University of Wollongong
Research Online
Graduate School of Medicine - Papers
Faculty of Medicine
2010
Role of fesoterodine in the treatment of overactive
bladder
Kylie J. Mansfield
University of Wollongong,
[email protected]
Publication Details
Mansfield, K. J. (2010). Role of fesoterodine in the treatment of overactive bladder. Open Access Journal of Urology, 2 1-9.
Research Online is the open access institutional repository for the
University of Wollongong. For further information contact Manager
Repository Services:
[email protected].
Role of fesoterodine in the treatment of overactive bladder
Abstract
Abstract: Muscarinic receptors have long been the target receptors for treatment of patients with overactive
bladder (OAB). These patients experience symptoms of urgency, urinary frequency and nocturia, with or
without urge incontinence (the involuntary leakage of urine associated with urge). Fesoterodine, a pro-drug,
structurally and functionally related to tolterodine, is the newest agent developed for the treatment of OAB.
Fesoterodine is broken down to the active metabolite, 5-hydroxy-methyl-tolterodine (5-HMT) by nonspecific esterases. This metabolism results in the complete breakdown of the parent compound and is
responsible for dose related improvements in clinical efficacy and health related quality of life. Like other
antimuscarinic agents including tolterodine, fesoterodine is associated with improvements in clinical variables
related both to bladder filling (decreasing micturition frequency and increasing mean voided volume) and
urgency (urgency and urge incontinence episodes). Improvements in health related quality of life following
treatment with fesoterodine is indicated by improvements in 7 of the 9 variables measured by the King’s
Health Questionnaire. Also like other antimuscarinic agents, fesoterodine use is associated with adverse
events including dry mouth. However the incidence of dry mouth is reduced with fesoterodine, compared to
oxybutynin, due to the improved bladder selectivity of 5-HMT.
Keywords
fesoterodine, role, bladder, treatment, overactive
Publication Details
Mansfield, K. J. (2010). Role of fesoterodine in the treatment of overactive bladder. Open Access Journal of
Urology, 2 1-9.
This journal article is available at Research Online: http://ro.uow.edu.au/medpapers/25
Open Access Journal of Urology
Dovepress
open access to scientific and medical research
Open Access Full Text Article
review
Role of fesoterodine in the treatment
of overactive bladder
Kylie J Mansfield
Graduate School of Medicine,
University of Wollongong, NSW,
Australia
Abstract: Muscarinic receptors have long been the target receptors for treatment of patients with
overactive bladder (OAB). These patients experience symptoms of urgency, urinary frequency
and nocturia, with or without urge incontinence (the involuntary leakage of urine associated
with urge). Fesoterodine, a pro-drug, structurally and functionally related to tolterodine, is the
newest agent developed for the treatment of OAB. Fesoterodine is broken down to the active
metabolite, 5-hydroxy-methyl-tolterodine (5-HMT) by non-specific esterases. This metabolism
results in the complete breakdown of the parent compound and is responsible for dose related
improvements in clinical efficacy and health related quality of life. Like other antimuscarinic
agents including tolterodine, fesoterodine is associated with improvements in clinical variables
related both to bladder filling (decreasing micturition frequency and increasing mean voided
volume) and urgency (urgency and urge incontinence episodes). Improvements in health related
quality of life following treatment with fesoterodine is indicated by improvements in 7 of the
9 variables measured by the King’s Health Questionnaire. Also like other antimuscarinic agents,
fesoterodine use is associated with adverse events including dry mouth. However the incidence
of dry mouth is reduced with fesoterodine, compared to oxybutynin, due to the improved bladder selectivity of 5-HMT.
Keywords: fesoterodine, 5-hydroxymethyl-tolterodine, muscarinic, overactive bladder, urgency,
incontinence
Overview of the overactive bladder
Correspondence: Kylie Mansfield
Graduate School of Medicine,
University of Wollongong,
Northfields Ave, Wollongong,
2522, NSW, Australia
Email
[email protected]
submit your manuscript | www.dovepress.com
Dovepress
Overactive bladder (OAB) is a debilitating chronic disorder experienced by
approximately 17% of both men and women over the age of 40 years, with the
prevalence increasing with increasing age.1,2 Patients with OAB typically experience
symptoms of urgency, usually with frequency and nocturia, with or without urge
incontinence (the involuntary leakage of urine associated with urge).3 Results from
the National Overactive Bladder Evaluation (NOBEL) Programme conducted in the
US indicated that OAB was more prevalent than other chronic conditions such as heart
disease, sinusitis, and asthma.2
OAB is a chronic disease with a major negative influence on quality of life,
especially associated with the limitations it places on physical and emotional roles,
vitality and social functioning.4 The symptoms of OAB affect all aspects of life including: social (limiting outings due to frequent need to urinate), psychological (loss of
self esteem associated with incontinence), physical (limitations of physical activities
due to fear of incontinence) and occupational (decreased productivity).3 OAB can
also be associated with economic costs including; the personal costs of managing
Open Access Journal of Urology 2010:2 1–9
© 2010 Mansfield, publisher and licensee Dove Medical Press Ltd. This is an Open Access article
which permits unrestricted noncommercial use, provided the original work is properly cited.
Mansfield
Dovepress
incontinence, the treatment costs associated with managing
symptoms and providing care for incontinent nursing home
residents and costs associated with decreased work productivity. In the US, the economic costs associated with OAB
in the year 2000, were estimated to be between US$12.02
billion and US$17.5 billion.5 This makes the economic impact
of OAB, comparable to the economic impact of influenza,
arthritis and osteoarthritis.5
The cornerstone symptom of OAB is urgency1,6 which is
defined as the complaint of a sudden compelling desire to pass
urine which is difficult to defer.3 This symptom is associated
with frequency of urination and nocturia. Approximately 66%
of patients with OAB do not have urge incontinence and are
classified as OAB dry.2 The remaining 33% of patients with
OAB have urgency associated with incontinence and are
classified as OAB wet.2 Urodynamic testing demonstrates
that patients with OAB wet have detrusor overactivity, where
urine leakage arises from involuntary detrusor contraction.3
The etiology of these involuntary detrusor contractions
remains uncertain.
