Male and Female Sexual Dysfunction-072343266X
Male and Female Sexual Dysfunction-072343266X
Male and Female Sexual Dysfunction-072343266X
2
-agonist clonidine induced a profound decit in intro-
missive and ejaculatory behavior.
43
In contrast, a polar
analog of clonidine, which does not readily enter the
CNS, had no effect on male sexual behavior. Con-
versely, yohimbine, an
2
-antagonist with serotonergic
activity, which has long been used as an aphrodisiac
in humans (for review, see
44
) was shown to stimulate
sexual motivation in male rats. Similar facilitating
effects on sexual behavior were obtained with other
2
-antagonists, idazoxan and imiloxan. The central
origin of the prosexual effect of yohimbine was best
demonstrated by Sala and coworkers, who showed that
the most efcient dose delivered within the cerebral
ventricles facilitating sexual behavior was more than
one order of magnitude lower than the most efcient
dose delivered intraperitoneally.
45
It is noteworthy that
immunoreactivity associated with both
2
a and c
adrenoceptors was widely distributed within the brain,
including the hypothalamic nuclei involved in the
control of penile erections. The more selective
2
-
antagonist delequamine also increased sexual behavior
in mating experiments in a dose-related manner (for
example, it increased the percentage of naive male
rats intromitting and ejaculating), but, in contrast to
yohimbine, it did not decrease ejaculation latency.
Indeed, a combination of delequamine and 8-OH-
DPAT reproduced the effect of yohimbine, suggesting
that yohimbine also acts as a 5-HT1A receptor
agonist.
46
Although two metaanalyses concluded that yohim-
bine was superior to placebo in the treatment of erec-
tile dysfunction, yohimbine is much less efcient as a
proerectile drug in humans than in rats.
47
The exact
mechanism by which yohimbine can increase sexual
motivation is unknown at this time. It has been
postulated that such an
2
-antagonist would overall
increase noradrenergic transmission, which would
result in increased arousal. In a relevant context, this
would favor sexual arousal. An implication of central
-adrenoceptors in the antierectile role of -blockers
cannot be ruled out. For example, the mixed
1
- and
2
-adrenergic antagonists propranolol and pindolol
profoundly reduced ejaculatory behavior and other
parameters of sexual behavior in rats.
48
Dopamine
The prosexual effect of dopamine in humans was rst
suggested by the observation of increased sexual
activity in Parkinsons patients treated with dopamine
agonists.
49
Since then, double-blind placebo-controlled
studies have shown that 24 mg doses of apomorphine
delivered sublingually could increase the likelihood of
penile erections in men with erectile dysfunction.
50
It
is noteworthy that, whereas in rats, apomorphine is a
strong inducer of penile erection, it has only a facili-
tatory effect in humans, so that a sexually relevant
context is necessary to increase the likelihood of penile
erection occurring. Projections of dopaminergic nuclei
in the rat brain are represented in Figure 1.3B.
Nucleus accumbens
In mating experiments, injections of apomorphine in
the nucleus accumbens decreased the latency to begin
10 Central Neurophysiology of Penile Erection
copulating, whereas injections of apomorphine in the
striatum had no effect. Conversely, injections of apo-
morphine in the ventral tegmental area increased intro-
mission latency, likely by inhibiting the mesolimbic
dopamine pathway by stimulating autoreceptors, and
thus decreasing dopamine transmission at the level of
the nucleus accumbens (Fig. 1.3B).
16
Specic tests have been developed to discriminate
appetitive from consummatory components of sexual
behavior, such as the bilevel chambers test. In this test,
the male rat chases the female from one level to
another after each intromission. The number of level
changes in a xed time before the introduction of the
female is considered a measure of the anticipatory
phase (or motivation) of sexual activity. Such tests
proved that dopamine plays a role in the anticipatory
or appetitive phase of the sexual behavior in male rats,
as bilateral infusions of haloperidol into the nucleus
accumbens reduced the number of level changes (or
appetitive phase) but did not affect consummatory
measures of copulation.
16
Extracellular dopamine increased in the nucleus
accumbens when an estrous female was presented
behind a barrier, as well as during copulation, support-
ing the hypothesis that dopamine plays a positive role
in the anticipatory phase in male rats at the level of
the nucleus accumbens in physiological conditions.
51
Control experiments indicate that neither novelty nor
locomotor activity can account for the increased extra-
cellular dopamine concentrations observed in the
nucleus accumbens of male rats during the presentation
of a sexually receptive female and during copulation.
Moreover, exposure to a non-estrous female did not
elicit dopamine release in the nucleus accumbens in
male rat.
Median preoptic area
The MPOA receives dopaminergic innervation from
the incertohypothalamic pathway, originating from the
A13 cell group, and from the periventricular A14 cell
group. In mating experiments, injection of apomorphine
in the MPOA displayed clear prosexual effects. Injec-
tions of dopamine antagonist (cis-flupentixol) had the
opposite effect (increased intromission and ejaculation
latency, decreased number of rats that copulate and
ejaculate) and prevented the effects of apomorphine
injected in this area.
52
In the bilevel-chamber paradigm,
infusion of haloperidol in the MPOA reproduced all
the effects of its systemic administration, i.e., decrease
of the anticipatory/preparatory and consummatory
phase of copulatory behavior.
16
The stimulatory role of
dopamine in the MPOA in physiological conditions
has been conrmed, as dopamine and 3,4-
dihydroxyphenylacetic acid (DOPAC) were shown
to be increased during the precopulatory phase and
during copulation, and declined after ejaculation.
53
Hull and coworkers reported a consistent relationship
between precopulatory dopamine release in the MPOA
and the ability to copulate subsequently.
54
Further-
more, microinjections of a dopamine agonist into the
MPOA of animals with lesions of the amygdala restored
the copulatory behavior affected by the lesions.
55
Paraventricular nucleus of the
hypothalamus
Injection of doses of apomorphine as low as 5 ng in the
PVN can induce penile erections in a freely moving rat
without the presence of a female. Similar responses are
obtained when the selective D2-like agonist LY171555
is injected in place of apomorphine, but not with the
D1-like agonist SKF38393. Erections induced by
apomorphine injections in the PVN are abolished by
systemic pretreatment with the central D1-like antag-
onist, SCH23390, and D2 antagonist, sulpiride.
56
In
anesthetized rats (Fig. 1.6), penile erections induced by
peripheral delivery of apomorphine are antagonized
by a preceding injection in the PVN of SCH23390
or sulpiride.
57
It is noteworthy that apomorphine
injection in other hypothalamic structures, such as the
ventromedial and DM nucleus, the preoptic area, or the
nucleus accumbens, did not induce penile erection.
56
Conversely, lesions of the PVN strongly reduced the
proerectile activity induced by apomorphine in male
rats.
58
Those experiments therefore strongly suggest
that activation of dopaminergic receptor in the PVN are
responsible for the proerectile effect of apomorphine.
Nevertheless, dopamine is not the sole neurotrans-
mitter exogenously applied which is able to activate
proerectile neurons: injection of glutamate receptor
agonists as well as oxytocin in the PVN was also
shown to induce penile erection in anesthetized rats.
Altogether, activation of neurons in the PVN can be
considered as a sine qua non condition for the pro-
erectile effect of apomorphine to occur. According to
the available data, it cannot be determined whether
this effect is mediated by either dopamine D1-like and
D2-like receptors, or both. The measurement of some
dopamine release within the PVN concomitant to
penile erection during copulation would be a denite
argument for the physiological implication of dopamine
in the PVN in the induction of penile erection but, to
our knowledge, it has not yet been performed before,
during, or after copulation.
Spinal cord
It has recently been demonstrated that apomorphine
delivered at the lumbosacral level with an intrathecal
catheter, or systemically in spinalized animals, elicited
erectile activity in anesthetized rats.
59
The proerectile
activity of apomorphine was also shown to be con-
served in conscious spinalized rats.
60
These results
suggest that there may be a dopamine spinal component
The Aminergic Control of Sexual Behavior 11
in the control of penile erection. Accordingly, there
are dopamine projections from the A11 cell group to
the spinal cord as well as an intrinsic dopamine
innervation within the spinal cord.
61
Immunocyto-
chemical studies revealed that dopamine bers and
terminals exist in virtually all laminae throughout the
spinal cord.
62
Furthermore, studies using ligand-
binding techniques have shown the presence of D1
and D2 receptors in the spinal cord.
63
In male rats, D2
receptors identied with immunochemistry and in situ
hybridization have been located in the parasympathetic
nucleus of the lumbosacral spinal cord, which contains
the cellular bodies of the proerectile autonomic
neurons innervating the penis. D2 receptors have also
been found to be particularly abundant in the DM and
DL nucleus, which innervate the bulbospongiosus and
ischiocavernosus striated muscles involved in penile
rigidity in the rat.
64
OTHER NEUROTRANSMITTERS
INVOLVED IN THE CONTROL OF
SEXUAL BEHAVIOR
Nitric oxide
NO has been involved as an important proerectile
messenger in the CNS, and especially within the PVN.
An increase in NO production in the paraventricular
nucleus of the hypothalamus seems to be the primary
condition for the proerectile effect of apomorphine, n-
methyl aspartic acid and oxytocin, centrally delivered.
Dopaminergic agonists crossing the bloodbrain
barrier injected systemically induced penile erection
and increased the reaction products of NO with O
2
,
NO
2
and NO
3
and NO
3
and NO
3
and NO
3
1
-agonist, a calcium-channel blocker, and inhibits
platelet aggregation. Price and Grunhaus reported
reversal of clomipramine-induced anorgasmia with a
dose of 10 mg administered 90 min before coitus.
193
In a placebo-controlled study of 15 patients with
fluoxetine-induced anorgasmia, Jacobsen reported a
73% response rate to yohimbine.
194
Hollander reported
yohimbine reversal of anejaculation in ve of six men
with intercourse and/or masturbation.
195
The response
to yohimbine is typically delayed, taking up to 8 weeks,
and is often associated with adverse effects, including
nausea, headache, dizziness, and anxiety. Careful dose
titration is important as the extremes of dose have less
prosexual effect.
Buspirone is a benzodiazepine-class anxiolytic
which possesses 5HT-1A receptor agonist activity.
196
Othmer et al. reported normalization of sexual func-
tion in eight of 10 men with a generalized anxiety
disorder and associated sexual dysfunction using a
dose range of 1560 mg daily.
197
Bupropion is a novel
antidepressant which prolongs the action of dopamine
by reducing its uptake from the synaptic cleft.
198
Ashton and Rosen described reversal of SSRI-induced
anorgasmia in 66% of patients studied. An improve-
ment in sexual function was noted by Rowland in 14
non-depressed diabetic men with erectile dysfunction
with on-demand doses of 75150 mg.
199
Several authors have reported induction of pre-
mature ejaculation in rats following administration of
apomorphine, a central and peripheral dopamine-2
receptor agonist, at a dose of 50 g/kg. Dopamine
receptor antagonists block this effect.
200,201
A potential
role for apomorphine in the management of erectile
dysfunction was rst highlighted by Seagraves et al. and
more recently Heaton et al. have reported an efcacy
in excess of 50% in patients with psychogenic erectile
dysfunction when administered sublingually.
202,203
Adverse effects of nausea, vomiting, and dizziness are
minimized with this sublingual route of administration.
Aizenberg et al. examined the effect of the 5-HT2a/
2c and a
2
-antagonist mianserin in the treatment of
patients with sexual dysfunction induced by SSRIs.
204
Nine of the 15 subjects reported a marked improve-
ment in their sexual functioning in the areas of orgasm
and satisfaction, usually within the rst and second
week of mianserin treatment. The authors suggested
that coadministration of low-dose mianserin might
be an additional option in the treatment of sexual
dysfunction induced by SSRIs.
Quinelorane is a highly selective, potent dopamine-
2 agonist, which was extensively studied in animals in
the early part of this decade. Foreman and Hall observed
increased mounting, intromission, and ejaculation in
both sexually inactive and sluggish rats following
administration of quinelorane.
205
Prior administration
of a dopamine antagonist eliminated these stimulatory
effects, conrming that these sexual effects were due
to stimulation of dopamine receptors. They reported
that many rats failed to ejaculate at the extremes of
doses, with low doses causing sedation and high doses
causing hyperactive behavior, such as chewing or
snifng. Animals appears to become more sensitive to
dopamine agonists with increased use, suggesting that
abuse may eliminate any sexual benets. Eaton et al.
76 Ejaculatory Dysfunction
TABLE 5.5 Mechanism of action of drugs which increase dopamine neurotransmission.
Mechanism of increasing dopamine neurotransmission Drug
Prolong action by decreasing uptake Bupropion, cocaine
Prolong action by decreasing metabolism L-deprenyl
Increased release of dopamine Amfetamine
Direct stimulation of dopamine receptors with substitute neurotransmitters Bromocriptine, quinelorane,
apomorphine
Increase dopamine synthesis by providing precursors L-dopa
injected quinelorane directly into the rat paraventricular
nucleus and MPOA and reported different responses
with different doses.
206
At extremes, quinelorane could
cause paradoxical PE, reduced sexual desire, and erec-
tile dysfunction.The reduced sexual response observed
at low doses is due to stimulation of dopamine
autoreceptors which decrease dopamine activity
and respond to lower doses than do the stimulatory
dopamine-2 receptors. In theoretical clinical use,
lowering the dose to avoid excess excitement may
result in worse sexual dysfunction than prior to treat-
ment. Human double-blind placebo-controlled clinical
studies of quinelorane were commenced in the late
1980s, involving multiple sites and more than 500
men and women with erectile dysfunction, reduced
sexual desire, and reduced arousal. The US Food and
Drug Administration review of the trial data was
inconclusive and concern was expressed over the more
than 50% incidence of nausea and hypotension and
the indirect negative sexual adverse effects. Clinical
studies were terminated and the results remain con-
dential and unpublished.
In patients who do not achieve antegrade ejaculation
with either surgery or medication, sperm retrieval and
articial insemination is an alternative approach. The
basic method of sperm retrieval involves recovery of
urine by either catheter or voiding after masturbation,
and then centrifugation and isolation of the sperm.
There are several more invasive methods of sperm
retrieval available. Sperm can be harvested directly
from the vas percutaneously or during microsurgical
vasotomy under local anesthetic. Ejaculation can be
produced with the use of vibration or transrectal
electrostimulation. This is most commonly performed
in men with spinal cord damage. Approximately 70%
of spinal cord-injured men can obtain ejaculation with
this method but each man must be closely monitored
for a hypertensive crisis due to autonomic dysreflexia.
Sperm banking can be conducted in men with
testicular cancer prior to surgery. However more than
50% of these men are subfertile at the time of diag-
nosis and this may explain the relatively poor results
from articial insemination with stored sperm in these
patients.
EJACULATORY DYSFUNCTION IN
SPINAL CORD-INJURED PATIENTS
The ability to ejaculate is severely impaired by spinal
cord injury (SCI). Bors and Comarr highlighted the
impact of the level and completeness of SCI on post-
injury erectile and ejaculatory capacity (Table 5.6).
207,208
Unlike erectile capacity, the ability to ejaculate
increases with descending levels of spinal injury. Fewer
than 5% of patients with complete upper motor neuron
lesions retain the ability to ejaculate. Ejaculation rates
are higher (15%) in patients with both a lower motor
neuron lesion and an intact thoracolumbar sympa-
thetic outflow. Approximately 22% of patients with an
incomplete upper motor neuron lesion and almost all
men with incomplete lower motor neuron lesions will
retain the ability to ejaculate. In those patients who
are capable of successful ejaculation, the sensation of
orgasm may be absent and retrograde ejaculation often
occurs.
Several techniques for obtaining semen from spinal
cord-injured men with ejaculatory dysfunction have
been reported. The intrathecal administration of the
anticholinesterase inhibitors neostigmine and subcu-
taneous physostigmine to induce ejaculation is more of
historical interest and is no longer used due to a 60%
risk of autonomic dysreflexia, especially in men with
injuries above the T5 level.
209,210
The use of electro-
ejaculation to obtain semen by electrical stimulation of
efferent sympathetic bers of the hypogastric plexus
is an effective and safe method of obtaining semen.
Brindley reported that 71% of men with SCI who
underwent electroejaculation achieved ejaculation.
211
Ohl et al. reported that sperm density and motility
were higher in those with incomplete lesions.
212
Vibratory stimulation is successful in obtaining
semen in up to 70% of men with SCI.
213
This tech-
nique induces a reflexogenic ejaculation via the sacral
roots and the ejaculatory coordination center in the
upper thoracolumbar spinal cord. It is however
associated with a signicantly higher risk of autonomic
dysreflexia than electroejaculation. Pretreatment with a
fast-acting vasodilator such as nifedipine will minimize
the risk of severe hypertension should autonomic
TABLE 5.6 Correlation of erection, ejaculation, and intercourse with level and severity of spinal cord injury
1
Cord lesion Reflexogenic Psychogenic Successful Ejaculation
erections (%) erections (%) coitus (%) (%)
Upper motor neuron lesion
Complete 92 9 66 1
Incomplete 93 48 86 22
Lower motor neuron lesion
Complete 0 24 33 15
Incomplete 0 1 100 100
Ejaculatory Dysfunction in Spinal Cord-injured Patients 77
dysreflexia occur with either form of treatment.
214
Percutaneous aspiration of semen from the vas deferens
has also been reported as a means of harvesting semen
for use with articial reproductive techniques.
215
Semen collected from men with SCI is often initially
senescent and of poor quality, with a low sperm count
and reduced sperm motility, but may improve with
subsequent ejaculations. This poor semen quality may
be due to chronic urinary tract infection, sperm content
with urine, chronic use of various medications, elevated
scrotal temperature due to prolonged sitting, and stasis
of prostatic fluid. Testicular biopsies in spinal cord-
injured men demonstrate a wide range of testicular
dysfunction, including hypospermatogenesis, maturation
arrest, atrophy of seminiferous tubules, germinal cell
hypoplasia, interstitial brosis and Leydig cell hyper-
plasia. In addition prostatitis secondary to prolonged
catheterization, epididymitis, and epididymoorchitis
can precipitate obstructive ductal lesions and testicular
damage.
INHIBITED MALE ORGASM
Inhibited male orgasm is the psychogenic variant of
retarded ejaculation, also called ejaculatory incom-
petence by Masters and Johnson.
27
It may be dened
as recurrent and persistent inhibition of the male
orgasm as manifested by delay in or absence of ejacu-
lation following an adequate phase of sexual excite-
ment. It may range in severity from very severe forms
in which a man has never been able to experience
waking climax, even with masturbation, to milder
forms in which intravaginal climax occurs, but only
after prolonged thrusting. Clinically it is the least
common sexual disorder and it most often presents as
a primary disorder. In most cases, however, the problem
is situational. Orgasm occurs readily with masturbation
but not during intercourse. Usually only the rare global
case of retarded ejaculation presents any difculty
with differential diagnosis. Secondary retarded ejacu-
lation, when its situational, strongly suggests a
problematic relationship. Global secondary retarded
ejaculation suggests the development of some psycho-
physiologic or pharmacologic cause such as sedative
hypnotic abuse, narcotic or alcohol abuse. In very rare
instances neurologic disease or neurologic trauma may
account for this disorder.
The prevailing wisdom holds that inhibited male
orgasm is analogous to female anorgasmia. According
to this theory, psychogenically mediated reflex
inhibition occurs despite high levels of sexual tension.
Apfelbaum has proposed, however, that the disorder
is best understood as the surface manifestation of an
underlying disorder of sexual desire.
216
He has theorized
that these patients, though they have erections, never
pass from initial penile engorgement to the plateau
level with high levels of sexual tension. There are
patients who typify both of the above perspectives but
Martin believes that the majority of patients do reach
plateau at some point during thrusting, only to
experience reflex inhibition, and subsequently subside
back to preplateau levels of tension.
217
A wide variety
of psychologic factors may be responsible for the
inhibition, including fear of impregnating the partner,
religion, guilt, depressed or repressed hostility towards
the partner, oedipal fears of retaliation, and fears of
soiling or deling the partner with semen.
As a rule, treatment outcome with behavioral sex
therapy tends to be less successful for orgasm
inhibition than for other sexual disorders. The basic
treatment strategy requires that the man move by the
method of successive approximation from extravaginal
ejaculation to ejaculation in the vagina. A treatment
sequence might involve a progression from solitary
masturbation to masturbation with his wife in the next
room, to masturbation in her presence but with her
back turned, to masturbation with her looking on, to
wife-assisted masturbation to orgasm. Once he has
traversed these steps with successful ejaculatory
outcome the patient is asked to insert his penis into
the lubricated vagina just at the point of ejaculatory
inevitability.After several repetitions of this maneuver,
which is designed to desensitize the man to the anxiety
associated with intravaginal orgasm, he is asked to
insert at plateau but before ejaculatory inevitability. If
he can proceed to ejaculation he is given permission to
insert yet earlier in the sexual response cycle. The
couple is encouraged to do everything possible to
enhance the erotic aspects of the sexual experience
and the wife is taught how to cup her husbands
testicles for an extra sensation when he is at high levels
of erotic tension. Liberal use of fantasy is encouraged,
as is the use of commercially available erotica. It is
particularly important that the man should not
attempt insertion until he has reached high levels of
erotic tension during sex play.
REFERENCES
1. Lipshultz LI, McConnell J, Benson GS. Current concepts
of the mechanisms of ejaculation. Normal and abnormal
states. J Reprod Med 1981; 26:499.
2. Yeates W. Ejaculatory disturbances. In: Pryor JP, Lipschultz
L, eds. Andrology. London: Butterworths; 1987:183216.
3. Reading A, Wiest W. An analysis of self-reported sexual
behavior in a sample of normal males. Arch Sex Behav 1984;
13:69.
4. Nathan SG.The epidemiology of the DSM-III psychosexual
dysfunctions. J Sex Marit Ther 1986; 12:267.
5. Spector KR, Boyle M. The prevalence and perceived
aetiology of male sexual problems in a non-clinical sample.
Br J Med Psychol 1986; 59:351.
78 Ejaculatory Dysfunction
6. Spector IP, Carey M. Incidence and prevalence of the sexual
dysfunctions: a critical review of the empirical literature.Arch
Sex Behav 1990; 19:389.
7. Grenier G, Byers ES. The relationships among ejaculatory
control, ejaculatory latency, and attempts to prolong hetero-
exual intercourse. Arch Sex Behav 1997; 26:27.
8. American Psychiatry Association. Diagnostic and statistical
manual of mental disorders, DSM-IV, 4th edn. Washington,
DC: American Psychiatric Association; 1994:509.
9. Gross S. Practical treatise on impotence and sterility.
Edinburgh: YJ Pentland; 1887.
10. von Krafft-Ebing RF. Psychopathia sexualis, 11th edn.
Stuttgart: Publishing Hause Enke; 1901.
11. Abraham K. Ueber Ejaculatio Praecox. Z Aerztl
Psychoanal 1917; 4:171.
12. Stekel W. Impotence in the male, vol. 2. New York:
Liveright Publishing; 1927.
13. Schapiro B. Premature ejaculation: a review of 1130
cases. J Urol 1943; 50:374.
14. Masters W, Johnson V. Human sexual response. Boston:
Little, Brown; 1966.
15. Kotin J, Wilbert DE, Verburg D et al. Thioridazine and
sexual dysfunction. Am J Psychiatry 1976; 133:82.
16. Deveaugh-Geiss J, Landau P, Katz R. Preliminary results
from a multicentre trial of clomipramine in obsessive com-
pulsive disorder. Psychopharmacol Bull 1989; 25:36.
17. Monteiro WO, Noshirvani HF, Marks IM et al.Anorgasmia
from clomipramine in obsessive-compulsive disorder: a
controlled trial. Br J Psychiatry 1987; 151:107.
18. Waldinger MD, Hengeveld MW, Zwinderman AH.
Paroxetine treatment of premature ejaculation: a double-
blind, randomized, placebo-controlled study. Am J Psychiatry
1994; 151:1377.
19. McMahon CG, Touma K. Treatment of premature ejacu-
lation with paroxetine hydrochloride as needed: 2 single-
blind placebo controlled crossover studies. J Urol 1999;
161:1826.
20. Waldinger MD, Hengeveld M. Neuroseksuologie en
seksuele psychofarmacologie. Tijdschr Psychiatr 2000; 8.
21. Olivier B, van Oorschot R, Waldinger MD. Serotonin
serotonergic receptors, selective serotonin reuptake inhibitors
and sexual behavior. Int Clin Psychopharmacol 1998;
(suppl. 13):s9.
22. Waldinger MD, Olivier B. Hersenonderzoek en
farmacologie: serotonine, seks en agressie. In: Wolters-
Schweitzer MHJ, Beuger CL, eds. Het brein belicht: opstellen
over niet-aangeboren hersenletsel. Utrecht: Uitgeverij
Lemma: 2001:5563.
23. Waldinger MD, Rietschel M, Nothen MM et al. Familial
occurrence of primary premature ejaculation. Psychiatr Genet
1998; 8:37.
24. Colpi GM, Fanciullacci F, Beretta G et al. Evoked sacral
potentials in subjects with true premature ejaculation.
Andrologia 1986; 18:583.
25. Fanciullacci F, Colpi GM, Beretta G et al. Cortical
evoked potentials in subjects with true premature ejaculation.
Andrologia 1988; 20:326.
26. Kaplan HS, Kohl RN, Pomeroy WB et al. Group treat-
ment of premature ejaculation. Arch Sex Behav 1974; 3:443.
27. Masters W, Johnson V. Human sexual inadequacy. Boston:
Little Brown, 1970.
28. Segraves RT, Saran A, Segraves K et al. Clomipramine vs
placebo in the treatment of premature ejaculation. Paper
presented at the meeting of the International Academy of Sex
Research, Prague, Czechoslovakia, 1992.
29. Waldinger MD, Hengeveld M, Zwinderman AH. An
empirical operationalization study of DSM-IV diagnostic
criteria for premature ejaculation. Int J Psych Clin Pract
1998; 2:287.
30. Segraves RT, Saran A, Segraves K et al. Clomipramine
versus placebo in the treatment of premature ejaculation: a
pilot study. J Sex Marit Ther 1993; 19:198.
31. Cooper AJ, Magnus RV.A clinical trial of the beta blocker
propranolol in premature ejaculation. J Psychosom Res 1984;
28:331.
32. Spiess WF, Geer JH, ODonohue WT. Premature
ejaculation: investigation of factors in ejaculatory latency.
J Abnorm Psychol 1984; 93:242.
33. Strassberg DS, Mahoney JM, Schaugaard M et al. The
role of anxiety in premature ejaculation: a psychophysio-
logical model. Arch Sex Behav 1990; 9:251.
34. Strassberg DS, Kelly MP, Carroll C et al. The psycho-
physiological nature of premature ejaculation. Arch Sex
Behav 1987; 16:327.
35. LoPiccolo J. Direct treatment of sexual dysfunction in
the couple. In: Money J, Mesaph H, eds. Handbook of
sexology, vol. 5. Selected syndromes and therapy. New York:
Elsevier; 1978:1227.
36. Kilmann PR, Auerbach R. Treatments of premature ejacu-
lation and psychogenic impotence: a critical review of the
literature. Arch Sex Behav 1979; 8:81.
37. Trudel G, Proulx S. Treatment of premature ejaculation by
bibliotherapy: an experimental study. Sex Marit Ther 1987;
2:163.
38. Zeiss RA, Christensen A, Levine AG. Treatment for
premature ejaculation through male-only groups. J Sex Marit
Ther 1978; 4:139.
39. Schover LR, Friedman JM, Weiler SJ et al. Multiaxial
problem-oriented system for sexual dysfunctions. Arch Gen
Psychiat 1982; 39:614.
40. Gebhard PH. Factors in marital orgasm. J Soc Issues
1966; 22:88.
41. Darling CA, Davidson JK, Cox RP. Female sexual response
and the timing of partner orgasm. J Sex Marit Ther 1991; 17:3.
42. Kinsey AC, Pomeroy WB, Martin CE. Sexual behavior in
the human male. Philadelphia: WB Saunders; 1948.
43. Westoff C. Coital frequency and contraception. Fam
Plann Perspect 1974; 6:136.
References 79
44. McMahon CG. Long term results of treatment of
premature ejaculation with selective serotonin re-uptake
inhibitors. Int J Impot Res 2002; 14:S19.
45. Lawrance K, Byers ES. Validation of the interpersonal
exchange model of sexual satisfaction in long-term relation-
ships. In: Byers ES (chair) Testing common assumptions about
human sexuality. Symposium presented at the meeting of the
Canadian Psychological Association Montreal, 1993.
46. McCarthy B. Cognitive-behavioral strategies and tech-
niques in the treatment of early ejaculation. In: Leiblum S,
Rosen, eds. Principles and practices of sex therapy: Update
for the 1990s. New York: Guilford Press; 1988:141167.
47. Vandereycken W. Towards a better delineation of
ejaculatory disorders. Acta Psychiatr Belg 1986; 86:57.
48. Zilbergeld B. Male sexuality. Toronto: Bantam; 1978.
49. Gospodinoff ML. Premature ejaculation: clinical sub-
groups and etiology. J Sex Marit Ther 1989; 15:130.
50. Grenier G, Byers ES. Controlling ejaculation: Reality or
pipe dream. In: Byers ES (chair) Testing common assumptions
about human sexuality. Symposium presented at the meeting
of the Canadian Psychological Association Montreal, 1993.
51. Rowland DL, Cooper SE, Schneider M. Dening pre-
mature ejaculation for experimental and clinical investigations.
Arch Sex Behav 2001; 30:235.
52. Williams W. Secondary premature ejaculation. Aust NZ J
Psychiatry 1984; 18:333.
53. Zilbergeld B. The new male sexuality. Toronto: Bantam;
1992.
54. Kockott G, Feil W, Ferstl R et al. Psycho-physiological
aspects of male sexual inadequacy: results of an experimental
study. Arch Sex Behav 1980; 9:477.
55. Segraves RT. Treatment of premature ejaculation with
lorazepam. Am J Psychiatry 1987; 144:1240.
56. Kaplan HS. PE How to overcome premature ejacu-
lation. New York: Brunner/Mazel; 1989.
57. Kockott G, Feil W, Revenstorf D et al. Symptomatology
and psychological aspects of male sexual inadequacy: results
of an experimental study. Arch Sex Behav 1980; 9:457.
58. Crooks R, Baur K. Our sexuality. Don Mills, Ontario:
Benjamin/Cummings; 1990.
59. Hong LK. Survival of the fastest: on the origin of pre-
mature ejaculation. J Sex Res 1984; 20:109.
60. Bixler RH. Of apes and men (including female!). J Sex
Res 1986; 22:255.
61. Ellis H. Studies in the psychology of sex. New York:
Random House; 1936.
62. Rowland DL, Greenleaf W, Mas M et al. Penile and nger
sensory thresholds in young, aging, and diabetic males. Arch
Sex Behav 1989; 18:1.
63. Xin ZC, Chung WS, Choi YD et al. Penile sensitivity in
patients with primary premature ejaculation. J Urol 1996;
156:979.
64. Xin ZC, Choi YD, Rha KH et al. Somatosensory evoked
potentials in patients with primary premature ejaculation.
J Urol 1997; 158:451.
65. Paick JS, Jeong H, Park MS. Penile sensitivity in men
with premature ejaculation. Int J Impot Res 1998; 10:247.
66. Rowland DL, Haensel SM, Blom JH et al. Penile
sensitivity in men with premature ejaculation and erectile
dysfunction. J Sex Marit Ther 1993; 19:189.
67. Edwards A, Husted J. Penile sensitivity, age and sexual
behavior. J Clin Psychol 1976; 32:697.
68. Newman HF. Vibratory sensitivity of the penis. Fertil
Steril 1970; 21:791.
69. Yang CC, Bradley WE. Neuroanatomy of the penile
portion of the human dorsal nerve of the penis. Br J Urol
1998; 82:109.
70. Semans JH. Premature ejaculation: new approach. South
Med J 1956; 49:353.
71. Pirke KM, Kockott G, Aldenhoff J et al. Pituitary gonadal
system function in patients with erectile impotence and
premature ejaculation. Arch Sex Behav 1979; 8:41.
72. Cohen PG. The association of premature ejaculation and
hypogonadotropic hypogonadism. J Sex Marit Ther 1997;
23:208.
73. Cohen PG, Holbrook JM. Effects of fenfluramine on
ejaculatory function, luteinizing hormone and testosterone
levels in men with hypogonadotropic hypogonadism and
premature ejaculation. Int Clin Psychopharmacol 1999;
14:91.
74. Marson L, McKenna KE.The identication of a brainstem
site controlling spinal sexual reflexes in male rats. Brain Res
1990; 515:303.
75. Yells DP, Prendergast MA, Hendricks SE et al.
Fluoxetine-induced inhibition of male rat copulatory behavior:
modication by lesions of the nucleus paragigantocellularis.
Pharmacol Biochem Behav 1994; 49:121.
76. MacLean PD. Brain mechanisms of primal sexual
functions and related behavior. In: Sandler M, Gessa GL, eds.
Sexual behavior: pharmacology and biochemistry. New York:
Raven Press; 1975.
77. Yells DP, Hendricks SE, Prendergast MA. Lesions of the
nucleus paragigantocellularis: effects on mating behavior in
male rats. Brain Res 1992; 596:73.
78. Marson L, McKenna KE. A role for 5-hydroxytryptamine
in descending inhibition of spinal sexual reflexes. Exp Brain
Res 1992; 88:313.
79. Coolen LM, Peters HJ,Veening JG. Fos immunoreactivity
in the rat brain following consummatory elements of sexual
behavior: a sex comparison. Brain Res 1996; 738.
80. Coolen LM, Olivier B, Peters HJ et al. Demonstration of
ejaculation-induced neural activity in the male rat brain using
5-HT1A agonist 8-OH-DPAT. Physiol Behav 1997; 62:881.
81. Coolen LM, Peters HJ, Veening JG. Anatomical
interrelationships of the medial preoptic area and other brain
80 Ejaculatory Dysfunction
regions activated following male sexual behavior: a combined
fos and tract-tracing study. J Comp Neurol 1998; 397:421.
82. Ahlenius S, Larsson K, Svensson L et al. Effects of a new
type of 5-HT receptor agonist on male rat sexual behavior.
Pharmacol Biochem Behav 1981; 15:785.
83. Waldinger MD, Berendsen HH, Blok BF et al. Premature
ejaculation and serotonergic antidepressants-induced delayed
ejaculation: the involvement of the serotonergic system.
Behav Brain Res 1998; 92:111.
84. Waldinger MD. The neurobiological approach to pre-
mature ejaculation. J Urol 2002; 168:2359.
85. De Amicis LA, Goldberg DC, LoPiccolo J et al. Clinical
follow-up of couples treated for sexual dysfunction. Arch Sex
Behav 1985; 14:467.
86. Hawton K, Catalan J, Martin P et al. Long-term outcome
of sex therapy. Behav Res Ther 1986; 24:665.
87. Patterson WM. Fluoxetine-induced sexual dysfunction.
J Clin Psychiatry 1993; 54:71.
88. Hsu JH, Shen WW. Male sexual side effects associated
with antidepressants: a descriptive clinical study of 32 patients.
Int J Psychiatry Med 1995; 25:191.
89. Labbate LA, Grimes JB, Arana GW. Serotonin reuptake
antidepressant effects on sexual function in patients with
anxiety disorders. Biol Psychiatry 1998; 43:904.
90. Montejo-Gonzalez AL, Llorca G, Izquierdo JA et al.
SSRI-induced sexual dysfunction: fluoxetine, paroxetine,
sertraline, and fluvoxamine in a prospective, multicenter, and
descriptive clinical study of 344 patients. J Sex Marit Ther
1997; 23:176.
91. Ahlenius S, Heimann M, Larsson K. Prolongation of
the ejaculation latency in the male rat by thioridazine and
chlorimipramine. Psychopharmacology (Berl) 1979; 65:137.
92. Colpi GM, Fanciullacci F, Aydos K et al. Effectiveness
mechanism of chlomipramine by neurophysiological tests in
subjects with true premature ejaculation. Andrologia 1991;
23:45.
93. Steiger A. Effects of clomipramine on sleep EEG and
nocturnal penile tumescence: a long-term study in a healthy
man. J Clin Psychopharmacol 1988; 8:349.
94. Aizenberg D, Zemishlany Z, Hermesh H et al. Painful
ejaculation associated with antidepressants in four patients.
J Clin Psychiatry 1991; 52:461.
95. Goodman RE.An assessment of clomipramine (Anafranil)
in the treatment of premature ejaculation. J Int Med Res
1980; 8:53.
96. Girgis SM, El-Haggar S, El-Hermouzy S. A double-blind
trial of clomipramine in premature ejaculation. Andrologia
1982; 14:364.
97. Althof SE, Levine SB, Corty EW et al. A double-blind
crossover trial of clomipramine for rapid ejaculation in 15
couples. J Clin Psychiatry 1995; 56:402.
98. Montorsi F, Guazzoni L, Barbieri L et al. Chlomipramine
for premature ejaculation: a randomized double blind placebo
controlled study. Abstract D74. Proceedings of the VI World
Meeting on Impotence, Singapore 1994.
99. Haensel SM, Rowland DL, Kallan KT. Clomipramine and
sexual function in men with premature ejaculation and
controls. J Urol 1996; 156:1310.
100. Strassberg DS, de Gouveia Brazao CA, Rowland DL
et al. Clomipramine in the treatment of rapid (premature)
ejaculation. J Sex Marit Ther 1999; 25:89.
101. Kim SC, Seo KK. Efcacy and safety of fluoxetine,
sertraline and clomipramine in patients with premature
ejaculation: a double-blind, placebo controlled study. J Urol
1998; 159:425.
102. Hsieh JT, Chang HC, Law HS et al. In vivo evaluation
of serotonergic agents and alpha-adrenergic blockers on pre-
mature ejaculation by inhibiting the seminal vesicle pressure
response to electrical nerve stimulation. Br J Urol 1998;
82:237.
103. Berendsen H, Broekkamp C. Behavioral evidence for
functional interactions between 5-HT receptor sub-types in
rats and mice. Br J Pharmacol 1990; 101:667.
104. Hillegaart V, Ahlenius S. Facilitation and inhibition of
male rat ejaculatory behavior by the respective 5-HT1A and
5-HT1B receptor agonists 8-OH-DPAT and anpirtoline, as
evidenced by use of the corresponding new and selective
receptor antagonists NAD-299 and NAS-181. Br J Pharmacol
1998; 125:1733.
105. Bergstrom RF, Lemberger L, Farid NA et al. Clinical
pharmacology and pharmacokinetics of fluoxetine: a review.
Br J Psychiatry 1998 (suppl):47.
106. Renyi L. The effect of selective 5-hydroxytryptamine
uptake inhibitors on 5-methoxy-N,N-dimethyltryptamine-
induced ejaculation in the rat. Br J Pharmacol 1986; 87:639.
107. Mos J, Mollet I, Tolboom JT et al. A comparison of the
effects of different serotonin reuptake blockers on sexual
behavior of the male rat. Eur Neuropsychopharmacol 1999;
9:123.
108. Vega Matuszcyk J, Larsson K, Eriksson E. The selective
serotonin reuptake inhibitor fluoxetine reduces sexual moti-
vation in male rats. Pharmacol Biochem Behav 1998; 60:527.
109. Yilmaz U, Tatlisen A, Turan H et al. The effects of
fluoxetine on several neurophysiological variables in patients
with premature ejaculation. J Urol 1999; 161:107.
110. Crenshaw RT. Treatment of premature ejaculation with
fluoxetine. In: Proceedings of American Psychiatric Association
Meeting May 1992.
111. Power-Smith P. Benecial sexual side-effects from
fluoxetine. Br J Psychiatry 1994; 164:249.
112. Forster P, King J. Fluoxetine for premature ejaculation.
Am J Psychiatry 1994; 151:1523.
113. Lee HS, Song DH, Kim CH et al. An open clinical trial
of fluoxetine in the treatment of premature ejaculation. J Clin
Psychopharmacol 1996; 16:379.
114. Grazziotin A. Prozac in the treatment of premature
ejaculation. J Urol 1995; 153:474A.
References 81
115. Kara H, Aydin S, Yucel M et al. The efcacy of fluoxetine
in the treatment of premature ejaculation: a double-blind
placebo controlled study. J Urol 1996; 156:1631.
116. Kindler S, Dolberg OT, Cohen H et al. The treatment of
comorbid premature ejaculation and panic disorder with
fluoxetine. Clin Neuropharmacol 1997; 20:466.
117. Haensel SM, Klem T, Hop WC et al. Fluoxetine and
premature ejaculation: a double-blind, crossover, placebo-
controlled study. J Clin Psychopharmacol 1998; 18:72.
118. Murray MJ, Hooberman D. Fluoxetine and prolonged
erection. Am J Psychiatry 1993; 150:167.
119. Benazzi F. Involuntary sperm emission with fluoxetine.
Can J Psychiatry 1995; 40:431.
120. Koe BK, Lebel A, Burkhart CA et al. Sertraline: a
potent inhibitor of (+) [3H]3-PPP binding to brain sigma ()
receptors. Society for Neuroscience Abstracts 19th Meeting
1989; 15 (Pt.2):1235.
121. Swartz DA. Sertraline hydrochloride for premature
ejaculation. J Urol 1994; 1 (suppl.): 51.
122. Balbay MD, Yildiz M, Salvarci A et al. Treatment of
premature ejaculation with sertralin. Int Urol Nephrol 1998;
30:81.
123. McMahon CG. Treatment of premature ejaculation with
sertraline hydrochloride. Int J Impot Res 1998; 10:181.
124. Kaplan PM. The use of serotonergic uptake inhibitors in
the treatment of premature ejaculation. J Sex Marit Ther
1994; 20:321.
125. Wise TN. Sertraline as a treatment for premature ejacu-
lation. J Clin Psychiatry 1994; 55:417.
126. Mendels J, Camera A, Sikes C. Sertraline treatment for
premature ejaculation. J Clin Psychopharmacol 1995;
15:341.
127. McMahon CG. Treatment of premature ejaculation with
sertraline hydrochloride: a single-blind placebo controlled
crossover study. J Urol 1998; 159:1935.
128. Biri H, Isen K, Sinik Z et al. Sertraline in the treatment
of premature ejaculation: a double-blind placebo controlled
study. Int Urol Nephrol 1998; 30:611.
129. Waldinger MD, Hengeveld MW, Zwinderman AH
et al. Effect of SSRI antidepressants on ejaculation: a
double-blind, randomized, placebo-controlled study with
fluoxetine, fluvoxamine, paroxetine, and sertraline. J Clin
Psychopharmacol 1998; 18:274.
130. Kaye CM, Haddock RE, Langley PF et al. A review of
the metabolism and pharmacokinetics of paroxetine in man.
Acta Psychiatr Scand 1991; 80:60.
131. Waldinger MD, Hengeveld MW, Zwinderman AH.
Ejaculation-retarding properties of paroxetine in patients with
primary premature ejaculation: a double-blind, randomized,
doseresponse study. Br J Urol 1997; 79:592.
132. Ludovico GM, Corvasce A, Pagliarulo G et al. Paroxetine
in the treatment of premature ejaculation. Br J Urol 1996;
77:881.
133. Giammusso B, Morgia G, Spampinato A et al.
[Paroxetine in the treatment of premature ejaculation.] Arch
Ital Urol Androl 1997; 69:11.
134. McMahon CG, Touma K. Treatment of premature ejacu-
lation with paroxetine hydrochloride. Int J Impot Res 1999;
11:241.
135. Isaksen PM. [The effect of an antidepressive agent on
premature ejaculation.] Tidsskr Nor Laegeforen 1995;
115:1616.
136. Hiemke C. [Paroxetine: pharmacokinetics and pharma-
codynamics.] Fortschr Neurol-Psychiatr 1994; 62:2.
137. Bloomer JC, Woods FR, Haddock RE et al. The role of
cytochrome P450 2D6 in the metabolism of paroxetine by
human liver microsomes. Br J Clin Pharmacol 1992; 33:532.
138. Preskorn S. Pharmacokinetics of anti-depressants: why
and how they are relevant to treatment. J Clin Psychiatry
1993; 54:14.
139. McMahon CG. High dose sildenal citrate as a salvage
therapy for severe erectile dysfunction. Int J Impot Res 2002;
14:533.
140. Murat Basar M, Atan A, Yildiz M et al. Comparison of
sertraline to fluoxetine with regard to their efcacy and side
effects in the treatment of premature ejaculation. Arch Esp
Urol 1999; 52:1008.
141. Dawson T, Snyder SH. Gases as biological messengers:
nitric oxide carbon monoxide in the brain. J Neurosci 1994;
14:5147.
142. Garthwaite J, Charles SL, Chess-Williams CR.
Endothelium-derived relaxing factor release on activation of
NMDA receptors suggests role as intercellular messenger in
the brain. Nature 1988; 336.
143. Bredt DS, Glatt CE, Hwang PM et al. Nitric oxide
synthase protein and mRNA are discretely localized in
neuronal populations of mammalian CNS together with
NADPH diaphorase. Neuron 1991; 7.
144. Nelson RJGE, Demas PL, Huang MC et al. Behavioral
abnormalities in male mice lacking neuronal nitric oxide
synthase. Nature 1995; 378:383.
145. Vincent SR, Kimura H. Histochemical mapping of nitric
oxide synthase in the rat brain. Neuroscience 1992; 46:755.
146. Melis MR, Argiolas A. Nitric oxide synthase inhibitors
prevent apomorphine- and oxytocin-induced penile erection
and yawning in male rats. Brain Res Bull 1993; 32:71.
147. Melis MR, Stancampiano R, Argiolas A. Nitric oxide
synthase inhibitors prevent N-methyl-D-aspartic acid-induced
penile erection and yawning in male rats. Neurosci Lett 1994;
179:9.
148. Sato Y, Horita H, Kurohata T et al. Effect of the nitric
oxide level in the medial preoptic area on male copulatory
behavior in rats. Am J Physiol 1998; 274:R243.
149. Lorrain DS, Hull EM. Nitric oxide increases dopamine
and serotonin release in the medial preoptic area. Neuroreport
1993; 5:87.
82 Ejaculatory Dysfunction
150. Lorrain DS, Matuszewich L, Howard RV et al. Nitric
oxide promotes medial preoptic dopamine release during
male rat copulation. Neuroreport 1996; 8:31.
151. Giuliano F, Rampin O, Brown K et al. Stimulation of
medial preoptic area of hypothalamus in the rat elicits increase
in intracavernous pressure. Neurosci Lett 1996; 209:209.
152. Hull EM, Lumley LA, Matuszewich L et al. The roles
of nitric oxide in sexual function of male rats. Neuro-
pharmacology 1994; 33:1499.
153. Kriegsfeld LJ, Demas GE, Huang PL et al. Ejaculatory
abnormalities in mice lacking the gene for endothelial nitric
oxide synthase (eNOS/). Physiol Behav 1999; 67:561.
154. Tome AR, da Silva JC, Souza AA et al. Possible involve-
ment of nitric oxide in pilocarpine induced seminal emission
in rats. Gen Pharmacol 1999; 33:479.
155. Giuliani DOA, Ferrari F. Influence of sildenal on
copulatory behavior in sluggish or normal ejaculator male rats:
a central dopamine mediated effect? Neuropharmacology
2002; 42:562.
156. Ottani AGD, Ferrari F. Modulatory activity of sildenal
on copulatory behavior of both intact and castrated male rats.
Pharmacol Biochem Behav 2002; 72:717.
157. Abdel-Hamid IA, El Naggar EA, El Gilany AH.
Assessment of as needed use of pharmacotherapy and the
pausesqueeze technique in premature ejaculation. Int J
Impot Res 2001; 13:41.
158. Salonia A, Maga T, Colombo R et al.A prospective study
comparing paroxetine alone versus paroxetine plus sildenal
in patients with premature ejaculation. J Urol 2002;
168:2486.
159. Chen J, Mabjeesh NJ, Matzkin H et al. Efcacy of
sildenal as adjuvant therapy to selective serotonin reuptake
inhibitor in alleviating premature ejaculation. Urology 2003;
61:197.
160. Stefanick ML, Smith ER, Davidson JM. Penile reflexes
in intact rats following anesthetization of the penis and
ejaculation. Physiol Behav 1983; 31:63.
161. Sachs BD, Liu YC. Maintenance of erection of penile
glans but not penile body, after transection of rat cavernous
nerves. J Urol 1991; 146:900.
162. Wieder JA, Brackett NL, Lynne CM et al. Anesthetic
block of the dorsal penile nerve inhibits vibratory-induced
ejaculation in men with spinal cord injuries. Urology 2000;
55:915.
163. Aycock L. The medical management of premature
ejaculation. J Urol 1949; 62:361.
164. Damru F. Premature ejaculation: use of ethyl
aminibenzoate to prolong coitus. J Urol 1963; 89:936.
165. Berkovitch M, Keresteci AG, Koren G. Efcacy of
prilocainelidocaine cream in the treatment of premature
ejaculation. J Urol 1995; 154:1360.
166. Sahin H, Bircan MK. Re: Efcacy of prilocainelidocaine
cream in the treatment of premature ejaculation. J Urol 1996;
156:1783.
167. Atan A, Basar MM, Aydoganli L. Comparison of the
efcacy of fluoxetine alone vs. fluoxetine plus local lidocaine
ointment in the treatment of premature ejaculation. Arch Esp
Urol 2000; 53:856.
168. Xin ZC, Choi YD, Choi HK. The effects of SS-cream and
its individual components on rabbit corpus cavernosal
muscles. Yonsei Med J 1996; 37:312.
169. Xin ZC, Choi YD, Lee SH et al. Efcacy of a topical
agent SS-cream in the treatment of premature ejaculation:
preliminary clinical studies. Yonsei Med J 1997; 38:91.
170. Xin ZC, Choi Y, Seong DH. Sensory evoked potential
and effect of SS-cream in premature ejaculation. Yonsei Med
J 1995; 36:397.
171. Abrahams JI, Solish GI, Boorjian P et al. The surgical
correction of retrograde ejaculation. J Urol 1975; 114:888.
171a. DeBusk R, Drory Y, Goldstein I et al. Management of
sexual dysfunction in patients with cardiovascular disease:
recommendations of the Princeton Consensus Panel. Am J
Cardiology 2000; 86:175181.
172. Kedia K, Markland C. The effect of pharmacological
agents on ejaculation. J Urol 1975; 114:569.
173. Nijman JM, Jager S, Boer PW et al. The treatment of
ejaculation disorders after retroperitoneal lymph node
dissection. Cancer 1982; 50:29672971.
174. Fossa SD, Ous S, Abyholm T et al. Post-treatment
fertility in patients with testicular cancer. I. Influence of retro-
peritoneal lymph node dissection on ejaculatory potency.
Br J Urol 1985; 57:204.
175. Menendez Abraham E, Moran Viesca P, Velasco Plaza A
et al. Modications of the sexual activity in male rats follow-
ing administration of antiserotoninergic drugs. Behav Brain
Res 1988; 30:251.
176. Kimura Y, Kisaki N, Sakurada S et al. On the brain
monoaminergic systems relating to ejaculation. I. Brain
dopamine and ejaculation. Andrologia 1976; 8:313.
177. Kimura Y, Tadano T, Urano S et al. On suppression
of ejaculation by alpha-methyldopa. Andrologia 1984;
16:118.
178. Harrison W, Stewart J, McGrath PJ et al. Unusual side
effects of clomipramine associated with yawning. Can J
Psychiatry 1984; 29:546.
179. Wawrose FE, Sisto TM. Clomipramine and a case of
exhibitionism. Am J Psychiatry 1992; 149:843.
180. McCormick S, Olin J, Brotman AW. Reversal of
fluoxetine-induced anorgasmia by cyproheptadine in two
patients. J Clin Psychiatry 1990; 51:383.
181. Ashton K, Hamer R, Rosen RC. Serotonin reuptake
inhibitor-induced sexual dysfunction and its treatment: a
large-scale retrospective study of 596 psychiatric outpatients.
J Sex Marit Ther 1997; 23:165.
182. Feder R. Reversal of antidepressant activity of fluoxetine
by cyproheptadine in three patients. J Clin Psychiatry 1991;
52:163.
References 83
183. Lauerma H. Successful treatment of citalopram-induced
anorgasmia by cyproheptadine. Acta Psychiatr Scand 1996;
93 (1):6970.
184. Cohen A. Fluoxetine-induced yawning and anorgasmia
reversed by cyproheptadine treatment. J Clin Psychiatry
1992; 53:174.
185. Aizenberg D, Zemishlany Z,Weizman A. Cyproheptadine
treatment of sexual dysfunction induced by serotonin
reuptake inhibitors. Clin Neuropharmacol 1995; 18:320.
186. Ferraz MR, Santos R. Amantadine stimulates sexual
behavior in male rats. Pharmacol Biochem Behav 1995;
51:709.
187. Yells DP, Prendergast MA, Hendricks SE et al.
Monoaminergic influences on temporal patterning of sexual
behavior in male rats. Physiol Behav 1995; 58:847.
188. Balon R. Intermittent amantadine for fluoxetine-induced
anorgasmia. J Sex Marit Ther 1996; 22:290.
189. Shrivastava RK, Shrivastava S, Overweg N et al.
Amantadine in the treatment of sexual dysfunction
associated with selective serotonin reuptake inhibitors. J Clin
Psychopharmacol 1995; 15:83.
190. Balogh S, Hendricks S, Kang J. Treatment of fluoxetine-
induced anorgasmia with amantadine. J Clin Psychiatry
1992; 53:212.
191. Valevski A, Modai I, Zbarski E et al. Effect of amantadine
on sexual dysfunction in neuroleptic-treated male schizophrenic
patients. Clin Neuropharmacol 1998; 21 (6): 355357.
192. Gitlin MJ. Treatment of sexual side effects with
dopaminergic agents. J Clin Psychiatry 1995; 56:124.
193. Price J, Grunhaus L.Treatment of clomipramine-induced
anorgasmia with yohimbine: a case report. J Clin Psychiatry
1990; 51:32.
194. Jacobsen FM. Fluoxetine-induced sexual dysfunction
and an open trial of yohimbine. J Clin Psychiatry 1992;
53:119.
195. Hollander E, McCarley A. Yohimbine treatment of
sexual side effects induced by serotonin reuptake blockers.
J Clin Psychiatry 1992 53 (6):207209.
196. Witkin JM, Perez LA. Comparison of effects of
buspirone and gepirone with benzodiazepines and antagonists
of dopamine and serotonin receptors on punished behavior
of rats. Behav Pharmacol 1989; 3:247.
197. Othmer EOS. Effect of buspirone on sexual dysfunction
in patients with generalized anxiety disorder. J Clin Psychiatry
1987; 48:201.
198. Cooper BR, Hester TJ, Maxwell RA. Behavioral and
biochemical effects of the antidepressant bupropion
(Wellbutrin): evidence for selective blockade of dopamine
uptake in vivo. J Pharmacol Exp Ther 1980; 215:127.
199. Ashton AK, Rosen R. Bupropion as an antidote for
serotonin reuptake inhibitor-induced sexual dysfunction.
J Clin Psychiatry 1998; 59:112.
200. Napoli-Farris L, Fratta W, Gessa GL. Stimulation of
dopamine autoreceptors elicits premature ejaculation in
rats. Pharmacol Biochem Behav 1984; 20:69.
201. Kaplan JM, Hao JX, Sodersten P. Apomorphine induces
ejaculation in chronic decerebrate rats. Neurosci Lett 1991;
129:205.
202. Segraves RT, Bari M, Segraves K et al. Related articles,
links: effect of apomorphine on penile tumescence in men with
psychogenic impotence. J Urol 1991; 145 (6):11741175.
