throbber
CENTER FOR DRUG EVALUATION AND
`
`0 RESEARCH
`
`APPLICATIUN NUMBER.-
`
`22-192
`
`PHARMACOLOGY REVIEW
`
`Vanda Exhibit 2031 - Page 1
`
`VNDA 02700028
`
`Vanda Exhibit 2031 - Page 1
`
`

`
` ES
`PUBLIC HEALTH SERVICE
`FOOD AND DRUG ADMINISTRATION
`CENTER FOR DRUG EVALUATION AND RESEARCH
`
`.
`
`PHARMACOLOGY/TOXICOLOGY REVIEW AND EVALUATION
`
`NDA NUMBER! SERIAL NUMBER:
`
`22-192 / N-0000
`
`DATE RECEIVED BY CENTER:
`
`9/27/2007
`
`PRODU_CT:
`
`Iloperidone
`
`SPONSOR:
`
`DOCUMENTS REVIEWED:
`
`Vanda Pharmaceuticals
`
`REVIEW :IVISIOII 0 syc Iatry Drug Products (ITFD-130)
`
`PHARMII‘OX REVIEWER:
`Sonia Tabacova, Ph.D.
`
`PHARM/TOX SUPERVISOR:
`DIVISION DIRECTOR:
`PROJECT MANAGER:
`
`I
`
`Barry Rosloff, Ph.D.
`Thomas Laughren, M.D.
`Kimberly Updegraff, R.Ph.
`
`Date oi revlew submrssxon to DIVISIOH File Syétem (DFS): 673U7ZOU8
`
`Vanda Exhibit 2031 - Page 2
`
`VNDA 02700029
`
`Vanda Exhibit 2031 - Page 2
`
`

`
` Nfl
`
`EXECULPPVES ....
`
`..........................................._............................... .. 3
`
`2.6 PHARMACOLOGY/TOXICOLOGY REVIEW .......
`
`11
`
`........................................................................ .. 13
`2.6.2 PHARMACOLOGY ..................._......
`' 2.6.2.1
`Brief summary .................................................................................................................... .. 13
`2.6.2.2
`Primary pharmacoclynamics .............................................................................. ..
`15
`2523
`Secondary pharmacodynamics
`26
`2.624 ......................... ..
`28
`2.6.2.5
`Pharmacodynamic drug interactions ...........................
`................................................... 36
`
`
`
`40
`
`44
`
`66
`
`2.6.3 PHARMACOLOGY TABULATED SUMMARY...............................................
`
`.......
`2.6.4 .......
`2.6.4.1
`Brief summary ........................................................................................... .. 44
`2.6.4.2
`Methods ofAnalysis
`........ .. 46
`2.6.4.3
`Absorption .. . . . . . .. . .. . .. . . . . .. .. . . . . .. . . .. .. .. .. . . . . . . .. . . .
`. .. ..
`. 47
`2.6.4.4
`Distribution ... . .... . . . . .... . . .. . . . . . .. ..... ... . .... . . . . .. .. .. . .
`. .. . . .. . .. 51
`2.6.71.5
`Metabolism ......
`54
`2.6.4.6
`Excretion........................................ ..
`62
`2.6.4.7
`Pharmacokinetic drug interactions...
`................................. .. 64
`2.6.4.8
`Other Pharmacokinetic Studies.. .. . .
`.. . . .. . . . . . . .. . . . . . . .. . ... .. . . . .... 64
`2.6.4.9
`Discussion and Conclusions ................................................. ..
`64
`2.1S.4.l0
`Taliles and’figures to include comparative TK summary ............................................... .. 66
`
`
`
`2.6.5 PHARMACOKINETICS TABULATED
`
`..............................
`2.6.6 TOXICOLOGY.............................
`2.6.6.1
`Overall toxicology summary ................................................................. .. 67
`
`2.6.6.2
`Single-dose toxicity ................ ..
`75
`Repeat-dose toxicity . .. ... . . .. .. . . . . .. . . . . .. .. . .
`. . . . . .. 77
`2.6.6.3
`2.6.6.4
` mmWm..................................................................................... H3
`2.6.6.5
`Carcinogenicity...................................................................................................... .............J36
`2.6.6.6
`Reproductive and developmental toxicology.
`............... 167
`2.6.6.7
`Local tolerance .... . ... . . ....... . .. .. . . .. . .. . ... . . . . ... .... . .
`. . ... 213
`2.6.6.8
`Special toxicology studies ........................................................................................ 216
`
`
`
`2.6.6.10
`
`Tables and Figures .......................................................................................................... 217
`
`2.6.7 TOXICOLOGY TABULATED SUMMARY
`
`........
`
`217
`
`
`
`Vanda Exhibit 2031 - Page 3
`
`VNDA 02700030
`
`Vanda Exhibit 2031 - Page 3
`
`

