throbber
LEADING ARTICLE
`
`Drugs 2008; 68 (16): 2269-2292
`0012-6667/08/0016-2269/$53.45/0
`
` 2008 Adis Data Information BV. All rights reserved.
`
`Upcoming Agents for the Treatment
`of Schizophrenia
`Mechanism of Action, Efficacy and Tolerability
`
`Delia Bishara and David Taylor
`Pharmacy Department, Maudsley Hospital, London, UK
`
`Abstract
`
`Since the introduction of a group of atypical antipsychotics in the 1990s, there
`has been a decline in the rate of new antipsychotics being introduced into clinical
`practice. However, with increasing safety and efficacy concerns over currently
`available drugs and a dearth of options available for atypical depot formulations,
`there is a considerable need for the development of new formulations and agents.
`This review examines the profile of seven antipsychotic drugs currently in the
`premarketing stage of development and summarizes their mechanism of action,
`clinical potential and safety.
`Asenapine is an antipsychotic with activity for multiple receptors and has
`potential to improve negative and cognitive symptoms of schizophrenia. Bifepru-
`nox is a partial dopamine D2 and serotonin 5-HT1A receptor agonist showing a
`less than convincing efficacy profile, but which may offer safety advantages over
`available agents by means of a reduced risk of metabolic complications. Iloper-
`idone is a D2 and 5-HT2A receptor antagonist requiring further studies to establish
`its effectiveness. It has a high affinity for α1-adrenoceptors, which can lead to
`associated haemodynamic adverse effects. Nemonapride is essentially a typical
`antipsychotic drug, similar in structure to sulpiride, which has been available for
`some time in Japan. It has efficacy against positive symptoms and has shown
`some antidepressant and anxiolytic properties, although efficacy data for it are
`somewhat limited. Norclozapine (N-desmethylclozapine) is a major metabolite of
`clozapine formed by its demethylation. Its partial agonist activity at D2 receptors
`has raised interest in it as an antipsychotic in its own right. In addition, it appears
`to have muscarinic agonist activity, which is believed to be responsible for the
`observed positive effects it has on cognition. It was envisaged to be effective as an
`adjunct to other agents or at high doses in the treatment of refractory schizo-
`phrenia, although a recent randomized, controlled study showed that it was no
`more effective than placebo in patients with schizophrenia experiencing an acute
`psychotic episode. Olanzapine pamoate depot injection has shown comparable
`efficacy to oral olanzapine in several studies. However, it has provoked considera-
`ble safety concerns by its association with inadvertent intravascular injection
`events in numerous patients. This accidental intravascular administration of
`olanzapine pamoate leads to excessive sedation, confusion, dizziness and altered
`speech. Post-injection observation periods and postmarketing surveillance are
`planned following the introduction of the depot. Paliperidone palmitate is the
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1038, p. 001
`

