You are on page 1of 13

Expert Opinion on Drug Discovery

ISSN: 1746-0441 (Print) 1746-045X (Online) Journal homepage: http://www.tandfonline.com/loi/iedc20

The preclinical discovery and development of


cariprazine for the treatment of schizophrenia

Anna Wesołowska, Anna Partyka, Magdalena Jastrzębska-Więsek & Marcin


Kołaczkowski

To cite this article: Anna Wesołowska, Anna Partyka, Magdalena Jastrzębska-Więsek & Marcin
Kołaczkowski (2018): The preclinical discovery and development of cariprazine for the treatment of
schizophrenia, Expert Opinion on Drug Discovery, DOI: 10.1080/17460441.2018.1471057

To link to this article: https://doi.org/10.1080/17460441.2018.1471057

Published online: 03 May 2018.

Submit your article to this journal

Article views: 40

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iedc20
EXPERT OPINION ON DRUG DISCOVERY, 2018
https://doi.org/10.1080/17460441.2018.1471057

DRUG DISCOVERY CASE HISTORY

The preclinical discovery and development of cariprazine for the treatment of


schizophrenia
Anna Wesołowskaa, Anna Partykaa, Magdalena Jastrzębska-Więseka and Marcin Kołaczkowskib
a
Department of Clinical Pharmacy, Jagiellonian University Medical College, Kraków, Poland; bDepartment of Pharmaceutical Chemistry, Jagiellonian
University Medical College, Kraków, Poland

ABSTRACT ARTICLE HISTORY


Introduction: Cariprazine is approved in the United States and Europe for the treatment of manic or Received 1 February 2018
mixed episodes associated with bipolar I disorder and for the treatment of schizophrenia in adult Accepted 26 April 2018
patients. It is typically administered orally once a day (a dose range 1.5 – 6 mg/day), does require KEYWORDS
titration, and may be given with or without food. It has a half-life of 2 – 4 days with an active metabolite Abuse; bipolar mania;
that has a terminal half-life of 2 – 3 weeks. cariprazine; cognition;
Areas covered: This review article focuses on the preclinical discovery of cariprazine providing details depression; rodent models;
regarding its pharmacological, behavioral, and neurochemical mechanisms and its contribution to schizophrenia
clinical therapeutic benefits. This article is based on the available literature with respect to the
preclinical and clinical findings and product labels of cariprazine.
Expert opinion: Cariprazine shows highest affinity toward D3 receptors, followed by D2, 5-HT2B, and 5-
HT1A receptors. It also shows moderate affinity toward σ1, 5-HT2A, and histamine H1 receptors. Long-
term administration of cariprazine altered the abundance of dopamine, serotonin, and glutamate
receptor subtypes in different brain regions. All these mechanisms of cariprazine may contribute toward
its unique preclinical profile and its clinically observed benefits in the treatment of schizophrenia,
bipolar mania, and possibly other psychiatric disorders.

1. Introduction Cariprazine is well absorbed after oral administration and


reaches the maximum peak plasma levels within approxi-
Cariprazine (US brand name: Vraylar®) was approved by the US
mately 3–8 h following multiple-dose administration. A high-
Food and Drug Administration (FDA) on 17 September 2015 for
fat meal (900–1000 calories) did not significantly affect Cmax or
the treatment of acute manic or mixed episodes associated with
the area under the curve (AUC) of cariprazine, which indicates
bipolar I disorder and for the treatment of schizophrenia in adults
that the effect of food on the exposure of cariprazine’s two
[1]. Cariprazine capsules are currently available in four strengths,
primary metabolites (desmethylcariprazine (DCAR) and dides-
in the following color: 1.5 mg (white cap and body imprinted
methylcariprazine (DDCAR)) was also minimal. Absolute bioa-
with ‘FL 1.5’), 3 mg (green to blue-green cap and white body
vailability of cariprazine is still unknown. The parent
imprinted with ‘FL 3’), 4.5 mg (green to blue-green cap and body
compound and its major active metabolites are highly
imprinted with ‘FL 4.5’), and 6 mg (purple cap and white body
bound (96–97% for cariprazine, 94%–97% for DCAR, and
imprinted with ‘FL 6’). In Europe, cariprazine (EU brand name:
92%–97% for DDCAR) to plasma proteins [1,2], an important
Reagila®) has been used for the treatment of schizophrenia in
fact to be considered during the treatment of comorbidities.
adult patients since 6 June 2017, in the form of hard capsules
The half-life of terminal disposition of cariprazine was found to
containing 1.5, 3, 4.5, and 6 mg of the active substance for oral
be 5–6 days and the elimination of its active metabolite
use, to be taken once daily at the same time of the day with or
DDCAR was found to be prolonged with an apparent terminal
without food [2]. The recommended starting dose of cariprazine
half-life of 2–3 weeks in healthy male volunteers [3]. A similar
is 1.5 mg. Thereafter, the dose can be increased slowly in 1.5 mg
pharmacokinetic profile has been observed for cariprazine in
increments to a maximum dose of 6 mg/day, if needed. Because
male patients with schizophrenia [4,5].
of the long half-life of cariprazine and its active metabolites,
Cariprazine is metabolized by CYP3A4 and, to a lesser extent,
changes in a dose will not be fully reflected in plasma for several
by CYP2D6 enzymes [6,7]. The parent compound and its major
weeks. Switching from other antipsychotics to cariprazine should
active metabolites are not substrates of P-glycoprotein, the
be made with caution and gradual cross-titration should be
organic anion transporting polypeptides 1B1 and 1B3, and the
considered, with gradual discontinuation of the previous anti-
breast cancer resistance protein. This suggests that cariprazine
psychotic while cariprazine treatment is initiated. Concomitant
does not interact with inhibitors of the aforementioned proteins.
smoking following oral administration had no effect on the
However, at its theoretical maximum intestinal concentration,
dosage of cariprazine.

CONTACT Anna Wesołowska a.wesolowska@uj.edu.pl Jagiellonian University Medical College, Department of Clinical Pharmacy, 9 Medyczna Street, 30-688
Kraków, Poland
© 2018 Informa UK Limited, trading as Taylor & Francis Group
2 A. WESOŁOWSKA ET AL.

receptors as the most promising compounds [11] because it


Article highlights can cross the blood–brain barrier, has a better safety profile,
● Cariprazine is approved in the United States and Europe for the
and has unique receptor actions. Binding affinities of cariprazine
treatment of manic or mixed episodes associated with bipolar I were determined using cloned human receptors or membrane
disorder and the treatment of adult patients with schizophrenia. It fractions prepared from rat tissues. It displayed picomolar affinity
is typically administered orally once a day, does require titration, may
be given with or without food, has active metabolites with a half-life
toward human dopamine D3 (pKi = 10.07) and subnanomolar
of 2-3 weeks and highly bound to plasma proteins. affinity toward rat D3 (pKi = 9.15) receptors, both D2L (pKi = 9.31)
● Clinical trials reported that cariprazine is generally safe when admi- and D2S (pKi = 9.16) subtypes of human D2 as well as human 5-
nistered at a recommended dose range of 1.5 - 6 mg/day for
schizophrenia.
HT2B (pKi = 9.24) receptors, and slightly lower for human
● Cariprazine shows highest affinity toward D3 receptors, followed by (pKi = 8.59) and rat (pKi = 8.34) 5-HT1A ones [12]. However,
D2, 5-HT2B, and 5-HT1A receptors. It also shows moderate affinity another study demonstrated a similar affinity of cariprazine
toward σ1, 5-HT2A, and histamine H1 receptors, and negligible affinity
toward a wide range of neurotransmitter receptors, transporters, and
toward the cloned human 5-HT1A and D2 receptors (both pKi
enzymes. It is D3/D2 receptor partial agonist. around 9.0) [13]. As it is shown above, the preference of caripra-
● Behavioral studies indicated that cariprazine is active in different zine for rat and human D3 versus D2 receptors is similar. This
animal models predictive of antipsychotic, antidepressant and anxio-
lytic activity. It also displays procognitive effects in several animal
difference is not observed for binding to human and rat 5-HT1A
models of memory, cognitive and executive functions in rodents. receptors. Moreover, the affinity of cariprazine toward the human
● Following are the advantages of cariprazine treatment: clinical ability D3 receptors was not found to be affected by the Ser9Gly poly-
in improving negative symptoms, ability to be useful in young
patients with a relatively short duration of schizophrenia, ability to
morphism [14], which has been associated with tolerability and
decrease substance abuse, and/or ability to maintain efficacy in cases response to antipsychotic treatment [15].
of missed doses. Cariprazine binds with moderate affinity to human σ1
● Further trials are warranted to evaluate the long-term efficacy of
cariprazine and its metabolic profile. Preclinical discovery and clinical
(pKi = 7.74), 5-HT2A (pKi = 7.73), and histamine H1
development of cariprazine should expand the therapeutic options (pKi = 7.63) receptors and shows low affinity (pKi<7.0) for 5-
available for improved treatment of schizophrenia and bipolar mania, HT7, 5-HT2C, α1A, and α1C receptors [12]. According to Kiss et al.
and other psychiatric/cognitive disorders.
[12], cariprazine displays very low affinity (pKi<6.0) toward 5-
HT6, α1B-, α2A-, β-adrenoceptors, and dopamine and serotonin
transporters and negligible affinity toward a variety of neuro-
transmitter receptors, ion channels, and transporters, includ-
cariprazine has shown effectiveness as an in vitro P-glycoprotein ing dopamines D1, D4.2, and D5; adenosine; cannabinoid;
inhibitor. Although the clinical consequences of this effect is not cholecystokinin; corticotropin; estrogen; GABAA and GABAB;
fully understood, the use of P-glycoprotein substrates with a glutamate AMPA, kainate, and NMDA subtypes; histamines
narrow therapeutic index such as dabigatran (an anticoagulant) H2, H3, and H4; muscarinic M1-5; nicotinic; opiate δ, κ, and μ;
and digoxin (used for treating heart failure) might require extra potassium channel HERG; serotonin 5-HT3, 5-HT4, and 5-HT5A
monitoring and dose adjustment [2]. Cariprazine may show sig- subtypes; sigma σ2; Ca2+ L-type and N-type channels; Na+ site-
nificant interaction with strong to moderate CYP3A4 inhibitors or 1 and site-2 channels; and norepinephrine, adenosine, and
inducers. CYP2D6 inhibitors are unlikely to have a clinically rele- choline transporters, and enzymes.
vant effect on the pharmacokinetic properties of cariprazine. It is In vitro functional receptor activity using different assays
not necessary to adjust the dose of cariprazine in patients with has been studied on both G-protein-dependent and
mild to moderate hepatic or renal impairment. However, it is not G-protein-independent signaling pathways. The results
recommended for patients with severe hepatic or renal impair- demonstrated that cariprazine either stimulates or inhibits
ment [2]. Cariprazine is generally well tolerated. The pooled the activity of D3 and D2 receptors. According to Kiss et al.
analysis of four short-term clinical trials showed that cariprazine [12], in the GTPγS-binding assay, cariprazine demonstrated
at doses of 1.5–12 mg/day was generally safe. Patients receiving silent antagonistic activity against both CHO-hD3 and native
a dose over 6 mg/day showed a greater likelihood for adverse rat striatal D2 receptors with the highest antagonist potency
events, akathisia and Parkinsonism, increase in weight, and detected (pKb = 9.50 and pKb = 8.5, respectively) among the
changes in blood pressure, as well as increased rates of creatine various antipsychotic drugs tested, having no agonist activity.
phosphokinase and transaminases [8]. Hence, the recommended In CHO cells expressing human D3 receptors, cariprazine
dose range for schizophrenia is 1.5–6 mg/day. Moreover, caripra- showed partial agonistic activity, as it inhibited forskolin-
zine in randomized Phase II [9] and Phase III [10] 6-week clinical induced cAMP accumulation presenting 10-fold higher
trials showed no clinically relevant effects on sedation, QT pro- potency than aripiprazole [12]. In mouse A9 cells expressing
longation, or prolactin elevation, issues commonly encountered human D2L receptors and cotransfected with Gqo5 protein,
with antipsychotic treatment. cariprazine was found to be a partial agonist, stimulating IP
formation (Emax ~ 30%) and preventing IP formulation
(pKb = 9.22) evoked by quinpirole, the selective D2 receptor
2. Pharmacology of cariprazine agonist [12]. Taking into account the results of in vitro func-
Cariprazine (trans-N-[4-[2-[4-(2,3-dichlorophenyl)piperazin-1-yl] tional studies, it must be confirmed that, depending on the
ethyl]cyclohexyl]-N’,N’-dimethyl-urea hydrochloride) was signaling pathway linked to D3/D2 receptors, the profile of
selected from a series of new piperazine/piperidine derivatives cariprazine corresponds to a biased agonist activity which
demonstrating high affinity to both dopamine D3 and D2 was already proposed by De Deurwaerdere [16].
EXPERT OPINION ON DRUG DISCOVERY 3