`
`EXECUTIVE SUMMARY
`
`1.
`
`Recommendations
`
`A. Recommendation on approvability: Approvable
`
`B. Recommendation for nonclinical studies: Adequate
`
`C.
`
`H.
`
`Summary of nonclinical findings
`
`A. Brief overview of nonclinical findings
`Pharmacology: Iloperidone has high affinity for serotonin 5-HT2A, adrenergic (ll,
`adrenergic (12, .D2, D3, and 5-HTlA receptors in humans, and acts as an antagonist at
`selected dopaminergic, serotonergic, and noradrenergic receptor subtypes. Affinity was
`highest for 5-HT2 and adrenergic 0.] receptors, and lower for dopamine D2, which is a
`profile of an atypical antipsychotic. Iloperidone metabolites P88 and P89 have a profile
`similar to that of iloperidone in receptor-binding studies, with potential to exert CNS
`effects mediated by dopaminergic, serotonergic, and noradrenergic antagonism. P95
`exhibits a similar affinity to iloperidone for human 5-HT2A and adrenergic receptor
` mmmmmm,m,mm»
` .P%bb$Hkdym$mC wm,m
`shown by whole—body autoradiography,
`it apparently does not cross the blood-brain
`barrier. The high affinities of iloperidone and its metabolites for 411- adrenergic receptors
`m wwmmfi$u%m&%mmm P%,P%,md
`no
`I
`Illl
`.‘_.‘
`|‘.‘u
`cg.
`.
`-5-
`_ n.
`--gu
`
`In vitro evaluation of iloperidone effects in isolated dog Purkinje fibers and in
`mammalian cells expressing the cloned hERG showed that iloperidone has the capacity to
`prolong action potential duration and to block hERG currents;
`this indicates that
`iloperidone has the capacity to prolong QTc interval. Iloperidone metabolite PS8, but not
`P95, also exhibited this potential. In hemodynamic evaluations conducted in rats and
`dogs, iloperidone was found to dose-dependently decrease blood pressure and to induce
`transient increases in heart rate; however, cardiac output and ECG par_ameters_ were not
`affected. Neither iloperidone nor its metabolite P95 was associated with any adverse
`respiratory effects as evaluated in rats.
`’
`Pharmacokinetics:
`Iloperidone was rapidly absorbed in all animal species tested
`following oral and iv. administrations, but its bioavailability was very low due to a
` -pass effect. Oral bioavailability was <T% in rat, 5% in mouse, I9% in
`both rabbit and
`og, an
`a
`x'
`ey
`0 i
`u a
`.
`e absorption profiles of
`metabolites P88 and P95 were similar to the parent compound; their absorption was
`rapid after either oral or i.v. administration. At equal oral doses, bioavailability of P95
`P%3 (5%) in mice-
`levels genfl dose-
` (Cmax and AUC)
`proportionally in -the tested animal species, except for the rat in which exposure increased
`over—proportiona1ly possibly due to inhibitory activity of iloperidone to CYP enzymes.
`
`Vanda Exhibit 2031 - Page 4
`
`VNDA 02700031
`
`Vanda Exhibit 2031 - Page 4
`
`