`

`

`2270
`
`Bishara & Taylor
`
`palmitate ester of paliperidone, the major metabolite of risperidone, and is
`formulated as a long-acting injection for intramuscular use. Its pharmacology is
`comparable to risperidone, having D2 and 5-HT2A receptor antagonist activity.
`Efficacy studies have shown positive results, and because paliperidone has no
`antagonistic activity at cholinergic receptors, it has low potential for anticholiner-
`gic adverse effects, including cognitive dysfunction. However, with higher doses,
`the frequency of extrapyramidal side effects and orthostatic hypotension have
`been shown to be greater than with placebo.
`
`such as weight gain[7,8] and impaired glucose meta-
`Schizophrenia is a severely debilitating psychiat-
`bolism.[9] Furthermore, similar to conventional anti-
`ric disorder observed worldwide. It often results in
`lengthy hospitalizations and is a considerable bur-
`psychotics, atypical antipsychotics have not proved
`den upon medical resources.[1] Prevalence amongst
`to be effective in treatment-refractory patients. Until
`adults tends to vary between studies but is usually
`recently, clozapine, discovered shortly after chlor-
`reported to be in the range of 0.5–1.5%.[2] Since the
`promazine, remained the only drug to have demon-
`introduction of chlorpromazine in the 1950s, the
`strated clinical superiority to other agents in treat-
`number of antipsychotic drugs available has notably ment-resistant schizophrenia[10] and in suicidali-
`increased. By the 1980s, several conventional or
`ty.[11] Attempts to develop new drugs based on its
`first-generation antipsychotics had been developed
`pharmacology in an effort to mimic its superior
`and were found to be effective in treating the posi-
`efficacy have so far largely been unsuccessful.
`tive symptoms of schizophrenia, such as delusions However, a recent paper reported that higher than
`and hallucinations. However, the negative symp-
`typically prescribed doses of olanzapine may be as
`toms of the illness, (emotional withdrawal, apathy,
`effective as conventional doses of clozapine in treat-
`avolition and cognitive dysfunction) were not effec- ment-resistant schizophrenia.[12]
`tively managed by these drugs. In addition, these
`In the last decade, antipsychotic drug develop-
`antipsychotics were found to produce a high burden
`ment has remained somewhat static, at least in terms
`of extrapyramidal side effects (EPS)[3] and adverse
`of new drug introductions. This may be because of
`effects related to elevation of serum prolactin.[4]
`poorly defined pathophysiology of the disorder and
`Moreover, their tendency to cause tardive dyskine-
`incomplete understanding of the pharmacology of
`sia[5] in the longer term made their continued use
`available drugs,[13] resulting in confusion as to the
`problematic.
`ideal mode of action required. In addition, failure of
`In an effort to develop novel agents that would
`some drugs in early clinical trials and the increased
`treat both the positive and negative symptoms of
`costs of drug development may have contributed to
`schizophrenia while affording a low propensity for
`the recent dearth of new agents being launched.
`movement disorders, the pharmaceutical industry
`Since many currently available atypical antipsychot-
`developed several antipsychotics in the 1990s, re-
`ics will soon lose patent protection, there is in-
`ferred to as second-generation antipsychotics, con-
`creased pressure on the pharmaceutical industry to
`sidered to be ‘atypical’. While these drugs have
`develop novel treatments, and so a renewed interest
`probably been of some benefit to patients, full ex-
`in drug development for schizophrenia has emer-
`pectations have not been realized. Despite having a
`ged.[13]
`somewhat improved (although debatable) efficacy
`This article reviews the antipsychotic agents that
`in treating the negative symptoms of schizophrenia
`have undergone extensive clinical development for
`as well as a lower propensity to cause movement
`the treatment of schizophrenia and reached the
`disorders, these benefits have been accompanied by
`other effects, including metabolic adverse effects[6]
`premarketing stage of development, and examines
`
` 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (16)
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1038, p. 002
`

