throbber
Clin Pharmacokinet
`DOI 10.1007/s40262-016-0465-5
`
`R E V I E W A R T I C L E
`
`Pharmacokinetics and Pharmacodynamics of Lurasidone
`Hydrochloride, a Second-Generation Antipsychotic: A Systematic
`Review of the Published Literature
`
`William M. Greenberg1,2
`
`• Leslie Citrome3
`
`Ó Springer International Publishing Switzerland 2016
`
`Abstract Lurasidone
`benzisothiazol
`a
`hydrochloride,
`derivative, is a second-generation (atypical) antipsychotic
`agent that has received regulatory approval for the treat-
`ment of schizophrenia in the US, Canada,
`the EU,
`Switzerland, and Australia, and also for bipolar depression
`in the US and Canada. In addition to its principal antago-
`nist activity at dopamine D2 and serotonin 5-HT2A recep-
`tors, lurasidone has distinctive 5-HT7 antagonistic activity,
`and displays partial agonism at 5-HT1A receptors, as well
`as modest antagonism at noradrenergic a2A and a2C
`receptors. Lurasidone is devoid of antihistaminic and
`anticholinergic activities. It
`is administered once daily
`within the range of 40–160 mg/day for schizophrenia and
`20–120 mg/day for bipolar depression, and its pharma-
`cokinetic profile requires administration with food. In adult
`healthy subjects and patients, a 40 mg dose results in peak
`plasma concentrations in 1–3 h, a mean elimination half-
`life of 18 h (mostly eliminated in the feces), and apparent
`volume of distribution of 6173 L; it is approximately 99 %
`bound to serum plasma proteins. Lurasidone’s pharma-
`cokinetics are approximately dose proportional in healthy
`adults and clinical populations within the approved dosing
`range, and this was also found in a clinical study of chil-
`dren and adolescents. Lurasidone is principally metabo-
`lized by cytochrome P450 (CYP) 3A4 with minor
`metabolites and should not be coadministered with strong
`
`CYP3A4 inducers or inhibitors. Lurasidone does not sig-
`nificantly inhibit or induce CYP450 hepatic enzymes.
`
`Key Points
`
`Lurasidone, a second-generation antipsychotic
`approved for the treatment of patients with
`schizophrenia or bipolar depression, is a potent
`antagonist at dopamine D2, serotonin 5-HT2A and
`5-HT7 receptors, and partial agonist at 5-HT1A
`receptors, but is devoid of antihistaminic or
`anticholinergic activities.
`
`Lurasidone has limited bioavailability after oral
`ingestion, and should be taken with at least 350 kcal
`of food.
`
`Lurasidone does not significantly affect the
`metabolism of other pharmaceuticals, but is
`dependent on CYP3A4 for its metabolism and should
`not be coadministered with very strong CYP3A4
`inducers or inhibitors.
`
`& William M. Greenberg
`wmg1019@optonline.net
`
`1
`
`St. George’s University School of Medicine, True Blue,
`Grenada
`
`2 Mental Health Association of Rockland County,
`Valley Cottage, NY, USA
`
`3 New York Medical College, Valhalla, NY, USA
`
`1 Introduction
`
`Despite the availability of multiple approved antipsychotic
`medications, it remains challenging to optimize therapies for
`patients with severe and persistent mental illness, in view of
`heterogeneity in therapeutic response among individual
`patients [1], differences in expectable adverse event profiles
`among the various options [2], and differences in potential
`
`Par Pharm., Inc.
`Exhibit 1052
`Page 001
`
`

`

`drug–drug and disease–drug interactions for an individual
`patient’s current regimen [3, 4]. In general,
`in clinical
`practice, patients’ unique experiences of, and tolerances for,
`specific medications often presents the practical need for
`complex trade-offs and careful individualization of treat-
`ment regimens. Therefore, a substantial number of thera-
`peutic options that offer variability in pharmacokinetic and
`pharmacodynamic profiles is very desirable.
`
`2 Methods
`
`An initial search using the US National Library of Medi-
`cine PubMed.gov resource conducted on 16 February 2016
`using the terms ‘lurasidone AND (pharmacokinetic OR
`pharmacodynamic OR receptor)’ yielded 78 entries, and
`when updated on 9 June 2016, two additional records were
`found. Both authors reviewed the results of the search with
`regard to the relevance of the identified articles, as well as
`the articles referenced in those publications, focusing pri-
`marily on English-language studies involving human data,
`although some preclinical animal data were reviewed
`where applicable. Original data were from pharmacokinetic
`and pharmacodynamic studies conducted by, or supported
`by, the manufacturer. Additional information was obtained
`from the prescribing information and the US FDA drug
`approval package and European Medicines Agency (EMA)
`assessment report [5–7].
`
`3 Physicochemical Properties
`
`Lurasidone [(3aR,4S,7R,7aS)-2-{(1R,2R)-2-[4-(1,2-ben-
`zisothiazol-3-yl)piperazin-1-ylmethyl]cyclohexylmethyl]
`hexahydro-4,7-methano-2H-isoindole-1,3-dione hydrochlo-