`
`Gender differences in exposure were present in the rat, the mean AUC in female rats
`being significantly greater than that in males.
`‘
`Distribution of iloperidone and its metabolites after oral administration was rapid; the
`highest drug concentrations were observed in the liver, kidney, gastrointestinal system,
`and secretory glandular tissues; placental transfer was limited; and drug concentration in
`the brain was very low. P95 metabolite did not pass the Blood-brain barrier in the rat
`(whole-body autoradiography). Afier oral administration to lactating rats, iloperidone was
`excreted in milk; Cmax was attained 4 hours post dosing when iloperidone concentration
`
`
`
`metabolites in humans (P95 and P88) are found in the species used in toxicology studies.
`However, in rodents, P95 and P88 are only minor circulating metabolites, in contrast to
`'
`P
`mmms.P 88ini=oelentsanddogsislower
`gg‘
`IQI‘
`I
`' II II.l
`_u'_,_
`
`
`thanthatof
`.
`,
`,
`,_ .
`.
`_
`‘
`-=-
`-=e
`-19:1-
`
`compound. For P95, the differences between humans and animals are even greater than
`for P88. Results of pharmacology and pharmacokinetic studies that have bearing on the
`potential toxicological characteristics of metabolite P95, include the following:
`- While P95 is the predominant circulating metabolite of iloperidone in humans,
`comprising 25% to 54% of its total metabolism, in rodents it represents only 3.9%
`to 5.7% of the total measurable exposure to iloperidone and its metabolites.
`— Although P95 did not appear to cross the blood-brain barrier as assessed in the
`whole-body autoradiography study in rats, in general toxicity studies in rodents
`and dogs with direct oral administration of P95, it induced CNS clinical signs
`similar to those induced by iloperidone, which suggests that the blood-brain
`barrier is not impenetrable to P95.
`P95 is rapidlyd min in mice, 40 min
`inSmagw- a half-life of
`23-26 hours for P95 in humans. _
`
`-
`
`In vitro metabolic studies showed that iloperidone has stronger inhibitory activity to
`CYP2D6 and CYP3A4/3A5 compared with either PS8 or P95; neither iloperidone nor its
`metabolites had potential to induce cytochrome P450 enzymes.
`Excretion profiles of iloperidone, P85 and P99 were similar. They are mainly eliminated
`through the feces,
`in contrast
`to humans in which urinary excretion is the major
`elimination pathway.
`Toxicology: Repeat-dose studies of general toxicity and corresponding toxicokinetic
`parameters were conducted with iloperidone in mice,
`rats,
`rabbits, and dogs.
`Additionally, toxicology studies were performed in rats and mice with the predominant
`'
`,
`,
`r c arac erize i s safety and
`inviewoftlrelower
`'
`i
`ioeione
`P
`
`"
`
`'
`
`administration in animal species vs. humans.
`General toxicology: Among all the repeat-dose general toxicology studies on iloperidone
`-“.ae‘a~e.
`.6‘
`e
`-‘
`i‘:‘
`‘ "““ “‘i“‘i‘
`
`to safety rat study and th<.L12-month dog study%
`with iloperidone, and the 6-month rat study conducted with P95 metabolite. These studies
`are the subject of the present review.
`.
`_
`'
`
`Vanda Exhibit 2031 - Page 5
`
`VNDA 02700032
`
`Vanda Exhibit 2031 - Page 5
`
`

`
`iloperidone 6-month oral administration to rats (Sprague-Dawley) at doses of 0,
`I2, 24, and 48 mg/kg/fl induced dose-related clinical signs indicative of C'NS7lepression
`(ptosis, decreased motor activity, relaxation of the scrotum, anus, vaginal opening) and
`decrease of mean body weight at all dose levels; hematological changes (lower total
`.
` mummLD,MfimmHBmmbwa satMD
`m@);%%— mgmmmkmmmam
`doses and in MD and HD males. Prolactin was not determined. Increased incidence and
`severity of vacuolization of glandular epithelium in the mammary glands of males and
`&mmww%wwmw®mgoups, m&mfl%m
`
`bone marrw sections i
`op. rng the 5-week ecovery period, an incomplete
`reversibility was seen for decreased body weight, hematology and mammary glandular
`epithelium changes. The MTD was 12m
`(18-22% vs. control) at the next higher dose tested (24 m%gd was not
`reached in this study, as the lowest tested dose (12 mg/kg/d) induced a decrease in body
`and organ weights, hematological and clinical chemistry changes, and histopathology
`changes in the mammary glands of males and females. This dose is about 5 times the
`human dose at MRHD (24 mg/day) on an mg/m2 basis.
`Iloperidone l-year oral administration to beagle dogs at 6, 12, and 24 mg/kg/d ,
`induced drug-related clinical signs at all dosages (decreased spontaneous activity,
`tremors, bizarre behaviors, labored breathing, ptosis, slow response times and/or lack of
` mw reflex); firm and high-dose induced ataxia, loss of righting and toe pinch
`reflex (in single
`Bfily weight decreases of 7.3% and $276 vs.
`control were registered over
`the treatment period at LD and HI),
`respectively.
`Hematology and clinical chemistry changes were induced dose-dependently at MD and
`I=ED,he., ‘ ‘“'-“ ~‘- e‘-'‘‘--‘ 2-:
`- “”i‘ii
`as
`.0
`"" In
`malesandfemales;lower ‘e
`-e
`- =='
`'
`
`
`alanin aminotransferase in HD males. No abnrmalities were found in any dose group on
`ECG and auditory examination. Higher mean absolute and relative liver weights and
`hepatocellularh ,I‘1.IIl
`‘
`I;
`III oo'.n Q
`n- ‘n!!!..
`' '
`
`were found in males in the HDgrou
`MTD was 6 mg/kg/d in view of severe clinical signs and emaciation induced at and
`above the next higher dose of 12 mg/kg/d. NOAEL was not reached in this study as the
`lowest tested dose (6 mg/kg/d) induced decreased body weight and neurological clinical
`signs. This dose is 8 times the human dose at MRHD (24 mg/day) on an mg/m2 basis.
`Iloperidone metabolite P95 six-month administration to rats (Wistar) at oral doses
`of 50 and 500 mg/kg/day (yielding P95 plasma exposures of about 2 to 3x and 150 to
`400x, respectively, the human P95 plasma exposure at iloperidone MRHD of 24 mg/d),
`induced dose—dependent CNS clinical signs at both dose levels throughout the entire
`treatment period, similar to those induced by iloperidone (ptosis, decreased motor
`activity, relaxation of the scrotum, anus, vaginal opening) that are attributable to
`pharmacological effect. Body weight and weight gain decreases were induced at 1-H)
`oniy. There were no drug-reiated wmmm chemistry
` plasma leveis), or urine anaiysis. ogic
`reproductive system changes were induced in both genders. In females, dose—dependent
`cycle prolongation occurred at both LD and HD, consistent with the finding of vaginal
`
`Vanda Exhibit 2031 - Page 6
`
`VNDA 02700033
`
`Vanda Exhibit 2031 - Page 6
`
`