`

`

`Upcoming Agents for Schizophrenia
`
`2271
`
`their pharmacology, clinical potential and tolerabili-
`ty.
`
`1.2 Atypical Antipsychotics
`
`1. Pharmacology of Currently
`Available Antipsychotics
`
`1.1 Conventional (Typical) Antipsychotics
`
`Because of the problems encountered with con-
`ventional antipsychotics, the atypical antipsychotics
`were developed based to a large extent on the com-
`plex pharmacology of clozapine. Clozapine has af-
`finity for a diverse range of receptors including D1,
`D2 and D4 dopaminergic, α1 and α2 adrenergic, H1
`histaminergic, muscarinic and various serotonin re-
`Since the dopamine hypothesis was first pro-
`ceptor subtypes.[21-23] Its supported clinical superior-
`posed in the 1960s, it has remained the central pivot
`ity[10] and near absence of the debilitating EPS has
`around which antipsychotic agents have been devel-
`fuelled an intense effort over the last 20 years to
`oped.[14] Conventional antipsychotic drugs all show
`develop similar agents.
`at least some affinity for the dopamine D2 receptors
`In the development of atypical antipsychotics,
`and there is a strong correlation between clinical
`efficacy of the drugs and their binding affinities.[15]
`researchers have tried to mimic the pharmacological
`action of clozapine while trying to avoid its own
`Positive symptoms of schizophrenia are believed to
`serious adverse effects, such as agranulocytosis.[24]
`result from dopaminergic hyperactivity,[16] while
`Like conventional antipsychotics, atypical antipsy-
`negative symptoms have been attributed to reduced
`chotics are also antagonists at D2 receptors. How-
`functioning in the prefrontal cortex, mainly resulting
`ever, they do show an additional range of binding
`from underactivity of prefrontal dopaminergic neu-
`activity at various other receptor sites. In particular,
`rons. Therefore, these symptoms are potentially im-
`their antagonist activity at serotonin receptors was
`proved by agents that reduce serotonergic function
`thought to account for the differences between the
`(by serotonin 5-HT1A receptor agonist activity),
`two classes of agents. Atypical drugs show a higher
`thus promoting increased dopamine activity in the
`affinity for 5-HT2A receptors compared with D2
`prefrontal cortex, and by drugs that block presynap-
`tic dopamine receptors.[17] Such an effect may also
`receptors, and this ratio of affinities has been hy-
`pothesized to account for their enhanced efficacy
`improve cognitive impairment because of the result-
`and fewer EPS.[25,26] Antagonism at 5-HT2A recep-
`ing stimulation of D1 receptors.[18] These concepts
`tors leading to an increase of dopamine activity in
`are consistent with the clinical profile of conven-
`the prefrontal cortex has also been suggested to
`tional antipsychotics, which are effective in treating
`account for the beneficial effects that atypical anti-
`positive symptoms, presumably by reducing overac-
`psychotics have against negative symptoms.[26]
`tivity in the mesolimbic pathways, but offer little
`benefit to the negative symptoms or cognitive defi- However, amisulpride has no affinity for 5-HT2A
`receptors[27] but clearly has atypical antipsychotic
`cits because of inadequate or adverse effects in the
`properties,[28] suggesting that this 5-HT2A/D2 recep-
`mesocortical pathways (where dopamine activity is
`already decreased).[19]
`tor hypothesis may not hold true for all drugs or that
`other receptor systems also play an important role in
`The high rates of movement disorders caused by
`the atypicality of antipsychotics.
`conventional antipsychotics are believed to arise
`It has also been suggested that atypical anti-
`from dopamine antagonism in the nigrostriatal path-
`ways. The subsequent reduced dopamine activity
`psychotic activity may be explained by differences
`leads to a relative increase in cholinergic activity,
`in the occupancy and dissociation of antipsychotics
`from D2 receptors.[29-31] Agents showing a relatively