There are numerous treatment options available for
patients with OAB including biofeedback, electrical stimulation, bladder training and pharmacotherapy, either alone
or in combination. However, the primary treatment for the
OAB is pharmacotherapy with muscarinic receptor antagonists7–9 which have been used for many years. Oxybutynin
(Ditropan®) was the first muscarinic receptor antagonist to
be introduced to OAB therapy. Newer antimuscarinic agents
include the M3 selective antagonists, darifenacin (Enablex®)
and solifenacin (VESIcare) and the relatively non-subtype
selective antagonists, tolterodine (Detrol®) and its related
compound fesoterodine (Toviaz®), which has only been
recently introduced.
The role of muscarinic receptors
in bladder physiology
The traditional dogma behind the treatment of OAB with
muscarinic receptor antagonists was based on our understanding of the nerves controlling the physiological functions of the
bladder. During filling the bladder detrusor muscle expands
at low pressure. During this time the stretch of the bladder
wall initiates the release of mediators (such as ATP) from the
urothelium that signals bladder fullness via the underlying
afferent nerves.10 Signals from these afferent nerves are processed in the Pontine micturition centre in the brain and, at an
appropriate time, efferent parasympathetic nerves are activated.
The efferent parasympathetic nerves release acetylcholine
onto muscarinic receptors located on the detrusor muscle.11
submit your manuscript | www.dovepress.com
Dovepress
There are 5 individual subtypes of muscarinic receptors
(M1–M5) that have been cloned and pharmacologically
characterized.12 In the urinary bladder, as in other smooth
muscles, multiple muscarinic receptor subtypes have been
identified.13 Binding and immunoprecipitation studies,14–17
have demonstrated that the majority of muscarinic receptors present in human detrusor muscle, are M 2 receptor
(∼70%) with smaller populations of M 3 (20%) and M1
(10%) receptors.17 Activation of muscarinic receptors by
acetylcholine leads to contraction of the detrusor muscle and
subsequent emptying of the bladder. Functional studies carried out in M3 knockout mice18 and human detrusor strips19,20
have demonstrated that muscarinic M3 receptors are the
receptor subtype responsible for contraction of the detrusor
muscle. Nevertheless, there is some evidence that M2 receptors also have some functional importance.21,22 Traditionally
the muscarinic antagonists used to treat OAB were thought
to inhibit activation of the muscarinic receptors responsible
for detrusor contractions. Since both muscarinic M2 and M3
receptor subtypes are associated with detrusor contraction,
muscarinic receptor antagonists, have been characterized
according to their affinity for these receptor subtypes.
Antimuscarinic therapy for OAB
OAB therapy began when oxybutynin was shown to reduce
contractions of the rabbit detrusor in response to the muscarinic agonist carbachol23 although oxybutynin is not selective
for any individual muscarinic receptor subtype (Table 1).
Oxybutynin was then shown to be clinically effective at
preventing detrusor spasms following transurethral surgery24
which provided the impetus for antimuscarinic therapy for
OAB. This was soon followed in the early 1980s with reports
of oxybutynin providing symptomatic relief for patients with
detrusor instability.25,26
In 1998 tolterodine, another muscarinic receptor
antagonist was first introduced. Like oxybutynin, toltero-
Table Range of Ki values (in nM) reported for antimuscarinic agents
in cell lines expressing human muscarinic receptor subtypes
Compound
M
M
M3
M4
M5
Oxybutynin
a
4.5
36.5
3.3
5.2
19.6
Tolterodineb
6.9
6.7
6.4
6.8
5.9
Fesoterodinec
11.9
5.1
26.9
8.9
4.5
5.9
5.6
5.7
5.8
6.1
5-HMT
d
Data summarized from
Data summarized from27,87–92
c
Data summarized from36,92
d
Data summarized from27,36
a
36,85–89
b
Open Access Journal of Urology 2010:2
Fesoterodine for overactive bladder
Dovepress
dine, is relatively non-selective for individual muscarinic
receptor subtypes (Table 1, Ki at M3 and M2 receptors of 6.4
nM and 6.7 nM respectively).27 And also like oxybutynin,
tolterodine is efficacious in the treatment of OAB.28–32
However, the clinical efficacy of tolterodine has been
demonstrated to be associated both with tolterodine itself,
and with the generation of an active metabolite, 5-hydroxymethyl-tolterodine (5-HMT).33–35 Like tolterodine, 5-HMT
demonstrates similar affinity for the individual muscarinic
receptor subtypes (Table 1 Ki at M3 and M2 receptors of
5.7 nM and 5.6 nM respectively).27,36 The newest muscarinic antagonist for therapy of OAB is fesoterodine.
Fesoterodine, which is structurally related to tolterodine,
also results in the formation of the same active metabolite,
5-HMT37,38 although the mechanism underlying production
of 5-HMT are vastly different.
Pharmacokinetics of fesoterodine
and tolterodine
The active metabolite of tolterodine, 5-HMT, is formed by
cytochrome P450 2D6 (CYP2D6)34 (Figure 1) which is subject to polymorphism.39 The polymorphism of CYP2D6 is
highly clinical relevant as it is responsible for variability in
metabolism of more than 100 different drugs.40 Based on their
CYP2D6 phenotype patients are characterized as extensive
metabolizers, if they have two functional CYP2D6 alleles, or
poor metbaolizers who lack functional CYP2D6 alleles.40 Up
to 10% of white populations and 19% of black populations are
characterized as poor metabolizers.40 In patients classified as
extensive metabolizers, 81% of the absorbed tolterodine, is
extracted during first pass metabolism through the liver,39 and
hydrolyzed to 5-HMT with an average maximal plasma concentration of tolterodine and 5-HMT being similar (5.2 and 4.8
ng/mL respectively).39 In contrast, in poor metabolizers only
18% of tolterodine is extracted during first pass metabolism
through the liver39 and the average maximal plasma concentration of tolterodine of is increased to 38 ng/mL while 5-HMT
is not detectable.39 Furthermore the concentration of 5-HMT
released from metabolism of tolterodine is highly variable
(1 to 100 ng/mL)41 and this variability in the generation of
the active metabolite makes individual tailoring of tolterodine
dose necessary in some patients.39
Fesoterodine has been developed as a sustained release
preparation with maximal plasma concentrations of 5-HMT
detected approximately 4 to 6 hours after oral administration.42
In contrast to tolterodine, the formation of the active metabolite from fesoterodine is not dependent on CYP2D6 activity
(Figure 1) but rather occurs due to hydrolysis of fesoterodine
O
O
N
H
OH
Fesoterodine
non-specific
esterases
OH
OH
N
H
N
CYP2D6
OH
H
CYP2D6
Inactive
metabolite
5-HMT
Tolterodine
CYP3A4
Inactive
metabolite
Figure Metabolic pathways responsible for the generation of 5-hydroxymethyl-tolterodine (5-HMT) from tolterodine and fesoterodine.