203. Heaton JP, Morales A., Adams MA et al. Recovery of
erectile function by the oral administration of apomorphine.
Urology 1995; 45:200.
204. Aizenberg D, Gur S, Zemishlany Z et al. Mianserin, a
5-HT2a/2c and alpha 2 antagonist, in the treatment of sexual
dysfunction induced by serotonin reuptake inhibitors. Clin
Neuropharmacol 1997; 20:210.
205. Foreman MM, Hall JL. Effects of D2-dopaminergic
receptor stimulation on male rat sexual behavior. J Neural
Transm 1987; 68:153.
206. Eaton RC, Markowski VP, Lumley LA et al. D2 receptors
in the paraventricular nucleus regulate genital responses and
copulation in male rats. Pharmacol Biochem Behav 1991;
39:177.
207. Bors E, Comarr AE. Neurological disturbances of sexual
function with special reference to 529 patients with spional
cord injury. Urol Surv 1960; 10:191.
208. Comarr AE. Sexual function among patients with spinal
cord injury. Urol Int 1970; 25:134.
209. Guttman L. Fourth International Neurology Conference
1949; 2:69.
210. Spira R.Artical insemination after intrathecal injection
of neostigmine in a paraplegic. Lancet 1956; 1:670.
211. Brindley G. Sexual and reproductive problems of
paraplegic men. Oxf Rev Reprod Biol 1986; 8:214.
212. Ohl DA, Bennett CJ, McCabe M et al. Predictors of
success in electroejaculation of spinal cord injured men.
J Urol 1989; 142:1483.
213. Brindley GS. The fertility of men with spinal injuries.
Paraplegia 1984; 22: 337.
214. Steinberger RE, Ohl DA, Bennett CJ et al. Nifedipine
pretreatment for autonomic dysreflexia during electro-
ejaculation. Urology 1990; 36:228.
215. Hovatta O, von Smitten K. Sperm aspiration from vas
deferens and in-vitro fertilization in cases of non-treatable
anejaculation. Hum Reprod 1993; 8:1689.
216. Apfelbaum B. The diagnosis and treatment of retarded
ejaculation. In: Leiblum SR, Pervin LA, eds. Principles and
practice of sex therapy. New York: Guilford Press;
1980:263296.
217. Martin L. Treatment of male sexual dysfunction with
sex therapy. In: Montague DK, ed. Disorders of male sexual
function. Chicago: Year Book Medical, 1988:142153.
84 Ejaculatory Dysfunction
INTRODUCTION
The testes produce spermatozoa and the androgenic
steroid hormones. Both processes are under complex
feedback control by the hypothalamicpituitary system.
Testicular androgen hormones are responsible for the
formation of the basic male phenotype during embryo-
genesis and regulate male sexual function.
Androgens are hormones that are based on the struc-
ture of testosterone, the major male sex hormone, and
are capable of developing and maintaining masculine
sexual characteristics, fertility, and the anabolic status
of somatic tissues. All androgens act through the single
androgen receptor and have similar biological effects.
Their effects in different target tissues are diversied
by metabolism of testosterone to its active metabolites
dihydrotestosterone (DHT) and estradiol.
ANATOMY OF THE TESTES
The testes lie loose within the cavity of the scrotum,
are capable of great mobility, and are suspended by the
spermatic cords. The left testis hangs somewhat lower
than the right (Fig. 6.1). In early fetal life the testes are
located intraperitoneally within the abdominal cavity.
Before birth they descend and enter, pass through and
emerge from the inguinal canal with the spermatic
cord at the external inguinal ring to enter the scrotum.
During their course coverings derived from the serous,
Androgens in Male Sexual Physiology
Chris G. McMahon
CHAPTER 6
Figure 6.1 Anatomy of the male genitals.
Testicular
artery
Testicular
vein
Inguinal ligament
Penis
Superficial
inguinal ring
Spermatic cord
Scrotal septum
Cremaster muscle
Superficial scrotal fascia
Dartos muscle
Scrotal
skin (cut)
Urinary bladder
Inguinal canal
Genitofemoral
nerve
Deferential artery
Ductus deferens
Pampiniform plexus
Testicular artery
Epididymis
Scrotal
cavity
Testis (covered by
visceral layer of
tunica vaginalis)
Raphe
Ureter
muscular, and brous layers of the abdominal parietes
invest them. The average dimensions of the testis are
45 cm in length, 2.5 cm in breadth, and 3 cm in the
anteroposterior diameter; its weight varies from 10.5
to 14 g. Each testis is of an oval form. Lying upon the
lateral edge of this posterior border is a long, narrow,
flattened body, named the epididymis.
The spermatic cord is composed of arteries, veins,
lymphatics, nerves, and the excretory duct of the testis
and is invested by the layers brought down by the
testis in its descent. The arteries of the cord are the
internal and external spermatic artery and the artery
to the ductus deferens. The spermatic veins emerge
from the back of the testis, receive tributaries from the
epididymis, and unite to form a convoluted plexus, the
pampiniform plexus. This forms the chief mass of the
spermatic cord and ascends, eventually uniting in a
single vein as it passes through the inguinal canal to
open into the inferior vena cava on the right side, and
the left renal vein on the left side. The lymphatic
vessels of the testes consist of two sets, supercial and
deep, the former commencing on the surface of the
tunica vaginalis, the latter in the epididymis and body
of the testis. They ascend with the spermatic veins in
the spermatic cord and end in the lateral and preaortic
groups of lumbar glands. The nerves are the spermatic
plexus from the sympathetic thoracolumbar outflow,
joined by laments from the pelvic plexus, which
accompany the artery of the ductus deferens.
The glandular structure of the testis consists of
numerous conical-shaped lobules, the apex being
directed toward the mediastinum (Fig. 6.2). Each lobule
consists of one to three, or more, minute convoluted
tubes, the seminiferous tubules. The total number of
tubules is estimated to be 840, and the average length
of each is 7080 cm. Each tubule consists of a base-
ment layer containing three different groups of epithe-
lial cells arranged in several irregular layers. Among
these cells may be seen the spermatozoa in different
stages of development as spermatogonia, spermatoblasts
or spermatids or spermatozoon, and the supporting
cells or Sertoli cells. In the apices of the lobules, the
tubules unite together to form 2030 larger ducts, the
tubuli recti. The tubuli recti enter the mediastinum to
form a close network of anastomosing tubes, the rete
testis. The rete testis eventually terminates in 1215 or
20 ducts, the efferent ductules, and ultimately in the
epididymis.
REGULATION OF TESTICULAR
FUNCTION
Androgenic steroids are synthesized in the Leydig
cells. Leydig cell function is regulated by the posterior
pituitary gonadotropin, luteinizing hormone (LH).
Spermatozoa are produced in the Sertoli cells of the
spermatogenic tubules. A second posterior pituitary
gonadotropin, follicle-stimulating hormone (FSH),
controls Sertoli cell function and spermatogenesis.
Gonadotropin regulation
The production of the pituitary gonadotropins, LH and
FSH, is regulated by hypothalamic gonadotropin-
releasing hormone (GnRH), or luteinizing hormone-
releasing hormone (LHRH). Plasma concentrations of
LH, FSH, and testosterone fluctuate in a pattern that
parallels the pulsatile release of GnRH (Fig. 6.3). This
pulsatile gonadotropin release is most apparent for
LH due to its short plasma half-life compared to FSH.
Pulses of LH secretion occur every 12 h in adult men.
The pulsatile secretion of testosterone in response to
LH release is dampened and less obvious due to con-
current stimulation of steroid synthesis and secretion
by the Leydig cell. Although testosterone secretion is
primarily regulated by pituitary LH, a FSH stimulated
Sertoli-cell-derived factor may enhance testosterone
production.
1
Testosterone regulates the hypothalamicpituitary
production of LH in a negative-feedback manner. By
decreasing the hypothalamic GnRH pulse frequency
and reducing pituitary sensitivity to GnRH, testos-
terone lowers LH release. Although testosterone is
86 Androgens in Male Sexual Physiology
Figure 6.2 Anatomy of the testis.
Head of
epididymis
Efferent
ductules
Rete
testis
Seminiferous
tubule
Scrotal
cavity
Tunica
albuginea
Spermatic
cord
Body of
epididymis
Ductus
deferens
Tail of
epididymis
Testis
converted to DHT and to estrogens in the pituitary,
pituitary gonadotropin secretion is primarily regulated
by testosterone. LH secretion is exquisitely sensitive
to testosterone negative feedback. Administration of
exogenous androgen that approximates the normal
daily secretory rate of testosterone (~20 mol or 6 mg)
results in almost complete suppression of LH secretion.
However, pituitary sensitivity to testosterone feedback
is reduced in chronic androgen deciency.
Pituitary FSH release is also regulated by the gonadal
peptide inhibins and activins.
2
Gonadal inhibins A and
B are heterodimeric proteins, produced by the Sertoli
cell.
3
They selectively suppress FSH without affecting
LH and provide feedback control of FSH production.
Activins, which are also produced by the pituitary,
are homodimeric proteins that selectively stimulate
FSH production through an autocrineparacrine
mechanism.
4
In summary, hormonal feedback signals from Leydig
cells and the spermatogenic tubules feedback and
precisely regulate the hypothalamicpituitary release
of LH/FSH and thereby their own function.
Synthesis of testosterone
About 95% of plasma testosterone in men is produced
by the Leydig cells of the testes from cholesterol and
the remainder is derived from conversion of adrenal
androgens, largely androstenedione. The biochemical
pathway by which the cholesterol is converted to
androgens and estrogens is depicted in Figure 6.4.
Cholesterol is synthesized either de novo in the Leydig
cell or derived from plasma lipoproteins, and is
converted to testosterone as the result of ve enzy-
matic reactions: (1) cholesterol side-chain cleavage
(CYP11A1); (2) 3-hydroxysteroid dehydrogenase/
isomerase 2 (3-HSD2); (3) 17-hydroxylase (CYP17);
(4) 17,20-lyase (CYP17); and (5) 17-hydroxysteroid
dehydrogenase 3 (17-HSD3). The rst four reactions
occur in the adrenal gland as well as the testis. The
presence of enzyme cofactors and posttranslational
modication (phosphorylation) of the enzyme confers
17,20-lyase activity, allowing androgen synthesis in
both the testis and zona reticularis of the adrenal
gland.
Delivery of cholesterol by the steroid acute
regulatory (StAR) protein to the inner mitochondrial
membrane for side-chain cleavage by CYP11A1 to
form pregnenolone is the rate-limiting step in testos-
terone synthesis. LH stimulates the activity of StAR
protein and the enzymes in the steroid pathway. Small
amounts of other steroid hormones including estradiol
are synthesized in the Leydig cell.
Transport of testosterone
The testes of a healthy young man secrete 1720 mol
(56 mg) of testosterone into the plasma each day.
Almost all testosterone is transported in plasma bound
to protein, largely to albumin and to a specic trans-
port protein, sex hormone-binding globulin (SHBG).
Approximately 12% of testosterone is unbound in the
plasma and circulates as free testosterone. The bound
and unbound fractions are in dynamic equilibrium.
5
Testosterone binds strongly to SHBG, whereas its
afnity for albumin is weak.
6
Testosterone rapidly
dissociates from albumin, adding to the fraction of
circulating testosterone available for entry into tissues
(bioavailable testosterone).
7
Whether non-SHBG-
bound testosterone is truly the fraction that is available
to all androgen target tissues is not clear, however.
Non-SHBG-bound testosterone does appear to be
available to the brain and the prostate.
8
However,
DHT, which is converted from testosterone via
Regulation of Testicular Function 87
Figure 6.3 Hypothalamic
pituitary regulation of
testosterone and
spermatogenesis.
Hypothalamus
Pituitary
Leydig cells
Testes
GnRH (+)
Sperm
Seminiferous
tubules
LH (+)
FSH (+)
T ()
Inhibitin
()
Activin
(+)
5-reductase locally in the androgen target tissue, is
the predominant androgen in most androgen target
organs, including the prostate. Furthermore, bone
mineral density measurements in older men correlate
signicantly better with free testosterone levels than
they do with total testosterone levels.
9
Metabolism of androgens
Testosterone is a precursor hormone which is converted
into two other hormones, DHT and estrogens. These
two hormones mediate most of the peripheral androgen
physiologic actions. DHT is synthesized by 5-reduction
of testosterone in androgen target tissues. It is respon-
sible for most of the differentiative, growth-promoting,
and functional aspects of male sexual differentiation
and virilization. Estrogens are principally synthesized
in adipose tissue and to a lesser extent in other extra-
glandular tissues, from circulating plasma testosterone
by the action of aromatase enzyme (CYP19).
10
Extra-
glandular estrogen formation increases with age and
with increased mass of adipose tissue. Estrone is
88 Androgens in Male Sexual Physiology
Figure 6.4 The biochemical pathway for the synthesis of testosterone.
Cholesterol
Pregnenolone
A
d
r
e
n
a
l
a
n
d
t
e
s
t
e
s
T
e
s
t
e
s
P
e
r
i
p
h
e
r
a
l
t
i
s
s
u
e
s
Progesterone
17OHProgesterone
17,20 lyase
CYP10 (aromatase)
CYP17
CYP17
CYP11A1
Cholesterol side-chain
cleavage enzyme
Androstendione
Testosterone
3-HSD3
(5-reductase)
3-HSD2
(17-hydroxysteroid
dehydrogenase)
17-hydroxylase
(3-hydroxysteroid
dehydrogenase/isomerase2)
Dihydrotestosterone Estradiol
HO
OH
HO
OH
HO
HC
HO
H
OH
CH
3
CH
3
CH
3
CH
2
CH CH
2
CH
2
produced from circulating precursors. Estradiol pro-
duction is derived directly from estrone (50%), from
circulating testosterone (35%), and from direct
synthesis in the testis (15%).
Testosterone and its active metabolites are
catabolized in the liver and excreted predominantly in
the urine as the 17-ketosteroids, androsterone and
etiocholanolone, or as the polar metabolites, diols,
triols, and conjugates. As opposed to testosterone,
estradiol secretion by the testes increases when
pituitary gonadotropin levels increase. Peripheral
androgen effects are the result of the combined actions
of testosterone and its active androgen and estrogen
metabolites. These androgen and estrogen metabolites
of testosterone exert local (paracrine) actions in the
tissues in which they are formed or enter the circu-
lation and act as hormones at other sites.
Actions of androgens
Androgens are responsible for the formation of the
male phenotype during sexual differentiation, regu-
lation of LH secretion, and induction of sexual
maturation at puberty. The cellular process by which
androgens perform these functions is schematized in
Figure 6.5. Testosterone passively diffuses into the
target tissue cell and is converted to DHT predomi-
nantly by steroid 5-reductase 2 and to a lesser extent
by steroid 5-reductase 1.Androgen-receptor proteins of
the cell nucleus bind testosterone or DHT. The formed
hormonereceptor complex binds to specic DNA
sequences and regulates messenger RNA transcription
to synthesize specic cellular proteins. The androgen
receptor, which is encoded by a gene on the long arm
of the X chromosome, contains 917 amino acids and
has a molecular mass of about 110 kDa. The poly-
morphic region in the amino terminus, which contains
a variable number of glutamine repeats, regulates the
activity of the receptor. The androgen receptor has
distinct hormone-binding, DNA-binding, and transcrip-
tional regulatory domains. Estradiol acts by a similar
mechanism on its own distinct estrogen receptors.
The testosteronereceptor complex regulates LH
secretion, spermatogenesis, and the virilization of the
wolfan ducts during sexual differentiation. The DHT
receptor complex is responsible for external virilization
during embryogenesis and for most androgen actions
during sexual maturation and adult sexual life. The
process by which two hormones can bind to the same
receptor but have different physiologic effects is
unknown. However, DHT appears to bind to the
receptor much more tightly than does testosterone,
which serves to amplify its hormonal signal.
Spermatogenesis
The normal adult testes produce in excess of 100
million sperm each day. Both androgen production by
the Leydig cells and pituitary release of FSH effect
spermatogenesis.The Sertoli cell, which regulates germ
cell proliferation and maturation in the seminiferous
tubules, is the major site of FSH action. FSH, whilst
not essential for spermatogenesis, increases the number
and maturation of sperm. Androgen, acting through
receptors in the seminiferous tubules, is essential for
spermatogenesis. The Sertoli cells are unable to syn-
Regulation of Testicular Function 89
Figure 6.5 The cellular mechanism of action of androgens. LH, luteinizing hormone.
Gonadotropin
regulation
Sexual
differentiation
Testis
Testosterone
Receptor
5-reductase
Dihydrotestosterone
Androgen target tissue
Estradiol
LH
Wolfian stimulation
External virilization
Sexual maturation
at puberty
thesize steroid hormones de novo but can convert
testosterone that diffuses from adjacent Leydig cells to
DHT and estradiol. Spermatogenesis is also regulated
by several paracrine and autocrine mechanisms
involving multiple cytokines and growth factors, some
of which are produced by the Sertoli cell itself.
EVALUATION OF TESTICULAR
FUNCTION
The diagnosis of androgen deciency involves the
recognition of appropriate clinical features of absent or
diminished androgenization, with conrmation by bio-
chemical testing.
11
The assessment of androgen status
should include specic enquiry about the reproductive
history, including the presence of developmental
abnormalities of the urogenital tract and the extent of
sexual maturation at puberty.
Reduced androgen action during embryogenesis
may result in hypospadias, cryptorchidism, or micro-
phallus. Sexual maturation will not occur with pre-
pubertal Leydig cell failure and the features termed
eunuchoidism will occur. These include an infantile
amount and distribution of body hair, poor develop-
ment of skeletal muscles, and delayed closure of the
epiphyses. Postpubertal Leydig cell failure is associated
with changes in sexual function, including hypoactive
sexual desire and erectile/ejaculatory dysfunction,
reductions in fertility, beard and body hair growth and
distribution, testicular volume, musculature, strength
and energy, and the presence of gynecomastia.
12
Evaluation of the testes is an essential part of the
physical examination. The prepubertal testis measures
about 2 cm in length and 2 ml in volume and grows
during puberty to reach the adult proportions by age
1617 years. Testes in adult men average 4.6 cm in
length (range 3.55.5 cm), corresponding to a volume
of 1225 ml. Approximately 60% of testicular mass is
contributed by the the seminiferous tubules. The testes
of prepubertal seminiferous tubular damage are small
and rm, whereas postpubertal damage is associated
with testes that are soft as the capsule, once enlarged,
does not contract to its previous size. Testicular size
remains unchanged as men age, even in advanced age.
The presence of a varicocele should be sought by
palpation with the patient standing.
The diagnosis of postpubertal androgen deciency
requires a high index of suspicion and appropriate
laboratory assessment because some androgen func-
tions regress very slowly and other functions may
remain unchanged.
Plasma testosterone levels
Plasma testosterone is measured by immunoassay. The
plasma testosterone level in normal adult men ranges
from 10 to 35 nmol/l (310 ng/ml). Testosterone
secretion is pulsatile and occurs every 6090 min in
response to pulsatile hypothalamicpituitary LH
secretion. Men with prolonged intervals between LH
pulses may have testosterone levels that transiently fall
below the normal range. Furthermore, plasma testos-
terone levels also vary throughout the day and at dif-
ferent times of the year, and may be as much as 30%
higher in the morning than in the evening. As such, a
single random testosterone sample is unreliable in the
diagnosis of androgen deciency. A pool of two or
three samples spaced 1520 min apart on at least two
separate days and preferably in the morning minimizes
the effects of random and laboratory fluctuations and
diurnal rhythms and provides a more accurate
assessment.
13
Measurements of free testosterone by immunoassay
may assist in the diagnosis of androgen deciency, but
require extensive validation. Indirect measurements
of free testosterone, such as the free androgen index
(testosterone/SHBG ratio), correspond poorly with
direct measurements and lack empirical validation as a
diagnostic test.
DHT can also be measured by immunoassay. In
young men the plasma DHT level is about one-tenth the
value for testosterone, averaging ~2 nmol/l (0.6 ng/ml).
In older men with benign prostatic hyperplasia (BPH),
plasma DHT levels average ~3 nmol/l (0.9 ng/ml).
Testicular function cannot be assessed by the measure-
ment of urinary 17-ketosteroids as most of the urinary
17-ketosteroids in men is derived from adrenal
androgens and only approximately 40% is derived
from the testes.
Plasma LH levels
Plasma LH is also measured by immunoassay. Because
LH secretion is pulsatile in fashion, assay of a pool of
two or three samples drawn 1520 min apart, as is the
case with testosterone, provides a more accurate assess-
ment. In early puberty, sleep-related nocturnal gonado-
tropin surges result in levels of plasma testosterone
and LH which are higher at night than during the day.
By the age of 17, the pulsatile LH secretion is of similar
magnitude during sleep and waking periods and
daytime levels of plasma testosterone have gradually
increased to reach adult levels. The presence of hypo-
gonadism can only be assessed by the simultaneous
interpretation of a low plasma testosterone level and
the plasma LH. A low plasma testosterone level
associated with an elevated LH level implies primary
testicular insufciency or primary hypogonadism
whereas a low plasma testosterone level and a reduced
LH level suggests hypothalamic or pituitary disease
or secondary hypogonadism (hypogonadotropic
hypogonadism).
90 Androgens in Male Sexual Physiology
Response to gonadotropin stimulation
Leydig cell capacity in prepubertal boys where both
LH and testosterone levels are low can be evaluated
by measuring the plasma testosterone response to
gonadotropin stimulation. Normal prepubertal boys
respond within 35 days of injection of human chorionic
gonadotropin (hCG) with an increase in the plasma
testosterone level to ~7 nmol/l (2 ng/ml). This response
increases with the start of puberty and peaks in early
puberty.
Response to GnRH
In prepubertal boys, acute administration of GnRH
produces a minimal increase in plasma LH and FSH as
the pituitary has not been primed by previous expo-
sure to GnRH or gonadal steroids. The LH response to
acute administration of GnRH increases after puberty,
while the FSH response is less robust.The extent of the
LH response to GnRH challenge reflects the amount of
LH stored in the pituitary. The extent of LH response
is broad but most men demonstrate a four- to vefold
increase in LH levels with the peak level at 30 min,
following either subcutaneous or intravenous GnRH
challenge.
The determination of testosterone and basal LH is
usually sufcient for the diagnosis of primary testicular
failure, and the assessment of GnRH response is rarely
required. Men with secondary hypogonadism due to
either pituitary failure or hypothalamic disease can
have either a normal or an abnormal LH response to
GnRH challenge.Although a normal response does not
clearly distinguish these causes of hypogonadism, a
suboptimal response indicates that an abnormality
exists but fails to characterize the site of the defect.
The presence of a normal LH response after pulsatile
or daily GnRH challenge for a week suggests that
hypothalamic disease is present.
SEMEN ANALYSIS
The ejaculate can be divided into several fractions by
serial biochemical analysis.
14
It comprises secretions
from the seminal vesicles, prostate and bulbourethral
(Cowpers) glands, and spermatozoa. It is produced
when the combination of the secretions of the prostate
and the contents of the ampullary parts of the vasa
deferentia is washed out by fluid from the seminal
vesicles and expelled from the urethra.
15
The sperma-
tozoa are stored in the tails of the epididymides and
the vas deferens ampullae. Approximately 5080% of
the entire ejaculatory volume is contributed by the
seminal vesicles, 1530% by the prostate gland, and a
small contribution is derived from the bulbourethral
(Cowpers) glands; this is rich in enzymes and plasmin-
ogen activator.
16
Spermatozoa normally constitute less
than 0.1% of the ejaculatory volume. The rst fraction
of the ejaculate contains the maximum number of
spermatozoa, and subsequent fractions contain
sequentially less. Acid phosphatase, citric acid, and
zinc, emanating from the prostate, are also in highest
concentration in the initial fractions of the ejaculate.
Subsequent fractions contain fructose from the seminal
vesicles, which increases in concentration towards the
end of the ejaculatory process. The pH of the ejaculate
increases in successive fractions as the acid component
provided by the prostate is serially mixed with the
more alkaline contribution of the fructose-rich fluid
from the seminal vesicles.
Seminal fluid, obtained by masturbation and after
ejaculatory abstention for 12 days on at least three
separate occasions, should be examined within an hour
of collection. The normal ejaculate volume is 26 ml.
Sperm density should exceed 20 million per ml. At
least 60% of the sperm should be motile and of normal
morphology. What denes an adequate sperm count is
unclear and some men with low sperm counts are
fertile. The confusion surrounding the lower limit of
normal sperm density, motility, and morphology
relates to the presence of multiple factors that produce
temporary aberrations in sperm count and the absence
of any true measure of sperm functional capacity in
routine semen analysis.
Plasma FSH levels
Plasma FSH is measured by immunoassay. When
damage to the germinal epithelium of the seminiferous
tubules is severe, plasma levels of Sertoli cell inhibin B
fall and plasma levels of FSH increase.
TESTICULAR BIOPSY
Open or percutaneous needle testicular biopsy is an
aid to diagnosis in some men with oligospermia and
azoospermia. The presence of a normal testicular
histology on biopsy and a normal FSH level in an
azoospermic man suggests obstruction of the vas
deferens. Sperm harvesting can be performed during
testicular biopsy for intracytoplasmic sperm injections
(ICSI) into oocytes.
17
PLASMA ESTROGEN LEVELS
Estrogens are principally synthesized in adipose tissue
and to a lesser extent in other extraglandular tissues,
from circulating plasma testosterone by the action of
aromatase enzyme. Extraglandular estrogen formation
increases with age and with increased mass of adipose
tissue.
18
Estrone is produced from circulating precursors.
Plasma Estrogen Levels 91
Estradiol production is derived directly from estrone
(50%), from circulating testosterone (35%), and
from direct synthesis in the testis (15%). The plasma
level of estradiol is usually <180 pmol/l (50 pg/ml) in
normal men whereas the plasma estrone level is some-
what higher but is usually <300 pmol/l (80 pg/ml).
Abnormally high estrogen production and elevated
plasma estrogen levels can be due to increased pro-
duction or reduced metabolism of plasma precursors
in liver failure or adrenal disease, to increased extra-
glandular aromatization seen in adipose tissue of
obese men, or to increased production by the testes
due to hormone-secreting testicular tumors, androgen
resistance, or gonadotropin stimulation.
PHASES OF NORMAL TESTICULAR
FUNCTION
Testicular production of testosterone in the male
embryo commences at about 78 weeks of gestation
and is partly stimulated by placental hCG. A high level
of plasma testosterone is maintained until late
gestation, at which stage the level falls and is only
slightly greater in males than in females at birth. The
plasma testosterone level in the male infant is
transiently raised for the rst 3 months of life due to a
short-lived increase in pituitary gonadotropins, but
again falls to low levels by age 6 months to 1 year and
remains low until puberty. After puberty, the plasma
testosterone level progressively increases to reach adult
levels by the age of 17 years. The level of bioavailable
testosterone declines at a rate of approximately 1%
per year after the age of 40 years. However, there is a
negligible decline in total testosterone until the later
decades of life, as the level of SHBG increases by
approximately 1% per year in parallel with the decline
in bioavailable testosterone.
19
ABNORMALITIES OF TESTICULAR
FUNCTION
Puberty
The hypothalamicpituitary system, the testes, or the
adrenals are probably responsible for the control of
puberty.
20
Before the onset of puberty, gonadotropin
secretion by the pituitary is low, but prepubertal
castration causes a rise in plasma gonadotropin levels.
The small amount of circulating plasma testosterone
present before puberty exerts sensitive negative-
feedback control of pituitary gonadotropin, resulting
in low secretion of gonadotropins. Sleep-related
nocturnal gonadotropin surges occur in early puberty
and result in levels of plasma testosterone and LH
which are higher at night than during the day. As
puberty proceeds, gonadotropin secretion increases
due to an increase in both GnRH secretion and pituitary
sensitivity to GnRH and the hypothalamicpituitary
system becomes less sensitive to negative-feedback
control. By the age of 17, the pulsatile LH secretion is
of similar magnitude during sleep and waking periods,
and daytime levels of plasma testosterone have
gradually increased to reach adult levels.
The onset, duration, and sequence of normal male
puberty are highly variable.
21
Testicular enlargement
forecasts puberty at the age of 1112 years. The rise in
plasma testosterone is responsible for the growth of
the testes, penis, the prostate, the seminal vesicles, and
the epididymides. Muscle and connective tissue growth,
particularly androgen-sensitive muscles of the pectoral
region and the shoulder, occurs. The pitch of the voice
lowers as the larynx and vocal cords enlarge. Growth
of the beard, regression of the scalp hair line, and the
development of truncal, limb, axillary, and pubic hair
occur as part of the typical hair growth of male
puberty.
Precocious puberty
Precocious puberty is sexual development before the
age of 9. It may be either complete with virilization and
premature activation of the hypothalamicpituitary
system with resultant spermatogenesis, or incomplete
with virilization only and hypothalamicpituitary
activity appropriate for age.
Virilizing syndromes are associated with plasma
testosterone which is inappropriately elevated for age
and can occur with Leydig cell tumors, Leydig cell
hyperplasia, hCG-secreting tumors, adrenal tumors,
congenital adrenal hyperplasia (most commonly 21-
hydroxylase deciency), or androgen administration.
Precocious puberty with asymmetric-sized testes
suggests the presence of a Leydig cell tumor. The local
androgen production seen in Leydig cell tumors may
result in limited localized spermatogenesis.
Virilizing adrenal tumors predominantly secrete
androstenedione and dehydroepiandrosterone, which
are partly converted to testosterone. Levels of 17-
hydroxyprogesterone and, as a result, androgen levels
are elevated in congenital adrenal hyperplasia.Although
both congenital adrenal hyperplasia and adrenal
tumors are associated with increased 17-ketosteroid
excretion, treatment with glucocorticoid administration
will promptly decrease the 17-ketosteroid excretion
of the former to normal but fails to normalize 17-
ketosteroid excretion in the latter.
Autonomous Leydig cell hyperplasia results in adult
levels of testosterone and virilization by the age of
2 years, and occurs either sporadically or can be
inherited as a male-limited autosomal disorder. LH
receptor mutations result in premature receptor acti-
vation in the absence of LH due to receptor mutations.
92 Androgens in Male Sexual Physiology
LH levels and the LH response to GnRH are
prepubertal.
Premature activation of the hypothalamicpituitary
system can be due to brain tumors, brain injury, or
infection but is most often idiopathic. It is associated
with the normal physical, hormonal, and biochemical
features of puberty, including sleep-related nocturnal
gonadotropin surges, elevated plasma LH levels, and
enhanced LH response to GnRH.
Management of sexual precocity is etiology-specic.
The elevated plasma testosterone levels seen in Leydig
cell hyperplasia can be partly lowered by treatment with
either medroxyprogesterone acetate or ketoconazole,
or its androgen action partly reduced with spirono-
lactone.The gonadotropin production and testosterone
synthesis of idiopathic precocious puberty and
precocious puberty due to inoperable central nervous
systems lesions is best treated with long-acting GnRH
analog therapy.
Delayed or incomplete puberty
The onset, duration, and sequence of normal male
puberty are highly variable and distinguishing delayed
or failed puberty from variants of normal develop-
ment can be problematic. Separating failure of puberty
from variants of normal development is one of the
most difcult problems in endocrinology. Most boys
fail to demonstrate a normal spurt of growth and sexual
development at 1213 years of age but a small number
of boys may not commence puberty until 16 years of
age or older, continue slow pubertal development until
age 2022, and may have a family history of delayed
puberty.
Pubertal failure is seen in prepubertal hypo-
thyroidism and panhypopituitarism. Absent puberty in
the presence of low plasma testosterone levels and
elevated FSH and LH levels suggests primary testicular
disease. Hereditary androgen resistance, in which both
plasma testosterone and LH levels are high, often
causes male pseudohermaphroditism but in a mild
form may present as absent or incomplete puberty.
The majority of boys with absent puberty have low
plasma levels of testosterone and gonadotropins and
must be distinguished from idiopathic hypogonado-
trophic hypogonadism (Kallmann syndrome) or
isolated gonadotropin deciency. Cryptorchidism and
microphallus are features of hypogonadotrophic hypo-
gonadism. Anosmia or hyposmia is common in hypo-
gonadotropic hypogonadism and is due to abnormal
development of the olfactory tracts which share
progenitor cells with GnRH neurons.
22
Several distinct
abnormalities of GnRH formation or action are
involved in the pathogenesis of hypogonadotropic
hypogonadism. An X-linked recessive trait associated
with defects in a neural cell adhesion molecule (KAL)
involved in the migration of GnRH neurons into the
olfactory bulb may be inherited. An autosomal domi-
nant trait with variable expression or rare autosomal
recessive traits may impair the GnRH receptor. The
plasma testosterone level is low for age, plasma FSH
and LH levels are usually below the normal male
range, and the secretion of other pituitary hormones is
normal. In patients with GnRH deciency, endocrine
abnormalities are corrected and spermatogenesis is
initiated following pulsed GnRH treatment.
Hypogonadotropic hypogonadism, due to decient
GnRH production and abnormal gonadotropin func-
tion, and primary adrenal insufciency are features of
X-linked adrenal hypoplasia congenita. The extent of
LH and/or FSH deciency varies in isolated gonado-
tropin deciency and clinical features can range from
eunuchoidal features and prepubertal-sized testes to
a degree of testicular enlargement and pubertal
development in boys with only partial LH and/or FSH
deciency.
Adult males
Androgen production and spermatogenesis can be
influenced by a variety of factors at testicular and
hypothalamicpituitary levels.
23,24
An increase in scrotal
temperature due to a varicocele, a transient increase
in either core or peripheral temperature, as might
occur in a hot bath, diet, drugs, alcohol, environmental
agents, and psychological stress may be associated
with a temporary decrease in spermatogenesis.
Hypothalamicpituitary disorders, testicular defects,
or abnormalities of sperm transport may affect Leydig
cell function or spermatogenesis jointly or selectively,
and lead to chronic abnormalities of testicular func-
tion. Most often, androgen production and fertility are
jointly affected due to dependence of spermatogenesis
on androgens.
HYPOTHALAMICPITUITARY
DISORDERS
Impaired gonadotropin secretion can occur in either
generalized anterior pituitary disease or as an isolated
defect. In hypogonadotropic hypogonadism, secretion
of both LH and FSH is impaired. Gonadotropin secre-
tion can also be affected by several external factors.
The elevated plasma cortisol of Cushings disease can
suppress LH secretion. Patients with untreated
congenital adrenal hyperplasia have elevated levels of
adrenal androgens, suppressed gonadotropin secretion,
and consequent infertility. Critical illness also sup-
presses plasma gonadotropin levels. Androgen misuse
or abuse in eugonadal men can inhibit gonadotropin
secretion and impair spermatogenesis. Hyperpro-
lactinemia due to a pituitary adenoma or as an adverse
Hypothalamicpituitary Disorders 93
effect of drugs such as spironolactone or phenothiazines
can inhibit LH and FSH secretion and cause combined
Leydig cell and seminiferous tubule dysfunction.
Elevated prolactin can occasionally be associated with
normal gonadotropin and androgen levels but still
impair fertility by a direct action of spermatogenesis.
Hemochromatosis usually impairs testicular function
as the result of effects on the pituitary; less often it
affects the testis directly. Morbid obesity is associated
with reduced levels of SHBG and of total and bio-
available testosterone but normal LH levels, which
return toward normal with weight loss. Obesity may
also contribute to the decreased testosterone levels in
the subset of such men with Pickwickian syndrome.
TESTICULAR DISORDERS
Developmental defects
The diagnosis of Klinefelter syndrome (XXY) and the
XX male syndrome is not usually made until the
recognition that puberty is delayed. Varicocele, crypt-
orchidism, germinal cell aplasia, and deletions or muta-
tions of the azoospermia factor (AZF) genes on the Y
chromosome may cause infertility in the presence of
normal androgen production.
2527
Varicocele occurs in approximately 10% of men, most
often on the left side (90%), and may be the causative
factor in as many as 30% of couples with infertility.
A varicocele is due to dilation of the pampiniform
plexus of veins that surrounds the testis and is caused
by incompetence of the venous valve between the
internal spermatic vein and the renal vein, allowing
retrograde flow of blood into the internal spermatic
vein. Varicocele occurs in ~1015% of men in the
general population and in 2040% of men with
infertility. The testes are normally 12C cooler than
the core body temperature. A varicocele increases the
blood flow and consequently the temperature of both
testes as a result of the extensive anastomoses of the
venous systems and may impair spermatogenesis, non-
specically affecting all sperm parameters. Open ligation
or percutaneous transfemoral balloon embolization of
the internal spermatic vein restores fertility in 50%
of men whose preoperative sperm counts exceed 10
million per milliliter.
Unilateral cryptorchidism, even when corrected
before puberty, is often associated with impaired
spermatogenesis, demonstrating that both testes can be
abnormal in unilateral cryptorchidism. Some patients
with germinal cell aplasia have absent germinal
epithelium and resultant azoospermia, elevated FSH
levels, but normal plasma testosterone and LH levels.
28
Kartagener syndrome is an autosomal recessive defect
associated with situs inversus, chronic sinusitis, and
bronchiectasis due to absent or impaired motility of
the cilia of the airways and of the sperm. Electron
microscopic studies demonstrate structural defects in
the dynein arms, spokes, or microtubule doublets of the
cilia of the epithelia of the airways and of the sperm.
Acquired testicular disorders
Viral orchitis caused by the mumps virus, echovirus,
lymphocytic choriomeningitis virus, and group B
arboviruses is a common cause of testicular failure.
Mumps orchitis occurs in approximately 2025% of
mumps, is usually unilateral but may be bilateral
(3035%), and develops 23 days after the onset of
parotitis. The testes are swollen and painful and will
either return to normal size within 710 days or
undergo atrophy over the next 26 months. Unilateral
atrophy occurs in approximately 30% of cases and
bilateral atrophy occurs in about 1015%. Atrophy is
thought to be due to ischemia secondary to pressure
and edema within the tunica albuginea and to the
direct effects of the virus on the seminiferous tubules.
Normal seminiferous tubular function is restored in
70% of men with unilateral mumps orchitis but in only
30% of men with bilateral orchitis.
Testicular torsion and direct, severe testicular
trauma in contact sports and in men with hazardous
occupations can cause testicular atrophy. Testicular
irradiation reduces testicular blood flow and secretion
of testosterone, and spermatogenesis in a dose-
dependent fashion. Plasma levels of FSH and LH are
increased and azoospermia may develop as the ger-
minal epithelium is progressively destroyed. Recovery
of sperm density is also dose-related, may require as
long as 45 years, and may be incomplete despite
shielding of the testes. Cryopreservation and banking
of sperm are vital in men who wish to father children
prior to further radiotherapy or chemotherapy.Although
direct testicular radiation therapy for boys with acute
lymphoblastic leukemia causes permanently low
plasma testosterone levels, lifelong androgen deciency
is uncommon in adult men after radiotherapy.
Testicular function can be impaired by prescribed or
recreational drugs by a variety of mechanisms. Testos-
terone synthesis can be inhibited by spironolactone
and ketoconazole, which interferes with the nal steps
in androgen synthesis. Spironolactone and cimetidine
also block androgen target organ actions by compe-
titive inhibition of androgen receptor site binding.
The cirrhosis of long-term alcohol abuse is associated
with feminization, including testicular atrophy and
gynecomastia due to impaired hepatic catabolism of
estrogens. Long-term alcohol abuse also decreases
plasma testosterone levels in the absence of liver
disease. Marijuana, heroin, or methadone abuse may
be associated with low plasma testosterone and elev-
ated estradiol levels through unknown mechanisms.
94 Androgens in Male Sexual Physiology
Treatment with digitalis may occasionally cause
increased plasma estradiol and decreased plasma
testosterone levels.
Chemotherapeutic agents often impair spermato-
genesis. Cyclophosphamide often causes azoospermia
within a few weeks of starting therapy and return of
spermatogenesis occurs in 50% of men within 3 years
of ceasing chemotherapy. Leydig cell function and
androgen synthesis may also be damaged with com-
bination chemotherapy containing alkylating agents.
This is manifested by reduced plasma testosterone and
elevated LH levels in prepubertal boys, and normal
plasma testosterone levels but an enhanced LH
response to GnRH in adult men.
Occupational and recreational exposure to chemicals
or agents toxic to spermatogenesis such as lead, cad-
mium, microwaves, or ultrasound may cause infertility.
Increased exposure to environmental antiandrogens
or estrogens may be responsible for the 40% decline
in sperm density observed over the past 50 years.
Testicular abnormalities associated with
systemic disease
Androgen synthesis and spermatogenesis are decreased
in chronic renal failure despite the high plasma LH
level caused by both increased production and reduced
clearance but does not restore normal testosterone
production. Furthermore, hyperprolactinemia is present
in 30% of men with renal failure and decreases testos-
terone production. Gynecomastia, erectile dysfunction
(ED), and hypoactive sexual desire due to androgen
deciency and increased plasma estrogen levels are
common in chronic renal failure. Successful transplan-
tation and, to a much lesser extent, chronic hemo-
dialysis may return testicular function to normal.
In cirrhosis of the liver, the presence of an elevated
LH level inappropriately lower than that expected for
the degree of androgen deciency probably results
from inhibition of LH secretion by elevated estrogens.
Increased estrogen production results from impaired
hepatic extraction of adrenal androstenedione and
subsequent increased extraglandular conversion by
aromatization to estrone and estradiol. ED, testicular
atrophy, and gynecomastia are common in men with
cirrhosis and successful liver transplantation reverses
these effects of cirrhosis. Advanced metastatic cancer
and Hodgkins disease are often associated with
reduced Leydig cell function characterized by lowered
plasma testosterone levels and normal to increased
plasma LH levels, and reduced spermatogenesis.
Similar non-specic hormone changes occur after
surgery, acute myocardial infarction, and severe burns.
In men with a human immunodeciency virus (HIV)-
related illness, 3550% of men eventually develop low
testosterone levels.
29
Gonadotropins may initially be
elevated as part of a compensated state of hypo-
gonadism prior to the development of overt androgen
deciency. Many of the hormonal changes seen in HIV-
related illness are non-specic and related to severe
illness. Androgen replacement therapy (ART) may
increase muscle and lean body mass.
30
Neurologic diseases associated with altered testicular
function include myotonic dystrophy, spinobulbar
muscular atrophy, and paraplegia. In myotonic
dystrophy, small testes may be associated with both
androgen deciency, due to impaired Leydig cell func-
tion, and impaired spermatogenesis. The function of
the androgen receptor is impaired in spinobulbar
muscular atrophy and reduced androgenization, infer-
tility, and the hormonal features of androgen resistance
are late manifestations. Spinal cord injury may lead
to a temporary decrease in testosterone levels and
persistent defects in spermatogenesis.
Androgen resistance
Androgen receptor defects result in resistance to the
action of androgen in target organs and may be
associated with a spectrum of symptoms, ranging from
oligo- or azoospermia with mild resistance, to a
defective male phenotypic development, infertility,
and under androgenization with more severe degrees
of androgen resistance.
Treatment
Oral testosterone preparations have relatively poor
bioavailability. Relatively small amounts reach the
systemic circulation as it is rapidly absorbed into the
portal blood and promptly degraded by the liver.
Parenteral testosterone, unless modied to retard
absorption or degradation, is also rapidly degraded.
Effective ART requires either a slowly absorbed form
such as a transdermal patch system or micronized
oral testosterone, or modied analogs which retard
absorption or degradation, or enhance the androgenic
potency.
Three types of modication have had widespread
clinical application: (1) esterication of the 17-hydroxyl
group; (2) alkylation at the 17 position; and (3) alter-
ation of the ring structure, particularly by substitutions
at the 2, 9, and 11 positions. Most pharmacologic
agents actually have combinations of ring structure
alterations and either 17-alkylation or esterication of
the 17-hydroxyl group.
Esterication of the 17-hydroxyl group decreases
the polarity of the molecule, rendering the steroid
more soluble in the fat injection vehicles and leading to
slower release into the circulation after intramuscular
injection. Absorption is more retarded and drug action
prolonged, if larger acids are esteried. Esters such as
Testicular Disorders 95
testosterone cypionate and testosterone enanthate can
be injected intramuscularly every 2 weeks.
31
Most orally active androgens feature 17-methyl or
-ethyl alkylation which retards hepatic catabolism and
allows the alkylated derivatives to reach the systemic
circulation. Unfortunately, all 17-alkylated steroids
can cause abnormal liver function, and have a limited
role in therapy. Other alterations of the ring structure
have been adopted empirically; some slow the rate of
inactivation, others enhance the potency of a given
molecule, and some alter the conversion to other
active metabolites. For example, the potency of
fluoxymesterone may be due to the fact that, unlike
most androgens, it is a poor precursor for conversion
to estrogens in peripheral tissues.
Three transdermal preparations are available in
which a testosterone-loaded patch is applied to the
skin each day and provides physiologic testosterone
levels that mimic the normal diurnal variation with
higher levels in the morning hours.
3234
One is applied
to a shaved scrotum (Testoderm) and has poor patient
acceptance. The other two systems, Androderm and
Testosterone TTS, contain proprietary skin absorption
enhancers and are applied to the trunk, arms, or
thighs. Androderm is associated with a high incidence
of skin irritation.
Adverse effects of androgens
Adverse effects of physiological androgen doses are
uncommon in adult men. All androgens can induce
virilization in women and children and use in these
settings requires expert management. Acne, coarsening
of the voice, hirsutism, weight gain, gynecomastia,
male-pattern hair loss, and menstrual irregularities
may occur and will slowly resolve once treatment is
discontinued. Gynecomastia is common in children
receiving androgens, due to a greater capacity to con-
vert androgens (aromatized) in extraglandular tissues
to estradiol in childhood. The administration of
testosterone esters to men results in an increase in
plasma estrogen levels, but in men with normal liver
function. Although chronically ART in adult men with
testosterone esters results in high plasma estrogen
levels, gynecomastia only develops in adult men with
abnormal liver function or following treatment with
high doses.With the supraphysiologic doses of androgen
misuse or abuse, gonadotropin secretion is inhibited,
the testes atrophy, and spermatogenesis diminishes,
often for up to 12 months after cessation of the
androgen.
In older men, ART with testosterone ester injections
may cause polycythemia.
35
Symptoms of obstructive
sleep apnea may occur in predisposed men or worsen
in men receiving ART.
3638
Minor sodium retention is
common with ART and may result in edema in men
with controlled or insipient cardiac failure, or when
androgens are administered in large supraphysiological
doses.
The transient supraphysiological peak plasma
testosterone levels of testosterone esters or the high
rst-pass portal testosterone concentrations of oral
testosterone undecanoate increase hepatic testosterone
exposure, and may be associated with reduced levels
of SHBG, high-density lipoprotein cholesterol, and
other hepatic proteins.
39
This is rarely seen with trans-
dermal patches or implants where the plasma testos-
terone level is relatively constant.
All 17-alkylated synthetic androgens (oxandrolone,
fluoxymesterone, danazol) are hepatic and can cause
cholestatic hepatitis, peliosis hepatis, and hepatic
tumors with abnormal liver function tests, including
elevated plasma levels of alkaline phosphatase and
conjugated bilirubin. Although preexisting chronic liver
disease increases the risk of androgen hepatotoxicity,
jaundice may occur in the absence of men with normal
hepatic function. Peliosis hepatis (blood-lled cysts in
the liver) and hepatoma are the most serious compli-
cations of androgens and may either follow a benign
course after cessation of androgens or be rapidly fatal.
Other synthetic androgens, such as 19-nortestosterone
derivatives (nandrolone) and the 1-methyl androgens
(mesterolone, methenolone), are not hepatotoxic.
ART is contraindicated in men with prostate or
breast cancer. Prostate cancer should be excluded in
all men over the age of 40 prior to initiating ART, with
a digital rectal examination and determination of
prostate-specic antigen. In men with an elevated
prostate-specic antigen level, further investigation
with transurethral ultrasonography (TRUS) and biopsy
of the prostate should be performed prior to initiation
of ART. ART should be used cautiously in older men
with symptoms of BPH as it may precipitate urinary
obstruction. These men should also be warned about
the possibility of experiencing unfamiliar increases in
sexual desire. Precautions should also be exercised in
the use of ART in men with bleeding disorders,
androgen-sensitive epilepsy, migraine, sleep apnea or
polycythemia, and in men with cardiac or renal failure
or severe hypertension susceptible to fluid overload
from sodium and fluid retention.
40
Screening for
cardiovascular and prostate disease in men receiving
ART need be no more intensive than for age-matched
men not receiving ART.
41
Androgen replacement therapy
ART is indicated in androgen deciency of any cause
to restore normal male secondary sexual characteristics
and male sexual behavior and to mimic the anabolic
hormonal effects on muscle and bone mass, bone
marrow, nitrogen retention, and other androgen-
responsive tissues. Response to ART in hypogonadal
96 Androgens in Male Sexual Physiology
men is almost always successful.
42
All the clinical
features of androgen deciency, except decreased
spermatogenesis, usually respond within 12 months
of starting ART, although the full effect may take
longer. However, suppression of plasma LH to the
normal range may not occur for many weeks in men
with primary, long-standing hypogonadism. ART, after
puberty, is lifelong as the underlying disorders are
almost always permanent.
Deep intramuscular injections of a long-acting ester
such as testosterone enanthate or mixed testosterone
esters as 250 mg in 1 ml oil at 14-day intervals are
usually given into the upper and outer quadrant of the
buttock, resulting in a sustained increase in plasma
testosterone to the normal male range.
43
Alternatively,
transdermal testosterone patchs, which are available
in different doses and are replaced daily, can be used.
Testosterone and its esters should be used in
preference to 17-alkylated synthetic androgens and
19-nortestosterone derivatives, because of their
established safety and efcacy and ease of dose titration
and monitoring of testosterone levels. ART should
start with injections, but the proven patient preference
for the stable testosterone levels and smoother clinical
effects provided by implants or transdermal formu-
lations as opposed to the wide fluctuations in testos-
terone levels and symptoms during intramuscular
testosterone ester injections offers superior compliance
for long-term treatment.
44,45
Previously untreated elderly men may require lower
starting doses of ART. Poor compliance with dosing
frequency may be associated with more, higher peaks
and lower trough blood testosterone levels, and
exaggerated fluctuations of symptoms. An inadequate
response to ART, indicated by absent or poor sympto-
matic improvement and low trough testosterone levels
with or without persistently elevated LH levels in
primary hypogonadism, may indicate a need for 10-day
injection intervals. Increased dosage of ART is rarely
required, with the exception of men with mild andro-
gen resistance due to androgen-receptor mutations.
Persistently inadequate clinical responses suggest that
recalcitrant symptoms are not due to androgen
deciency.
In prepubertal hypogonadism, commencement of
low-dose ART at the time of puberty (age 1214 years),
followed by gentle dose escalation, results in a normal
progression of the events of puberty. ART should be
designed to replicate normal development and not
shorten the process. If ART is delayed until after the
expected time of puberty, the extent of virilization is
variable but the majority of patients achieve relatively
complete anatomic and functional sexual maturation.
Prepubertal hypogonadal boys with microphallus
require intermittent low-dose androgen therapy to
achieve normal external genitalia, and rarely experience
premature epiphyseal closure and shortened stature.
Although the time frame for the appearance of second-
ary sex characteristics is variable, penile development,
deepening of the voice, and the appearance of other
secondary sexual characteristics usually commence
during the rst year of treatment.
Adequacy of ART can be monitored by clinical
observation for resolution of the major symptoms of
androgen deciency and for improved clinical well-
being, combined with a limited number of hormonal
assays. Restoration of adequate potency and sexual
desire has a low threshold for androgen action and
does not indicate adequate androgen replacement.
The achievement of trough blood testosterone levels
within the normal range can be a valuable guide to the
adequacy of parenteral androgen replacement but is of
limited use in monitoring with oral ART. Suppression
of blood LH levels into the eugonadal reference range
indicates adequate ART in men with primary hypo-
gonadism. ART is either inadequate or the patient is
non-compliant if elevation of LH persists after 46
months of ART. In contrast to this, blood LH levels are
of no use in monitoring the response to ART in hypo-
gonadotropic hypogonadism.
The adequacy of long-term androgen effects on
bone mass can be evaluated by baseline and annual
bone densitometry of vertebral trabecular bone with
dual-photon absorptiometry.
Use of androgens in eugonadal men
The use of androgens has been touted as a treatment
for manifold disorders in eugonadal men. In some of
these disorders, the use of androgen therapy has been
rationalized on the basis that potential non-virilizing
benets, including improved nitrogen retention, muscle
mass, and hemoglobin, might outweigh any deleterious
actions. The proposed benets include improved
nitrogen balance in catabolic states, such as HIV-related
illness, enhanced erythropoiesis in the anemia of renal
failure and other refractory anemias, the treatment of
endometriosis, improved growth in growth retardation
of various etiologies, and self-administration by
athletes to increase muscle mass and consequently
performance.
46,47
These benets are rarely achieved, as
negligible physiologic effects occur in eugonadal men
with normal testicular androgen production unless
extremely high androgen doses are used. Furthermore,
as all androgens act upon a single receptor, the
prospect of the development of a synthetic androgen
that produces only the anabolic and not the virilizing
effects seems remote.
Fewer than 2% of men with ED have an androgen
deciency. In men with ED, androgen deciency should
be excluded with screening plasma testosterone levels
Testicular Disorders 97
and androgens should only be administered to men
with a proven androgen deciency.
At this stage, there is no convincing evidence that
the modest midlife decreases in plasma testosterone
levels have any clinical importance. This progressive
decline in androgen production with aging has been
inappropriately called the male menopause or
andropause but clearly differs from female meno-
pause, when ovarian failure effectively ends reproduc-
tive capacity.These terms are confusing and misleading
and have no basis in evidence-based medicine.
Androgen abuse by male and female athletes is now
epidemic, in the expectation that muscle development
and athletic performance will be improved.
48
In
controlled studies, androgens do improve nitrogen
balance and muscle mass.
49
The androgen doses
frequently taken by athletes may exceed 10 times the
replacement dose, and there are no data to conrm
that athletic performance is enhanced and that increased
muscle mass is sustained.
50
The risks associated with
multiple side-effects of high-dose androgen abuse,
particularly marked virilization in women, far outweigh
the presumed benets.
51
The prescription of androgens
with the sole purpose of enhancing the performance of
elite athletes is regarded as a breach of professional
standards by medical boards in most countries.
TESTOSTERONE SUPPLEMENTATION
IN THE AGING MALE
There is a large amount of clinical evidence to support
the role of hormone replacement therapy (HRT) in
postmenopausal women. Over the past two decades,
the concept of the male menopause or andropause
and the potential role for male HRT in reversing some
aspects of aging and prolonging the quality of life in
older men, by preventing or reducing a variety of age-
related degenerative symptoms, has generated increasing
interest in both physicians and special-interest patient
groups. The major androgen target organs of interest
with regard to the benecial effects of male HRT
include bone, muscle, adipose tissue, the cardiovascular
system, and the central nervous system (libido and
aspects of mood). Serum testosterone levels decline
slowly with normal aging in men and, although all men
are not destined to become hypogonadal as they age,
the prevalence of androgen deciency in the older
male is signicant.
ANDROGEN LEVELS IN THE AGING
MALE
Most recent studies have demonstrated a progressive
decline in plasma levels of total testosterone, free
testosterone and bioavailable testosterone, and an
increase in SHBG plasma levels as men age.
5255
Serum
levels of DHT do not alter as men age. The increase in
SHBG results in a greater rate of decline in serum
levels of bioavailable testosterone than that seen for
total testosterone.
56,57
The circadian rhythm in plasma levels of testos-
terone seen in young men, with peak levels occurring
in the morning, attenuates in older men.
58
Testosterone
declines with age due to decreased production. Leydig
cell mass and the activity of the enzymes in the testos-
terone production pathway reduce as men age.
