throbber
CENTER FOR DRUG EVALUATION AND
`RESEARCH
`
`APPLICA TION NUMBER:
`
`22-030
`
`OFFICE DIRECTOR MEMO
`
`

`

`MEMORANDUM
`
`DEPARTMENT OF HEALTH AND HUMAN SERVICES
`PUBLIC HEALTH SERVICE
`FOOD AND DRUG ADMINISTRATION
`CENTER FOR DRUG EVALUATION AND RESEARCH
`
`DATE:
`
`October 31, 2008
`
`FROM:
`
`Julie Beitz, MD
`
`SUBJECT:
`
`Office Director Memo
`
`TO:
`
`NDA 22—030 Toviaz (fesoterodine fumarate) extended-release tablets;
`Pfizer, Inc.
`
`Summafl
`Fesoterodine fumarate is a muscarinic receptor antagonist that inhibits detrusor contractions and enhances
`bladder capacity. This memo documents my concurrence with the Division of Reproductive and Urologic
`Product’s (DRUP’s) decision that fesoterodine fumarate for the treatment of adults with overactive bladder
`with symptoms of urge urinary incontinence,-urgency, and urinary frequency be approved. The
`recommended starting dose is 4 mg once daily. Based upon individual response and tolerability, the dose
`may be increased to 8 mg once daily. The daily dose should not exceed 4 mg in patients with severe renal
`insufficiency or in patients taking potent CYP3A4 inhibitors. Fesoterodine fumarate is not recommended
`for use in patients with severe hepatic impairment.
`
`‘
`Regulatory Histog;
`NDA 22—030 was originally submitted by Schwarz Pharma Inc. on March 17, 2006, and received on March
`27, 2006. The sponsor’s API manufacturing facility in Shannon, Ireland, was not ready for FDA’s pre-
`approval inspection during the initial review cycle. On January 25, 2007, an approvable letter was issued
`stipulating that a satisfactory inspection of this site would be needed prior to approval and requesting that
`an updated summary of safety be submitted with the sponsor’s future response. On May 1, 2007, Schwarz
`Pharma Inc. submitted a complete response to the approvable letter. On August 1, 2007, ownership of this
`NDA was transferred from Schwarz Pharma, Inc. to Pfizer, Inc. During this review cycle, inspection of the
`Ireland manufacturing site was completed and it was found to be acceptable.
`
`This application was not referred to an FDA advisory committee because fesoterodine fumarate is a
`member of the anti-muscarinic class of products used to treat overactive bladder, including previously
`approved products. Fesoterodine fumarate did not pose unique concerns beyond those applicable to other
`members of this class.
`
`.
`Efficacy
`The efficacy of fesoterodine fumarate is supported by two 12-week randomized, placebo-controlled phase 3
`trials that enrolled adult patients with overactive bladder (with a combined total of 554 patients on
`fesoterodine fumarate 4 mg daily, 566 on fesoterodine fumarate 8 mg daily, and 554 on placebo). Patients
`were required to have overactive bladder symptoms for at least 6 months prior to study entry. The majority
`of patients were female (79%) with a mean age of 58 years. Both trials showed a statistically significant
`reduction from baseline to endpoint (Week 12) in the number of urge urinary incontinence episodes per 24
`hours for the 4 and 8 mg fesoterodine fumarate doses compared to placebo treatment. Both trials also
`showed a statistically significant reduction from baseline to endpoint (Week 12) in the number of
`micturitions per 24 hours for the 4 and 8 mg fesoterodine fumarate doses compared to placebo treatment.
`A reduction compared to placebo in the number of incontinence episodes was seen as early as 2 weeks
`following the start of fesoterodine fumarate treatment.
`
`For the secondary endpoint of voided volume per micturition, one of the phase 3 trials showed significant
`increases for both daily doses of fesoterodine fumarate compared to placebo treatment. In the second trial,
`
`