Cariprazine acts as a neutral antagonist of the rat 5-HT2B reduced serotonin turnover in all three regions studied: olfactory
receptor as it potently inhibited serotonin-induced contrac- tubercles, striatum, and frontal cortex [12].
tion of isolated rat stomach fundus strips (IC50 = 12.9 nM)
having no effect by itself [12]. Moreover, the compound
4. Chronic effects of cariprazine
potently stimulated [35S]GTPγS binding in membranes pre-
pared from rat hippocampus containing 5-HT1A receptors Extrapyramidal, limbic, and cortical regions of the brain
and showed moderate intrinsic activity (Emax = 38.6%) as that mediate motor, emotional, and cognitive behaviors,
compared to 8-OH-DPAT, a full agonist of these receptors. respectively, are typically disturbed in patients with schizo-
In in vitro assays, cariprazine also showed moderate antag- phrenia [25]. Furthermore, clinically effective antipsychotic
onistic activity toward 5-HT2A (inhibition of IP formation drugs produce region-specific changes in monoaminergic
(IC50 = 403 nM) in CHO cells evoked by the 5-HT2A receptor and glutamatergic receptor subtypes’ levels. Chronic effects
agonist DOI) and histamine H1 (inhibition of the histamine- of cariprazine on different neurotransmitter receptors were
induced contraction (IC50 = 149 nM) of isolated guinea pig evaluated in male Sprague Dawley rats receiving daily
trachea) receptors [12]. intraperitoneal injections of three doses of cariprazine
A positron emission tomography (PET) study in monkeys active in behavioral paradigms, that is, 0.06, 0.20, or
showed that administration of cariprazine at increasing doses 0.60 mg/kg [26], for 28 days [27,28]. At the end of the
(30–300 µg/kg) resulted in a dose-dependent and saturable treatment, brains were collected and then processed for
striatal D2/D3 receptor occupancy approximately 94% after the quantitative in vitro receptor autoradiography using radi-
highest dose, whereas in the raphe nuclei, the occupancy of the oligands that selectively label receptors of interest. Sections
5-HT1A receptor was about 30%. These data confirmed in vitro were obtained from different brain regions that represent
affinity cited above [17]. In a previous PET analysis, by utilizing a extrapyramidal, limbic, and cortical systems. Cariprazine-
D3 receptor-preferring radioligand such as 4-propyl-9-hydroxy- evoked changes in different receptors after repeated treat-
naphthoxazine ([3H]-(+)-PHNO), the authors demonstrated that ment can be regarded as a functional index of its actions
cariprazine at a dose of 1 mg/kg completely inhibited the uptake because specific chronic drug–receptor interactions conse-
of this radioligand in rat cerebellum [18]. In healthy subjects, quently trigger changes in signal transduction cascades
cariprazine occupied 3–79% of striatal D2/D3 receptors when and downstream neuronal elements and pathways, and
measured 4 h after 0.5–1 mg doses repeated for 2–12 days [19]. contribute to the clinical efficacy, safety, and tolerability
Using PET with [11C]-(+)-PHNO scans performed in patients with of the studied drug.
schizophrenia, Girgis et al. [20] demonstrated a dose–occupancy
relationship for D3 and D2 receptors after 2 weeks of cariprazine
4.1. Dopamine receptors
treatment. At a high dose of 12 mg/kg of cariprazine, nearly
100% occupancies were observed for both receptors. Repeated doses of cariprazine induced a significant increase in
Selectivity for D3 over D2 receptors were noted after lower D3 receptors localized in forebrain regions [27], where they are
doses. An exposure-response analysis presented an ~ 3-fold highly expressed. This effect appears to be unique for caripra-
difference in EC50 (D3 = 3.84 nM and D2 = 13.03 nM) in plasma zine since other (both typical and atypical) antipsychotics
after 2 weeks of dosing [20]. All these results show the unique (including haloperidol, fluphenazine, clozapine, olanzapine,
receptor signature and functional activity of cariprazine and risperidone, and asenapine) did not evoke such changes in
support its distinct psychopharmacological profile for the all regions examined [29–32]. Moreover, cariprazine differen-
improved treatment of schizophrenia. tially altered D3 receptor levels in the subdivisions of the
nucleus accumbens (NAc), shell and core, which are associated
with limbic and motor systems, respectively [33]. Cariprazine
significantly induced greater increases in D3 receptors loca-
3. Neurochemical effects of cariprazine
lized in the NAc-shell versus NAc-core [27]. This finding further
Atypical antipsychotics are able to preferentially increase extra- supports the atypical antipsychotic profile of the drug because
cellular concentrations of monoamines and acetylcholine in cor- it was reported that atypical antipsychotics (e.g. clozapine)
tical and limbic brain regions [21–24]. In vivo studies with preferentially induce changes in dopamine levels in the NAc-
cariprazine focused primarily on dopaminergic and serotonergic shell, whereas typical drugs (e.g. haloperidol) preferentially act
neuronal activity [12]. Cariprazine enhanced the turnover and in the NAc-core [34]. Potent affinity of cariprazine for D3
biosynthesis of dopamine in mouse brain, preferentially in olfac- receptors, its considerable D3 receptor occupancy
tory tubercles (i.e. limbic region) compared to the striatum. [12,17,18,20], partial agonist activity at these sites, and their
Moreover, this compound potently and dose-dependently, but upregulation by the drug support its unique action among
only partially, reduced the dopamine biosynthesis in the striatum antipsychotics that may be responsible for the normalization
of reserpinized mice and normalized a metabolite of dopamine of disturbances in D3 receptor-mediated neurotransmission in
(3,4-dihydroxyphenylacetic acid) accumulation to the control patients with schizophrenia. In addition, unique actions of
levels, reaching a plateau at the highest dose used. Cariprazine cariprazine at these receptors may provide therapeutic bene-
showed lower potential in inhibiting dopamine neurotransmis- fits in the treatment of negative symptoms of the disease,
sion in the striatum relative to the limbic area, which suggests cognitive, and executive deficits, and thanks to a concomitant
that it has low propensity toward causing extrapyramidal symp- preference to the NAc-shell over the NAc-core – in the treat-
toms [12]. In addition, cariprazine moderately but significantly ment of mood symptoms as well as lesser extrapyramidal side
4 A. WESOŁOWSKA ET AL.