59,60
The
increase in testosterone production in response to
increased gonadotropin stimulation is also attenuated
in older men.
61
An age-related decline in hypothalamic
pituitary function may also contribute to the decline in
testosterone production. Older men fail to demonstrate
an appropriate increase in LH secretion in response to
a hypoandrogenic state, and many older men with low
testosterone levels have normal LH levels, resulting in
a relative hypogonadotropic hypogonadism.
The rate of testosterone decline among individual
men can vary as men age. There is a wide variation in
the reported prevalence of hypogonadism in older
men in published studies (Table 6.1). This relates to a
general lack of agreement on the criteria for dening
hypogonadism in older men. Well-designed clinical
research involving male HRT is limited and is at least
20 years behind that for HRT in the postmenopausal
female. Most of the current ART in studies of aging
98 Androgens in Male Sexual Physiology
TABLE 6.1 Prevalence of androgen deciency in aging men.
Study Age range n Plasma total % of older men
(years) testosterone (ng/ml) in study
Vermeulen and Kaufman (1996)
95
2000 300 <32 22% (6080 years)
36% (80100 years)
Lunglmayr (1997)
96
5087 817 <30 11.4%
Morley et al. (1997)
55
75101 77 <25 33%
Smith et al. (2000)
97
4069 1660 <32 20.4% (4079 years)
males have enrolled men with testosterone levels either
at or below the lower limit of the normal range for
young adult men and have assessed the responsiveness
of androgen target organ responses to ART.
ANDROGEN TARGET ORGAN
CHANGES IN THE AGING MALE
Age-related degenerative organ changes that occur in
the aging male include a decline in bone mass and an
increased incidence of osteoporosis and minimal-
trauma fractures, a decrease in muscle tissue mass and
a decline in muscle strength, an increase in adipose
mass, particularly in intraabdominal fat, a decline in
sexual desire and erectile function, and a decreased
sense of well-being. All of these changes, with the
exception of increased adipose mass and ED, can be
improved with ART in the younger adult hypogonadal
male.
POTENTIAL BENEFITS OF MALE
HORMONE REPLACEMENT THERAPY
The potential benets and risks of male HRT in the
aging male are listed in Table 6.2.
Bone
The effects of androgen therapy on bone mineral den-
sity or other biochemical parameters of bone turnover
has been reported by several authors in studies lasting
318 months.
6266
Some of these studies included
osteoporotic older men and evaluated androgen
therapy in men undergoing chronic treatment with
glucocorticoids. The majority of these studies demon-
strated that androgen therapy increased bone mineral
density and slowed down the rate of bone degradation.
However, at this stage there are no data as to whether
the increased bone density is maintained in the long
term, the optimal level of testosterone therapy required
to improve bone mass, and whether testosterone
therapy lowers the rate of osteoporotic fractures in
older men.
Muscle mass and strength
The effects of testosterone therapy in older men on
body composition changes and muscle strength have
been evaluated in several published trials.
61,64,6770
Results from these studies are inconsistent and difcult
to interpret (Table 6.3). A decline in body fat was
reported by Marin et al.
67
and Katznelson et al.,
65
but
other authors reported no change. Most studies demon-
strated an increase in lean body mass and muscle
strength.The magnitude of the decline in body fat mass
was similar to that seen with testosterone replacement
in young hypogonadal men. Improvements in lean
body mass and muscle strength were less pronounced,
suggesting that the muscles of older men may be less
responsive to the anabolic effects of testosterone than
are the muscles of young men.Although muscle strength
was improved in most studies, grip strength was the
primary strength measure. One study assessed the
effects of androgen therapy on mobility and function
and demonstrated reductions in stair-climbing and
distance walk times. One study assessed the effects
of androgen therapy on mobility and function and
demonstrated reductions in stair-climbing and distance
walk times.
71
Sexual function and mood
The effect of testosterone therapy on sexual function
in healthy eugonadal older men has not been reported.
The effects of androgen therapy in eugonadal men
with sexual dysfunction has been reported by several
TABLE 6.2 Potential benets and risks of male hormone replacement therapy in the aging male.
Benets Risks
Increased bone mass, reduced osteoporosis and Increased risk of prostate cancer and benign
prevent fractures prostatic hyperplasia
Increased muscle strength Development of gynecomastia
Increased physical capacity Precipitate or worsen sleep apnea
Increased muscle mass Development of polycythemia
Improved libido and sexual function Fluid overload
Improved well-being and mood Increased incidence of cardiovascular disease
Decreased incidence of cardiovascular disease
Potential Benefits of Male Hormone Replacement Therapy 99
authors. ED is seldom improved but men with hypo-
active sexual desire often report improvement.
7275
Several blinded, placebo-controlled testosterone
replacement studies in older men have evaluated effects
on sense of well-being or other aspects of mood.Testos-
terone therapy in older men was associated with an
improved sense of well-being and mood which was
superior to placebo in several blinded, placebo-
controlled studies.
61,67
Neuropsychological performance
scores for visual memory and visuospatial performance
were lower in hypogonadal men, suggesting a possible
relationship between circulating plasma testosterone
levels and cognitive function.
76
Cardiovascular system
The incidence of cardiovascular disease and cardio-
vascular mortality increases as men age. However,
most epidemiological studies demonstrate that higher
serum testosterone levels are associated with a lower
risk of cardiovascular disease in men.
77
There are scant
published data on the effects of testosterone therapy
on cardiovascular risk factors in older men. Decreased
platelet aggregation or decreased vascular smooth-
muscle tone in men receiving androgen therapy has
been reported in some small preliminary studies. In
older men, testosterone administration by intra-
muscular injections or transdermal patch results in a
modest decrease in total or low-density lipoprotein-
cholesterol levels, and no change or a minimal decrease
in high-density lipoprotein-cholesterol levels.
73
Although the impact of these changes on cardiovascular
disease is unknown, one recent study demonstrated
that transdermal patch replacement therapy in hypo-
gonadal men over the age of 65 did not affect brachial
artery vascular reactivity.
78
Dehydroepiandrostenedione
replacement
Testosterone is not the only androgen that declines
with age. Dehydroepiandrostenedione (DHEA) also
declines with age. Serum DHEA levels are highest in
the third decade of life and then decline at a rate of
approximately 2% per year, so that by the eighth
decade, levels might be only 120% of normal values
among young people.
79
Decreased adrenal secretion of
DHEA is responsible for this decline, with the rate of
decline highest before age 60 and then later becoming
more gradual.
80,81
Data on the role of DHEA supplementation in the
aging male are conflicting. Morales et al. reported that
DHEA supplementation in aging men returned levels
of DHEA and DHEA sulfate to normal values and was
associated with signicant improvements in subjects
muscle strength and body fat mass, sense of well-
being, including better sleep, increased energy, and
better ability to handle stress.
82,83
More recently, in a
crossover study of 39 older men given 100 mg per
day of DHEA, there were no changes in measures of
body composition or subjective measurements of well-
being.
84
The effect of DHEA on bone mineral density is also
unclear. Villareal et al. reported an increase in total
bone mineral density (1.6%) and lumbar spine bone
mineral density (2.5%), as well as a decrease in total
and truncal fat and an increase in fat-free mass in men
and women who received 50 mg per day of DHEA for
6 months.
85
Labrie et al. reported similar changes in
women aged 6070 treated with a skin cream contain-
ing DHEA.
86
However, the Morales group did not nd
a change in bone mineral density after 6 months of
DHEA 100 mg/day.
82
There is inadequate evidence to
100 Androgens in Male Sexual Physiology
TABLE 6.3 Body fat, lean muscle mass, and muscle strength in the aging male on hormone replacement
therapy.
Study Treatment Study Body fat Lean body Muscle strength
(months) (n) (%) mass
Tenover (1992)
61
3 13 NC + 3.2% NC (grip)
Morley et al. (1993)
64
3 8 NC + (grip)
Marin et al. (1993)
67
9 31 6.4% NC
Urban et al. (1995)
68
1 6 + (LE)
Katznelson et al. (1996)
65
18 29 14.0% +5.0%
Sih et al. (1997)
69
12 17 NC + (grip)
Brill et al. (2002)
71
10 1 NC + (excluding time)
Ferrando et al. (2002)
70
6 12 + + (LE)
NC, no change; LE, leg extensors.
support the use of DHEA in the prevention or treat-
ment of age-related osteoporosis.
The relationship between cognition and DHEA has
been reported in a limited number of studies. The
Rotterdam study found that the ratio of free cortisol
over DHEA sulfate was signicantly related to cogni-
tive impairment, as measured by the Mini-Mental
State Examination healthy elderly subjects.
87
However,
no studies have demonstrated that DHEA supplemen-
tation improves cognition. Although short-term supple-
mentation produced mild changes in event-related
potentials, an improvement in memory or mood was
not demonstrated.
88
Claims that DHEA can prevent
memory loss and slow the progression of age-related
conditions such as Alzheimers or Parkinsons disease
are not supported by available scientic evidence.
DHEA sulfate concentration is independently and
inversely related to death from any cause and death
from cardiovascular disease in men over age 50.
89
The
Massachusetts Male Aging Study also suggests that low
DHEA and DHEA sulfate might predict coronary
artery disease in men.
90
These androgens can interfere
with the atherogenic process by several mechanisms.
They influence enzymes such as glucoso-6-phosphate
dehydrogenase, which can modify the lipid spectrum.
Furthermore, they can inhibit human platelet aggre-
gation, enhance brinolysis, slow down cell prolifer-
ation, and reduce plasma levels of plasminogen activator
inhibitor type 1 and tissue plasminogen activator
antigen. We suggest that all these DHEA (sulfate)
actions are dependent on sex hormone metabolic path-
ways. However, there are still insufcient data to advise
routine DHEA supplementation in elderly men.
91
ANDROGEN ADVERSE EFFECTS IN
OLDER MEN
As mentioned earlier, ART is contraindicated in men
with prostate or breast cancer; prostate cancer should
be excluded in all men over the age of 40 prior to
initiating ART, with a digital rectal examination and
determination of prostate-specic antigen. Many of the
potential risks of male HRT are increased in older men
and can be predicted by the baseline medical history,
examination, or laboratory testing.
Fluid overload from sodium and fluid retention may
occur during the rst few months of ART but is usually
minimal and transient. Caution should be used with
ART in chronically ill or frail older man due to the
risk that fluid overload may cause an exacerbation of
hypertension or congestive cardiac failure. ART may
exacerbate sleep apnea and sleep apnea may con-
tribute to low serum testosterone levels.
3638
As the
prevalence of sleep apnea is higher in older men,
patients should be specically questioned regarding
the presence of symptoms of sleep apnea before and
during ART.
Breast tenderness and gynecomastia may develop in
a small number of older men on ART. Both are more
common adverse effects of ART in older men than
younger men, due to the effects on breast tissue of the
increase in extraglandular estrogen formation, which
increases as men age.A signicant increase in red blood
cell mass and hemoglobin levels often occurs with ART
in older men and exceeds those changes seen in young
hypogonadal men receiving ART.
92
The development
of polycythemia in men on ART usually responds to a
reduction in the testosterone replacement dose but
may occasionally require cessation of therapy.
69,93
This
effect on red cell mass is less with the more uniform
and physiological plasma testosterone levels achieved
with ART using implants or transdermal patchs.
BPH and prostate cancer are common diseases in
older men. Both are promoted by androgens and
androgen deprivation therapy has been used for the
treatment of both of these diseases. ART should be
used cautiously in older men with symptoms of BPH
as it may precipitate urinary obstruction.
Prostate cancer is now the most commonly diagnosed
cancer in men, is second only to lung cancer as the
leading cause of cancer death in men and, as such, is a
major public health issue in all western countries. The
lifetime probability of prostate cancer is 1 in 6, and 1
in 3 men over the age of 50 years will die from prostate
cancer. The American Cancer Society estimated that in
1998 about 184 500 new cases of prostate cancer would
be diagnosed and 39 200 men will die of this disease
in the USA. In most countries, the age-standardized
incidence of prostate cancer increased dramatically
in the early 1990s coincident with increased public
awareness and the introduction of early detection and
screening programs based on regular digital rectal
examination and measurement of prostate-specic
antigen levels.
Whilst there is little doubt that androgens promote
the growth of diagnosed prostate cancer, the increased
risk of ART promoting the development of clinically
signicant prostate disease from preexisting, but sub-
clinical, disease is speculative. The incidence of prostate
cancer, prostate size, serum levels of prostate-specic
antigen, TRUS-guided prostatic needle biopsies, and
urodynamic parameters such as urine flow rates or
urinary bladder residual volumes in men aged 4089
years receiving ART exhibit either no change or minimal
change but remain within the normal range.
94
How-
ever, caution must be exercised as there are no long-
term safety data and the current data are insufcient
to rule out ART denitely as a causal risk of prostate
cancer.
The older man should be screened for conditions
that increase the potential risks of androgen therapy
Androgen Adverse Effects in Older Men 101
prior to initiating treatment. Prostate cancer should be
excluded in all men over the age of 40 prior to initiating
ART, with a digital rectal examination and deter-
mination of prostate-specic antigen. In men with an
elevated prostate-specic antigen level, further investi-
gation with TRUS and biopsy of the prostate should be
performed prior to initiation of ART.
Although larger longitudinal studies of male HRT
are lacking, the currently published studies indicate
that male HRT may improve mineral density, increase
muscle mass and strength, and, in some men, improve
libido and mood. There are no data, however; it can
only be speculated that ART is associated with a
reduced long-term fracture risk, improved mobility
and function, and improved overall quality of life.
Short- and intermediate-term adverse effects are well
characterized, relatively predictable, and manageable,
but long-term adverse effects on the prostate and
cardiovascular system are unclear. Despite the assertions
of special-interest groups, the media, and the pharma-
ceutical industry that male HRT reverses aging, the
duty of care of the treating physician is to offer older
men only evidence-based medical treatment and coun-
seling that the risk/benet ratio for the male HRT has
yet to be determined.
REFERENCES
1. Levalle O, Zilbersztein C, Aszpis S et al. Recombinant
human follicle-stimulating hormone administration increases
testosterone production in men, possibly by a Sertoli cell-
secreted nonsteroid factor. J Clin Endocrinol Metab 1998;
83:3973.
2. Gnessi L, Fabbri A, Spera G. Gonadal peptides as mediators
of development and functional control of the testis. Endocrine
Rev 1997; 18:541.
3. Hayes FJ, Hall JE, Boepple PA et al. Differential control
of gonadotropin secretion in the human: endocrine role of
inhibin. J Clin Endocrinol Metab 1998; 83:1835.
4. Boujrad N, Ogwuegbu SO, Garnier M et al. Identication
of a stimulator of steroid hormone synthesis isolated from
testis. Science 1995; 268:1609.
5. Dunn JF, Misula BC, Rodbard D. Transport of steroid
hormones: binding of 21 endogenous steroids to both
testosterone-binding globulin and corticosteroid-binding
globulin in human plasma. J Clin Endocrinol Metab 1981;
53:5868.
6. Pardridge WM, Landaw EM. Testosterone transport in
brain: primary role of plasma protein-bound hormone. Am J
Physiol 1985; 249:E534E542.
7. Manni A, Pardridge WM, Cefalu M et al. Bioavailability of
albumin-bound testosterone. J Clin Endocrinol Metab 1983;
61:705710.
8. Sakiyama R, Pardridge WM, Musto NA. Influx of
testosterone-binding globulin (TeBG) and TeBG-bound sex
steroid hormones into rat testis and prostate. J Clin Endocrinol
Metab 1988; 67:98103.
9. Khosla S, Melton LJ, Atkinson EJ et al. Relationship of
serum sex steroid levels and bone turnover markers with
bone mineral density in men and women: a key role for
bioavailable estrogen. J Clin Endocrinol Metab 1998;
83:2266.
10. MacDonald PC et al. Origin of estrogen in normal men
and in women with testicular feminization. J Clin Endocrinol
Metab 1979; 49:905.
11. Behre HM, Yeung CH, Nieschlag E. Diagnosis of male
infertility and hypogonadism. In: Nieschlag E, Behre HM,
eds. Andrology: male reproductive health and dysfunction.
Berlin: Springer, 1997:87111.
12. Plymate SR. Male hypogonadism. In: Becker KL, ed.
Principles and practice of endocrinology and metabolism, 2nd
edn. Philadelphia: JB Lippincott; 1995:10561082.
13. Goldzieher JW, Dozier TS, Smith KD et al. Improving the
diagnostic reliability of rapidly fluctuating plasma hormone
levels by optimized multiple-sampling techniques. J Clin
Endocrinol Metab 1976; 43:824.
14. Eliasson R, Lindholmer C. Functions of male accessory
genital organs. In: Hafez ESE, ed. Human semen and fertility
regulation in men. St Louis: CV Mosby; 1976:4450.
15. Gil-Vernet JM Jr, Alvarez-Vijande R, Gil-Vernet A et al.
Ejaculation in men: a dynamic endorectal ultrasonographical
study. Br J Urol 1994; 73:442448.
16. Mann T, Lutwak-Mann C. Male reproductive function and
semen. Berlin: Springer-Verlag; 1981:171193.
17. Palermo GD, Cohen J, Alikani M et al. Intracytoplasmic
sperm injection: a novel treatment for all forms of male factor
infertility. Fertil Steril 1995; 63:1231.
18. MacDonald PC, Madden JD, Brenner PF et al. Origin of
estrogen in normal men and in women with testicular
feminization. J Clin Endocrinol Metab 1979; 49:905.
19. Gray A, Feldman HA, McKinlay JB et al. Age, disease
and changing sex hormone levels in middle-aged men: results
of the Massachusetts male aging study. J Clin Endocrinol
Metab 1991; 73:1016.
20. Grumbach MM, Styne DM. Puberty: ontogeny, neuro-
endocrinology, physiology, and disorders. In: Williams RH,
Foster DW, Kronenberg HW et al., eds. Williams textbook
of endocrinology, 9th edn. Philadelphia: Saunders; 1998:
15091626.
21. Marshall WA, Tanner JM. Variation in the pattern of
pubertal changes in boys. Arch Dis Child 1970; 45:13.
22. Rugarli EI, Ballabio A. Kallmann syndrome. From genetics
to neurobiology. JAMA 1993; 270:2713.
23. Grifn JE, Wilson JD. Disorders of the testes and male
reproductive tract. In: Williams RH, Foster DW, Kronenberg
HW et al., eds. Williams textbook of endocrinology, 9th edn.
Philadelphia: Saunders; 1998: 819876.
24. Handelsman DJ. Testicular dysfunction in systemic
disease. J Clin Endocrinol Metab North Am 1994; 23:839.
102 Androgens in Male Sexual Physiology
25. Kobayashi K, Mizuno K, Hida A et al. PCR analysis of
the Y-chromosome long arm in azoospermic patients: evidence
for a second locus required for spermatogenesis. Hum Mol
Genet 1995; 3:1965.
26. Foresta C, Ferlin A, Garolla A et al. Y-chromosome
deletions in idiopathic severe testiculopathies. J Clin
Endocrinol Metab 1997; 82:1075.
27. Najmabadi H, Huang V, Yen P et al. Substantial
prevalence of microdeletions of the Y-chromosome in infertile
men with idiopathic azoospermia and oligospermia detected
using a sequence-tagged site-based mapping strategy. J Clin
Endocrinol Metab 1996; 81:1347.
28. Foresta C, Ferlin A, Garolla A et al. High frequency of
well-dened Y-chromosome deletions in idiopathic Sertoli
cell-only syndrome. Hum Reprod 1998; 13:302.
29. Salehian B. Testicular pathologic changes and the
pituitarytesticular axis during human immunodeciency
virus infection. Endocrinol Pract 1999; 5:1.
30. Grinspoon S, Corcoran C, Anderson E et al. Sustained
anabolic effects of long-term androgen administration in men
with AIDS wasting. Clin Infect Dis 1999; 28:634.
31. Snyder PF, Lawrence DA. Treatment of male hypo-
gonadism with testosterone enanthate. J Clin Endocrinol
Metab 1980; 51:1335.
32. Meikle AW, Mazer NA, Moellmer JF et al. Enhanced
transdermal delivery of testosterone across nonscrotal skin
produces physiological concentrations of testosterone and its
metabolites in hypogonadal men. J Clin Endocrinol Metab
1992; 74:623628.
33. Bals-Pratsch M, Langer K, Place VA et al. Substitution
therapy of hypogonadal men with transdermal testosterone
over one year. Acta Endocrinol 1988; 118:713.
34. Parker S, Armitage M. Experience with transdermal
testosterone replacement therapy for hypogonadal men. Clin
Endocrinol 1999; 50:57.
35. Drinka PJ, Jochen AL, Cuisinier M et al. Polycythemia as
a complication of testosterone therapy in nursing home men
with low testosterone levels. J Am Geriatr Soc 1995; 43:899.
36. Sandblom RE, Matsumoto AM, Schoene RB et al.
Obstructive sleep apnea syndrome induced by testosterone
administration. N Engl J Med 1983; 308:508510.
37. Schneider BK, Pickett CK, Zwillich CW et al. Influence
of testosterone on breathing during sleep. J Appl Physiol
1986; 61:618.
38. Santamaria JD, PriorJC, Fleetham JA. Reversible repro-
ductive dysfunction in men with obstructive sleep apnoea.
Clin Endocrinol (Oxf) 1988; 28:461470.
39. Barrett-Connor E. Testosterone, HDL-cholesterol and
cardiovascular disease. In: Bhasin S, Gabelnick HL, Spieler JM
et al., eds. Pharmacology, biology, and clinical applications of
androgens: current status and future prospects. New York:
Wiley-Liss; 1996:215223.
40. Gooren LJ, Polderman KH. Safety aspects of androgen
therapy. In: Nieschlag E, Behre HM, ed. Testosterone: action,
deciency and substitution. Berlin: Springer-Verlag; 1990:
182203.
41. Alexandersen P, Haarbo J, Christiansen C. The relation-
ship of natural androgens to coronary heart disease in males:
a review. Atherosclerosis 1996; 125:113.
42. Conway AJ, Boylan LM, Howe C et al. A randomised
clinical trial of testosterone replacement therapy in hypo-
gonadal men. Int J Androl 1988; 11:247264.
43. Snyder PF, Lawrence DA. Treatment of male hypo-
gonadism with testosterone enanthate. J Clin Endocrinol
Metab 1980; 51:1335.
44. Cantrill JA, Dewis P, Large DM et al. Which testosterone
replacement therapy? Clin Endocrinol (Oxf) 1984; 24:97107.
45. Handelsman DJ, Conway AJ, Boylan LM. Pharmaco-
kinetics and pharmacodynamics of testosterone pellets in
man. J Clin Endocrinol Metab 1990; 71:216222.
46. Grinspoon S, Corcoran C, Anderson E et al. Sustained
anabolic effects of long-term androgen administration in men
with AIDS wasting. Clin Infect Dis 1999; 28:634.
47. Wilson JD. Androgen abuse by athletes. Endocrinol Rev
1988; 9:181199.
48. Lin GC, Erinoff L, eds. Anabolic steroid abuse. National
Institute on Drug Abuse Research monograph series. Rockville,
MD: US Department of Health and Human Services; 1990.
49. Bhasin S, Storer TW, Berman N et al.The effects of supra-
physiologic doses of testosterone on muscle size and strength
in normal men. N Engl J Med 1996; 335:17.
50. Young NR, Baker HWG, Liu G, Seeman E. Body com-
position and muscle strength in healthy men receiving testos-
terone enanthate for contraception. J Clin Endocrinol Metab
1993; 77:10281032.
51. Handelsman DJ, Gupta L. Prevalence and risk factors for
anabolic-androgenic steroid abuse in Australian secondary
school students. Int J Androl 1997; 20:159164.
52. Nankin HR, Calkins JH. Decreased bioavailable testos-
terone in aging normal and impotent men. J Clin Endocrinol
Metab 1986; 63:14181420.
53. Gray A, Berlin JA, McKinlay JB et al. An examination of
research design effects on the association of testosterone and
male aging: Results of a meta-analysis. J Clin Epidemiol 1991;
44:671684.
54. Vermeulen A. Clinical review 24: androgens in the aging
male. J Clin Endocrinol Metab 1991; 73:221224.
55. Morley JE, Kaiser FE, Perry HM et al. Longitudinal
changes in testosterone, luteinizing hormone, and follicle-
stimulating hormone in healthy older men. Metabolism 1997;
46:410413.
56. Stearns EL, MacDonnell JA, Kaufman BJ et al. Declining
testicular function with age: hormonal and clinical correlates.
Am J Med 1974; 57:761766.
57. Tenover JS, Matsumoto AM, Plymate SR et al. The effects
of aging in normal men on bioavailable testosterone and
luteinizing hormone secretion: response to clomiphene citrate.
J Clin Endocrinol Metab 1987; 65:11181126.
References 103
58. Bremner WJ, Vitiello MV, Prinz PN Loss of circadian
rhythmicity in blood testosterone levels with aging in normal
men. J Clin Endocrinol Metab 1983; 56:12781281.
59. Neaves WB, Johnson L, Porter JC et al. Leydig cell
numbers, daily sperm production, and serum gonadotrophin
levels in aging men. J Clin Endocrinol Metab 1984;
55:756763.
60. Takahashi J, Higashi Y, LaNasa JA et al. Studies of the
human testis XVIII. Simultaneous measurement of nine
intratesticular steroids: evidence for reduced mitochondrial
function in testis of elderly men. J Clin Endocrinol Metab
1983; 56:11781187.
61. Tenover JS. Male hormonal changes with aging. In:
Morley JE, Korenman SG, eds. Endocrinology and metabolism
in the elderly. Boston: Blackwell Scientic; 1992:243261.
62. Jackson JA, Kleerekoper M, Partt AM et al. Bone
histomorphometry in hypogonadal and eugonadal men
with spinal osteoporosis. J Clin Endocrinol Metab 1987;
65:5358.
63. Greenspan SL, Oppenheim DS, Klibanski A. Importance of
gonadal steroids to bone mass in men with hyperprolactinemic
hypogonadism. Ann Intern Med 1989; 110:526531.
64. Morley JE, Perry HM, Kaiser FE et al. Effects of testos-
terone replacement therapy in old hypogonadal males: a
preliminary study. J Am Geriatr Soc 1993; 41:129152.
65. Katznelson L, Finkelstein JS, Schoenfeld DA et al. Increase
in bone density and lean body mass during testosterone
administration in men with acquired hypogonadism. J Clin
Endocrinol Metab 1996; 81:43584365.
66. Reid IR, Wattie DJ, Evans MC et al. Testosterone therapy
in gluco-corticoid treated men. Arch Intern Med 1996;
156:11731177.
67. Marin P, Holmang S, Gustafsson E et al. Androgen
treatment of abdominally obese men. Obesity Res 1993;
1:245251.
68. Urban RJ, Bodenburg YH, Gilkison C et al. Testosterone
administration to elderly men increases skeletal muscle
strength and protein synthesis. Am J Physiol 1995; 269:
E820E826.
69. Sih R, Morley JE, Kaiser FE et al. Testosterone replace-
ment in older hypogonadal men: a 12-month randomized
controlled trial. J Clin Endocrinol Metab 1997; 82:16611667.
70. Ferrando AA, Shefeld-Moore M, Yeckel CW et al.
Testosterone administration to older men improves muscle
function: molecular and physiological mechanisms. Am J
Physiol Endocrinol Metab 2002; E601E607.
71. Brill KT,Weltman AL, Gentili A et al. Single and combined
effects of growth hormone and testosterone administration on
measures of body composition, physical performance, mood,
sexual function, bone turnover, and muscle gene expression
in healthy older men. J Clin Endocrinol Metab 2002;
87:50015007.
72. OCarroll R, Bancroft J. Testosterone therapy for low
sexual interest and erectile dysfunction in men: a controlled
study. Br J Psychiatry 1984; 145:146151.
73. Carani CM, Zini D, Baldini A. Effects of androgen treat-
ment in impotent men with normal and low levels of free
testosterone. Arch Sex Behav 1990; 19:223234.
74. Guay AT, Bansal S, Heatley GJ. Effect of raising
endogenous testosterone levels in impotent men with
secondary hypogonadism: double blind placebo-controlled
trial with clomiphene citrate. J Clin Endocrinol Metab 1995;
80:35463552.
75. Rhoden EL, Teloken C, Sogari PR et al. The relationship
of serum testosterone to erectile function in normal aging
men. J Urol 2002; 167 (4):17451748.
76. Moffat SD, Zonderman AB, Metter EJ et al. Longitudinal
assessment of serum free testosterone concentration predicts
memory performance and cognitive status in elderly men.
J Clin Endocrinol Metab 2002; 87:50015007.
77. Bagatell CJ, Bremner WJ. Androgen and progestagen
effects on plasma lipids. Prog Cardiovasc Dis 1995;
38:255271.
78. Kenny AM, Prestwood KM, Gruman CA et al. Effects of
transdermal testosterone on lipids and vascular reactivity in
older men with low bioavailable testosterone levels. J Gerontol
Biol Sci Med Sci 2002; 57 (7):M460M465.
79. Orentreich N, Brind JL,Rizer RL et al. Age changes and
sex differences in serum DHEAS concentrations throughout
adulthood. J Clin Endocrinol Metab 1984; 59:551555.
80. Baulieu EE. Dehydroepiandrosterone (DHEA): a fountain
of youth? J Clin Endocrinol Metab 1996; 81:31473151.
81. Labrie F, Belanger A, Cusan L et al. Marked decline in
serum concentrations of adrenal C19 sex steroid precursors
and conjugated androgen metabolites during aging. J Clin
Endocrinol Metab 1997; 82:23962402.
82. Morales AJ, Haubrich RH, Hwang JY et al. The effect of
six months treatment with 100 mg daily dose dehydro-
epiandrosterone (DHEA) on circulating sex steroids, body
composition, and muscle strength in age-advanced men and
women. Clin Endocrinol 1998; 49:421432.
83. Yen SSC, Morales AJ, Khorram O. Replacement of DHEA
in aging men and women: potential remedial effects. Ann NY
Acad Sci 1995; 774:128142.
84. Flynn MA, Weaver-Osterholtz D, Sharpe-Timms KL et al.
Dehydroepiandrosterone replacement on aging humans. J Clin
Endocrinol Metab 1999; 84:15271533.
85. Villareal DT, Holloszy JO, Kohrt WM. Effects of DHEA
replacement on bone mineral density and body composition in
elderly women and men. Clin Endocrinol 2000; 53:561568.
86. Labrie F, Diamond P, Cusan L et al. Effect of 12-month
dehydroepiandrosterone replacement therapy on bone,
vagina, and endometrium in postmenopausal women. J Clin
Endocrinol Metab 1997; 82:34983505.
87. Kalmijn S, Launer LJ, Stolk RP et al. A prospective study
on cortisol, dehydroepiandrosterone sulfate, and cognitive
function in the elderly. J Clin Endocrinol Metab 1998;
83:34873492.
88. Wolf OT, Naumann O, Hellhammer DH et al. Effects
of dehydroepiandrosterone replacement in elderly men on
104 Androgens in Male Sexual Physiology
event-related potentials, memory, and well-being. J Gerontol
1998; 53:M385M390.
89. Barrett-Connor E. Lower endogenous androgen levels
and dyslipidemia in men with non-insulin-dependent diabetes
mellitus. Ann Intern Med 1992; 117:870811.
90. Feldman HA, Johannes CB, Araujo AB et al. Low
dehydroepiandrosterone and ischemic heart disease in
middle-aged men: prospective results from the Massachusetts
Male Aging Study. American Journal of Epidemiology 2001;
513:7989.
91. Porsova-Dutoit I, Sulcova J, Starka L. Do DHEA/DHEAS
play a protective role in coronary heart disease? Physiol Res
2000; 49 (suppl. 1):S43.
92. Drinka PJ, Jochen AL, Cuisiner M et al. Polycythemia as
a complication of testosterone therapy in nursing home men
with low testosterone levels. J Am Geriatr Soc 1995; 43:899.
93. Tenover JL. Effects of androgen supplementation in the
aging male. In: Oddens JB, Vermeulen A, eds. Androgens and
the aging male. New York: Parthenon; 1996:191204.
94. Behre HM, Bohmeyer J, Nieschlag E. Prostate volume
in testosterone-treated and untreated hypogonadal men in
comparison to age-matched normal controls. Clin Endocrinol
(Oxf) 1994; 40:341349.
95. Vermeulen A, Kaufman JM. Androgens and the aging
male. New York: Parthenon; 1996:202.
96. Lunglmayr G. (1997) Trial on androgen supplementation
in aging men. In: Waites GMH, Frick J, Baker GWH, eds.
Current advances in andrology. Bologna: Monduzzi Editore;
1997:289292.
97. Smith KW, Feldman HA, MacKinlay JB. Construction and
eld validation of a self administered screener for testosterone
deciency. Clin Endocrinol (Oxf) 2000; 53 (6):703711.
References 105
MALE ERECTILE DYSFUNCTION
Introduction
A recent milestone in medicine allowed for a break-
through in the eld of erectile dysfunction (ED).
This event was the March 1998 Food and Drug
Administrations (FDAs) approval of the rst ever oral
agent used to treat ED. Unfortunately, prior to this
time, many ED complaints were attributed to psycho-
logical conditions and accurately diagnosed by either
urologists or primary care providers. Dr. Francois Eid
puts the impact of sildenal into perspective:
It is useful to remember that in 1997; prior to FDA
approval, only 1.7 million men were evaluated and
treated for ED. Urologists evaluated most of those
patients. Of that 1.7 million, 39% were prescribed
yohimbine, which is as efcacious as placebo.
Another 30% were prescribed the Medicated
Urethral System for Erection (MUSE), an alprostadil
suppository therefore, less than 30% of the 1.7
million men were given effective treatment in the
form of penile prosthesis, intracorporal injection
therapy, or an external vacuum device.
1
The World Health Organization (WHO) has under-
scored the impact of ED as a worldwide public health
issue. An international consensus meeting asserted
fundamental individual rights to sexual health that
include: (1) a capacity to enjoy and control sexual and
reproductive behavior in accordance with a personal
social ethic; (2) freedom from fear, shame, guilt, false
beliefs, and other factors that inhibit sexual response
and impair sexual relationships; and (3) freedom from
organic disorders, diseases, and deciencies that inter-
fere with sexual and reproductive functioning.
2
The
current WHO guidelines on ED offer a stepwise
protocol for recognizing, assessing, and treating ED
and for categorizing treatments as rst-line (oral
agents), second-line (intracavernous injection therapy
and intraurethral therapy), and third-line (surgical
prosthesis).Vacuum constriction devices can be used at
any point throughout the treatment process. In addition,
hormonal therapy and penile revascularization have
been proven successful for a small number of patients
with specic etiologies. Sex therapy can be used alone
in specic cases or in combination with other treat-
ments.
3
This chapter reviews current information on
phosphodiesterase type 5 (PDE5) inhibitors and their
role in the treatment of male and female sexual
dysfunction.
Epidemiology of ED
A literature review completed in 1999 demonstrated
a 52% prevalence of ED in non-institutionalized men
between 40 and 70 years of age in the Boston,
Massachusetts area.
4
Identied risk factors for ED
include atherosclerosis, diabetes mellitus, chronic renal
failure, liver failure, multiple sclerosis (MS),Alzheimers
disease, chronic obstructive lung disease, and endocrine
disorders. As expected, advancing age was also a pre-
dominant risk factor, with the prevalence nearly
doubling from 39% of 40-year-olds to 67% of men in
their 70s.
4
A cross-national study of over 2400 men in
Malaysia, Italy, Brazil, and Japan also reinforced the
astounding prevalence of ED. In the Japanese cohort,
71% of the 700 men reported some degree of dys-
function. Again, a higher prevalence was seen with
increasing age.
5
Physiology of erection
Erectile function is the result of a complex interaction
of hemodynamic, neurologic, neuropeptide, electro-
physiologic, and pathophysiologic factors. The primary
hemodynamic events of penile tumescence/detu-
escence are regulated by corporal smooth-muscle relax-
ation/contraction, respectively.Tumescence (erection) is
mediated by: (1) the parasympathetic nervous systems
release of acetylcholine, and (2) the non-adrenergic,
non-cholinergic cavernous nerve release of nitric oxide
(NO) with subsequent activation of cyclic guanosine
monophospate (cGMP). This NO/cGMP mechanism
appears to play a major role in modulating erectile
function. So much so, that organic or psychogenic
factors causing pathway alterations can impair the
relaxation of smooth muscle or even increase its
contraction, thereby causing ED.
4
A pivotal event in tumescence is the active clearance
of calcium from the corpora smooth muscle. This
Oral Type 5 Phosphodiesterase Therapy for
Male and Female Sexual Dysfunction
J. C. Trussell, Aristotelis G. Anastasiadis, Harin Padma-Nathan, and Ridwan Shabsigh
CHAPTER 7
smooth muscle is in a tonic state of contraction that,
upon relaxation (removal of intracellular calcium),
allows for sinusoidal enlargement, helicine artery
dilation, and eventually, emissary vein compression.
Smooth-muscle contraction relies on the interaction
between thin and thick muscle laments. This inter-
action (which results in force generation) can only occur
after the phosphorylation of a 20 kDa light chain of
myosin by a Ca
2+
/calmodulin-dependent myosin light-
chain kinase.Without the presence of calcium, thin and
thick muscle laments are held apart, allowing for
relaxation. In the normal corpora this smooth-muscle
relaxation results in tumescence.
In an effort to promote tumescence, the removal of
intracellular calcium requires energy. The corpora have
two different energy generators used to pump calcium
to the extracellular space via calcium channels, or to
sequester calcium in the cells endoplasmic reticulum
storage spaces. These two energy sources are: (1) cyclic
adenosine monophosphate (cAMP), mediated by
prostaglandin E
1
, and (2) cGMP, mediated by NO. By
inhibiting degradation, the effect of these two energy
sources can be potentiated. This is accomplished by
blocking phosphodiesterase (PDE), which acts to con-
vert active cAMP/cGMP into inactive 5AMP/5GMP
respectively. Along this line, sildenal blocks PDE type
5 (PDE5), which specically inhibits cGMP break-
down, while papaverine blocks PDE24, acting more
specically within the cAMP pathway.
Sildenal can only potentiate cGMP activity after
cGMP is manufactured. This is why sexual stimulation
is necessary to enable sildenal to be effective. Sexual
stimulation is what triggers the non-adrenergic, non-
cholinergic release of NO, thereby initiating a rise in
cGMP levels. This rise is then additionally potentiated
by sildenal.
Pharmacology of sildenal
Sildenal is an orally active, potent, and selective
inhibitor of cGMP-specic PDE5.
6
Although four PDE
isoenzymes have been identied in penile tissues
(PDE2, 3, 4, and 5) the predominant isoform is PDE5.
Since the discovery of sildenal, ve additional human
PDE families have been discovered (PDE711).
7
Fortunately, sildenal has a 1010 000-fold greater
selectivity for this particular isoform (PDE5). It also
has a 4000-fold greater selectivity for PDE5 than
PDE-3 the isoform involved in the control of cardiac
contractility.
6
Moreover, PDE5 is not expressed in
isolated cardiac myocytes, and as a result, sildenal
has no known direct effects on the myocardium. Its
potential effects on coronary circulation, specically
vasodilation, may be benecial in minimizing angina,
or even a myocardial infarction.
After oral administration, sildenal is rapidly
absorbed with an absolute bioavailability of 40%. The
time to peak plasma concentration (t
max
) while fasting
is 30120 min with a median time of 60 min.
6,8
On
the other hand, after a high-fat meal, not only is the
time to peak plasma concentration prolonged (by an
additional 60 min), but the peak plasma concentration
is reduced by 29%. The terminal half-life of sildenal
is 35 h.
6,8
Clinically, responses to the medication may
be observed for upwards of two to three half-lives
(812 h).
Sildenal is metabolized by hepatic microsomal
cytochrome P450 isoenzymes 3A4 (major route) and
2C9 (minor route).
6,8
Potent P450 3A4 inhibitors, such
as cimetidine, erythromycin, and ketaconazole, may
retard metabolism. Because of this, drug levels may be
two to eight times higher in these patients. Interestingly,
even with higher serum drug concentrations, side-effect
proles were no different from that of the general
study population. Patients receiving ritonavir, a
protease inhibitor sharing two metabolic pathways
with sildenal, should not be given sildenal at doses
greater than 25 mg or at a frequency of greater than
48 h. Since an active metabolite of sildenal is excreted
in the feces (80%) and urine (13%),
8
downward dose
adjustments (starting with 25 mg) are recommended
for patients who are over 65 years of age, have hepatic
impairment, or have severe renal insufciency.
Dosage
The recommended dose of sildenal ranges from 25 to
100 mg. Due to increased plasma concentrations and
efcacy (and possible side-effects), a 25 mg starting
dose is recommended for patients over 65 years of age,
those with liver failure, or severe renal insufciency,
and for individuals who are concomitantly taking
drugs that inhibit cytochrome P450 isoenzyme 3A4
(see above). Otherwise, the initial starting dose should
be 50 mg taken orally 1 h before sexual activity. Note,
however, that some patients have reported the onset of
activity as early as 1119 min.
6,8
Following this initial
dose, adjustments may be made upward (maximum
of 100 mg), or downward based on efcacy and
tolerability.
In the American flexible-dose study, 75% of patients
ultimately chose the 100 mg dose, 23% the 50 mg
dose, and 2% the 25 mg dose.
9
Similar results were
reported in a worldwide flexible-dose study where
60% chose 100 mg, 30% chose 50 mg, and 10% chose
the 25 mg dose.
8
Contraindications
Sildenal was initially developed and tested as a
vasodilator to be used in cardiac patients. Since small
108 Oral Type 5 Phosphodiesterase Therapy for Male and Female Sexual Dysfunction
amounts of PDE5 are also found on vascular endo-
thelium, an expected event resulting from PDE5
inhibition is the potentiation of the hypotensive (vaso-
dilatory) effects of nitrates. Because of this, sildenal is
contraindicated in patients taking organic nitrates or
other NO donors (such as nitroprusside). Be cautious
of patients taking nitrates on an intermittent basis, or
even recreationally (amyl nitrate, poppers).
10
This, of
course, includes all routes for nitrate administration:
sublingual, transnasal, transdermal, and orally. In
addition, although no case has been reported to date,
the second contraindication is a known hypersensitivity
to tablet components. Cardiovascular events, such as
myocardial ischemia/infarction, are more related to the
physical exertion of sexual activity rather than a direct
effect of sildenal. Patient stratication and recommen-
dations will be discussed later in this chapter.
Precautions
An update on product labeling by the US FDA has
cautioned against the use of sildenal in patient popu-
lations that have generally been excluded in clinical
trials. This includes patients who have sustained a
myocardial infarction, stroke, or life-threatening
arrhythmia within the past 6 months; patients with
resting hypotension (<90/50 mmHg) or hypertension
(>170/110 mmHg); patients with cardiac failure or
coronary artery disease causing unstable angina (see
later in chapter); and patients with retinitis pigmentosa.
8
Excluding the last group, these precautions also apply
to the use of any of the other vasoactive drugs used to
treat ED.
Safety of sildenal
Flexible-dose studies
The most common adverse events in sildenal clinical
trials have included vasodilatory effects such as head-
aches, flushing, and nasal congestion from hyperemic
nasal mucosa. In addition, dyspepsia has been observed.
A dog model has demonstrated that PDE5 may have
a role in maintaining gastroesophageal sphincter
integrity. Therefore, blocking PDE5 can allow for
reflux, and thereby, result in dyspepsia. A recent review
by Morales of more than 3700 patients in 18 of 21
clinical trials examined adverse experiences from
sildenal in a total exposure of 1631 man-years.
11
The
most common adverse events in 2% or more of the
placebo-controlled studies are listed in Table 7.1.
12
A total of 574 adverse events occurred in the 734
sildenal-treated patients, with most events reported
as transient in nature and described as mild (62%) or
moderate (31%).
13
Overall discontinuation rates due
to adverse events were comparable in the sildenal
(2.5%) and placebo (2.3%) treatment groups. Head-
aches (1.1%), flushing (0.4%), and nausea (0.4%) were
the most common causes leading to discontinuation.
The nausea side-effect presumably arises from
dyspepsia, which results from a relaxing effect on the
gastroesophageal sphincter. Lastly, only 1 of the 2722
patients treated with (up to) 100 mg of sildenal
discontinued treatment due to abnormal vision.
11
It is interesting to note that respiratory tract
infection, back pain, and a flu-like syndrome were
noted in 2% or more of the patients, but occurred in
equal frequency with active drug and placebo groups.
In early sildenal studies, back pain occurred in nearly
10% of patients with daily and thrice-daily dosing.
8
This effect has also been observed with other PDE5
inhibitors and is apparently not associated with the
vasculitis picture, or abnormal serum chemistries such
as creatinine kinase.
Fixed-dose studies
The American xed-dose, randomized, placebo-
controlled study provides further insight into the dose
relationship of adverse events.
14
Headaches, flushing,
dyspepsia, nasal congestion, abnormal vision, and
dizziness were not only found to be the most common
adverse-events, but were also found to be dose-
related.
11,15
Once again, headache was the most
common adverse event leading to treatment discon-
tinuation (0.6% of those in the 100 mg group). Discon-
tinuation rates due to adverse events were similar
between the 25 and 50 mg groups (0.6 and 0.4%
respectively), while they were slightly higher with the
100 mg dose (1.2%).The placebo-related adverse event
discontinuation rate was comparable at 1.0%.
12,15
Male Erectile Dysfunction 109
TABLE 7.1 Sildenal adverse event experience
in worldwide flexible-dose studies
11
Percentage of patients
Adverse event Sildenal Placebo
(n = 734) (n = 725)
Headache 16 4
Flushing 10 1
Dyspepsia 7 2
Nasal congestion 4 2
Abnormal vision 3 0
Diarrhea 3 1
Dizziness 2 1
Urinary tract infections 3 2
Long-term, open-label studies
Adverse events experienced by the 2199 patients in
10 separate long-term studies were headache (10%),
flushing (9%), dyspepsia (6%), and respiratory tract
infection (6%). Abnormal vision was reported by 2%
of the patients. In a 1-year analysis, adverse event and
lack of efcacy accounted for 2% and 4% of with-
drawals, respectively.
Headache continues to be the number-one reason
for discontinuation.
16
Although no reports of priapism,
prolonged erections, or penile brosis occurred during
the worldwide clinical trials,
8,11
after FDA approval,
there were at least 25 cases of prolonged erections (>4,
<6 h) or priapism. Notably, many of these cases were
related to combinations of sildenal with other phar-
macologic therapies for ED (either injection or intra-
urethral). The safety and efcacy of such combinations
have yet to be established.
Finally, the impact of adverse events may wane with
prolonged usage. A review of 17 studies comparing
adverse events (a biweekly evaluation over 4 months)
suggested that adverse events might be transient and
eventually decrease in frequency to levels indistin-
guishable from those experienced by placebo-controlled
arms. After 2 months of treatment, differences in
adverse events between sildenal users and placebo
were no longer statistically signicant.
17
Efcacy
Unless otherwise noted, the following efcacy statistics
are based on the sildenal New Drug Application to
the FDA. These data were based on 4526 patients: 576
in phase I studies, 3003 in phase IIIII studies, 769 in
long-term extension studies (with 550+ patients treated
for longer than 1 year), and 178 in Japanese studies.
The mean patient age was 55 years, ranging from 18 to
87. Patients were diagnosed with an organic etiology
51.8% of the time; 18% were psychogenic, and the
remaining 25.7% were considered mixed. As can be
expected with ED, comorbid conditions are common:
24% had hypertension, 16% had diabetes mellitus,
14% had cardiovascular disease, 14% had hyper-
lipidemia, 6% were individuals with spinal cord injury
(SCI), 5% had depression, and 4% had a radical
prostatectomy.
1415,16,18
Sildenal clinical trials employed four different tools
to measure efcacy (some of which have been sub-
sequently modied for efcient clinical use). These
include: (1) the International Index of Erectile Function
(IIEF): a 15-item questionnaire addressing relevant
domains of sexual function. In general these questions
address erectile function, orgasm, desire, satisfaction
with intercourse, and overall sexual satisfaction. This
questionnaire has been validated in 20 different
languages and is both sensitive and specic for detect-
ing treatment-related changes;
19
(2) a Global Efcacy
Question (GEQ). This requires a simple yes or no
response to: Did the treatment improve your
erections?;
9,20
and (3) a log of erectile activity
whereby the patient would record information on the
date and dose taken, the presence of sexual stimu-
lation, and whether successful sexual intercourse took
place. In addition, the 24-week, xed-dose, American
trial included a four-point scale to measure penile
rigidity,
9,20
where, for example, grade three was an
erection hard enough for penetration but not com-
pletely hard, in comparison to grade four a fully
rigid erection.
15
The last survey was optional and
assessed the partners perception of the patients ability
to achieve, and then maintain, an erection.
20
International Index of Erectile Function
IIEF survey results were compiled from 3000+ patients
from 21 American and European randomized, double-
blinded, placebo-controlled, phase III trials lasting up
to 6 months. To reflect the National Institutes of Health
denition of ED,
16
primary endpoints for measuring
efcacy were based on the results of only two items on
this IIEF questionnaire: item 3, the ability to achieve
an erection, and item 4, the ability to maintain an
erection.
19
In all 21 studies, sildenal produced a
signicantly improved erection when compared to
placebo. Regarding survey results, signicant improve-
ments were reported for both items 3 (attain erection)
and 4 (maintain erection), where increases of 100%
and 130% were recorded respectively.
9,20
In flexible-
dose studies, 69% (94/134) of patients taking sildenal
reported erections sufcient for vaginal penetration on
most to all occasions, compared with 23% (32/138)
taking placebo.
20
In addition, 62% (85/132) of
sildenal users were able to maintain their erections
after penetration on most to all occasions, in com-
parison with 15% (21/138) of the placebo group.
Overall, 59% (81/137) treated with sildenal reported
that they were able to achieve and maintain their
erections on most to all occasions compared to 15%
(21/138) of placebo-treated patients.
20
On subset
analysis, these results were consistent regardless of
age, race, baseline severity, or etiology of dysfunction.
Furthermore, sildenal was found to be effective for
those patients with each of the following ED risk
factors: coronary artery disease, hypertension, periph-
eral vascular disease, diabetes mellitus, coronary artery
bypass grafting, radical prostatectomy, transurethral
resection of the prostate (TURP), and SCI. This held
true even for patients currently being medicated for
those conditions. In addition, effective treatment was
also seen in depressed patients, as well as those taking
antidepressants and antipsychotic medication.
8
110 Oral Type 5 Phosphodiesterase Therapy for Male and Female Sexual Dysfunction
In addition to items 3 and 4, the remaining questions
on the IIEF questionnaire were also evaluated, and,
as expected, secondary improvements were seen in
the sildenal group regarding intercourse, orgasm, and
overall sexual satisfaction. The domain of desire,
however, was not increased by sildenal, implying that
it lacks aphrodisiac qualities.
Global Efcacy Question and diary data
General population
Overall erectile improvement with sildenal, as
evaluated by the GEQ, was 74% (101/136) compared
to 16% (23/118) in the placebo group (P < 0.0001).
20
Once again, improved erections were seen in all patient
groups regardless of etiology, with the most success
(80% of patients) seen in the psychogenic group. In
addition, improvements were noted in nearly 70% of
men with organic and 75% of mixed ED.
8,14,20
Sildenal
dose escalation also resulted in more patients with
improved erections. Compared to 24% of patients
taking placebo, 63%, 74%, and 82% of patients taking
25, 50, and 100 mg of sildenal respectively reported
improvement in erections (n = 1797).
8
A similar dose
response was extracted from the patients diary, part
of which used a four-point grading scale for penile
rigidity. On this scale grade 3 represented an erection
hard enough for penetration, but not fully hard and
grade 4 was considered a fully rigid erection.
Seventy-ve percent of patients receiving 25 mg, 80%
receiving 50 mg, and 85% of patients receiving 100 mg
of sildenal demonstrated a grade 3 or 4 erection in
comparison to 50% of patients receiving placebo.
15
In these two groups of treated patients, 80% of those
responding at grade 3 and 94% of those reaching
grade 4 also reported successful sexual intercourse.
15
Patients with diabetes
An initial pilot study of 21 diabetic patients (types 1
and 2) with a mean age of 50 years and a 3-year median
duration of ED examined the efcacy of sildenal in
doses of up to 50 mg.
21
ED improvement (measured
by diary, questionnaire, and RigiScan) was noted in
48% and 52% of the men receiving 25 and 50 mg of
sildenal, respectively, compared to 10% of patients
receiving placebo. More recently, a large study of 268
diabetic men (mean age of 57 years, mean diabetes
duration of 12 years, 21% with type I and 79% with
type 2) with a 12-year mean duration of ED was
randomized for 12 weeks of treatment.
13
This flexible-
dose study started at 50 mg with dose adjustments (up
and down) based on efcacy and adverse events. At
the end of the 12-week study, improved erections were
noted in 56% of sildenal patients compared with
22% receiving placebo (P < 0.001). Likewise, sexual
intercourse success rates were 48% versus 12%.
13
Patients with spinal cord injury
The efcacy of sildenal in the population of spinal
cord-injured individuals has also been examined in
several studies, including a single-dose, double-blinded,
two-way crossover study using RigiScan evaluation in
27 patients.
22,23
In that study 65% of sildenal patients,
compared with 8% of placebo patients, had penile
base rigidity greater than 60%. No patient discontinued
sildenal due to adverse events. A larger randomized,
double-blind, placebo-controlled, crossover, flexible-
dose study of 178 patients employed the IIEF and
partners questionnaire.
24
Fifteen percent (27/178) of
these patients had no prestudy erectile function. Erectile
function improved in 83% of patients taking sildenal,
versus 12% of those taking placebo. The sexual inter-
course success rate was greater than 70% on sildenal,
and 0% for those taking placebo (n = 45).
24
Patients with multiple sclerosis
A double-blind, placebo-controlled, flexible-dose study
of sildenal in men with MS and ED demonstrated
improved erections in nearly 90% of the sildenal
group, in contrast to a 24% response in the placebo
group.
25
Patients after radical prostatectomy
Unlike the traditional radical prostatectomy, the
nerve-sparing approach has signicantly improved
postoperative erectile function. None the less, when
objectively evaluated, up to 80% of men sustain some
degree of ED following nerve-sparing surgery. In a sub-
group analysis of men in American trials having had
prostatectomies (n = 42), 42.5% of those receiving
sildenal demonstrated improved erections compared
to 14.6% of patients receiving placebo.
8,26
The resulting
intercourse success rate was nearly 30%, compared
to 5% taking placebo. Since this study lumped all
prostatectomy patients together (both nerve-spared
and non-nerve-spared), a more accurate response rate
can be provided based on a patients specic operative
outcome. Doing this, Zippi and colleagues
27
have
demonstrated an improvement of erections in 71.7%
(38/50), 50% (6/12), and 15.4% (4/26) of men follow-
ing a bilateral nerve-sparing, unilateral nerve-sparing,
and non-nerve-sparing procedure, respectively. To
validate further the importance of sparing nerves to
improve postoperative erectile function, closer scrutiny
of the IIEF (questions 34) responses were similarly
poor for the non-nerve-sparing and unilateral nerve-
sparing, in contrast to the excellent responses seen in
bilateral nerve-spared patients.
27
In an attempt to preserve healthy corporal tissue,
more recent studies have suggested expanding the
indication of sildenal (and other non-surgical inter-
ventions) to include the immediate postop period.