`and the resulting imbalance accounts for these
`troublesome adverse effects.[3] Elevation of prolac-
`low D2 receptor affinity, such as clozapine,[32,33]
`quetiapine[34] and olanzapine,[35] and fast dissocia-
`tin caused by these drugs stems from their dopamine
`antagonist effects on the tuberoinfundibular path-
`tion from the receptor, have atypical properties. This
`way.[20]
`loose D2 receptor binding may also account for the
`
` 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (16)
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1038, p. 003
`

`

`

`2272
`
`Bishara & Taylor
`
`Table I. New antipsychotic drugs
`
`Drug
`Asenapine
`Bifeprunox
`
`Iloperidone
`
`Norclozapine
`
`Nemonapride
`
`Mechanism of action
`Multiple receptors
`Partial D2 and 5-HT1A
`receptor agonist
`D2 and 5-HT2A receptor
`antagonist
`Partial D2 and muscarinic
`receptor agonist
`D2, D3 and D4 receptor
`antagonist
`D2 and 5-HT2A receptor
`antagonist
`Paliperidone palmitate D2 and 5-HT2A receptor
`antagonist
`
`Olanzapine pamoate
`
`Manufacturer
`Organon
`Lundbeck/Solvay
`
`Development status
`Phase III
`Phase III
`
`Expected launch date
`2009
`2010/2011
`
`Titan Pharmaceuticals
`
`Phase III
`
`ACADIA Pharmaceuticals
`
`Phase II
`
`Not known
`
`Not known
`
`Astellas
`
`Eli Lilly
`
`Janssen-Cilag
`
`Launched in Japan
`
`Phase III
`
`Phase III
`
`No plans for launch in
`US, UK or Europe
`Late 2008
`
`2009
`
`observed limbic selectivity observed for some drugs
`such as clozapine.[36]
`
`pathways, their propensity to cause EPS and elevat-
`ed prolactin levels may be reduced.[19]
`
`1.3 Dopamine Partial Agonists
`
`2. New Antipsychotics
`
`2.1 Asenapine
`
`Asenapine (figure 1) is a novel psychotropic
`agent being developed for the treatment of schizo-
`phrenia and bipolar disorder. Its chemical structure
`and pharmacological action are distinct from cur-
`rently available drugs.
`
`We are now approaching an exciting and chal-
`More recently, a new class of antipsychotics has
`lenging time for the development of new treatments
`been introduced, the dopamine partial agonists, of
`for schizophrenia. With continual, if somewhat
`which aripiprazole is the only one currently avail-
`slow, improvements in our understanding of the
`able in clinical practice. Aripiprazole is a potent
`pathophysiology of the disease and complex phar-
`partial agonist at D2 and 5-HT1A receptors and acts macology of the drugs, novel approaches for drug
`as an antagonist at 5-HT2A receptors.[37,38] While
`discovery are being studied. The following agents
`both typical and atypical antipsychotics act as full
`(table I) are presently in their developmental stages
`antagonists at dopamine receptors, schizophrenia as
`and are due to be introduced for clinical practice in
`outlined is thought to arise from a combination of
`the near future.
`over- and under-activity in different dopamine path-
`ways. Thus, blocking dopamine activity in all parts
`of the system may account for the problems encoun-
`tered with drug therapy already discussed. There-
`fore, in theory, the capacity for an agent to alter
`dopamine neurotransmission differently in separate
`areas of the dopaminergic system may have both
`therapeutic and safety advantages.
`Partial agonists are thought to exert their effects
`by acting effectively as dopamine antagonists in the
`mesolimbic pathway. However, in the mesocortical
`pathway, where reduced dopamine activity is
`thought to produce negative symptoms and cogni-
`tive impairment, they effectively act as dopamine
`agonists. Furthermore, because dopamine partial ag-
`onists do not produce complete dopamine activity
`blockade in the nigrostriatal and tuberoinfundibular
`
`O
`
`CI
`
`H
`
`H
`
`N
`Fig. 1. Structural formula of asenapine ((3aS,12bS)-5-chloro-2,3,3-
`a,12b-tetrahydro-2-methyl-1H-dibenz[2,3:6,7]
`oxepino[4,5-c]pyr-
`role).
`
` 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (16)
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1038, p. 004
`