effects. In addition, in comparison to aripiprazole, cariprazine 4.3. Glutamate receptors


produced more profound increases in D3 receptors in many
Continuous administration (28 days) of cariprazine significantly
regions studied, especially in the NAc-shell [28] which is pro-
decreased the binding of an appropriate radioligand to NMDA
posed to be a common neurobiological mechanism of anti-
receptors in NAc and the medial and lateral caudate putamen
depressant treatment.
[28] evoking a similar effect to those reported for atypical (includ-
Long-term administration of cariprazine showed dose-
ing aripiprazole [28]), but not typical haloperidol [43–45] anti-
dependent upregulation of D2 receptors in the medial pre-
psychotics. Reduction in striatal NMDA receptors may result from
frontal cortex, NAc, and caudate putamen [27], which is char-
indirect interaction with dopamine and serotonin systems that
acteristic of both typical and atypical antipsychotics [28–
modulate glutamatergic transmission [46,47] in basal ganglia
30,32]. Cariprazine differs from typical neuroleptics in upregu-
which, at least in part, could be responsible for the benign
lating D2 receptors in the hippocampal areas as well, which
extrapyramidal effects of cariprazine. Repeated doses of caripra-
may explain some differences between the two classes of
zine also downregulated NMDA receptors in the hippocampal
antipsychotics. An increase in striatal D2 binding sites may
CA1 and CA3 subregions, the effect that may normalize the
account for higher clinical incidence of akathisia (9% versus
abnormalities in glutamatergic transmission occurring in the
15%) and extrapyramidal side effects (12% versus 5%) in car-
hippocampus of patients with schizophrenia, and subsequently
iprazine-treated patients than that of placebo-treated patients
improve their psychotic symptoms [48].
[35,36]. Long-term treatment with cariprazine also increased
Long-term treatment with cariprazine at doses of 0.2 and
the striatolimbic D4 receptor subtype [27], which appears to
0.6 mg/kg increased AMPA receptors in the hippocampal CA1
be a common mechanism of action for typical and atypical
and CA3 areas only [28] similar to aripiprazole [28] and asena-
antipsychotics [29–32].
pine [44]. Upregulation of ionotropic AMPA receptors may
Repeated treatment with different doses of cariprazine did
further contribute to the improvement of psychotic symptoms
not induce any changes in binding to D1 receptors in all the
of schizophrenia by direct action of cariprazine on D2 recep-
examined regions [27], which is a common feature of typical and
tors colocalized with AMPA in the brain [49]. Moreover, oppo-
atypical antipsychotics [30–32], except asenapine [29]. Because
site effects on NMDA versus AMPA receptors of cariprazine
of negligible affinity of cariprazine for D1 receptors, it is possible
suggest different ways of action of this D3/D2 partial agonist
that the drug did not occupy these sites to the extent required
on ionotropic glutamatergic binding sites.
to induce their upregulation as is the case with D4 receptors.
Moreover, similarly to atypical antipsychotics [37,38],
chronic (21 days) treatment with cariprazine suppressed pre-
5. Activity of cariprazine in behavioral models of
ferentially spontaneous activity of mesolimbic dopamine neu-
positive symptoms of schizophrenia
rons (45–66% at doses of 0.1–1.0 m/kg), but had no effects on
their activity in mesostriatal areas [39]. These data further 5.1. Apomorphine-induced climbing
support the utility of cariprazine as an antipsychotic drug
This model is considered as an animal test for screening anti-
effective in patients with schizophrenia.
psychotic activity of standard and experimental compounds
characterized by D1 and/or D2 antagonist activity [50].
Potential antipsychotics attenuate climbing behavior of mice
4.2. Serotonin receptors evoked by apomorphine which enhances dopaminergic neu-
rotransmission in the mesolimbic system. Cariprazine, orally
An increase in 5-HT1A binding receptors after repeated
administered, potently inhibited apomorphine-induced climb-
(28 days) treatment of cariprazine or aripiprazole was mea-
ing in mice with a median effective dose (ED50) of 0.27 mg/kg
sured in the medial and dorsal prefrontal cortex, which is
[26], which supports cariprazine’s high ability to block D2
shared with atypical antipsychotics [40,41]. In addition, the
receptors in vivo [12].
measured 5-HT1A receptor upregulation in the hippocampal
CA1 and CA3 subregions after cariprazine treatment is
shared with aripiprazole [28] and asenapine only [41].
5.2. Novelty or psychostimulant-induced hypermotility
However, both latter compounds downregulate 5-HT2A
receptors in all the examined forebrain regions but caripra- Cariprazine dose-dependently attenuated novelty-evoked
zine failed to alter that [28]. This lack may be due to the locomotor activity both in mice (ED50 = 0.18 mg/kg) and in
lower in vitro affinity of cariprazine toward 5-HT2A receptors rats (ED50 = 0.11 mg/kg) reaching a plateau at approximately
resulting in insufficient occupancy of these binding sites to 70% inhibition as shown by its sigmoidal fit [26]. Isolation
the level required to induce their changes. This prolong rearing of rats alone or combined with neonatal phencyclidine
action of cariprazine on 5-HT1A receptors might contribute, (PCP) administration causes developmental induction of a
at least in part, to its beneficial therapeutic effects in mini- reproducible hyperactivity in response to placement in a
mizing the incidence of extrapyramidal side effects and in novel arena. This effect is regarded as a marker for striatal
improving cognitive functions in patients with schizophre- dopaminergic hyperactivity akin to positive symptoms of schi-
nia acting indirectly via neocortical glutamate pyramidal zophrenia. Moreover, it is attenuated by several antipsychotic
neurons, where these receptors are located [42]. Serotonin drugs [51–53]. Cariprazine at doses of 0.1 and 0.3 mg/kg
5-HT2A receptors are less likely to contribute to the in vivo attenuated the hyperactivity of rats induced by neonatal PCP
mechanism of cariprazine’s action. and social isolation rearing [54]. Since most typical and
EXPERT OPINION ON DRUG DISCOVERY 5

atypical antipsychotics attenuate or block the stimulation accompanied by attentional impairments [63]. Another model
evoked by amphetamine in locomotor activity [55], cariprazine used in the development of potential antipsychotics constitutes
was tested after oral administration in rats using that test. It neonatal treatment of rats with PCP that disrupts neuronal devel-
showed blockade of amphetamine-induced hypermotility with opment and synaptogenesis and also reproduces many behavioral
ED50 = 0.12 mg/kg [26]. In glutamatergic models of positive features akin to schizophrenia in adult animals such as hyperactiv-
symptoms of schizophrenia, that is, locomotor activity induced ity to psychostimulants, attenuated sensorimotor gating, and cog-
by MK-801 or PCP, cariprazine demonstrated potent inhibition nitive deficits [64,65]. Furthermore, combining neonatal PCP
against both psychostimulants, MK-801 in mice administration with subsequent isolation rearing causes additional
(ED50 = 0.049 mg/kg) and PCP in rats (ED50 = 0.09 mg/kg) attenuation of social interaction [66,67], imitating social withdra-
[26]. As Leriche et al. [56] demonstrated that MK-801-induced wal observed in patients. One of the advantages of such afore-
hyperactivity is very sensitive to D3 receptor antagonist/partial mentioned models is evaluation of behavioral deficits in a drug-
agonist action, the above effect of cariprazine supports its free state because after PCP treatment there is washout period
ability to block/partially block D3 receptors in vivo. included, lasting for 1 week or longer. Then, any potentially con-
founding effects on behavior as a result of the PCP present in the
animal are eliminated.
5.3. Serotonergic activity models in rats
Because of its high affinity toward 5-HT1A receptors and mod-
6.1. Novel object recognition paradigm (NOR)
erate affinity toward 5-HT2A [12], cariprazine was further stu-
died in rat lower lip retraction and mouse DOI-induced head NOR task, a measure of visual recognition memory, disrupted
twitch tests, respectively. Cariprazine induced lower lip retrac- by PCP is associated with hypodopaminergic dysfunction in
tion after oral administration of 2 mg/kg and dose-depen- the prefrontal cortex [59]. Cariprazine (0.05 mg/kg) reversed
dently reduced head twitch behavior evoked by DOI, this impairment, evoking no effects on line crossings confirm-
yielding an ED50 value of 0.27 mg/kg [26]. These finding con- ing specificity of this action. A dose of 0.1 mg/kg cariprazine
firm cariprazine agonist/partial agonist 5-HT1A activity and 5- significantly reversed PCP-induced deficits in NOR in spite of
HT2A antagonist action, respectively. the significant reduction in line crossings. In addition, the
compound had no effects on the total object exploration.
The highest dose tested (0.25 mg/kg) evoked significant seda-
5.4. Catalepsy
tive effects by attenuating the number of line crossings and
Up to a dose of 85 mg/kg cariprazine did not induce catalepsy object exploration [62]. In the ‘dual-hit’ model of the disease,
in rats [26], which could reflect its weak propensity toward cariprazine (0.03 and 0.3 mg/kg) dose-dependently reversed a
extrapyramidal side effects in the clinical-setting. Several delay-induced impairment in NOR in rats given neonatal PCP
mechanisms may underlie this feature, that is, mixed D3/D2 and housed in social isolation [54]. This effect may be due, at
antagonist activity with D3 preference, partial D2 agonist, and/ least in part, to cariprazine’s D3 and/or D2 blockade as well as
or 5-HT1A receptor partial agonist efficacy [12]. 5-HT1A partial agonist activity [12].