`
`epithelium mucilication and decreased uterine weight in the treated groups. In males,
`atrophy of testicular seminiferous tubule epithelium (in 2 animals) and an increased
`incidence of mixed cell inflammation of prostate gland with associated degenerative
`changes were found at HD.
`In both genders,
`increased cellular proliferation in the
` mw gland (ah/Tlar hyperplasia, increased secretion and dilatation of mammary
`ducts) occurred with dose-related severity at LD and HD, non-reversible after the
`recovery period. Drug—related proliferative histopathology changes, demonstrable by
`routine histology and/or immunohistochemical method (BrdU labeling) were induced in
`me adrenals in
`, thyroid in females, and pancreas in
`
`
`
`
`-
`:A- --rear
`both
`-
`:
`,
`--.4.----.-L 5.-e as - _-_’-=-
`organs (ovary, uterus,
`testes, prostate). Statistically significant, treatment—related increase in cell proliferation
`(increased proportion of cells in S phase of the cell cycle, as assessed by BrdU labeling)
`was found in
`gland (duetandalveoli) inbeth
`genders (LDM, I-IDJM, HDF), and the , HDF).
`Most of these histopathology deviations (with the exception of the adrenal, testicular and
`secondary sex organ pathology in males) were induced in a dose-dependent manner at
`both tested dose levels. An NOAEL was not reached in either male or femal
`pathomorphological proliferative changes in multiple organs/tissues were present at the
`lowest tested dose of 50 mg/kg/day, corresponding to plasma exposure (AUC 0-24)
`approximately 2 to 3x the human P95 plasma exposure at MRI-ID of 24 mg
`iloperidone/day.
`Genetic toxicology: Iloperidone was clastogenic in one in vitro test (chromosomal
`alftenaion assay in Chinese Hamster Ovary (CHO) cells). ITis likely that the positive
`results obtained in the chromosomal aberration assay in vitro are of little biological
`relevance, having in mind the negative results obtained in the in vivo micronucleus
`assays in rat hepatoeytes and mouse bone marrow. Iloperidone metabolite P95 was
`‘
`-- ‘ ‘ . ,achromosomal
`
`aberration test
`in CHO cells, and a bone marrow micronucleus test
`in rats. For
`
`iloperidone genotoxic and potentially genotoxic impurities ".1..-
`
`theacceptancecriteriaaresetatthelevelef.e— each,sethatthe
`ommH®® :-~e.gdw.
`Carcinogenicity: Two—year carcinogenicity studies of iloperidone were conducted in mice '
`and rats of both genders.
`Iloperidone administration tc -~—‘,D-l (ICR) ‘BR mice ‘at oral doses of L5, 5, and L0
`mg/kg/d (causing an increased mortality in males at HD and in females at all dose levels),
`did not exert carcinogenic effect
`in males. In females,
`the incidence of malignant
`mammary tumors was significantly increased above the concurrent and historical control
`range in the low dose group only. On an mg/m2 basis, there is no safety margin between
`the low dose employed in the study (2.5 mg/kg/day) and the maximal recommended dose
`in humans (24 mg/day). However, mammary tumor incidences were not increased in the
`mid— and high—dose groups, although the duration of treatment was the same in the mid—
`dose and low dose groups. It is not clear why similar increases in mammary tumor
`J1 seen at the higher doses employed in this study. Drugs which elevate
` y cause mammary tumors in rodents.
`Iloperidone administration to ~«~-=-- 3D(SD)BR rats at oral doses of 4, 8, and 16 mg/kg/d
`for 24 months (inducing a dose-related, significant decrease in mean body weights of
`
`b(4)
`
`Vanda Exhibit 2031 - Page 7
`
`VNDA 02700034
`
`Vanda Exhibit 2031 - Page 7
`
`

