varicoceles. In general, a prevalence of 15–20% able to corroborate any increase in the prevalence of
is assumed in the male population. Seventeen percent varicocele with the age of patients (for the proportion
of three million recruits for the German army, born of patients with varicocele is just as high in those
between 1937 and 1945, were found to have a varico- patients over 50 years as in 30-year-old patients).
cele (Nöske and Weidner 1999). When, however, stricter
diagnostic criteria were applied to sperm donors
(Handelsman et al. 1998) and volunteers in clinical
studies (Lemcke et al. 1996), a prevalence of 20% was
Thus to date there is no indisputable evidence
found. It is generally assumed that the proportion of
that varicocele itself reduces fertility, and the
men with varicocele and disturbed fertility is even
hypothesis that it is at most a cofactor for infertility
higher than that in the normal population. Here too esti-
in combination with other genetic and molecular
mates range up to 40%, depending on the composition
events is gaining importance (Marmar 2001).
of the investigator’s population and it stands to reason
that these estimates are higher due to referral bias when
made by surgically orientated urologists than by endo-
crinologists. We found a varicocele in 14.8% of nearly 13.4.3 Clinical Picture
13,000 consecutive patients attending our institute
(Table 4.2). This corresponds to statistics from other Infertility observed with varicocele manifests itself in
comparable centers of reproductive medicine. While a spermiogram characterized by oligo-, astheno- or
this makes varicocele the second most frequent path- teratozoospermia, or a variable combination of these
ological finding after idiopathic infertility, it does not findings, without a causal connection necessarily
give any indication of the importance of the finding and existing between these conditions and varicocele.
whether is it really a cause of infertility at all. Varicoceles can also be found in azoospermic patients
In general, a certain degree of uncertainty surrounds (Table 4.2). Varicoceles may also be associated with
the prevalence of varicocele in the normal population increased FSH values, indicating damage of the ger-
and in men attending a fertility clinic. There is urgent minal epithelium and a poor prognosis. It should be
need for clarification. The situation is even less clear emphasized that not all men bearing a varicocele show
concerning the influence of varicocele on fertility. decreased sperm parameters and impaired fertility.
The varying frequency of varicocele observed in proven Some patients mention feelings of pressure or some-
fathers and in patients attending the infertility clinic times pain in the afflicted testis or in the scrotum
prompted some investigators to assume a connection which can worsen after longer periods of standing or
between fertility and varicocele. The fact that varico- sitting in unchanged position. The cut of trousers and
cele does not exclude paternity, however, causes some underwear may also cause pain. Occasionally a varico-
investigators to deny this connection. This overlooks cele can become so enlarged as to represent a mechan-
the complexity of a couple’s fertility. Slight disturbance ical problem, especially in the elderly.

You might also like