`ride] (brand name Latuda
`) is a benzisothiazol derivative
`approved by the US FDA for the treatment of schizophrenia
`(recommended dose 40–160 mg/day), and for depressive
`episodes associated with bipolar I disorder (bipolar depres-
`sion) as monotherapy or adjunctive treatment with lithium or
`valproate (recommended dose 20–120 mg/day) [5, 6]. It has
`also been approved by the EMA for the treatment of
`schizophrenia in adults, and by Health Canada for the acute
`treatment of patients with schizophrenia, and for bipolar
`depression [7, 8]. Lurasidone has been generally referred to as
`an ‘atypical’ or ‘second-generation’ antipsychotic, but a
`recent proposed nomenclature recommends adopting func-
`tional classifications, and would categorize it as a dopamine
`and serotonin antagonist [9]. More specifically, lurasidone is
`principally pharmaceutically active as a D2 and 5-HT2A
`
`W. M. Greenberg, L. Citrome
`
`antagonist, characteristics that are believed to underlie
`antipsychotic efficacy, and with the latter action also helping
`limit extrapyramidal adverse effects and perhaps improve
`negative symptoms of schizophrenia [10, 11]. It is available in
`the form of immediate-release lurasidone hydrochloride
`(molecular formula C28H36N4O2S.HCl; molecular weight
`529.14) tablets in 20, 40, 60, 80 and 120 mg strengths. Inac-
`tive excipients include mannitol, pregelatinized starch,
`croscarmellose sodium, hypromellose, magnesium stearate,

`Opadry
`and carnuba wax. The 80 mg tablet also contains
`yellow ferric oxide and FD&C Blue No. 2 Aluminum Lake
`coloring [6]. It is only very slightly soluble in water.
`
`4 Pharmacokinetics in Adults
`
`4.1 Absorption
`
`Lurasidone hydrochloride is poorly soluble in water
`(0.224 mg/ml at 20 °C), and is poorly absorbed after oral
`ingestion in a healthy volunteer population; bioavailability
`is estimated to be 9–19 % when taken with a recommended
`meal of at least 350 calories [5]. The most definitive data
`on food effects derive from two open-label, randomized,
`crossover,
`bioavailability
`studies
`in
`patients
`aged
`18–65 years with schizophrenia or schizoaffective disorder
`who were clinically stable, had a body mass index of
`19.5–37.0 kg/m2, and were not taking medications known
`to be inhibitors or inducers of CYP3A4. These patients
`were administered a single daily dose of 120 mg of
`lurasidone administered with and without food, and max-
`imum concentration (Cmax) and area under the concentra-
`tion–time curve during a dosing interval (AUCs) were
`examined on the fifth day [12]. The first study of 16 indi-
`viduals compared ingesting lurasidone in a fasting state
`with taking it with meals of 100 kcal with medium fat,
`200 kcal of medium fat, and 800–1000 kcal with high fat.
`The geometric Cmax and AUCs were approximately twice
`as high for the 800–1000 kcal meal compared with the
`fasted state; bioavailability was significantly better with the
`high-calorie meal compared with the two other low-calorie
`meals. The second study, in 26 subjects, compared the
`bioavailability of lurasidone taken in the fasting state with
`that taken with meals of 350 kcal with high fat, 500 kcal
`with low fat, 500 kcal with high fat, 800–1000 kcal with
`low fat, and 800–1000 kcal with high fat. This study also
`demonstrated Cmax values that were approximately three
`times higher, and AUCs that was approximately two times
`higher, for the fed conditions of meal sizes of at least
`350 kcal compared with the fasting condition. These
`
`Par Pharm., Inc.
`Exhibit 1052
`Page 002
`
`