`
`over 10% in all treated groups), did not exert carcinogenic effect in male rats. In females,
`the combined incidences for pancreatic islet cell adenomas and islet cell carcinomas were
`increased at HD (2, 2, 0, 3, and 7 for the two controls, LD, MD and HD, respectively).
`The incidence value at HD was within historical control range for this species and strain;
`the dose—response trend analysis showed a p-value ofTH)051 that approachecfbut did not
`reach the level of statistical significance required for common tumors (alpha=0.003).
`Having in mind that the incidences of pancreatic islet cell tumors in this study were
`within the reported historical control range for this species and strain and that there was
`mema fimmm,
`
`elo mental toxici was assessed in a Fertility (Segment 1) study in
`;andinaPi=e—and
`"'
`" ' ,
`
`'
`
`Segment I rat fertilifl study: Iloperidone oral administration at doses of O, 4, 12,
`36 mg/kg/day to Sprague'Dawley male and female rats
`for a period starting 10 weeks
`prior to mating (males) or 2 weeks prior to mating (females) and continuing mmug
`mating, gestation and lactation,
`resulted in the following drug-related effects:
`'
`pharmacological clinical signs at all doses, significant decreases in body weight of both
`males and females at MD and HD, female estrous cycle disturbances (all doses, dose-
`dependently) and reduction in male reproductive organs’ weight
`(prostate weight
`decreased in all dosed groups; testis and epididymis weights decreased at HD), lower
`fertility indices (72% and 88% at HD and MD, respectively, vs. 100% in control), lower
`pregnancy rates at MD and HD groups (86%, and 60%, respectively, vs. 100% in
`control), reduction of corpora lutea count at HD in comparison to control; increased
`m neonatalldeaths at
`M33andI-EB.ArtI-H3,
`'
`"o raes
`(dilatation of lateral and third brain ventricles, dilatation of heart ventricles) were
`increased, but no external malformations were observed in the treated groups. There were
`1219' ::I.:.' e: :9:--2-;
`.--e--.
`e ' -nee‘-.-e.
`-
`efthe
`
`
`" 0:, In." I,.
`.1:
`-.--9'
`I -I --
`a.-.7
`and behavioral evaluations because of the low pregnancy rate and neonatal deaths).
`Reproductive performance of F1 generation was apparently not affected. The NOAEL
`was 4 mg/kg/day (1.6 times the human dose at MRHD (24 2 basis).
`Although this dose induced estrous cycle disturbances and a decrease in prostate weight,
`it did not affect parental fertility or the prenatal and postnatal survival, development and
`reproductive capacity of the progeny.
`Segment II Prenatal developmental toxicity studies in rats:
`Iloperidone administration to pregnant Wistar rats at oral doses of 0, 4, 16, and 64
`mg/kg/day during the period of major organogenesis (Gestation Days 7 through 18)
`induced developmental toxicity (expressed as embryofetal lethality, retarded intrauterine
`development and minor skeletal abnormalities) at oral doses above 16 mg/kg/day. Signs
`of maternal toxicity (reduced weight and weight gain, reduced placental weight) were
`premtfit and above T6 mg/kg/day. The NOAEL for developmental toxicity was 16
`mg/kg/day (6 times the human dose at MRHD of 24 mg/day on an mg/m2 basis).
`
`Vanda Exhibit 2031 - Page 8
`
`VNDA 02700035
`
`Vanda Exhibit 2031 - Page 8
`
`