Male Erectile Dysfunction 111
Considering sildenal acts to promulgate the effects
of NO, then conceptually it should work best when
cavernous nerves are intact, to allow for its release and
subsequent tumescence. In contrast, if neither caver-
nous nerve could be spared during a prostatectomy,
sildenal would have no effect on the severed nerve
bundles. In this case, injectable vasoactive agents might
work best. Reducing cavernosal brosis has been
attempted with both sildenal and PGE injections. A
study from Connecticut looked at corpora cavernosal
biopsies during and 6 months following a bilateral
nerve-sparing radical prostatectomy in 40 potent
patients. Every other night sildenal (50 or 100 mg)
was started just after Foley removal. This early sildenal
treatment resulted in preservation of corporal smooth
muscle while lessening the development of corporal
brosis. In fact, those patients taking the higher
100 mg dose seemed to develop much less brosis, and
may have even developed more corporal smooth
muscle!
28
For patients recovering after non-nerve-
sparing radical prostatectomies, consider early intra-
cavernous injections as soon as the rst postoperative
month. Waiting longer may result in cavernous veno-
occlusive dysfunction, which is the most likely expla-
nation for subsequent non-response to PGE in this
patient population.
29
These studies do not demonstrate
a return of potency, but maintenance of proerectile
corporal ultrastructure should be an integral part in
the preservation of postradical erectile function.
Patients after radiation therapy for
prostate cancer
In contrast to the sudden onset of ED in men following
radical prostatectomy, the onset of ED in those receiving
external beam radiation is more insidious. The extent
of impaired sexual function following radiation was
the most important predictor of response to sildenal.
Ninety percent of men who had partial erections
following radiotherapy also had signicant responses
to sildenal compared with only 52% of men who had
flaccid erections (P = 0.02).
30
Another study noted
an increased response to sildenal over time (40%
responded the rst week, while 77% responded at the
sixth week), warning that the drug should not be
discontinued prematurely for this group of patients
because of presumed ineffectiveness.
31
Lastly, sildenal
is also useful in treating patients with ED occurring
after prostate brachytherapy.
32
Patients with transurethral resection of
the prostate
In a subpopulation analysis (after TURP), 60% of those
who had ED after their resection saw an improvement
with sildenal, compared to 33.9% of men receiving
placebo (n = 171).
26
Patients with depression
Beyond direct effects on sexual function, ED also leads
to depressive symptoms, low self-esteem, and other
signs of psychological distress.
3336
A recent landmark
study of 152 men with untreated minor depression
(according to DSM-IV) and concomitant ED were
treated with sildenal.
37
Of the patients receiving
sildenal, 73% were considered responders, in contrast
to 14% of patients receiving placebo.The study showed
that, regardless of the treatment received (placebo or
sildenal), patients who reported a positive treatment
also claimed a signicant improvement in depression
parameters! In those patients the depression was most
likely a secondary factor to ED, but an important fact
is that the study clearly demonstrated that sildenal
treatment resulted in improvements in both sexual and
non-sexual aspects of life quality.
Patients with increased age
ED represents an important quality-of-life issue for
many aging men. Studies seem to conflict regarding
the efcacy of sildenal. For instance, a study in 1998
compared IIEF responses in men younger or older
than 65 years of age. No difference in erectile function
between the two age groups was demonstrated.
18
In
contrast, a more recent study (2002) from Japan did
nd a statistically signicant difference between the
under- and over-65-year-old groups (89.1% versus
65.75% efcacy, respectively). It is noteworthy, how-
ever, that this second study used a modied IIEF
questionnaire (the IIEF-5, which asked only ve
questions), and used only 25 and 50 mg doses of
sildenal.
38
The fact that 100 mg doses of sildenal
were not used may be signicant in light of evidence
that, as men age, the resting tone of cavernosal smooth
muscle is reset to a higher level. This higher level of
smooth-muscle tone may require a greater degree of
relaxation to permit tumescence.
39
Although the older
patients (from Japan) showed a higher prevalence of
diabetes mellitus, hypertension, and benign prostatic
hyperplasia, only the diagnosis of diabetes appeared
to decrease the efcacy of sildenal (P = 0.019).
38
Age-related changes in sexual function did not appear
to be directly related to either testicular testosterone
production or serum testosterone concentration in
most older men. Whether or not testosterone declines
with age seems controversial. Some studies demon-
strate an age-related decrease in testosterone produc-
tion related to decreased testis sensitivity to luteinizing
hormone,
4042
while others have reported no effect of
age on total testosterone concentrations.
38,43
The Japan
study did demonstrate a signicant decrease in mean
serum luteinizing hormone and follicle-stimulating
hormone concentrations in the group of men over 65
years of age.
38
112 Oral Type 5 Phosphodiesterase Therapy for Male and Female Sexual Dysfunction
Patients with premature ejaculation
Studies are just starting to look at sildenal in treating
premature ejaculation. In two studies sildenal was
added to selective serotonin uptake inhibitor (SSRI)
therapy, with signicant improvements in ejaculatory
delay.
44,45
At this time, the mechanism of action is
unclear. It is important to remember that increased
adverse events may be seen when combining sildenal
and SSRIs.
45
Partner data
In most studies, partner input was optional. However,
whenever data were present, partner ratings of the
patients ability to achieve and maintain an erection
during sexual activity were signicantly higher for
the sildenal groups compared to those taking
placebo.
8,20,46
Long-term efcacy
In long-term follow-up studies of 1, 2, and 3 years,
more than 95% of patients continued to demonstrate
not only improved erections, but also an improved
ability to engage in sexual activity.
11
More specically,
a 3-year review of efcacy after radical prostatectomy
(using the modied, ve-item IIEF questionnaire)
demonstrated that 71% (29/41) still responded to
sildenal. There was no statistical signicance between
the 1- and 3-year postop questionnaire results, regard-
less of the specic surgical category (bilateral, unilateral,
or non-nerve-sparing). Thirty-one percent (9/29) of
patients responding to sildenal did eventually require
the 100 mg dosage; on the other hand, 50% of those
who dropped out of the study (6/12) did so because
of an eventual return to natural erections. Overall,
85% of the patients were sexually satised, and 95%
were able to achieve and maintain erections during
more than 65% of their attempts.
47
Salvage of sildenal non-responders
Sildenal is an effective rst-line treatment for ED,
with 65% of initial prescriptions written by primary
care providers. Not infrequently, patients are referred
to a urologist because of sildenal failure or non-
response. Studies have shown that careful patient
reeducation with supplemental written information
and possible video instruction can turn many such
patients into responders. Many of the individuals
reporting initial failure had tried the drug only one
time, had only used 25 or 50 mg doses, and had lacked
concomitant sexual stimulation.
48,49
It is important
for patient education to include: (1) reminders that
absorption is best on an empty stomach (at least a low-
fat diet); (2) information that tobacco cessation will
improve responses; and (3) advice to avoid the
sedative effects of alcohol. In a prospective study of
622 patients referred to a urologist, 38% (98/622)
reverted to responders after higher doses (100 mg)
and reeducation.
48
A separate study reported a 54%
(41/76) salvage rate. Those with diabetes, active
smokers, and hypertensive men taking multiple agents
were most unlikely to respond to salvage efforts.
49
It
will be interesting to see if future studies include doses
of sildenal greater than 100 mg. In phase I clinical
studies of healthy male volunteers, doses of up to
800 mg were not associated with adverse events which
were different from those occurring with doses ranging
from 25 to 100 mg.
8
FEMALE SEXUAL DYSFUNCTION
Disclosure
Sildenal is currently not FDA-approved for the treat-
ment of female sexual dysfunction (FSD).
Introduction
In contrast to the plethora of information and wide-
spread interest in the research and treatment of male
sexual dysfunction, much less attention has been given
to similar female difculties. This eld of study is
relatively new, with a limited number of tools and
protocols available to help in understanding the
complex psychological and physiological interactions
that comprise normal female sexual function. In fact,
it was not until 1999 that an international consensus
panel developed a classication of FSD (Table 7.2),
which, for the rst time, included possible organic
causes. This is in contrast to classications from the
WHO International Classication of Diseases-10
(ICD-10) and Diagnostic and Statistical Manual
of Mental Disorder (DSM-IV) that rely more on
psychiatric disorder nomenclature.
50,51
Female Sexual Dysfunction 113
TABLE 7.2 Classication of female sexual
dysfunction according to the 1999 Consensus
Classication System
50
I. Sexual desire disorders:
Hypoactive sexual desire disorder
Sexual aversion disorder
II. Sexual arousal disorder
III. Orgasmic disorder
IV. Sexual pain disorders:
A. Dyspareunia
B. Vaginismus
Other sexual pain disorders
Unfortunately, medical interventions are extremely
limited.Aside from hormone replacement therapy, there
are no FDA-approved medical treatments for FSD.
52
On the other hand, several studies have demonstrated
successful treatment using sildenal (see below).
Epidemiology
FSD is a multidimensional problem that is age-related,
progressive, and highly prevalent, affecting 3050% of
American women.
53
Based on epidemiological data
from the National Health and Social Life Survey which
included 1749 women under the age of 60 years, 43%
had complaints of sexual dysfunction, 33% of women
lacked sexual interest, and nearly 25% did not
experience orgasm.
54
In addition, nearly 20% of the
women reported either lubrication difculties, and/or
nding sex not pleasurable.
50,54
Pathophysiology and oral
pharmacotherapy
Characterizing the female sexual response has evolved
several times, with the rst description by Masters and
Johnson in 1966. These researchers described four
successive phases: excitement, plateau, orgasm, and
resolution.
55
Later, Kaplan introduced desire as part
of a three-phase model: desire, arousal, and orgasm. In
this model, desire comes rst and acts to incite the
overall response cycle.
56
More recently, others have
suggested that sexual function should be viewed as a
circuit, with four main domains: libido, arousal, orgasm,
and satisfaction. Each of these can overlap and act as
negative- or positive-feedback interactions on the other
three domains.
53,57
This sexual function cycle, or circuit, is dependent
on normal anatomy and physiology. Disruptions in
one or more of the following areas may lead to FSD:
vasculogenic, neurogenic, hormonal, musculogenic,
pain conditions, and psychogenic.
The same neurogenic disorders that cause ED in men
can also cause sexual dysfunction in women. Although
women with incomplete SCI do retain the capacity for
psychogenic arousal and vaginal lubrication,
58
they do
have signicantly more difculty achieving orgasm.
In a placebo-controlled, double-blind, crossover study
of 19 premenopausal SCI patients receiving 50 mg
sildenal, a signicant increase in subjective arousal
was seen. Consistent with previous ndings in men, the
best results were observed after conditions of optimal
stimulation. The authors of this research state: further
large-scale studies of sildenals effects in women with
neurogenic sexual dysfunction are strongly indicated.
58
In women, despite the presence or absence of organic
disease, emotional and relational issues signicantly
affect sexual arousal. Sexual response is either
enhanced by self-esteem, positive body image, and a
good partner relationship, or inhibited by mood dis-
orders and depression (as well as the medications used
to treat them). In particular, one of the most commonly
used medications to treat uncomplicated depression
is SSRIs. Women receiving these medications often
complain of decreased desire, decreased arousal, and
decreased genital sensation, as well as difculty
achieving orgasm. This response can be explained, in
part, by the SSRIs peripheral mechanism of action
(at the level of NO synthase). NO synthesis is tightly
regulated by the activity of NO synthase. Nerve ter-
minal excitement, either from central input (fantasy)
or peripheral stimulation (foreplay), will cause the
events necessary for NO release.As shown in Figure 5.3,
the action of NO synthase is inhibited by SSRIs.
5557
Because of NO inhibition and the subsequent effect on
cGMP production, it is best to consider treating these
patients with sildenal in attempt to enhance whatever
cGMP effects may be present. Several studies have,
in fact, documented postsildenal improvements in
SSRI-induced sexual dysfunction in women.
59
Specic
SSRIs implicated in FSD include: fluoxetine, sertraline,
paroxetine, and fluvoxamine.
59
Safety
Keeping in mind that sildenal is not currently FDA-
approved for FSD, several studies have not only
reported efcacy, but also include an adverse event
prole very similar to that seen with the male
population,
53,59,60
except for one caveat. In a study of
33 women using 50 mg sildenal, 21% (7/33) reported
clitoral discomfort and hypersensitivity, causing
three of these patients to drop out of the study.
60
CARDIOVASCULAR EFFECTS OF
SILDENAFIL
ED is not uncommon among men who have cardio-
vascular disease. In fact, it may be among the rst signs
of small-vessel disease of the penis. Somehow, these
vessels appear to be more sensitive to atherosclerotic
occlusion than blood vessels in the heart or other areas
of the body. Because of this, physicians must consider
ED as a predictive factor for ischemic heart disease,
61
and consider the emergence of ED, in a previously
asymptomatic male, as a marker for occult coronary
artery disease.
11
As anticipated, extensive coronary
artery disease reflects worse ED. Patients whose
ischemic heart disease involves more than one vessel
often have greater trouble achieving erections than
those patients with single-vessel disease.
62
In addition
to the effects of cardiovascular disease, certain
medications used to treat heart disease (including
114 Oral Type 5 Phosphodiesterase Therapy for Male and Female Sexual Dysfunction
beta-blockers and thiazide diuretics) have been
associated with the development of ED.
62
The possibility of PDE5-inhibitors affecting the
heart has been studied extensively. To date, there is no
evidence that sildenal directly affects myocytes, or
that PDE5-inhibitors have any direct effect on either
the myocardium or the cardiac conduction system.
Recent studies have veried that sildenal is not
inotropic and does not alter cardiac output.
63,64
In men
with stable coronary artery disease, as assessed by
exercise echocardiography, sildenal had no effect on
symptoms, exercise duration, or the presence or extent
of exercise-induced ischemia.
65
In contrast, the effect
of sildenal on coronary circulation is potentially
benecial as it provides small increases in coronary
blood flow. Unfortunately, this small increase in
coronary blood flow is not globally protective.
The American Heart Association and American
College of Cardiology consensus document clearly
addressed the issue of sildenal use in male patients
with cardiovascular disease and ED, but failed to
address management of cardiac risk associated with
sexual activity.This issue was more specically addressed
at a Consensus Conference on Sexual Activity and
Cardiac Risk at Princeton University.
71
In general, it
was reported that patients can be placed into one of
three major categories (Table 7.3) at the time of initial
assessment, based on their cardiovascular status: low-
risk, high-risk, or intermediate/indeterminate-risk.
Those in the low-risk category have no specic cardiac
risk associated with the treatment of ED and sexual
activity. These patients may be treated for ED without
the need for additional cardiovascular evaluation. On
the other hand, the high-risk category includes patients
with cardiovascular disease, which requires specialized
cardiac consultation, evaluation, and priority cardio-
vascular management. Sexual activity and the manage-
ment of sexual dysfunction in such patients should be
deferred until the patients cardiac condition has been
evaluated, treated, and fully stabilized. Patients with
an intermediate or indeterminate level of risk should
not resume sexual activity, or undergo treatment of
sexual dysfunction, until a cardiac evaluation aimed at
facilitating the restratication of these patients into the
high- or low-risk category has been performed.
Similar agents in development
The success of sildenal has resulted in an explosive
growth in the eld of sexual pharmacology. Not only
are new agents being developed that act at different
locations (either centrally or peripherally), but also
additional PDE5-inhibitors are being developed in
attempts to improve efcacy and PDE-isoform speci-
city. The pharmacokinetic properties of the different
PDE5-inhibitors are shown in Table 7.4.
IC351 (tadalal, Cialis)
IC351 (tadalal, trade name Cialis, Lilly ICOS,
Indianapolis, IN) is a new compound of the PDE5-
inhibitor class, which has proven in in vitro trials to
have a higher selectivity to the PDE5 enzyme than
sildenal.
72
Compared to sildenal, IC351 has a longer
half-life of 17.5 h.
72
Clinical human experiences with
IC351 are reviewed in Porst.
72
They include a double-
blind, placebo-controlled, single-crossover RigiScan
Cardiovascular Effects of Sildenafil 115
TABLE 7.3 Cardiovascular risk groups
according to the Princeton guidelines
71
Low-risk
Asymptomatic, fewer than three risk factors for
coronary artery disease (excluding gender)
Controlled hypertension
Mild, stable angina (prior cardiovascular
assessment)
Post successful coronary revascularization
Uncomplicated past myocardial infarction
(>68 weeks)
Mild valvular disease
Congestive heart failure (New York Heart
Association (NYHA) class I)
Intermediate- or indeterminate-risk
Three or more risk factors for coronary artery
disease (excluding gender)
Moderate stable angina
Recent myocardial infarction (>2, <6 weeks) or
cerebrovascular accident
Left ventricular dysfunction/congestive heart failure
(NYHA class II)
Arrhythmia of unknown cause
High-risk
Unstable or refractory angina
Uncontrolled hypertension
Congestive heart failure (NYHA class III, IV) and
cardiomyopathies
Recent myocardial infarction (<2 weeks),
cerebrovascular accident
High-risk arrhythmias
Hypertrophic obstructive cardiomyopathy or
idiopathic hypertrophic subaortic stenosis and
other cardiomyopathies
Moderate/severe valvular disease
study in 44 men with mild to moderate ED after visual
sexual stimulation,
73
two large dose-ranging studies
in the USA and Canada,
74,75
and a Spanish 12-week,
randomized, double-blind, parallel study taken as
needed in 216 diabetic patients.
76
The various trials
demonstrated the effectiveness of IC351 in a broad-
spectrum population of ED patients, including dia-
betics. Major differences compared to sildenal are a
higher PDE5 selectivity, especially in terms of PDE6,
resulting in the avoidance of visual disturbances, and
the longer half-life time, leading to a longer window of
opportunity.
72
The trials also showed a favorable
adverse effect prole with less headaches, flushing, and
dyspepsia than reported in the rst large sildenal
trial.
72
An update on IC351 trials was recently
presented at the 2002 annual meeting of the American
Urological Association (AUA): a double-blind, placebo-
controlled study of 348 men taking 20 mg tadalal
demonstrated efcacy for at least a 36-h time period.
At 24 h postdose, 57.3% of intercourse attempts by
tadalal-treated men were successful versus 31.3% of
placebo-controlled attempts (P < 0.001). The effect was
maintained at 36 h, with 60.4% of intercourse attempts
successful, versus 29.9% of attempts on placebo
(P < 0.001).
77
Tadalal, however, may have an effect
for 6080 h and is in the blood stream for up to 100 h
in men over 60 years of age.This is also associated with
a potential for a longer period of nitrate interaction.
Tadalal was well tolerated: most adverse events
reported were only mild to moderate in intensity. Once
again, dyspepsia and headache were the most common
(>5%) treatment-emergent adverse events of this
study.
77
Little is known about tadalals direct cardiac
effect. This is important since it is an inhibitor of
PDE11, a cardiac PDE. Tadalal dosing (from 2.5 to
20 mg) was evaluated in ve randomized, double-blind,
placebo-controlled phase III trials enrolling 1112 men.
Compared with placebo, tadalal signicantly improved
all primary and secondary endpoints (IIEF, GAQ,
diaries), with 81% of tadalal-treated patients report-
ing improved erections, and the mean percentage of
successful intercourse attempts improved to 75%.
Adverse events were similar to the study above, except
that an additional 45% of men reported back pain.
78
Since some outlying subsets of patients experienced
hypotension, tadalal should not be used in combi-
nation with nitrates.
79
Vardenal
Vardenal (Bayer Corp., West Haven, CT) is another
new PDE5-inhibitor that has high potency in vitro (50%
inhibitory concentration, IC
50
, 0.1 nmol/l) and selec-
tivity for PDE5 compared with other isoenzymes.
80,81
It has been shown to be active for inducing penile
erection in a conscious rabbit model when given
orally.
82
Pharmacokinetic data in humans were
obtained in two randomized, double-blind, placebo-
controlled threefold crossover studies with a single
oral dose of 10, 20, or 40 mg of vardenal in men with
mild to moderate ED.
83,84
RigiScan data showed a
signicant difference in rigidity at the base of the penis
between placebo and vardenal at all doses, but there
was no signicant difference between 20 and 40 mg of
vardenal.The duration of rigidity greater than 60% at
the base and the tip of the penis increased from 25 min
to more than 60 min with 40 mg of vardenal. Again,
there was no signicant difference between the 20 and
40 mg doses. At-home oral administration of vardenal
was carried out in Europe, the USA, and South Africa
in randomized, double-blind, placebo-controlled studies
of 601 men with ED (reviewed in Pryor
80
). Efcacy
was assessed using the IIEF, the FuglMeyer quality
of life questionnaire as well as a global assessment
question (GAQ: Has the treatment improved your
erections?). Compared to placebo, vardenal showed
signicant improvement in the questionnaires as well
as in the GAQ.
82
Vardenal was well tolerated, with
headache (818%), vasodilation (613%), rhinitis
(18%), and dyspepsia (26%) listed as the most
common adverse events.
85
In addition, two pivotal
double-blind phase III trials were conducted in North
America (n = 749) and Europe (n = 636) and were able
to verify that vardenal signicantly improved erectile
function in patients with hypertension, hyperlipidemia,
or diabetes. Following FDA approval, vardenal had a
116 Oral Type 5 Phosphodiesterase Therapy for Male and Female Sexual Dysfunction
TABLE 7.4 Pharmacokinetic parameters of
selective PDE5-inhibitors
81
Parameter Sildenal Tadalal Vardenal
In vitro IC
50
3.5 6.7 0.1
(nmol/l)
C
max
(ng/ml) 560 378 209
T
max
(h) 0.70.8 2.0 0.70.8
t
1
2
(h) 3.7 17.5 3.9
AUC 1685 8066 74.5
(ng h/ml)
Metabolism Hepatic Hepatic Hepatic
Protein 96 94
binding (%)
Bioavailability 40 Unknown 15
(%)
In vitro IC
50
, concentration of the drug that inhibits a given
response (PDE5) by 50%; C
max
, maximum total plasma
concentration; T
max
, time to C
max
; t
1/2
, half-life; AUC, area
under the plasma concentrationtime curve.
contraindication in men receiving either nitrates or
any -blocker.
CONCLUSION
Sildenal has revolutionized the treatment of ED. Not
only is it an effective oral agent treating 70% of men
with a wide range of ED risk factors, but it also comes
with a limited adverse event prole (headache, flushing,
dyspepsia). A word of caution, however, is necessary
for those patients taking nitrates (a sildenal contra-
indication) and for those patients with exercise limi-
tations who should receive cardiac clearance before
assuming the physical strain of sexual activity.
Recently, the use of sildenal has expanded into the
eld of FSD, particularly for sexual arousal disorder.
With newer, more selective PDE5 agents on the horizon,
ED treatment options will continue to expand, and
have longer efcacy and fewer adverse events.
REFERENCES
1. Eid JF. Present and future management of erectile dys-
function: treatment options.AUA Annual Convention 2001;12
Suppl 4:S626.
2. Jardin A, Wagner G, Khoury S et al., eds. Erectile dys-
function the rst international consultation. Plymouth, UK:
Health Publication; 2000.
3. Padma-Nathan H. A new era in the treatment of erectile
dysfunction. Am J Cardiol 1999; 84:18N23N.
4. Melman A, Gingell JC. The epidemiology and patho-
physiology of erectile dysfunction. J Urol 1999; 161:511.
5. McKinlay JB, Digruttolo L, Glasser D et al. International
differences in the epidemiology of male erectile dysfunction.
Int J Clin Pract 1999; (suppl 102): 35.
6. Boolell M, Allen MJ, Ballard SA et al. Sildenal: an orally
active type 5 cyclic GMP-specic phosphodiesterase inhibitor
for the treatment of penile erectile dysfunction. Int J Impot
Res 1996; 8:4752.
7. Gbekor E, Bethell S, Fawcett L et al. Phosphodiesterase 5
inhibitor proles against all human phosphodiesterase families:
implication for use as pharmacological tools. J Urol 2002;
167 (suppl):246, abstract 967.
8. Viagra labeling information, NDA submission. New York,
NY: Pzer; 1997.
9. Goldstein I, Lue T, Padma-Nathan H et al. Oral sildenal
in the treatment of erectile dysfunction. N Engl J Med 1998;
338:13971404.
10. Webb DJ, Freestone S, Allen MJ et al. Sildenal citrate
and blood pressure lowering drugs: results of drug interaction
studies with organic nitrate and a calcium antagonist. Am J
Cardiol 1999; 83 (5A):21C28C.
11. Morales A, Gingell C, Collins M et al. Clinical safety of
sildenal citrate (Viagra) in the treatment of erectile
dysfunction. Int J Impot Res 1998; 10:6973.
12. Padma-Nathan H, Giuliano F. Oral drug therapy for erec-
tile dysfunction. Urol Clin North Am 2001; 28(2):321334.
13. Rendell MS, Fajfer J, Wicker PA et al. Sildenal for the
treatment of erectile dysfunction in men with diabetes. JAMA
1999; 281:421426.
14. Fugl-Meyer AR, Lodnert G, Branholm IB et al. On life
satisfaction in male erectile dysfunction. Int J Impot Res
1997; 9:141148.
15. Padma-Nathan H, the Sildenal Study Group. A 24-week,
xed-dose study to assess the efcacy and safety of sildenal
(Viagra) in men with erectile dysfunction. J Urol 1998; 159
(suppl):238A.
16. NIH consensus development panel on impotence.
Impotence. JAMA 1993; 270:8390.
17. Carson CC, Siegel RL, Orazem J. Sildenal citrate treat-
ment for erectile dysfunction: rate of adverse events decreases
over time. J Urol 2002; 167 (suppl.):179, abstract 717.
18. Wagner G, Maytom M, Smith M et al. Analysis of the
efcacy of sildenal (Viagra) in the treatment of male erectile
dysfunction in elderly patients. J Urol 1998; 159 (suppl):
238A.
19. Rosen RC, Riley A, Wagner G et al. The International
Index of Erectile Function (IIEF): a multidimensional scale
for assessment of erectile dysfunction. Urology 1997; 49:
822830.
20. Padma-Nathan H, Steere WD, Wicker PA. Efcacy and
safety of oral sildenal in the treatment of erectile dysfunction:
a double blind, placebo-controlled study of 329 patients. Int
J Clin Pract 1998; 52:14.
21. Price D, Gingell C, Gepi-Attee S et al. Sildenal, a novel
oral therapy for penile erectile dysfunction in patients with
diabetes. Diabet Med 1997; 14:A6.
22. Derry FA, Dinsmore WW, Fraser M et al. Efcacy and
safety of oral sildenal in men with erectile dysfunction caused
by spinal cord injury. Neurology 1998; 51:16291633.
23. Derry F, Gardner B, Glass C et al. Sildenal: a double
blind placebo controlled single dose 2-way crossover study in
men with erectile dysfunction caused by traumatic spinal cord
injury. J Urol 1997; 157 (suppl.):181.
24. Holmgren E, Ciuliano F, Hultling C et al. Sildenal in the
treatment of erectile dysfunction caused by spinal cord injury:
a double blind placebo controlled flexible dose 2-way cross-
over study. Neurology 1998; 50:A127.
25. Unpublished data. New York, NY: Pzer; 2000.
26. Padma-Nathan H, and the Sildenal Study Group. Efcacy
of Viagra (sildenal citrate) in the treatment of erectile dys-
function (ED) in men with transurethral or radical prostatec-
tomy. J Urol 1999; annual meeting.
27. Zippi CD, Jhaveri FM, Klein EA et al. Role of Viagra after
radical prostatectomy. Urol 2000; 55: 241245.
28. Schwartz EJ, Wong P, Graydon RJ. Sildenal preserves
intracorporal smooth muscle content after bilateral nerve
sparing radical retropubic prostatectomy. J Urol 2002; 167
(4):278, abstract 1096.
References 117
29. Gontero P, Pretti G, Bagnasacco A et al. A prospective
study on the optimal timing for intracavernous PGE rehabili-
tation following non nerve sparing radical prostatectomy.
J Urol 2002; 167 (4):147, abstract 590.
30. Zelefsky MJ, McKee AB, Lee H et al. Efcacy of oral
sildenal in patients with erectile dysfunction after radio-
therapy for carcinoma of the prostate. Urology 1999; 53:
775778.
31. Weber DC, Bieri S, Kurtz JM et al. Prospective pilot
study of sildenal for treatment of post radiotherapy erectile
dysfunction in patients with prostate cancer. J Clin Oncol
1999; 17:34443449.
32. Merrick GS, Butler WM, Lief JH et al. Efcacy of sildenal
citrate in prostate brachytherapy patients with erectile dys-
function. Urology 1999; 53:11121116.
33. Althof SE, Turner LA, Levine SE. Intracavernosal
injection in the treatment of impotence: a prospective study
of sexual, psychological, and marital functioning. J Sex Marit
Ther 1987; 13:155167.
34. Derogatis LR, Meyer JK. A psychological prole of the
sexual dysfunctions. Arch Sex Behav 1979; 8:201223.
35. Jonler M, Moon T, Brannan W et al. The effect of age,
ethnicity and geographical location on impotence and quality
of life. Br J Urol 1995; 75:651655.
36. Krane RJ, Goldstein I, Saenz DeTejada I. Impotence.
N Engl J Med 1989; 321:16481659.
37. Menza M, Rosen R, Sidmena S et al. The effect of
sildenal in men with erectile dysfunction and depression.
Presented at the American Psychiatric Association Annual
Meeting: 1999.
38. Tsujimura A, Yamanaka M, Takahashi T et al. The clinical
studies of sildenal for the ageing male. Int J Androl 2002;
25:2833.
39. Melman A, Ahao W, Rehman J et al. The successful long-
term treatment of age-induced erectile dysfunction with the
HSLO/Maxi-K gene. J Urol 2002; 167 (suppl):235, abstract
921.
40. Buvat J, Lemaire A. Endocrine screening in 1022 men
with erectile dysfunction: clinical signicance and cost-effective
strategy. J Urol 1977; 158:17641767.
41. Korenman SG, Morley JE, Mooradian AD et al. Secondary
hypogonadism in older men: its relation to impotency. J Clin
Endocrinol Metab 1990; 71:963969.
42. Guay AT, Bansal S, Hodge MB. Possible hypothalamic
impotence. Male counterpart to hypothalamic amenorrhea?
Urology 1991; 38:317322.
43. Harman SM, Blackman MR. Is there an andropause, the
analogue to menopause and if so what tissue are affected and
how? In: Robaire B, Pryor JL, Trasler JM, eds. Handbook of
andrology. 7275.
44. Chen J, Greenstein A, Mabjeesh NJ et al. Role of sildenal
in the treatment of premature ejaculation (PE). J Urol 2002;
167 (suppl.):280, abstract 1101.
45. Salonia A, Montorsi F, Zanoni M et al. A prospective
study comparing paroxetine alone versus paroxetine plus
sildenal in patients with premature ejaculation. J Urol 2002;
167 (suppl):279, abstract 1097.
46. Althof SE, Stecher VJ, Tseng L. Evaluation of partner
satisfaction with sildenal citrate treatment in men with
erectile dysfunction. J Urol 2002; 167 (suppl.): 280, abstract
1102.
47. Raina R, Nelson DR, Agarwal A et al. Long term efcacy
of sildenal citrate following radical prostatectomy: 3-year
follow-up. J Urol 2002; 167 (suppl):279, abstract 1100.
48. Sklar GN, Szostak MJ. Urology salvage of sildenal
failures from primary care physicians. J Urol 2002; 167
(suppl.):177, abstract 712.
49. Barada JH. Leadership role of urologist in optimizing
patient outcomes. J Urol 2001; (suppl): abstract 724.
50. Basson R, Berman J, Burnett A et al. Report of the inter-
national consensus development conference on female sexual
dysfunction: denitions and classications. J Urol 2000; 163:
888899.
51. American Psychiatric Association. DSM-IV: Diagnostic and
statistical manual of mental disorders, 4th edn. Washington,
DC: American Psychiatric Press; 1994.
52. Berman JR, Berman LA, Lin H et al. Effect of sildenal
on subjective and physiologic parameters of the female sexual
response in women with sexual arousal disorder. J Sex Marit
Ther 2001; 27:411420.
53. Berman JR, Bassuk J. Physiology and pathophysiology of
female sexual function and dysfunction. World J Urol 2002;
20:111118.
54. Laumann EO, Paik A, Rosen RC. Sexual dysfunction in
the United States: prevalence and predictors. JAMA 1999;
281:537544.
55. Masters ED, Johnson JV. Human sexual response. Boston,
MA: Little, Brown; 1966.
56. Kaplan H.The new sex therapy. London: Baillire Tindall;
1974.
57. Pelletier G. Localization of androgen and estrogen
receptors in rat and primate tissues. Histol Histopathol 2000;
15:12611270.
58. Sipski ML, Rosen RC, Alexander CJ et al. Sildenal
effects on sexual and cardiovascular responses in women with
spinal cord injury. Urology 2000; 55:812815.
59. Shen W, Urosevich Z, Clayton DO. Sildenal in the
treatment of female sexual dysfunction induced by selective
serotonin reuptake inhibitors. J Reprod Med 1999; 44:
535542.
60. de Groat WC, Booth AM. Physiology of male sexual
function. Ann Intern Med 1980; 92:329331.
61. Montorsi F, Salonia A, Montorsi P et al. May erectile
dysfunction predict ischemic heart disease? J Urol 2002; 167
(suppl.):148, abstract 591.
62. Jackson G. Erectile dysfunction and cardiovascular
disease. Int J Clin Pract 1999; 53:363368.
118 Oral Type 5 Phosphodiesterase Therapy for Male and Female Sexual Dysfunction
63. Herrmann HC, Chang G, Klugherz BD et al. Hemo-
dynamic effects of sildenal in men with severe coronary
artery disease. N Engl J Med 2000; 342:16221626.
64. Chen Y, Du R, Traverse JH et al. Effect of sildenal on
coronary active and reactive hyperemia. Am J Physiol Heart
Circ Physiol 2000; 279:H2319-H2325.
65. Arruda-Olson AM, Mahoney DW, Nehra A et al. Cardio-
vascular effects of sildenal during exercise in men with
known or probable coronary artery disease. JAMA 2002;
287:719725.
66. Feenstra J, Van Drie-Pierik RJ, Lade CF et al. Acute
myocardial infarction associated with sildenal. Lancet 1998;
352:957958.
67. Porter A, Mager A, Birnbaum Y et al. Acute myocardial
infarction following sildenal citrate (Viagra) intake in a
nitrate free patient. Clin Cardiol 1999; 22:762763.
68. Shah PK. Sildenal in the treatment of erectile dys-
function. N Engl J Med 1998; 339:699.
69. Hayashi K, Minekaki KK, Narukawa M et al. Atrial
brillation and continuous hypotension induced by sildenal
in an intermittent WPW syndrome patient. Jpn Heart J 1999;
40:827830.
70. Cheitlin MD, Hutter AM Jr, Brindis RG et al. Use of
sildenal in patients with cardiovascular disease. Circulation
1999; 99:168177.
71. DeBusk R, Drory Y, Goldstein I et al. Management of
sexual dysfunction in patients with cardiovascular disease:
recommendation of the Princeton Consensus Panel. Am J
Cardiol 2000; 86:175181.
72. Porst H. IC351 (tadalal, Cialis): update on clinical
experience. Int J Impot Res 2002; 14 (suppl.):S57S64.
73. Meuleman E et al. Effect of IC 351 on erectile response
to visual sexual stimulation. J Urol 1999; 161 (suppl.):212,
abstract 814.
74. Padma-Nathan H, McMurray JG, Pullman WE et al. for
the IC351 on demand dosing study group. On demand IC351
(Cialis
TM
) enhances erectile function in patients with erectile
dysfunction. Int J Impot Res 2001; 13:29.
75. Brock G et al. Efcacy and safety of IC351 treatment for
ED. Eur Urol 2001; 39 (suppl.):106, abstract 414.
76. Saenz de Tejada I et al. The effect of on-demand IC351
treatment of erectile dysfunction in men with diabetes. Eur
Urol 2001; 39 (suppl.):16, abstract 56.
77. Porst H, Rosen RC, Padma-Nathan H et al.Tadalal allows
men with erectile dysfunction to have successful intercourse
up to 36 hours post dose. J Urol 2002; 167 (suppl.):177,
abstract 709.
78. Brock G, McMahon C, Chen K et al. Efcacy and safety
of tadalal in men with erectile dysfunction: an integrated
analysis of registered trials. J Urol 2002; 167 (suppl.):178,
abstract 713.
79. Kloner RA, Mitchell MI, Beddiing A et al. Pharmaco-
dynamic interactions between tadalal and nitrates compared
with sildenal. J Urol 2002; 167 (suppl.):176, abstract 708.
80. Pryor J. Vardenal: update on clinical experience. Int J
Impot Res 2002; 14 (suppl.):S65S69.
81. Corbin JD, Francis SH. Pharmacology of phosphodiesterase-
5 inhibitors. IJCP 2002; 56:453459.
82. Bischoff E, Niewoehner U, Haning H et al.The oral efcacy
of vardenal hydrochloride for inducing penile erection in a
conscious rabbit model. J Urol 2001; 165:13161318.
83. Klotz T, Sachse R, Heidrich A et al. Vardenal increases
penile rigidity and tumescence in erectile dysfunction
patients: a RigiScan and pharmacokinetic study. World J Urol
2001; 19:3239.
84. Stark S, Sachse R, Liedl T et al.Vardenal increases penile
rigidity and tumescence in men with erectile dysfunction
after a single oral dose. Eur Urol 2001; 40:181188.
85. Donatucci C, Eardley I, McVary KT et al. Vardenal
improves erectile function regardless of etiology or baseline
severity in men with erectile dysfunction. J Urol 2002; 167
(suppl.):178, abstract 715.
86. Goldscher E, Eardley I, Segerson T. Vardenal improves
erectile function in men with signicant comorbidities
associated with erectile dysfunction. J Urol 2002; 167
(suppl):178, abstract 714.
References 119
INTRODUCTION
The modern era of pharmacotherapy for the treatment
of erectile dysfunction (ED) was ushered into clinical
practice by intracavernous vasoactive drugs. Prior to
the dramatic demonstrations and reports by Brindley
1
and Virag
2
in the early 1980s that direct injection of
vasoactive agents could induce penile rigidity, the only
effective means of restoring erectile function was either
surgical or by external appliances. The numbers of
men willing to undergo these forms of treatment and
their attendant costs limited treatment to a small
minority of those affected. The availability of a rever-
sible therapy, able to simulate a natural erection with a
rapid onset of action and detumescence, represented
an important medical advance. This landmark finding,
that direct local injection into the corpora cavernosa,
intracavernosal injection (ICI), could create a useful
erection, was the spark that ignited research into the
physiology of erection worldwide.
3,4
Over the past two decades, medical research into the
underlying etiology of ED has flourished. We currently
have an expanded understanding of the role of the
nitric oxide (NO)-cyclic guanosine monophoshate
(cGMP) second messenger pathway essential for
normal erectile function.
5
These advances have led to
investigators evaluating a number of potential intra-
cavernous agents in clinical trials using human volun-
teers and animal models. These include NO donors,
6
potassium channel openers,
7
vasoactive polypeptide
8
and, most recently, guanylate cyclase activators.
9
Although a large number of vasoactive compounds
have been tested, the most clinically important agents
remain prostaglandin E
1
(PGE-1), phentolamine, and
papaverine. These vasoactive agents have been reported
alone and in combination at various doses and
concentrations.
10
Local pharmacological therapies for the treatment of
male ED are logical, effective, inexpensive, and widely
available. The fact that oral therapies have largely
usurped these treatments in usage points to their
greatest weakness the need for local delivery via
needle or intraurethral application. Based on recent
North American data it is clear that the generally good
efcacy and safety of oral and sublingual pharmaco-
therapy have relegated injection therapy to a second-
line therapy.
11,12
Currently greater than 90% of new
treatment prescriptions are being written for sildenal.
In most current management algorithms a minimal
investigation is suggested with a trial of oral medi-
cation. If successful, no further diagnostic maneuvers
are recommended and ongoing use of oral agents is
suggested. If oral agents do not succeed, an attempt
with second-line therapies such as injection therapy,
intraurethral therapy, and vacuum devices are offered.
This approach is both time- and cost-effective and
allows the vast majority of men to achieve an effective
result. The Canadian Urology Association acting
through its guidelines committee has recently published
its management algorithm for ED
13
(Fig. 8.1).
In spite of the clear dominance of oral rst-line
therapy, the potential advantages of local vasoactive
treatment for ED are easily apparent. It reduces the
chance of systemic complications, is nerve-independent,
has a faster onset of action and smaller chance for drug
interaction, and is able to achieve high local concen-
trations even in situations of severe vascular insuf-
ciency (Box 8.1). An extensive clinical experience with
their use currently exists. These agents preceded oral/
sublingual ED therapy development and remain an
important treatment option for men who cannot take
or tolerate rst-line treatments.
The future of this category of agents is bright. Use
of novel agents delivered directly into the penile
circulation has been reported recently and includes
MUSE and Intracavernosal Therapies
Gerald B. Brock
CHAPTER 8
Box 8.1 Ideal candidates for local therapy
Unsuccessful at rst-line oral therapy
Nitrate use or potential for nitrate use
Neural injury from pelvic surgery, radiation, or
trauma
Desires rapid onset of erection following therapy
Diabetic or severe vasculopaths (usually after
failed rst-line therapy)
Wishes greater rigidity than achievable with oral
agents
guanylate cyclase activators, potassium-channel
openers, as well as standard vasoactive agents such
as PGE-1, where their impact on cavernous smooth
muscle and vascular function have been evaluated.
Their role in disease modication, especially in the
postradical prostatectomy patient who frequently
undergoes a period of neuropraxia postoperatively,
may prove essential to optimize erectile function.
14
The
potential of local injections to upregulate NO synthase
activity via a high-flow effect is currently under study.
15
The potential for gene therapy, delivered by direct
injection into the penile circulation, makes the role of
ICI in the future seem bright.
16
In this chapter a comprehensive review of local
treatments for ED is presented. This should not be
considered a historical review, as it is the belief of the
author that local therapy remains an important
treatment option for many men with erectile problems
unresponsive to less invasive approaches.
ESSENTIAL ANATOMY
The human penis has many design features that are
important for normal function and which the clinician
should be aware of, for optimal use of ICI therapy. In
contrast to the dog, where the two corporal bodies are
completely separated, in man they communicate freely
via an incomplete middle septum (Fig. 8.2). This fact
is essential for use with intracavernosal vasoactive
agents. Injection into a single side produces a rapid
bilateral increase in cavernous blood flow that creates
equalized pressure in both corpora, even in the face
of asymmetrical cavernous artery flow or unilateral
injury. The cavernous arteries are located asymmetri-
cally within the corpora, just lateral to the midline
septum, in a position that is unlikely to be injured by
the needle puncture. The surrounding tunica albuginea
is often a difcult anatomical structure to penetrate for
patients but can serve as an important landmark,
allowing them to recognize the proper depth of needle
advancement prior to drug delivery.
17,18
Deep to the tunica albuginea lie the cavernosal
smooth muscles, arranged in a series of potential spaces
or sinusoids lined by endothelium and separated by
trabeculae. Injection of the agent should be directly
into these open spaces, not into the muscle itself. A
misplaced needle can be easily recognized as a higher-
pressure injection or one that causes discomfort to the
patient.We routinely recommend that, if the drug does
not easily enter into the penis with minimal pressure,
the needle should be rotated; this usually dislodges the
beveled end from the trabeculae. The ideal injection
site is located at the lateral aspect of the shaft, away
from the dorsal nerve, artery, and vein.
122 MUSE and Intracavernosal Therapies
Figure 8.1 A cross sectional view of the human penis
depicting the extent of smooth muscle trabeculae located
below the fiberous tunica albuginea incomplete mid-line
section.
Figure 8.2 A longitudinal view of the penis with sites of
injection labelled.
*
*
*
*
*
*
The genius of ICI lies in its simplicity. Potentially, any
agent which induces an adequate degree of smooth-
muscle relaxation within the penis will be effective at
creating an erection. Neural stimulation of erection,
believed to be predominantly a non-adrenergic, non-
cholinergic phenomenon, plays no role in the efcacy
of injection therapy. Direct smooth-muscle relaxation
via altering the ion channels (Ca
2+
, K
+
) or through
accumulation of second-messenger molecules (decreasing
degradation or by direct activation) can produce an
erection. The neural-independent nature of ICI
allows for early use among postprostatectomy patients
as well as in the spinal cord-injured and diabetic
populations.
1921
The usual sequence of events when using intra-
cavernous agents is injection, sexual stimulation, and
erectile rigidity within 210 min mediated by a
dramatic increase in cavernous artery blood flow and
consequent compression of the subtunical venous out-
flow. Coincident with ejaculation and the attendant
increase in sympathetic tone, vasoconstriction occurs
with loss of penile rigidity. Use of penile constriction
bands or elastics, which were commonly recommended
early in the clinical experience with ICI, are now
largely of historical interest and believed to be of little
value.
22,23
THE AGENTS
Over the past two decades an extensive medical liter-
ature has developed around the use of intracavernous
vasoactive agents. A Medline search of intracavernous
penile injections, self-injection therapy for impotence,
ED and impotence reveals in excess of 1000 scientic
publications dealing with this topic in animal models
and human subjects in a variety of languages. Agents
which may provide enhanced efcacy and safety that
are currently undergoing clinical evaluations for
regulatory review will be presented in this chapter,
along with the limited number of agents approved for
intracavernous use.
Prostaglandin E
1
Independently, at the same impotence meeting in 1986,
both Adaikan
24
and Ishii
25,26
described the potential
for PGE-1 as an effective vasoactive agent for intra-
cavernosal use. It has gained widespread use and accept-
ance for ICI and intraurethral delivery worldwide.
Over the past 15 years a large number of clinical trials
and basic science evaluations with PGE-1 have been
undertaken and it represents the best studied of all the
drugs in current use.
PGE-1 is a prostanoid that has a variable impact on
vasulature throughout the systemic circulation. Within
the penis it upregulates the production of cyclic
adenosine monophosphate (cAMP) and leads to a
robust and rapid vasodilatation through alteration of
Ca
2+
ion channels. Additionally it is believed to have
an antiadrenergic effect, further enhancing its pro-
erectile function. It is a highly potent agent, requiring
only microgram amounts to induce this therapeutic
effect. Its small molecular size and ability to penetrate
the urethral mucosal layer made it the logical choice
for intraurethral delivery.
27,28
Injected PGE-1 acting on cAMP induces vaso-
dilatation mediated via gap junctions within the penis.
It is metabolized by a large rst-pass effect (90%) and,
to a smaller extent, by the liver and kidneys as well as
by local metabolism within the penis. Priapism, the
most devastating of the common adverse effects of ICI
therapy, is relatively low with PGE-1. Compilations
of recent reports describe rates of priapism between
1 and 3%. In our own experience these events are
frequently the result of rapid progression of dose by
the patient, missed initial injection, and a second
attempt or use among the young and/or neurogenic
patient. Priapism can be largely avoided through use of
a gradual and progressive dose escalation regime.
28
Our current protocol for use of PGE-1 is a starting
dose of 510 g in the vasculogenic male. In cases
where a normal vasculature is suspected or neurogenic-
based ED exists, a lower initial dose is recommended
2.5 g. After the rst injection associated with sexual
self-stimulation in the clinic, a higher or lower dose is
recommended based on response and duration of
action, with our goal of complete rigidity persisting for
1545 min. All men are informed of the technique
to optimize and increase their dose based on their
response at home. We generally recommend dose
increases of 2.55 g per attempt with a 24-h minimum
window between injections. Included in their initial
information is an explanation of priapism and how
likely they are to experience penile brosis or scarring.
All men are informed if brosis is detected on physical
examination at the start of treatment.
A vast literature exists describing efcacy and safety
of alprostadil across a wide spectrum of disease
processes and ages. Over the past 15 years since its
initial description for intracavernosal use, it has
become the dominant vasoactive agent. A wide range
The Agents 123
Box 8.2 Absolute and relative contraindications
for use
Previous priapism with vasoactive drug use
Severe penile brosis
Visual acuity which limits needle delivery
Monoamine oxidase inhibitors (would limit use of
phenylephrine for potential priapism)
of reported efcacy rates exists; however a reasonable
composite of these studies places PGE-1 intercourse
rates at 7075% among cohorts of men with mixed
etiologies, including diabetics, vasculopaths, and
situational ED. Most investigators report median doses
of 1215 g with common adverse events being
limited to bruising (common), priapism, and brosis at
13% (Boxes 8.2, 8.3 and 8.4).
2931
Recently some reports have described a potentially
important nding of increased incidence of spon-
taneous erections, disease modication, and improved
cavernous artery blood flow in men following long-
term use of PGE-1.
32
Although there exists animal
evidence reporting upregulation of the constitutive
form of nitric oxide synthase that supports these
clinical trials, much controversy continues, as there are
conflicting reports, which have failed to demonstrate
signicant objective evidence of improved nocturnal
erections. This exciting area probably requires further
studies to dene which patient populations are most
likely to experience genuine erectile enhancement
from vasoactive drug use. At present, we inform men
that ICI therapy results in a signicant minority of
men reporting improved spontaneous sexual function,
but few of these men reach the point of not needing
medication.
33,34
Among the largest groups of men considering ICI
and PGE-1 therapy are those who have failed rst-line
oral agents. Several recent reports have explored the
efcacy in this population specically. Shabsigh and
colleagues reported data from a large multicenter trial
using alprostadil alfadex among men who had failed
sildenal. Prior to entry into the study these men were
retried on sildenal at 100 mg and failed as assessed
by the International Index of Erectile Function (IIEF)
questionnaire. In 67 men, improvements in their ability
to achieve (Q3 of the IIEF) and maintain an erection
(Q4 of the IIEF) were observed in 89.6% and 85.1%
of men respectively. The investigators demonstrated a
signicant response rate among these sildenal non-
responders, indicating that progression to second-line
therapy is appropriate.
35
Discontinuation of treatment with intravenous
agents is high. Many long-term reports describe a drop-
out rate of 50%. Recently, Lehmann and coworkers
looked at a cohort of 86 men and found those most
likely to stop treatment are generally less motivated;
less satised with the treatment, and did not like the
drug-induced erection. Interestingly, side-effects such
as penile pain, brosis, and priapism were not a major
determining factor in choosing not to continue.
36
Wespes performed a novel histological review of his
penile biopsy specimens obtained from a small group
of men who had undergone between 150 and 250
PGE-1 injections in the 3-year period prior to
obtaining the sample. Among these men there was no
evidence of smooth-muscle alteration or excessive
brosis
37
(Box 8.5).
Intracavernous PGE-1 is frequently used as a
diagnostic test to assess vascular flow in men with ED.
A complete erection following injection indicates a
reasonable response to the vasoactive agent but
provides no information about the neural axis. A non-
response appears from work recently published by
Elhanbly and colleagues to indicate incompetent veno-
occlusive function and/or severe arterial insufciency.
38
Papaverine
Merck discovered papaverine in 1848: it is a
benzylquinoloine synthesized from tyrosine. Initially
124 MUSE and Intracavernosal Therapies
Box 8.5 Common steps to correct inadequate
therapeutic response
Review injection technique
Reassess dose and increase until a reasonable
therapeutic response is achieved
Evaluate timing of injection in relation to sexual
stimulation
Change to more potent agent, if at maximum
recommended dose
If pain is a limiting factor, use combination therapy
Involve partner and reassure
Box 8.3 Strategies to optimize intracavernous
therapy
Inject agent directly into the proximal corpora
Apply gentle local pressure for 2 min to injection
site
Sexual stimulation following injection
Comfortable, unstressful environment
If unsuccessful, repeat attempt at slightly higher
dose in 23 days time; increase dose until
recommended maximum achieved
Box 8.4 Common causes for inadequate
response to intracavernosal injection
Inadequate dose
Injection into wrong location (trabeculae,
subcutaneous)
Leakage of agent prior to injection
Inadequate sexual stimulation
Premature ejaculation
Stress
reported by Virag in 1982, this agent was in common
use in the early to mid-1980s until the description of
PGE-1. It acts via a non-specic phosphodiesterase
action inducing increased levels of second messengers.
Delivered as an intracavernous agent, doses between
5 and 160 mg have been reported. It is a convenient
vasoactive agent that is inexpensive, stable at room
temperature, and effective. A compilation of reports
yields a general efcacy of 60%. It has a high rate of
priapism (16%), elevated rate of penile brosis
compared to other agents (5.7%) in a study of 1056
cases, and has never been approved for this medical
indication, which limits its use as monotherapy. It is
metabolized in the liver and in its papaverine hydro-
chloride formulation is acidic, commonly producing
pain at injection
39,40
(Table 8.1).
Phentolamine
Phentolamine is primarily an alpha-adrenergic antag-
onist. Early animal studies have shown it is a highly
effective agent at blocking the epinephrine (adrenaline)
pressor response. It also has a smaller reported
sympatholytic effect. It produces a generalized direct
vasodilatation of muscle walled vessels independent of
size in all vascular regions.Although phentolamine has
been widely used as a component of the triple mix
(phentolamine, PGE-1, papaverine), its use, as mono-
therapy for ED, has been disappointing. Stief et al.
have reported an inadequate therapeutic response in
14 of 15 consecutive men injected with phentolamine
who had responded to a trimix preparation.
41,42
Papaverine, phenoxybenzamine, and phentolamine
were all reported to have proerectile effects well
before the rise of PGE-1 as the predominant vaso-
active agent. They continue to be used largely in com-
bination preparations as bimix or trimix compounded
drugs. The advantage of the bimix (papaverine and
phentolamine) as opposed to the trimix (papaverine,
PGE-1, and phentolamine) is its stability without
refrigeration. Although not approved in most western
countries by the local regulatory drug boards, these
drug combinations are widely available. Results of
multiple studies demonstrate a good efcacy and safety
record (Table 8.1).
YC-1 is a soluble guanylate cyclase (sGC) activator,
which has been shown in recent publications to
increase intracavernous pressure in rabbit and rat
penile preparations. Delivered by the intracavernous
route, it produced erections comparable to those seen
with direct cavernous nerve electrostimulation, working
via an NO-independent pathway. Activation of sGC
induces an increased concentration of cGMP with con-
sequent smooth-muscle relaxation. Clinical trials
remain in the future but this agent may provide a novel
means of inducing erection among those men refractory
to other therapies.
4345
Recently, use of combination therapy involving
PGE-1 acting on the cAMP second-messenger system
with NO spontaneously releasing compounds such as
sodium nitroprusside have been reported. This syner-
gistic impact may salvage some men who may other-
wise remain unresponsive.The potential for hypotension
with sodium nitroprusside remains to be determined in
further work.
46
MUSE
Using a novel approach to deliver high local
concentrations of vasoactive drug (PGE-1), MUSE is a
medicated urethral system for erection. This non-
needle system has been widely approved and in use
in most developed countries for the past 5 years. A
1.4 36 mm white pellet of PGE-1 is delivered into
The Agents 125
TABLE 8.1 Comparison of drug characteristics based on published reports.
Drug Dose Efcacy Priapism Fibrosis Increased Drop-out Satisfaction
(%) >6 hours (%) liver enzymes rate (%) rate (%)
(%) (%)
Papaverine 30110 mg 61 6.87.1 5.7 1.6 4.6 Not stated
Prostaglandin 540 g 72.6 0.250.36 0.8 0 37 9095
E
1
Pain 11.5
MUSE 2501000 g 3050 0 <1 0 Not stated 6771
Pain 30
Papaverine/ 15 mg/ 68.5 6 12.4 5.4 45 Not stated
phentolamine 1.25 mg60 mg/
2 mg
Adapted from Chapter 9 International consensus on ED Virag/Porst tables.
the urethra by a single-use disposable plastic appli-
cator placed into the distal urethra, which has been
moistened by a recent urination. Following placement
of the pellet, the patient is instructed to elevate the
penis to prevent the PGE-1 from falling out, and
massage of the glans and urethra for 25 min is
suggested. The composition of the pellet allows for a
rapid dissolution and delivery of the vasoactive agent
into the local penile circulation. Transfer from the
corpus spongiosum to the corpora cavernosa occurs
via retrograde venous flow.