`

`

`Upcoming Agents for Schizophrenia
`
`2273
`
`9.4
`
`1.8
`
`0.31
`
`0.26
`
`0.16
`
`NEM
`
`4898
`
`0.4
`
`42.8
`
`5.6
`
`93.1
`
`25
`
`7.1
`
`21.4
`
`216
`ILO
`
`>>
`
`>>
`
`>>
`
`>>
`
`>>
`
`10.0
`
`1.6
`
`0.6
`
`3.2
`
`BIF
`
`>>
`
`1.0
`
`1.2
`
`0.034
`
`0.07
`
`2.7
`
`1.1
`
`0.42
`
`1.3
`
`1.4
`ASE
`
`>1500
`
`1.9
`
`>1000
`
`440
`
`6
`
`>10000
`
`45
`
`1100
`
`2
`
`3
`
`0.7
`
`210
`HAL
`
`8
`
`7
`
`6
`
`12
`
`770
`
`9–12
`
`473
`
`125
`
`85
`CLO
`
`50
`
`11
`
`1
`
`0.4
`
`32
`
`5
`
`7
`
`high Ki, therefore no appreciable affinity for receptor.
`Ki = dissociation constant; M = muscarinic; NEM = nemonapride; OLA = olanzapine; PAL = paliperidone; QUE = quetiapine; RIS = risperidone; ZIP = ziprasidone; >> indicates very
`5-HT = serotonergic; ARI = aripiprazole; ASE = asenapine; BIF = bifeprunox; CLO = clozapine; D = dopaminergic; H = histamine; HAL = haloperidol; ILO = iloperidone;
`
`3
`
`525
`ZIP
`
`120
`
`11
`
`7
`
`615
`
`220
`
`2450
`
`1600
`
`340
`
`160
`
`455
`QUE
`
`32
`
`1.0
`
`590
`
`30
`
`6.9
`
`4.8
`
`PAL
`
`>10000
`
`20
`
`0.7
`
`25
`
`0.5
`
`7
`
`2
`
`19
`
`11
`
`4
`
`210
`
`>1000
`
`9
`
`10
`
`4
`
`430
`RIS
`
`27
`
`49
`
`11
`
`31
`OLA
`
`>>
`
`61
`
`57
`
`15
`
`3.4
`
`1.7
`
`44
`
`0.8
`
`0.34
`
`M1
`
`H1
`α1-Adrenergic
`5-HT2C
`
`5-HT2A
`
`5-HT1A
`
`D4
`
`D3
`
`D2
`
`D1
`
`ARI
`Ki (nmol/L)
`
`Receptor
`
`Table II. Antipsychotic receptor-binding profiles[39-50] (adapted from Chou,[51] with permission)
`
`Asenapine has higher affinity for a variety of
`serotonergic (5-HT2A, 5-HT2C, 5-HT6, 5-HT7), nor-
`adrenergic (α2A, α2B, α2C) and dopaminergic (D3,
`D4) receptors than D2 receptors, but minimal affini-
`ty for muscarinic receptors (see table II for compari-
`son of binding affinities).[39]
`One of the mechanisms for alleviating negative
`symptoms is believed to be the blockade of 5-HT2A
`and 5-HT2C receptors. Asenapine binds to these
`receptors 19-fold and 38-fold, respectively, higher
`than D2 receptors, suggesting a potential for improv-
`ing negative symptoms.[39] Asenapine is thought to
`maintain adequate but not excessive blockade of D2
`receptors, and so it may allow control of positive
`symptoms without the resulting EPS and elevation
`of prolactin. Similarly, activity at α-adrenergic re-
`ceptors has been suggested to improve negative and
`cognitive symptoms via α2-receptor antagonism
`and positive symptoms via α1-adrenoceptor ant-
`agonism.[52] Asenapine appears to have relatively
`high affinity for adrenergic receptors, which may
`offer potential therapeutic advantages, although
`there is no such evidence as yet. In contrast, it has
`been shown to have very low affinity for muscarinic
`and other CNS receptors. The D2 receptor affinity of
`asenapine is approximately 6000-fold greater than
`the affinity for M1 receptors, thus minimizing the
`risk of antimuscarinic adverse effects that are seen
`with many other agents.[39]
`
`2.1.1 Preclinical Studies
`Results from preclinical studies using animal
`models have been consistent with the receptor
`profiles described in the previous section. Using the
`conditioned avoidance response (CAR) test in rats,
`the dose-response relationship for the antipsychotic-
`like effect of asenapine was determined. For appar-
`ently adequate antipsychotic effect (i.e. 80% sup-
`pression of CAR[53]), the dose needed was 0.1–0.2
`mg/kg (dose that produces a 50% effective response
`= 0.12 mg/kg).[54] When tested in the catalepsy test,
`asenapine 0.1 and 0.2 mg/kg did not reach a score of
`2 (where catalepsy is considered to begin[53]) at any
`time interval examined. These findings suggest that
`asenapine may exhibit a potent antipsychotic effect
`without inducing EPS.[53,54]
`
` 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (16)
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1038, p. 005
`