`
`
`
`The predominant circulating iloperidone metabolite in humans (fifi) administered to
`pregnant rats at oral doses ofZ0, 80 and 200 mg/kg/day (Gestation Days 7 through T7),
`produced dose-dependent maternal pharmacological effect (signs of sedation) at all dose
`levels, but no maternal toxicity. Maternal plasma exposure (AUC) at the high dose was
` mm.mnmpbmmkUCfi P%MmflmpflWt
`
`not induce embryo/fetal mortality or congenital malformations but produced a dose-
`dependent increase in the incidence of retarded skeletal ossification vs. the concurrent
`connmatafltesteddoseleve1s,mngmgfi=om8%(LD)teM%(l=H9).Thesevalues,
`IQ.‘ ‘
`." .1: I‘: g.
`IIII
`.nI‘
`n
`u-_
`-_
`<1
`5- . ;n5
`
`ent II_Prenaa_l eve entltxi stud _in_rits:
`Se
`Iloperidone administration at oral (gavage) doses ‘of 0, 4, 10 and 25 mg/kg/day to
`‘ pregnant rabbits from gestationda Q1115) and
`decreased maternal body weight at the HD and dose-dependent dru -related clinic 1
`'
`(sedation) at all dose levels. Maternal food intake was reduced at MD and HD. The high
`dose induced increase in embryo/fetal
`intrauterine lethality and a decrease in fetal
`viability at term. No embryo/fetal toxicity or teratogenicity were observed at LD and
`MD. Based on these results, the NOAEL for embryo/fetal toxicity is 10 mg/kg/day (8x
`the human dose at MRHD of 24 mg/day on an mg/m2 basis).
`Segment III Prenatal and postnatal developmental toggicity -study in rats
`Iloperidone oral administration to pregnant CD rats from gestation day 17 through
` posmatal day 217 at doses of 4, T6 and 48736 mg/kg/day, caused matema]
`toxicitym mortality at HD and dose-
`dependent decrease in maternal body weight at HD, MD and LD), significantly prolonged
`.
`gestation and parturition, high perinatal- and postnatal mortality (stillbirths and neonatal
`
`deaths)inFlgeneratien' atHDandMD,andsememerease'in '
`
`‘
`
`was impaired at MD and HD, as demonstrated by the reduced pup weight at birth and
`weight gain through weaning. However, there was no apparent adverse effect on F1
`16
`I
`.
`1
`1.
`1
`I
`.
`I
`.
`1
`1
`.
`.
`of
`
`the administered dose levels. The NOAELwa (L5 times the human dose at
`MRHD of 24 mg/day on an mg/m2 basis).
`B. Pharmacologic activity
`The pharmacological profile of iloperidone is consistent with that of an atypical
`antipsychotic with a reduced potential for extrapyramidal side effects and therapeutic
`potential with regard to positive, negative, and social withdrawal symptoms of
`schizophrenia. Iloperidone has the potential to induce hypotensive effects and to prolong
`QTc interval duration
`
`C. Nonclinical safety ISSUCS relevant to clinical use
`
`— The high affinity for 0L1- adrenergic receptors in peripheral vascular tissues,
`mmm
` .
`Iloperidone prolongs action potential duration and block hERG currents in vitro;
`indicating a capacity to prolong QTc interval.
`In vivo,
`iloperidone dose-
`
`—
`
`Vanda Exhibit 2031 - Page 9
`
`VNDA 02700036
`
`Vanda Exhibit 2031 - Page 9
`
`

`
`dependently decreases blood pressure and induces transient increases in heart rate
`in rats and dogs; however, ECG parameters were not affected.
`the
`Iloperidone has the potential
`to inhibit CYP2D6 and CYP3A4/5 at
`recommended therapeutic dose in humans (12 mg BID); its metabolites P88 and
`
`and of iloperidone P95 metabolite to rats (6 months) induced general toxicity in
`all tested species. An NOAEL was not reached in either of these studies. The
`mwmm@$& ,wm5
`times
`on an mglm2 basis) induced a
`decrease in body and organ weights, hematological and clinical chemistry
`changes, and histopathology changes in the mammary glands of males and
`females. The lowest tested dose
`8 times the human dose at MRHD on an mg/m2 basis body
`weight and neurological clinical signs. The lowest tested dose of P95 in the 6-
`month rat study (50 mg/kg/day, corresponding to plasma exposure (AUC 0-24)
`approximately 2 to 3x the human exposure at MRHD) induced proliferative
`pathomorphological changes in multiple organs/tissues,
`i.e., endocrine glands
`(pituitary, thyroid, and pancreas), mammary gland (both genders) and ovary.
`Iloperidone was clastogenic in one in vitro test (chromosomal aberration assay in
`Chinese Hamster Ovary (CHO) cells) but was not clastogenic in the in vivo
`m’c onuc eus assays in rat hepatocytes and mouse bone marrow. The positive
`results wm afiterraion assay in CHO cells in vitro are of
`little biological relevance, having in mind the negative results obtained in vivo.
`Iloperidone
`genotoxic
`and potentially genotoxic
`impurities ._———.\
`
`
`pp sot
`ug/day.
`
`(female) administered oral doses of 2.5, 5 and 11) mglkglday for 2 years the
`incidence of malignant mammary tumors was significantly increased above the
`concurrent and historical control range in the low dose group only. On an mg/m2
`basis, there is no safety margin between the low dose employed in the study (2.5
`mg/kg/day) and the maximal
`recommended dose in humans (24 mg/day).
`However, mammary tumor incidences were not increased in the mid— and high-
`dose groups, although the duration of treatment was the same. It is not clear why
`similar increases in mammary tumor incidences were not seen at higher doses.
`In view ofthe proliferative effects seen with iloperidone metabolite P95 in the 6-
`month rat study, the Division required a 2-year carcinogenicity study with P95 in
`he rat which is ongoing (CAC meeting of March 25, 2008).
`lloperidone induces decreased fertility, prolonged gestation, increased prenatal
`and mmmd growth of the progeny upon oral
` wmmmmmmmammmm o
`mating (males) or 2 weeks prior to mating (females) and continuing through
`
`13(4)
`
`Vanda Exhibit 2031 - Page 10
`
`VNDA 02700037
`
`Vanda Exhibit 2031 - Page 10
`
`