47
Onset of a penile response occurs as early as 7 min
postdose with maximal engorgement recorded in some
reports by 2224 min. This is a time course similar to,
but slightly slower than, ICI. Duration of the effect of
drug is a dose-dependent phenomenon with the longest
duration noted at 1000 g, being 72 min from onset to
return to a non-erect state. In addition to the corpora
cavernosa becoming erect, the use of MUSE promotes
engorgement of the glans penis. This glans fullness is
frequently a noted complaint among the post penile
implant patient who wishes to have a fuller penis.
Studies have documented a potential role for this
combination therapy
48
(Box 8.6).
Early reports described a response rate of 65%,
which in subsequent clinical reports has been lowered
to roughly 30% of men with ED of mixed (organic and
situational) etiology. Problems of penile pain (32%),
urethral pain (12%), testicular pain (5%), and minor
urethral bleeding (5%) coupled with insufcient
erectile rigidity have limited its use. Compared to ICI,
MUSE has a lower incidence of priapism (<0.1%) and
cavernosal brosis (<1%).
Recent reports suggest that a 500 g starting dose
provides the greatest likelihood of success. Titration
upward or downward after the initial few attempts is
suggested. Rare hypotensive responses appear to be
dose-independent; however penile pain was reported
more commonly among the higher doses used (500
and 100 g).
49
MUSE remains an effective option for
treatment of neurogenic cases of ED, particularly
where some impairment of manual dexterity exists.
Unfortunately, it has proven to be less effective in men
with severe vascular disease and diabetes.
4952
THE FUTURE
Intracavernous delivery of vasoactive drugs remains an
essential part of the management of male ED. In the
near term, it likely will remain an important means of
salvaging those men who fail to respond to oral agents
or cannot take them out of concern for drug inter-
actions. In the future, however, use of direct injection
into the penile circulation to delivery gene therapy
and modify the ion channels may become a more
important reason to continue using this technique.
Ongoing investigations into the potential benet from
early ICI therapy to men following radical pelvic
surgery and the potential impact on cavernous artery
flow remain controversial but interesting areas of
study. This technique has been proven, through the
test of time, to be an effective and safe means of
restoring erectile function among patients who suffer
from a variety of comorbid conditions. New thera-
peutic agents able to provide greater efcacy, improved
patient tolerability, and rapid onset may make this
delivery approach more appealing in the years to come
(Box 8.7).
REFERENCES
1. Brindley G. Cavernosal alpha-blockage: a new technique
for investigating and treating erectile impotence. Br J
Psychiatry 1983; 143:332337.
2. Virag G, Intracavernous injection of papaverine for
erectile failure. Lancet 1982; ii:938.
3. Andersson KE, Wagner G. Physiology of penile erection.
Physiol Rev 1995; 75:191.
126 MUSE and Intracavernosal Therapies
Box 8.7 The future direction of local therapy
Administration of highly potent specic
vasoactive agents
Delivery of long-term pharmacological agents
altering smooth-muscle tone
Application of vascular endothelial growth factors
directly into the penile circulation
Neural enhancement agents
Delivery of gene-based erectile dysfunction
approaches:
Alteration of ion channels
Upregulation of guanylate cyclase
Increased synthesis of nitric oxide
Box 8.6 Strategies to optimize intraurethral
therapy
Urinate prior to placement of pellet
Advance device into proximal urethra gently
Deliver vasoactive agent
Check device to see that pellet has dislodged
Massage penis for 25 min in standing position
with penis slightly elevated
Sexual stimulation with partner
If unsuccessful at initial 500 g dose, increase
to 1000 g
4. Burnett AL. Nitric oxide in the penis: physiology and
pathology. J Urol 1997; 157:320.
5. Lue TF, Dahiya R. Molecular biology of erectile function
and dysfunction. Mol Urol 1997; 1:55.
6. Saenz De Tejada I, Cuevas P, Cuevas B et al. Nitrosylated-
adrenergic receptor antagonists as a potential drugs for the
treatment of erectile dysfunction (ED). AUA Abstract 1998.
7. Vick RN, Benevides M, Patel M et al. The efcacy, safety
and tolerability of intracavernous PNU-83757 for the treat-
ment of erectile dysfunction. J Urol 2002; 167:26182623.
8. Sazova O, Kadioglu A, Gurkan L et al. Intracavernous
administration of SIN-1 + VIP in an in vivo rabbit model for
erectile function. Int J Impot Res 2002; 14:44492.
9. Mizusawa H, Hedlund P, Brioni JD et al. Nitric oxide
independent activation of guanylate cyclase by YC-1 causes
erectile responses in the rat. J Urol 2002; 167:22762281.
10. Linet OI, Ogrinc FG. Efcacy and safety of intracavernosal
alprostadil in men with erectile dysfunction. The alprostadil
study group. N Engl J Med 1996; 334:873877.
11. Virag R, Becher E, Carrier S et al. Local pharmacological
treatment modalities. In: Erectile dysfunction; the rst inter-
national consensus consultation on erectile dysfunction.
1999: 305354.
12. IMS prescription tracking data 2001.
13. The Canadian Urology Association Guidelines Committee
Report on Management of Erectile Dysfunction. Can J Urol
2002; (in press).
14. Montorsi F, Guazzoni G, Strambi LF et al. Recovery of
spontaneous erectile function after nerve-sparing radical
retropubic prostatectomy with and without early intra-
cavernous injections of alprostadil: results of a prospective,
randomized trial. J Urol 1997; 158:14081410.
15. Escrig A, Marin R, Mas M. Repeated PGE-1 treatment
enhances nitric oxide and erection responses to nerve
stimulation in the rat penis by upregulating constutive NOS
isoforms. J Urol 1999; 162:22052210.
16. Christ GJ, Rehman J, Day N et al. Intracorporal injection
of hSlo cDNA in rats produces physiologic relevant alteration
in penile function. Am J Physiol 1998; 275:H600.
17. deGroat WC, Steers WD. Neuroanatomy and neuro-
physiology of penile erection. In: Tanagho EA, Lue TF,
McClure RD, eds. Contemporary management of impotence
and infertility. Baltimore: Williams & Wilkins; 1988:327.
18. Hsu GL, Brock GB, Martinez-Pineiro L et al. The three
dimensional structure of the human tunica albuginea:
anatomical and ultrastructural levels. Int J. Impot Res 1992;
4:117129.
19. Ismail M, Abbott L, Hirsch IH. Experience with intra-
cavernous PGE-1 in the treatment of erectile dysfunction: dose
considerations and efcacy. Int J Impot Res 1997; 1:3942.
20. Richter S, Vardi Y, Ringel A et al. Intracavernous
injections: still the gold standard for treatment of erectile dys-
function in elderly men. Int J Impot Res 2001; 13:172175.
21. Heaton JP, Lording D, Liu SN et al. Intracavernosal
alprostadil is effective for the treatment of erectile dysfunction
in diabetic men. Int J Impot Res 2001; 13:317321.
22. Brock GB, Breza J, Lue TF: High-flow arterial priapism:
a spectrum of disease. J Urol 1993; 150:968-971.
23. Ledda A. Erectile dysfunction: intracavernous treatment.
Curr Med Res Opin 2000; 16:S59S62.
24. Adaikan PG, Kottegoda SR, Ratnam SS. A possible role
for PGE-1 in human erection. In: Abstract book of the second
world meeting on impotence. Prague, Czechosolakia: 1986.
25. Ishii N, Watanabe H, Irisawa C et al. Therapeutic trial
with PGE-1 for organic impotence. In: Abstract book of the
second world meeting on impotence. Prague, Czechosolakia:
1986.
26. Ishii N, Watanabe H, Irisawa C et al. Intracavernous
injection of prostaglandin E-1 for the treatment of erectile
dysfunction. J Urol 1989; 141:323325.
27. Choi HK, Adimoelja A, Kim SC et al. A doseresponse
study of alprostadil sterile powder (S.Po) (Caverject) for the
treatment of erectile dysfunction in Korean and Indonesian
men. Int J Impot Res 1997; 1: 4751.
28. Cawello W, Schweer H, Dietrich B et al. Pharmacokinetics
of prostaglandin E-1 and its main metabolites after intra-
cavernous injection and short term infusion of prostaglandin
E-1 in patients with erectile dysfunction. J Urol 1997;
158:14031407.
29. Kunelius P, Lukkarinen O. Intracavernous self-injection
of prostaglandin E-1 in the treatment of erectile dysfunction.
Int J Impot Res 1999; 11:2124.
30. Porst H, Buvat J, Meuleman E et al. Intracavernous
alprostadil alfadex an effective and well tolerated treatment
for erectile dysfunction. Results of a long-term European
study. Int J Impot Res 1998; 10:225231.
31. Hauck EW, Altinkilic BM, Schroder-Printzen I et al.
Prostaglandin E1 long term self-injection programme for
treatment of erectile dysfunction a follow up of at least
5 years. Andrologia 1999; 31:S99S103.
32. Maniam P, Seftel AD, Corty EW et al. Nocturnal penile
tumescence activity unchanged after long-term intracavernous
injection therapy. J Urol 2001; 165:830832.
33. Wespes E, Sattar AA, Noel JC et al. Does prostaglandin
E-1 therapy modify the intracavernous musculature? J Urol
2000; 163:464466.
34. Brock G, Linet OI. Return of spontaneous erection
during long-term intracavernosal alprostadil (Caverject)
treatment. Urology 2001; 57:536541.
35. Shabsigh R, Padma-Nathan H, Gittleman M et al. Intra-
cavernous alprostadil alfadex (Edex/Viridal) is effective and
safe in patients with erectile dysfunction after failing sildenal.
Urology 2000; 55:477480.
36. Lehmann K, Casella R, Blochlinger A and et al. Reasons
for discontinuing intracavernous injection therapy with
prostaglandin E-1 (alprostadil). Urology 1999; 53:397400.
References 127
37. Wespes E. Smooth muscle pathology and erectile dys-
function. Int J Impot Res 2002; 14:S17S21.
38. Elhanbly S, Schoor R, Elmogy M et al.What nonresponse
to intracavernous injection really indicates: a determination
by quantitative analysis. J Urol 2002; 167:192196.
39. Earle CM, Keogh EJ, Wisniewski ZS et al. Prostaglandin
E-1 therapy for impotence, comparison with papaverine. J Urol
1990; 143:5759.
40. Sahin M, Basar MM, Bozdogan O et al. Short-term histo-
pathologic effects of different intracavernous agents on corpus
cavernosum and antibrotic activity of intracavernosal
verapamil: an experimental study. Urology 2001; 58:487492.
41. Stief CG, Wetterauer U. Erectile responses to intra-
cavernous papaverine and phentolamine administration in
patients with neuropathic pain. Anesth Analg 1993;
76:10081011.
42. Brock GB. Oral phentolamine (Vasomax). Drugs Today
2000; 36:121124.
43. Brioni JD, Nakane M , Hsieh GC et al. Activators of
soluble guanylate cyclase for the treatment of male erectile
dysfunction. Int J Impot Res 2002; 14:814.
44. Nakane M, Hsieh G, Miller LN et al.Activation of soluble
guanylate cyclase causes relaxation of corpus cavernosum
tissue: synergism of nitric oxide and YC-1. Int J Impot Res
2002; 14:121127.
45. Stone JR, Marletta MA. Synergistic activation of soluble
guanylate cyclase by YC-1 and carbon monoxide: implications
for the cleavage of the ironhistidine bond during activation
of nitric oxide. Chem Biol 1998; 5:255261.
46. Martinez-Pinerio L, Cortes R , Cuervo E et al.
Prospective comparative study with intracavernous sodium
nitroprusside and prostaglandin E-1 in patients with erectile
dysfunction. Eur Urol 1998; 4:350354.
47. Hellstrom WJ, Bennett AH, Gesundheit N et al. A double
blind placebo controlled evaluation of the erectile response to
transurethral alprostadil. Urology 1996; 48:851856.
48. Padma-Nathan H, Hellstrom WJ, Kaiser FE et al.
Treatment of men with erectile dysfunction with transurethral
alprostadil. Medicated urethral system for erection (MUSE)
study group. N Engl J Med 1997; 336:17.
49. Ekman P, Sjogren L, Englund G et al. Optimizing the
therapeutic approach of transurethral alprostadil. Br J Urol
2000; 86:6874.
50. Werthman P, Rajfer J. MUSE therapy: preliminary clinical
observations. Urology 1997; 50:809811.
51. Williams G, Abbou CC, Amar ET et al. The effect of
transurethral alprostadil on the quality of life of men with
erectile dysfunction, and their partners. MUSE Study Group.
Br J Urol 1998; 82:847854.
52. Fulgham PF, Cochran JS, Denman JL et al. Disappointing
initial results with transurethral aplrostadil for erectile
dysfunction in a urology practice setting. J Urol 1998;
160:20412046.
128 MUSE and Intracavernosal Therapies
VACUUM DEVICES
Epidemiology (history)
In 1974, Geddings D. Osbon Sr., a businessman from
Augusta, Georgia, formed a company to market the rst
commercial vacuum constriction therapy for erections.
He was later awarded a US patent for this device,
which he had originally intended for his personal
use.
1,2
This invention, as well as those of a few other
inventors, was based on John Kings report in 1874 of
applying negative pressure for obtaining an erection as
well as the more detailed description by Otto Lederer,
for which the US patent ofce had issued a patent in
1917.
36
Despite the positive inroads, acceptance of the
vacuum erection device (VED) was far from universal
in its initial days. The company that was originally
started by Osbon (the Youth Equivalent Company)
had to challenge the US Postal Service in court when
it classied the companys literature on erectile dys-
function (ED) as pornography and ordered the company
to cease and desist. After overcoming this initial
hurdle, the company was again ordered to close its
doors after the Food and Drug Administration (FDA)
classied the VED as a medical device rather than a
marital aid. In 1982, the FDA granted permission for
marketing the device after it was ruled that all federal
guidelines had been satised.The rst scientic presen-
tations of the vacuum device were made by Drs Roy
Witherington and Perry Nadig, who later published on
the subject and made inroads in the acceptance of this
mechanical device in the medical community.
79
The
American Urological Association Clinical Guidelines
Panel on the treatment of organic ED listed the VEDs
as a major treatment alternative in 1996.
10
Mechanism of action and indications
The devices offered in the USA are manufactured by a
variety of companies and may be manual or battery-
operated (Fig. 9.1).The main components are the pump,
cylinder, and constriction rings to decrease venous
outflow. Many of the currently marketed devices fea-
ture a safety mechanism whereby pressures in excess
of 300350 mmHg may not be achieved. The device is
assembled by rst placing the constriction rings on to
the proximal end of the apparatus and subsequently
connecting the pump to the distal cylinder. After
lubricating the proximal open end for a better seal
around the base of the penis, negative pressure is
obtained by manual or automatic (battery) activation
of the pump and simultaneous application of pressure
on the cylinder toward the pubic area.
It has been shown that the increase in penile volume
during application of the VED is caused not only by
arterial inflow but also by venous backflow.
11
The
preplaced constriction rings on the proximal aspect of
the device are glided on to the penis once an adequate
negative pressure (and hence penile rigidity) has been
reached. Application of the constriction ring causes a
relative ischemia in the penis 30 min after application
of the constriction ring, although no study has demon-
strated related specic adverse long-term effects.
11
Indications for the use of VEDs are broad and
include all categories of ED. VEDs may be combined
with other modalities for the treatment of ED, such as
intracavernous pharmacotherapy.
12
Other investigators
have reported variable degrees of success in cases of
venoocclusive dysfunction.
13
Signicant success has also been reported in more
difcult patient populations, including those with veno-
occlusive disorders and explanted penile prostheses.
In 1989, Moul and McLeod
14
described VED use in
patients who had undergone prosthesis explantation
and concluded that VEDs may be a useful therapy for
ED in the challenging explant population despite a
history of corporeal infection and presumed brosis.
Others have reported successful use of the VED in
spinal cord injury patients, with 41% of the men still
satised with the device at 6-month follow-up.
15
Lue
and El-Sakka
16
recently reported on chronic inter-
mittent stretching with a VED after circumferential
tunical incision and circular venous grafting in 4
patients with penile shortening from severe Peyronies
disease. Although the study evaluated only a very
small cohort, all patients were satised and reported
improved psychological well-being as well as relation-
ships with partners.
In a study of the safety and efcacy of the VED in
patients with corporal venoocclusive disease (CVOD),
Vacuum Devices and Penile Implants
Hossein Sadeghi-Nejad and Allen D. Seftel
CHAPTER 9
the authors found no relationship between the severity
of disease and the rating of erection or satisfaction
with the device and recommended an initial trial of
VED regardless of the degree of CVOD. They reported
success rates similar to intracavernosal injection (ICI)
therapy.
17
In another study, satisfactory results were obtained
in 69% of patients with venous leakage based on a
questionnaire evaluation.
13
These encouraging results,
however, must be weighed against those showing lack
of efcacy in more severe cases of ED. A study of the
use, efcacy, and acceptance of the VED among 60
impotent men not satised with ICI therapy revealed
that more than 80% of those studied abandoned the
VED due to lack of efcacy. The authors concluded
that, in a group of men who have failed intracavernous
injection therapy, VED is not likely to be highly efca-
cious or widely accepted.
18
Economics
Tan
19
examined the economic cost of ED for a hypo-
thetical managed-care model based on 1998 US dollars
and concluded that, from a purely economic stand-
point, sildenal and the VED should be considered as
rst-line management strategies whereas ICI therapy,
transurethral alprostadil suppository, and penile
prosthesis implant should be reserved for second- or
third-line therapy. Overall, VED was found to be the
least costly alternative and, since at the time of publi-
cation in 2000 VED was the only treatment covered
under Medicare part B, with a 20% co-payment, Tan
concluded that VED may be the most affordable choice
to consider for seniors who live on a xed monthly
income.
Complications
The VED is usually well tolerated and free of major
complications when used correctly. Minor side-effects
are common, but most studies have not found these
to be a major deterrent. Contraindications to the use
of vacuum therapy are few and primarily include
patients with unexplained intermittent priapism and
bleeding disorders. Side-effects such as occasional
numbness, pain, penile bruising, or petechiae have a
low incidence.
1
Penile necrosis has been documented in at least one
case report with frequent and prolonged use of the
device in spinal cord injury patients.
20
It is therefore
recommended that device use be restricted to once
per day and constriction band application limited to a
maximum of 30 min.
Other unusual complications include severe urethral
bleeding in a diabetic patient, development of a penile
130 Vacuum Devices and Penile Implants
Figure 9.1ac Manual and battery-operated vacuum
erection devices with constriction bands. (Courtesy of
Endo Care.)
A
B
C
cystic mass that was seen only with VED use, but not
during the flaccid state (later found to be scrotal tunica
vaginalis!), and dorsal penile plaque formation after
4 years of VED use.
21
Patient satisfaction, future outcomes
and the effects of oral therapies
Dutta and Eid
22
evaluated the satisfaction rate,
attrition rate, and follow-up treatment of well-trained
patients with organic ED using an external VED. The
overall drop-out rate was 65% and was lowest among
patients with moderate ED (55%). One hundred per
cent of those with mild dysfunction discontinued use,
and a large number (70%) of patients with complete
dysfunction also discontinued use. Of the patients who
discontinued, the majority stopped treatment early
(median 1 month, mean 4 months) and 63% sought no
further treatment. Only 35% of patients were satised
with the device and continued long-term use (mean
37 months).
22
Cookson and Nadig
23
report the long-
term drop-out rate for VED to be 30% while Turner
et al.
24
report a 19% discontinuation rate for the rst
6 months of treatment.
A study of 61 impotent men who participated in a
clinical trial indicated that, among 32 patients with
arteriogenic impotence, 28 (88%) had satisfactory
results and had an improvement in their capacity for
spontaneous erections with the device.
25
Soderhal
et al.
26
compared the Osbon ErecAid system to ICI
therapy in relation to satisfaction, effectiveness, and
side-effects. Of the 44 patients who completed the
study, the ability to attain orgasm and the overall
satisfaction of the patient and partner with the sexual
experience were signicantly better when using
injections. In this report, those with a shorter duration
of impotence (<12 months) and those impotent
secondary to radical prostatectomy strongly favored
injection therapy.
Chen et al.
27
evaluated the preference of patients
with ED who had been effectively treated with a VED
and then switched to sildenal. Of the 36 participants
in whom the efcacy of sildenal was similar to that of
a VED, 12 (33.3%) decided to resume use of a VED
while 24 (66.6%) preferred to continue sildenal.There
were no statistically signicant differences between
the groups regarding patient age or the etiology and
duration of ED. The adverse side-effects of sildenal
were the main reasons for preferring a VED. Fewer
ejaculatory difculties, efcacy, comfort, and ease of
use were the main reasons for choosing sildenal. This
investigation revealed that a substantial number of
patients who respond to both oral pharmacotherapies
and the VED may choose the latter as their preferred
treatment modality.
PENILE IMPLANTS
Historical perspective
Following attempts at penile implant surgery using rib
cartilage and bone in the 1930s by Nicolai Bogoras,
Goodwin and Scott as well as Loeffler and Sayegh
described the use of acrylic implants for the treatment
of impotence.
2830
Similar to Pearmans Silastic rod,
proposed in 1967,
31
these devices were not implanted
intracavernosally and were not well received due to a
high erosion rate, lymphatic edema, glans irritability,
and glans slippage over the prosthesis. The use of
silicone rubber as a material for implantation was rst
suggested by Lash in 1968
32
in the plastic surgery
literature and later proposed as a device for penile
prosthesis surgery. A major improvement was reported
by Beheri in 1966
33
when he published his series of
700 intracavernosal polyethylene prostheses placed
through a midline dorsal penile incision followed by
tunical incision and corporal dilation. Morales et al.
34
described a relatively similar technique and prosthesis
in 1973. However, because of the rigidity of the device
and its rm, narrow proximal end, penile pain as
well as crural or midshaft (septal) perforations were
reported.
The next milestone in penile prosthesis surgery
was the description in 1973 by Scott et al. of intra-
cavernosal, inflatable silicone cylinders.
35
Two years
later, Small published a paper on the implantation
of the SmallCarrion prosthesis in 31 patients and
reported favorable results in 28.
30
The author described
a much easier surgical implantation technique through
a perineal surgical approach with a pair of sponge-
lled silicone prostheses and later reported a more
extensive follow-up on 160 cases.
36
Although less
extensive surgery and lower chances of mechanical
failure due to absence of hydraulic and mechanical
components were cited as advantages, it was conceded
that the Scott prosthesis, which was based on the
mechanisms of an implantable fluid transfer system
developed by Kothari et al.,
37
had excellent potential
and more closely approximated the natural physiologic
state. The permanent erectile state problem was
overcome by Finney
38
and Subrini and Couvelaire,
39
who independently introduced the concept of a hinged
device and soft silicone in the middle of the implant.
In 1986, Dr. Robert Krane reported on the
Omniphase, which was composed of a pair of silicone
rods that were able to move back and forth due to the
spring-like properties of its central cable.
40
Although
the Omniphase and its successor, the Duraphase, were
popular and combined the desirable properties of easy
concealment, rigidity for intercourse, and a straight-
forward surgical technique, they were handicapped
by relatively frequent cable failure and the need for
replacement.
41
Penile Implants 131
The improved American Medical Systems (AMS)
model in 1983 incorporated a three-layer design that
included a woven fabric layer between two silicone
layers. Although this change improved the aneurysmal
dilation problems, it limited the ability for girth
expansion.
42
Yet another change for the AMS device in
1987 came in the form of substituting polypropylene
for the fabric and the launch of the CX (controlled
expansion) device which allowed cylinder expansion.
The Mentor three-piece inflatable prosthesis is made
of Bioflex, a polyurethane material that has the advan-
tage of resisting aneurysmal dilation. First introduced
in 1983, the Bioflex material used in the Mentor
device offered signicantly higher durability than the
existing silicone cylinders. None the less, the early
versions suffered a high number of mechanical failures
in the silicone tubing.
43
Additional reinforcement of
the silicone tubing resulted in a 93% 5-year survival
from revision for mechanical reasons in the enhanced
Mentor Alpha-1 prosthesis, as reported by Wilson et al.
in 1999.
44
Unitary hydraulic devices consist of a non-disten-
sible inner chamber, which becomes rigid when lled.
These devices were ingenious in their design concept
and combined the cosmetic advantages of inflatable
multicomponent devices with the implantation ease of
semirigid implants. Unfortunately, both the Surgitek
(Flexi-flate) and AMS (Hydroflex/Dynaflex) devices
had unacceptable mechanical malfunction or patient
dissatisfaction rates and were discontinued.
45
Indications
Penile prostheses are indicated for the treatment of
organic ED due to a variety of causes. Most surgeons
recommend a trial of less invasive modalities, including
oral pharmacotherapy or ICI, before moving on to
prosthesis surgery. Despite many surgeons fear of
increased complications in radiated patients, it has
been shown that penile prosthesis surgery can be safely
and effectively performed after radiation therapy for
prostate cancer with minimal intraoperative and post-
operative adverse events.
46
Similarly, there is a denite
role for penile prostheses in the management of ED
that is refractory to conservative measures following
radical retropubic prostatectomy and some investi-
gators have advocated immediate sexual rehabilitation
by simultaneous placement of a penile prosthesis in
patients undergoing radical prostatectomy with no
apparent increase in morbidity.
47
Experienced implant
surgeons have reported successful prosthesis place-
ment in severely brotic corpora by utilizing a variety
of techniques and cavernotomes.
4851
Penile modeling over an inflatable implant was rst
described by Wilson and Delk in 1994
52
and has since
been accepted as an effective therapeutic option for
patient with simultaneous Peyronies disease and
ED.
53,54
Levine and Dimitriou
55
developed an algorithm
for surgical management and placement of penile
prostheses in this group of patients. They attempted
manual molding initially, followed by tunica incision
for insufcient straightening. For tunical defects greater
than 2 cm, polytetrafluoroethylene (PTFE) patch
grafting was performed to prevent prosthesis cylinder
herniation and recurrent deformity from cicatrix con-
traction. Full erectile capacity with a straight phallus
was achieved in all patients.
Prosthesis types
Semirigid/rod prostheses
These prostheses are manufactured by the AMS
(Malleable 650), Mentor (Acu-Form and Malleable)
and Timm Medical Technologies (Dura II) and are
constructed of two solid prostheses that are indepen-
dently placed in each corpus cavernosum. They are
ideal for patients in whom the cosmetic advantages of
the inflatable devices are not as important as the ease
of use and the lower chances of mechanical failure in
semirigid implants. Spinal cord-injured patients who
may not accept or respond to ICI therapy or those who
require external condom drainage for incontinence are
also candidates for these prostheses.
56
Another group
well served by the semirigid devices (or a two-piece
rather than a three-piece inflatable device) is pelvic
organ transplant recipients in whom traditional con-
servative therapy for ED has failed. Three-piece
prostheses should be avoided in this group because of
a signicantly higher incidence or reservoir compli-
cations in the retroperitoneal space in these patients.
57
The semirigid devices are typically made of pure
silicone rubber (e.g., the Mentor and the AMS devices)
which may be wrapped around a central coiled wire or
have a core construction of articulating segments with
metallic cables running through them (e.g., the Dura II
device). The PTFE-coated rings are interlocked and
connected by a spring-loaded cable which can lock the
rings in a straight column when activated and unlock
for a relaxed flaccid state.
The Dura II is a third-generation derivative of the
Omniphase (Dacomed, Minneapolis, MN, USA) pros-
thesis discussed in the historical review. Further
improvements achieved in the latest-generation Dura
II included substitution of the more durable high-
molecular-weight polyethylene for polysulfone and
strengthening of the cable strands (Fig. 9.2).
The current device is offered in 10 and 12 mm widths
and a 13 cm length which can be individually tailored
with distal tips of varying sizes. (image) Because of its
superb flexibility and ease of operation, the device is
particularly well suited to those with poor manual
dexterity. Conversely, it is a relatively poor choice for
132 Vacuum Devices and Penile Implants
those with a large-diameter penis or a very short penis,
since the smallest device is 15 cm long.
41
The Acuform and Malleable are manufactured by the
Mentor Corporation (Santa Barbara, CA: Fig. 9.3). The
former device has an outer helical wire surrounding a
silver wire core in the distal end and has greater
flexibility than the Malleable. The company offers a
lifetime warranty for mechanical failure.These implants
are available in three diameters (9, 11, 13 mm) with
standard, 0.5 cm and 1.0 cm tail caps and may be
trimmed using a standard #10 scalpel blade to 30
various sizes (the caps are tted on to the trimmed
ends).
The AMS 600 was introduced in 1983 and had a
twisted stainless-steel wire surrounded by solid
silicone. Despite excellent durability, the device had
signicant springback and was redesigned as the AMS
650 (Fig. 9.4). The improved prosthesis featured ner
strands in a spiral conguration in a polyester covering
and further encased in a solid silicone body.
41
This
prosthesis has been marketed since 1996 and is avail-
able in 13 or 11 mm girths and 12, 16, or 20 cm lengths
that are further adjustable with rear tip extenders.
Semirigid prostheses may be placed through an
infrapubic or penoscrotal incision, as described below,
with the caveat that the corporotomy incisions are
slightly larger than those used for the inflatable
cylinders. Excessive bending of the device during place-
ment through a small corporotomy may damage the
cylinders and cause poor rigidity.
58
Alternatively, the
semirigid devices may be placed through a limited sub-
coronal incision.
59
Corporal and urethral complications
encountered intraoperatively are similar to those
described in more detail in the inflatable prostheses
section, below. Other reported complications include
infection, prolonged pain, mechanical failure, and
erosion. Erosion is more frequently encountered in
spinal cord injury patients compared to the general
population (11% versus 1%).
60
Erosion of a malleable
penile prosthesis into bladder has been reported in a
patient with spinal cord injury and recurrent urinary
tract infections.
61
Penile Implants 133
Figure 9.2 Dura II semirigid penile prosthesis.
(Courtesy of Dacomed.)
Figure 9.3 Acuform semirigid penile prosthesis.
(Courtesy of Mentor.)
Figure 9.4 AMS 650 semirigid prosthesis.
(Courtesy of AMS.)
Inflatable prostheses
Unitary self-contained cylinders
The Hydroflex prosthesis was introduced in 1985
(AMS) and later succeeded by the Dynaflex. These
prostheses combined the easy implantability of the rod
prostheses with the cosmetic advantages of inflatable
devices.
62,63
Despite the elegant simplicity of design,
inferior mechanical reliability and patient satisfaction
compared to the multicomponent devices resulted in a
gradual phasing-out of the unitary devices. Wilson and
Delk have stated that the combination of semirigidity
and ability to partially inflate acted as a tissue
expander by compressing corporal tissue.
42
Over
time, this resulted in an inadequate size for a complete
rigid erection.
Two- and three-piece multicomponent
prostheses
In the USA, the multicomponent inflatable devices
are made by AMS and Mentor. Currently marketed
devices in this category by AMS are the AMS 700 CX
and CXM (smaller components than the CX; useful for
implantation in brotic corpora), the AMS 700 Ultrex
and Ultrex Plus, and the Ambicor, which is a two-piece
device with a combination scrotal pump/reservoir.
The Ambicor is currently the only marketed two-piece
inflatable prosthesis in the USA. Although its small
scrotal pump is unable to produce girth expansion,
activating the pump transfers fluid from the proximal
to the distal portion of the cylinders to obtain adequate
rigidity (Fig. 9.5).
The AMS and Mentor three-piece prostheses share a
similar general structural design: an intraabdominal
fluid reservoir to be placed in the perivesical space, a
pair of cylinders for intracavernosal implantation, a
scrotal pump for fluid transfer between the reservoir
and the cylinders, and silicone tubing for connecting
these components.
The Ultrex was introduced in 1990 and was designed
to provide combined girth and length expansion.
64
There was a higher incidence of cylinder failure with
the Ultrex as compared with the same companys CX
device and the 5-year survival from mechanical revision
was reported to be as low as 66%.
65
However, a recent
report indicates that cylinder modication in 1993
appeared to have signicantly decreased the propensity
of cylinder failure of the premodication device.
66
Additional reported problems included buckling and
accelerated wear of the Ultrex cylinders due to limi-
tations of length expansion after the natural process of
capsule formation around the cylinders.
42
Montorsi et
al. have also corroborated these ndings and reported
that in the long-term evaluation of cylinders, the CX
appears to be more mechanically reliable than the
Ultrex.
67
The AMS 700CXM with medium controlled expan-
sion cylinders was introduced in 1990 to provide an
inflatable prosthesis with controlled expansion in girth
and tness for Asian men.
68
The CX and CXM have
recently been marketed with an antibiotic coating
(inhibizone: Fig. 9.6). Long-term data regarding infec-
tion rates with the new antibiotic-coated device are
not available at the time of this writing.
134 Vacuum Devices and Penile Implants
Figure 9.5 Ambicor inflatable penile prosthesis.
(Courtesy of AMS.)
Figure 9.6 Antibiotic-coated inflatable AMS prosthesis.
(Courtesy of AMS.)
Mentor limits its inflatable products to the Mentor
Alpha-1 and the narrow-base version of the same
prosthesis, which is particularly well-suited for difcult
repeat implantations or postradiation surgery in brotic
corpora. Both of the Mentor three-piece prostheses are
now available with the recently introduced lock-out
valve, a mechanical enhancement of the scrotal pump
designed to inhibit undesired spontaneous auto-
inflation of the cylinders (Fig. 9.7).
Perioperative care
Adherence to strict infection control protocol, including
antibiotic prophylaxis, intraoperative shaving, careful
coverage of ostomy sites with an Ioband adhesive
layer, minimization of operating room trafc during
the procedure, 10-min antibacterial scrubbing of the
operative site with iodophore prior to the iodophore
paint skin preparation, double-gloving of all involved
personnel, and an antibacterial shower the night or
morning before surgery will reduce the incidence of
infectious complications. The presence of urinary tract
infection or any other infection at the time of surgery
is cause for rescheduling the procedure. Antibiotic pro-
phylaxis should be administered 12 h prior to surgery.
Agents effective against the most common organisms,
Staphylococcus epidermidis and Escherichia coli,
must be chosen and may include an aminoglycoside
and vancomycin or a rst-generation cephalosporin.
Fluoroquinolones have also been shown to be equally
effective against these organisms and may be
administered orally.
69
The decision to perform the procedure in an
outpatient or inpatient setting is governed by surgeon
and patient preference as well as the inescapable
realities of managed-care and insurance guidelines in
some areas. Garber has reported on successful penile
prosthesis placement on an outpatient basis with no
apparent increase in morbidity except for a 4% urinary
retention rate that was effectively managed by over-
night catheterization.
70
When the patient is kept over-
night, the Foley catheter may be left indwelling until
the following morning. Oral antibiotics (cephalosporins
or fluoroquinolones) are continued for 710 days and
the patient is instructed to refrain from heavy lifting or
using the device for 46 weeks. It is also important to
Penile Implants 135
Figure 9.7a Hydrophilic-coated Mentor Alpha-1 inflatable
penile prosthesis with lock-out valve. (Courtesy of Mentor.)
b Detail of the lock-out valve mechanism for prevention of
autoinflation. (Courtesy of Mentor.)
c Detail of the pump and release bar mechanism for the
Mentor Alpha-1 inflatable penile prosthesis. (Courtesy of
Mentor.)
A
B
C
keep the reservoir full and the cylinders deflated most
of the time during the initial 8-week period until
capsule formation around the reservoir is complete. If
the capsule forms around a deflated reservoir, auto-
inflation or difculty deflating the cylinders may be
experienced. Instructional brochures and videos on
inflation/deflation techniques are an important
educational aid for the implant recipient and may be
viewed during the healing period.
Surgical aproaches
Both inflatable and semirigid devices may be implanted
through an infrapubic or penoscrotal incision. The
choice of incision is based on surgeon preference and
the individual patients special needs and body habitus.
Comparison of infection rates shows no statistically
signicant difference between the two incisions.
71
In the infrapubic approach popularized by Furlow,
the cylinders are placed through an incision in the space
between the pubic bone and the penis. Dissection to
the level of the tunica albuginea and reservoir place-
ment in the perivesical space are relatively simple in
non-obese patients and require minimal dissection
with excellent exposure. Following midline separation
of the rectus muscles, the reservoir is placed in a
subrectus pouch under direct vision. When dissecting
on to the surface of the shiny white tunica albuginea
covering the corpora, it is critical to identify and
preserve the dorsal nerves, which are typically found
23 mm lateral to the deep dorsal vein. Absorbable
stay sutures (2-0) are placed on to the tunica and a
corporotomy is performed with a #15 scalpel blade.
It is important to avoid cautery use to minimize the
chances of nerve injury. After gentle introduction of
Metzenbaum scissors into the corpora for determi-
nation of the corporal course prior to dilation, Brooks
or Hagar dilators may be used (up to size 14) to expand
the intracavernosal space both proximally and distally.
Proximal and distal measurements are made using the
Furlow passer or the Dilamezinsert to the level of the
preplaced stay sutures. The cylinders are pulled into
their proper corporal position using the Furlow passer.
The corpora are then closed with the preplaced 2-0
sutures. Pump placement in the dependent scrotum is
performed through the same incision. After connecting
the tubing with the provided tools and connectors,
the prosthesis is once again inflated at the end of the
case to insure proper mechanical function as well as
symmetry, rigidity, and absence of leakage.
Proximal exposure (often critical in repeat implan-
tations or brotic corpora) is also limited when using
the infrapubic approach. In the authors opinion, the
added disadvantage of possible irreversible dorsal
nerve injury makes this incision less desirable than the
penoscrotal incision except in cases where blind
reservoir placement (as in the penoscrotal approach) is
truly complicated or risky (as in a patient with an
orthotopic intestinal bladder).
The penoscrotal incision allows for superb corporal
exposure throughout the corporal length and may be
vertical, longitudinal, or transverse. A transverse
incision is preferred when optimal proximal corporal
exposure (as in brotic cases) is desired. It may also be
used for combined articial urethral sphincter and
inflatable penile prosthesis placement when refractory
incontinence and ED coexist (as in a postradical
prostatectomy patient).
In the vertical midline approach, after Foley
catheterization, a 2.55 cm incision is made in the
midline raphe through the skin and the dartos layer.
Popularized by Wilson, this incision is then pulled up
on to the penis and secured in position by skin hooks
that are symmetrically attached to a Scott retractor.
After skin hook placement for exposure and dis-
section through the tissue layers on to the unmis-
takable shiny white surface of the tunica albuginea,
2-0 absorbable stay sutures (CT2 needle; detachable)
are placed into the tunica in a medial and lateral
position along the long axis of the right or left corpus
approximately 1 cm lateral to the urethra. The tunica
albuginea is then incised with a #15 scalpel blade and
further stay sutures are placed on each side of the
incision proximally.The extended incision will allow for
less tubing travel alongside the cylinders and provide
additional length for optimal pump positioning in the
dependent scrotum. Careful insertion of Metzenbaum
scissors (laterally pointed, to avoid urethral injury) will
determine the plane of distal intracorporal dilation,
which is subsequently accomplished to size 14 with
Brooks dilators. Proximal dilation to the level of the
crus is similarly performed, although proximal
Metzenbaum scissor insertion is usually unnecessary.
Experienced surgeons may use the Rossello or
Mooreville cavernotomes in brotic corpora to dilate
up to size 11 or 12. Others may use a Dilamezinsert
which is distally placed and subsequently dilated with
a prepackaged disposable dilator. Caution must be used
when using this device in brotic cases as the blind
distal dilation may cause tunical tear rather than
dilation. After proximal and distal dilation, cylinder
sizing is accomplished with sizers (Furlow passer or
the Dilamezinsert may be used) by measuring corporal
length from the proximal and distal stay sutures. The
exact same procedure is done on the contralateral
tunica and corpus.
Cylinders and rear-tip extenders (if necessary) are
then selected and positioned in the corpora using the
Furlow passer and needle, as described in the infra-
pubic section, above. They are then inflated to check
for symmetry, rigidity, absence of leakage, or kinking.
Again, it is important to provide manual support in the
136 Vacuum Devices and Penile Implants
area of corporotomy to prevent temporary buckling
if this maneuver is performed before corporotomy
closure. Pump positioning in the dependent scrotum is
much easier than in the infrapubic approach, but the
same principles and the creation of a dartos pouch
apply. A Babcock clamp may be used to secure the
pump in position until the end of the procedure and
the time of closure, at which time one or two sutures
may be placed just supercial to the pump to anchor it
in place and prevent pump migration.
After corporotomy closure, the Scott retractor is
removed and attention is focused on perivesical reser-
voir placement. This task is best accomplished using
small Deaver retractors (infant Deavers) to allow
cephalad retraction of the incision to the external
inguinal ring. Gentle blunt dissection medial to the
spermatic cord and over the pubic tubercle will lead to
the identication of the tranversalis fascia. Blunt dis-
section is often all that is necessary in rst implant
cases. It is advisable to empty the bladder by applying
suprapubic pressure before entering the perivesical
space. This is even more important in patients who
have had previous pelvic surgery, since it is unlikely
that blunt dissection will allow passage through the
brotic layers. In these cases, sharp dissection with
Metzenbaum scissors or a Kelly clamp is performed to
incise the fascia.The area is subsequently enlarged with
the surgeons index nger and the reservoir balloon is
positioned. It was previously recommended that all
reservoirs be lled and allowed to equilibrate. The new
Mentor lock-out valve device will prevent sponta-
neous reservoir deflation under normal intraabdominal
pressures and equilibration is therefore not necessary.
The practice is still recommended when using the
AMS prostheses. It is important to ensure proper sub-
fascial reservoir placement to minimize the chances of
intrascrotal or inguinal reservoir migration, a rare com-
plication that is almost exclusive to the penoscrotal
approach.
72
The tubing between the cylinders, reser-
voir, and pump is then trimmed, cleaned, and connected
as previously described using the manufacturers
connector tools.
Important technical considerations include lateral,
rather than medial, angulation of the scissors during
the initial passage in the corpus, gradual dilation up to
size 14 for virgin cases and size 1112 for brotic cases,
and gentle proximal pressure to avoid tunical tear.
Skin closure is performed in layers using 3-0 running
synthetic absorbable suture for the dartos layer and
interrupted 3-0 or 4-0 absorbable sutures for the skin.
Complications
Mechanical malfunction
Wilson et al. reported the results of a prospective study
of 1381 Mentor Alpha-1 penile prostheses implanted
to treat impotence, and compared the mechanical
reliability of the original and enhanced penile pros-
thesis. The 5-year survival rate increased from 75.3%
for the original to 92.6% for the enhanced model. The
failure rate of the enhanced model implants was about
0.8% per year during the rst 3.5 years and increased
to approximately 3.1% per year thereafter.
44
In
another publication, the same group reported that in
Peyronies disease cases in whom modeling was used
to straighten the penis after implantation, mechanical
survival of the Mentor Alpha-1 was superior to that
of the AMS 700CX and concluded that modeling
may predispose the AMS 700CX to earlier mechanical
failure.
52
A long-term multicenter study of the AMS 700CX
three-piece inflatable penile prosthesis reported that
mean device mechanical reliability plus or minus
standard deviation was 92.1% 3.3% after 3 years and
86.2% 4.6% after 5 years. Postoperative infection
and device malfunction developed in 3.2% and 17.5%
of the cases, respectively. Of the 207 men interviewed
by a neutral observer, 86% still had an AMS 700CX
penile prosthesis implanted, including 87.1% with
erection suitable for coitus.
73
In 1998, Dubocq and Dhabuwalas group compared
ve different types of devices and reviewed mechanical
complication rates in 83 patients with two-piece
and 283 patients with three-piece inflatable penile
prostheses for a mean time of 66 months. All device-
related complications were secondary to fluid leakage.
They noted a trend toward all three-piece prostheses
being more mechanically reliable than the two-piece;
the Mentor Alpha-1 device had a higher cumulative
proportional survival (0.957) than all other devices.
74
Goldstein et al. conducted a multiinstitutional retro-
spective study to assess safety and efcacy outcome
pertaining to the Mentor Alpha-1.With a mean follow-
up of 22.2 months, there were no morbidities of any
type in approximately 90% of implant recipients.
Fluid leak and autoinflation were reported in 2.5%.
No cylinder aneurysms were reported and only 2.5%
required revision surgery for approximately 2 years
from the original implant date. Cumulative survival of
the prosthesis at 36 months was 85 7% until device
malfunction and 75 7% until surgical intervention
(revision or explantation).
75
Corporal cross-over
Both distal and proximal corporal cross-over may
be encountered during corporal dilation or cylinder
placement, although the latter is more unusual. Neither
occurrence is catastrophic and may be easily recog-
nized and corrected during the procedure. The initial
correct lateral angulation of the Metzenbaum scissors
during distal tunneling and gradual corporal dilation
Penile Implants 137
using laterally directed Brooks dilators will help avoid
cross-over. The best means of testing either proximal
or distal cross-over is side-by-side placement of the
Brooks dilators in each corpus to check for symmetry
and proper positioning. If a cross-over is detected, the
dilator may simply be redirected with the contralateral
dilator left in place to prevent repeat cross-over.
Corporal and urethral perforation
This complication may be encountered both distally
and proximally and is more likely to occur during
dilation of brotic corpora. Careful use of specialized
cavernotomes such as the Rossello or Mooreville will
prevent uncontrolled tearing of the brotic corpus and
tunica and avoid the need for extensive corporal
excision, but these instruments have sharp edges and
may cause signicant injury if used by inexperienced
implanters.
48,49
A proximal crural perforation is
suspected when there is asymmetry of proximally
positioned dilators (i.e., when placed side-by side) or a
signicant length differential.
A variety of techniques have been described for
addressing this problem intraoperatively and include
direct repair of the perforation, placement of a wind-
sock patch of Gore-Tex or Dacron on the proximal
end of the cylinder, anchoring of cylinder tubing to the
tunica, and placement of an absorbable polyglycolic
acid patch in the defect (the plug and patch
technique).
76,77
Continued dissection to the level of
the rear-tip extenders and anchoring the extender to
the tunica are safe and widely adopted. This technique
is employed after rerouting the proximal dilation in
the affected corpus toward the proper crural tip against
the tuberosity. Attachment of the rear tip to the tunica
will prevent cylinder migration back into the per-
foration until healing is complete.
Management of distal corporal perforation and
urethral perforation is more likely to involve termi-
nation of the procedure and return at a later date,
particularly if distal perforation occurs during dilation
of the rst side. If corporal dilation has been success-
fully accomplished on one side and it is the second
side that is perforated, a single cylinder may be placed
on the non-perforated side. The tubing from the other
cylinder is removed and plugged with a standard
metallic plug provided by the manufacturers. A single
functioning cylinder may provide adequate rigidity for
penetration and the patient may elect not to undergo
a second procedure for contralateral cylinder place-
ment. Similarly, if urethral perforation occurs after one
side has been successfully dilated, the cylinder from the
perforated side is removed and the tubing is plugged.
The urethral tear may be directly repaired or, if small,
allowed to heal over a urethral catheter. Alternatively,
a perineal urethrostomy or a suprapubic urethrostomy
may be performed for urinary diversion. Because of
the potential for device infection, many surgeons will
recommend abandoning the case if there is any indi-
cation of urethral injury. However, this decision must
be balanced against the difculty of repeat surgery in
brotic corpora at a later date, especially in view of
ndings that implicate increased operative time to a
higher incidence of infections.
78
Infection
Infection is a devastating and dreaded complication of
penile prosthesis surgery which occurs in approximately
23% of rst-time implants in most series, but the
reported range is widely variable. Most infections
appear within the rst 3 months after surgery and the
vast majority will manifest within the rst year after
implantation, although delayed infections beyond
1 year have been reported.
79,80
Predisposing factors include inadequate perioperative
prophylaxis and lax sterile protocol, prolonged
hospitalization, prolonged operative time, and repeat
implantations. Carson reports that the combination of
any other procedure (e.g., hernia repair, circumcision)
with penile prosthesis surgery is associated with a
signicant increase in infections in his series.
81
The severity of infections may range from simple
supercial infections that can be managed by conser-
vative measures and wound care to the extreme of
penile gangrene which may be life-threatening. The
latter complication is fortunately rare and may be due
to Gram-negative organisms with or without anaerobic
superinfection. It may be initiated by factors including
local infection, pressure dressing, presence of a urethral
catheter, edema, and ischemia of the corpus caver-
nosum and appears to be more common in insulin-
dependent diabetics.
8285
Most infections are caused by Gram-positive
organisms that colonize normal skin, but 20% have
been attributed to Gram-negative bacteria in some
reports.
79
The latter are typically early infections that
manifest within the rst 30 days after surgery. The
causes of infection are controversial, but there is
universal agreement that weakened host defense
mechanisms and the presence of a synchronous infec-
tion at another site in the body will increase the risk.
Late infections may be due to hematogenous spread or
reemergence of bacteria previously embedded in the
biolm.
81
Jarow
78
reported an infection rate of 1.8% in men
without previous penile surgery compared to 21.7%
for procedures requiring reconstruction of the corpora.
The infection rate after revision of a penile prosthesis
was 13.3%, which was signicantly greater than that
following primary uncomplicated implantation, but
not different from that for patients requiring recon-
struction. This and other studies have not provided an
explanation for the increased incidence of infection in
138 Vacuum Devices and Penile Implants
patients undergoing revision who have an operative
time similar to rst-time implants.
Lynch et al.
86
reported a higher incidence of
infection in those receiving an inflatable device and
also reported a 22% incidence of infection in diabetic
patients compared to 6.7% in non-diabetics. Neither
of these ndings has been corroborated by other
investigators.
79,87
Although Bishop et al.
88
reported on
the direct relation between the degree of metabolic
control in diabetics as measured by glycosylated
hemoglobin and the risk of prosthesis infection, others
have found no meaningful difference in the median or
mean level of glycosylated hemoglobin A1C in infected
and non-infected patients regardless of diabetes.
89
The latter study also found no correlation between
elevation of fasting sugar or insulin dependence and an
increased risk of infection in diabetics undergoing
prosthesis implantation.
An interesting study by Licht et al.
90
evaluated the
presence of bacteria on implants undergoing mechanical
revision and isolated low colony counts of Staphy-
lococcus epidermidis in 40% of uninfected penile
prostheses. The authors concluded that low colony
counts of this organism are unlikely to cause an overt
clinical infection and that the role of S. epidermidis in
infections is likely to be overestimated. This reasoning
is further supported by documentation of ample
vancomycin, gentamicin, and aztreonam levels in the
corporal tissue of patients receiving antibiotic prophy-
laxis 12 h prior to device implantation.
91
Preventive measures are critical in the management
of prosthesis infections and have been addressed in the
section on perioperative care. The patient is instructed
to report any signs or symptoms of infection, which
may include a purulent exudate, a pattern of increasing
pain instead of gradual improvement, worsening
erythema and induration, and high-grade fever. Pain is
not unusual for as long as 46 weeks after surgery.
However, the normal course involves a gradual
improvement rather than deterioration. Increasing
pain, especially in the presence of chills, fever, or
leukocytosis, is an indication of possible infection.
Other clinical signs include erosion of device com-
ponents and fluctuance in the scrotum or along the
penile shaft. Most authorities recommend removal of
the entire device in severely infected cases with overt
purulence.
Some authors have advocated an early salvage and
rescue procedure for mild to moderate infections.
92,93
The technique has gained increasing popularity among
urologists for infections diagnosed in the earlier stages.
Successful salvage and return to function have been
reported in more than 80% of cases.
9395
However,
Carson
81
has warned that the procedure is not indicated
in insulin-dependent diabetics, immunocompromised
patients, or those with copious purulent drainage. The
salvage procedure involves complete removal of all
prosthesis components, copious irrigation of all affected
chambers through a rubber catheter placed in each
area with 5 liters of vancomycingentamicin solution,
and possible use of the water-pik or a similar high-
pressure irrigation device, as reported by Brant et al.
93
Erosion
Prosthesis erosion is often a telltale sign of device
infection, but device extrusion beneath the penile skin
may occur as an isolated phenomenon. Erosion is more
common in semirigid prostheses and in those with
distressed tissue and vascular supply, as may be seen
in brittle diabetics or redo implants. Erosion has
lso been described as a complication of urethral
catheterization.
Erosion presented as a late complication several
months after implantation in 80% of patients with
indwelling urethral catheters or who were using inter-
mittent clean catheterization and who had received a
penile prosthesis. The incidence of this complication
may be greatly reduced by using inflatable rather than
semirigid prostheses and by construction of a perineal
or suprapubic cystostomy.
96
Mulcahy has described a
distal corporoplasty for lateral extrusion of penile
prosthesis cylinders whereby the cylinder may be
repositioned in a more medial and secure position
under the glans penis by creating a new cavity for the
cylinder behind the back wall of the brotic sheath
that contains it.
97,98
Other complications
Reservoir herniation is an unusual complication of
three-piece inflatable prosthesis surgery through the
penoscrotal approach (Fig. 9.8). It occurs in approxi-
mately 0.7% of cases and may be caused by vigorous
postoperative coughing or failure of proper initial
reservoir placement under the transversalis fascia by
Figure 9.8 Three-piece inflatable prosthesis reservoir
herniation clinical presentation. (Courtesy of
Dr. Steven Wilson.)
Penile Implants 139
less experienced implanters. It may also be caused by
reservoir protrusion through an unrecognized existing
hernia or a large tranversalis defect created intra-
operatively.
72
The latter scenario is intuitively less
likely with decreased spontaneous autoinflation of the
cylinders in the immediate postoperative period (as
with Mentors lock-out valve mechanism). When
recognized in the immediate postoperative period, the
herniated intrascrotal reservoir may be repositioned
through the original penoscrotal incision. Alternatively,
a small inguinal incision may be used to place the reser-
voir in the perivesical space and close the defect from
above (Fig. 9.9). In the absence of clinical symptoms, a
few surgeons have reported leaving the reservoir in its
herniated intrascrotal position without adverse effects.
Glans bowing (supersonic transport
deformity)
Supersonic transport deformity is aptly named, after
the angulated tip of these aircraft. The problem may
be secondary to small prosthesis sizing or incomplete
distal dilation of the corpora. If the problem is recog-
nized intraoperatively and adequate distal dilation
has been achieved, a larger rear-tip extender may be
placed to lengthen the cylinder and see if the defect is
corrected. If the problem is noted in the immediate
postoperative period, it is wise to wait a few weeks and
allow for complete healing and scar formation, which
may result in glans xation and resolution of the
supersonic transport deformity.
81
Refractory cases may
be surgically corrected by placement of two 3/0 PDS
or permanent Prolene sutures on the underside of the
glans on each side through a circumcoronal incision.
By tying these sutures to the tunica albuginea near the
cylinder tip and away from the neurovascular bundle,
the glans is effectively secured against the cylinder
tip.
99,100
FUTURE OUTCOMES (THE EFFECTS
OF ORAL THERAPIES)
Although many implant surgeons feared a gradual
dwindling of penile prosthesis surgery with the intro-
duction of effective oral pharmacotherapeutic agents
such as sildenal, the signicant increase in the
number of men who present for the treament of ED
has further secured a role for surgical intervention.
Some investigators have postulated that the number
of penile prosthesis surgeries may actually increase in
the coming years.
101
Comparison of trends in penile
prosthesis procedures at a tertiary medical center
before and after the introduction of sildenal revealed
no signicant change in the number of penile implant
procedures done annually, although both the com-
plexity of procedures and severity of illness
increased.
102
These ndings will surely encourage
the implant surgeons, as well as the companies and
engineers, who are in pursuit of further renements
and improvements in the design of future prostheses
and the art of penile prosthesis surgery.