`

`PK/PD of Lurasidone
`
`studies did not find a differentiation in absorption between
`low- and high-fat meals, and did not demonstrate a sig-
`nificant difference in absorption for meals across the range
`of 350–1000 kcal, which led to the recommendation in
`labeling to take lurasidone with a meal of at least 350 kcal,
`without regard to the fat content [6].
`Lurasidone’s time to reach Cmax (tmax) is approximately
`1–3 h after an oral dose of 40 mg (reported as 1.5 h after a
`single dose and 3 h after multiple-dose administration),
`with steady-state concentrations achieved within 7 days of
`dosing. Lurasidone AUC and Cmax are approximately
`proportional in the range of 20–160 mg/day dose admin-
`istration [5, 6, 13]. Single- and multiple-dose statistics for
`the Cmax, AUC from time zero to 24 h (AUC24) and min-
`imum concentration (Cmin) were approximately linear [5].
`
`4.2 Distribution
`
`Lurasidone is very highly plasma protein-bound, reported
`as 99.8 %,
`independent of concentration, principally
`binding to albumin and a1-acid glycoprotein [7, 14].
`Although plasma albumin decreases with age, and a1-acid
`glycoprotein increases with age, population pharmacoki-
`netic analyses suggested that lurasidone use in the elderly
`would not incur significant safety or efficacy issues based
`on these changes [7]. The plasma protein binding of the
`two active metabolites was reported as 98.8–99.0 % [5, 7],
`and the mean fraction of lurasidone in whole blood dis-
`tributed in red blood cells is approximately 12 % [5]. The
`mean apparent volume of distribution after single-dose
`administration ranged from 4182 to 11,236 L, and between
`3220 and 4410 L after multiple-dose administration [5].
`A preclinical study of the intravenous administration of
`lurasidone in male Sprague–Dawley rats
`found that
`lurasidone was widely distributed in all nine tissues
`examined (brain, liver, kidneys, heart, spleen, lungs, small
`intestine, muscle, and adipose tissues), with tissue-to-
`plasma concentration ratios ranging from 1.04 for the brain
`to the highest values in the small intestine (3.84) and adi-
`pose tissue (9.16). Estimated brain uptake clearance sup-
`ported lurasidone’s good penetration but somewhat slow
`distribution to the brain, presumably related to its very high
`plasma-protein binding and thus very low unbound con-
`centration. This intravenous study, as well as a similar oral
`administration study with rats, supported the conclusion
`that absorption, distribution and elimination of lurasidone
`were mediated by linear processes,
`in the dose range
`examined [15].
`11C-raclopride was used to measure the dopamine D2
`receptor occupancy in the brain, in an open-label positron
`
`emission tomography (PET) study, using either 10, 20, 40,
`60 or 80 mg single oral doses of lurasidone in 20 healthy
`male volunteers, 30 min after they had a meal. D2 receptor
`occupancy levels were measured in the putamen, caudate
`nucleus and cerebellum, with similar findings in these
`areas. A dose-dependent increase in D2 occupancy levels
`up to the 60 mg dose was observed, which was associated
`with a maximal D2 receptor occupancy of 75–85 %; the
`80 mg dose was associated with slightly lower values [16].
`Although it has been posited that at least 60 % or so
`occupancy is needed for a therapeutic clinical response in
`patients with schizophrenia, and that higher occupancy
`levels are more often associated with more adverse effects
`[17, 18], it is also true that clozapine, accepted as the most
`effective antipsychotic, is associated with lower occupancy
`levels [19, 20]. A PET study using 18F-fallypride was also
`conducted in 17 patients with schizophrenia or schizoaf-
`fective disorder who were treated with a dose of lurasidone
`80, 120, or 160 mg for 1 week before the PET scans. This
`study did not find significant correlations between the dose
`group and brain D2 occupancy at tmax, despite good cor-
`relations between serum lurasidone concentrations and D2
`occupancy [21]. Although the two radioligands used in
`these studies were non-selective with regard to D2 and D3
`receptors, as lurasidone’s relative affinity for D2 compared
`with D3 receptors is in the order of 16-fold, these findings
`realistically reflect D2 occupancy values.
`In animal models, lurasidone is widely distributed, but
`particularly retained in pigmented tissues, such as the eyes.
`It is excreted in breast milk and crosses over into the pla-
`centa and fetus in Sprague–Dawley rats [5].
`
`4.3 Metabolism
`
`Metabolism is principally by CYP3A4, mostly via oxida-
`tive N-dealklyation, hydroxylation of the norbornane ring
`or cyclohexane ring, S-oxidation, reductive cleavage of the
`isothiazole ring, followed by S-methylation, or combina-
`tions of these pathways [5]. There are two active metabo-
`lites, but only one is present to a significant degree: the
`mean Cmax and AUC24 of ID-14283 were 23–26 and
`24–29 % of lurasidone,
`respectively, while the corre-
`sponding values for the other active metabolite, ID-14326,
`were 2–3 and 2–4 % (as determined in a study of lurasi-
`done administration in Japanese male and female patients
`with schizophrenia at steady state) [5]. CYP3A5 has
`polymorphisms that could affect metabolism of substrates,
`but
`lurasidone is not significantly metabolized by the
`CYP3A5 enzyme [7]. Lurasidone and its principal
`metabolite ID-14283 are reportedly not substrates for
`
`Par Pharm., Inc.
`Exhibit 1052
`Page 003
`
`