Open Access Journal of Urology 2010:2
submit your manuscript | www.dovepress.com
Dovepress
3
Mansfield
Dovepress
by non-specific esterases. No fesoterodine is detected in the
plasma of patients indicating that metabolism of fesoterodine
is rapid and complete.41,43 Also, in contrast to tolterodine, there
is little inter-subject variability in the generation of 5-HMT
from fesoterodine, as the activity of the non-specific esterases
is similar in all people.37,44 Oral administration of a single dose
of 4 mg fesoterodine, results in a maximal plasma concentration of 5-HMT between 1 and 10 ng/mL,41,45 with plasma
concentrations of 5-HMT overlapping in patients characterized
as extensive (0.9 to 5.6 ng/mL) or poor metabolizers (2.0 to
10.9 ng/mL).41 In addition, plasma concentrations of 5-HMT
increases linearly with increasing fesoterodine dose.41 A large
proportion of the active metabolite, 5-HMT, is transported in the
plasma unbound (36 to 54%),39,42 in contrast to tolterodine which
is almost entirely bound to serum albumin (3.7% unbound).39
Breakdown of 5-HMT, generated from either tolterodine
or fesoterodine, to inactive metabolites occurs via cytochrome P450 3A4 and CYP2D6 (Figure 1) and is therefore
varied in extensive and poor metabolizers.45,46 Approximately
70% of the fesoterodine dose is eliminated via the urine with
approximately 16% being eliminated as 5-HMT42 the rest
as inactive metabolites.43 The urine elimination of 5-HMT
increasing proportionally with fesoterodine dose.43 Excretion of 5-HMT is slowed in patients with renal impairment,
however this delay in excretion was not associated with a
significant increase in adverse events in these patients.42
Efficacy of fesoterodine
and tolterodine in clinical trials
Both fesoterodine and tolterodine have been associated with
clinical efficacy that exceeds placebo in a number of important
clinical variables. Four recent randomized controlled trials
have compared the clinical efficacy of fesoterodine with
placebo,37,47–49 two of which also compared fesoterodine
with tolterodine.47,49 Selected results from these clinical trials
are summarized in Table 2. These results indicate that both
fesoterodine and tolterodine have clinical efficacy against
symptoms related to bladder filling (micturition frequency
and mean voided volume) and urgency (urgency episodes,
urge incontinence episodes).
In regards to symptoms related to bladder filling, treatment for 12 weeks with a once daily dose of fesoterodine
(4 mg) resulted in a significant decrease in micturition
frequency (5.5% greater than the average placebo effect)
which corresponds to 1.7 less micturitions per 24 hours.
Fesoterodine (4 mg) was also associated with an increase
in mean voided volume of 25.1 mL compared to an average
increase in placebo of 9.3 mL. These changes in clinical outcomes with 4 mg fesoterodine were comparable to tolterodine
(4 mg) (Table 2).47,49
Fesoterodine was also effective against symptoms related
to urgency. Treatment with 4 mg fesoterodine resulted in
a significant decrease in urge incontinence episodes and
urgency episodes. In OAB wet patients the decrease in urge
incontinence episodes was 26.6% greater than the average
placebo effect; that is 1.9 less episodes of urge incontinence
per 24 hours (Table 2). In addition there was a significant
decrease in urgency episodes which was 8.2% greater than
the average placebo effect (Table 2). This corresponds to
2 less urgency episodes per 24 hours.37,47–49
One interesting feature of fesoterodine treatment is
that the improvements in clinical efficacy are shown to be
Table Efficacy of fesoterodine and tolterodine in 12 week clinical trials for OAB therapy
Range of
baseline values
Placeboa,b
4 mg
8 mg
Micturition frequency
11.5–12.9
−9.3%
−15.5%*,†
−16.9%*,†
Fesoterodinea
Tolterodine ERb
4 mg
−15.6%*
MVV (mL)
150–160
+9.04
+23.0*
+33.2* **
+25.1*
Urgency episodes
11–12.5
−7.5%
−16.9%*,†
−18.8%*,†
−17.5%*
UUI episodes
3.7–4.0
−40.7%
−67.5%*,†
−77.9%*,†,**
−56.3%
Continent days/week
0.6–0.8
+1.7
+2.6*,†
+3.0*,†,**
+2.6*
,†
,†,
Notes: Statistical significance reported: *P 0.05 vs placebo, †P 0.001 vs placebo and **P 0.05 vs 4 mg fesoterodine treatment.
Definition of measures:
Micturition frequency is the number of times a patient passed urine (including incontinence episodes) in a 24-hour period.
MVV is the mean voided volume (mL) per micturition determined from a 1-day collection period.
Urgency episodes is the number of times a patient recorded an urgency episode with or without incontinence per day determined from a 3-day bladder diary.
Urge urinary incontinence is the number of times the patient experiences involuntary leakage of urine accompanied by or immediately preceded by urgency in a 24-hour
period determined from a 3-day bladder diary.