REFERENCES
1. Lewis RW, Witherington R. External vacuum therapy for
erectile dysfunction: use and results. World J Urol 1997;
15:7882.
2. Osbon G, inventor Erection Aid Device. United States
patent US patent number 4 378 008. 1983 March 29, 1983.
3. King J. The American physician domestic guide to
health. Indianapolis: Streight and Douglass; 1874.
4. Lederer O, inventor Surgical Device. United States patent
US patent number 1 225 241. 1917 May 8, 1917.
5. Sell F, inventor Erector. United States patent 2 874 698.
1959 February 24, 1959.
6. Wilson E, inventor Apparatus for Obtaining an Articial
Erection. United States patent 3 744 486. 1973 July 10, 1973.
7. Nadig PW, Ware JC, Blumoff R. Noninvasive device to
produce and maintain an erection-like state. Urology 1986;
27:126131.
8. Witherington R. Vacuum constriction device for manage-
ment of erectile impotence. J Urol 1989; 141:320322.
9. Witherington R. Vacuum devices for the impotent. J Sex
Marit Ther 1991; 17:6980.
10. Montague DK, Barada JH, Belker AM et al. Clinical
guidelines panel on erectile dysfunction: summary report on
the treatment of organic erectile dysfunction. The American
Urological Association. J Urol 1996; 156:20072011.
11. Bosshardt RJ, Farwerk R, Sikora R et al. Objective
measurement of the effectiveness, therapeutic success and
dynamic mechanisms of the vacuum device. Br J Urol 1995;
75:786791.
140 Vacuum Devices and Penile Implants
Figure 9.9 Reservoir herniation: removal of the reservoir
through inguinal approach prior to herniorrhaphy and
repositioning of the reservoir in the perivesical space.
(Courtesy of Dr. Steven Wilson.)
12. Chen J, Godschalk MF, Katz PG et al. Combining intra-
cavernous injection and external vacuum as treatment for
erectile dysfunction. J Urol 1995; 153:14761477.
13. Blackard CE, Borkon WD, Lima JS et al. Use of vacuum
tumescence device for impotence secondary to venous
leakage. Urology 1993; 41:225230.
14. Moul JW, McLeod DG. Negative pressure devices in the
explanted penile prosthesis population. J Urol 1989; 142:
729731.
15. Denil J, Ohl DA, Smythe C. Vacuum erection device in
spinal cord injured men: patient and partner satisfaction.
Arch Phys Med Rehabil 1996; 77:750753.
16. Lue TF, El-Sakka AI. Lengthening shortened penis
caused by Peyronies disease using circular venous grafting
and daily stretching with a vacuum erection device. J Urol
1999; 161:11411144.
17. Kolettis PN, Lakin MM, Montague DK et al. Efcacy of
the vacuum constriction device in patients with corporeal
venous occlusive dysfunction. Urology 1995; 46:856858.
18. Earle CM, Seah M, Coulden SE et al. The use of the
vacuum erection device in the management of erectile impo-
tence. Int J Impot Res 1996; 8:237240.
19. Tan HL. Economic cost of male erectile dysfunction
using a decision analytic model: for a hypothetical managed-
care plan of 100 000 members. Pharmacoeconomics 2000;
17:77107.
20. Meinhardt W, Kropman RF, Lycklama a Nijeholt AA et al.
Skin necrosis caused by use of negative pressure device for
erectile impotence. J Urol 1990; 44:983.
21. Ganem JP, Lucey DT, Janosko EO et al. Unusual compli-
cations of the vacuum erection device. Urology 1998; 51:
627631.
22. Dutta TC, Eid JF.Vacuum constriction devices for erectile
dysfunction: a long-term, prospective study of patients with
mild, moderate, and severe dysfunction. Urology 1999; 54:
891893.
23. Cookson MS, Nadig PW. Long-term results with vacuum
constriction device. J Urol 1993; 149:290294.
24. Turner LA, Althof SE, Levine SB et al. Treating erectile
dysfunction with external vacuum devices: impact upon
sexual, psychological and marital functioning. J Urol 1990;
144:7982.
25. Baltaci S, Aydos K, Kosar A et al. Treating erectile dys-
function with a vacuum tumescence device: a retrospective
analysis of acceptance and satisfaction. Br J Urol 1995;
76:757760.
26. Soderdahl DW, Thrasher JB, Hansberry KL. Intra-
cavernosal drug-induced erection therapy versus external
vacuum devices in the treatment of erectile dysfunction. Br J
Urol 1997; 79:952957.
27. Chen J, Mabjeesh NJ, Greenstein A. Sildenal versus
the vacuum erection device: patient preference. J Urol 2001;
166:17791781.
28. Bogoras N. Uber die volle plastiche wiederherstellung
einer zum koitus fahigen penis (penilplastica totalis). Zentralbl
Chir 1936; 63:1271.
29. Loeffler RASE. Perforated acrylic implants in manage-
ment of organic impotence. J Urol 1960; 84:559562.
30. Small MP, Carrion HM, Gordon JA. SmallCarrion penile
prosthesis. New implant for management of impotence.
Urology 1975; 5:479486.
31. Pearman RO. Treatment of organic impotence by implan-
tation of a penile prosthesis. J Urol 1967; 97:716719.
32. Lash H. Silicone implant for impotence. J Urol 1968;
100:709710.
33. Beheri GE. Surgical treatment of impotence. Plast
Reconstr Surg 1966; 38:9297.
34. Morales PA, Suarez JB, Delgado J et al. Penile implant
for erectile impotence. J Urol 1973; 109:641645.
35. Scott FB, Bradley WE, Timm GW. Management of erectile
impotence. Use of implantable inflatable prosthesis. Urology
1973; 2:8082.
36. Small MP. SmallCarrion penile prosthesis: a report on
160 cases and review of the literature. J Urol 1978; 119:
365368.
37. Kothari DR, Timm GW, Frohib DA et al. An implantable
fluid transfer system for treatment of impotence. J Biomech
1972; 5:567.
38. Finney RP. New hinged silicone penile implant. J Urol
1977; 118:585587.
39. Subrini L, Couvelaire R. [Surgery of male impotence
intracavernous prosthesis of the penis (authors translation).]
J Urol Nephrol (Paris) 1974; 80:269276.
40. Krane RJ. Omniphase penile prosthesis. Semin Urol 1986;
4:247251.
41. Mulcahy J. Unitary inflatable, mechanical, and malleable
penile implants. Oxford: Isis Medical Media; 1999.
42. Wilson SK, Delk JR, 2nd. Historical advances in penile
prostheses. Int J Impot Res 2000; 12 (suppl. 4):S101S107.
43. Merrill DC. Mentor inflatable penile prostheses. Urol Clin
North Am 1989; 16:5166.
44. Wilson SK, Cleves MA, Delk JR, 2nd. Comparison of
mechanical reliability of original and enhanced Mentor Alpha
I penile prosthesis. J Urol 1999; 162:715718.
45. Wilson SK, Cleves M, Delk JR, 2nd. Long-term results
with Hydroflex and Dynaflex penile prostheses: device
survival comparison to multicomponent inflatables. J Urol
1996; 155:16211623.
46. Dubocq FM, Bianco FJ Jr, Maralani SJ et al. Outcome
analysis of penile implant surgery after external beam
radiation for prostate cancer. J Urol 1997; 158:17871790.
47. Khoudary KP, DeWolf WC, Bruning CO, 3rd et al. Imme-
diate sexual rehabilitation by simultaneous placement of
penile prosthesis in patients undergoing radical prostatectomy:
initial results in 50 patients. Urology 1997; 50:395399.
48. Mooreville M, Adrian S, Delk JR, 2nd et al. Implantation
of inflatable penile prosthesis in patients with severe corporeal
References 141
brosis: introduction of a new penile cavernotome. J Urol
1999; 162:20542057.
49. Rossello Barbara M, Carrion H. [Cavernotomy.] Arch Esp
Urol 1991; 44:185186.
50. Montorsi F, Salonia A, Maga T et al. Reconguration of
the severely brotic penis with a penile implant. J Urol 2001;
166:17821786.
51. Carbone DJ, Jr, Daitch JA, Angermeier KW et al.
Management of severe corporeal brosis with implantation of
prosthesis via a transverse scrotal approach. J Urol 1998;
159:125127.
52. Wilson SK, Delk JR, 2nd. A new treatment for Peyronies
disease: modeling the penis over an inflatable penile
prosthesis. J Urol 1994; 152:11211123.
53. Wilson SK, Cleves MA, Delk JR, 2nd. Long-term followup
of treatment for Peyronies disease: modeling the penis over
an inflatable penile prosthesis. J Urol 2001; 165:825829.
54. Carson CC. Penile prosthesis implantation in the treatent
of Peyronies disease and erectile dysfunction. Int J Impot Res
2000; 12 (suppl. 4):S122S126.
55. Levine LA, Dimitriou RJ. A surgical algorithm for penile
prosthesis placement in men with erectile failure and Peyronies
disease. Int J Impot Res 2000; 12:147151.
56. Gross AJ, Sauerwein DH, Kutzenberger J et al. Penile
prostheses in paraplegic men. Br J Urol 1996; 78:262264.
57. Cuellar DC, Sklar GN. Penile prosthesis in the organ
transplant recipient. Urology 2001; 57:138141.
58. Lockyer R, Gingell C. Spontaneous breakage of malleable
prosthesis. Int J Impot Res 1999; 11:237.
59. Mills RD, Sethia KK. Limited sub-coronal incision for
insertion of semi-rigid penile prostheses. Br J Urol 1997;
79:802803.
60. Collins KP, Hackler RH. Complications of penile
prostheses in the spinal cord injury population. J Urol 1988;
140:984985.
61. Swana HS, Foster HE, Jr. Erosion of malleable penile
prosthesis into bladder. J Urol 1997; 157:22592260.
62. Mulcahy JJ. The Hydroflex penile prosthesis. Urol Clin
North Am 1989; 16:3338.
63. Mulcahy JJ. The Hydroflex self-contained inflatable
prosthesis: experience with 100 patients. J Urol 1988;
140:14221423.
64. Montague DK, Lakin MM. Early experience with the
controlled girth and length expanding cylinder of the
American Medical Systems Ultrex penile prosthesis. J Urol
1992; 148:14441446.
65. Wilson SK, Cleves MA, Delk JR, 2nd. Ultrex cylinders:
problems with uncontrolled lengthening (the S-shaped
deformity). J Urol 1996; 155:135137.
66. Milbank AJ, Montague DK, Angermeier KW et al.
Mechanical failure of the American Medical Systems Ultrex
inflatable penile prosthesis: before and after 1993 structural
modication. J Urol 2002; 167:25022506.
67. Montorsi F, Rigatti P, Carmignani G et al. AMS three-
piece inflatable implants for erectile dysfunction: a long-term
multi-institutional study in 200 consecutive patients. Eur Urol
2000; 37:5055.
68. Deuk Choi Y, Jin Choi Y, Hwan Kim J et al. Mechanical
reliability of the AMS 700CXM inflatable penile prosthesis
for the treatment of male erectile dysfunction. J Urol 2001;
165:822824.
69. Schwartz BF, Swanzy S, Thrasher JB. A randomized
prospective comparison of antibiotic tissue levels in the
corpora cavernosa of patients undergoing penile prosthesis
implantation using gentamicin plus cefazolin versus an oral
fluoroquinolone for prophylaxis. J Urol 1996; 156:991994.
70. Garber BB. Outpatient inflatable penile prosthesis
insertion. Urology 1997; 49:600603.
71. Garber BB, Marcus SM. Does surgical approach affect the
incidence of inflatable penile prosthesis infection? Urology
1998; 52:291293.
72. Sadeghi-Nejad H, Sharma A, Irwin RJ et al. Reservoir
herniation as a complication of three-piece penile prosthesis
insertion. Urology 2001; 57:142145.
73. Carson CC, Mulcahy JJ, Govier FE. Efcacy, safety and
patient satisfaction outcomes of the AMS 700CX inflatable
penile prosthesis: results of a long-term multicenter study.
AMS 700CX study group. J Urol 2000; 164:376380.
74. Dubocq F, Telli MV, Gheiler EL et al. Long-term
mechanical reliability of multicomponent inflatable penile
prosthesis: comparison of device survival. Urology 1998;
52:277281.
75. Goldstein I, Newman L, Baum N et al. Safety and
efcacy outcome of mentor alpha-1 inflatable penile pros-
thesis implantation for impotence treatment. J Urol 1997;
157:833839.
76. Mulcahy JJ. A technique of maintaining penile prosthesis
position to prevent proximal migration. J Urol 1987;
137:294296.
77. Szostak MJ, DelPizzo JJ, Sklar GN. The plug and patch:
a new technique for repair of corporal perforation during
placement of penile prostheses. J Urol 2000; 163:12031205.
78. Jarow JP. Risk factors for penile prosthetic infection.
J Urol 1996; 156:402404.
79. Montague DK. Periprosthetic infections. J Urol 1987;
138:6869.
80. Carson CC, Robertson CN. Late hematogenous infection
of penile prostheses. J Urol 1988; 139:5052.
81. Carson C. Complications of penile prostheses and
complex implantations. Oxford: Isis Medical Media; 1999.
82. McClellan DS, Masih BK. Gangrene of the penis as a
complication of penile prosthesis. J Urol 1985; 133:862863.
83. Walther PJ, Andriani RT, Maggio MI et al. Fourniers
gangrene: a complication of penile prosthetic implantation in
a renal transplant patient. J Urol 1987; 137:299300.
84. Kardar A, Pettersson BA. Penile gangrene: a compli-cation
of penile prosthesis. Scand J Urol Nephrol 1995; 29:355356.
142 Vacuum Devices and Penile Implants
85. Kumbkarni Y, Magsino CH, Jr, Dandona P. A diabetic
patient with a black penile tip. J Diabetes Complications
1999; 13:316319.
86. Lynch MJ, Scott GM, Inglis JA et al. Reducing the loss of
implants following penile prosthetic surgery. Br J Urol 1994;
73:423427.
87. Wilson SK, Delk JR, 2nd. Inflatable penile implant infec-
tion: predisposing factors and treatment suggestions. J Urol
1995; 153:659661.
88. Bishop JR, Moul JW, Sihelnik SA et al. Use of
glycosylated hemoglobin to identify diabetics at high risk for
penile periprosthetic infections. J Urol 1992; 147:386388.
89. Wilson SK, Carson CC, Cleves MA et al. Quantifying risk
of penile prosthesis infection with elevated glycosylated
hemoglobin. J Urol 1998; 159:15371539; discussion
15391540.
90. Licht MR, Montague DK, Angermeier KW et al. Cultures
from genitourinary prostheses at reoperation: questioning the
role of Staphylococcus epidermidis in periprosthetic infection.
J Urol 1995; 154:387390.
91. Walters FP, Neal DE, Jr, Rege AB et al. Cavernous tissue
antibiotic levels in penile prosthesis surgery. J Urol 1992;
147:12821284.
92. Furlow WL, Goldwasser B. Salvage of the eroded inflat-
able penile prosthesis: a new concept. J Urol 1987; 138:
312314.
93. Brant MD, Ludlow JK, Mulcahy JJ.The prosthesis salvage
operation: immediate replacement of the infected penile
prosthesis. J Urol 1996; 155:155157.
94. Mulcahy JJ. Long-term experience with salvage of
infected penile implants. J Urol 2000; 163:481482.
95. Carson CC. Lecture 2. Penile prosthesis implantation and
infection for Sexual Medicine Society of North America. Int J
Impot Res 2001; 13 (suppl. 5):S35S38.
96. Steidle CP, Mulcahy JJ. Erosion of penile prostheses:
a complication of urethral catheterization. J Urol 1989;
142:736739.
97. Mulcahy JJ. Distal corporoplasty for lateral extrusion of
penile prosthesis cylinders. J Urol 1999; 161:193195.
98. Mulcahy JJ. Surgical management of penile prosthesis
complications. Int J Impot Res 2000; 12 (suppl. 4):S108S111.
99. De Stefani S, Simonato A, Capone M et al. The benet of
glans xation in prosthetic penile surgery. J Urol 1994;
152:15331534.
100. Ball TP, Jr. Surgical repair of penile SST deformity.
Urology 1980; 15:603604.
101. Montague DK, Angermeier KW. Penile prosthesis
implantation. Urol Clin North Am 2001; 28:355361, x.
102. Stanley GE, Bivalacqua TJ, Hellstrom WJ. Penile pros-
thetic trends in the era of effective oral erectogenic agents.
South Med J 2000; 93:11531156.
References 143
INTRODUCTION
Prostate cancer is the most frequently diagnosed solid
tumor in US men with 80% of newly diagnosed cases
occurring in men older than 65 years. In 2000, it caused
an estimated 180 400 new cases and 31 900 deaths.
1
The patient faced with the diagnosis of prostate cancer
is often focused on the eradication of his disease.
Quality-of-life issues that follow the disease process
or therapies for prostate cancer may not be initially of
concern to the individual devastated by the news of
cancer. However, improvements in cancer detection
rates and longer life expectancy in the industrialized
nations have translated into signicant long-term
survivorship. Along with great strides in the direction
of cancer control for localized disease, signicant rene-
ments in surgical technique and radiation delivery have
decreased the incidence of adverse events following
therapy. The physician counseling his patient on appro-
priate treatment modalities for prostate cancer must
also understand patient expectations relating to the
aforementioned quality-of-life issues.Thus, the issue of
quality of life posttherapy for prostate cancer is of
paramount importance.This chapter will explore issues
related to sexuality and erectile dysfunction (ED) in
the context of prostate cancer.
EPIDEMIOLOGY
The average age at diagnosis of prostate cancer is 69.2
years for white men and 67.3 years for black men.
2
It follows that, in the vast majority of patients, the
impact of prostate cancer diagnosis and treatment on
erectile function must be considered in an age group
that is already at high risk for ED. Equally important is
the knowledge that, although men younger than 55
constitute only 3% of diagnosed prostate cancer cases,
there has been a striking increase of prostate cancer
detection in this population.
3,4
Quality-of-life issues
will likely assume an even greater importance in this
latter group, who will carry the burden of their diag-
nosis for a much longer period.
The National Health and Social Life Survey
(NHSLS) reported an ED prevalence rate of 18% for
men aged 5059 years in the general population.
5
The Massachusetts Male Aging Study indicated that
the probability of complete ED increased from 5.1%
to 15% between the ages of 40 and 70 years and the
probability of moderate ED increased from 17% to
34%.
6
In a prospective evaluation of patient-reported
symptoms in prostate cancer, ED, dened as having
either no erections or erections usually inadequate for
sexual intercourse, was reported by 32% of presurgical
and 45% of preradiation therapy patients.
7
The better
pretreatment erectile function in the surgical group is
likely a function of the younger age and better overall
health status of the surgical candidates. This disparity
reflects an ongoing issue in the eld of ED as it relates
to prostate cancer. The large difference in the pretreat-
ment ED rates is reflective of the inconsistencies seen
in the follow-up data in these men. Reports on the
prevalence of ED in the posttherapy prostate cancer
patients are widely variable and range from to 20 to
90%.
717
Siegel et al. evaluated pre- and posttreatment ED in
patients with localized prostate cancer who were
managed with irradiation, prostatectomy, or watchful
waiting.
13
Validated questionnaires were not used.
Sixty-nine percent of the overall cohort had pretreat-
ment erections that were sufcient for intercourse and,
as in the Talcott study, the pretherapy potency was
signicantly higher in those who later underwent
radical prostatectomy (RP) as compared to those who
received irradiation. Patients selecting watchful wait-
ing had the lowest risk of ED and 38% of this group
reported adequate erections at last follow-up (mean
follow-up 53 months). No signicant difference was
noted in posttreatment erectile function between
patients treated with RP or external beam radiation
(XRT: 10% versus 15%) and the two groups were
similar in patient age, clinical stage, and pathological
grade. The authors further note that, due to the differ-
ing times of onset of ED after XRT as compared to
RP (delayed onset versus early onset), studies with
shorter follow-up may erroneously document higher
ED rates in the prostatectomy group. The decline in
potency in the watchful waiting group was 17%, which
is similar to an age-matched population without pros-
tate cancer.
18
The incidence of ED after RP has been the subject of
many investigations in the past 1015 years. There is
Prostate Cancer and Erectile Dysfunction
Hossein Sadeghi-Nejad and Allen D. Seftel
CHAPTER 10
considerable variability between the reported series
from experienced urological surgeons and centers of
urological excellence compared to other epidemio-
logical surveys. For example, a study by Walsh et al.
11
to determine patient-reported rates of potency after
RP performed by an experienced surgeon found that,
by 18 months, 86% of patients were potent and 84%
considered sexual bother as none or small. Potency
was dened as the ability to have unassisted intercourse
with or without the use of sildenal, and improved
gradually over the course of the study. The authors
further commented that, although one-third of patients
at 18 months were using sildenal intermittently, only
two patients were not able to have intercourse without
its use. A similar center of excellence report from
Washington University by Catalona et al. indicated
that recovery of erections occurred in 68% of pre-
operatively potent men treated with bilateral (543 of
798) and 47% treated with unilateral (28 of 60) nerve-
sparing surgery. Recovery of erections was more likely
with bilateral than with unilateral nerve-sparing surgery
in patients less than 70 years old (71 versus 48%)
compared with patients aged 70 years old or older (48
versus 40%).
15
Similarly, in a study of factors predicting recovery of
erections after RP, Rabbani et al.
19
reported that age,
quality of preoperative erectile function, and extent of
neurovascular bundle (NVB) preservation, but not
pathological stage, were predictive of potency recovery
after radical retropubic prostatectomy (RRP). A total
of 149 of the 314 evaluated patients recovered potency
and 75% of this cohort had recovered satisfactory
erections by 11.8 months postoperatively. Median time
to potency was 3.9 months (before sildenal) with
bilateral nerve-sparing, 4.1 months with unilateral or
bilateral nerve damage, and 9.7 months with uni-
lateral nerve resection. Compared to the younger men,
those 6065 years old were only 56% and those older
than 65 years were 47% as likely to recover potency.
Patients with partial erections were only 47% as likely
to recover potency as men with full erections pre-
operatively. The authors determined the probability of
return of spontaneous erections at 24 and 36 months
for any given combination of predictive variables
from the coefcients of the Cox regression model for
variables having statistical signicance. With a median
follow-up of 25.4 months, the difference in potency
rates was statistically signicant for unilateral versus
bilateral nerve-sparing and there was a fourfold
reduction in recovery of potency rates with unilateral
versus bilateral nerve-sparing. In addition, these and
other authors have reported that pathological stage is
not an independent predictive parameter for erection
recovery.
15,19
In contrast to the above studies at urological centers
of excellence, Stanford et al.
12
found lower potency
rates in their report on the prostate cancer outcomes.
At 18 months or more following RP, 59.9% of the
1291 evaluated patients were impotent after RP.
Among men who were potent before surgery, the
proportion of men reporting impotence at 18 or more
months after surgery varied according to whether the
procedure was nerve-sparing (65.6% of non-nerve-
sparing, 58.6% of unilateral, and 56.0% of bilateral
nerve-sparing). At 18 or more months after surgery,
41.9% reported that their sexual performance was a
moderate-to-large problem. In another prospective
study, inadequate erections, present in one-third of men
prior to treatment, were nearly universal at 3 months
after surgery (96%), although some improvement,
primarily in men under 65 years of age, was evident at
12 months.
7
A study of ED in men with prostate cancer
before and after RP reported potency (unassisted inter-
course with vaginal penetration) in 2 of 187 (1.1%)
undergoing surgery without nerve-sparing, and 27 of
203 (13.3%) undergoing unilateral and 22 of 69 (31.9%)
undergoing bilateral nerve-sparing prostatectomy.
Less than half of the patients who were sexually active
postoperatively were satised with the erections or
achieved intercourse at least once a month.
20
Fowler et al. evaluated Medicare patients 24 years
after RP and reported that about 60% of patients
reported having no full or partial erections since their
surgery, and only 11% had any erections sufcient
for intercourse during the month prior to the survey.
9
Interestingly, the same authors reported on the quality-
of-life measures in this same population 2 years later
and noted that, similar to a cohort of patients with
benign prostatic hyperplasia who had undergone trans-
urethral resection of the prostate, postsurgical patients
scored comparatively high on the quality-of-life mea-
sures such as feeling positive about the results (81%),
and that most would choose surgical treatment again
(89%).
10
Despite the conflicting data, most experts agree that
Walsh and Donkers introduction of the nerve-sparing
technique in 1982 has had a signicant impact on
reducing postoperative ED.
21
The observed differences
between the different series may be attributed to
patient selection and the volume of cases, inconsistent
denitions of ED, various reporting methodologies
(i.e., patient-reported versus physician-reported), the
multifactorial etiology of postoperative ED, comor-
bidities, and the experience of the operating surgeon
in preserving the NVB.
9,11,22
Table 10.1 lists a
number of studies reporting on the prevalence of ED
following RP.
The incidence of ED following XRT and brachy-
therapy has been reported by a number of investigators.
Litwin et al. measured the effect of treatment choice
(pelvic irradiation (XRT) versus RP with or without
nerve-sparing) on sexual function and sexual bother
146 Prostate Cancer and Erectile Dysfunction
during the rst 2 years after treatment.
23
Patients
undergoing XRT or RP with or without nerve-sparing
all showed comparable rates of improvement in sexual
function during the rst year after treatment for early-
stage prostate cancer. However, in the second year
after treatment, patients treated with XRT began to
show declining sexual function; patients treated with
RP did not. Sexual function was measured by the
UCLA Prostate Cancer Index on a scale of 0100 with
higher scores representing better outcomes. Whereas
at 0 months after treatment, the RP with nerve-sparing
group had a score of 15 1.5 compared to 28 2.5 in
the XRT group, the same scoring system documented
24 1.8 (RP) versus 37 2.9 (XRT) at 12 months
and 28 2.6 (RP) versus 30 4.0 (XRT) at 24 months,
indicating the declining sexual function scores in the
XRT group with the passage of time. Otherwise stated
and similarly shown in Talcotts study, they found that
although short-term sexual function differs between
the two groups with an apparent advantage in the
XRT group, long-term function becomes increasingly
similar due to gradually worsening function in the XRT
group.
7
Zelefsky et al.
24
evaluated the incidence and
predictors of late toxicity in patients with localized
prostate carcinoma treated with high-dose three-
dimensional conformal radiotherapy (3D-CRT) and
found that 39% of patients who were potent before
treatment became impotent after 3D-CRT. The 5-year
actuarial risk of potency loss was 60% and the median
time to the development of impotence was 19 months.
Patients with pretherapy potency who received
75.6 Gy had a 68% actuarial likelihood of impotence
compared to 52% in those who received 70.2 Gy.
In addition to higher doses, the use of androgen depri-
vation therapy increased the likelihood of permanent
impotence (69% ED compared to 56% in those not
receiving neoadjuvant hormonal therapy (HT)). Stock
et al.
25
assessed erectile function after prostate brachy-
therapy and analyzed those factors affecting potency
preservation. In 313 patients who were potent before
therapy, the actuarial freedom from any decrease in
erectile function score was 64% and 30% at 3 and
6 years, respectively. The two factors found to have a
signicant negative effect on potency were high implant
dose and a lower pretreatment erectile function score.
Similar ndings were reported by Turner et al.,
26
who
showed that, while 62% of men (90 of 146) who were
potent before XRT preserved their potency 12 months
after XRT, this gure was 41% at 24 months.
Fulmer et al.
27
performed a comparative prospective
study on the effects of RP and hormone plus XRT on
ED. Initially RP patients experienced worse sexual
function scores; however, scores for RP patients
changed over time and approached the levels seen in
HT plus XRT cohort at 18 months. All of these studies
are in agreement with Litwin and colleagues report on
the worsening of erectile function with the passage of
time after XRT.
23
In a study by Sanchez-Ortiz et al.,
28
potency was
maintained in 49% of men (40/81) who were potent
prior to brachytherapy. ED rates were signicantly lower
in younger patients (48%) versus older patients (55%).
There was no difference in posttreatment potency
between men who received neoadjuvant HT and those
who did not. Two years following brachytherapy, 25%
of patients complained of complete (20/81) or partial
(26%, 21/81) ED, for an overall rate of 51% (41/81).
Short-term neoadjuvant HT (36 months) did not
increase the likelihood of posttreatment ED.
The effects of HT in combination with radiation
were also investigated by Chen et al.,
29
who evaluated
the effect of 3D-CRT with or without HT on sexual
function (SF) in prostate cancer patients whose SF was
known before all treatment. Before 3D-CRT, 87 (60%)
of 144 men were totally potent as compared to only
47 (47%) of 101 at 1-year follow-up. Of the 60 men
totally potent at baseline and followed for at least
1 year, 35 (58%) remained totally potent. Patients
who had 3D-CRT alone were more likely to be totally
potent at 1 year than those receiving 3D-CRT with
HT. Of those receiving 3D-CRT, 71% were potent pre-
therapy and 56% of this group retained potency 1 year
after therapy. For those receiving HT + CRT, 44%
were potent prior to therapy and 31% retained their
potency at 1-year follow-up.
29
However, those receiving
CRT alone (i.e., no HT) were also more likely to be
Epidemiology 147
TABLE 10.1 Prevalence of erectile dysfunction
after radical prostatectomy.
Author Year n Erectile
dysfunction (%)
Quinlan et al.
80
1991 503 32.0
Fowler et al.
9
1993 739 79.0
Geary et al.
20
1995 459 44.4
Catalona et al.
15
1999
Total 858 33.5
Bilateral nerve-
sparing 798 32.0
Unilateral nerve-
sparing 60 53.0
Stanford et al.
12
2000 1291 59.0
Walsh et al.
11
2000 70 14.0
Kao et al.
14
2000 887 89.0
Siegel et al.
13
2001 419 90.0
potent at baseline (compared to 71% in the non-HT
group, only 44% of patients receiving HT + 3D-CRT
were potent before administration of any therapy). The
authors concluded that, while the use of HT was a
signicant predictor of ED before therapy, it did not
appear to increase the risk of sexual dysfunction after
treatment. Although the study did not provide answers
regarding the length of time after which HT will
adversely affect sexual function, the authors stated
that the degree of decrement in sexual function after
CRT +HT did not appear to differ signicantly from
that in those receiving CRT alone. Table 10.2 lists a
number of studies with variable follow-up reporting on
the prevalence of ED after XRT and brachytherapy.
PATHOPHYSIOLOGY
The etiology of ED following RP and irradiation for the
treatment of prostate cancer is multifactorial. Branches
of the prostatovesicular artery and vein run together
with the cavernosal nerve branches as the NVB. The
relationship of the prostate and urethra to the caver-
nous nerve entering the perineum deep to the prostatic
venous plexus on each side is shown in Figure 10.1.
Following the pioneering work in 1982 of Walsh and
Donker,
21
who described the precise delineation of
anatomic relationship of the NVB to the bladder neck
and prostate, surgeons performing RP developed
nerve-sparing techniques in hopes of preserving sexual
function.
30
Anatomic variability in the course of caver-
nous nerves and difculties with reliable intraoperative
identication of the nerves led to the development
of CaverMap (Uromed, Boston, MA), a device that
stimulates the cavernosal nerves and measures minute
variations in penile girth. A multicenter evaluation of
the device by experienced surgeons demonstrated an
87.8% sensitivity and 54% specicity in locating the
NVB.
31
However, widespread use of this device has not
occurred due to poor specicity.
Despite signicant improvements in sexual function
and quality of life following nerve-sparing RP compared
to non-nerve-sparing RP, it became clear that a signi-
cant number of patients whose nerves were spared
developed ED after surgery. A vasculogenic etiology in
this group of patients was suggested by Bahnson and
Catalona, who demonstrated absent response to intra-
cavernosal papaverine injection.
32
Although the poor
causal relation between vasculogenic pathology and
absence of erectile response to erectogenic agents has
been demonstrated in other investigations, this work
prompted further investigations in search of a vascular
etiology for post-RP ED. In 1989, the anatomy of
penile neurovascular supply was described by Breza
et al.,
33
who found a 70% prevalence of an accessory
pudendal artery coursing over the anterolateral surface
of the prostate in a study of 10 cadavers. The high
prevalence reported was not supported by previous
reports indicating a range of 621%,
34,35
but the report
highlighted the variable origin of the artery from the
obturator (50%) or vesical arteries (47%). Polascik
and Walsh later reported their own ndings relating to
the influence of the accessory pudendal artery on
the recovery of sexual function.
36
They identied the
artery in 4% of 835 men and concluded that, because
potency rates are similar in men with or without
preservation of accessory arteries, routine preservation
may not be productive and accessory arteries that may
be identied for preservation occur infrequently.
Another possible source of vasculogenic compromise
during RP are the arteries identied beneath the
anterior capsule of the prostate under the dorsal vein.
Preservation of these arteries is difcult and potentially
complicated by massive blood loss due to their close
communication with the dorsal venous complex.
The multifactorial nature of the post-RP ED is further
demonstrated by wide variability in response to intra-
148 Prostate Cancer and Erectile Dysfunction
TABLE 10.2 Erectile dysfunction (ED) after radiation therapy (XRT).
Author Year n XRT versus brachytherapy ED (%)
Talcott et al.
7
1998 135 XRT 61.0
a
Merrick et al.
81
2001 34 Brachytherapy 35.0
b
Stock et al.
25
2001 313 Brachytherapy 41.0
c
Talcott et al.
82
2001 105 Brachytherapy 73.0
d
Siegel et al.
13
2001 319 XRT 85.0
a
Erections inadequate for sex at 12-month follow-up.
b
13-month median follow-up.
c
59% actuarial preservation of potency at 6-year follow-up.
d
Erections not rm enough for penetration without manual assistance.
cavernosal injections in different reported series. For
example, whereas Aboseif et al.
37
note vascular com-
promise (reduction in both the diameter and velocity
of blood flow within cavernosal arteries) and poor
response to injections in 40% of their patients (mean
age 65), Polascik and Walsh report that most of their
patients (mean age 57) have a positive response to
intracavernosal injection therapy.
36
These differences
are likely due to the younger age of patients and
different selection criteria in the latter report.
Aboseif et al.
37
used duplex ultrasonography to
evaluate possible vascular mechanisms responsible
for postoperative ED and documented signicantly
decreased cavernosal artery diameter and peak systolic
velocities in 40% of post-RP patients who had had a
poor response to intracavernosal injections. The role of
corporovenoocclusive dysfunction was not evaluated
in this study. After elucidation of a possible vascular
contribution to post-RP sexual dysfunction, a number
of researchers focused their attention on further
denition of the exact vasculogenic pathophysiology
and evaluation of relative contributions from arterio-
genic versus venogenic factors. DeLuca et al. reported
11% prevalence (5 patients) of postsurgical veno-
occlusive dysfunction (VOD) with impotence following
radical cystectomy and a 5% prevalence (2 patients) of
impotence following RP in a retrospective study.
38
Kim
et al. documented increased cavernosal artery end-
diastolic flow velocity and decreased resistive index on
spared and non-spared sides after RP.
39
Although these
ndings did not reach statistical signicance, involve-
ment of both VOD and arterial insufciency was
suggested as a possible cause for post-RP ED.
Two years later, Mulhall and Graydon
40
presented
pre- and postoperative cavernosometric data on post-
RP patients with ED that corroborated previous nd-
Pathophysiology 149
Figure 10.1A (schematic) and B (dissection) The relationship of the cavernosal nerve and the cavernosal vessels to the
corpora cavernosa and the urethra. (Reproduced with permission from Hinman F Jr. Atlas of Urosurgical Anatomy.
Philadelphia: WB Saunders; 1993:445.)
Dorsal artery
Dorsal nerve
Circumflex vein
Retrocoronal
plexus
Deep dorsal vein
Tunica albuginea
Circumflex artery
Lateral vein
Cavernous
vein
Cavernous artery
Carvernous
nerve
Bulbourethral
artery
Crural
vein
Periprostic
plexus
Internal
pudendal
vein
Penile artery
Prostate Endopelvic
fascia
Levator ani
muscle
Membranous
urethra
Aberrant r.
cavernous artery
R. crus
Dorsal artery
Deep dorsal vein
(ligated)
Dorsal nerve
Communication
between
cavernous and
dorsal nerve
Cavernous
nerve
ings suggestive of a vascular mechanism. The authors
found the mechanism to be predominantly arterial in
nature, but reported that some men have a mixed
pattern, with both arterial and venous components.
They further speculated that postoperative arterial
insufciency may be the result of intraoperative injury
to the accessory pudendal artery, but did not have an
explanation for the postoperative VOD.
In order to evaluate molecular changes pertaining to
erection post-RP, Podlasek et al.
41
performed bilateral
cavernous nerve resections in a rat model and
investigated changes in nitric oxide synthase (NOS)
isoform expression and distribution. A profound
decrease in smooth-muscle and endothelium NOS-I
protein was observed in the corpora. Their work high-
lighted the importance of maintaining at least partial
innervation of the penis after surgical intervention and
further demonstrated that endothelial and smooth-
muscle changes resulting from loss of innervation
may account for the ED observed in prostatectomy
patients.
Goldstein et al.
42
evaluated the erectile function in
23 patients (mean age 65 years) who received radia-
tion therapy for prostate cancer and concluded that
vasculogenic impotence is the most consistent organic
erectile abnormality in irradiated patients. All 15
patients in the study who experienced post-XRT ED
had vascular abnormalities. In two, arteriography
revealed bilateral occlusive disease in the distal internal
pudendal and penile arteries overlying the pelvic
radiation eld. The authors also found a history of
cigarette smoking as a signicant risk factor in those
who developed ED after XRT.
Cavernosal artery insufciency has been implicated
in other studies involving irradiated patients. Zelefsky
and Eid
43
performed duplex sonography to determine
the etiology of ED after treatment for prostate cancer.
In those who underwent 3D-CRT, 24 (63%) had
arteriogenic dysfunction, 12 (32%) had cavernosal
VOD, one (2.5%) was classied as mixed, and one
(2.5%) as having neurogenic dysfunction.
Mulhall et al. undertook a study to evaluate the
magnitude of the radiation to the corporal bodies
when 3-D conformal delivery techniques are used.
44
Although these techniques have lessened the radiation
delivery to periprostatic tissues, the exact degree of
proximal corporal body exposure to radiation had not
been investigated prior to this study. Based on an
outline of the proximal corpora on axial computed
tomography, the authors used computer modeling to
calculate the dose delivered to this area. It was found
that the mean radiation delivered to the most proximal
2 cm of the corporeal bodies was 31 12.8 Gy (43%
of the total dose delivered to the target areas in
prostate and seminal vesicles). The data illustrated the
high doses delivered to the proximal corpora despite
improved techniques in 3D-CRT.
44
The mechanism of ED following brachytherapy (Fig.
10.2) has not been dened and is likely multifactorial.
Merrick et al.
45
evaluated the potential relationship
between radiation dose to the NVB and the develop-
ment of ED following prostate brachytherapy. They
found no relationship between radiation dose to the
NVB and the development of postbrachytherapy ED
(median follow-up 37 months), but speculated that
such a difference may become evident with additional
follow-up.The only difference in demographics between
those who developed ED and those who did not was
that the latter were, on average, 4 years younger. Their
ndings refuted earlier reports by DiBiase et al.,
46
who
had reported that higher doses of radiation therapy
after brachytherapy were predictive of ED and that
those with ED had received NVB doses that were
signicantly higher than the prescription dose. The
proximity of the NVBs to the peripheral zone, an area
that requires the full prescription dose, restricts the
ability of radiation oncologists to reduce the dose to
those structures.
47
Stock et al.
25
assessed erectile func-
tion after prostate brachytherapy and analyzed those
factors affecting potency preservation. The authors
provided a 6-year follow-up and found pretreatment
ED as the factor with the most signicant effect on
posttreatment potency. The other factor found to have
a signicant negative effect on potency in univariate
and multivariate analyses was high implant dose. If
the D90 (dose delivered to 90% of the gland on a
dosevolume histogram from the 1-month computed
tomography-based dosimetric analysis) was greater
than 160 Gy for I-125 and greater than 100 Gy for
Pd-103, then dose had a signicant negative impact on
preservation of potency (58% preservation for those in
the high-dose versus 64% in the low-dose group).
Potters et al. assessed potency after brachytherapy
and accounted for such confounding variables as the
150 Prostate Cancer and Erectile Dysfunction
Figure 10.2 Radioactive seeds used in prostate
brachytherapy. 2002, Bruce Fritz. Reproduced by kind
permission of Bruce Fritz.
use of XRT, neoadjuvant androgen ablation, and patient
age.
47
This investigation is the largest prospective study
to date on ultrasound-guided transperineal brachy-
therapy and reported a 5-year actuarial potency rate of
76% for those treated with monotherapy. Addition of
XRT decreased potency rates by 20%. Potency was
further reduced to 52% when neoadjuvant androgen
ablation was added. With a median age of 69, only
41% of the patients in this study were potent pretreat-
ment and age was a signicant factor for predicting
posttherapy potency.
47
In 1992, Migliari et al. evaluated sleep-related
erections in 5 patients with stage T3N0M0 prostate
cancer treated solely with 50 mg once daily of the non-
steroidal antiandrogen, Casodex.
48
Mean follow-up
was 6 months. No signicant modications in regard to
number of nocturnal penile tumescence (NPT) episodes,
maximum penile circumference, and total rigidity time
were found before and after therapy. Only one patient
reported a decrease in sexual drive and libido. The
authors concluded that pure antiandrogen therapy did
not seem to interfere signicantly with the erectile
capability of men with prostate cancer. The study is
weakened by a small number of patients and con-
lusions that are based on a self-reported non-validated
questionnaire and NPT testing. In contrast to pure
antiandrogens that exert their effects by competitive
inhibition of androgen binding to the cytosolic
androgen receptor, administration of a luteinizing
hormone-releasing hormone agonist and reduction of
serum testosterone concentrations to a castrate level
have been shown to exert a signicant inhibitory effect
on sexual desire, sexual interest, and sexual intercourse,
with signicant changes in frequency, magnitude,
duration, and rigidity of nocturnal erections observed
in all patients.
49
Cryoablation of the prostate has been shown to have
a signicant adverse impact on the quality of erection
after treatment.
50
However, there is a paucity of data on
the exact mechanisms responsible for the dysfunction.
Aboseif et al.
51
prospectively examined erectile func-
tion before and 6 months after therapy in 15 sexually
active men who underwent cryoablation by using high-
resolution ultrasonography and color pulsed-Doppler
spectral analysis. At 6-months follow-up, ED persisted
in nine, with minimal or no response to the intra-
cavernosal prostaglandin E
1
injections. The authors
documented a signicant decrease in the peak velocity
of blood flow within cavernosal arteries and a signi-
cant increase in the time to achieve peak arterial flow.
It was concluded that vascular factors play a signicant
role in the etiology of postcryotherapy ED.
FOLLOW-UP AND TREATMENT
Both the diagnosis of prostate cancer and the variety
of existing treatments can have a devastating negative
impact on the psychological state of the patient.
Although ED can exacerbate the negative outlook for
the affected patient, there are very few, if any, patients
who cannot be treated by one of the available
modalities. It has been reported that, despite highly
successful treatment modalities for ED following RP,
fewer than 50% of patients seek therapy.
52
For patients
with ED who have had RP, Baniel et al. advocate a
progressive local treatment protocol whereby a four-
phase treatment regimen is instituted: phase I vacuum
erection device (VED); phase II sildenal; phase III
intracorporal injection; and phase IV intracorporal
injection plus the VED.
53
With a 1-year follow-up, this
progressive treatment method gave a positive response
in 94% of patients. The authors found intracorporal
injection to be the most effective method. In this study,
the positive response rate to sildenal (30% in phase
II) was lower than that reported by Zippe et al., who
reported a 72% success rate with sildenal in those
who had undergone bilateral nerve-sparing RP and
50% in those after unilateral nerve-sparing RP.
54
The
lower success rate in the former study is likely due to
a higher percentage of patients with non-nerve-sparing
RP. Indeed, introduction of sildenal (Viagra: Pzer,
New York, NY) in early 1998 revolutionized the
management strategies for patients with ED. Sildenal
citrate, an inhibitor of the enzyme phosphodiesterase
type 5, augments cavernosal smooth-muscle relaxation
through a cascade of events that involves increased
concentrations of cyclic guanosine monophosphate
and decreased intracellular calcium concentrations.
The whole mechanism, however, is dependent on the
presence of nitric oxide released by cavernous nerve
endings. Therefore, it is only intuitive that poor
sildenal efcacy would be expected under conditions
where the cavernosal nerves have been compromised.
Zagaja et al.
55
surveyed 170 men who had undergone
RP, but had not recovered natural erections sufcient
for intercourse, and had subsequently received
sildenal (50100 mg). The overall response rate was
29% in the 120 men who began taking the sildenal at
least 12 months after surgery; 80% of those younger
than 55 years of age in whom both nerves had been
preserved had a successful response. In contrast,
regardless of age, none of the patients in whom both
nerves had been excised reported a successful
response. Similarly, Feng et al.
56
reported that 71% of
bilateral nerve-sparing and 80% of unilateral nerve-
sparing groups had a positive response to sildenal,
compared to 6% of patients who had undergone a
non-nerve-sparing operation.
Montorsi et al. reported on intracavernous injections
of alprostadil administered in the early postoperative
phase after RP to facilitate corporal oxygenation and
limit the development of hypoxia-induced tissue
Follow-up and Treatment 151
damage.
57
The authors instituted this treatment
strategy 1 month after the surgical procedure (three
times a week for 3 months). Their prospective study
has shown that early postoperative administration of
alprostadil injections signicantly increases the recovery
rate of spontaneous erections after nerve-sparing RP.
The authors were able to demonstrate that at the end
of the 3-month course of empiric alprostadil injec-
tions, 67% (8 of 12) recovered spontaneous erections
sufcient for satisfactory sexual intercourse, as com-
pared with only 20% in the group that did not receive
postoperative alprostadil injections. Based on the results
of nocturnal testing and color Doppler sonographic
studies, failure of recovery of spontaneous erections in
the non-alprostadil group was attributed to cavernous
VOD (53%), arterial insufciency (13%), and nerve
injury (13%).
57
Of note, the authors documented a
17% incidence of complications ranging from priapism
to cavernous hematoma in the alprostadil group and
cautioned about the need for exhaustive documentation
of these possibilities in the consent form. Although the
study is limited by a lack of pretreatment evaluation
of erectile function, it is signicant in demonstrating
the positive impact of early postoperative cavernous
oxygenation induced by alprostadil injections.
The use of VEDs after RRP has been reported by a
number of authors. Baniel et al. evaluated response to
various modalities offered for the treatment of post-
RRP ED in 85 patients.
53
In this stepped-care model,
patients were allowed to progress to the next level of
care if they failed earlier more conservative measures.
Interestingly, a large percentage of the patients (92%)
responded to therapy with the VED. However, only
14% agreed to continue with this treatment regimen
at home. Zippe et al. have reported encouraging pre-
liminary results with early institution of daily VED
therapy soon after RRP.
58
The compliance rate was
found to be high (80%) in this early therapy group,
the complications were low, and 80% of patients
reported having sexual activity by using VED at a
frequency of twice per week.
Most experts agree that recovery of nerve function
beyond 2 years following RRP is unlikely and failure of
conservative means is an indication for more aggressive
measures. Patients who fail oral pharmacotherapy or
cavernosal injection therapy after RP may be candi-
dates for semirigid or inflatable penile prosthesis place-
ment. The use of penile prostheses in this group is
associated with low morbidity and patient satisfaction
rates around 85%.
59
Patients with a history of penile
prosthesis insertion after RP comprised 17% of a
group evaluated by Levine et al.
60
Overall patient and
partner satisfaction was 96.4% and 91.2%, respectively.
Of the respondents, 92.9% of patients and 90.1% of
partners would recommend the device to others. Of
the 85 men (65%) and 46 partners who completed the
modied Erectile Dysfunction Inventory of Treatment
Satisfaction survey, 90.6% and 82.6%, respectively,
were satised or very satised overall with the penile
prosthesis.
60
Most experienced implant surgeons place
the reservoirs for inflatable three-piece prostheses in
the retropubic space after RP. In these cases, the fascia
may be incised sharply using the Mezenbaum scissors
to enter the paravesical retropubic space. However,
some surgeons are reluctant to do this procedure blindly
and choose to make a second incision. The develop-
ment of the lock-out valve system incorporated in the
newer Mentor alpha-1 devices decreases the chance of
autoinflation and is especially helpful in cases where
an ectopic (i.e., non-retropubic) reservoir location is
chosen.
61
Although immediate sexual rehabilitation
with a prosthesis placed at the same time as the RP has
been suggested by some authors,
62
most experts do
not advocate this strategy since many patients will
regain sexual function with less invasive measures.
Nerve grafting has been advocated as a viable
alternative in men who are not candidates for nerve-
sparing during RP. Kim et al.
63
reported the results of
using an interposition sural nerve graft at the time of
RRP in an extended series of men with at least 1 year
of follow-up. Of the 23 men, 26% had spontaneous,
medically unassisted erections sufcient for sexual
intercourse with vaginal penetration and an additional
26% described 40% to 60% spontaneous erections
(fullness, no rigidity, not able to penetrate). Forty-three
percent had intercourse with sildenal and the greatest
return of function was observed at 18 months after
surgery. Despite the enthusiasm of the authors for this
technique, others have expressed reservations due to
technical difculties in precise identication of caver-
nous nerve endings and the distinct possibility that
patients who require resection of both NVBs (hence
the proposed candidate group for sural nerve grafting)
may not be curable with RP and are better served with
other treatment modalities.
64
Zelefsky et al. investigated the efcacy of sildenal
for patients with ED after radiotherapy for localized
prostate cancer.With a median follow-up of 19 months,
signicant improvement in the rmness of the erection
after sildenal was reported in 74%. Patients with
poor erectile function prior to radiation were less
likely (52%) to respond to oral pharmacotherapy with
sildenal whereas 90% of those with partial erections
had a signicant response to the medication.
65
A
prospective study by Weber et al.
66
documented 77%
with improved erectile function (sildenal 100 mg)
while Valicenti et al.
67
reported restoration of pre-XRT
sexual function in 21 of 23 patients who used sildenal
100 mg after radiation therapy. The latter study
evaluated 24 men with median age of 68 years (range
5177) who had 3D-CRT for localized prostate cancer
(median prescribed dose 70.2 Gy) and started taking
152 Prostate Cancer and Erectile Dysfunction
sildenal for relief of sexual dysfunction at a median
time of 1 year after completing 3D-CRT. Sexual func-
tion was assessed with the OLeary Brief Sexual Func-
tion Inventory before XRT, before sildenal therapy
(50 or 100 mg), and after completion of sildenal
therapy with a minimum of 2 months follow-up. Of
the 20 patients who were potent prior to any treat-
ments, 13 (65%) remained potent after XRT, with only
11% being fully potent. Potency was achieved in 91%
of the cohort after sildenal, with 30% reported as
being fully potent.
67
Incrocci et al. evaluated the efcacy of sildenal
citrate (Viagra) in patients with ED after 3D-CRT for
prostate cancer. Overall, 50% of the patients responded
to sildenal.
68
The authors cited lack of pretherapy
erectile function data as a potential weakness in their
study. They found a positive signicant correlation
between baseline scores after therapy and scores after
sildenal. However, no statistically signicant cor-
relation in efcacy of sildenal with age, concomitant
medication, or time after RT was noted. The authors
additionally commented that the higher dose of 100 mg
appeared to be needed for treatment of post-RT ED,
since 90% of their patients were on this dose.
68
Merrick et al. evaluated the efcacy of sildenal
citrate (Viagra) in patients with ED either before or
after prostate brachytherapy by an open-label, non-
randomized study. Fifty (80.6%) of 62 patients
responded favorably to sildenal.
69
Potters et al.
reported the largest prospective study to date on
potency after ultrasound-guided transperineal brachy-
therapy with a median follow-up of 34 months and
a median age of 68 years. In this study sildenal
improved erectile function in 83% treated with either
brachytherapy or brachytherapy + XRT. However, the
response rate was only 46% for those treated with
neoadjuvant androgen ablation and brachytherapy.
47
These ndings are illustrated in Table 10.3.
Penile prosthesis surgery is a viable option with
excellent patient satisfaction rates in post-XRT patients
who are not candidates for (or do not respond to) more
conservative measures. Outcome analysis of penile
implant surgery in 43 men following XRT for prostate
cancer has shown no evidence of infection or device
erosion with a 40-month mean follow-up.
70
The
authors reported that three of the patients were at
increased risk of infection due to reoperation for
device malfunction or previously failed operations at
other institutions. However, none of the redo
operations resulted in infections and the reoperation
was not due to previous irradiation in any of the cases.
They further concluded that penile prosthesis surgery
has no increased risk of infection or erosion and that
it is a safe and efcacious treatment modality for this
subpopulation.
FUTURE OUTCOMES
A number of studies have evaluated the psychological
and physiological impact of therapies for prostate
cancer on sexuality parameters other than erectile
function. While there is near unanimous agreement on
the psychological impact of the cancer diagnosis and
treatment on the quality of erections, the effects on
sexual desire and orgasm are more controversial.
Schover reported normal sexual desire and orgasm
after surgery despite disruption of the genital vaso-
congestion accompanying sexual arousal.
71
In contrast,
Koeman and colleagues evaluation of post-RP patients
indicated that half the patients reported diminished
sexual desire (libido) and arousal after the operation
and reported the same to occur in their partners.
72
During their dry orgasm postoperatively, none of
the patients experienced the exquisite sensation of
inevitability, the so-called point of no return. Among
those who did experience orgasm, 50% complained
that their orgasmic sensation was weakened. Four
patients reported normal pleasure and sensation com-
pared to that experienced preoperatively. The authors
also reported involuntary loss of urine in 9 of 14
patients at orgasm and this was sufcient reason to
avoid any sexual contact with their partner in 5 of
these patients.
Helgason et al.
73
performed a similar study in
patients treated with RT and found that sexual desire
diminished among 77% after treatment. Furthermore,
of those retaining orgasm after treatment, 47% reported
decreased orgasmic pleasure and 91% reduced ejacu-
lation volume. Of all men, 50% reported that quality
Future Outcomes 153
TABLE 10.3 Patients who developed impotence
after permanent brachytherapy treated with
sildenal.
Treatment n Successful
outcome to
sildenal
All patients 84 52 (62%)
All non-NAAD patients
Brachytherapy as monotherapy 15 12 (80%)
EBT and brachytherapy 21 18 (86%)
All NAAD patients
Brachytherapy as monotherapy 25 11 (44%)
EBT and brachytherapy 23 11 (48%)
Adapted from Potters et al.
47
NAAD, neoadjuvant androgen deprivation; EBT, external
beam radiation.
of life had decreased much or very much due to a
decline in the erectile capacity following XRT. The
authors concluded that XRT for prostate cancer is
associated with a reduction in sexual desire, erectile
capacity, and orgasmic functions and that this reduces
quality of life in the majority of patients.
In recent years, health-related quality-of-life (HRQOL)
outcomes have assumed a more important role as an
adjunct to survival in the assessment of the success
of therapies for prostate cancer. HRQOL discussions
related to the adverse effects of prostate cancer therapy
typically focus on incontinence and ED. Sexual func-
tion and sexual bother are two distinct, but related,
domains in HRQOL that have a signicant impact on
treatment decisions in early-stage prostate cancer. In
the context of HRQOL issues, sexual function
includes the broad domains of desire, the quality of
erections, and the ability to be physically intimate,
whereas sexual bother is dened as the degree of
interference or annoyance due to altered sexual
function.