`

`significant improvement in voided volume was observed for the 8 mg daily close but not for the 4 mg daily
`dose.
`
`Safety
`A total of 2288 patients with overactive bladder received fesoterodine fumarate in phase 2 and 3 clinical
`trials. Of these, 782, 785, and 222 patients received fesoterodine fumarate doses of 4 mg, 8 mg, or 12 mg
`respectively, in Phase 2 and 3 trials with treatment periods of 8 to 12 weeks. Approximately 80% of these
`patients had > 10 weeks of exposure in these trials. Longer exposures to fesoterodine fumarate were
`observed in 857 patients for Z 6 months, in 701 patients for Z 12 months, in 529 patients for 3 24 months,
`and in 105 patients for 3 36 months. The safety profile for fesoterodine fumarate is consistent with that of
`other anti-muscarinics marketed for the treatment of overactive bladder; in fact, fesoterodine fumarate
`produces the same active metabolite as does tolterodine, a drug already marketed for this use. The most
`common adverse events reported were dry mouth, constipation and urinary retention. These events were
`generally mild or moderate in intensity. The incidence of dry mouth was highest in those taking 8 mg‘daily
`(35%) as compared to 4 mg daily (19%) or placebo treatment (7%). As with other anti-muscarinics, use of
`fesoterodine fumarate is contraindicated in patients with urinary retention, gastric retention, or uncontrolled
`narrow-angle glaucoma.
`
`A randomized, double-blind, placebo- and moxifloxacin-controlled study involving 261 subjects and
`designed to evaluate the risk of QT prolongation at therapeutic and supratherapeutic doses of fesoterodine
`fumarate (4 and28 mg daily doses) did not demonstrate abnormalities in cardiac repolarization related to
`fesoterodine fumarate use.
`
`Fesoterodine fumarate is associated with an increase in heart rate. In the phase 3 trials, the mean increase
`in heart rate compared to placebo was 3-4 beats/minute in patients treated with 4 mg daily and 3-5
`beats/minute in patients treated with 8 mg daily.
`
`No evidence of drug—related carcinogenicity was found in 24-month studies of oral administration to mice
`and rats. Fesoterodine fumarate was not mutagenic or genotoxic in vitro or in viva.
`
`Fesoterodine fumarate produced no dose—related teratogenic effects in mice and rabbits. Following oral
`doses 6-27 times the maximum human recommended dose (MHRD), female mice demonstrated increased
`resorptions and decreased live fetuses; fetuses with cleft palate were also noted at an incidence within the
`background range. Following oral administration to female rabbits, incomplete ossification of the
`stemebrae (retardation of bone development) was noted in fetuses. There have been no adequate and well—
`controlled studies of fesoterodine fumarate conducted in pregnant women. DRUP has proposed, and I
`concur, with a Pregnancy Category C, i.e., that this product should be used during pregnancy only if the
`potential benefit justifies the potential risk to the fetus.
`
`Pharmacokinetic Considerations
`
`Following oral administration, fesoterodine fumarate is rapidly and extensively hydrolyzed to its active
`metabolite, 5-hydroxymethyl tolterodine, which is responsible for the anti-muscarinic activity of
`fesoterodine fumarate. The active metabolite is further metabolized in the liver with involvement of
`CYP2D6 and CYP3A4. Exposure to the active metabolite is doubled in poor metabolizers of CYP2D6 as
`compared to extensive metabolizers. Poor metabolizers of CYP2D6 were also more likely than extensive
`metabolizers to experience anti-muscarinic adverse events (e.g., dry mouth), particularly at the 8 mg daily
`dose, however, no dosing adjustments are recommended based solely on intrinsic CYP2D6 metabolizer
`status. Co-administration of ketoconazole, a potent inhibitor of CYP3A4, with fesoterodine fumarate also
`increased exposure to the active metabolite two-fold; dry mouth occurred in up to 43% of patients c0-
`administered fesoterodine fumarate 8 mg daily and CYP3A4 inhibitors. Exposure was calculated as being
`nearly 6-fold higher in CYP2D6 poor metabolizers taking ketoconazole as compared to CYP2D6 extensive
`metabolizers not taking ketoconazole. Therefore, doses of fesoterodine fumarate greater than 4 mg are not
`recommended in patients taking potent CYP3A4 inhibitors.
`
`In patients with moderate (Child-Pugh B) hepatic impairment, Cmax and AUC of the active metabolite are
`1.4- and 2.1—fold higher respectively, as compared to healthy subjects. No dose adjustment is
`
`