`

`P-glycoprotein (P-gp), although one study in transfected
`cells did demonstrate some potential P-gp inhibition
`[5, 22]. The major metabolites ID-20219 and ID-20220
`were described as not active, as was the very minor
`metabolite ID-11614. ID-14283, the major active metabo-
`lite, has a shorter elimination half-life compared with
`lurasidone, thus lurasidone’s pharmacodynamic activities
`are believed to be mainly due to lurasidone itself [5, 14].
`Although lurasidone AUC and Cmax were reported to be
`approximately linear in the dose range of 20–160 mg, in
`human microsome studies lurasidone was reported to be a
`modest inhibitor of CYP3A4 (50 % maximal inhibitory
`concentration [IC50], Ki = 17 lM), therefore some non-
`linear increase in exposure might conceivably be expected
`at higher doses. There was a similar finding for CYP2C19
`(Ki = 10 lM), and weaker inhibitory effects on CYP2C8,
`CYP2C9 and CYP2B6, as well as negligible effects on
`CYP1A2, CYP2D6, CYP1A2 and CYP2E1, although the
`FDA-approved product insert simply states that lurasidone
`is not a substrate of CYP1A1, CYP1A2, CYP2A6,
`CYP4A11, CYP2B6, CYP2C6, CYP2C8, CYP2C9,
`CYP2C19, CYP2D6, or CYP2E1 enzymes [5–7]. It was
`also reported that when coadministered with digoxin,
`midazolam, or an oral contraceptive containing ethinyl
`estradiol and norelgestromin, the concentrations of these
`pharmaceuticals were not significantly affected, suggesting
`that the in vitro moderate inhibition may generally not
`result in clinically significant drug interactions.
`
`4.4 Elimination
`
`Elimination is mostly gastrointestinal. As studied, 80.1 %
`of a total dose was recovered in feces and 9.2 % in urine,
`with 10.7 % unrecovered [5]. Elimination time as mea-
`sured by half-life is somewhat increased for higher doses,
`being 18 h for the 40 mg dose, 22 h for the 80 mg dose,
`and 31 h for the 120 mg dose [5]. The mean (percentage
`coefficient of variation [%CV]) apparent clearance after
`administration of a 40 mg dose was 3902 (18.0) mL/min
`[6].
`
`5 Effects of Intrinsic Factors on Lurasidone
`Pharmacokinetics
`
`5.1 Effects of Hepatic Impairment
`
`Compared with matched controls with normal hepatic
`function, inpatients with mild, moderate or severe hepatic
`
`W. M. Greenberg, L. Citrome
`
`impairment were demonstrated to have lurasidone Cmax
`increased by 26, 20 and 25 %, and AUC more significantly
`by 35–49, 66–75 % and threefold, respectively [5]. A dose
`adjustment is not recommended for patients with mild
`hepatic impairment;
`for moderate and severe hepatic
`impairment it is recommended that the dose should initially
`be 20 mg/day, with a maximum of 80 mg/day for those
`with moderate hepatic impairment and 40 mg/day for those
`with severe hepatic impairment [6].
`
`5.2 Effects of Renal Impairment
`
`Compared with matched controls with normal renal func-
`tion, patients with mild, moderate and severe renal
`impairment had lurasidone Cmax increased by 40, 92 and
`54 %, and AUC by 53, 91 and 103 %, respectively [5]. A
`dose adjustment is not recommended for patients with mild
`renal impairment; for moderate and severe renal impair-
`ment, it is recommended that the dose should initially be
`20 mg/day, with a maximum dosage of 80 mg/day [6].
`
`6 Effects of Age
`
`Although a formal study in the elderly was not conducted,
`the FDA’s analysis of lurasidone studies submitted for the
`initial product approval found a modest 21 % higher AUC
`in the elderly population (age 65–85 years), compared with
`the non-elderly adult study population. A dose adjustment
`in the elderly was not recommended based on this finding
`[5, 6].
`The pharmacokinetics of lurasidone were studied in a
`multicenter, open-label, single- and multiple-ascending-
`dose study in 105 enrolled child and adolescent patients
`who had a variety of psychiatric diagnoses (mainly atten-
`tion deficit/hyperactivity disorder with aggressive behavior
`or bipolar spectrum disorder). These patients were stratified
`into four different age groups (6–9, 10–12, 13–15 and
`16–17 years of age). Both Cmax and AUC24 for lurasidone
`and its three active metabolites (ID-14283, ID14326 and
`ID-11614) increased linearly with dose across the range of
`20–160 mg/day, and were similar to the exposure values
`found in the adult population, although exposure levels
`were somewhat higher in the 6- to 9-year-old cohort.
`Apparent oral clearance ranged from 317 to 346 L/h, and
`apparent volume of distribution at terminal phase ranged
`from 6940 to 8700 L, respectively, across the dose groups;
`these values were also similar to those reported for the
`adult population [23].
`
`Par Pharm., Inc.
`Exhibit 1052
`Page 004
`
`