23
Perez et al. compared self-reports of global quality
of life, sexuality, urinary continence, and physical
capabilities in 86 nerve-sparing patients, 89 standard-
prostatectomy patients, 74 prostatectomy patients who
used erectile aids, and a comparison group of 45
patients awaiting RP.
74
They showed that the best
outcomes in sexuality were reported by patients who
used erectile aids. Non-nerve-sparing prostatectomy
patients who did not use erectile aids scored worse in
most areas of sexuality than nerve-sparing patients
who did not use erectile aids. Interestingly, although
most patients reported problems in sexual and urinary
function, global quality of life did not appear to be
compromised following surgery.
Mazur and Merz assessed whether patients report
willingness to trade-off urologic adverse outcomes
urinary incontinence and total impotence for a
better chance of 5-year survival in the clinical setting
of prostate cancer.
75
Ninety-four percent of patients
were willing to choose worse short-term and better
long-term survival. The results suggested than many
patients were more concerned with long-term survival
in the clinical setting of prostate cancer than with
short-term treatment risks and that patients are more
willing to accept an impotence outcome (83%) than a
urinary incontinence outcome (62%), irrespective of
the patients reported frequency of sexual activity.
Other studies have shown that the general domain
of HRQOL which measures a patients own perception
of his well-being remains unchanged after surgery and
that, given the option, most patients would chose to
have surgery again.
8,10,22
Similarly, Kao et al.
14
report
on morbidity in men who underwent RP, mainly in the
late 1980s and early 1990s, performed by different
surgeons in the US military health care system. The
authors demonstrate that, despite negative effects on
the self-reported quality of life, most patients would
choose the same therapy again. Table 10.4 provides
descriptive statistics of multicenter patient self-reported
questionnaires on RP with illustration of patient satis-
faction measures and willingness to undergo repeat
surgery.
In a study by Braslis et al., 81% of those who saw
sexuality as a major issue and were unable to achieve
erections postoperatively stated that they would prob-
ably or denitely have their surgery again and felt that
the perceived survival benet of surgery outweighs the
loss of sexual function.
16
However, this choice must
not mask the frustration that is surely experienced at
the loss of sexual function. Additionally, the majority
154 Prostate Cancer and Erectile Dysfunction
TABLE 10.4 Descriptive statistics of multicenter
patient self-reported questionnaires on radical
prostatectomy.
Number of patients
(%)
Age at surgery (years)
70 or younger 853 (84.2)
Older than 70 160 (15.7)
Married 633 (62.5)
Not married 370 (36.5)
Unknown 10 (1.0)
Before 1990 145 (14.3)
After 1990 868 (85.7)
Impotence
Yes 780 (77.0)
No 94 (9.3)
Unknown 139 (13.7)
Sexual function satisfaction
Very satised 98 (9.7)
Somewhat satised 190 (18.8)
Somewhat unsatised 219 (21.6)
Very unsatised 362 (35.7)
Unknown 144 (14.2)
Quality of life
Better 149 (14.7)
Same 446 (44.0)
Worse 373 (36.8)
Unknown 45 (4.4)
Willing to be treated again
Yes 785 (77.5)
No 168 (16.6)
Unknown 60 (5.9)
Adapted from Kao et al.
14
of patients surveyed after surgery indicated that the
risk of impotence had been understated in preoperative
counseling.
16
Comparison of the nerve-sparing and non-nerve-
sparing techniques demonstrated that scores for sexual
function, sexual bother, physical function, and physical
limitation domains were signicantly better in the
nerve-sparing group.
76
Fowler et al. evaluated the
effects of urinary incontinence and sexual dysfunction
on the quality of life in a large group of Medicare
patients and reported that, overall, incontinence
appeared to have a more signicant impact on the
quality of life than sexual dysfunction.
10
Knowledge of the pretreatment sexual dysfunction
is critical in the ability to counsel patients properly on
individualized ideal therapies for prostate cancer. A
study of patient attitudes regarding treatment-related
ED at the time of diagnosis of cancer showed that
33.9% of patients reported either partial or complete
lack of erections and 31.1% were not sexually active
or active less than once per month. Although 55.4%
would be affected or very affected by lack of erections,
73.6% chose denitive treatment despite a 50%
chance of ED. Finally, 47.4% found such treatment-
induced ED to be an important or very important
problem.
77
Madalinska et al. performed a study within the
framework of a screening trial to compare the HRQOL
outcomes of RP and XRT. Patients referred for primary
radiotherapy were signicantly older than prostatec-
tomy patients. Analyses revealed poorer levels of
generic HRQOL after radiotherapy, although prostatec-
tomy patients reported signicantly higher posttreat-
ment incidences of ED.
78
However, evaluation of long-
term sexual function outcomes has shown similar rates
regardless of whether radiation or surgery is chosen as
treatment.
7,23
Additionally, Talcott et al.
7
demonstrated
that treatment choice signicantly affects sexual
function, but not sexual bother, suggesting that these
two are separate but independent psychometric con-
structs.Whereas sexual function is intimately related to
the ability to obtain and maintain erections satisfactory
for intercourse, sexual bother is related to a mans
resignation to loss of potency, availability of a sexual
partner, and the relative value of sexual function.
23
Their ndings also corroborated those by Perez et al.,
who had shown an overall quality of life after treat-
ment that was uniformly good.
74
Finally, Derby et al. have shown that physical
activity status is associated with ED, with the highest
risk among men who remain sedentary and the lowest
among those who remained active or initiated physical
activity.
79
They advocate early adoption of healthy
lifestyles as the best approach to reducing the burden
of ED on the health and well-being of older men, a
recommendation that is equally applicable to those
undergoing therapy for prostate cancer.
REFERENCES
1. Greenlee RT, Murray T, Bolden S et al. Cancer statistics,
2000. CA Cancer J Clin 2000; 50 (1):733.
2. Farkas A, Schneider D, Perrotti M et al. National trends in
the epidemiology of prostate cancer, 1973 to 1994: evidence
for the effectiveness of prostate-specic antigen screening.
Urology 1998; 52 (3):444448; discussion 448449.
3. Stanford JL, Stephenson, RA, Coyle LM et al. Prostate
cancer trends 19731995. Report no. 99. Bethesda, MD:
NIH; 1999.
4. Post PN, Stockton D, Davies TW et al. Striking increase in
incidence of prostate cancer in men aged 60 years without
improvement in prognosis. Br J Cancer 1999; 79 (1):1317.
5. Laumann EO, Paik A, Rosen RC. The epidemiology of
erectile dysfunction: results from the National Health and
Social Life Survey. Int J Impot Res 1999; 11 (suppl 1):
60S64.
6. Feldman HA, Goldstein I, Hatzichristou DG et al.
Impotence and its medical and psychosocial correlates:
results of the Massachusetts Male Aging Study. J Urol 1994;
151 (1):5461.
7. Talcott JA, Rieker P, Clark JA et al. Patient-reported
symptoms after primary therapy for early prostate cancer:
results of a prospective cohort study. J Clin Oncol 1998; 16
(1):275283.
8. Litwin MS, Nied RJ, Dhanani N. Health-related quality of
life in men with erectile dysfunction. J Gen Intern Med 1998;
13 (3):159166.
9. Fowler FJ Jr, Barry MJ, Lu-Yao G et al. Patient-reported
complications and follow-up treatment after radical
prostatectomy.The national Medicare experience: 19881990
(updated June 1993). Urology 1993; 42 (6):622629.
10. Fowler FJ Jr, Barry MJ, Lu-Yao G et al. Effect of radical
prostatectomy for prostate cancer on patient quality of life:
results from a Medicare survey. Urology 1995; 45
(6):10071013; discussion 10131015.
11. Walsh PC, Marschke P, Ricker D et al. Patient-reported
urinary continence and sexual function after anatomic radical
prostatectomy. Urology 2000; 55 (1):5861.
12. Stanford JL, Feng Z, Hamilton AS et al. Urinary and
sexual function after radical prostatectomy for clinically
localized prostate cancer: the Prostate Cancer Outcomes
Study. JAMA 2000; 283 (3):354360.
13. Siegel T, Moul JW, Spevak M et al. The development of
erectile dysfunction in men treated for prostate cancer. J Urol
2001; 165 (2):430435.
14. Kao TC, Cruess DF, Garner D et al. Multicenter patient
self-reporting questionnaire on impotence, incontinence and
stricture after radical prostatectomy. J Urol 2000; 163
(3):858864.
15. Catalona WJ, Carvalhal GF, Mager DE et al. Potency,
References 155
continence and complication rates in 1870 consecutive
radical retropubic prostatectomies. J Urol 1999; 162
(2):433438.
16. Braslis KG, Santa-Cruz C, Brickman AL et al. Quality of
life 12 months after radical prostatectomy. Br J Urol 1995; 75
(1):4853.
17. Benoit RM, Naslund MJ, Cohen JK. Complications after
radical retropubic prostatectomy in the Medicare population.
Urology 2000; 56 (1):116120.
18. Johannes CB, Araujo AB, Feldman HA et al. Incidence of
erectile dysfunction in men 40 to 69 years old: longitudinal
results from the Massachusetts Male Aging Study. J Urol
2000; 163 (2):460463.
19. Rabbani F, Stapleton AM, Kattan MW et al. Factors
predicting recovery of erections after radical prostatectomy.
J Urol 2000; 164 (6):19291934.
20. Geary ES, Dendinger TE, Freiha FS et al. Nerve-sparing
radical prostatectomy: a different view. J Urol 1995; 154
(1):145149.
21. Walsh PC, Donker PJ. Impotence following radical
prostatectomy: insight into etiology and prevention. J Urol
1982; 128 (3):492497.
22. Shekarriz B, Upadhyay J, Wood DP. Intraoperative,
perioperative, and long-term complications of radical
prostatectomy. Urol Clin North Am 2001; 28 (3):639653.
23. Litwin MS, Flanders SC, Pasta DJ et al. Sexual function
and bother after radical prostatectomy or radiation for
prostate cancer: multivariate quality-of-life analysis from
CaPSURE. Cancer of the Prostate Strategic Urologic
Research Endeavor. Urology 1999; 54 (3):503508.
24. Zelefsky MJ, Cowen D, Fuks Z et al. Long term tolerance
of high dose three-dimensional conformal radiotherapy in
patients with localized prostate carcinoma. Cancer 1999; 85
(11):24602468.
25. Stock RG, Kao J, Stone NN. Penile erectile function after
permanent radioactive seed implantation for treatment of
prostate cancer. J Urol 2001; 165 (2):436439.
26. Turner SL, Adams K, Bull CA et al. Sexual dysfunction
after radical radiation therapy for prostate cancer: a
prospective evaluation. Urology 1999; 54 (1):124129.
27. Fulmer BR, Bissonette EA, Petroni GR et al. Prospective
assessment of voiding and sexual function after treatment for
localized prostate carcinoma: comparison of radical
prostatectomy to hormonobrachytherapy with and without
external beam radiotherapy. Cancer 2001; 91 (11):20462055.
28. Sanchez-Ortiz RF, Broderick GA, Rovner ES et al.
Erectile function and quality of life after interstitial radiation
therapy for prostate cancer. Int J Impot Res 2000; 12
(suppl. 3):S18S24.
29. Chen CT, Valicenti RK, Lu J et al. Does hormonal
therapy influence sexual function in men receiving 3D
conformal radiation therapy for prostate cancer? Int J Radiat
Oncol Biol Phys 2001; 50 (3):591595.
30. Walsh PC, Lepor H, Eggleston JC. Radical prostatectomy
with preservation of sexual function: anatomical and
pathological considerations. Prostate 1983; 4 (5):473485.
31. Walsh PC, Marschke P, Catalona WJ et al. Efcacy of
rst-generation Cavermap to verify location and function of
cavernous nerves during radical prostatectomy: a multi-
institutional evaluation by experienced surgeons. Urology
2001; 57 (3):491494.
32. Bahnson RR, Catalona WJ. Papaverine testing of impo-
tent patients following nerve-sparing radical prostatectomy.
J Urol 1988; 139 (4):773774.
33. Breza J, Aboseif SR, Orvis BR et al. Detailed anatomy of
penile neurovascular structures: surgical signicance. J Urol
1989; 141 (2):437443.
34. Ginestie J, Ginestie JF, Romieu A. [Arteriography of
internal pudendal arteries and revascularization of the corpus
cavernosum in impotence of vascular etiology.] J Urol
Nephrol (Paris) 1976; 82 (suppl 2):509510.
35. Gray RR, Keresteci AG, St Louis EL et al. Investigation of
impotence by internal pudendal angiography: experience
with 73 cases. Radiology 1982; 144 (4):773780.
36. Polascik TJ, Walsh PC. Radical retropubic prostatectomy:
the influence of accessory pudendal arteries on the recovery
of sexual function. J Urol 1995; 154 (1):150152.
37. Aboseif S, Shinohara K, Breza J et al. Role of penile
vascular injury in erectile dysfunction after radical
prostatectomy. Br J Urol 1994; 73 (1):7582.
38. De Luca V, Pescatori ES, Taher B et al. Damage to
the erectile function following radical pelvic surgery:
prevalence of veno-occlusive dysfunction. Eur Urol 1996;
29 (1):3640.
39. Kim ED, Blackburn D, McVary KT. Post-radical
prostatectomy penile blood flow: assessment with color flow
Doppler ultrasound. J Urol 1994; 152 (6):22762279.
40. Mulhall JP, Graydon RJ. The hemodynamics of erectile
dysfunction following nerve-sparing radical retropubic
prostatectomy. Int J Impot Res 1996; 8 (2):9194.
41. Podlasek CA, Gonzalez CM, Zelner DJ et al. Analysis of
NOS isoform changes in a post radical prostatectomy model
of erectile dysfunction. Int J Impot Res 2001; 13 (suppl. 5):
S1S15.
42. Goldstein I, Feldman MI, Deckers PJ et al. Radiation-
associated impotence. A clinical study of its mechanism.
JAMA 1984; 251 (7):903910.
43. Zelefsky MJ, Eid JF. Elucidating the etiology of erectile
dysfunction after denitive therapy for prostatic cancer. Int J
Radiat Oncol Biol Phys 1998; 40 (1):129133.
44. Mulhall JP, Yonover P, Sethi A et al. Radiation exposure
to the corporeal bodies during 3-dimensional conformal
radiation therapy for prostate cancer. J Urol 2002; 167
(2):539542.
45. Merrick GS, Butler WM, Dorsey AT et al. A comparison
of radiation dose to the neurovascular bundles in men with
and without prostate brachytherapy-induced erectile
dysfunction. Int J Radiat Oncol Biol Phys 2000; 48
(4):10691074.
156 Prostate Cancer and Erectile Dysfunction
46. DiBiase SJ, Wallner K, Tralins K et al. Brachytherapy
radiation doses to the neurovascular bundles. Int J Radiat
Oncol Biol Phys 2000; 46 (5):13011307.
47. Potters L, Torre T, Fearn PA et al. Potency after per-
manent prostate brachytherapy for localized prostate cancer.
Int J Radiat Oncol Biol Phys 2001; 50 (5):12351242.
48. Migliari R, Muscas G, Usai E. Effect of Casodex on sleep-
related erections in patients with advanced prostate cancer.
J Urol 1992; 148 (2):338341.
49. Marumo K, Baba S, Murai M. Erectile function and
nocturnal penile tumescence in patients with prostate cancer
undergoing luteinizing hormone-releasing hormone agonist
therapy. Int J Urol 1999; 6 (1):1923.
50. Chaikin DC, Broderick GA, Malloy TR et al. Erectile
dysfunction following minimally invasive treatments for
prostate cancer. Urology 1996; 48 (1):100104.
51. Aboseif S, Shinohara K, Borirakchanyavat S et al. The
effect of cryosurgical ablation of the prostate on erectile
function. Br J Urol 1997; 80 (6):918922.
52. McCullough AR. Prevention and management of erectile
dysfunction following radical prostatectomy. Urol Clin North
Am 2001; 28 (3):613627.
53. Baniel J, Israilov S, Segenreich E et al. Comparative
evaluation of treatments for erectile dysfunction in patients
with prostate cancer after radical retropubic prostatectomy.
Br J Urol Int 2001; 88 (1):5862.
54. Zippe CD, Jhaveri FM, Klein EA et al. Role of Viagra
after radical prostatectomy. Urology 2000; 55 (2):241245.
55. Zagaja GP, Mhoon DA, Aikens JE et al. Sildenal in the
treatment of erectile dysfunction after radical prostatectomy.
Urology 2000; 56 (4):631634.
56. Feng MI, Huang S, Kaptein J et al. Effect of sildenal
citrate on post-radical prostatectomy erectile dysfunction.
J Urol 2000; 164 (6):19351938.
57. Montorsi F, Guazzoni G, Strambi LF et al. Recovery of
spontaneous erectile function after nerve-sparing radical
retropubic prostatectomy with and without early intra-
cavernous injections of alprostadil: results of a prospective,
randomized trial. J Urol 1997; 158 (4):14081410.
58. Zippe CD, Raina R, Thukral M et al. Management of
erectile dysfunction following radical prostatectomy. Curr
Urol Rep 2001; 2 (6):495503.
59. Mulcahy JJ. Erectile function after radical prostatectomy.
Semin Urol Oncol 2000; 18:7175.
60. Levine LA, Estrada CR, Morgentaler A. Mechanical
reliability and safety of, and patient satisfaction with the
Ambicor inflatable penile prosthesis: results of a 2 center
study. J Urol 2001; 166 (3):932937.
61. Wilson SK, Henry GD, Delk JR Jr et al. The mentor
Alpha 1 penile prosthesis with reservoir lock-out valve:
effective prevention of auto-inflation with improved
capability for ectopic reservoir placement. J Urol 2002; 168
(4):14751478.
62. Khoudary KP, DeWolf WC, Bruning CO 3rd et al.
Immediate sexual rehabilitation by simultaneous placement
of penile prosthesis in patients undergoing radical
prostatectomy: initial results in 50 patients. Urology 1997; 50
(3):395399.
63. Kim ED, Nath R, Slawin KM et al. Bilateral nerve grafting
during radical retropubic prostatectomy: extended follow-up.
Urology 2001; 58 (6):983987.
64. Walsh PC. Nerve grafts are rarely necessary and are
unlikely to improve sexual function in men undergoing
anatomic radical prostatectomy. Urology 2001; 57
(6):10201024.
65. Zelefsky MJ, McKee AB, Lee H et al. Efcacy of oral
sildenal in patients with erectile dysfunction after
radiotherapy for carcinoma of the prostate. Urology 1999; 53
(4):775778.
66. Weber DC, Bieri S, Kurtz JM et al. Prospective pilot
study of sildenal for treatment of postradiotherapy erectile
dysfunction in patients with prostate cancer. J Clin Oncol
1999; 17 (11):34443449.
67. Valicenti RK, Choi E, Chen C et al. Sildenal citrate
effectively reverses sexual dysfunction induced by three-
dimensional conformal radiation therapy. Urology 2001; 57
(4):769773.
68. Incrocci L, Koper PC, Hop WC et al. Sildenal citrate
(Viagra) and erectile dysfunction following external beam
radiotherapy for prostate cancer: a randomized, double-blind,
placebo-controlled, cross-over study. Int J Radiat Oncol Biol
Phys 2001; 51 (5):11901195.
69. Merrick GS, Butler WM, Lief JH et al. Is brachytherapy
comparable with radical prostatectomy and external-beam
radiation for clinically localized prostate cancer? Tech Urol
2001; 7 (1):1219.
70. Dubocq FM, Bianco FJ Jr, Maralani SJ et al. Outcome
analysis of penile implant surgery after external beam
radiation for prostate cancer. J Urol 1997; 158 (5):17871790.
71. Schover LR. Sexuality and fertility in urologic cancer
patients. Cancer 1987; 60 (suppl 3):553558.
72. Koeman M, van Driel MF, Schultz WC et al. Orgasm after
radical prostatectomy. Br J Urol 1996; 77 (6):861864.
73. Helgason AR, Fredrikson M, Adolfsson J et al. Decreased
sexual capacity after external radiation therapy for prostate
cancer impairs quality of life. Int J Radiat Oncol Biol Phys
1995; 32 (1):3339.
74. Perez MA, Meyerowitz BE, Lieskovsky G et al. Quality
of life and sexuality following radical prostatectomy in
patients with prostate cancer who use or do not use erectile
aids. Urology 1997; 50 (5):740746.
75. Mazur DJ, Merz JF. Older patients willingness to trade
off urologic adverse outcomes for a better chance at ve-year
survival in the clinical setting of prostate cancer. J Am Geriatr
Soc 1995; 43 (9):979984.
76. Gralnek D, Wessells H, Cui H et al. Differences in sexual
function and quality of life after nerve-sparing and nonnerve-
sparing radical retropubic prostatectomy. J Urol 2000; 163
(4):11661169; discussion 11691170.
References 157
77. Karakiewicz PI, Aprikian AG, Bazinet M et al. Patient
attitudes regarding treatment-related erectile dysfunction at
time of early detection of prostate cancer. Urology 1997; 50
(5):704709.
78. Madalinska JB, Essink-Bot ML, de Koning HJ et al.
Health-related quality-of-life effects of radical prostatectomy
and primary radiotherapy for screen-detected or clinically
diagnosed localized prostate cancer. J Clin Oncol 2001; 19
(6):16191628.
79. Derby CA, Mohr BA, Goldstein I et al. Modiable risk
factors and erectile dysfunction: can lifestyle changes modify
risk? Urology 2000; 56 (2):302306.
80. Quinlan DM, Epstein JI, Carter BS et al. Sexual function
following radical prostatectomy: influence of preservation of
neurovascular bundles. J Urol 1991; 145 (5):9981002.
81. Merrick GS, Wallner K, Butler WM et al. Short-term
sexual function after prostate brachytherapy. Int J Cancer
2001; 96 (5):313319.
82. Talcott JA, Clark JA, Stark PC et al. Long-term treatment
related complications of brachytherapy for early prostate
cancer: a survey of patients previously treated. J Urol 2001;
166 (2):494499.
158 Prostate Cancer and Erectile Dysfunction
INTRODUCTION
Erectile dysfunction (ED) is increasingly being con-
strued as an organic condition with vascular patho-
physiologic foundations. With the recent elucidation
of the molecular physiology of penile erection and
advanced diagnostic techniques, ED is now understood
to result from organic causes in up to 80% of cases,
1
with vascular causes accounting for the majority.
Conceptually, ED, like myocardial infarction (MI) and
stroke, may be viewed as a distinct end-organ con-
sequence of a shared vascular pathology.
Dened as the inability to achieve and maintain
penile rigidity sufcient for sexual intercourse, ED is
often a neurovascular disorder. In the healthy male,
relaxation of smooth-muscle cells (SMC) within the
penile vasculature and corpus cavernosum, along with
engorgement of corporal sinusoids, causes penile blood
pressure to approach or exceed mean arterial pressure
(MAP) (Fig. 11.1). A venoocclusive mechanism is
instrumental in the maintenance of penile erection.
This involves venous outflow obstruction as a result of
stretching of the tunica albuginea with compression of
the subtunical venous plexus. SMC relaxation within
Cardiovascular Safety of Sexual Activity and
Phosphodiesterase Type 5 Inhibition
Robert A. Kloner
CHAPTER 11
Figure 11.1 Anatomy and mechanisms of penile erection. (Adapted with permission from Lue TF. Erectile dysfunction.
N Engl J Med 2000; 342:18021813. Copyright 2000 Massachusetts Medical Society. All rights reserved.)
Prostate Deep
dorsal
vein
Dorsal
artery
Dorsal nerve
(somatic)
Circumflex
artery
Circumflex
vein
Cavernous
nerve
(autonomic)
Dorsal
artery
Dorsal nerve
(somatic)
Sinusoidal
spaces
Helicine
arteries
Trabecular
smooth
muscle
Subtunical
venular plexus
Deep
dorsal vein
Tunica
albuginea
Corpora
cavernosa
Cavernous
artery
Erect
Cavernous
artery
Flaccid
the penile vasculature and trabecular erectile tissue is
pivotal in generating a normal physiologic erection.
Either arterial pathology or a suboptimal venoocclusive
mechanism (e.g., venous leak) underlies vasculogenic
ED.
24
Nitric oxide (NO), previously termed endothelium-
derived relaxing factor, is the central mediator of
penile erection. A complex signal-transduction cascade
involving NO results in SMC relaxation with increased
penile blood flow and tissue expansion.
Produced from L-arginine by NO synthase (NOS),
NO is released at cavernous non-adrenergic, non-
cholinergic nerve terminals as a result of sexual arousal
and increased parasympathetic (cholinergic) input;
NO is also released from endothelial cells within the
vasculature of the corpora cavernosa. NO then binds to
and activates soluble guanylate cyclase (sGC) in SMC,
and the synthesis of cyclic guanosine monophosphate
(cGMP) is upregulated. Membrane hyperpolarization
with closure of gated transmembrane calcium (Ca
2+
)
channels via phosphorylation results in a reduced influx
of Ca
2+
across the cell membrane. In addition, Ca
2+
is
sequestered by cellular organelles (e.g., sarcoplasmic
reticulum) through an energy-dependent process, and
the decline in intracellular Ca
2+
is associated with
reduced formation of actomyosin complexes and
diminished smooth-muscle tone. SMC relaxation with
vasodilation of arteries and arterioles occurs; these
structures ll with blood, penile tissue expansion
occurs, and these processes result in penile erection
(Fig. 11.2).
3,5
The NOsGCcGMP signal-transduction cascade has
been recognized as a potential pharmacotherapeutic
target in ED.
6
Phosphodiesterase type 5 (PDE5)
degrades (inactivates) intracellular cGMP, and this
enzymatic hydrolysis terminates its smooth-muscle-
relaxing effects. Conversely, inhibition of this process
by selective PDE5 inhibitors prevents the breakdown
of cGMP, resulting in higher concentrations of this
substance with subsequent improved vasodilation and
enhanced erections.
SHARED RISK FACTORS BETWEEN
CARDIOVASCULAR DISEASE AND
ERECTILE DYSFUNCTION
Epidemiologic, physiologic, and clinical links exist
between coronary artery disease (CAD) and ED. With
advancing median ages in industrialized societies,
including the USA, the prevalences of both conditions
are projected to increase.
7,8
Risk factors for CAD,
including hypertension, dyslipidemia, diabetes, smoking,
obesity, and lack of physical activity, also either
directly or indirectly elevate the risk of developing
ED.
912
Hypertension
Abundant evidence lends credence to the close associ-
ation between hypertension and ED. In an early study
of 302 patients, ED was present in 6.9% of normo-
tensive men, 17.1% of men with untreated hyper-
tension, and 24.6% of men with treated hypertension
(P < 0.001).
13
The Massachusetts Male Aging Study
(MMAS), a random-sample observational study of
men aged 4070 years, showed that ED was more
160 Cardiovascular Safety of Sexual Activity and Phosphodiesterase Type 5 Inhibition
Hyperpolarization
Relaxation
+
+
-
-
NO
GC
PLC PL
3
Endothelium
Nerves
Na
+
/K
+
-ATPase
Ca
2+
K
+
PKA
PLB cGMP PKG
Ca
2+
-channels
Ca
2+
-ATPase
IP
3
-receptor
K
+
-channels
[Ca
2+
]
Ca
2+
Figure 11.2 Schematic
representation of the
mechanisms involved in penile
smooth-muscle relaxation by the
cyclic guanosine monophosphate
(cGMP) pathway. GC, guanylate
cyclase; IP
3
, inositol triphosphate;
PKA, protein kinase A; PKG,
protein kinase G; PLB,
phospholipase B; PLC,
phospholipase C; NO, nitric
oxide. - indicates inhibitory effect;
+ indicates stimulating effect.
(Adapted with permission from
Senz de Tejada I. Molecular
mechanisms for the regulation of
penile smooth muscle
contractility. Int J Impot Res 2000;
12 (suppl 4):S34S38. Copyright
2000 Nature Publishing
Group.)
prevalent in men with hypertension than the age-
matched general population.
11
The age-adjusted
probability of complete ED was 15% in patients with
treated hypertension compared with 9.6% in the
entire sample.
11
Further, in a hypertension clinic outpatient study
that used the International Index of Erectile Function,
68.3% of men with hypertension exhibited some
degree of ED, and ED was more severe in patients with
hypertension than in the general population.
9
Finally,
a 4-month study of 101 patients from a Danish out-
patient hypertension clinic revealed that 27% of men
with hypertension experienced ED compared with 4%
of men in the general regional population (P < 0.001).
14
Of note from this study, the cause of ED in 89% of
patients with hypertension was arterial dysfunction,
14
which was assessed using Doppler ultrasonography of
the penile arteries with or without papaverine (60 mg)
injection. In addition, the prevalence of ED was related
to the degree of secondary organ manifestation.
Advanced hypertension according to World Health
Organization classications (P = 0.01), intermittent
claudication (P = 0.001), and ischemic heart disease
(P = 0.005) were the most reliable determinants of ED.
Pathophysiologic connections between ED and
hypertension implicate endothelial dysfunction as a
shared mechanism. Endothelial dysfunction implies
that the blood vessels are incapable of normal vaso-
dilation through a defect in the NOcGMP pathway.
Indeed, the term ED may stand for endothelial dys-
function almost as readily as erectile dysfunction.
The diminished vascular compliance, increased vascular
thickness, and endothelial dysfunction associated with
hypertension can lead to increased vascular tissue
resistance within the corpus cavernosum, inadequate
penile blood flow, and, hence, ED.
Recent in vivo and in vitro studies performed in
spontaneously hypertensive rats demonstrated that
hypertension markedly compromised erectile function.
15
Endothelial dysfunction was responsible for impaired
SMC relaxation in both the aortas and corpora caver-
nosa of these organisms. An imbalance of endogenous
vasoactive substances, including relative declines in
NO bioavailability and the cyclic nucleotide signal-
transduction cascade in general, occurs in both the
systemic circulation and penile vascular beds in the
setting of hypertension. The NO-decient state is a
potential unifying theme in patients with ED secondary
to cardiovascular disease or diabetes, among other
conditions.
16
Certain drugs prescribed to treat hypertension,
including certain thiazide diuretics, are also considered
to be risk factors for the development of ED.
11
However, the wide variety of antihypertensive agents
(with different mechanisms of action) that have been
reported to induce ED suggests that hypertension per se
contributes more signicantly to the development of
ED than administration of individual antihypertensive
drugs in particular.
14,17
All antihypertensive drugs do not exert equal effects
on erectile function. In a cross-over study of 160 men
aged 4049 years with newly diagnosed hypertension
and no history of ED, sexual activity signicantly
declined with administration of the -blocker carvedilol
but not the angiotensin-receptor blocker (ARB)
valsartan over 16 weeks.
18
In addition, a study of men
and women at 110160% of their ideal body weight
who took chlorthalidone showed that adherence to a
weight-loss diet signicantly improved well-being and
measures of distress, including sexual problems.
19
Atherosclerosis
Atherosclerosis is the most common cause of vasculo-
genic ED,
1
partly because atheromata, which constitute
a form of endothelial insult, can reduce penile perfusion.
In an in vivo study, 93% of animals with experimen-
tally induced stenosis (luminal occlusion >50%) of the
iliohypogastric arteries exhibited ED, although other
contributing factors, such as hypertension-induced
alterations of broelastic properties of the corpus
cavernosum, could not be ruled out.
20
Even in the
absence of stenotic lesions, hypercholesterolemia was
associated with an increased probability of ED.
Balloon de-endothelialization of the common and
internal iliac arteries and hypercholesterolemia induced
by a high-cholesterol diet of 1.6% cholesterol and 4%
triglyceride over 8 weeks resulted in impaired erectile
function in 16 (76%) of 21 animals.
20
Advancing age, which is also a risk factor for athero-
sclerosis, correlates with reduced NOSNOsGC
cGMP signaling.
2,21,22
Downregulation of this signal-
transduction mechanism in the setting of vascular
disease with aging renders PDE5 inhibition a highly
rational pharmacotherapeutic approach.
6
Dyslipidemia
High levels of total cholesterol and low levels of high-
density lipoprotein (HDL) cholesterol are risk factors
for ED as well as CAD. In the MMAS,
11
the probability
of ED was inversely correlated with HDL-cholesterol
levels. In men aged 4055 years, the age-adjusted
probability of moderate ED increased from 6.7% to
25% as HDL cholesterol decreased from 90 to
30 mg/dl. In men aged 5670 years, the probability of
complete ED increased from close to 0 to 16% as HDL
cholesterol decreased from 90 to 30 mg/dl (Fig. 11.3).
Further epidemiologic evidence supports the associ-
ation between dyslipidemia and ED. In a longitudinal
study, 3250 men aged 2583 (mean 51) years were
followed for 648 (mean 22) months after rst
Shared Risk Factors between Cardiovascular Disease and Erectile Dysfunction 161
presentation with dyslipidemia, at which time they did
not have ED.
10
In these men, every 1 mmol/l increase
in total cholesterol was associated with a 1.32 times
increase in the risk of incident ED, whereas every
1 mmol/l increase in HDL cholesterol was associated
with a 0.38 times decrease in the risk of ED. Men with
HDL cholesterol levels of greater than 1.55 mmol/l
(60 mg/dl) had 0.30 times the risk of ED compared
with men with HDL cholesterol levels of less than
0.78 mmol/l (30 mg/dl). Men with total cholesterol
levels greater than 6.21 mmol/l (240 mg/dl) had 1.83
times the risk of ED compared with men who had
total cholesterol values of less than 4.65 mmol/l
(180 mg/dl).
10
Diabetes
Approximately 3550% of men with diabetes mellitus
suffer from ED,
23,24
and ED is often the rst sign of
diabetes. The age-adjusted probability of complete ED
is 28% in men with treated diabetes versus 9.6% in
the general population;
11
the prevalence of ED in
3034-year-old men with diabetes has been estimated
at 15% and in men aged 60 years at 55%. The onset of
ED tends to occur earlier in patients with diabetes
than in the general population.
11
In a cohort study using self-report that evaluated
365 patients under age 30 years at diagnosis of type 1
diabetes, had diabetes for 10 years or longer, and took
162 Cardiovascular Safety of Sexual Activity and Phosphodiesterase Type 5 Inhibition
Probability
None Minimal Moderate Complete
Degree of impotence
0.0 A 0.5 1.0
Excessive alcohol: No
Yes
Age 4055 HDL-C (mg/dl): 30
60
90
Age 5670 HDL-C (mg/dl): 30
60
90
DHT (ng/ml): 0.01
0.1
1
Cortisol (g/dl): 5
25
75
DHEAS (g/ml): 0.5
1
5
10
Age-adjusted Probability
Degree of impotence
0.0 B 0.5 1.0
Hypoglycemic agents: No
Yes
Vasodilators: No
Yes
Cardiac drugs: 30
Yes (non-smoker)
Yes (smoker)
Antihypertensives: No
Yes (non-smoker)
Yes (smoker)
Figure 11.3 Association of
excessive alcohol consumption,
high-density lipoprotein
cholesterol (HDL-C), serum
hormone levels and medication
use with probability of erectile
dysfunction, imputed by
discriminant analyses in 1290
respondents to sexual activity
questionnaires. Probabilities for
HDL-C are stratied by age.
Other probabilities are age-
adjusted by regression. DHEAS,
dehydroepiandrosterone sulfate;
DHT, dihydrotestosterone.
(Adapted with permission from
Feldman HA, Goldstein I,
Hatzichristou DG et al. Impotence
and its medical and psychosocial
correlates: results of the
Massachusetts Male Aging Study.
J Urol 1994; 151(1):5461.)
insulin,
25
the prevalence of ED increased sharply with
advancing age. A more than 40-fold increase in ED
prevalence was evident in men from ages 21 to 30
years (1.1%) to those aged 43 years or older (47.1%;
P <0.0001). Moreover, 10 (83%) of 12 diabetes patients
aged 65 years or older experienced ED.
25
The prevalence of ED also increased dramatically
with increasing duration of diabetes: men with diabetes
for more than 35 years were 7.2 times as likely to
report ED as men with a duration of 1014 years
(P < 0.0001; Fig. 11.4). Other factors signicantly
associated with an increased likelihood of ED in men
with diabetes included higher body mass index and
systolic blood pressure (SBP), treated hypertension
(including diuretic treatment), and smoking.
25
A multi-
center study involving 1460 patients with diabetes
also demonstrated signicant associations among ED,
diabetes, and depression.
26
Putative pathophysiologic mechanisms linking
diabetes and ED include impaired neurogenic and
endothelium-mediated smooth-muscle relaxation; NO
deciency secondary to the formation of advanced
glycation end-products, which form protein cross-links
that scavenge NO; autonomic insufciency; and gonadal
dysfunction.
1,27
Mechanisms that may underlie the
association between hyperglycemia and vasculopathy
are presented in Figure 11.5.
Smoking
Both clinical and epidemiologic evidence suggests that
cigarette smoking exerts deleterious effects on erectile
function and is an independent risk factor for the
development of vasculogenic ED. These adverse effects
may be synergistically amplied in the presence of other
risk factors, such as hypertension and CAD.
11,12,28,29
A comprehensive review of the literature found that
smoking increased the likelihood of moderate to com-
plete ED twofold.
12
Smoking also increased the age-
adjusted probability of complete ED in patients taking
cardiac drugs (from 14% to 41%), antihypertensive
agents (from 7.5% to 21%), or vasodilators (from 21%
to 52%).
11
In a follow-up to the MMAS that analyzed
513 men aged 4070 years, baseline cigarette smoking,
passive cigarette smoke exposure, and cigar smoking
were associated with odds ratios of approximately
2.0 for developing ED over a 10-year period (P < 0.05
versus general population for all variables).
30
The pathophysiologic mechanisms responsible for
the damaging effects of smoking on erectile function
may also be related to endothelial dysfunction.
12
In
animal models, inhalation of cigarette smoke caused
failure of the venoocclusive mechanism and a decline
in arterial blood flow;
28
smoking also exerts well-
documented adverse effects on the vascular
endothelium in general.
12
Adding to any existing NO
deciency, free radicals and aromatic compounds from
cigarette smoke decrease endothelial NOS (eNOS)
activity and elicit superoxide-mediated NO degra-
dation, which tends to increase penile smooth-muscle
tone and promote flaccidity (Fig. 11.6).
12
Other puta-
tive mechanisms for the chronic adverse effects of
nicotine on erectile function include hypercoagulability,
increased platelet aggregation, release of free fatty
acids and catecholamines, and direct toxic effects of
nicotine and carbon monoxide on the vascular
endothelium.
28
Shared Risk Factors between Cardiovascular Disease and Erectile Dysfunction 163
Duration (years)
P
r
e
v
a
l
e
n
c
e
o
f
e
r
e
c
t
i
l
e
d
y
s
f
u
n
c
t
i
o
n
(
%
)
0
>1015 >1520 >2025 >2530 >3035 >3540 >40
100
25
50
75
Figure 11.4 Duration of diabetes
and prevalence of erectile
dysfunction. (Adapted with
permission from Klein R, Klein
BE, Lee KE et al. Prevalence of
self-reported erectile dysfunction
in people with long-term IDDM.
Diabetes Care 1996; 19:135141.
Copyright 1996 American
Diabetes Association.)
164 Cardiovascular Safety of Sexual Activity and Phosphodiesterase Type 5 Inhibition
Hyperglycemia
Glucose
auto-oxidation
Oxidative stress
O
2
/NO
Polyol
pathway
Protein
glycation
Oxidative
factors
Antioxidant
defense
Heparan
sulfate
NCV
Endoneural
blood flow
LDL
oxidation
Vasculopathy Retinopathy Neuropathy Nephropathy
NO-dependent
vasodilation
Ca
2+
VSMC
proliferation
Hemorrheologic
alterations
Coagulation
activation
Hypoxia
l
w
i
t
h
o
r
w
i
t
h
o
u
t
d
i
f
f
e
r
e
n
t
c
l
a
s
s
e
s
o
f
a
n
t
i
h
y
p
e
r
t
e
n
s
i
v
e
a
g
e
n
t
s
a
.
D
i
u
r
e
t
i
c
-
B
l
o
c
k
e
r
1
-
B
l
o
c
k
e
r
A
C
E
i
n
h
i
b
i
t
o
r
C
a
l
c
i
u
m
-
c
h
a
n
n
e
l
N
o
n
e
b
l
o
c
k
e
r
A
d
v
e
r
s
e
e
v
e
n
t
P
b
o
S
i
l
d
P
b
o
S
i
l
d
P
b
o
S
i
l
d
P
b
o
S
i
l
d
P
b
o
S
i
l
d
P
b
o
S
i
l
d
(
A
E
)
(
n
=
8
3
)
(
n
=
1
2
3
)
(
n
=
7
2
)
(
n
=
1
3
)
(
n
=
7
9
)
(
n
=
1
2
4
)
(
n
=
1
6
1
)
(
n
=
3
0
3
)
(
n
=
1
5
0
)
(
n
=
2
7
8
)
(
n
=
1
0
4
4
)
(
n
=
1
8
3
7
)
%
w
i
t
h
a
n
y
A
E
6
3
5
1
1
3
4
1
0
3
1
6
3
6
7
3
3
1
1
3
8
H
e
a
d
a
c
h
e
0
1
3
0
1
5
1
1
1
1
1
5
0
1
3
4
1
6
F
l
u
s
h
i
n
g
1
1
1
3
1
1
1
1
1
0
1
6
2
1
0
1
1
4
D
y
s
p
e
p
s
i
a
1
4
1
7
0
7
0
7
0
5
1
7
A
b
n
o
r
m
a
l
v
i
s
i
o
n
0
7
0
4
1
4
0
6
0
7
<
1
5
H
y
p
o
t
e
n
s
i
o
n
0
0
0
0
0
<
1
0
<
1
0
0
<
1
<
1
D
i
z
z
i
n
e
s
s
0
2
1
3
1
2
<
1
2
2
1
<
1
2
S
y
n
c
o
p
e
0
0
0
0
0
0
0
0
0
0
0
0
A
C
E
,
a
n
g
i
o
t
e
n
s
i
n
-
c
o
n
v
e
r
t
i
n
g
e
n
z
y
m
e
;
P
b
o
,
p
l
a
c
e
b
o
;
S
i
l
d
,
s
i
l
d
e
n
a
l
.
a
P
e
r
c
e
n
t
a
g
e
o
f
p
a
t
i
e
n
t
s
.
R
e
p
r
i
n
t
e
d
w
i
t
h
p
e
r
m
i
s
s
i
o
n
o
f
E
l
s
e
v
i
e
r
S
c
i
e
n
c
e
f
r
o
m
K
l
o
n
e
r
R
A
,
B
r
o
w
n
M
,
P
r
i
s
a
n
t
L
M
e
t
a
l
.
E
f
f
e
c
t
o
f
s
i
l
d
e
n
a
l
i
n
p
a
t
i
e
n
t
s
w
i
t
h
e
r
e
c
t
i
l
e
d
y
s
f
u
n
c
t
i
o
n
t
a
k
i
n
g
a
n
t
i
h
y
p
e
r
t
e
n
s
i
v
e
t
h
e
r
a
p
y
.
S
i
l
d
e
n
a
l
s
t
u
d
y
g
r
o
u
p
.
A
m
J
H
y
p
e
r
t
e
n
s
2
0
0
1
;
1
4
:
7
0
7
3
.
C
o
p
y
r
i
g
h
t
2
0
0
1
A
m
e
r
i
c
a
n
J
o
u
r
n
a
l
o
f
H
y
p
e
r
t
e
n
s
i
o
n
L
t
d
.
Phosphodiesterase Type 5 Inhibitornitrate Interactions 175
randomized to concomitant treatment with the PDE5
inhibitor together with these antihypertensive agents.
Arrhythmia
There is no convincing evidence from randomized,
double-blind, placebo-controlled clinical trials moni-
toring cardiovascular adverse events that therapy with
sildenal, tadalal, or vardenal is arrhythmogenic.
71,8386
PHOSPHODIESTERASE TYPE 5
INHIBITORNITRATE INTERACTIONS
Both clinical and in vivo evidence demonstrates that
sildenal can potentiate the hypotensive effects of
nitrates and NO donors. Sildenal causes a slight
decrease in systemic arterial pressure (8.0 mmHg
systolic and 5.5 mmHg diastolic) in most men, but
synergistic, profound decreases in systemic arterial
pressure have been reported in patients taking
sildenal with nitrates or NO donors.
87
In a double-blind, placebo-controlled cross-over
study that evaluated the effects of oral sildenal 50 mg
on BP in men with a history of stable angina taking the
NO donor isosorbide mononitrate (ISMN) or glyceryl
trinitrate (GTN) for stable angina,
88
sildenalISMN
reduced standing mean maximum SBP/DBP from
baseline by 52/29 mmHg versus a 25/15 mmHg
decline for placeboISMN (P < 0.001). Corresponding
decreases were 36/21 mmHg for sildenalGTN
versus 26/11 mmHg for placeboGTN (P < 0.01).
In an animal model, sildenal caused vasodilation in
normal coronary arteries, but the combined effect of
sildenal and isosorbide dinitrate resulted in large
and protracted declines in mean SBP (46.2%) and
coronary blood flow in coronary arteries with critical
stenoses.
89
Cases of MI in patients taking sildenal and nitrates
have been reported.
90,91
Such reports and other
evidence prompted the publication of a joint con-
sensus statement by the American Heart Association
and the American College of Cardiology
65
(Table 11.6)
as well as a statement in the manufacturers labeling
92
that sildenal is clearly contraindicated in patients
taking short- or long-acting nitrates.
It is important to note, however, that, in a sub-
sequent prescription event monitoring study involving
5601 patients ranging in age from 18 to 90 (mean 57.4)
years, there was no evidence of a higher incidence of
fatal MI or ischemic heart disease among men taking
sildenal as compared with the general population.
93
Other postmarketing studies have also suggested that
the incidences of MI and death among patients in
sildenal clinical trials were within the expected ranges
for age.
83
In fact, the incidence of serious cardiovascular
adverse events was comparable for patients who
176 Cardiovascular Safety of Sexual Activity and Phosphodiesterase Type 5 Inhibition
TABLE 11.6 Summary of clinical recommendations.
Administration of sildenal clearly contraindicated Concurrent nitrate therapy
Nitroglycerin
Isosorbide mononitrate
Isosorbide dinitrate
Pentaerythritol tetranitrate
Erythrityl tetranitrate
Isosorbide dinitrate/phenobarbital
Illicit drugs containing organic nitrates
Cardiovascular effects of sildenal may be Active coronary ischemia (not taking nitrates)
hazardous; prescribe based on individual CHF and borderline low blood pressure and
assessment borderline low volume status
Complicated, multidrug antihypertension regimen
Concurrent drugs that may prolong half-life of sildenal
Antibiotic/antifungal
Cardiovascular
HMG-CoA reductase inhibitors
Central nervous system
Other
CHF, congestive heart failure; HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A.
Adapted with permission from Cheitlin MD, Hutter AM Jr, Brindis RG et al. ACC/AHA expert consensus document. Use of
sildenal (Viagra) in patients with cardiovascular disease. American College of Cardiology/American Heart Association.
J Am Coll Cardiol 1999; 33:273282. Copyright 1999 American College of Cardiology and American Heart Association Inc.
Permission granted for one-time use. Further reproduction is not permitted without permission of the ACC/AHA.
received sildenal in placebo-controlled trials (or
open-label studies) and those who received placebo
94
(Table 11.7).
According to preliminary evidence, the combination
of the investigational PDE5 inhibitor tadalal with
nitroglycerin led to higher frequencies of BP
outliers;
95
in a subset of patients, tadalal augmented
the BP-lowering effects of nitrates. In clinical studies,
the incidence of MI in tadalal-treated patients was
0.39/100 person-years as compared with 1.1/100
person-years in placebo-treated patients.
86
Partly because of the prospect of life-threatening
PDE5 inhibitornitrate interactions, the importance of
taking a thorough medical history and performing
both a focused physical examination and selected
laboratory tests (e.g., lipids, glucose) before prescribing
PDE5 inhibitor (or any other) therapy for ED cannot
be overstated.
96
Such a workup can also help identify
possible underlying cardiovascular diseases or diabetes
associated with ED, while use of the Princeton guide-
lines for risk stratication can help to reassure both the
patient and clinician about the advisability of resuming
sexual activity.
62
Emergent presentation with angina related to
sexual exertion should prompt suspicion concerning
coadministration of sildenal and nitrates. In addition,
the health-care provider should remember to address,
and urge caution concerning, occult nitrate use,
including recreational nitrates such as amyl nitrate
(poppers) and nitrate sprays. Patient counseling
should also cover the matter of recommended bed-
room preparations, such as warming the room and
bedsheets to a comfortable temperature and banishing
any previously prescribed nitrates so that the patient is
not tempted to use them in the event of angina during
sexual activity after taking sildenal.
POTENTIAL FUTURE INDICATIONS
FOR PHOSPHODIESTERASE TYPE 5
INHIBITORS IN CARDIOVASCULAR
DISEASE
Preliminary evidence suggests that PDEs, enormously
heterogeneous enzymes with wide organ-tissue distri-
butions that mediate diverse physiologic functions,
are possible pharmacotherapeutic targets for clinical
indications apart from ED. Numerous PDE isozymes
belonging to seven families based on genetic and
functional characteristics have been identied in
mammals,
97
and elucidation of their physiologic roles
may culminate in the development of other therapeutic
uses for PDE inhibitors.
Potentially relevant to the management of cardio-
vascular diseases, PDEs are widely distributed through-
out the systemic vasculature. For instance, PDE1C
may represent an inducible PDE in atherosclerosis
because it is expressed by proliferating, but not
quiescent, vascular SMC.
66
Selective inhibitors of PDE3
may play roles as antithrombotic or cardiac inotropic
agents.
98
Experimental data also suggest a putative
role for PDE5 inhibitors in enhancing BP regulation
through effects on vascular architecture, endothelial
function, and large-artery compliance, among other
mechanisms.
21,22,99,100
In addition, lung tissue is richly endowed with
PDE5,
98
and preliminary clinical evidence suggests
that PDE5 inhibitors may be useful in the treatment of
pulmonary hypertension, which is suboptimally treated
TABLE 11.7 Incidence of serious cardiovascular events/myocardial infarction in patients receiving
sildenal or placebo in phase II and III studies.
Incidence (95% CI)
a
Sildenal Placebo
Phase II/III placebo-controlled studies
Serious cardiovascular events
b
4.1 (2.75.5) 5.7 (3.38.2)
Myocardial infarction 1.7 (0.82.6) 1.4 (0.22.6)
Phase II/III open-label extension studies
Serious cardiovascular events
b
3.5 (2.34.7)
Myocardial infarction 1.0 (0.31.6)
a
Incidence expressed as rate per 100 person-years of treatment. CI, condence interval.
b
Serious cardiovascular events include myocardial infarction, angina, and coronary artery disease.
(Reprinted with permission from Morales A, Gingell C, Collins M et al. Clinical safety of oral sildenal citrate (Viagra) in the
treatment of erectile dysfunction. Int J Impot Res 1998; 10:6973. Copyright 1998 Nature Publishing Group.)
Potential Future Indications for Phosphodiesterase Type 5 Inhibitors in Cardiovascular Disease 177
at present and has a generally dismal prognosis. In
a randomized controlled trial, sildenal attenuated
hypoxia-induced rises in PAP without signicant effects
on systemic BP in humans.
101
Further, a case report described a 21-year-old man
with pulmonary hypertension who was successfully
treated with sildenal at a dose of up to 100 mg ve
times a day.
102
After 3 months of such treatment, PAP
declined from 120 mmHg at presentation to 90 mmHg.
In addition, a small clinical trial demonstrated that
coadministration of sildenal with the prostacyclin
analog iloprost lowered mean PAP signicantly more
than iloprost alone: by 13.8 1.4 compared with 9.4
1.3 mmHg (P < 0.009).
103
Another therapeutic area in which PDE5 inhibitors
have potential value is CHF, the pathophysiology of
which may involve impaired endothelium-dependent,
NO-mediated vasodilation through downregulation of
the NOSNOsGCcyclic nucleotide signaling path-
way. In 48 patients with NYHA class II or III CHF,
single oral doses of sildenal 25 or 50 mg signicantly
elevated brachial-artery flow-mediated vasodilation
versus placebo.
104
CONCLUSIONS
Given the recent elucidation of the molecular mech-
anisms underlying physiologic erection and ED, which
involves the same NOSNOsGCcyclic-nucleotide
signal-transduction mechanism that influences both
systemic and penile vascular smooth-muscle relaxation,
it is not surprising that this condition has patho-
physiologic, clinical, and epidemiologic links with
cardiovascular disease. A largely vasculogenic disorder
in many patients, ED shares risk factors with CAD and
stroke, including hypertension, dyslipidemia, diabetes,
and smoking. Emerging evidence suggests that endo-
thelial dysfunction may underlie both ED and cardio-
vascular diseases.
Abundant ndings from studies concerning the
physiologic costs to the heart of sexual activities,
which are not dissimilar to those of daily activities,
support the essential safety of sex in men who do not
have serious cardiovascular conditions, such as very
recent MI or class III or IV CHF. Coitus is, in general,
a quite weak precipitant of coronary events.
Although the relative risks of coronary events and/or
death are transiently elevated after coitus, the absolute
risk during or shortly after any sexual encounter is
low. In addition, this exceedingly low absolute risk may
be further reduced through lifestyle changes, including
aerobic exercise appropriate to the patients overall
cardiac status. The Princeton guidelines are useful
when counseling cardiovascular patients who wish to
engage in, or resume, sexual activity and/or receive
treatment for ED.
On the other hand, concomitant administration of
sildenal and nitrates or NO donors is contraindicated
because sildenal can potentiate the hypotensive
effects of these agents. With adequate, individualized
diagnostic workup, including a medical history, focused
physical examination, and selected laboratory tests,
the vast majority of ED patients, including those with
stable CAD not managed with nitrates, are suitable
candidates for sildenal treatment of ED.
Large clinical trials involving sildenal, as well as the
PDE5 inhibitor tadalal, have demonstrated that these
agents do not signicantly elevate the incidence of MI
or other adverse cardiovascular events (e.g., syncope,
dizziness) as compared with placebo controls or men
on a number of agents with hypotensive effects (aside
from nitrates). Indeed, preliminary evidence suggests
that sildenal might have therapeutic utility in the
management of other cardiovascular conditions,
including pulmonary hypertension, systolic hyper-
tension, and CHF.
ACKNOWLEDGMENTS
Editorial assistance was provided by Stephen W.
Gutkin, Rete Biomedical Communications Corp.,
Ridgewood, New Jersey, USA.
ADDENDUM
Since this chapter was written it is recommended that
sildenal >25 mg not be given within 4 h of -blocker
administration, as some patients exhibit orthostatic
hypotension. The PDE5 inhibitor vardenal is contra-
indicated with -blocker use, and the PDE5 inhibitor
tadalal is contraindicated with -blockers other than
tamsulosin 0.4 mg.
REFERENCES
1. Sullivan ME, Keoghane SR, Miller MAW. Vascular risk
factors and erectile dysfunction. Br J Urol Int 2001;
87:838845.
2. Rehman J, Melman A. Pathophysiology of erectile
dysfunction. In: Lue TF, Goldstein M, eds. Impotence and
infertility. Atlas of clinical urology, vol. 1. Philadelphia:
Current Medicine; 1999:1.11.16.
3. Senz de Tejada I. Molecular mechanisms for the regulation
of penile smooth muscle contractility. Int J Impot Res 2000;
12 (suppl 4):S34S38.
4. Lue TF. Erectile dysfunction. N Engl J Med 2000;
342:18021813.
5. Lincoln TM. Cyclic GMP and mechanisms of vasodilation.
Pharmacol Ther 1989; 41:479502.
6. ckert S, Kthe A, Stief CG et al. Phosphodiesterase
isoenzyme as pharmacological targets in the treatment of
male erectile dysfunction. World J Urol 2001; 19:1422.