5.5. Conditioned avoidance response (CAR) 6.2. Executive function models


CAR assay features high construct validity with respect to Patients who suffer from frontal lobe deficiencies can easily
efficacy and potency on the positive symptoms of schizophre- learn and follow individual rules, but have great difficulty
nia [57] and has shown a high degree of predictive validity for modifying their responses to new rules [68–70]. Patients with
screening typical and atypical antipsychotic activity [58]. schizophrenia do not adapt normally to changes in their
Cariprazine dose-dependently inhibited rat CAR in the shuttle environments, especially in social and emotional contexts,
box with ED50 = 0.84 mg/kg [26]. Inhibition of the CAR effect and they exhibit an inability to modify responses in testing
by cariprazine shows a good correlation with its striatal D2 situations [71–74]. There are behavioral tasks requiring animals
receptor occupancy [12]. to show cognitive flexibility, attention, and motivation [59,75].
Usually PCP is used to impair the aforementioned features in
animal models. In reversal learning task, rats maintained at
6. Activity of cariprazine in behavioral models of
80–90% free-feeding weight were trained to respond for a
negative symptoms and cognitive dysfunction
food reward on a fixed ratio schedule of reinforcement, so
occurring in schizophrenia
that pressing either of the two active levers delivers food
Repeated PCP administration appears to produce robust and pellets. Once animals had acquired a lever-pressing response,
enduring effects that are more consistent with the chronic symp- they were trained to press either the left or the right lever
toms observed in patients with schizophrenia [59]. Chronic PCP (only one was active then) for food delivery. The active lever
exposure has been associated with lasting neurochemical changes varied from day-to-day and such training lasted a couple of
in the prefrontal cortex, including decreased cerebral blood flow, weeks. Next, the rats were trained to press either the left or
glucose utilization, and decreased dopamine neurotransmission the right lever for food delivery according to the presence or
[60,61]. Moreover, subchronic PCP (e.g. 7 days [62]) exposure was absence of a visual cue until they reach ≥90% of the correct
linked to highly localized morphological abnormalities in the brain responding. In the reversal learning task, rats are tested for
regions associated with the pathogenesis of schizophrenia and their ability to abandon a previously learned reward
6 A. WESOŁOWSKA ET AL.

contingency and acquire a new strategy to obtain reward. PCP rats. Lower (0.01 mg/kg) and higher doses (0.15–0.3 mg/kg)
in that test impairs the ability of rats to switch learning stra- of the drug did not evoke significant responses [26].
tegies in the reversal phase. Cariprazine at doses of 0.1 and Cariprazine’s unique receptor profile may support its procogni-
0.25 mg/kg significantly attenuated such PCP impairments tive activity observed in the most described paradigms, in parti-
showing efficacy in improving executive function [62]. The cular antagonism at D3 receptors. This, in combination with
attentional set-shifting task (ASST) employs both relevant appreciable affinity toward D2 receptors, high affinity toward 5-
and irrelevant stimuli (e.g. odor and texture) with the help of HT2B, and moderate affinity toward 5-HT1A binding sites as well as
which animals are trained to retrieve a food reward. During partial agonist/antagonist action toward these receptors may help
separate phases of the task, animals learn to reach an appro- to explain cariprazine’s procognitive effects in preclinical models.
priate criterion of consecutive correct trials using first an
intradimensional and then an extradimensional shift of atten-
tion which is disturbed (e.g. by PCP). In wild-type mice, car- 7. Activity of cariprazine in behavioral models of
iprazine at doses of 0.01 and 0.02 mg/kg significantly reverses anxiety and depression
deficits induced by PCP administration, while showing no 7.1. Elevated plus-maze (EPM)
activity in D3-receptor knockout mice [76].
EPM is a behavioral test that is widely employed to assess
anxiety-like behaviors in rodents. This test is based on
6.3. Conditioned freezing response (CFR) approach/avoidance conflict, with rodents perceived as ‘less
anxious’ being more willing to explore the brighter, open, and
CFR is performed using a two-chamber shuttle box with light
elevated arms of the apparatus as opposed to remaining in
and dark sides and with wire grid floors, separated by an
the darkened and enclosed arms. Measures of the time spent
automated door. Conditioned (light and tone) and uncondi-
in open arms and the number of open arm entries serve as an
tioned (foot shock) stimuli are delivered according to different
index of anxiety-like behavior. Cariprazine in the dose range of
protocols. Reduction in freezing following reexposure to the
0.005–0.15 mg/kg was tested in EPM in wild-type mice having
context or cue may result from altered hippocampal and
no anxiolytic-like action. Only at the highest doses of 0.08 and
amygdala functions [76,77]. Cariprazine given at doses of 0.1
0.15 mg/kg, the drug decreased the number of open and
and 0.3 mg/kg did not reverse the CFR deficit in the ‘dual-hit’
closed arm entries showing sedative action [76].
model [54], indicating that it lacks the effect on associative
learning in the clinical setting.
7.2. Chronic mild stress (CMS)
6.4. Social interaction Anhedonia is a core component of negative symptoms in
schizophrenia and one of the most challenging-to-treat symp-
Subchronic treatment of PCP produces significant increases in toms associated with depression [78,79]. A rodent model that
avoiding social behavior of rats such as sniffing, following conspe- mimics anhedonic symptoms and the lack/decrease in the
cifics, and other interactions. Cariprazine at a dose of 0.05 mg/kg ability of such patients to experience pleasure is CMS. In this
reversed PCP-induced social behavior deficits in the absence of any model, animals (usually rats or mice) are chronically exposed
effect on line crossings. Higher doses of the drug (0.1 and 0.25 mg/ to a constant bombardment of unpredictable and different
kg) produced nonspecific activity in this test due to moderate microstressors (e.g. forced swim, cage tilt, light/dark distur-
sedative effects [62]. In another study [54], the total social interac- bance, no/wet bedding, odor, and strobe light), resulting in
tion was unaffected by cariprazine administered at doses of 0.1 the development of behavioral changes, including decreased
and 0.3 mg/kg. PCP administered at a single dose in mice pro- response to rewards. A rodent version of rewards is access to a
duced reduction in social interaction time and social recognition highly preferred sweet solution, or to a choice between a
memory. Cariprazine at doses of 0.01 and 0.02 mg/kg significantly sweet solution (sucrose) and plain water. Consumption of, or
reversed social recognition/interaction and social recognition preference for, the sweet reward decreases over several weeks
memory impaired by PCP in wild-type mice, but not in D3-receptor of exposure to stressors, but can be restored to normal levels
knockout mice [76], confirming that dopamine D3 receptors con- by chronic treatment with antidepressant drugs from many
tribute to the action of cariprazine. therapeutic classes, acting by different intracellular and sys-
temic mechanisms [79]. CMS, sometimes also called chronic
unpredictable/varied stress, is considered one of the best-
6.5. Spatial working memory models
validated animal models available to test novel compound
PCP given at a single dose evoked significant impairments in treatments for anhedonic-like symptoms [80].
the performance of mice in a delayed alternation test per- Repeated treatment (5 weeks) with cariprazine in the dose
formed in a T-maze. Cariprazine in the dose range of 0.005– range of 0.01–1 mg/kg reversed the CMS-induced reduction in
0.2 mg/kg significantly reversed PCP-induced deficits in wild- sucrose consumption in rats reaching an ED50 dose of
type mice, but not in D3-receptor knockout mice [76]. 0.052 mg/kg. Only the lowest (0.01 mg/kg) and the highest
Similarly, in water-labyrinth learning performance, when an (1 mg/kg) doses of cariprazine did not statistically reverse
amnestic drug, scopolamine, was used in order to induce anhedonia in rats, possibly due to insufficient levels of D2
deficits in memory, cariprazine in the dose range of 0.02– and D3 receptors’ engagement or sedative effects, respec-
0.075 mg/kg significantly reversed the amnestic effects in tively. This reversal was similar to that observed after
EXPERT OPINION ON DRUG DISCOVERY 7