`
`gestation, parturition and lactation. The NOAEL is 4 mg/kg/day (1.6 times the
`huvlose at MRHD (24 mg/day) on an mg/m2 basis).
`Iloperidone oral administration to pregnant rats and rabbits during the period of
`major organogenesis induces developmental
`toxicity (embryofetal
`lethality in
`
`'
`
`'
`
`'
`
`'
`
`m$em8a%mmfim
`toxicity is 16 mg/kg/day in rats and 10 mg/kg/day in rabbits (6- and 8 times,
`respectively, the human dose at MRI-ID of 24 mg/day on an mg/m2 basis).
`
`.-. 2..
`---—e-
`
`--
`..
`-
`-
`
`
`gestation and parturition, incresdincdence o sillirths, neonatal mortality, d
`retarded growth of progeny up to weaning, but did not affect neurobehavioral and
`"reproductive development of the survivingpu 's.'The'
`_
`(L6
`times the human dose at MRHD (24 mg/day) on anmg/m2’ basis). ''
`
`
`
`'
`
`APPEARS THIS WAY ON ORIGINFL
`
`l0
`
`Vanda Exhibit 2031 - Page 11
`
`VNDA 02700038
`
`Vanda Exhibit 2031 - Page 11
`
`

`
`2. 6 PHARMA COLOGY/TOXICOLOGY RE VIEW
`
`2.6.1
`
`INTRODUCTION AND DRUG HISTORY
`
`NDA number: 22-192
`Review number:
`1
`
`Sequence number/date/type of submission: N-0000
`Information to sponsor: No (x)
`Sponsor and7or agent: Vanda Rharmaceuticals
`Manufacturer for drug substance: Vanda Pharmaceuticals
`
`Reviewer name: Sonia Tabacova
`
`IH?‘l}#- B0
`Revievv completion date:
`
`June 2008
`
`Drug:
`
`Trade name: None provided
`Generic name: Iloperidone
`Code name: ILO522 (Novartis); ILO522—NXA (Novartis); VYV-683 (Vanda)
`C nafiie: J -[4-[3-FL (?$—Fiuor0benzo[a7isoxaz0l—3—yl)-I -
` ahanone V
`Chemical Abstract Service_(CAS) Number: 133454-47-4
`Mole file number:
`
` m weight: C24H27N2O4F F4265 ‘
`Structure:
`
`04333
`
`Relevant INDs/NDAs/DMFS: IND 36827
`
`Drug class: Antipsychotic
`Intended clinical population: Adults with schizophrenia
`Clinical formulation: Tablets
`Route of administration: oral
`
`Disclaimer: Tabular and graphical information is from sponsor’s submission unless
`stated otherwise.
`
`11
`
`Vanda Exhibit 2031 - Page 12
`
`VNDA 02700039
`
`Vanda Exhibit 2031 - Page 12
`
`

`
`Studies reviewed Wifllill this‘ submission: All submitted studies, except for the non-
`pivotal floperidone repeat—dose general toxicology studies
`
`Studies go_t reviewed within this submission: Non-pivotal
`1.]
`1.
`
`iloperidone repeat-dose
`
`INPPBNRSTFIIS WAY ON ORIGINAL
`
`12
`
`Vanda Exhibit 2031 - Page 13
`
`VNDA o27ooo4o
`
`Vanda Exhibit 2031 - Page 13
`
`