178 Cardiovascular Safety of Sexual Activity and Phosphodiesterase Type 5 Inhibition
7. Foot DK, Lewis RP, Pearson TA et al. Demographics
and cardiology, 19502050. J Am Coll Cardiol 2000; 35
(5 suppl B):66B80B.
8. Ayta IA, McKinlay JB, Krane RJ. The likely worldwide
increase in erectile dysfunction between 1995 and 2025 and
some possible policy consequences. Br J Urol Int 1999;
84:5056.
9. Burchardt M, Burchardt T, Baer L et al. Hypertension is
associated with severe erectile dysfunction. J Urol 2000;
164:11881191.
10. Wei M, Macera CA, Davis DR, et al. Total cholesterol and
high density lipoprotein cholesterol as important predictors
of erectile dysfunction. Am J Epidemiol 1994; 140:930937.
11. Feldman HA, Goldstein I, Hatzichristou DG et al.
Impotence and its medical and psychosocial correlates:
results of the Massachusetts Male Aging Study. J Urol 1994;
151:5461.
12. McVary KT, Carrier S, Wessells H. Smoking and erectile
dysfunction: evidence based analysis. J Urol 2001;
166:16241632.
13. Bulpitt CJ, Dollery CT, Carne S. Change in symptoms of
hypertensive patients after referral to hospital clinic. Br Heart
J 1976; 38:121128.
14. Jensen J, Lendorf A, Stimpel H et al. The prevalence and
etiology of impotence in 101 male hypertensive outpatients.
Am J Hypertens 1999; 12:271275.
15. Behr-Roussel D, Chamiot-Clerc P, Bernab J et al.
Hypertension per se drastically impairs erectile function in
spontaneously hypertensive rats. J Urol. 2002; 167:237
(abstract 929).
16. Sullivan ME, Thompson CS, Dashwood MR et al. Nitric
oxide and penile erection: is erectile dysfunction another
manifestation of vascular disease? Cardiovasc Res 1999;
43:658665.
17. Grimm RH Jr, Grandits GA, Prineas RJ et al. Long-term
effects on sexual function of ve antihypertensive drugs and
nutritional hygienic treatment in hypertensive men and
women. Treatment of Mild Hypertension Study (TOMHS).
Hypertension 1997; 29:814.
18. Fogari R, Zoppi A, Poletti L et al. Sexual activity in hyper-
tensive men treated with valsartan or carvedilol: a crossover
study. Am J Hypertens 2001; 14:2731.
19. Wassertheil-Smoller S, Blaufox MD, Oberman A et al.
Effect of antihypertensives on sexual function and quality of
life: the TAIM study. Ann Intern Med 1991; 114:613620.
20. Azadzoi KM, Goldstein I. Erectile dysfunction due to
atherosclerotic vascular disease: the development of an
animal model. J Urol 1992; 147:16751681.
21. Ruetten H, Zabel U, Linz W et al. Downregulation of
soluble guanylyl cyclase in young and aging spontaneously
hypertensive rats. Circ Res 1999; 85:534541.
22. Kl S, Bouloumi A, Mulsch A.Aging and chronic hyper-
tension decrease expression of rat aortic soluble guanylyl
cyclase. Hypertension 2000; 35:4347.
23. McCulloch DK, Campbell IW,Wu FC et al.The prevalence
of diabetic impotence. Diabetologia 1980; 18:279283.
24. Guay AT. Sexual dysfunction in the diabetic patient. Int J
Impot Res 2001; 13 (suppl. 5):S47S50.
25. Klein R, Klein BE, Lee KE et al. Prevalence of self-
reported erectile dysfunction in people with long-term IDDM.
Diabetes Care 1996; 19:135141.
26. De Berardis G, Franciosi M, Belglio M et al. Erectile
dysfunction and quality of life in type 2 diabetic patients: a
serious problem too often overlooked. Diabetes Care 2002;
25:284291.
27. Senz de Tejada I, Goldstein I, Azadzoi K et al. Impaired
neurogenic and endothelium-mediated relaxation of penile
smooth muscle from diabetic men with impotence. N Engl J
Med 1989; 320:10251030.
28. Shabsigh R, Fishman IJ, Schum C et al. Cigarette smoking
and other vascular risk factors in vasculogenic impotence.
Urology 1991; 38:227231.
29. Bortolotti A, Parazzini F, Colli E et al. The epidemiology
of erectile dysfunction and its risk factors. Int J Androl 1997;
20:323334.
30. Feldman HA, Johannes CB, Derby CA et al. Erectile
dysfunction and coronary risk factors: prospective results
from the Massachusetts Male Aging Study. Prev Med 2000;
30:328338.
31. Virag R, Bouilly P, Frydman D. Is impotence an arterial
disorder? A study of arterial risk factors in 440 impotent men.
Lancet 1985; 8422:181184.
32. OKane PD, Jackson G. Erectile dysfunction: is there
silent obstructive coronary artery disease? Int J Clin Pract
2001; 55:219220.
33. Kirby M, Jackson G, Betteridge J et al. Is erectile dys-
function a marker for cardiovascular disease? Int J Clin Pract
2001; 55:614618.
34. Goldstein I. The mutually reinforcing triad of depressive
symptoms, cardiovascular disease, and erectile dysfunction.
Am J Cardiol 2000; 86(suppl):41F45F.
35. Shabsigh R, Klein LT, Seidman S et al. Increased incidence
of depressive symptoms in men with erectile dysfunction.
Urology 1998; 52:848852.
36. Burchardt M, Burchardt T, Anastasiadis AG et al. Erectile
dysfunction is a marker for cardiovascular complications and
psychological functioning in men with hypertension. Int J
Impot Res 2001; 13:276281.
37. Buvat J, Lemaire A, Buvat-Herbaut M et al. Comparative
investigations in 26 impotent and 26 nonimpotent diabetic
patients. J Urol 1985; 133:3438.
38. Greenstein A, Chen J, Miller H et al. Does severity of
ischemic coronary disease correlate with erectile function?
Int J Impot Res 1997; 9:123126.
39. Carney RM, Rich MW, Freedland KE et al. Major
depressive disorder predicts cardiac events in patients with
coronary artery disease. Psychosom Med 1988; 50:627633.
References 179
40. Frasure-Smith N, Lesprance F, Talajic M. Depression
following myocardial infarction: impact on 6-month survival.
JAMA 1993; 270:18191825.
41. Derby CA, Mohr BA, Goldstein I et al. Modiable risk
factors and erectile dysfunction: can lifestyle changes modify
risk? Urology 2000; 56:302306.
42. Ralph D, McNicholas T. UK management guidelines for
erectile dysfunction. Br Med J 2000; 321:499503.
43. Davey Smith G, Frankel S, Yarnell J. Sex and death: are
they related? Findings from the Caerphilly Cohort Study.
Br Med J 1997; 315:16411645.
44. Palmore EB. Predictors of the longevity difference: a
25-year follow-up. Gerontologist 1982; 22:513518.
45. Persson G. Five-year mortality in a 70-year-old urban
population in relation to psychiatric diagnosis, personality,
sexuality and early parental death. Acta Psychiatr Scand
1981; 64:244253.
46. Drory Y, Shapira I, Fisman EZ et al. Myocardial ischemia
during sexual activity in patients with coronary artery disease.
Am J Cardiol 1995; 75:835837.
47. Hellerstein HK, Friedman EH. Sexual activity and the
postcoronary patient. Arch Intern Med 1970; 125:987999.
48. Larson JL, McNaughton MW, Kennedy JW et al. Heart
rate and blood pressure responses to sexual activity and a
stair-climbing test. Heart Lung 1980; 9:10251030.
49. Bohlen JG, Held JP, Sanderson MO et al. Heart rate, rate-
pressure product, and oxygen uptake during four sexual
activities. Arch Intern Med 1984; 144:17451748.
50. Stein RA. The effect of exercise training on heart rate
during coitus in the post myocardial infarction patient. Circu-
lation 1977; 55:738740.
51. Nemec ED, Manseld L, Kennedy JW. Heart rate and
blood pressure responses during sexual activity in normal
males. Am Heart J 1976; 92:274277.
52. DeBusk RF. Evaluating the cardiovascular tolerance for
sex. Am J Cardiol 2000; 86 (suppl):51F56F.
53. DeBusk RF. Sexual activity triggering myocardial
infarction: one less thing to worry about. JAMA 1996;
275:14471448.
54. Muller JE, Mittleman MA, Maclure M et al. Triggering
myocardial infarction by sexual activity: low absolute risk
and prevention by regular physical exertion. Determinants of
Myocardial Infarction Onset Study Investigators. JAMA 1996;
275:14051409.
55. Drory Y, Fisman EZ, Shapira Y et al. Ventricular
arrhythmias during sexual activity in patients with coronary
artery disease. Chest 1996; 109:922924.
56. Parzeller M, Raschka C, Bratzke H. Sudden cardio-
vascular death in correlation with sexual activity: results of a
medicolegal postmortem study from 19721998. Eur Heart J
2001; 22:610611.
57. Ueno M. The so-called coition death. Jpn J Legal Med
1963; 17:333340.
58. Anderson KM, Wilson PW, Odell PM et al. An updated
coronary risk prole: a statement for health professionals.
American Heart Association scientic statement. Circulation
1991; 83;356362.
59. Anderson KM, Odell PM,Wilson PW et al. Cardiovascular
disease risk proles. Am Heart J 1993; 121:293298.
60. Mller J, Ahlbom A, Hulting J et al. Sexual activity as a
trigger of myocardial infarction: a case-crossover analysis in
the Stockholm Heart Epidemiology Programme (SHEEP).
Heart 2001; 86:387390.
61. Kimmel SE. Sex and myocardial infarction: an epidemio-
logic perspective.Am J Cardiol 2000; 86 (suppl 2A):10F13F.
62. DeBusk R, Drory Y, Goldstein I et al. Management of
sexual dysfunction in patients with cardiovascular disease:
recommendations of the Princeton Consensus Panel. Am J
Cardiol 2000; 86:175181.
63. Zusman RM, Morales A, Glasser DB et al. Overall
cardiovascular prole of sildenal citrate. Am J Cardiol 1999;
83:35C44C.
64. Phillips BG, Kato M, Pesek CA et al. Sympathetic
activation by sildenal. Circulation 2000; 102:30683073.
65. Cheitlin MD, Hutter AM Jr, Brindis RG et al. ACC/AHA
expert consensus document. Use of sildenal (Viagra) in
patients with cardiovascular disease. American College of
Cardiology/American Heart Association. J Am Coll Cardiol
1999; 33:273282.
66. Rybalkin SD, Bornfeldt KE, Sonnenburg WK et al.
Calmodulin-stimulated cyclic nucleotide phosphodiesterase
(PDE1C) is induced in human arterial smooth muscle cells of
the synthetic, proliferative phenotype. J Clin Invest 1997;
100:26112621.
67. Wallis RM, Corbin JD, Francis SH et al.Tissue distribution
of phosphodiesterase families and the effects of sildenal on
tissue cyclic nucleotides, platelet function, and the contractile
responses of trabeculae carneae and aortic rings in vitro. Am
J Cardiol 1999; 83 (suppl 8A):3C12C.
68. Kloner RA. Sex and the patient with cardiovascular risk
factors: focus on sildenal. Am J Med 2000; 109 (suppl. 9A):
13S21S.
69. Montorsi F, McDermott TED, Morgan R et al. Efcacy
and safety of xed-dose oral sildenal in the treatment of
erectile dysfunction of various etiologies. Urology 1999;
53:10111018.
70. Brock GB, McMahon CG, Chen KK et al. Efcacy and
safety of tadalal in the treatment of erectile dysfunction:
results of integrated analyses. J Urol 2002; 168:13321336.
71. Porst H, Rosen R, Padma-Nathan H et al.The efcacy and
tolerability of vardenal, a new, oral, selective phosphodi-
esterase type 5 inhibitor, in patients with erectile dysfunction:
the rst at-home clinical trial. Int J Impot Res 2001;
13:192199.
72. Mittleman MA, Glasser DB, Orazem J et al. Incidence of
myocardial infarction and death in 53 clinical trials of Viagra
(sildenal citrate). J Am Coll Cardiol 2000; 35 (suppl. A):302
(abstract 807-6).
180 Cardiovascular Safety of Sexual Activity and Phosphodiesterase Type 5 Inhibition
73. Jackson G, Benjamin N, Jackson N et al. Effects of
sildenal citrate on human hemodynamics. Am J Cardiol
1999; 83 (suppl 5A):13C20C.
74. Herrmann HC, Chang G, Klugherz BD et al. Hemo-
dynamic effects of sildenal in men with severe coronary
artery disease. N Engl J Med 2000; 342:16221626.
75. Arruda-Olson AM, Mahoney DW, Nehra A et al. Cardio-
vascular effects of sildenal during exercise in men with
known or probable coronary artery disease: a randomized
crossover trial. JAMA 2002; 287:719725.
76. Fox KM, Thadani U, Ma PTS et al. Time to onset of
limiting angina during treadmill exercise in men with erectile
dysfunction and stable chronic angina: effect of sildenal
citrate. Circulation 2001; 104 (17 suppl. II):II-601II-602
(abstract 2846).
77. Conti CR, Pepine CJ, Sweeney M. Efcacy and safety of
sildenal citrate in the treatment of erectile dysfunction in
patients with ischemic heart disease. Am J Cardiol 1999;
83:29C34C.
78. Przyklenk K, Kloner RA. Sildenal citrate (Viagra) does
not exacerbate myocardial ischemia in canine models of
coronary artery stenosis. J Am Coll Cardiol 2001; 37:286292.
79. Kloner RA, Brown M, Prisant LM et al. Effect of sildenal
in patients with erectile dysfunction taking antihypertensive
therapy. Sildenal Study Group. Am J Hypertens 2001;
14:7073.
80. Zusman RM, Prisant LM, Brown MJ. Effect of sildenal
citrate on blood pressure and heart rate in men with erectile
dysfunction taking concomitant antihypertensive medication.
Sildenal Study Group. J Hypertens 2000; 18:18651869.
81. Webb DJ, Freestone S, Allen MJ et al. Sildenal citrate
and blood-pressure-lowering drugs: results of drug interaction
studies with an organic nitrate and a calcium antagonist. Am
J Cardiol 1999; 83 (suppl 5A):21C28C.
82. Kloner RA, Mitchell M, Emmick JT. Cardiovascular
effects of tadalal in patients on common antihypertensive
therapies. Am J Cardiol 2003; 92 (suppl 9A): 47M57M.
83. Sadovsky R, Miller T, Moskowitz M et al. Three-year
update of sildenal citrate (Viagra
1
2
Adenylyl
Decreased
cAMP
Endothelial
cell
Endothelins
Prostaglandin
F
2
Ca
2+
calmodulin
Myosin light-
chain kinase
Inactive
Active
Ca
2+
-calmodulin-myosin
light-chain kinase complex
Myosin light-chain
phosphorylation
Cycling of myosin
cross-bridges along actin
Smooth-muscle
contraction
Stimulation
Inhibition
cyclase-activity peptide, adenosine triphosphate (ATP),
serotonin (5-HT), dopamine, oxytocin, and histamine.
12
The roles of these molecules in the normal erectile
response remain controversial.
Changes in aging that may have
negative effects on erectile function
Many systemic changes and alterations in the structure
and function of the penis are independent of disease
and can have a negative effect on erectile function.
Systemic changes
As men age, they undergo a number of hormonal
changes (Table 18.1
13
), including a marked decrease in
the serum levels of testosterone and free testosterone.
14
Schiavi et al.
15
evaluated both sexual and erectile
function and levels of pituitary and gonadal hormones
in 77 healthy married men aged 4574 years. Aging
was negatively correlated with bioavailable testos-
terone (bT) and positively correlated with luteinizing
hormone (LH) levels. bT and the ratio of bT to LH
showed a close association with sexual behavior,
244 Erectile Dysfunction in the Elderly: Epidemiology, Etiology, and Approaches to Treatment
Figure 18.3 (Contd) B Mechanism of relaxation of the penile smooth-muscle cell. Nitric oxide (NO) is synthesized in the
non-adrenergic, non-cholinergic nerve terminal, as well as the endothelium, and diffuses through into the smooth cell.
NO stimulates formation of cyclic guanosine monophospate (cGMP), which produces smooth-muscle relaxation.
cAMP, cyclic adenosine monophosphate; ATP, adenosine triphosphate; PDE, phosphodiesterase; GTP, guanosine
triphosphate; eNOS, endothelial nitric oxide synthase. (Reprinted with permission from Lue TF. Erectile dysfunction.
N Engl J Med 2000; 2:1802.)
Cavernous
nerve
Adrenergic
Endothelial
cell
Cavernous
nerve
Non-adrenergic
non-cholinergic
Nitric
oxide
l-Arginine
Increased
inositol
triphosphate
Ca
2+
O
2
Acetylcholine
Prostaglandin E
1
Receptors
Smooth-muscle
cell
Forskolin
G-protein
Adenylyl
cyclase
ATP
cAMP
5' AMP
PDE
2, 3, 4
Papaverine
Ca
2+
K
+
cAMP specific
protein kinase
Endoplasmic
reticulum
Ca
2+
Decreased
Ca2+
Myosin
head detaches
from actin
cGMP
specific protein
kinase
Guanylyl
cylase
GTP
cGMP
5' GMP
PDE 5
K
+
Ca
2+
Sildenafil
papaverine
zaprinast
Smooth-
muscle
relaxation
eNOS
Stimulation
Inhibition
whereas total testosterone, estradiol, and prolactin did
not. However, the age-related effect of bT was a more
important determinant of reported sexual behavior
than was bT, independent of age. These investigators
concluded that changes in circulating hormones have
relatively little influence on erectile function in healthy
aging men.
Others have noted that a decline in serum testos-
terone occurs as part of the normal aging process and
that a small fraction (about 2%) of aging men have
subnormal serum testosterone levels. However, it is
unclear whether these decreases are associated with
the development of ED.
16
Thus, although testosterone
and free testosterone, like sexual function, decline with
age, the contribution of these hormonal changes to the
development of ED is thought to be only minor.
17
Penile structure
Signicant changes in penile structure occur with aging.
Collagen and elastic bers in the tunica albuginea are
key structures that permit increases in girth and length
of the penis during tumescence, and ultrastructural
analysis of penile biopsies has shown that the con-
centration of elastic bers decreases with age.
18
This
decrease results in a reduction in elasticity that could
contribute to ED in elderly men. Additional studies
have shown that there is a decrease of up to 35% in
the smooth-muscle content of the penis in men older
than 60 years of age. Decreases in the ratio between
corpus cavernosum smooth muscle and connective
tissue has been associated with increased likelihood of
diffuse venous leak that may contribute to ED.
19
It has
also been noted that the concentration of type III
collagen decreases and that for type I collagen increases
(Fig. 18.4) in the aging penis.
20
It has been suggested
that this change makes the corpus cavernosum less
compliant, reduces lling of vascular spaces, and also
contributes to venoocclusive dysfunction.
20
It has also
been hypothesized that changes in the collagen
content of the penis may result in chronic ischemia
that leads to loss of smooth-muscle cells.
21
Penile function
A large number of physiological and biochemical
changes in the aging penis may contribute to the
development of ED in older men. Not surprisingly,
Age-related Changes in Erectile Function 245
Figure 18.4 Collagen III (A) and collagen I (B) in the human penile corpus cavernosum tissue (400). The antibodies are
commercial and are used in a 1/30 dilution. The technique is standard immunohistochemistry.
A B
TABLE 18.1 Hormonal changes in the aging
male.
Hormone Change in the
aging male
Testosterone Decreased
(especially after
age 70)
Free testosterone Decreased
Bioavailable testosterone Decreased
Sex hormone-binding globulin Increased
Luteinizing hormone Increased
(especially after
age 70)
Follicle-stimulating hormone Increased
Dihydrotestosterone No change
Estradiol No change
Dehydroepiandrosterone Decreased
Dehydroepiandrosterone sulfate Decreased
Triiodothyronine Decreased
Insulin Decreased
Reprinted with permission from Margolese HC. The male
menopause and mood: testosterone decline and depression
in the aging male is there a link? Geriatr Psychiatry Neurol
2000; 13:93. Copyright 2000 by Sage Publications Inc.
mechanical sensitivity of the penis is decreased.
Rowland and associates
22
used psychophysical methods
to demonstrate marked increases in the threshold
for response to vibrotactile stimulation in both older
(mean age, 67.3 years) and diabetic men. Surprisingly,
the decline in penile sensory function in older men
was not matched by a corresponding reduction in the
sensitivity of the nger to vibrotactile stimulation (Fig.
18.5). It is not known whether this functional change
results from a decrease in the afferent nerve supply
to the penis or an increase in the thresholds for an
unchanged number of bers.
Animal studies have provided conflicting evidence
regarding changes in the innervation of the penis with
aging. Carrier et al.
23
reported that the number of NOS
immunoreactive bers is decreased in the penis of
aging rats, while Warburton and Santer
24
and Amenta
et al.
25
reported no signicant changes in the inner-
vation of the penis by CGRP- and VIP-containing
axons, respectively. It is important to note that these
studies provide limited insight regarding age-related
changes in the innervation density in the penis, because
results reflect density of immunostaining for products
within nerve bers that may change independently of
the number of penile sensory or sympathetic axons.
Increased understanding of the molecular pathways
involved in erectile function has prompted consider-
able research into the ways specic molecules related
to erection may change with age. Much of this attention
has focused on NO and NOS. As noted immediately
above, there is evidence that NOS-positive axons are
decreased in the penis of aged rats.
25
Garbn et al.
26
also noted that NOS activity per gram of penile tissue
was decreased by 63% in senescent (30-month-old)
rats.
Simple conclusions about age-related changes in the
amount or activity of NOS in the penis with aging are
complicated by the fact that there are three NOS
species: endothelial NOS (eNOS), inducible NOS
(iNOS), and neuronal NOS (nNOS).
27
eNOS (Fig. 18.6)
and nNOS are constitutive and activated in part by
an increased concentration of intracellular calcium and
calmodulin binding to the enzyme. iNOS is thought to
be associated primarily with macrophages and is
activated by specic cytokines as part of inflammatory
and immune responses.
28
Both eNOS and iNOS are
expressed in human cavernosal smooth-muscle cells,
and nNOS is expressed in nerve bers innervating the
penis.
29
Ferrini and associates reported that iNOS exhibits a
3.6-fold upregulation (as demonstrated by immuno-
staining) in penile smooth muscle of aged (24- to
30-month-old) rats. This rise was associated with a 3.6-
fold increase in apoptotic cells and a 2.0-fold increase
246 Erectile Dysfunction in the Elderly: Epidemiology, Etiology, and Approaches to Treatment
Figure 18.6 Endothelial nitric oxide synthase (eNOs) polyclonal antibody staining in the rat corpus cavernosum
(A, 200, 1:200 dilution) and the human corpus cavernosum (B, 400). The technique is standard immunohistochemistry
and the antibody commercial. Note the intense staining in the helicine artery and the smooth muscle (B).
A B
Figure 18.5 Rowland and associates
22
used
psychophysical methods to demonstrate marked increases
in the threshold for response to vibrotactile stimulation in
both older (mean age, 67.3 years) and diabetic men. This
decline in penile sensory function in older men is not
matched by a corresponding reduction in the sensitivity of
the nger to vibrotactile stimulation.
A
m
p
l
i
t
u
d
e
(
)
0
Finger Penis
5
1
2
3
4
Young
Aging
Diabetic
in collagen bers. These investigators suggested that
induction of iNOS and the resulting peroxynitrite for-
mation in the aging penis may result in both apoptosis
and proteolysis.
30
This suggestion is consistent with
the above-noted loss of smooth muscle in the aging
penis.
20
It is also reasonable to speculate that increased
iNOS expression may be associated with inflammatory
processes that could impair erectile function.
Haas et al.
31
reported that eNOS was upregulated in
the penile corporal tissue of aging rabbits. Despite this
increase, acetylcholine (ACh)-NO-mediated smooth-
muscle relaxation was markedly impaired in these
animals. These investigators showed further that caver-
nosal smooth-muscle relaxation in old, but not young,
rabbits was enhanced in the presence of the calcium
ionophore A23187. This suggests that the decreased
smooth-muscle relaxation observed in the aging penis
may be due to altered calcium flux, possibly at the
level of the calciumeNOS interaction. This speculation
is consistent with the recent suggestion that aging-
associated ion channel abnormalities may play a role
in the development of ED.
32
The age-related upregulation in eNOS activity in the
penis may be a response to reduced NO in this tissue.
Advanced glycation endproducts (AGEs) (Fig. 18.7)
accumulate in the aging penis in both corporal tissues
and the tunica albuginea.
33
AGEs form protein and
DNA adducts and cross-links. The cross-links may func-
tion as NO scavengers, reducing levels of this mediator
in corporal tissue. Studies in other tissues have shown
clearly that decreasing NO levels with a scavenger
(oxyhemoglobin) increases eNOS activity.
34
Thus, the
apparently paradoxical age-related rise in an enzyme
known to be involved in erectile function may actually
reflect lower levels of its product, NO.
One additional change in the aging penis is worthy
of mention. As already noted, CGRP appears to play a
signicant role in erectile function. Several years ago,
Wimalawansa
35
reported that levels of this peptide are
decreased in the penis of aged rats.
Some or all of the above-described changes in
structure, physiology, and molecular function may
underlie penile hemodynamic changes observed in
normal aging men. For example, Chung et al. showed
that advancing age was associated with a signicant
decrease in cavernous arterial flow velocity in response
to intracavernous injection of 10 g prostaglandin E
1
(Fig. 18.8).
36
Importantly, all of the men in this study
were free of any cardiovascular risk factors, so it is
unlikely that systemic disease was responsible for the
observed hemodynamic change.
In summary, studies to date strongly suggest that
the normal aging process produces changes in penile
anatomy and in the molecular and cellular processes
involved in erectile function. It thus seems reasonable
to suggest that aging, in the absence of disease, is
sufcient to produce ED in at least some men. This
conclusion is consistent with results reported by
Mulligan et al.,
7
who showed that age alone was a
signicant risk factor for ED. Advancing age also raises
the risk for a number of other diseases that in turn
increase the chance of ED.
COMORBIDITIES IN ED PATIENTS
ED is often associated with a number of other
conditions that are common in aging men. The most
important of these involve the cardiovascular system.
However, psychiatric disease can also increase the risk
Comorbidities in ED Patients 247
Figure 18.7 The advanced glycation endproduct, pyrraline
(1:400 dilution, 200 magnication) in the human penile
corpus cavernosum. Note the intense staining in the
smooth muscle and the media of the blood vessel.
Figure 18.8 Relationship between age and maximum
peak systolic velocity (PSV) in the cavernosal artery.
(Reprinted with permission from Chung WK, Park YY,
Kwon SW. The impact of aging on penile hemodynamics in
normal responders to pharmacological injection: a Doppler
sonographic study. J Urol 1997; 157:2129.)
Age (years)
P
S
V
m
a
x
(
c
m
/
s
)
0
20 30 40 50 60 70 80
100
20
40
60
80
P = 0.003 by simple regression test
for ED.
37
Diseases that occur with increasing frequency
in older men may interact with the normal age-related
changes in penile structure and function to increase
the risk for ED. For example, it was noted above that
levels of NO may be reduced in the aging penis, and
that this may increase the risk for ED. This suggestion
is consistent with ndings indicating that production of
NO by the vascular endothelium in other portions of
the body decreases with age.
38
Vascular NO levels are
also decreased in patients with hypertension.
39
NO
reductions associated with normal aging and those
resulting from elevated blood pressure may interact to
magnify the risk for ED signicantly.
Cardiovascular disease and ED
The prevalence of cardiovascular disease increases with
age,
40
and such disease markedly elevates the risk for
ED. Chew and associates assessed the prevalence of
ED and comorbidities in 1240 men between 18 and
91 years of age.
41
Of these, 488 men reported some
degree of ED. Hypertension, ischemic heart disease,
and peripheral vascular disease were all frequently
associated with ED in this cohort.
42
The results of the
Massachusetts Male Aging Study also showed that the
age-adjusted risk for ED was signicantly increased by
the presence of heart disease or hypertension.
42
These
ndings are supported by the results of several other
smaller-scale epidemiologic studies. Parazzini et al.
43
showed that a history of cardiopathy, hypertension,
peripheral vascular disorder, and thrombotic or hemor-
rhagic stroke all increased the risk of ED in a cohort of
2010 men. Martin-Morales et al.
44
also showed that
high blood pressure, peripheral vascular disease, and
cardiac disorders were associated with signicantly
increased risk for ED in a sample of 2476 men between
25 and 70 years of age.
The link between cardiovascular disease and ED
is strongly supported by results from a study by
Greenstein et al., who assessed the relationship
between ED severity and ischemic heart disease in 40
men who underwent coronary angiography.
45
They
found a signicant correlation between the severity of
ED and the number of obstructed coronary vessels.
Patients with one-vessel disease had more and rmer
erections with fewer difculties in achieving erection
than did men with two- or three-vessel disease. They
also noted a signicant association between hyper-
tension and ED.
Kawanishi and colleagues provided evidence that the
presence of ED is predictive of undiagnosed ischemic
heart disease.
46
They performed echocardiograms,
treadmill tests, thallium exercise scintigrams, and
coronary angiograms in 58 patients with ED. Fourteen
patients (24.1%) were diagnosed with ischemic heart
disease. Although six patients had already been diag-
nosed at the time of testing, eight were newly diagnosed
by the exercise tests.
46
While the relationship between vascular disease and
ED would seem straightforward, the reason for the
strong independent relationship between hypertension
and ED is less clear. For example, Jaffe and associates
reported that penile peak systolic velocity, resistance
index, and penile brachial index did not differ signi-
cantly between hypertensive and normotensive men
with ED.
47
However, NO production is reduced in
patients with essential hypertension,
48
and it is reason-
able to speculate that this decrease may contribute to
the increased risk for ED in patients with high blood
pressure.As noted above, NO production by the vascular
endothelium also decreases with age independent of
disease,
38
and it is possible that a hypertension-related
decrease in NO may exacerbate the normal age-
associated fall and potentially increase the risk for ED
in older men with elevated blood pressure.
Diabetes
The risk of diabetes increases with age,
49
and the
presence of diabetes signicantly raises the risk for
ED.
4144
It has also been shown that ED severity is cor-
related with the degree of impaired glycemic control.
50
Diabetes is associated with both neural and micro-
vascular disease, which may contribute to the develop-
ment of ED.
50
Diabetes also results in increased
formation of AGEs in the human tunica albuginea and
corpus cavernosum.
51
As noted above, AGE-associated
changes in DNA and proteins can result in scavenging
of NO and thus decreased erectile function. Unlike
aging, however, diabetes is associated with a marked
reduction in eNOS in the cavernosal endothelium and
smooth muscle but upregulation of iNOS.
51
As with
cardiovascular disease, molecular events that occur in
patients with diabetes may interact additively or even
synergistically with those that result from normal
aging. AGEs accumulate in the aging penis
33
as well as
in many other tissues.
52
Thus, in elderly men, diabetes
may have the potential substantially to increase
normally occurring AGE formation and perhaps
elevate the risk for ED.
Benign prostatic hyperplasia
The prevalence of benign prostatic hyperplasia (BPH)
increases with age,
53
and the presence of BPH increases
the risk for ED.
54
Namasivayam et al. noted that over
one-half of a cohort of 140 men with BPH also had
some degree of ED.
54
However, it is not clear whether
the presence of BPH increases ED risk independent
of age, or whether the increased risk of ED in men
with BPH is due to the fact that the prevalence of
both conditions is increased in the elderly. Current
248 Erectile Dysfunction in the Elderly: Epidemiology, Etiology, and Approaches to Treatment
evidence appears to support the latter of these two
possibilities.
55
Depression
The association between age and the risk for depression
is controversial, but there appears to be a complex
relationship between depression and advancing age,
with a peak prevalence in the years before retirement,
a low prevalence during the rst 1015 years there-
after, and another increase in those older than 75 years
of age.
56
There is a signicant age-independent cor-
relation between the presence of depression and ED.
57
Several investigators have suggested that the age-
related decrease in the production of testosterone
and perhaps other hormones (e.g., growth hormone,
thyroxine, dehydroepiandrosterone, melatonin) may
contribute to the development of both conditions.
58
THERAPIES FOR ED IN THE ELDERLY
There is a wide range of treatments for ED, and
virtually all of them can be used in older patients.These
therapies can be divided into two broad categories:
non-oral and oral.
Non-oral treatments and early oral
therapies
There are a variety of non-oral approaches to the treat-
ment of ED, including vacuum constriction devices,
penile self-injection therapy, hormone injections, and
penile prostheses.
59
While all of these approaches can
result in signicant improvements in male sexual func-
tion, none are completely satisfactory. For example,
self-injection treatments may be associated with side-
effects such as prolonged erection, priapism, and brosis
of the corpus cavernosum.
59
While intercavernosal
injection therapy consistently produces erections in up
to 87% of patients, there is a dropout rate of about
50%.
59
Finelli et al.
60
evaluated treatment preferences
and commitment to specic therapeutic choices in a
cohort of 88 elderly men (mean age, 69.5 years) with
ED. The treatment options available to these patients
included older oral medications (e.g., yohimbine,
trazodone), vacuum devices, injection therapy, penile
prostheses, counseling, and testosterone replacement
(when indicated). Patients were followed from 1 to
63 months after initial consultation (median follow-up,
9 months).The most popular initial treatment choice in
this group of older men with ED was injection therapy
(30%), followed by vacuum devices (27%) and oral
medication (20%; yohimbine in 16 patients and
trazodone and sildenal each in one patient). Of the
84 patients treated, 40% switched therapies over the
course of follow-up. The dropout rate was signicantly
higher for patients who began with oral therapy (78%)
than for those who selected intracavernosal injection
(48%) or a vacuum device (29%). Another long-term
(3-year) study of ED treatment in 69 men (mean age
60.5 years) indicated that injection therapy with prosta-
glandin E
1
was effective and generally well-tolerated,
and that it resulted in the development of spontaneous
erections in 35% of these patients.
61
These results
appear to support the view that injection therapy
was the best ED treatment for older men, at least until
the time when new, highly effective oral treatments
became available.
Newer oral therapies
A wide range of oral treatments have been used
in men with ED, including sildenal, apomorphine,
oral phentolamine, trazodone, and yohimbine.
62
New
drugs in late-phase development include several new
PDE5 inhibitors and agents with other mechanisms of
action.
63,64
As noted above, older oral drugs, such as
yohimbine and trazodone, appear to be relatively
ineffective in older men.
60
The PDE5 inhibitor sildenal (Figs 18.918.11) is by
far the best studied oral therapy for the treatment of
men with ED, and it has been shown to be effective in
elderly men, including those with comorbidities likely
to be present in this age group. Rendell et al.
65
recently
reported that sildenal signicantly improved the
ability of men 65 years of age or older to achieve and
maintain erections (Table 18.2). While the response
rate to sildenal (based on the response to a general
efcacy question) was similar in men 65 years of age
and those 5064 years old (50% versus 53%), these
response rates were lower than those for men 1849
years of age (72%).
Wagner et al.
66
analyzed data from ve double-
blind, 12-week to 6-month placebo-controlled studies
to evaluate the efcacy and tolerability of sildenal in
411 patients 65 years of age or older having ED with
a broad spectrum of etiologies. Efcacy was assessed
using a global efcacy question, questions three and
four of the International Index of Erectile Function
(IIEF),
67
and the ve sexual function domains of the
IIEF. All efcacy assessments indicated that sildenal
signicantly improved erectile function both in elderly
patients with ED of broad-spectrum etiology and in
elderly patients with ED and diabetes. The most
common adverse events in these men were mild-to-
moderate headache, flushing, and dyspepsia. The rates
of discontinuation due to adverse events were low and
comparable to the rates with placebo.
Sildenal has also been shown to be effective in men
with ED and hypertension,
68
and in men with both ED
and ischemic heart disease.
69
Conti et al.
69
reported
that 70% of patients who received sildenal had
Therapies for ED in the Elderly 249
250 Erectile Dysfunction in the Elderly: Epidemiology, Etiology, and Approaches to Treatment
Figure 18.9 Oral type 5 phosphodiesterase inhibitors. All drugs have a similar chemical composition to cyclic guanosine
monophosphate (cGMP). That is their mechanism of action. They are all competitive inhibitors of cGMP.
Sildenafil
cGMP
Tadalafil
N
S
O
HN
O
O
O
OH
P
O
O
H
H
N
NH
O
O
O
N
N
N
O
OH
HN
O
O
N
N
N
O
N
H
3
C
H
2
C
CH
3
CH
3
CH
3
CH
3
Vardenafil
N
S
O
HN
O
O
N
N O
N
H
3
C
CH
3
CH
3
CH
3
Figure 18.10 Mechanism of
phosphodiesterase type 5 (PDE5)
inhibition. The oral PDE5
inhibitors competitively inhibit the
PDE5 enzyme to prolong the
effects of cyclic guanosine
monophospate (cGMP) and yield
a longer-lasting erection. GTP,
guanosine triphosphate; nNOS,
neuronal nitric oxide synthase;
eNOS, endothelial nitric oxide
synthase; cAMP, cyclic adenosine
monophosphate. (Reprinted with
permission from Lue TF. Erectile
dysfunction. N Engl J Med 2000;
2:1802.)
Guanylyl
cyclase
cGMP-specific
protein kinase
cAMP-specific
protein specific
Adenylyl
cylase
GTP
cGMP
5' GMP
Ca
2+
Ca
2+
PDE5
eNOS
nNOS
eNOS
improved erections, versus 20% in the placebo group,
in patients with both ED and ischemic heart disease
who were not taking nitrates.
Sildenal is also effective in patients with depression.
In a retrospective analysis that included 134 patients
with a medical history, past or present, of depression,
76% of the patients who received sildenal reported
improved erections versus 18% for placebo.
70
Although sildenal has been shown to be efcacious
in a wide range of individuals with ED, it is not effec-
tive in all patients. Martnez-Jabaloyas et al.
71
recently
evaluated risk factors for treatment failure with
sildenal and noted that diabetes, non-nerve-sparing
radical prostatectomy, and high baseline disease
severity, as reflected by Sexual Health Inventory for
Men (SHIM) scores, were all predictors of a lower
success rate for sildenal therapy.
The new agent tadalal (IC351), a reversible inhibitor
of PDE5, is more selective for PDE5 versus PDE6 than
is sildenal.
64
An assessment of the safety and effective-
ness of tadalal in patients older than 65 years of age
versus younger patients indicated that it was safe and
effective in both groups, with improved erections in up
to 81% of men in the combined population.
72
Vardenal, another novel selective PDE5 inhibitor,
has also been shown to be safe and effective for the
treatment of ED in a 12-week, multicenter, randomized,
double-blind, placebo-controlled trial that included
601 men with mild-to-severe ED.
73
While the study was
not specically directed toward evaluating the efcacy
of vardenal in older individuals, results showed that
the 20-mg dose improved erections in 80% of patients.
Oral therapies with mechanisms of action that differ
from those of sildenal and the other PDE5 inhibitors
are in development. These include an oral agent that
opens large-conductance calcium-activated potassium
channels (maxi-K), and agents that block the actions of
the catecholaminergic input to the penis (phentolamine
mesylate and sibutramine).
64
The importance of AGEs
in cardiovascular disease and ED has prompted
development and testing of ALT-711, an orally bio-
available non-enzymatic breaker of AGE cross-links.
Treatment with this agent signicantly increased
arterial compliance and reduced pulse pressure in
patients with systolic hypertension.
74
It seems reason-
able to suggest that this treatment might also be
effective in men with ED, especially in those patients
with diabetes who have been shown to have increased
concentrations of AGEs in penile tissues.
51
Gene therapy
Increased understanding of the molecular mechanisms
involved in penile erectile function and how they may
be impaired in men with ED has prompted investi-
gations into gene therapy. The importance of NO in
erectile function has prompted transfer of the gene for
eNOS in aged rats.
75
Champion and associates showed
that adenoviral-mediated gene transfer of eNOS could
reverse at least some symptoms of age-related ED in
60-week-old rats. Five days after gene transfer, eNOS
protein, mRNA, and cGMP levels in the corpora caver-
nosa were signicantly increased, and the erectile
Figure 18.11 Successful endothelial nitric oxide synthase
(eNOS) transfection in the animal produces an improvement
in intracavernosal pressure upon electrical stimulation.
C
h
a
n
g
e
i
n
c
a
v
e
r
n
o
s
a
l
p
r
e
s
s
s
u
r
e
(
m
m
H
g
)
0
Control eNOS
transfected
P <0.05
80
20
40
60
TABLE 18.2 PDE5 inhibitors: selectivity for
PDE5 vs other PDE isoenzymes.
PDE Sildenal Tadalal Vardenal
Isoenzyme
PDE1 80 >4500 500
PDE2 >8750 >14 800 44 290
PDE3 4630 >14 800 >7140
PDE4 2190 >14 800 43 570
PDE6 (rod) 11 187 25
(cone) 10 193 4
PDE7 6100 >14 800 >214 000
PDE8 8500 >14 800 >214 000
PDE9 750 >14 800 4150
PDE10 2800 >14 800 21 200
PDE11 780 5 1160
Therapies for ED in the Elderly 251
response to cavernosal nerve stimulation was signi-
cantly increased. These results raise the possibility that
transfer of the gene for eNOS may provide a new
therapeutic intervention for ED.
76
This same group evaluated the effects of gene transfer
for prepro-CGRP as therapy for ED in another group
of aged rats.
76
As noted above, levels of this peptide are
decreased in the penis of old rats.
35
Five days after
transfection, the aged rats had an approximately three-
fold increase in cavernosal CGRP levels. In addition,
cAMP levels in the corpora cavernosa were signicantly
increased. More importantly, there was a signicant
increase in the erectile response to cavernosal nerve
stimulation in the transfected animals.
It was noted above that aging-associated ion-channel
abnormalities may play a role in the development of
ED. Specically, Melman and Christ suggested that
alterations in the expression, regulation, and/or func-
tion of potassium (K
+
) channels and gap junctions may
be involved in the etiology of diabetes- and/or age-
related ED.
32
They tested the potential importance of a
specic K
+
channel, maxi-K, in diabetes-associated ED
in the following way. First, they rendered rats diabetic
with a single subcutaneous injection of streptozotocin.
Two months after the streptozotocin injection and
verication of a profound penile neuropathy, animals
received a single intracavernous injection of the DNA
encoding the K
+
channel maxi-K or the pcDNA vector
only. After an additional 23 months, the rats were
tested with an intracavernous nerve stimulation
protocol. Despite the presence of signicant neuro-
pathy, the nerve stimulation responses were dramati-
cally greater in the maxi-K-transfected animals than in
controls that received only the pcDNA. This result is
consistent with the conclusion that increasing maxi-K
channel expression in cavernosal tissues may reverse
diabetes-associated pathologic changes in penile
function.
All of the results summarized in this section are
consistent with the possibility that gene therapy may
provide effective treatment for patients with ED. Given
that ED may be a heterogeneous disease, it is likely
that transfer of different genes may be required in
different patients.
CONCLUSIONS
ED is a common condition with a prevalence that
increases dramatically with age. It seems likely that
both the normal aging process and the age-related
accumulation of risk factors for ED contribute to this
increased prevalence in older men. At present,
inhibition of PDE5 with oral agents such as sildenal
appears to be the treatment of choice. These drugs have
been shown to be safe and effective in elderly men
with ED, and sildenal has demonstrated efcacy in
patients who have many of the comorbidities observed
in older men with ED. New treatments, in particular
transfection with genes for key mediators of erectile
function that are known to be downregulated in elderly
men, also hold signicant promise. The results reviewed
here underscore the remarkable progress in the treat-
ment of ED that has taken place in the past decade. It
is likely that further research into the neural and
vascular mechanisms involved in penile erection will
continue to result in new safe, effective, and convenient
therapies for men with this condition.
REFERENCES
1. McKinlay JB.The worldwide prevalence and epidemiology
of erectile dysfunction. Int J Impot Res 2000; 12 (suppl. 4):S6.
2. Feldman HA, Goldstein I, Hatzichristou DG et al.
Impotence and its medical and psychosocial correlates: results
of the Massachusetts Male Aging Study. J Urol 1994; 1:54.
3. Masumori N, Tsukamoto T, Kumamoto Y et al. Decline of
sexual function with age in Japanese men compared with
American men results of two community-based studies.
Urology 1999; 54:335.
4. Marumo K, Nakashima J, Murai M.Age-related prevalence
of erectile dysfunction in Japan: assessment by the Inter-
national Index of Erectile Function. Int J Urol 2001; 8:53.
5. Blanker MH, Ruud Bosch JLH, Groeneveld FPMJ et al.
Erectile and ejaculatory dysfunction in a community-based
sample of men 50 to 78 years old: prevalence, concern, and
relation to sexual activity. Urology 2001; 57:763.
6. Braun M,Wassmer G, Klotz T et al. Epidemiology of erectile
dysfunction: results of the Cologne Male Survey. Int J Impot
Res 2000; 12:305.
7. Mulligan T, Retchin SM, Chinchilli VM et al. The role of
aging and chronic disease in sexual dysfunction. J Am Geriatr
Soc 1988; 36:520.
8. Lue TF. Erectile dysfunction. N Engl J Med 2000; 2:1802.
9. Maggi M, Filippi S, Ledda F et al. Erectile dysfunction: from
biochemical pharmacology to advances in medical therapy.
Eur J Endocrinol 2000; 143:143.
10. ckert S, Kthe A, Stief CG et al. Phosphodiesterase
isoenzymes as pharmacological targets in the treatment of
male erectile dysfunction. World J Urol 2001; 19:14.
11. Lin CS, Lau A, Tu R et al. Expression of three isoforms of
cGMP-binding cGMP-specic phosphodiesterase (PDE5) in
human penile cavernosum. Biochem Biophys Res Commun
2000; 268:628.
12. Garcia-Reboll L, Mulhall JP, Goldstein I. Drugs for the
treatment of impotence. Drugs Aging 1997; 11:140.
13. Margolese HC. The male menopause and mood: testos-
terone decline and depression in the aging male Is there a
link? Geriatr Psychiatry Neurol 2000; 13:93.
14. Kaiser FE, Viosca SP, Morley JE et al. Impotence and
aging: clinical and hormonal factors. J Am Geriatr Soc 1988;
36:511.
252 Erectile Dysfunction in the Elderly: Epidemiology, Etiology, and Approaches to Treatment
15. Schiavi RC, Schreiner-Engel P,White D et al. The relation-
ship between pituitarygonadal function and sexual behavior
in healthy aging men. Psychsom Med 1991; 53:363.
16. Nehra A. Treatment of endocrinologic male sexual dys-
function. Mayo Clin Proc 2000; 76 (suppl.):S40.
17. Davidson JM, Kwan M, Greenleaf WJ. Hormonal
replacement and sexuality in men. Clin Endocrinol Metab
1982; 11:599.
18. Akkus E, Carrier S, Baba K et al. Structural alterations in
the tunica albuginea of the penis: impact of Peyronies disease,
aging and impotence. Br J Urol 1997; 79:47.
19. Moreland RB. Pathophysiology of erectile dysfunction:
the contributions of trabecular structure to function and the
role of functional antagonism. Int J Impot Res 2000; 12
(suppl. 4):S39.
20. Vanegas JP, Raviv G, Kiss R et al. Intracavernous collagen
analysis in impotence. Acta Urol Belg 1996; 64:7.
21. Lin JS, Lin YM, Chow NH et al. Novel image analysis of
corpus cavernous tissue in impotent men. Urology 2000;
55:252.
22. Rowland DL, Greenleaf W, Mas M et al. Penile and nger
sensory thresholds in young, aging, and diabetic males. Arch
Sex Behav 1989; 18:1.
23. Carrier S, Nagaraju P, Morgan DM et al. Age decreases
nitric oxide synthase-containing nerve bers in the rat penis.
J Urol 1997; 157:1088.
24. Warburton AL, Santer RM. Sympathetic and sensory
innervation of the urinary tract in young adult and aged rats:
a semi-quantitative histochemical and immunohistochemical
study. Histochem J 1994; 26:127.
25. Amenta F, Cavallotti C, De Rossi M et al. Vasoactive
intestinal polypeptide levels and distribution in the penis of
old rats. J Neural Transm 1987; 70:137.
26. Garbn H, Vernet D, Freedman A et al. Effect of aging on
nitric oxide-mediated penile erection in rats. Am J Physiol
1995; 268:H467.
27. Andrew PJ, Mayer B. Enzymatic function of nitric oxide
synthases. Cardiovasc Res 1999; 43:521.
28. Hickey MJ. Role of inducible nitric oxide synthase in the
regulation of leucocyte recruitment. Clin Sci 2001; 100:1.
29. Rajasekaran M, Mondal D, Agrawal K et al. Ex vivo
expression of nitric oxide synthase isoforms (eNOS/iNOS)
and calmodulin in human penile cavernosal cells. J Urol
1998; 160:2210.
30. Ferrini M, Magee TR, Vernet D et al. Aging-related
expression of inducible nitric oxide synthase and markers of
tissue damage in the rat penis. Biol Reprod 2001; 64:974.
31. Haas CA, Seftel AD, Razmjouei K et al. Erectile dys-
function in aging: upregulation of endothelial nitric oxide
synthase. Urology 1998; 51:516.
32. Melman A, Christ GJ. Integrative erectile biology. The
effects of age and disease on gap junctions and ion channels
and their potential value to the treatment of erectile
dysfunction. Urol Clin North Am 2001; 28:217.
33. Jiann DB, Seftel AD, Fogarty J et al. Age-related increase
in an advanced glycation end product in penile tissue.
Potential role in erectile dysfunction. World J Urol 1995;
13:369.
34. Vaziri ND, Wang XQ. cGMP-mediated negative-feedback
regulation of endothelial nitric oxide synthase expression by
nitric oxide. Hypertension 1999; 34:1237.
35. Wimalawansa SJ. Age-related changes in tissue contents
of immunoreactive calcitonin gene-related peptide. Aging
Clin Exp Res 1992; 4:211.
36. Chung WK, Park YY, Kwon SW. The impact of aging on
penile hemodynamics in normal responders to pharmaco-
logical injection: a Doppler sonographic study. J Urol 1997;
157:2129.
37. Wandell PE, Brorsson B. Assessing sexual functioning in
patients with chronic disorders by using a generic health-
related quality of life questionnaire. Qual Life Res 2000;
9:1081.
38. Toprak M, zmen D, Mutaf I et al. Age-associated
changes in nitric oxide metabolites nitrite and nitrate. Int J
Clin Lab Res 2000; 30:83.
39. Paniagua OA, Bryant MB, Panza JA. Role of endothelial
nitric oxide in shear stress-induced vasodilation of human
microvasculature: diminished activity in hypertensive and
hypercholesterolemic patients. Circulation 2001; 103:1752.
40. Weber MA. Hypertension in the aging patient: new
imperatives, new options. Am J Geriatr Cardiol 2000; 9:12.
41. Chew KK, Earle CM, Stuckey BGA et al. Erectile dys-
function in general medicine practice: prevalence and clinical
correlates. Int J Impot Res 2000; 12:41.
42. Johannes CB, Araujo AB, Feldman HA et al. Incidence of
erectile dysfunction in men 40 to 69 years old: longitudinal
results from the Massachusetts Male Aging Study. J Urol
2000; 163:460.
43. Parazzini F, Menchini-Fabris F, Bortolotti A et al.
Frequency and determinants of erectile dysfunction in Italy.
Eur Urol 2000; 37:43.
44. Martin-Morales A, Sanchez-Cruz JJ, Saenz de Tejada I
et al. Prevalence and independent risk factors for erectile dys-
function in Spain: results of the Epidemiologia de la Disfuncion
Erectil Masculina study. J Urol 2001; 166:569.
45. Greenstein A, Chen J, Miller H et al. Does severity of
ischemic coronary disease correlate with erectile function?
Int J Impot Res 1997; 9:123.
46. Kawanishi Y, Lee KS, Kimura K et al. Screening of
ischemic heart disease with cavernous artery blood flow in
erectile dysfunctional patients. Int J Impot Res 2001; 13:100.
47. Jaffe A, Chen Y, Kisch ES et al. Erectile dysfunction in
hypertensive subjects: assessment of potential determinants.
Hypertension 1996; 28:859.
48. Klahr S. The role of nitric oxide in hypertension and renal
disease. Nephrol Dial Transplant 2001; 16 (suppl. 1):60.
49. Heine RJ. Current therapeutic options in type 2 diabetes.
Eur J Clin Invest 1999; 29 (suppl. 2):17.
References 253
50. Romeo JH, Seftel AD, Madhun ZT et al. Sexual function
in men with diabetes type 2: association with glycemic control.
J Urol 2000; 163:788.
51. Seftel AD, Vaziri ND, Ni Z et al. Advanced glycation end
products in human penis: elevation in diabetic tissue, site
of deposition, and possible effect through iNOS or eNOS.
Urology 1997; 50:1016.
52. Baynes JW. The role of AGEs is aging: causation or
correlation. Exp Gerontol 2001; 36:1526.
53. Boyle P, McGinn R, Maisonneuve P et al. Epidemiology
of benign prostatic hyperplasia: present knowledge and
studies needed. Eur Urol 1991; 20 (suppl. 2):3.
54. Namasivayam S, Minhas S, Brooke J et al. The evaluation
of sexual function in men presenting with symptomatic benign
prostatic hyperplasia. Br J Urol 1998; 82:842.
55. Green JS, Holden ST, Bose P et al. An investigation into
the relationship between prostate size, peak urinary flow rate
and male erectile dysfunction. Int J Impot Res 2001; 13:322.
56. Palsson S, Skoog I. The epidemiology of affective dis-
orders in the elderly: a review. Int Clin Psychopharmacol
1997; 12 (suppl. 7): S3.
57. Araujo AB, Durante R, Feldman HA et al. The relation-
ship between depressive symptoms and male erectile dys-
function: cross-sectional results from the Massachusetts Male
Aging Study. Psychosom Med 1998; 60:458.
58. Wespes E, Schulman CC. Male andropause: myth, reality,
and treatment. Int J Impot Res 2002; 14 (14 suppl. 1): S93.
59. Padma-Nathan H. Minimally invasive therapy for erectile
dysfunction: intracavernosal, oral, transdermal/transglanular,
and intraurethral approaches. In: Mulcahy JJ, ed. Topics in
clinical urology: diagnosis and management of male sexual
dysfunction. Tokyo: Igaku-Shoin; 1999:182185.
60. Finelli A, Hirshberg D, Radomski SB.The treatment choice
of elderly patients with erectile dysfunction. Geriatr Nephrol
Urol 1998; 8:15.
61. Kunelius P, Lukkarinen O. Intracavernous self-injection
of prostaglandin E1 in the treatment of erectile dysfunction.
Int J Impot Res 1999; 11:21.
62. Padma-Nathan H, Giuliano F. Oral drug therapy for erec-
tile dysfunction. Urol Clin North Am 2001; 28:321.
63. Padma-Nathan H, McMurray JG, Pullman WE et al.
On-demand IC351 (Cialis) enhances erectile function in
patients with erectile dysfunction. Int J Impot Res 2001; 13:2.
64. Pryor JL, Redmon B. New therapies and delivery
mechanisms for treatment of erectile dysfunction. Int J Impot
Res 2000; 12 (suppl. 4):S158.
65. Rendell MS, Rajfer J, Wicker P et al. Sildenal for
treatment of erectile dysfunction in men with diabetes: a
randomized controlled trial. JAMA 1999; 281:421.
66. Wagner G, Montorsi F, Auerbach S et al. Sildenal citrate
(Viagra