`

`recommended for patients with mild to moderate hepatic insufficiency. Patients with severe hepatic
`impairment have not been studied; fesoterodine fumarate is not recommended for use in these patients.
`
`In patients with mild to moderate renal insufficiency (creatinine clearance of 30-80 mL/min), Cmax and
`AUC of the active metabolite are 1.5- and 1.8-fold higher respectively, as compared to healthy subjects. In
`patients with severe renal insufficiency (creatinine clearance of < 30 mL/min), Cmax and AUC are 2.0- and
`23-fold higher, respectively. No dose adjustment is recommended for patients with mild or moderate renal
`insufficiency. Doses of fesoterodine fumarate greater than 4 mg are not recommended in patients with
`severe renal insufficiency.
`
`Tradename Review
`
`The tradename "Toviaz" was submitted for review on April 1, 2008 and found to be acceptable. The
`previously submitted proposed tradename “Ii—vs. was also found acceptable.
`
`4
`h£ )
`
`Postmarket Studies
`
`The pediatric study requirement under PREA will be waived for ages 0 months to 5 years, 11 months,
`because necessary studies are impossible or highly impracticable. This is because study endpoints are
`difficult to evaluate in this age group.
`
`Submission of the pediatric study for ages 6 to 16 years, 11 months, is deferred because the product is
`ready for approval for use in adults and the pediatric study has not been completed. The deferred study will
`evaluate fesoterodine fumarate for the treatment of overactive bladder in the subgroup of pediatric patients
`with neurologic disease.
`
`No postmarket studies or clinical trials will be required under Title IX, Subtitle A, Section 901 of the Food
`and Drug Administration Amendments Act of 2007.
`
`Julie Beitz, MD
`Director,
`Office of Drug Evaluation 111
`CDER, FDA
`
`

`

`This is a representation of an electronic record that was signed electronically and
`this page is the manifestation of the electronic signature.
`
`Julie Beitz
`
`10/31/2008 10:14:42 AM
`DIRECTOR
`
`