`

`PK/PD of Lurasidone
`
`7 Effects of Other Intrinsic Factors
`
`The FDA review noted that their statistical exploration of
`the effects of sex and race did not demonstrate any sig-
`nificant differences in lurasidone exposures. However, the
`FDA review did note that in the four pivotal clinical trials
`of patients with schizophrenia, patients from US sites had a
`median AUC of approximately 32–44 % lower compared
`with non-US patients, which was posited to be due to the
`lower body weight of study patients enrolled in the non-US
`regions. A reanalysis requested by the EMA supported that
`Asian race was more relevant in explaining these findings
`of increased exposure, rather than decreased body weight
`[7].
`Based on the submitted studies, the FDA did not rec-
`ommend dosage adjustments based on ethnicity or sex, and
`also determined that the pharmacokinetics of lurasidone
`did not differ in healthy volunteers, compared with patients
`with schizophrenia or schizoaffective disorder [5].
`
`8 Effects of Extrinsic Factors on Lurasidone
`Pharmacokinetics
`
`Human liver microsome studies found that quinidine and
`cimetidine did not significantly affect lurasidone metabo-
`lism [7]. Intravenous administration of the CYP3A4 sub-
`strates biperiden, flunitrazepam, haloperidol and diazepam
`did not affect the protein binding of lurasidone, nor was
`lurasidone metabolism inhibited by these pharmaceuticals
`when they were studied in human liver microsomes [7].
`However, lurasidone metabolism’s principal dependence
`on CYP3A4 would imply sensitivity to strong CYP3A4
`inhibitors or inducers, and this has been found to be the
`case. Coadministration of the strong CYP3A4 inhibitor
`ketoconazole caused a 9.3-fold increase in lurasidone
`AUC, and a 6.8-fold increase in Cmax. On the other hand,
`the moderate CYP3A4 inhibitor diltiazem increased
`lurasidone AUC by 116 % and Cmax by 110 % when
`coadministered, while the strong CYP3A4 inducer rifam-
`pin decreased lurasidone AUC by 83 % and Cmax by 85 %
`when coadministered (i.e. both by sevenfold) [5]. There
`was no significant effect of lithium coadministration on
`lurasidone AUC or Cmax [22]. It is recommended that
`lurasidone not be used in a patient
`receiving strong
`CYP3A4 inducers (such as rifampin) or inhibitors (such as
`ketoconazole); these are specifically listed as contraindi-
`cations in the FDA product label [6]. An example of a
`strong CYP3A4 inducer that is approved for use in persons
`with bipolar disorder is an extended-release (XR) formu-
`lation of carbamazepine [24]. For those taking a moderate
`
`CYP3A4 inhibitor, such as diltiazem, it is recommended
`that the dose of lurasidone be reduced in half; if starting
`lurasidone, it is recommended that the starting dose be
`20 mg/day, with a maximum recommended dose of
`80 mg/day. For those taking a moderate CYP3A4 inducer,
`the dose may need to be increased [5]. Examples of a
`moderate inducer of CYP3A4 are modafinil and armoda-
`finil, the latter having been extensively studied for the
`treatment of bipolar depression, and thus the combination
`of lurasidone and armodafinil may be encountered in
`clinical practice [25].
`
`9 Effect of Lurasidone on Other Drugs
`
`In vitro studies also explored the effect of lurasidone on
`transporters. Lurasidone was
`found to not
`inhibit
`OATP1B1, OATP1B3, OCT2, OAT1, OAT3, MATE1,
`MATE2-K and BSEP; but was shown to inhibit BCRP and
`OCT1 [7]. It was noted that at the pH of the small intestine
`lurasidone had very low solubility, and would be present at
`concentrations that should not inhibit intestinal P-gp in a
`clinically significant manner [7].
`The co-administration of steady-state lurasidone was
`studied in vivo for potential interactions on the pharma-
`cokinetics of digoxin, midazolam, and the oral contraceptive
`norgestimate/ethinyl estradiol (Ortho Tri-Cyclen). Digoxin
`was studied as its elimination is affected by the transporter
`P-gp, and an in vitro assay had found that lurasidone influ-
`enced digoxin transport; digoxin is also a medication that
`might be co-administered in clinical practice, the efficacy
`and toxicity of which are sensitive to its remaining in a
`therapeutic concentration range. Midazolam and the oral
`contraceptive were studied as they are metabolized by
`CYP3A4. Lurasidone 120 mg/day at steady state, adminis-
`tered to patients, increased the digoxin Cmax and AUC24 by 9
`and 13 %, respectively, and of midazolam Cmax and AUC0-
`24 by 21 and 44 %, respectively. Lurasidone 40 mg/day was
`co-administered to healthy female volunteers in a double-
`blind, placebo-controlled crossover study of its effect on
`ethinyl estradiol and norelgestromin exposure levels; the
`resulting geometric means were well within the prespecified
`90 % confidence level of 80–125 %. Data from two double-
`blind, placebo-controlled clinical trials of flexibly-adminis-
`tered lurasidone 20–120 mg/day as an adjunctive treatment
`for bipolar depression were available to examine the
`potential effects of lurasidone on exposure data for lithium
`and valproate. The 90 % confidence intervals for Ctrough
`blood values of lithium and valproate concentrations were
`well within the prespecified range of 80–125 %, indicating
`little potential for clinically meaningful interactions [22].
`
`Par Pharm., Inc.
`Exhibit 1052
`Page 005
`
`