treatment with the standard antidepressant medication such 9. Conclusion


as imipramine (10 mg/kg) or aripiprazole (ED50 = 4.4 mg/kg)
Cariprazine is an antipsychotic medication that received
[81]. Chronic administration (21–22 days) with cariprazine at
approval from the US FDA in September 2015 for the acute
doses of 0.2 and 0.4 mg/kg in mice exposed to chronic unpre-
treatment of manic or mixed episodes associated with bipolar
dictable stress (CUS) statistically reversed the attenuation of
I disorder and the treatment of schizophrenia in adults. It has
sucrose consumption in a novel environment, producing an
also received approval from the European Medicines Agency
antianhedonic-like effect on the same level as imipramine
in June 2017 for the treatment of schizophrenia in adult
(20 mg/kg) and aripiprazole (5 mg/kg). Interestingly, the
patients. It is formulated into capsules/hard capsules of differ-
same activity of cariprazine was not observed in D3-receptor
ent strengthens and administered orally, typically once daily at
knockout mice. Furthermore, the same authors also demon-
the same time of the day with or without food. Cariprazine
strated anxiolytic-like activity of cariprazine (0.2 and 0.4 mg/
and its major active metabolites (DCAR and DDCAR) are highly
kg) after repeated administration (24–25 days) to wild-type
bound (>90%) to plasma proteins which is an important factor
mice, but not D3-receptor knockout mice, exposed to CUS by
to be considered when treating comorbidities. The half-life of
significantly reducing latency to drink (diluted sweetened con-
terminal disposition of cariprazine is 5–6 days and elimination
densed milk) in the novelty-induced hypophagia test.
of active DDCAR is prolonged even to 2–3 weeks in both
Cariprazine produced the same level of activity as aripiprazole
healthy volunteers and patients with schizophrenic patients.
(1 and 5 mg/kg) in wild-type mice [82]. The above findings
Cariprazine is metabolized by the CYP3A4 pathway, and to a
show a clear involvement of D3 receptors in both the anxioly-
lesser extent, by CYP2D6. Hence, it should not be coadminis-
tic- and antidepressant-like activity of cariprazine. Moreover,
tered with CYP3A4 inducers or inhibitors. It is also not recom-
Duric et al. [82] observed that aripiprazole induced increases in
mended in patients with severe renal or hepatic impairment.
the total fluid consumption, which could be due to polydipsia
In patients with schizophrenia, cariprazine is recommended in
and/or polyuria, potential side effects of some antipsychotic
the dose range of 1.5–6 mg/day. The efficacy of cariprazine in
drugs [83,84]. In this regard, cariprazine had a more specific
acute schizophrenia was established in, three positive 6 week,
effect independent of changes in the overall liquid
Phase II/III, randomized controlled clinical trials which demon-
consumption.
strated improvement over placebo-treated patients [83].
Additionally, in 26-week, double-blind, randomized study in
8. Antimanic and antiabuse activities of cariprazine nonelderly patients with schizophrenia, and predominant
negative symptoms, cariprazine is superior to risperidone on
Intracerebroventricular single injection of ouabain is the
both the primary and the key secondary outcomes [83]. The
only available model of bipolar disorder and is among
most commonly encountered adverse effects are extrapyrami-
the best characterized rodent models of mania sensitive
dal symptoms and akathisia. Changes in body weight (small),
to lithium and other antimanic agents. Ouabain increases
glucose, lipid, and prolactin levels as well as in QT interval are
dopamine levels in the synapse and induces hyperactivity
not clinically meaningful.
measured in the open field test. It can also alter intracel-
Cariprazine exhibits the highest affinity toward dopamine
lular signaling pathways such as Akt-GSK-3 and D2 recep-
D3 (pKi = 10.7) and D2 (pKi = 9.31) receptors, followed by
tor/β-arrestin 2 [85,86]. Cariprazine (0.06–1 mg/kg), acting
serotonin 5-HT2B (pKi = 9.24) and 5-HT1A (pKi = 8.59). It has
as a D2 receptor partial agonist, attenuated hyperlocomo-
negligible affinity toward many other receptors, ion channels,
tor activity induced by ouabain. Subchronic (7 days)
and enzymes [12]. Interestingly, cariprazine does not share
administration of cariprazine at a sedative dose of 1 mg/
with other atypical antipsychotic drugs their higher affinity
kg statistically reduced an ouabain-evoked increase in
toward 5-HT2A (pKi = 7.73) over D2 receptors. But, its unique-
locomotor activity [86]. Although the above effect mea-
ness depends on having a higher potency for the D3 receptors
sured in animals appears to be unspecific, the results
than dopamine itself. Cariprazine demonstrated approximately
obtained from a recent meta-analysis of antimanic treat-
10-fold lower affinity toward histamine H1 and 5-HT2C recep-
ments showed that cariprazine demonstrates robust clinical
tors than for example, aripiprazole. Therefore, it is hypothe-
efficacy in patients with acute mania associated with bipo-
sized that cariprazine has low propensity toward causing
lar disorder [87].
sedation and body weight gain, side effects commonly asso-
Behavioral features of addiction including drug intake and
ciated with high H1 and 5-HT2C antagonist activity. Functional
relapse are amenable to modeling in animals. Rodents readily
in vitro studies found that cariprazine exhibits biased agonist
self-administer the same psychoactive drugs as humans
activity against D3/D2 receptors (depending on the assay/sig-
including cocaine, and following a period of abstinence, rein-
naling system involved), complete antagonistic activity toward
state their drug seeking when exposed to the same factors
5-HT2B, and partial agonist activity against 5-HT1A. Cariprazine
that put humans at risk of relapse. One of such paradigms is
has lower intrinsic activity at D2 receptors than aripiprazole
the cocaine self-administration model that mimics the situa-
which may translate into a better clinical profile because a bit
tion of abusers. Cariprazine reduced the rewarding effect of
higher agonist activity of aripiprazole seems to be responsible
cocaine (0.17 mg/kg) and attenuated relapse to cocaine-seek-
for its activation, agitation, and akathisia in some patients. In
ing with an ED50 value of 0.2 mg/kg in rats [88]. The antiabuse
this respect, cariprazine is more comparable to brexpiprazole
effect may enhance the therapeutic value of cariprazine as an
[90]. Whether the 5-HT2B receptor antagonism of cariprazine,
antipsychotic drug since drug abuse is a frequent comorbidity
in addition to its D3, D2, and 5-HT1A affinities, contributes to its
of both schizophrenia and mania.
8 A. WESOŁOWSKA ET AL.

antipsychotic activity and the side effect profile is presently Table 1. Summary of some literature data supporting the activity of cariprazine
in animal models of neurological disorders.
unclear. This explanation may be particularly important and
interesting given the recent data demonstrating that the Behavioral paradigm ED50/dose range efficacy
blockade of 5-HT2B receptors reduces significantly mesoac- Schizophrenia
Positive symptoms
cumbal dopamine transmission [91], whereas 5-HT2B receptor Apomorphine-induced 0.27 mg/kg in mice
deficiency in mice induces a wide spectrum of schizophrenic- climbing
like behavioral and psychopharmacological phenotypes [92]. DOI-induced head twitches 0.27 mg/kg in rats
Novelty-induced locomotor 0.18 mg/kg in mice
Clinical consequences of relatively low affinity of cariprazine hyperactivity 0.11 mg/kg in rats
toward 5-HT2A receptors are also unknown yet. Locomotor hyperactivity 0.12 mg/kg inhibited amphetamine effect in
Behavioral studies demonstrated cariprazine’s effectiveness rats
0.09 mg/kg inhibited PCP effect in rats
in different animal models of schizophrenia, mimic positive, 0.049 mg/kg inhibited MK-801 effect in mice
negative, and/or cognitive symptoms. It inhibited apomor- Conditioned avoidance 0.84 mg/kg in rats
phine-induced climbing behavior, CAR, and attenuated psy- response
Negative symptoms and
chostimulant or novelty-induced increases in locomotor cognitive impairments
activity (rodent models predictive of antipsychotic activity). Novel object recognition 0.05 and 0.1 mg/kg reversed PCP deficits in
In tests predictive of negative (social interaction), cognitive (NOR) rats
‘Dual-hit’ model in rats 0.1 and 0.3 mg/kg inhibited hyperactivity
(water-labyrinth learning performance and NOR), and execu- 0.03 and 0.3 mg/kg reversed PCP deficits in
tive functions (ASST and reversal learning task), cariprazine NOR
ameliorated subchronic or neonatal PCP and isolation-induced 0.1 and 0.3 mg/kg were inactive in
conditioned freezing response
deficits in rats. In animal models of anxiety and depression, 0.1 and 0.3 mg/kg were inactive in social
repeated cariprazine evoked anxiolytic- and antidepressant- interaction
like effects, in the novelty-induced hypophagia test and CMS Social interaction 0.05 mg/kg reversed PCP deficits in rats
0.01 and 0.02 mg/kg reversed PCP deficits in
model, respectively (Table 1). mice
Long-term cariprazine induced selective adaptive changes 0.01 and 0.02 mg/kg were inactive in D3-
in dopaminergic, serotonergic, and glutamatergic receptors in receptor knockout mice
Reversal learning task 0.1 and 0.25 mg/kg attenuated PCP deficits in
various rat brain regions. It produced regional and dose- rats
dependent upregulation of D2 and D4 receptor subtypes simi- Attentional set-shifting task 0.01 and 0.02 mg/kg reversed PCP deficits in
lar to other atypical antipsychotics. Hippocampal D4 receptors mice
T-maze 0.005–0.2 mg/kg reversed PCP deficits in mice
may constitute common targets that mediate the molecular 0.005–0.2 mg/kg were inactive in D3-receptor
actions of cariprazine, acting together with hippocampal D2 knockout mice
receptors to improve impaired emotional behavior that is Water-labyrinth learning 0.02–0.075 mg/kg reversed scopolamine
performance deficits in rats
commonly associated with psychosis. Additionally, cariprazine Anxiety
was unique in upregulating D3 receptors, which has not been Elevated plus-maze 0.005–0.15 mg/kg were inactive in mice
seen with other antipsychotics. The clinical consequences of Novelty-induced hypophagia 0.2 and 0.4 mg/kg reduced latency to drink in
mice
such a mechanism for patients are unknown, but preclinical 0.2 and 0.4 mg/kg were inactive in D3-
experiments suggest that such an action could be antidepres- receptor knockout mice
sant, anxiolytic, procognitive, active on negative symptoms of Depression
Chronic mild/unpredictable 0.052 mg/kg induced antianhedonic effect in
schizophrenia, and even be helpful in stimulant abuse stress rats
[26,76,80,88,90]. Action at 5-HT1A receptors is a mechanism 0.2 and 0.4 mg/kg induced antianhedonic
thought to enhance the neurochemical and behavioral effects effect in mice
0.2 and 0.4 mg/kg were inactive in D3-
of antidepressant drugs (e.g. selective serotonin reuptake inhi- receptor knockout mice
bitors) [93]. Activity toward glutamate NMDA and AMPA Antimania activity 0.06–1 mg/kg attenuated ouabain
receptors support cariprazine’s ability to improve psychotic hyperactivity in rats
Addiction
and cognitive symptoms visible, among others, through Cocaine self-administration 0.17 mg/kg reduced the rewarding effect in
restoring PCP-induced deficits in glutamatergic neurotrans- rats
mission. Both latter mechanisms are also responsible for mini- 0.2 mg/kg attenuated relapse to cocaine-
seeking in rats
mal extrapyramidal effects seen during cariprazine therapy. Side effects
The clinical consequences of the lack of 5-HT2A receptor action Catalepsy up to 85 mg/kg was inactive in rats
are not established yet. Summing up, the unique actions of Serotonergic syndrome 2 mg/kg induced lower lip retraction in rats
Sedative effect 0.1, 0.25, 1 mg/kg in rats
cariprazine on dopamine D3 receptors, combined with its 0.08 and 0.15 mg/kg in mice
effects on serotonin and glutamate receptor subtypes, may
confer the clinical benefits, safety, and tolerability of this novel
medicine in schizophrenia and bipolar mania.
metabolic side effects. In addition, the available compounds
show limited effect in the domains of negative and cognitive
10. Expert opinion symptoms. Owing to its unique pharmacodynamic profile,
Atypical antipsychotics are considered effective therapeutic cariprazine does not appear to be particularly associated
options in the treatment of schizophrenia, but they are with metabolic issues, increase in prolactin or QTc prolonga-
encumbered by a number of adverse effects, including tion, but extrapyramidal side effects and akathisia are more
EXPERT OPINION ON DRUG DISCOVERY 9