`

`MEMORANDUM
`
`DEPARTMENT OF HEALTH AND HUMAN SERVICES
`PUBLIC HEALTH SERVICE
`FOOD AND DRUG ADMINISTRATION
`CENTER FOR DRUG EVALUATION AND RESEARCH
`
`DATE:
`
`January 25, 2007
`
`FROM:
`
`Julie Beitz, MD
`
`SUBJECT:
`
`Office Director Memo
`
`b(4)
`
`m4;
`
`TO:
`
`NDA 22—030 K; (fesoterodine fumarate) extended release tablets;
`Shwarz Pharma, LLC
`
`b(4)
`
`m F
`
`esoterodine fumarate is a muscarinic receptor antagonist that inhibits detrusor contractions and enhances
`bladder capacity. The proposed starting dose is 4 mg administered orally once daily; based on individual
`response and tolerability, the daily dose may be increased to 8 mg. This memo documents my concurrence
`with the Division of Reproductive and Urologic Product’s (DRUP’s) decision that fesoterodine fumarate
`for the treatment of adults with overactive bladder with symptoms of urge urinary incontinence, urgency,
`and urinary frequency is approvable. The sponsor’s API manufacturing facility in Shannon, Ireland, was to
`be ready for FDA’s pre-approval inspection during the final 4 months of this review cycle. Due to
`decisions made by the sponsor toW
`Schwarz Pharma informed DRUP in writing on July 20, 2006 that it would not be ready for inspection
`during this review cycle. Before fesoterodine fumarate may be approved, a satisfactory inspection of this
`facility will be needed.
`
`‘
`Efficacy
`NDA 22-030 was submitted on March 17, 2006, and received on March 27, 2006. The sponsor's claim for
`efficacy is supported by two 12-week randomized, placebo-controlled phase 3 studies that enrolled adult
`patients with overactive bladder (with a combined total of 554 patients on fesoterodine fumarate 4 mg
`daily, 566 on fesoterodine fumarate 8 mg daily, and 554 on placebo). Patients were required to have
`overactive bladder symptoms for at least 6 months, including urinary urgency, and urinary frequency, and
`in most patients, urge urinary incontinence. Both studies showed a statistically significant reduction from
`baseline to endpoint (Week 12) in the number of urge urinary incontinence episodes per 24 hours for the 4
`and 8 mg fesoterodine fumarate doses compared to placebo treatment. Both studies also showed a
`statistically significant reduction from baseline to endpoint (Week 12) in the number of micturitions per 24
`hours for the 4 and 8 mg fesoterodine fumarate doses compared to placebo treatment. Improvement in the
`number of incontinence episodes ‘3 was seen as early as 2 weeks following the start of
`fesoterodine fumarate treatment, although the improvements at Week 2 for the 4 mg (starting) dose were
`statistically better than placebo for the urge incontinence endpoint only.
`
`For the key secondary endpoint of voided volume per micturition, one study showed significant increases
`for both daily doses of fesoterodine fumarate compared to placebo treatment. In the second study,
`significant improvement in voided volume was observed for the 8 mg daily dose but not for the 4 mg daily
`dose.
`
`Safety
`A total of 2288 patients with overactive bladder received fesoterodine fumarate in phase 2 and 3 clinical
`studies. Of these, 782, 785, and 222 patients received fesoterodine fumarate doses of 4 mg, 8 mg, or 12 mg
`respectively, in Phase 2 and 3 trials with treatment periods of 8 to 12 weeks. Most of these patients
`(approximately 80% in each group) received > 10 weeks of exposure. Longer expOsures to fesoterodine
`fumarate were observed in 858 patients for Z 6 months and in 570 patients for Z 12 months. The safety
`profile for fesoterodine fumarate derived from these 8- and 12-week, randomized, placebo-controlled
`studies and their 1-year open-label extension periods in patients with overactive bladder is consistent with
`
`