`

`

`2274
`
`Bishara & Taylor
`
`The behavioural effects of asenapine in rats, as more effective than placebo in treating negative
`assessed by in vivo microdialysis, have been found
`symptoms. This is not consistent with findings from
`to be accompanied by an increase in dopamine re-
`earlier trials showing risperidone 6 mg/day having
`lease in the three brain regions examined: the medial
`greater improvement on the PANSS negative sub-
`scale score compared with placebo.[58,59] Although
`prefrontal cortex, nucleus accumbens and stri-
`atum.[54] Asenapine induced a marked enhancement
`statistical comparisons between asenapine and ris-
`of dopamine efflux to a greater extent in the shell
`peridone were not performed, it is perhaps important
`region of the nucleus accumbens than in the core
`to note that discontinuations due to ineffectiveness
`region, thus sharing a similar profile to atypical
`of treatments were more common with risperidone
`drugs.[54] Microdialysis and electrophysiological as-
`than asenapine.[56]
`sessments showed that asenapine potentiated both
`The incidence of adverse effects was similar
`prefrontal dopaminergic and glutamatergic trans-
`across all three treatment groups, with the most
`mission. These effects may also contribute to im-
`frequent reports for asenapine being insomnia
`provement of negative symptoms and cognitive def-
`(11%), somnolence (11%), nausea (11%), anxiety
`icits.[54,55] Furthermore, the very low concentration
`(10%) and agitation (9%).[56] Asenapine showed a
`of asenapine required to facilitate glutamatergic
`placebo-equivalent risk of significant weight gain,
`transmission, even lower than that for clozapine, whereas risperidone was associated with a higher
`suggests an important cognitive-enhancing ac-
`incidence of clinically significant weight gain, con-
`tion.[54]
`sistent with previous reports,[60,61] along with a high
`incidence of hyperprolactinaemia.[56] Laboratory
`2.1.2 Clinical Studies
`findings in this study also showed that asenapine
`A randomized, double-blind, placebo- and risper- was not associated with metabolic disturbances or
`idone-controlled,
`fixed-dose, 6-week
`trial of
`adverse effects on cardiovascular function such as
`asenapine was carried out in the US.[56] Patients
`changes in blood pressure, heart rate or corrected
`were randomly assigned to receive sublingual QT interval prolongation.[56]
`asenapine 5 mg twice daily, placebo or oral risper-
`In summary, the receptor profile and combined
`idone 3 mg twice daily. Results for the primary
`data from preclinical and clinical studies predict that
`efficacy outcome measure, the Positive and Nega-
`asenapine has an ability to improve positive, nega-
`tive Syndrome Scale (PANSS) total score,[57] for the
`tive and cognitive symptoms of schizophrenia,
`intention-to-treat population showed mean changes while causing limited extrapyramidal, antimuscarin-
`at endpoint from baseline were –15.9 with asenapine
`ic and metabolic adverse effects. It may therefore
`and –5.3 with placebo (p < 0.005). The PANSS
`prove to be a useful option in patients with predomi-
`positive subscale score showed mean changes at
`nant or resistant negative symptoms. Further large-
`endpoint from baseline of –5.5 for asenapine com-
`scale studies will be needed in order to confirm these
`pared with –2.5 for placebo (p = 0.01) and –5.1 for
`findings and to distinguish it from other D2/5-HT2
`risperidone (also significant compared with placebo;
`receptor antagonists such as risperidone.
`p < 0.05). PANSS negative subscale score results
`showed mean changes at endpoint from a baseline of
`–3.2 for asenapine compared with –0.6 for placebo
`(p = 0.01).
`Bifeprunox (figure 2) is an antipsychotic show-
`The main efficacy findings from this study were
`ing partial agonist activity at D2, D4 and 5-HT1A
`that asenapine 5 mg twice daily was superior to
`receptors, and antagonism at D3 receptors. Unlike
`placebo in treating both the positive and negative
`symptoms of schizophrenia. Risperidone (3 mg many other antipsychotics, it shows no marked ac-
`twice daily), on the other hand, was superior to
`tivity at the 5-HT2A, 5-HT2C, noradrenergic, musca-
`rinic or histaminergic receptors.[46,62]
`placebo in treating positive symptoms, but it was not
`
`2.2 Bifeprunox
`
` 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (16)
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1038, p. 006
`