common. Cariprazine shares more similarities with the newer characterized by poor compliance. Thus, missing a dose here
antipsychotics such as the so-called dopamine stabilizers (e.g. and there would not have a ruinous effect on relapse, which is
aripiprazole and brexpiprazole), thanks to partial agonist yet to be proven in a clinical setting [90].
activity at D2 receptors, which translates into the stabilization Cariprazine is unique among antipsychotic drugs in terms
of dopaminergic neurotransmission in appropriate brain of having a higher potency for the D3 receptor than dopamine
regions. An already-crowded armamentarium of therapeutic itself and antagonist/partial agonist activity [12]. Although the
options for the treatment of acute schizophrenia makes pro- consequences of such an action for patients with schizophre-
spects of success for cariprazine difficult to achieve but not nia are unknown yet, preclinical findings indicate that such a
impossible. What elements of its activity should help in mechanism can be responsible for, among others, antidepres-
achieving such success? For sure, they are not its effective- sant/anxiolytic effects. Such data may support cariprazine’s
ness, tolerability, or favorable metabolic profile, but (i) car- utility in the treatment of major depressive disorder (MDD),
iprazine’s extra ability to improve negative symptoms, (ii) its at least as adjunctive therapy, which has been confirmed by
ability to be particularly useful in young patients with a randomized, double-blind, placebo-controlled, flexible-dose
relatively short duration of the disease, (iii) cariprazine’s abil- study in adult inadequate responders to antidepressant ther-
ity to decrease substance abuse, and/or (iv) its ability to apy [97]. Cariprazine’s partial agonist activity at 5-HT1A recep-
maintain efficacy in cases of missed dose. Meta-analysis as tors may confer additional benefits in the treatment of
well as a randomized placebo-controlled, double-blind trial depression and anxiety, since these receptors have been
showed that patients receiving cariprazine demonstrated involved in the etiology of both disorders. The ability of car-
greater improvement in prominent negative symptoms com- iprazine to evoke potential antidepressant and anxiolytic activ-
pared with patients receiving placebo or risperidone, in func- ity after long-term treatment in rodent models of depression
tioning, particularly with regard to self-care, interpersonal and anxiety [81,82] adds further support for its actions.
relationships, and socially useful activities [94,95]. Therefore, Nonetheless, further characterization of the comparative
cariprazine may be the treatment of choice for patients who effectiveness of cariprazine awaits the conduct of additional
have shown improvement in positive symptoms but continue and extended clinical trials to evaluate its procognitive and
to have disabling negative symptoms [96]. The effectiveness antidepressant/anxiolytic effects in patients with schizophre-
of cariprazine in various animal models of learning and nia and other psychiatric disorders characterized by such def-
memory, cognitive, and executive functions [54,62,76] sup- icits (e.g. MDD, Alzheimer’s disease, and ADHD). Only clinical
ports its potential clinical advantage in treating cognitive findings may further translate the unique preclinical profile of
deficits in patients with schizophrenia. In the rodent and cariprazine into benefits.
human studies, cariprazine’s superior improvement in cogni-
tive and social impairments was attributed to its activity at
Funding
D3 receptors. Moreover, in those aspects of efficacy, the
active doses of cariprazine used in animal studies correlate The authors are supported by funds for their statutory activity from the
with the dose range proposed in humans. Jagiellonian University Medical College (K/ZDS/006133).
Psychostimulant abuse is frequently comorbid in patients
with schizophrenia, especially young people, contributing to
worsening the disease and increased morbidity and mortality.
Declaration of interest
Cariprazine demonstrated the reduction in substance use and
relapse prevention against cocaine rewarding effect in rodents The authors have no relevant affiliations or financial involvement with any
organization or entity with a financial interest in or financial conflict with
[88], which can be attributed to its D2 receptor partial agonist
the subject matter or materials discussed in the manuscript. This includes
activity. Whether cariprazine’s antiabuse potential, detected in employment, consultancies, honoraria, stock ownership or options, expert
rat's model, can be extrapolated to humans remains to be testimony, grants or patents received or pending, or royalties. Peer
studied, but it might provide greater insights into this topic. reviewers on this manuscript have no relevant financial or other relation-
The aforementioned meta-regression data further refined ships to disclose.
cariprazine’s clinical profile, suggesting that the medicine is
particularly useful in young patients with a relatively short
duration of the disease [95]. As epidemiologic data show References
psychotic symptoms usually emerge in men in their late
teens and early 20s and in women in their mid-20s to Papers of special note have been highlighted as either of interest (•) or of
early 30s. considerable interest (••) to readers.
Cariprazine has a half-life of 2–4 days with an active meta- ®
1. Product information Vraylar (cariprazine). Initial US approval. 2017.
Reference ID: 4060067 - FDA.
bolite DDCAR that has a terminal half-life of 2–3 weeks and
systemic exposure to it several times higher than that of the
2. Summary of product characteristics Reagila
WC500234924 EMA.
® (cariprazine).

parent compound [3–5]. It means that the effective dose 3. Meszaros GP, Kapas M, Borsos M, et al. Pharmacokinetics of RGH-
188, a new dopaminę D3/D2 antagonist/partial agonist atypical
needs to be increased for many weeks to get to the steady
antipsychotic, in healthy subjects. Eur Neuropsychopharmacol.
state and clinicians could overshoot the ideal dose. However, 2007;217:S451–2.
upon stopping cariprazine treatment, it will be many weeks 4. Kapas M, Meszaros GP, Yu B, et al. Comparison of the pharmacoki-
before the active molecules are washed out, which could be a netic behaviour of RGH-188 in schizophrenic patients and healthy
potential advantage for patients with schizophrenia volunteers. Eur Neuropsychopharmacol. 2008;1:433.
10 A. WESOŁOWSKA ET AL.