`
`2.6.2 GGY
`
`2.6.2.1 Brief summary
` mMmw has an 1%vi ro and7or ex ‘vivo binding profile consistent with an atypical
`affinity for human dopamine (D), serotonergic (5-HT), ot-
`noradrenergic, and sigma receptors, and no affinity for glycine-binding site, N-methy1—D
`aspartate (Nl\/IDA) receptor channel, or muscarinic receptors. Affinity of iloperidone was
`fbundtebehighestferhumm15-HTl74\,5-HT2A,D2,D3,adrenergica1anda2
` b%rkrm,D5,mdema h$m
`agonist activity at any receptors evaluated (rat or human), but was found to have
`significant in vitro and in vivo antagonist activity at rat and human dopaminergic,
`serotomggam m%#ewmfi%mdmfiw
`functionalassa activities on 5-
`HT2 receptors and weak effects on D2 receptors as shown in an ex vivo study in rats. The
`high 5—HT2/D2 receptor binding ratio of iloperidone suggests that this compound has the A
`potential to act as an atypical antipsychotic agent with a relatively low potential to
`produce extrapyramidal side effects. Preferential affinity for (11 over I12 receptors
`suggests a potential for iloperidone to cause orthostatic hypotension.
`Behavioral assays conducted in mice,
`rats and monkeys compared the in vivo
`pharmacological activities of iloperidone with both typical
`(thioridazine and/or
`haloperidol) and atypical (clozapine) antipsychotic agents. The results indicated that
` as potential for atypical antipsychotic action with reduced capacity for
`inducing extrapyramidal (EPS) symptoms, potential anxiolytic properties at doses similar
`to its antipsychotic properties and a potential to increase social interaction. Overall, the in
`vivo
`irnimted properties conslstent with an
`
`The major metabolites of iloperidone in humans are P88, P89 and P95. It is important to
`note that P88 and P89 cross the blood-brain barrier, while P95 does not do so detectahlga
`Therefore, P95 is less likely to exert pharmacological effects on the central nervous
`system and was primarily evaluated for the potential
`to exert peripheral effects.
`Pharmacodynamic studies have been performed with P88, P89 and P95 metabolites
`directly and their receptor-binding profiles were separately characterized. P89 was found
`to binfi with high affinity to D2 and 5—HT2 receptors, whereas P88 was approximately
`36_—fi)ld and W- er at these sites, respectively. Both P88 and P89 also exhibited
`affinity for 5-HTlA receptors and oL2- noradrenergic receptors. P95 exhibited similar
`affinity for the human 5—HT2A receptor compared with iloperidone, and also exhibited
`higher or we tested as compared
`with
`D1, DZS, DZL, and
`D3 receptor subtypes compared with iloperidone. Neither iloperidone nor P95 showed
`appreciable affinity for histamine H1 receptors.
`TmmmmmaflmwmoflksMP95wdP%
`bmad-spectmmmceptoraffinityforhumamratandguineapigreeeptorsusing
`radiolabeled ligand-binding techniques. The results showed that the P88 metabolite had
`moderate to strong affinity at the adrenergic on,
`(1213, one, dopamine D1, D2, D3, D4,
`
`13
`
`Vanda Exhibit 2031 - Page 14
`
`VNDA o27ooo41
`
`Vanda Exhibit 2031 - Page 14
`
`

`
`histamine Hi, 5-HT113, and 5-HT2c sites. It had relatively weak affinity at the adrenergic
`(1213, 5-HT1A, and 5-I:ITs sites.
`In comparison,
`the P95 metabolite showed weak to
`moderate affinity at the adrenergic cu, a2A, a213, a.2c, dopamine D1, D3, D4, and 5-HT6
`sites. Compared with the receptor profile of iloperidone,
`these metabolites were
` mmWmm%akmammwmfimmmm aseparatesmdy,
`
`both compounds exhibit dopamine receptor antagonist properties. Additional behavioral
`a$%$ P%mdP%mm%,mB,mdmmkew_
`
`
`
`in contrast to the parent compound, nither
`profile similar to iloperidone. However,
`metabolite appeared to possess anxiolytic activity, as measured in behavioral assays.
`
`In a series of safety pharmacology studies, the cardiovascular safety of iloperidone was
`evaluated in vitro and in vivo in rats and dogs. In dog Purkinje fibers paced atstimulation
`frequencies of 0.5 and 1 Hz, iloperidone and P88 at a concentration of 0.1 uM and above
`induced a prolongation in action potential duration. The highest concentration of 10 uM
`induced amd 3 Hz,
`indicating a
` %mmpmure
`to P95 at concentration of 0.01, 0.1,
`1 and 10 p.M induced a prolongation in action
`
`
`toprolong’tl=ieQ5l'.41ntewal' atfieeplasma
`concentrations of 0.1 uM and above, while P95 has the potential to prolong the QT
`interval at free plasma concentrations of over 10 uM. This extent of exposure to P95 is
`unlikely, as administration of iloperidone doses of 24 mg given once-a-day in human
`patients yielded maximal P95 steady-state plasma levels of 55.5 ng/mL, or 0.13 uM. In
`vitro effects of iloperidone and its metabolites P95 and P88 in comparison with
`risperidone and ziprasidone on the cardiac ion channel hERG showed that all test articles
`produced rapid, reversible blockade of hERG currents. The block potency rank order was
`iloperidone > ziprasidone 3 P88 > risperidone > P95. These data suggest that P95 is
`unlikely to contribute to the QT prolongation potential of iloperidone.
`
`In vivo, iloperidone was found to have hypotensive and vasodilatory effects similar to
`mmed m$mdmem%mmmd
`
`preferential affinity for 111- over (12 adrenergic receptors in vit

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