`

`

`Upcoming Agents for Schizophrenia
`
`2275
`
`O
`
`HN
`
`O
`
`N
`
`N
`
`Fig. 2. Structural formula of bifeprunox (7-[4-[(3-phenylphenyl)-
`methyl]iperazin-1-yl]-3H-benzooxazol-2-one).
`
`phrenia. The efficacy, safety and tolerability of three
`fixed once-daily doses of bifeprunox (5, 10 and
`20 mg) were evaluated.
`In this study, only bifeprunox 20 mg showed a
`statistically significant reduction in PANSS total
`score[57] compared with placebo. In the last-observa-
`tion-carried-forward
`(LOCF)
`analysis, mean
`changes in PANSS total score were –11.3 and –5.3
`points for bifeprunox 20 mg and placebo, respec-
`2.2.1 Preclinical Studies
`tively (adjusted p = 0.031; treatment effect CI –11.1,
`In in vitro preclinical studies, bifeprunox demon-
`–0.4). Bifeprunox 5 and 10 mg produced mean
`strated partial dopamine agonist properties. Agonist
`changes of –9.7 and –5.0 points, respectively, but
`effects were seen to be induced when endogenous
`the difference compared with placebo was not statis-
`dopamine tone was low, for example in adenylate
`tically significant (adjusted p = 0.128, treatment
`cyclase activity assays in Chinese hamster ovary
`effect CI –9.2, 0.9 for 5 mg; p = 1.000, treatment
`cells and in rat striatal slices.[46] In contrast, in the
`effect CI –4.4, 5.5 for 10 mg). A statistically signif-
`presence of a full D2 receptor agonist (i.e. when
`icant –15.7-point change was observed in the risper-
`endogenous dopamine was high) bifeprunox acted
`idone arm (p < 0.0001 vs placebo; 95% CI –14.5,
`as a functional antagonist.[46,63] Results suggest a
`–6.5).[65] Bifeprunox 20 mg also produced statisti-
`distinct antipsychotic profile, showing functional
`cally significant changes versus placebo in the
`agonist activity in the prefrontal cortex region and a
`PANSS-positive (p = 0.037; treatment effect CI
`functional antagonist activity in brain regions such
`–2.9, –0.1) and PANSS-negative (p = 0.026; treat-
`as the nucleus accumbens where dopaminergic ment effect CI –2.6, –0.2) subscales.
`neurotransmission is thought to be increased in
`The incidence of patients withdrawing from the
`schizophrenia.[62]
`study because of adverse effects was similar in the
`In vivo preclinical studies on rats showed that
`bifeprunox and placebo groups. No statistically sig-
`bifeprunox, unlike conventional antipsychotics,
`nificant difference was observed at endpoint on any
`caused suppression of the CAR at near maximal D2 movement disorder scales between bifeprunox and
`receptor occupancy. This suppression was dose re-
`placebo. Treatment with bifeprunox at all doses was
`lated and, based on the relationship observed,
`associated with statistically significant weight de-
`bifeprunox was predicted to be clinically effective at
`creases, reduction in non-fasting total cholesterol
`doses ≥10 mg, producing >90% D2 receptor occu-
`and triglycerides, as well as decreases in prolactin
`pancy at these doses.[64] Receptor occupancy has
`levels compared with placebo.[65]
`also been studied in humans using positron emission
`tomography (PET). D2 receptor occupancy was also
`found to be dose related, with a plateau in occupan-
`cy observed at 90% for bifeprunox doses ≥10 mg
`(figure 3).[63]
`
`Observed
`Predicted
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`receptor occupancy (%)
`
`Caudate nucleus,
`
`0
`
`5
`
`10
`15
`20
`Bifeprunox (ng/mL)
`Fig. 3. Bifeprunox plasma concentration at 2 hours after administra-
`tion and dopamine D2 receptor occupancy in caudate nucleus.[63]
`
`25
`
`30
`
`2.2.2 Clinical Studies
`The following four randomized clinical trials
`have examined the efficacy and safety of bifepru-
`nox. Casey and co-workers[65] conducted a multi-
`centre, 6-week, randomized, placebo-controlled, ris-
`peridone-referenced, dose-finding study including
`589 patients with acute exacerbation of schizo-
`
` 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (16)
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1038, p. 007
`