`

`that of other anti-muscarinics; in fact, fesoterodine fumarate produces the same active metabolite as does
`tolterodine, a drug already marketed for this use. The most common adverse events reported were dry
`mouth, constipation and urinary retention. These events were generally mild to moderate in intensity. The
`incidence of dry mouth was highest in those taking 8 mg daily (35%) as compared to 4 mg daily (19%) or
`placebo treatment (7%). As with other anti-muscarinics, use of fesoterodine fumarate is contraindicated in
`patients with urinary retention, gastric retention, or uncontrolled narrow-angle glaucoma.
`
`A randomized, double-blind, placebo- and positive-controlled study designed to evaluate the risk of QT
`prolongation at therapeutic and supratherapeutic doses of fesoterodine fumarate (4 and 24 mg daily doses)
`did not demonstrate abnormalities in cardiac repolarization related to fesoterodine fumarate use.
`
`No evidence of drug-related carcinogenicity was found in 24-month studies of oral administration to mice
`and rats. Fesoterodine fumarate was not mutagenic or genotoxic in vitro or in vivo.
`
`Fesoterodine fumarate produced no frank teratogenic effects in mice. At exposures similar to or higher
`than the expected clinical exposure, female mice demonstrated an increased incidence of pre- and post-
`implantation fetal loss, including resorptions; fetuses with cleft palate were also noted. These findings are
`sometimes observed with maternal stress and have been seen with other anti-muscarinics. In a
`
`multigenerational study in female mice exposed from implantation through weaning, there were no
`occurrences of cleft palate. Following oral administration to female rabbits, post-implantation loss, early
`resorptions, and incomplete ossification of the stemebrae (retardation of bone development) were also
`noted. There have been no adequate and well-controlled studies of fesoterodine fumarate conducted in
`pregnant women. A search of FDA’s postmarketing adverse event reporting system (AERS) database
`revealed no cases of human teratogenicity in association with use of a similar product, tolterodine. DRUP
`has proposed, and I concur, with a Pregnancy Category C, i.e., that this product should be used during
`pregnancy only if the potential benefit justifies the potential risk to the fetus.
`
`Pharmacokinetic Considerations
`
`Following oral administration, fesoterodine fumarate is rapidly and extensively hydrolyzed to its active
`metabolite. The active metabolite is further metabolized in the liver with involvement of CYP2D6 and
`
`CYP3A4, with the CYP2D6 pathway being the major pathway. Exposure to the active metabolite is
`doubled in poor metabolizers of CYP2D6 as compared to extensive metabolizers. Poor metabolizers of
`CYP2D6 were also more likely than extensive metabolizers to experience anti-muscarinic adverse events
`(e.g., dry mouth), particularly at the 8 mg daily dose, however, no dosing adjustments are recommended
`based solely on intrinsic CYP2D6 metabolizer status. Co-administration of ketoconazole, a potent inhibitor
`of CYP3A4, with fesoterodine fumarate also increased exposure to the active metabolite two-fold; dry
`mouth occurred in up to 43% of patients co-administered fesoterodine fumarate 8 mg daily and CYP3A4
`inhibitors. Exposure was calculated as being approximately 4- to 5-fold higher in CYP2D6 poor
`metabolizers taking ketoconazole as compared to CYP2D6 extensive metabolizers not taking ketoconazole.
`Therefore, doses of fesoterodine fumarate greater than 4 mg are not recommended in patients taking potent
`CYP3A4 inhibitors.
`
`In patients with moderate (Child-Pugh B) hepatic impairment, Cmax and AUC of the active metabolite are
`1.4 and 2.1-fold higher compared to healthy subjects. No dose adjustment is recommended for patients
`with mild to moderate hepatic insufficiency. Patients with severe hepatic impairment have not been
`studied; fesoterodine fumarate is not recommended for use in these patients.
`
`In patients with mild to moderate renal insufficiency (creatinine clearance of 30-80 mL/min), Cmax and
`AUC of the active metabolite are 1.5 and 1.8-fold higher compared to healthy subjects. In patients with
`severe renal insufficiency (creatinine clearance of < 30 mL/min), Cmax and AUC are 2.0 and 23-fold higher
`compared to healthy subjects. No dose adjustment is recommended for patients with mild or moderate
`renal insufficiency. Doses of fesoterodine fumarate greater than 4 mg are not recommended in patients
`with severe renal insufficiency.
`
`Tradename Review
`The tradename 2-\ is acceptable.
`
`but}
`
`

`

`Phase 4 Studies
`No Phase 4 study commitments are requested at this time. Studies in pediatric patients will be waived for
`patients aged <‘years and deferred for patients aged—‘16 years.
`
`b“)
`
`Julie Beitz, MD
`Director,
`Office of Drug Evaluation III
`CDER, FDA
`
`“Us“
`
`

`

`This is a representation of an electronic record that was signed electronically and
`this page is the manifestation of the electrOnic signature.
`
`Julie Beitz
`
`1/25/2007 11:07:10 AM
`DIRECTOR
`
`