Continent days per week were normalized from a 3-day bladder diary.
a
Data summarized from37,47–49
b
Data summarized from47,49
4
submit your manuscript | www.dovepress.com
Dovepress
Open Access Journal of Urology 2010:2
Fesoterodine for overactive bladder
Dovepress
increased with increasing dose (8 mg compared to 4 mg)
(Table 2). Treatment with 8 mg fesoterodine resulted in a
significantly greater decrease in urge incontinence episodes
per 24 hours (9.1% greater than the average decrease with
4 mg fesoterodine), together with an increase in mean voided
volume (8 mL greater than the average improvement with
4 mg fesoterodine). In addition, treatment with 8 mg fesoterodine was also associated with an increase in the number of
continent days per week to 3.1 days compared to 2.7 days
with fesoterodine 4 mg.47–49
Fesoterodine is unusual in showing this dose response
relationship as a similar relationship has not been demonstrated for other antimuscarinic agents including
tolterodine,50–52 or the muscarinic M3 receptors selective
agents, darifenacin 53 and solifenacin. 54 It is likely that
this dose response relationship is a result of the simple
metabolism of fesoterodine by the non-specific esterases
and the associated linear relationship between fesoterodine
dose and plasma concentrations of the active compound,
5-HMT.41,43
In addition to the improvements in clinical outcomes
fesoterodine has also been associated with improvements
in Health Related Quality of life (HRQoL). The King’s
Health Questionnaire, which examines 9 domains related
to quality of life,55 has been used to assess improvements in
HRQoL in people who suffer from OAB following 12 week
treatment with fesoterodine.56,57 Significant improvement in
five or more domains of the King’s Health Questionnaire is
considered to indicate meaningful improvement in patient
quality of life.58 Similar to clinical efficacy, improvements
in HRQoL were related to fesoterodine dose. Treatment
with 8 mg fesoterodine showed significant improvements
(compared with placebo) in 8 of the 9 domains assessed by
the King’s Health Questionnaire. While 4 mg fesoterodine
(and tolterodine 4 mg), showed significant improvements
(compared with placebo) in 7 of the 9 domains. 59 The
domains where fesoterodine was associated with improvement include: severity/coping, emotions, role limitations,
physical limitations, social limitations, sleep/energy, personal relationship and incontinence impact.56,57 Of these
improvements all except benefit for personal relationship
were seen in patients who were classified as both OAB
wet and OAB dry.56 Treatment with 8 mg fesoterodine
also showed significantly greater improvement compared
to 4 mg fesoterodine in domains of severity/coping and
emotions.57
Adverse events associated with
fesoterodine and tolterodine
While muscarinic antagonists can be used to effectively
treat OAB in approximately 65% of patients, numerous
patients discontinue therapy long term due to adverse events
including dry mouth and constipation.59 These adverse events
occur due to a lack of organ selectivity of antimuscarinic
agents60 as muscarinic receptors are not only located on the
detrusor muscle but also in the salivary glands61 and smooth
muscle of the gastrointestinal tract.13,62 The incidence of these
adverse events in clinical trials of fesoterodine and tolterodine
are summarized in Table 3.37,47–49
Dry mouth was the most common adverse event
associated with fesoterodine use (Table 3), although most
patients described it as mild to moderate. Twenty percent of
patients being treated with 4 mg fesoterodine reported dry
mouth.37,47–49,63 This was increased to 35% in patients being
treated with 8 mg fesoterodine37,47–49 compared to an incidence
of 6% in placebo (Table 3). Although common, dry mouth
did not account for a large number of patients withdrawing
from the 12 week clinical trials (Table 3). The incidence of
dry mouth with fesoterodine (4 mg) was comparable to the
incidence in patients treated with tolterodine (4 mg)30,47,49,64
however it was considerably lower than that reported with
oxybutynin (Table 3).30,65
The reason for the decrease in incidence of dry mouth
with fesoterodine (and tolterodine) compared to oxybutynin
may lie in the selectivity of 5-HMT for the bladder over the
salivary gland (Table 4). Radiologand binding studies in
Table 3 Incidence of dry mouth and constipation in patients treated with oxybutynin, tolterodine, and fesoterodine
Antimuscarinic dose
Dry mouth
Constipation
Discontinuation due
to adverse events
Reference
Oxybutynin
5 and 10 mg
32.9%
7.3%
1.8%
Data summarized from30,65
Tolterodine ER
4 mg
19.8%
4%
2.5%
Data summarized from30,47,49,64
Fesoterodine
4 mg
20.2%
4.4%
5%
Data summarized from37,47–49,63
Fesoterodine
8 mg
34.7%
5.1%
6.8%
Data summarized from37,47–49
4.9%
2.5%
3.1%
Data summarized from37,47–49,65
Placebo
Open Access Journal of Urology 2010:2
submit your manuscript | www.dovepress.com
Dovepress
5
Mansfield
Dovepress
Table 4 Selectivity of antimuscarinic agents for bladder and salivary gland as determined by radioligand binding and in vivo functional studies
Compound
Radioligand binding studies
In vivo functional studies
Ki (nM)
ID50 (nmol/kg iv)a
Bladder
Salivary gland
Bladder
Salivary gland
Reference
Oxybutynin
9.8
3.02
215
76
Data summarized from66,86,93
Tolterodine
2.7
4.8
101
257
Data summarized from66,90,91
5-HMT
2.9
5.2
15
40
Data summarized from27
Note: data are not available for fesoterodine as it is completely metabolized to 5-HMT.
a
ID50 determined from in vivo functional studies where antagonists were infused into anesthetized animals. Bladder contraction was stimulated by intra-arterial acetylcholine.
Salivation was stimulated by electrical stimulation of the chorda-lingual nerve.27,66,91
salivary gland and bladder have demonstrated that oxybutynin has a three times higher affinity for salivary gland over
the bladder (Table 4). In contrast tolterodine and 5-HMT
have an affinity for the bladder that is twice that for the
salivary gland (Table 4). Further to this, functional studies
have examined the selectivity of oxybutynin, tolterodine and
5-HMT, for the bladder and salivary gland by comparing the
concentrations required to inhibit detrusor contractions and
salivation in vivo. Similar to the results from radioligand
binding studies, oxybutynin inhibits salivation at a concentration that is approximately one-third lower than that required
to inhibit bladder contraction (Table 4) indicating some
degree of selectivity for the salivary gland.66 In contrast,
5-HMT inhibits bladder contractions at a lower concentration than that required to inhibit salivation27 indicating some
degree of selectivity for the bladder. The reasons for this
bladder selectivity of 5-HMT is unknown and cannot be
explained simply by selectivity for individual muscarinic
receptor subtypes.