5. Citrome L. Cariprazine: chemistry, pharmacodynamics, pharmaco- profile of novel antipsychotics: frontocortical dopamine and hippo-
kinetics, and metabolism, clinical efficacy, safety, and tolerability. campal serotonin release in rat brain. J Pharmacol Exp Ther.
Expert Opinion Drug Metab Toxicol. 2013;9:193–206. 2005;315:265–272.
• Detailed review on pharmacological properties f cariprazine. 25. Baldessarini RJ, Tarazi FI. Pharmacotherapy of psychosis and mania.
6. Meszaros GP, Ágai-Csongor E, Kapas M. Sensitive LC-MS/MS meth- In: Brunton LL, Lazo JS, Parker K, editor. Goodman and Gilman’s the
ods for the quantification of RGH-188 and its active metabolites, pharmacological basis of therapeutics. New York: McGraw-Hill
desmethyl- and didesmethyl-cariprazine in human plasma and Press; 2005. p. 461–500.
urine. J Pharm Biomed Anal. 2008;48:388–397. 26. Gyertyán I, Kiss B, Sághy K, et al. Caripazine (RGH-188), a potent D3/
7. Kirschner N, Gemesi LI, Vastag M, et al. In vitro metabolism of RGH- D2 dopamine receptor partial agonist, binds to dopamine D3 recep-
188. Drug Metab Rev. 2008;40:128–129. tors in vivo and shows antipsychotic-like and precognitive effects
8. Earley W, Durgam S, Lu K, et al. Safety and tolerability of cariprazine in rodents. Neurochem Int. 2011;59:925–935.
in patients with acute exacerbation of schizophrenia: a pooled • Characterization of in vivo activity of cariprazine.
analysis of four phase II/III randomized, double-blind, placebo-con- 27. Choi YK, Adham N, Kiss B, et al. Long-term effects of cariprazine
trolled studies. Int Clin Psychopharmacol. 2017;32:319–328. exposure on dopamine receptor subtypes. CNS Spectrum.
9. Durgam S, Starace A, Li D, et al. An evaluation of the safety and efficacy f 2014;19:268–277.
cariprazine in patients with acute exacerbaton of schizophrenia: a phase • Characterization of chronic activity of cariprazine.
II, randomized clinical trial. Schizophrenia Res. 2014;152:450–457. 28. Choi YK, Adham N, Kiss B, et al. Long-term effects of aripiprazole
10. Kane JM, Zukin S, Wang Y, et al. Efficacy and safety of cariprazine in exposure on monoaminergic and glutamatergic receptor subtypes:
acute exacerbation of schizophrenia. J Clin Psychopharmacol. comparison with cariprazine. CNS Spectrum. 2017;22:484–494.
2015;35:367–373. • Preclinical comparisons of chronic effects of cariprazine and
11. Ágai-Csongor E, Domány G, Nógrádi K, et al. Discovery of caripra- aripiprazole.
zine (RGH-188): a novel antipsychotic acting on dopamine D3/D2 29. Tarazi FI, Moran-Gates T, Wong EH, et al. Differential regional and
receptors. Bioorg Med Chem Lett. 2012;22:3437–3440. dose-related effects of asenapine on dopamine receptor subtypes.
12. Kiss B, Horváth A, Némethy Z, et al. Cariprazine (RGH-188), a Pychopharmacology. 2008;198:103–111.
dopamine D3 receptor-preferring, D3/D2 dopamine receptor 30. Tarazi FI, Zhang K, Baldessarini RJ. Long-term effects of olanzapine,
antagonist-partal agonist antipsychotic candidate: in vitro and risperidone, and quetiapine on dopamine receptor types in regions
neurochemical profile. J Pharmacol Exp Ther. 2010;333:328–340. of rat brain: implications for antipsychotic drug treatment. J
•• Detailed characterization of pharmacological properties in Pharmacol Exp Ther. 2001;297:711–717.
vitro and in vivo of cariprazine. 31. Tarazi FI, Florijn WJ, Creese I. Differential regulation of dopamine
13. Newman-Tancredi A, Kleven MS. Comparative pharmacology of anti- receptors after chronic typical and atypical antipsychotic drug
psychotics possessing combined dopaminę D2 and serotonin 5-HT1A treatment. Neuroscience. 1997;78:985–996.
receptor properties. Psychopharmacology. 2011;216:451–473. 32. Tarazi FI, Yeghiayan SK, Baldessarini RJ, et al. Long-term effects of S(+)
14. Tadori Y, Forbes RA, McQuade RD, et al. In vitro pharmacology of N-n-propylnorapomorphine compared with typical and atypical anti-
aripiprazole, its metabolite and experimental dopamine partial psychotics: differential increases of cerebrocortcal D2-like and striato-
agonists at human dopamine D2 and D3 receptors. Eur J limbic D4-like dopamine receptors. Neuropsychopharmacology.
Pharmacol. 2011;668:355–365. 1997;17:186–196.
15. Arranz MJ, de Leon J. Pharmacogenetics and pharmacogenomics of 33. Deutch AY. Prefrontal cortical dopamine systems and the elabora-
schizophrenia: a review of last decade of research. Mol Psychiatry. tion of functional corticostriatal circuits: implications for schizo-
2007;12:707–747. phrenia and Parkinson’s disease. J Neural Transm. 1993;91:197–
16. De Deurwaerdère P. Cariprazine: new dopamine-biased agonist for 221.
neurpsychiatric disorders. Drug Today. 2016;52(2):1–14. 34. Marcus MM, Nomikos GG, Svensson TH. Differential actions of typical
17. Seneca N, Finnema SJ, Laszlovszky I, et al. Occupancy of dopamine D2 and atypical antipsychotic drugs on dopamine release in the core and
and D3 and serotonin 5-HT1A receptors by the novel antipsychotic drug shell of the nucleus accumbens. Eur Neuropsychopharmacol.
candidate, cariprazine (RGH-188), in monkey brain measured using 1996;6:29–38.
positron emission tomography. Psychopharmacology. 2011;218:579– 35. Citrome L. Cariprazine in schizophrenia: clinical efficacy, tolerability,
587. and place in therapy. Adv Ther. 2013;30:114–126.
18. Kiss B, Horti F, Bobok A, et al. Cariprazine, a D3/D2 dopamine 36. Citrome L. Cariprazine in bipolar disorder: clinical efficacy, toler-
receptor partial agonist antipsychotic, displays greater D3 receptor ability, and place in therapy. Adv Ther Feb. 2013;30:102–113.
accupancy in vivo compared with other antipsychotics. Biol 37. Chiodo LA, Bunney BS. Typical and atypical neuroleptics: differ-
Psychiatry. 2012;71:40S. ential effects of chronic administration on the activity of A9 and
19. Caccia S, Invernizzi RW, Nobili A, et al. A new generation of anti- A10 midbrain dopaminergic neurons. J Neurosci. 1983;3:1607–
psychotics: pharmacology and clinical utility of cariprazine in schi- 1619.
zophrenia. Ther Clin Risk Manag. 2013;9:319–328. 38. White FJ, Wang RY. Differential effects of classical and atypical
20. Girgis RR, Slifstein M, D’Souza D, et al. Preferential binding to antipsychotic drugs on A9 and A10 dopamine neurons. Science.
dopamine D3 over D2 receptors by cariprazine in patients with 1983;221:1054–1057.
schizophrenia using PET with the D3/D2 receptor ligand [11C]- 39. Adham-Parangi N, Kiss B, Gyertyán I, et al. RGH-188, a potential
(+)-PHNO. Psychopharmacology. 2016;233:3503–3512. antipsychotic with D3/D2 antagonist/partial agonist properties,
21. Kuroki T, Meltzer HY, Ichikawa J. Effects of antipsychotic drugs on exhibits an atypical antipsychotic profile in rats with respect to
extracellular dopamine levels in rat medial prefrontal cortex and effect on spontaneously active dopamine neurons. Int J
nucleus accumbens. J Pharmacol Exp Ther. 1999;288:774–781. Neuropsychopharmacol. 2008;11(Suppl 1):P–07.69.
22. Zhang W, Perry KW, Wong DT, et al. Synergistic effects of olanza- 40. Tarazi FI, Zhang K, Baldessarini RJ. Long-term effects of olanzapine,
pine and other antipsychotic agents in combination with fluoxetine risperidone, and quetiapine on serotonin 1A, 2A and 2C receptors
on norepinephrine and dopamine release in rat prefrontal cortex. in rat forebrain regions. Psychopharmacology. 2002;161:263–270.
Neuropsychopharmacology. 2000;23:250–262. 41. Tarazi FI, Moran-Gates T, Wong EH, et al. Asenapine induces differ-
23. Ichikawa J, Dai J, O’Laughlin IA, et al. Atypical, but not typical, antipsy- ential regional effects on serotonin receptor subtypes. J
chotic drugs increase cortical acetylcholine release without an effect in Pychopharmacol. 2010;24:341–348.
the nucleus accumbens or striatum. Neuropsychopharmacology. 42. Francis PT, Pangalos JM, Pearson RC, et al. 5-Hydroxytryptamine1A
2002;26:325–339. but not 5-hydroxytryptamine2 receptors are enriched on neocorti-
24. Assié MB, Ravailhe V, Faucillon V, et al. Contrasting contribution of cal pyramidal neurons destroyed by intrastriatal volkensin. J
5-hydroxytryptamine 1A receptor activation to neurochemical Pharmacol Exp Ther. 1992;261:1273–1281.
EXPERT OPINION ON DRUG DISCOVERY 11