`

`10 Pharmacodynamics
`
`lurasidone
`The receptor-binding pharmacodynamics of
`have been explored in assay systems using rat, guinea pig
`and human recombinant cells. Most prominently, lurasi-
`done is an antagonist with high affinity for dopamine D2L
`(Ki = 0.329 or 0.994), 5-HT2A (Ki = 0.357, 0.470 or
`2.03 nM), 5-HT7 receptors (Ki = 0.495 or 2.10 nM), and
`noradrenergic a2C receptors (10.8 or 16.2 nM), a partial
`agonist (maximum effect [Emax] of 33 % of serotonin) with
`high affinity for 5-HT1A receptors (Ki = 6.38 or 6.75 nM)
`[data from Ishibashi et al. and the FDA Summary Basis of
`Decision: multiple values indicate variation depending on
`the experimental system] [5, 26]. It has somewhat lower
`binding affinity for noradrenergic a1 (Ki = 47.9 nM) and
`a2A
`receptors
`(Ki = 40.7 nM),
`and
`dopamine D3
`(Ki = 15.7 nM) and D4.4 (Ki = 29.7 nM). In vitro func-
`tional studies and animal behavioral model studies were
`consistent with these findings, and supported the functional
`activities at receptors noted above. There was low affinity
`for dopamine D1 (Ki = 262 nM), the serotonin 5-HT2C
`receptor (Ki = 415 nM), the phenylalkylamine site of the
`L-type Ca2? channel (Ki = 641 nM) and the voltage-gated
`Na? channel (Ki = 398 nM). For other receptors studied,
`binding affinities were very low (Ki [ 1000 nM): adeno-
`sine A1 and A2; benzodiazepine, cholecystokinin CCKA
`and CCKB; L-type (dihydropyridine and benzothiazepine
`sites) and N-type Ca2? channel; dopamine D4, GABAA,
`AMPA, kainate and NMDA glutamate; glycine, histamine
`H1, muscarinic M1 and M2, nicotine, noradrenaline ß, ß1
`and ß2; opiate, 5-HT3 and 5-HT4; sigma, the dopamine and
`serotonin reuptake sites; and K? channels (KA, KATP, KV)
`[5, 14, 26–28]. No study regarding binding affinity to the
`noradrenaline reuptake receptor was identified. Binding
`site affinities for the active metabolites ID-14283 and ID-
`14326 are similar to those of lurasidone.
`Lurasidone inhibited the human ether-a-go-go-related
`gene (hERG) channel current with an IC50 of 108 nM, and
`caused a significant QTc prolongation at dosing associ-
`ated with a Cmax of 3 lg/ml in conscious dogs, but not in
`the cat or guinea pig [5]. A thorough QT study in patients
`with schizophrenia or schizoaffective disorder was con-
`ducted, employing a randomized, double-blind, double-
`dummy design, with three treatment condition arms of
`lurasidone 120 mg/day, a
`supratherapeutic dose of
`lurasidone 600 mg/day, and a positive control arm using
`ziprasidone 160 mg/day, administered over 11 days to
`ensure steady-state conditions. Using delta individual-
`specific corrected QT interval
`(QTcI) changes from
`baseline as the principal outcome, as per the International
`