Antimuscarinic agents show clinical
efficacy against the symptom of
urgency
Fesoterodine and tolterodine have demonstrated efficacy
against symptoms of urgency as demonstrated by improvements in the clinical variables of urge incontinence episodes
and urgency episodes per 24 hours (Table 2).37,47–49,67 This is
similar to reports from clinical trials with other muscarinic
receptor antagonists, including the M3 selective agent
solifenacin68–70 and trospium.71 However, these beneficial
effects, on the symptoms of urgency, raises the question as to
how urgency is sensed, what receptors are involved and why
antimuscarinic agents are effective against urgency. Answering these questions and understanding how these antimuscarinic agents are efficacious against urgency is important,
as urgency is the cornerstone symptom for OAB and the
symptom that patients identify as their most bothersome.6
submit your manuscript | www.dovepress.com
Dovepress
Recent reviews have suggested that at therapeutic doses,
muscarinic antagonists do not appear to inhibit bladder
contractility,8,72 and their activity is now thought to be during
bladder filling to increase bladder capacity and to decrease
urgency8 actions not attributable to inhibition of muscarinic
receptors located on the detrusor.
Radioligand binding studies in both pig73 and human
bladder17 have demonstrated M2 (70%) and M3 (30%) muscarinic receptors in the bladder mucosa. Mucosal muscarinic
receptors have also been demonstrated using molecular
RT-PCR studies which demonstrate expression of mRNA
for M1, M2, M3, and M5.17,74,75 Immunohistochemical studies
have localized muscarinic receptor immunoreactivity to
the bladder urothelium74–76 and to suburothelial myofibroblasts.76,77 The role of these mucosal muscarinic receptors
in bladder micturition remains unclear. However, they may
represent a site of action for the muscarinic receptor antagonists used to treat OAB.
Recent, in vivo studies in rat bladder have demonstrated
that intravesical administration of carbachol can induce
detrusor overactivity,78 where as intravesical instillation of
muscarinic antagonists including oxybutynin,79 tolterodine80
and darifenacin81 reduces stretch activated afferent nerve
firing in rat bladder, an effect also seen following systemic
administration of oxybutynin.82 Furthermore, clinical efficacy
has been associated, in patients with OAB, with intravesical
instillation of oxybutynin83,84 It is possible that following oral
administration of fesoterodine or tolterodine the presence
of the active metabolites, 5-HMT, in the urine as a result of
urinary excretion is partly responsible for the clinical efficacy
of these agents.
Conclusion
Fesoterodine is a pro-drug developed to produce the active
metabolite, 5-hydroxy-methyl-tolterodine (5-HMT) via the
actions of non-specific esterases. This metabolism of fesoterodine results in the complete breakdown of the parent
Open Access Journal of Urology 2010:2
Dovepress
compound and is responsible for dose related improvements in clinical efficacy and health related quality of life.
Fesoterodine, like tolterodine and other antimuscarinic
agents, has been shown to have clinical efficacy for the
treatment of patients suffering from OAB. Treatment with
fesoterodine is associated with improvements in clinical variables related both to bladder filling (decreasing micturition
frequency and increasing mean voided volume) and urgency
(urgency and urge incontinence episodes). Fesoterodine
is also associated with significant improvements in health
related quality of life as indicated by improvements in at
least 7 of the 9 variables measured by the King’s Health
Questionnaire. Fesoterodine, like other antimuscarinic
agents, is associated with adverse events such as dry mouth
and constipation. However the incidence of these adverse
events is reduced compared, to the original muscarinic
antagonist oxybutynin, due to the improved bladder selectivity of 5-HMT.
Disclosure
The author declares no conflicts of interest.
References
1. Milsom I, Abrams P, Cardozo L, et al. How widespread are the symptoms
of an overactive bladder and how are they managed? A population-based
prevalence study. BJU Int. 2001;87(9):760–766.
2. Stewart WF, Van Rooyen JB, Cundiff GW, et al. Prevalence and burden
of overactive bladder in the United States. World J Urol. 2003;20(6):
327–336.
3. Abrams P, Cardozo L, Fall M, et al; for Standardisation Sub-committee
of the International Continence Society. The standardisation of terminology of lower urinary tract function: report from the Standardisation
Sub-committee of the International Continence Society. Neurourol
Urodyn. 2002;21(2):167–178.
4. Tubaro A. Defining overactive bladder: epidemiology and burden of
disease. Urology. 2004;64(6 Suppl 1):2–6.
5. Hu TW, Wagner TH, Bentkover JD, et al. Estimated economic costs
of overactive bladder in the United States. Urology. 2003;61(6):
1123–1128.
6. Brubaker L. Urgency: the cornerstone symptom of overactive bladder.
Urology. 2004;64(6 Suppl 1):12–16.
7. Abrams P, Andersson KE. Muscarinic receptor antagonists for overactive
bladder. BJU Int. 2007;100(5):987–1006.
8. Andersson KE, Yoshida M. Antimuscarinics and the overactive detrusor-which is the main mechanism of action? Eur Urol.
2003;43(1):1–5.
9. Hegde SS, Muscarinic receptors in the bladder: from basic research to
therapeutics. Br J Pharmacol. 2006;147(Suppl 2):S80–S87.
10. Ferguson DR, Kennedy I, Burton TJ. ATP is released from rabbit urinary bladder epithelial cells by hydrostatic pressure changes – a possible
sensory mechanism? J Physiol. 1997;505(2):503–511.
11. Downie JW, Dean DM. The contribution of cholinergic postganglionic
neurotransmission to contractions of rabbit detrusor. J Pharmacol Exp
Ther. 1977;203(2):417–425.
12. Caulfield MP, Birdsall NJ. International Union of Pharmacology. XVII.
Classification of muscarinic acetylcholine receptors. Pharmacol Rev.
1998;50(2):279–290.
Open Access Journal of Urology 2010:2
Fesoterodine for overactive bladder
13. Eglen RM, Hegde S, Watson N. Muscarinic receptor subtypes and
smooth muscle function. Pharmacol Rev. 1996;48(4):531–565.
14. Nilvebrant L, Andersson KE, Mattiasson A. Characterization of the
muscarinic cholinoceptors in the human detrusor. J Urol. 1985;134(2):
418–423.
15. Wang P, Luthin GR, Ruggieri MR. Muscarinic acetylcholine receptor
subtypes mediating urinary bladder contractility and coupling to GTP
binding proteins. J Pharmacol Exp Ther. 1995;273(2):959–966.
16. Goepel M, Gronewald A, Krege S, et al. Muscarinic receptor subtypes
in porcine detrusor: comparison with humans and regulation by bladder
augmentation. Urol Res. 1998;26(2):149–154.
17. Mansfield KJ, Liu L, Mitchelson FJ, et al. Muscarinic receptor subtypes in human bladder detrusor and mucosa, studied by radioligand
binding and quantitative competitive RT-PCR: changes in ageing. Br J
Pharmacol. 2005;144(8):1089–1099.
18. Matsui M, Motomura D, Karasawa H, et al. Multiple functional defects
in peripheral autonomic organs in mice lacking muscarinic acetylcholine receptor gene for the M3 subtype. Proc Nat Acad Sci U S A.
2000;97(17):9579–9584.
19. Chess-Williams R, Chapple CR, Yamanishi T, et al. The minor
population of M3-receptors mediate contraction of human detrusor
muscle in vitro. J Auton Pharmacol. 2001;21(5–6):243–248.
20. Fetscher C, Fleichman M, Schmidt M, et al. M(3) muscarinic receptors mediate contraction of human urinary bladder. Br J Pharmacol.
2002;136(5):641–643.
21. Matsui M, Motomura D, Fujikawa T, et al. Mice lacking M2 and
M3 muscarinic acetylcholine receptors are devoid of cholinergic
smooth muscle contractions but still viable. J Neurosci. 2002;22(24):
10627–10632.
22. Ehlert FJ. Contractile role of M2 and M3 muscarinic receptors in
gastrointestinal, airway and urinary bladder smooth muscle. Life Sci.
2003;74(2–3):355–266.
23. Fredericks CM, Green RL, Anderson GF. Comparative in vitro effects
of imipramine, oxybutynin, and flavoxate on rabbit detrusor. Urology.
1978;12(4):487–491.
24. Paulson DF. Oxybutynin chloride in control of post-trasurethral vesical
pain and spasm. Urology. 1978;11(3):237–238.
25. Moisey CU, StephensonTP, Brendler CB. The urodynamic and
subjective results of treatment of detrusor instability with oxybutynin
chloride. Br J Urology. 1980;52(6):472–475.
26. Bary PR, Moisey CU, Stephenson TP, et al. The urodynamic and subjective results of treatment of detrusor instability with oxybutynin chloride.
Prog Clin Biol Res. 1981;78:313–319.
27. Nilvebrant L, Gillberg PG, Sparf B. Antimuscarinic potency and
bladder selectivity of PNU-200577, a major metabolite of tolterodine.
Pharmacol Toxicol. 1997;81(4):169–172.
28. Van Kerrebroeck P, Kreder K, Jonas U, et al; for Tolterodine Study
Group. Tolterodine once-daily: superior efficacy and tolerability in the
treatment of the overactive bladder. Urology. 2001;57(3):414–421.
29. Sussman D, Garely A. Treatment of overactive bladder with oncedaily extended-release tolterodine or oxybutynin: the antimuscarinic
clinical effectiveness trial (ACET). Cur Med Res Opin. 2002;18(4):
177–184.
30. Diokno AC, Appell RA, Sand PK, et al; for the OPERA Study Group.,
Prospective, randomized, double-blind study of the efficacy and
tolerability of the extended-release formulations of oxybutynin and
tolterodine for overactive bladder: results of the OPERA trial. Mayo
Clin Proc. 2003;78(6):687–695.
31. Kaplan SA, Roehrborn CG, Dmochowski R, et al. Tolterodine extended
release improves overactive bladder symptoms in men with overactive
bladder and nocturia. Urology. 2006;68(2):328–332.
32. Dmochowski R, Abrams P, Marschall-Kehrel D, et al. Efficacy and
tolerability of tolterodine extended release in male and female patients
with overactive bladder. Eur Urol. 2007;51(4):1054–1064.
33. Andersson SH, Lindgren A, Postlind H. Biotransformation of tolterodine, a new muscarinic receptor antagonist, in mice, rats, and dogs.
Drug Met Disp. 1998;26(6):528–535.
submit your manuscript | www.dovepress.com
Dovepress
Mansfield
Dovepress
34. Postlind HD, Lindgren A, Andersson SH. Tolterodine, a new muscarinic
receptor antagonist, is metabolized by cytochromes P450 2D6 and 3A
in human liver microsomes. Drug Met Disp. 1998;26(4):289–293.
35. Palmér L, Andersson L, Andersson T, et al. Determination of tolterodine and the 5-hydroxymethyl metabolite in plasma, serum and urine
using gas chromatography-mass spectrometry. J Pharm Biomed Anal.
1997;16(1):155–165.
36. Ney P, Pandita RK, Newgreen DT, et al. Pharmacological characterization of a novel investigational antimuscarinic drug, fesoterodine, in vitro
and in vivo. BJU Int. 2008;101(8):1036–1042.
37. Khullar V, Rovner ES, Dmochowski R, et al. Fesoterodine dose
response in subjects with overactive bladder syndrome. Urology.
2008;71:839–843.
38. Michel MC, Hedge S. Treatment of the overactive bladder syndrome
with muscarinic receptor antagonists: a matter of metabolites? Naunyn
Schmiedebergs Arch Pharmacol. 2006;374(2):79–85.
39. Brynne N, Dalén P, Alván G, et al. Influence of CYP2D6 polymorphism
on the pharmacokinetics and pharmacodynamic of tolterodine. Clin
Pharmacol Ther. 1998;63(5):529–539.
40. Xie HG, Kim RB, Wood AJ, et al. Molecular basis of ethnic differences
in drug disposition and response. Ann Rev Pharmacol Toxicol.
2001;41:851–850.
41. Simon HU, Malhotra B. The pharmacokinetic profile of fesoterodine: similarities and differences to tolterodine. Swiss Med Weekly.
2009;139(9–10):146–151.
42. Malhotra B, Gandelman K, Sachse R, et al. Assessment of the effects
of renal impairment on the pharmacokinetic profile of fesoterodine.
J Clin Pharmacol. 2009;49(4):477–482.
43. Sachse R, Cawello W, Haag C, et al. Pharmacodynamics and pharmacokinetics of ascending multiple doses of the novel bladder-selective
antimuscarinic fesoterodine. Eur Urol. 2003;2(1):30.
44. Cawello W, Auer S, Hammes W, et al. Multiple dose pharmacokinetics of fesoterodine in human subjects. Naunyn-Schmiedeberg’s Arch
Pharmacol. 2002;365(Suppl 1):R110.
45. Malhotra B, Sachse R, Wood N. Evaluation of drug-drug interactions
with fesoterodine. Eur J Clin Pharmacol. 2009;65(6):551–560.
46. Malhotra B, Guan Z, Wood N, et al. Pharmacokinetic profile of fesoterodine. Int J Clin Pharmacol Ther. 2008;46(11):556–563.
47. Chapple C, V Kerrebroeck P, Tubaro A, et al. Clinical efficacy, safety,
and tolerability of once-daily fesoterodine in subjects with overactive
bladder. Eur Urol. 2007;52(4):1204–1212.
48. Nitti VW, Dmochowski R, Sand PK, et al. Efficacy, safety and tolerability of Fesoterodine for overactive bladder syndromes. J Urol.
2007;178:2488–2494.
49. Sand PK, Morrow J, Bavendam T, et al. Efficacy and tolerability of
fesoterodine in women with overactive bladder. Int Urogynaecol J.
2009;20:827–835.
50. Larsson G, Hallén B, Nilvebrant L. Tolterodine in the treatment of
overactive bladder: analysis of the pooled phase II efficacy and safety
data. Urology. 1999;53(5):990–998.
51. Millard R, Tuttle J, Moore KH, et al. Clinical efficacy and safety
of tolterodine compared to placebo in detrusor overactivity. J Urol.
1999;161(5):1551–1555.
52. Van Kerrebroeck PE, Amarenco G, Thüroff JW, et al. Dose-ranging
study of tolterodine in patients with detrusor hyperreflexia. Neurourol
Urodyn. 1998;17(5):499–512.
53. Hill S, Khullar V, Wyndaele JJ, et al; for the Darifenacin Study Group.
Dose response with darifenacin, a novel once-daily M3 selective receptor
antagonist for the treatment of overactive bladder: results of a fixed dose
study. Int Urogynecol J Pelvic Floor Dysfunct. 2006;17:239–247.
54. Cardozo L, Lisec M, Millard R, et al. Randomized, double-blind
placebo controlled trial of the once daily antimuscarinic agent
solifenacin succinate in patients with overactive bladder. J Urol.
2004;172:1919–1924.
55. Kelleher CJ, Cardozo LD, Khullar V, et al. A new questionnaire to
assess the quality of life of urinary incontinent women. Br J Obstet
Gynaecol. 1997;104(12):1374–1279.
8
submit your manuscript | www.dovepress.com
Dovepress
56. Chapple CR, Van Kerrebroeck P, Jünemann KP, et al. Comparison of
fesoterodine and tolterodine in patients with overactive bladder. BJU
Int. 2008;102(9):1128–1132.
57. Kelleher CJ, Tubaro A, Wang JT, et al. Impact of fesoterodine on quality
of life: pooled data from two randomized trials. BJU Int. 2008;102(1):
56–61.
58. Kelleher CJ, Pleil A, Reese PR, et al. How much is enough and who
says so? BJOG. 2004;111(6):605–612.
59. Morris AR, Westbrook J, Moore KH. A longitudinal study over 5 to
10 years of clinical outcomes in women with idiopathic detrusor overactivity. BJOG. 2008;115(2):239–246.
60. Colli E, Digesu G, Olivieri L. Overactive bladder treatments in early
phase clinical trials. Exp Opin Invest Drugs. 2007;16(7):999–1007.
61. Martos F, Bermudez R, Gomez A, et al. Characterization of muscarinic
receptors in human submandibular salivary glands. Eur J Pharmacol.
1985;116(3):319–321.
62. Mansfield KJ, Mitchelson F, Moore KH, et al. Muscarinic receptor
subtypes in the human colon: lack of evidence for atypical subtypes.
Eur J Pharmacol. 2004;482(1–3):101–109.
63. Wyndaele JJ, Goldfischer ER, Morrow JD, et al. Effects of flexible-dose
fesoterodine on overactive bladder symptoms and treatment satisfaction:
an open-label study. Int J Clin Pract. 2009;63(4):560–567.
64. Chapple CR, Fianu-Jonsson A, Indig M, et al; for the STAR study group.
Treatment outcomes in the STAR study: a subanalysis of solifenacin
5 mg and tolterodine ER 4 mg. Eur Urol. 2007;52(4):1195–1203.
65. Zinner N, Tuttle J, Marks L. Efficacy and tolerability of darifenacin,
a muscarinic M3 selective receptor antagonist (M3 SRA), compared
with oxybutynin in the treatment of patients with overactive bladder.
World J Urol. 2005;23(4):248–252.
66. Nilvebrant L, Andersson KE, Gillberg PG, et al. Tolterodine – a
new bladder-selective antimuscarinic agent. Eur J Pharmacol. 1997;
327(2–3):195–207.
67. Freeman R, Hill S, Millard R, et al; for the Tolterodine Study Group.
Reduced perception of urgency in treatment of overactive bladder
with extended-release tolterodine. Obstet Gynaecol. 2003;102(3):
605–611.
68. Chapple CR, Araño P, Bosch JL, et al. Solifenacin appears effective and
well tolerated in patients with symptomatic idiopathic detrusor overactivity in a placebo- and tolterodine-controlled phase 2 dose-finding
study. BJU Int. 2004;93(1):71–77.
69. Millard RJ, Halaska M. Efficacy of solifenacin in patients with severe
symptoms of overactive bladder: a pooled analysis. Curr Med Res Opin.
2006;22(1):41–48.
70. Wagg A, Wyndaele JJ, Sieber P. Efficacy and tolerability of solifenacin
in elderly subjects with overactive bladder syndrome: a pooled analysis.
Am J Geriatr Pharmacother. 2006;4(1):14–24.
71. Zinner N, Gittelman M, Harris R, et al; for the Trospium Study Group.
Trospium chloride improves overactive bladder symptoms: a multicenter phase III trial. J Urol. 2004;171(6 pt 1):2311–2315.
72. Finney SM, Andersson KE, Gillespie JI, et al. Antimuscarinic drugs in
detrusor overactivity and the overactive bladder syndrome: motor or
sensory actions? BJU Int. 2006;98(3):503–507.
73. Hawthorn MH, Chapple CR, Cock M, et al. Urothelium-derived inhibitory factor(s) influences on detrusor muscle contractility in vitro. Br J
Pharmacol. 2000;129(3):416–419.
74. Bschleipfer T, Schukowski K, Weidner W, et al. Expression and distribution of cholinergic receptors in the human urothelium. Life Sci.
2007;80(24–25):2303–2307.
75. Tyagi S, Tyagi P, Van-le S, et al. Qualitative and quantitative expression profile of muscarinic receptors in human urothelium and detrusor.
J Urol. 2006;176(4 pt 1):1673–1678.
76. Mukerji G, YiangouY, Grogono J, et al. Localization of M2 and M3
muscarinic receptors in human bladder disorders and their clinical
correlations. J Urol. 2006;176(1):367–373.
77. Grol S, Essers P, van Koeveringe GA, et al. M(3) muscarinic receptor
expression on suburothelial interstitial cells. BJU Int. 2009;104(3):
398–405.
Open Access Journal of Urology 2010:2
Fesoterodine for overactive bladder
Dovepress
78. Kim Y, Yoshimura N, Masuda H, et al. Antimuscarinic agents exhibit
local inhibitory effects on muscarinic receptors in bladder-afferent
pathways. Urology. 2005;65(2):238–242.
79. de Wachter S, Wyndaele JJ. Intravesical oxybutynin: A local anaesthetic
effect on bladder C afferents. J Urol. 2003;169:1892–1895.
80. Yokoyama O, Yusup A, Miwa Y, et al. Effects of tolterodine on an
overactive bladder depend on suppression of C-fiber bladder afferent
activity in rats. J Urol. 2005;174(5):2032–2036.
81. Iijima K, de Wachter S, Wyndaele JJ. Effects of the M3 receptor selective muscarinic antagonist darifenacin on bladder afferent activity of
the rat pelvic nerve. Eur Urol. 2007;52(3):842–847.
82. de Laet K, de Wachter S, Wyndaele JJ. Systemic oxybutynin decreases
afferent activity of the pelvic nerve of the rat: new insights into the
working mechanism of antimuscarinics. Neurourol Urodyn. 2006;
25(2):156–161.
83. Madersbacher H, Jilg G. Control of detrusor hyperreflexia by the
intravesical instillation of oxybutynine hydrochloride. Paraplegia.
1991;29(2):84–90.
84. Lose G, Nørgaard JP. Intravesical oxybutynin for treating incontinence
resulting from an overactive detrusor. BJU Int. 2001;87(9):767–773.
85. Ikeda K, Kobayashi S, Suzuki M, et al. M(3) receptor antagonism
by the novel antimuscarinic agent solifenacin in the urinary bladder and salivary gland. Naunyn-Schmiedeberg’s Arch Pharmacol.
2002;366(2):97–103.
86. Moriya H, Takagi Y, Nakanishi T, et al. Affinity profiles of various muscarinic antagonists for cloned human muscarinic acetylcholine receptor
(mAChR) subtypes and mAChRs in rat heart and submandibular gland.
Life Sci. 1999;64(25):2351–2358.
87. Watson N, Daniels DV, Ford AP, et al. Comparative pharmacology of
recombinant human M3 and M5 muscarinic receptors expressed in
CHO-K1 cells. Br J Pharmacol. 1999;127(2):590–596.
88. McNamara A, Pulido-Rios MT, Sweazey S, et al. Pharmacological
properties of TD-6301, a novel bladder selective muscarinic receptor
antagonist. Eur J Pharmacol. 2009;605(1–3):145–153.
89. Ohtake A, Saitoh C, Yuyama H, et al. Pharmacological characterization
of a new antimuscarinic agent, solifenacin succinate, in comparison with
other antimuscarinic agents. Biol Pharm Bull. 2007;30(1):54–58.
90. Nilvebrant L, Sundquist S, Gillberg PG. Tolterodine is not subtype
(m1–m5) selective but exhibits functional bladder selectivity in vivo.
Neurourol Urodyn. 1996;15:310–311.
91. Gillberg PG, Sundquist S, Nilvebrant L. Comparison of the in vitro and
in vivo profiles of tolterodine with those of subtype-selective muscarinic
receptor antagonists. Eur J Pharmacol. 1998;349(2–3):285–292.
92. Mansfield KJ, Chandran J, Vaux KJ, et al. Comparison of receptor
binding characteristics of commonly used muscarinic antagonists
in human bladder detrusor and mucosa. J Pharmacol Exp Ther.
2009;328(3):893–899.
93. Ito Y, Oyunzul L, Yoshida A, et al. Comparison of muscarinic receptor
selectivity of solifenacin and oxybutynin in the bladder and submandibular gland of muscarinic receptor knockout mice. Eur J Pharmacol.
2009;615(1–3):201–206.
Open Access Journal of Urology
Dovepress
Publish your work in this journal
The Open Access Journal of Urology is an international, peer-reviewed,
open access journal publishing original research, reports, editorials,
reviews and commentaries on all aspects of adult and pediatric urology
in the clinic and laboratory including the following topics: Pathology,
pathophysiology of urological disease; Investigation and treatment of
urological disease; Pharmacology of drugs used for the treatment of
urological disease. The manuscript management system is completely
online and includes a very quick and fair peer-review system, which
is all easy to use. Visit http://www.dovepress.com/testimonials.php to
read real quotes from published authors.
Submit your manuscript here: http://www.dovepress.com/open-access-journal-of-urology-journal
Open Access Journal of Urology 2010:2
submit your manuscript | www.dovepress.com
Dovepress