43. Tarazi FI, Baldessarini J, Kula NS, et al. Long-term effects of olanza- • Characterization of cariprazine’s action in models of negative
pine, risperidone, and quetiapine ionotropic glutamate receptor symptoms of schizophrenia.
types: implications for antipsychotic drug treatment. J Pharmacol 63. Barnes SA, Sawiak SJ, Caprioli D, et al. Impaired limbic cortico-
Exp Ther. 2003;306:1145–1151. striatal structure and sustained visual attention in a rodent model
44. Tarazi FI, Choi YK, Gardner M, et al. Asenapine exerts distinctive of schizophrenia. Int J Neuropsychopharmacol. 2014;18:1–12.
regional effects on ionotropic glutamate receptor subtypes in rat 64. Bubenikova-Valesova V, Horacek J, Vrajova M, et al. Models of
brain. Synapse. 2009;63:413–420. schizophrenia in humans and animals based on inhibition of
45. Spurney CF, Baca SM, Murray AM, et al. Differentia effects of NMDA receptors. Neurosci Biobehav Rev. 2008;32:1014–1023.
haloperidol and clozapine on ionotropic glutamate receptors in 65. Mouri A, Noda Y, Enomoto T, et al. Phencyclidine animal models of
rats. Synapse. 1999;34:266–276. schizophrenia: approaches from abnormality of glutamatergic neu-
46. Aghajanian GK, Marek GJ. Serotonin model of schizophrenia: emer- rotransmission and neurodevelopment. Neurochem Int.
ging role of glutamate mechanisms. Brain Res Rev. 2000;31:302– 2007;51:173–184.
312. •• Review on design and development animal models based on
47. Carlsson A, Waters N, Holm-Waters S, et al. Interactions between neurodevelopment hypothesis of schizophrenia.
monoamines, glutamate, and GABA in schizophrenia: new evi- 66. McIntosh AL, Ballard TM, Steward LJ, et al. Clozapine reverses
dence. Ann Rev Pharmacol Toxicol. 2001;41:237–260. recognition memory but not social interaction deficits in a “dual-
48. Gao X, Sakai K, Roberts RC, et al. Ionotropic glutamate receptors hit” rat model of schizophrenia. Schizophr Res. 2012;136:S282.
and expression of N-methyl-d-aspartate receptor subunits in sub- 67. Goosens KA, Maren S. Contextual and auditory fear conditioning ae
regions of human hippocampus: effects of schizophrenia. Am J mediated by the lateral, basal, and central amygdaloid nuclei in
Psychiatry. 2000;157:1141–1149. rats. Learn Mem. 2001;8:148–155.
49. Ariano M, Larson ER, Noblett KL, et al. Coexpression of striatal 68. Cools R, Brouwer WH, de Jong R, et al. Flexibility, inhibition, and
dopamine receptor subtypes and excitatory amino acid subunits. planning: frontal dysfunctioning in schizophrenia. Brain Cogn.
Synapse. 1997;26:400–414. 2000;43:108–112.
50. Moore NA, Axton MS. The role of multiple dopamine receptors in 69. Jacobs R, Anderson V. Planning and problem solving skills follow-
apomorphine and N-n-propylnorapomorphine-induced climbing ing focal frontal brain lesions in childhood: analysis using the
and hypothermia. Eur J Pharmcol. 1990;178:195–201. Tower of London. Child Neuropsychol. 2002;8:93–106.
51. Fabricius K, Helboe L, Fink-Jensen A, et al. Pharmacological char- 70. Shamay-Tsoory SG, Tomer R, Goldsher D, et al. Impairment in
acterization of social isolaton-induced hyperactivity. cognitive and affective empathy in patients with brain lesions:
Psychopharmacology. 2010;215:257–266. anatomical and cognitive correlates. J Clin Exp Neuropsychol.
52. Fone KCF, Porkess MV. Behavioural and neurochemical effects of 2004;26:1113–1127.
post-weaning social isolation in rodents-relevance to developmen- 71. Bowie CR, Harvey PD. Treatment of cognitive deficits in schizophre-
tal neuropsychiatric disorders. Neurosci Biobehav Rev. nia. Curr Opin Investig. 2006;7:608–613.
2008;32:1087–1102. 72. Elliott R, McKenna PJ, Robbins TW, et al. Neuropsychological evi-
53. McIntosh AL, Ballard TM, Steward LJ, et al. The atypical antipsycho- dence for frontostriatal dysfunction in schizophrenia. Psychol Med.
tic risperidone reverses the recognition memory deficits induced 1995;25:619–630.
by post-weaning social isolation in rats. Psychopharmacology. 73. Leeson VC, Robbins TW, Matheson E, et al. Discrimination learning,
2013;228:31–42. reversal, and set-shifting in first-episode schizophrenia: stability
54. Watson DJG, King MV, Gyrtyán I, et al. The dopamine D3-preferring over six years and specific associations with medication type and
D2/D3 dopamine receptor partial agonist, cariprazine, reverses disorganization syndrome. Biol Psychiatry. 2009;66:586–593.
behavioural changes in a rat neurodevelopmental model for schi- 74. Pantelis C, Barber FZ, Barnes TR, et al. Comparison of set-shifting
zophrenia. Eur Neuropsychopharmacol. 2016;26:208–224. ability in patients with chronic schizophrenia and frontal lobe
• Characterization and comparison of cariprazine’s activity with damage. Schizophr Res. 1999;37:251–270.
aripiprazole’s effects in animal model of schizophrenia. 75. Bissonette GB, Powell EM. Reversal learning and attentional set-
55. Castagne´ V, Moser PC, Porsolt RD. Preclinical behavioral models for shifting in Mice. Neuropharmacology. 2012;62:1168–1174.
predicting antipsychotic activity. Adv Pharmacol. 2009;57:381–418. 76. Zimnisky R, Chang G, Gyertyán I, et al. Cariprazine, a dopamine D3-
56. Leriche L, Schwartz JC, Sokoloff P. The dopamine D3 receptor receptor-preferring partial agonist, blocks phencyclidine-induced
mediates locomotor hyperactivity induced by NMDA receptor impairments of working memory, attention set-shifting, and recog-
blockade. Neuropharmacology. 2003;45:174–181. nition memory in the mouse. Psychopharmacology. 2013;226:91–
57. Kapur S, Mizrahi, Li M. From dopamine to salience to psychosis – 100.
linking biology, pharmacology and phenomenology of psychosis. • Characterization of procognitive properties of cariprazine.
Schizophr Res. 2005;79:59–68. 77. Richmond MA, Yee BK, Pouzet B, et al. Dissociating context and
58. Natesan S, Reckless GE, Nobrega JN, et al. Dissociation between in space within the hippocampus: effects of complete, dorsal, and
vivo occupancy and functional antagonism of dopamine D2 recep- ventral excitotoxic hippocampal lesions on conditioned freezing
tors: comparing aripiprazole to other antipsychotics in animal and spatial learning. Behav Neurosci. 1999;113:1189–1203.
models. Neuropsychopharmacology. 2006;31:1854–1863. 78. Der-Avakian A, Markou A. The neurobiology of anhedonia and
59. Neill JC, Barns S, Cook S, et al. Animal models of cognitive dysfunc- other reward-related deficits. Trends Neurosci. 2012;35:68–77.
tion and negative symptoms of schizophrenia: focus on NMDA 79. Willner P. The chronic mild stress model of depression: history,
receptor antagonism. Pharmacol Ther. 2010;128:419–432. evaluation and usage. Neurobiol Stress. 2017;6:7–93.
60. Hertzmann M, Reba RC, Kotlyarov EV. Single photon emission •• Historical review on design and development of chronic mild
computed tomography in phencyclidine and related drug abuse. stress model of depression.
Am J Psychiatry. 1990;147:255–256. 80. Papp M. Models of affective illness: chronic mild stress in the rat.
61. Jentsch JD, Roth RH. The neuropsychopharmacology of phencycli- Curr Protoc Pharmacol. 2012.;57:5.9.1–5.9.11.
dine: from NMDA receptor hypofunction to the dopamine hypoth- 81. Papp M, Gruca P, Lasoń-Tyburkiewicz, et al. Attenuation of anhe-
esis of schizophrenia. Neuropsychopharmacology. 1999;20:201– donia by cariprazine in the chronic mild stress model of depression.
225. Behav Pharmacol. 2014;25:567–574.
62. Neill JC, Graysoon B, Kiss B, et al. Effects of cariprazine, a novel anti- 82. Duric V, Banasr M, Franklin T, et al. Cariprazine exhibits anxiolytic
psychotic, on cognitive and negative symptoms in a rodent model of and dopamine D3 receptor-dependent antidepressant effects in
schizophrenia symptomatology. Eur Neuropsychopharmacol. the chronic stress model. Int J Neuropsychopharmacol.
2016;26:3–14. 2017;20:78–96.
12 A. WESOŁOWSKA ET AL.

83. Bersani G, Pesaresi L, Orlandi V, et al. Atypical antipsychotics and •• Comparison of in vitro mechanism of cariprazine with aripi-
polydipsia: a cause or a treatment? Hum Psychopharmacol. prazole and brexpiprazole.
2007;22:103–107. 91. Auclair AL, Cathala A, Sarrazin F, et al. The central serotonin 2B recep-
84. Meulendijks D, Mannesse CK, Jansen PA, et al. Antipsychotic- tor: a new pharmacological target to modulate the mesoaccumbens
induced hyponatraemia: a systematic review of the published evi- dopaminergic pathway activity. J Neurochem. 2010;114(5):1323–1332.
dence. Drug Saf. 2010;33:101–114. 92. Pitychoutis P, Belmer A, Moutkine I, et al. Mice lacking the serotonin
85. Herman L, Hougland T, El-Mallakh RS. Mimicking human bipolar ion Htr2B receptor gene present an antipsychotic-sensitive schizophrenic-
dysregulation models mania in rats. Neurosci Biobehav Rev. like phenotype. Neuropsychopharmacology. 2015;40(12):2764–2773.
2007;31:874–881. 93. Celada P, Puig M, Amargós-Bosch M, et al. The therapeutic role of
86. Gao Y, Peterson S, Masri B, et al. Cariprazine exerts antimanic properties 5-HT1A and 5-HT2A receptors in depression. J Psychiatry Neurosci.
and interferes with dopamine D2 receptor β-arrestin interactions. 2004;29:252–265.
Pharmacol Res Perspect. 2014;3:1–10. 94. Németh G, Laszlovszky I, Czobor P, et al. Cariprazine versus risper-
87. Yildiz A, Vieta E, Leucht S, et al. Efficacy of antimanic treatments: a meta- idone monotherapy for treatment of predominant negative symp-
analysis of randomized, controlled trials. Neuropsychopharmacology. toms in patients with schizophrenia: a randomised, double-blind,
2011;36:375–389. controlled trial. Lancet. 2017;389:1103–1113.
88. Román V, Gyertyán I, Sághy K, et al. Cariprazine (RGH-188), a D3- 95. Corponi F, Serretti A, Montgomery S, et al. Cariprazine specificity
preferring dopamine D3/D2 receptor partial agonist antipsychotic profile in the treatment of acute schizophrenia: a meta-analysis and
candidate demonstrates anti-abuse potential in rats. meta-regression of randomized-controlled trials. Int Clin
Psychopharmacology. 2013;226:285–293. Psychopharmacol. 2017;32:309–318.
• Characterization of anti-abuse activity of cariprazine. 96. Scarff JR. The prospects of cariprazine in the treatment of schizo-
89. Citrome L. Cariprazine for the treatment of schizophrenia: a review phrenia. Ther Adv Psychopharmacol. 2017;7:237–239.
of this dopamine D3-preferring D3/D2 receptor partial agonist. Clin • Synthetic review of pharmacology and clinical efficacy/toler-
Schizophr Relat Psychoses. 2016;10(2):109–119. ability of cariprazine.
•• Recent review considering how pharmacological properties 97. Durgam S, Earley W, Guo H, et al. Efficacy and safety of adjunctive
translate into clinical efficacy and safety. cariprazine in inadequate responders to antidepressants: a rando-
90. Stahl SM. Mechanism of action of cariprazine. CNS Spectrums. mized, double-blind, placebo-controlled study in adult patients
2016;21(2):123–127. with major depressive disorder. J Clin Psychiatry. 2016;77:371–378.

You might also like