`

`

`2276
`
`Bishara & Taylor
`
`doses of bifeprunox produced statistically signif-
`A very similar study was conducted by Rapaport
`and colleagues,[66] which evaluated the efficacy and
`icant decreases in prolactin levels (p = 0.0001),
`safety of bifeprunox at once-daily doses of 30 and whereas placebo and olanzapine were associated
`40 mg compared with placebo and with a reference with non-significant increases from baseline.[67]
`risperidone arm. In the LOCF analysis, bifeprunox
`In order to assess long-term efficacy and safety of
`30 mg produced a statistically significant difference
`bifeprunox, a 6-month, randomized, double-blind,
`from placebo in the change in PANSS total score.[57]
`parallel-group, placebo-controlled, fixed-dose study
`Mean changes were –13.5 and –7.7, respectively
`was conducted.[68] Patients received bifeprunox 20
`(p = 0.020; 95% CI –10.3, –1.4). Bifeprunox 40 mg,
`or 30 mg/day or placebo. The primary endpoint was
`on the other hand, produced a mean –10.3-point
`the time from randomization to deterioration, de-
`change, but the difference from placebo was not
`fined as a Clinical Global Impression (CGI)-Im-
`statistically significant (adjusted p = 0.156; 95% CI
`provement[69] score ≥5 or PANSS[57] item P7 (hostil-
`–7.7, 1.2).
`ity) and/or G8 (uncooperativeness) score ≥5 for
`In terms of safety assessments, the most frequent
`2 consecutive days or ≥20% increase in PANSS
`adverse events seen in bifeprunox 30 and 40 mg
`total score from baseline. Other efficacy and safety
`groups compared with placebo and risperidone were
`assessments were similar to the studies mentioned
`gastrointestinal in nature. For bifeprunox 30 mg,
`previously. In this study, treatment with bifeprunox
`they included constipation (13%), dyspepsia (11%),
`20 and 30 mg resulted in statistically significant
`nausea (18%), vomiting (12%) and dizziness (8%).
`(p = 0.008 and p = 0.006, respectively) longer time
`Similar results were seen with the higher dose.[66]
`to deterioration compared with placebo (LOCF).[68]
`Decreases in weight were greater with bifeprunox
`The proportion of patients who deteriorated was
`compared with placebo. Non-fasting cholesterol,
`40.5% in the bifeprunox 20 mg group, 38.4% in the
`glucose and triglyceride levels were also lower with
`30 mg group and 59.0% in the placebo group. In the
`bifeprunox 30 and 40 mg. Bifeprunox was also
`placebo group, the risk of deterioration was found to
`associated with a lower incidence of EPS than the
`be approximately 1.5-fold higher than the bifepru-
`active reference drug, risperidone.[66]
`nox 20 and 30 mg groups (hazard ratio 0.656 and
`Barbato and colleagues[67] used the same study
`0.653, respectively).
`design but examined once-daily doses of bifeprunox
`20 and 30 mg versus placebo, this time with olanza-
`pine 15 mg/day as the reference drug. In this study,
`no statistically significant change in PANSS total
`score[57] was observed with bifeprunox 20 or 30 mg
`compared with placebo, whereas the reference agent
`olanzapine did show significant improvement on
`measures of efficacy over placebo.
`In contrast with these efficacy findings, bifepru-
`nox showed some safety advantages over olanza-
`pine. At the 6-week study endpoint, the groups
`treated with bifeprunox 20 mg, bifeprunox 30 mg
`and placebo showed mean weight decreases of 1.05,
`0.5 and 0.59 kg, respectively, whereas olanzapine
`was associated with a weight increase of 0.26 kg
`(p < 0.0001 vs placebo).[67] Movement-related ad-
`verse effects occurred with equal frequency across
`groups and ECG results were also comparable. Both
`
`Long-term safety data showed that treatment-
`emergent adverse events were 72.3%, 83.1% and
`57.8% in bifeprunox 20 mg, 30 mg and placebo
`groups, respectively. Movement-related disorders
`occurred in 10%, 15% and 4% in the bifeprunox
`20 mg, 30 mg and placebo groups, respectively,
`although there were no clinically relevant changes in
`the Barnes Akathisia Rating Scale (BARS),[70]
`Simpson-Angus Scale (SAS)[71] and Abnormal In-
`voluntary Movement Scale (AIMS)[72] total scores at
`endpoint in all three treatment groups. Bifeprunox
`30 mg reached statistical significance in reduction in
`adjusted mean weight (p = 0.027) and triglycerides
`(p = 0.006) versus placebo, but the other dose and
`safety parameters did not reach statistical signifi-
`cance. There was no difference between the groups
`in incidence of abnormal ECG findings.[68]
`
` 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (16)
`
`Mylan v. Janssen (IPR2020-00440) Ex. 1038, p. 008
`

`

`

`Upcoming Agents for Schizophrenia
`
`2277
`
`F
`
`N
`
`O
`
`O
`
`O
`
`O N
`Fig. 4. Structural formula of iloperidone (1-[4-[3-[4-(6-fluoro-1,2-benzisoxazol-3-yl)-1-piperidinyl]propoxy]-3-methoxyphenyl]ethanone).
`
`A pooled analysis from existing studies was per-
`formed in order to evaluate the metabolic effects of
`bifeprunox compared with placebo and the active
`reference agents used. Findings from this analysis
`reflected those shown in the studies discussed in this
`section. Bifeprunox treatment was associated with a
`reduction in weight, total cholesterol and triglycer-
`ide levels and minimal changes in plasma glucose
`levels.[73]
`
`and minimal activity at cholinergic receptors indi-
`cates that anticholinergic adverse effects may be
`avoided. Moreover, as a result of the low affinity for
`α2A adrenoceptors, iloperidone is not expected to
`induce convulsions.[47,76,77]
`
`2.3 Iloperidone
`
`2.3.1 Preclinical Studies
`In animal behavioural models, iloperidone exhib-
`ited potent, long-lasting ‘antipsychotic’ activity
`against the positive symptoms of schizophrenia as
`seen in the climbing mouse assay and CAR in rats.
`However, it was 300-fold less potent in causing
`Iloperidone (figure 4) is an atypical antipsychotic
`catalepsy and in inhibiting apomorphine-induced
`initially chosen for development because of its high
`stereotyped behaviour in rats. These two measures
`affinity for 5-HT2 receptors and moderate affinity
`evaluate activity

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