`

`MEMORANDUM
`
`DEPARTMENT OF HEALTH AND HUMAN SERVICES
`.
`PUBLIC HEALTH SERVICE
`FOOD AND DRUG ADMINISTRATION
`CENTER FOR DRUG EVALUATION AND RESEARCH
`
`DATE:
`
`January 22, 2007
`
`FROM:
`
`Julie Beitz, MD
`
`SUBJECT:
`
`Office Director Memo
`
`TO:
`
`i“(fesoterodine fumarate) extended release tablets;
`NDA 22-030
`Shwarz Pharma, LLC
`
`film:
`Fesoterodine fumarate is a muscarinic receptor antagonist that inhibits detrusor contractions and enhances
`bladder capacity. The proposed starting dose is 4 mg administered orally once daily; based on individual
`response and tolerability, the daily dose may be increased to 8 mg. This memo documents my concurrence
`with the Division of Reproductive and Urologic Product’s (DRUP’s) decision that fesoterodine fumarate
`for the treatment of adults with overactive bladder with symptoms of urge urinary incontinence, urgency,
`and urinary frequency is approvable. The sponsor’s API manufacturing facility in Shannon, Ireland, was to
`be ready for FDA’s pre-approval inspection during the final 4 months of this review cycle. Due to
`decisions made by the sponsor to \ww—m—M
`Schwarz Pharma informed DRUP on May 16, 2006 that it would not be ready for inspection during this
`review cycle. Before fesoterodine fumarate may be approved, a Satisfactory inspection of this facility will
`be needed.
`
`l1(4)
`
`Efficacy
`NDA 22—030 was received on March- 27, 2006. The sponsor's claim for efficacy is supported by two 12-
`week randomized, placebo-controlled phase 3 studies that enrolled adult patients with overactive bladder
`(with a combined total of 5 54 patients on fesoterodine fumarate 4 mg daily, 566 on fesoterodine fumarate 8
`mg daily, and 554 on placebo). Patients were required to have overactive bladder symptoms for at least 6
`months, including urinary urgency, and urinary frequency, and in most patients, urge urinary incontinence.
`Both studies showed a statistically significant reduction from baseline to endpoint (Week 12) in the number
`of urge urinary incontinence episodes per 24 hours for the 4 and 8 mg fesoterodine fumarate doses
`compared to placebo treatment. Both studies also showed a statistically significant reduction fiom baseline
`to endpoint (Week 12) in the number of micturitions per 24 hours for the 4 and 8 mg fesoterodine fumarate
`doses compared to placebo treatment. Improvement in the number of incontinence episodes (-
`was seen as early as 2 weeks following the start of fesoterodine fumarate treatment, although
`the improvements at Week 2 for the 4 mg (starting) dose were statistically better than placebo for the urge
`incontinence endpoint only.
`
`For the key secondary endpoint of voided volume per micturition, one study showed significant increases
`for both daily doses of fesoterodine fumarate compared to placebo treatment. In the second study,
`significant improvement in voided volume was observed for the 8 mg daily dose but not for the 4 mg daily
`dose.
`
`Safefl
`A total of 2288 patients with overactive bladder received fesoterodine fumarate in phase 2 and 3 clinical
`studies. Of these, 782, 785, and 222 patients received fesoterodine fumarate doses of 4 mg, 8 mg, or 12 mg
`respectively, in Phase 2 and 3 trials with treatment periods of 8 to 12 weeks. Most of these patients
`(approximately 80% in each group) received > 10 weeks of exposure. Longer exposures to fesoterodine
`fiimarate were observed in 858 patients for Z 6 months and in 570 patients for Z 12 months. The safety
`profile for fesoterodine fumarate derived from these 8- and 12-week, randomized, placebo-controlled
`studies and their 1-year open-label extension periods in patients with overactive bladder is consistent with
`
`