`W. M. Greenberg, L. Citrome
`
`Conference on Harmonisation (ICH) E14 guideline, the
`result was inconclusive as the maximum upper bound of
`the 90 % confidence interval exceeded 10 ms at the 2 and
`3 h post-dose time points for the 120 mg/day lurasidone
`condition, as well as
`several
`time points
`for
`the
`supratherapeutic 600 mg/day dose condition; the mean
`values for those treated in the ziprasidone arm was sig-
`nificantly higher. However, for the lurasidone conditions,
`no apparent dose–response relationship was observed, and
`no patients experienced clinically significant QTcI abso-
`lute values (QTcI [ 450 ms) or changes from baseline
`(delta QTcI [ 30 ms) [5, 6, 29, 30].
`
`11 Discussion: Distinguishing Features
`of Pharmacodynamic Profile
`
`As previously noted, lurasidone is a D2 and 5-HT2A antag-
`onist, shared characteristics of newer generation antipsy-
`chotics that effectuate antipsychotic efficacy, with the
`5-HT2A antagonism theoretically helping limit D2-antago-
`nist-induced extrapyramidal adverse effects (e.g. parkinso-
`nian muscle rigidity and tremor, dystonic reactions,
`akathisia) and prolactin elevation (associated with amenor-
`rhea, galactorrhea, sexual dysfunction), and perhaps improve
`negative symptoms (infra) of schizophrenia [31, 32].
`A relatively distinctive aspect of the pharmacodynamic
`profile of lurasidone is its potent activity at the 5-HT7
`receptor, which was of particular interest in developing this
`drug. Although the early therapeutic focus of attention in
`schizophrenia was targeting the so-called ‘positive symp-
`toms’ (e.g. delusions, hallucinations, disorganization in
`thought and speech), and later, in the 1990s, the ‘negative
`symptoms’ (e.g.
`lack of expressed emotions in facial
`expressions and tone of voice, lack of experienced plea-
`surable emotions, paucity of speech, and lack of motivation
`and volition), it became apparent that perhaps the greatest
`determinant limiting the work and social functioning for
`many were the cognitive deficits usually associated with
`this disorder (e.g. deficits in executive functioning, prob-
`lems maintaining focus or attention, deficits in working
`memory, declarative memory, processing speed, etc.),
`which persisted even in times of good clinical response or
`remission [33–38]. Although there is some evidence of the
`newer generation atypical antipsychotic medications being
`somewhat more beneficial in treating these cognitive defi-
`cits (e.g. risperidone, olanzapine, clozapine), it has been
`very challenging to demonstrate these as consistent and
`robust findings [39–43]. That functional activity studies
`established lurasidone as a very potent 5-HT7 antagonist
`
`Par Pharm., Inc.
`Exhibit 1052
`Page 006
`
`

`

`PK/PD of Lurasidone
`
`was of great interest because this mechanism of action is
`thought to have procognitive effects. These would pre-
`sumably be mediated by the inhibition of 5-HT7 receptors
`indirectly enhancing the glutamatergic output of prefrontal
`cortical pyramidal neurons, and perhaps similar actions via
`5-HT7 receptors in the hippocampus and dorsal raphe,
`although several animal models have also suggested that
`intact 5-HT7 activity is necessary for some cognitive tasks
`[26, 44–49]. Multiple animal studies have supported
`lurasidone’s value in improving cognition in specific con-
`texts. Moreover, 5-HT7 antagonism has been touted for
`possible antidepressant value, demonstrated in several
`animal models, as well as in stabilizing circadian rhythm
`synchronization [50–53].
`Lurasidone also has relatively strong binding affinity for
`the 5-HT1A receptor, at which it is a partial agonist. 5-HT1A
`partial agonism has been believed to have anxiolytic and
`antidepressant value, effected by actions at autoreceptors in
`the raphe nuclei and heteroreceptors elsewhere in cortical
`and limbic circuitry [54]. Buspirone is an azapirone
`5-HT1A partial agonist approved by the FDA to treat anx-
`iety disorders; other antidepressants such as trazodone and
`vilazodone, as well as antipsychotics such as aripiprazole,
`brexpiprazole and ziprasidone, also have 5-HT1A partial
`agonist activity, which may contribute to their therapeutic
`actions, although definitive demonstration of potential
`benefits in terms of superior efficacy, faster onset of action
`or decreased adverse sexual effects have not been estab-
`lished [55–57]. Buspirone has also been used as an
`adjunctive medication for treatment-resistant depression,
`and vilazodone has been posited to cause fewer serotonin
`reuptake inhibitor-related sexual adverse reactions because
`of its 5-HT1A partial agonism [58, 59]. Other psychotropic
`medications have different 5-HT1A activities: the antide-
`pressant vortioxetine is a 5-HT1A full agonist, and the
`antipsychotic risperidone is a 5-HT1A antagonist [60]. A
`principal problem in evaluating the contributions of
`available 5-HT1A pharmaceutical actions in clinical popu-
`lations is that most of these medications have various
`activities
`at
`alternative
`serotonergic,
`noradrenergic,
`dopaminergic, and other receptors and transporters; even
`buspirone, routinely referred to as a 5-HT1A partial agonist,
`has antidopaminergic and other serotonin receptor actions.
`Recently, gepirone, an antidepressant that acts solely as a
`full agonist at the presynaptic 5-HT1A autoreceptor and a
`partial agonist at the post-synaptic 5-HT1A receptor, has
`been the subject of a regulatory re-review [61]. Addition-
`ally, complex interactions have been demonstrated between
`5-HT1A and 5-HT7 receptor activities with regard to cog-
`nitive functioning and emotional memory, which may also
`be differentially influenced by whether the relevant sero-
`tonin release is tonic or phasic [62].
`
`Relevant to the current theory that the cognitive deficits
`in schizophrenia are related to hypofunction of prefrontal
`cortical dopaminergic and glutamatergic neurons, lurasi-
`done has been demonstrated to reverse the effects of
`administration of NMDA receptor antagonists, such as
`phencyclidine and MK-801, in animals, and to enhance
`NMDA receptor-mediated synaptic responses [63, 64].
`This has been hypothesized to be related to lurasidone’s
`5-HT7 receptor antagonism, but studies of cognition using
`the object retrieval detour task in marmosets were also
`interpreted as suggesting that lurasidone’s relatively weak
`D4 receptor binding affinity played a positive role in cog-
`nitive improvement in this model [65].
`Lurasidone has significant binding affinity and antago-
`nism at the noradrenergic a2c receptor, and this receptor
`has recently become of interest with regard to the treatment
`of patients with schizophrenia, with suggestions from ani-
`mal studies that noradrenergic a2c receptor blockade may
`have beneficial effects on cognit

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