`

`that of other anti-muscarinics; in fact, fesoterodine fumarate produces the same active metabolite as does
`tolterodine, a drug already marketed for this use. The most common adverse events reported were dry
`mouth, constipation and urinary retention. These events were generally mild to moderate in intensity. The
`incidence of dry mouth was highest in those taking 8 mg daily (3 5%) as compared to 4 mg daily (19%) or
`placebo treatment (7%). As with other anti-muscarinics, use of fesoterodine fumarate is contraindicated in
`patients with urinary retention, gastric retention, or uncontrolled narrow-angle glaucoma.
`
`A randomized, double-blind, placebo- and positive-controlled study designed to evaluate the risk of QT
`prolongation at therapeutic and supratherapeutic doses of fesoterodine fumarate (4 and 24 mg daily doses)
`did not demonstrate abnormalities in cardiac repolarization related to fesoterodine fumarate use.
`
`No evidence of drug-related carcinogenicity was found in 24-month studies of oral administration to mice
`and rats. Fesoterodine fumarate was not mutagenic or genotoxic in vitro or in viva.
`
`Fesoterodine fumarate had no effects on fertility and produced no frank teratogenic effects in mice given
`15 mg/kg/day, a dose which produced exposures about equal to the expected clinical exposure. At a dose
`of 75 mg/kg/day (producing exposures 6-30 times the expected clinical exposure of an 8 mg daily dose),
`female mice exposed during the period of organogenesis demonstrated an increased incidence of pre- and
`post-implantation fetal loss, including resorptions; fetuses with clefi palate were also noted. These findings
`are sometimes observed with maternal stress and have been seen with other anti-muscarinics. In a
`
`multigenerational study in femalemice exposed from implantation through weaning, there were no
`occurrences of cleft palate. Following oral administration to female rabbits, post-implantation loss, early
`resorptions, and incomplete ossification of the stemebrae (retardation of bone development) were also
`noted. There have been no adequate and well-controlled studies of fesoterodine fumarate conducted in
`pregnant women. A search of FDA’s postmarketing adverse event reporting system (AERS) database
`revealed no cases of human teratogenicity in association with use of a similar product, tolterodine. DRUP
`has proposed, and I concur, with a Pregnancy Category C, i.e., that this product should be used during
`pregnancy only if the potential benefit justifies the potential risk to the fetus.
`
`Pharmacokinetic Considerations
`
`Following oral, administration, fesoterodine fumarate is rapidly and extensively hydrolyzed to its active
`metabolite. The active metabolite is further metabolized in the liver with involvement of CYP2D6 and
`
`CYP3A4, with the CYP2D6 pathway being the major pathway. Exposure to the active metabolite is
`doubled in poor metabolizers of CYP2D6 as compared to extensive metabolizers. Poor metabolizers of
`CYP2D6 were also more likely than extensive metabolizers to experience anti-muscarinic adverse events
`(e.g., dry mouth), particularly at the 8 mg daily dose, however, no dosing adjustments are recommended
`based solely on intrinsic CYP2D6 metabolizer status. Co-administration of ketoconazole, a potent inhibitor
`of CYP3A4, with fesoterodine fumarate also increased exposure to the active metabolite two-fold; dry
`mouth occurred in up to 43% of patients co-administered fesoterodine fumarate 8 mg daily and CYP3A4
`inhibitors. Exposure was calculated as being approximately 4- to 5-fold higher in CYP2D6 poor
`metabolizers taking ketoconazole as compared to CYP2D6 extensive metabolizers not taking ketoconazole.
`Therefore, doses of fesoterodine fumarate greater than 4 mg are not recommended in patients taking potent
`CYP3A4 inhibitors.
`
`In patients with moderate (Child-Pugh B) hepatic impairment, Cmax and AUC of the active metabolite are
`1.4 and 2.1-fold higher compared to healthy subjects. No dose adjustment is recommended for patients
`with mild to moderate hepatic insufficiency. Patients with severe hepatic impairment have not been
`studied; fesoterodine fumarate is not recommended for use in these patients. ‘
`
`In patients with mild to moderate renal insufficiency (creatinine clearance of 30—80 mL/min), Cmax and
`AUC of the active metabolite are 1.5 and 1.8-fold higher compared to healthy subjects. In patients with
`severe renal insufficiency (creatinine clearance of < 30 mL/min), Cmax and AUC are 2.0 and 23-fold higher
`compared to healthy subjects. No dose adjustment is recommended for patients with mild or moderate
`renal insufficiency. Doses of fesoterodine fumarate greater than 4 mg are not recommended in patients
`with severe renal insufficiency.
`
`

`

`Tradename Review
`
`The tradename a”... is acceptable.
`
`M4)
`
`Phase 4 Studies
`
`.
`
`No Phase 4 study commitments are requested at this time. Studies in pediatric patients will be waived for
`patients aged ‘Nyears and deferred for patients aged\\-16 years.
`
`Julie Beitz, MD
`Director,
`Office of Drug Evaluation 111
`CDER, FDA
`
`

`

`This is a representation of an electronic record that was signed electronically and
`this page is the manifestation of the electronic signature.
`
`Julie Beitz
`
`1/22/2007 03:47:54 PM
`DIRECTOR
`'
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket