458
Views
2
CrossRef citations to date
0
Altmetric
Drug Discovery Case History

The preclinical discovery and development of cariprazine for the treatment of schizophrenia

, , &
Pages 779-790 | Received 01 Feb 2018, Accepted 26 Apr 2018, Published online: 03 May 2018

References

  • Product information Vraylar® (cariprazine). Initial US approval. 2017. Reference ID: 4060067 - FDA.
  • Summary of product characteristics Reagila® (cariprazine). WC500234924 EMA.
  • Meszaros GP, Kapas M, Borsos M, et al. Pharmacokinetics of RGH-188, a new dopaminę D3/D2 antagonist/partial agonist atypical antipsychotic, in healthy subjects. Eur Neuropsychopharmacol. 2007;217:S451–2.
  • Kapas M, Meszaros GP, Yu B, et al. Comparison of the pharmacokinetic behaviour of RGH-188 in schizophrenic patients and healthy volunteers. Eur Neuropsychopharmacol. 2008;1:433.
  • Citrome L. Cariprazine: chemistry, pharmacodynamics, pharmacokinetics, and metabolism, clinical efficacy, safety, and tolerability. Expert Opinion Drug Metab Toxicol. 2013;9:193–206.
  • Meszaros GP, Ágai-Csongor E, Kapas M. Sensitive LC-MS/MS methods for the quantification of RGH-188 and its active metabolites, desmethyl- and didesmethyl-cariprazine in human plasma and urine. J Pharm Biomed Anal. 2008;48:388–397.
  • Kirschner N, Gemesi LI, Vastag M, et al. In vitro metabolism of RGH-188. Drug Metab Rev. 2008;40:128–129.
  • Earley W, Durgam S, Lu K, et al. Safety and tolerability of cariprazine in patients with acute exacerbation of schizophrenia: a pooled analysis of four phase II/III randomized, double-blind, placebo-controlled studies. Int Clin Psychopharmacol. 2017;32:319–328.
  • Durgam S, Starace A, Li D, et al. An evaluation of the safety and efficacy f cariprazine in patients with acute exacerbaton of schizophrenia: a phase II, randomized clinical trial. Schizophrenia Res. 2014;152:450–457.
  • Kane JM, Zukin S, Wang Y, et al. Efficacy and safety of cariprazine in acute exacerbation of schizophrenia. J Clin Psychopharmacol. 2015;35:367–373.
  • Ágai-Csongor E, Domány G, Nógrádi K, et al. Discovery of cariprazine (RGH-188): a novel antipsychotic acting on dopamine D3/D2 receptors. Bioorg Med Chem Lett. 2012;22:3437–3440.
  • Kiss B, Horváth A, Némethy Z, et al. Cariprazine (RGH-188), a dopamine D3 receptor-preferring, D3/D2 dopamine receptor antagonist-partal agonist antipsychotic candidate: in vitro and neurochemical profile. J Pharmacol Exp Ther. 2010;333:328–340.
  • Newman-Tancredi A, Kleven MS. Comparative pharmacology of antipsychotics possessing combined dopaminę D2 and serotonin 5-HT1A receptor properties. Psychopharmacology. 2011;216:451–473.
  • Tadori Y, Forbes RA, McQuade RD, et al. In vitro pharmacology of aripiprazole, its metabolite and experimental dopamine partial agonists at human dopamine D2 and D3 receptors. Eur J Pharmacol. 2011;668:355–365.
  • Arranz MJ, de Leon J. Pharmacogenetics and pharmacogenomics of schizophrenia: a review of last decade of research. Mol Psychiatry. 2007;12:707–747.
  • De Deurwaerdère P. Cariprazine: new dopamine-biased agonist for neurpsychiatric disorders. Drug Today. 2016;52(2):1–14.
  • Seneca N, Finnema SJ, Laszlovszky I, et al. Occupancy of dopamine D2 and D3 and serotonin 5-HT1A receptors by the novel antipsychotic drug candidate, cariprazine (RGH-188), in monkey brain measured using positron emission tomography. Psychopharmacology. 2011;218:579–587.
  • Kiss B, Horti F, Bobok A, et al. Cariprazine, a D3/D2 dopamine receptor partial agonist antipsychotic, displays greater D3 receptor accupancy in vivo compared with other antipsychotics. Biol Psychiatry. 2012;71:40S.
  • Caccia S, Invernizzi RW, Nobili A, et al. A new generation of antipsychotics: pharmacology and clinical utility of cariprazine in schizophrenia. Ther Clin Risk Manag. 2013;9:319–328.
  • Girgis RR, Slifstein M, D’Souza D, et al. Preferential binding to dopamine D3 over D2 receptors by cariprazine in patients with schizophrenia using PET with the D3/D2 receptor ligand [11C]-(+)-PHNO. Psychopharmacology. 2016;233:3503–3512.
  • Kuroki T, Meltzer HY, Ichikawa J. Effects of antipsychotic drugs on extracellular dopamine levels in rat medial prefrontal cortex and nucleus accumbens. J Pharmacol Exp Ther. 1999;288:774–781.
  • Zhang W, Perry KW, Wong DT, et al. Synergistic effects of olanzapine and other antipsychotic agents in combination with fluoxetine on norepinephrine and dopamine release in rat prefrontal cortex. Neuropsychopharmacology. 2000;23:250–262.
  • Ichikawa J, Dai J, O’Laughlin IA, et al. Atypical, but not typical, antipsychotic drugs increase cortical acetylcholine release without an effect in the nucleus accumbens or striatum. Neuropsychopharmacology. 2002;26:325–339.
  • Assié MB, Ravailhe V, Faucillon V, et al. Contrasting contribution of 5-hydroxytryptamine 1A receptor activation to neurochemical profile of novel antipsychotics: frontocortical dopamine and hippocampal serotonin release in rat brain. J Pharmacol Exp Ther. 2005;315:265–272.
  • Baldessarini RJ, Tarazi FI. Pharmacotherapy of psychosis and mania. In: Brunton LL, Lazo JS, Parker K, editor. Goodman and Gilman’s the pharmacological basis of therapeutics. New York: McGraw-Hill Press; 2005. p. 461–500.
  • Gyertyán I, Kiss B, Sághy K, et al. Caripazine (RGH-188), a potent D3/D2 dopamine receptor partial agonist, binds to dopamine D3 receptors in vivo and shows antipsychotic-like and precognitive effects in rodents. Neurochem Int. 2011;59:925–935.
  • Choi YK, Adham N, Kiss B, et al. Long-term effects of cariprazine exposure on dopamine receptor subtypes. CNS Spectrum. 2014;19:268–277.
  • Choi YK, Adham N, Kiss B, et al. Long-term effects of aripiprazole exposure on monoaminergic and glutamatergic receptor subtypes: comparison with cariprazine. CNS Spectrum. 2017;22:484–494.
  • Tarazi FI, Moran-Gates T, Wong EH, et al. Differential regional and dose-related effects of asenapine on dopamine receptor subtypes. Pychopharmacology. 2008;198:103–111.
  • Tarazi FI, Zhang K, Baldessarini RJ. Long-term effects of olanzapine, risperidone, and quetiapine on dopamine receptor types in regions of rat brain: implications for antipsychotic drug treatment. J Pharmacol Exp Ther. 2001;297:711–717.
  • Tarazi FI, Florijn WJ, Creese I. Differential regulation of dopamine receptors after chronic typical and atypical antipsychotic drug treatment. Neuroscience. 1997;78:985–996.
  • Tarazi FI, Yeghiayan SK, Baldessarini RJ, et al. Long-term effects of S(+)N-n-propylnorapomorphine compared with typical and atypical antipsychotics: differential increases of cerebrocortcal D2-like and striatolimbic D4-like dopamine receptors. Neuropsychopharmacology. 1997;17:186–196.
  • Deutch AY. Prefrontal cortical dopamine systems and the elaboration of functional corticostriatal circuits: implications for schizophrenia and Parkinson’s disease. J Neural Transm. 1993;91:197–221.
  • Marcus MM, Nomikos GG, Svensson TH. Differential actions of typical and atypical antipsychotic drugs on dopamine release in the core and shell of the nucleus accumbens. Eur Neuropsychopharmacol. 1996;6:29–38.
  • Citrome L. Cariprazine in schizophrenia: clinical efficacy, tolerability, and place in therapy. Adv Ther. 2013;30:114–126.
  • Citrome L. Cariprazine in bipolar disorder: clinical efficacy, tolerability, and place in therapy. Adv Ther Feb. 2013;30:102–113.
  • Chiodo LA, Bunney BS. Typical and atypical neuroleptics: differential effects of chronic administration on the activity of A9 and A10 midbrain dopaminergic neurons. J Neurosci. 1983;3:1607–1619.
  • White FJ, Wang RY. Differential effects of classical and atypical antipsychotic drugs on A9 and A10 dopamine neurons. Science. 1983;221:1054–1057.
  • Adham-Parangi N, Kiss B, Gyertyán I, et al. RGH-188, a potential antipsychotic with D3/D2 antagonist/partial agonist properties, exhibits an atypical antipsychotic profile in rats with respect to effect on spontaneously active dopamine neurons. Int J Neuropsychopharmacol. 2008;11(Suppl 1):P–07.69.
  • Tarazi FI, Zhang K, Baldessarini RJ. Long-term effects of olanzapine, risperidone, and quetiapine on serotonin 1A, 2A and 2C receptors in rat forebrain regions. Psychopharmacology. 2002;161:263–270.
  • Tarazi FI, Moran-Gates T, Wong EH, et al. Asenapine induces differential regional effects on serotonin receptor subtypes. J Pychopharmacol. 2010;24:341–348.
  • Francis PT, Pangalos JM, Pearson RC, et al. 5-Hydroxytryptamine1A but not 5-hydroxytryptamine2 receptors are enriched on neocortical pyramidal neurons destroyed by intrastriatal volkensin. J Pharmacol Exp Ther. 1992;261:1273–1281.
  • Tarazi FI, Baldessarini J, Kula NS, et al. Long-term effects of olanzapine, risperidone, and quetiapine ionotropic glutamate receptor types: implications for antipsychotic drug treatment. J Pharmacol Exp Ther. 2003;306:1145–1151.
  • Tarazi FI, Choi YK, Gardner M, et al. Asenapine exerts distinctive regional effects on ionotropic glutamate receptor subtypes in rat brain. Synapse. 2009;63:413–420.
  • Spurney CF, Baca SM, Murray AM, et al. Differentia effects of haloperidol and clozapine on ionotropic glutamate receptors in rats. Synapse. 1999;34:266–276.
  • Aghajanian GK, Marek GJ. Serotonin model of schizophrenia: emerging role of glutamate mechanisms. Brain Res Rev. 2000;31:302–312.
  • Carlsson A, Waters N, Holm-Waters S, et al. Interactions between monoamines, glutamate, and GABA in schizophrenia: new evidence. Ann Rev Pharmacol Toxicol. 2001;41:237–260.
  • Gao X, Sakai K, Roberts RC, et al. Ionotropic glutamate receptors and expression of N-methyl-d-aspartate receptor subunits in subregions of human hippocampus: effects of schizophrenia. Am J Psychiatry. 2000;157:1141–1149.
  • Ariano M, Larson ER, Noblett KL, et al. Coexpression of striatal dopamine receptor subtypes and excitatory amino acid subunits. Synapse. 1997;26:400–414.
  • Moore NA, Axton MS. The role of multiple dopamine receptors in apomorphine and N-n-propylnorapomorphine-induced climbing and hypothermia. Eur J Pharmcol. 1990;178:195–201.
  • Fabricius K, Helboe L, Fink-Jensen A, et al. Pharmacological characterization of social isolaton-induced hyperactivity. Psychopharmacology. 2010;215:257–266.
  • Fone KCF, Porkess MV. Behavioural and neurochemical effects of post-weaning social isolation in rodents-relevance to developmental neuropsychiatric disorders. Neurosci Biobehav Rev. 2008;32:1087–1102.
  • McIntosh AL, Ballard TM, Steward LJ, et al. The atypical antipsychotic risperidone reverses the recognition memory deficits induced by post-weaning social isolation in rats. Psychopharmacology. 2013;228:31–42.
  • Watson DJG, King MV, Gyrtyán I, et al. The dopamine D3-preferring D2/D3 dopamine receptor partial agonist, cariprazine, reverses behavioural changes in a rat neurodevelopmental model for schizophrenia. Eur Neuropsychopharmacol. 2016;26:208–224.
  • Castagne´ V, Moser PC, Porsolt RD. Preclinical behavioral models for predicting antipsychotic activity. Adv Pharmacol. 2009;57:381–418.
  • Leriche L, Schwartz JC, Sokoloff P. The dopamine D3 receptor mediates locomotor hyperactivity induced by NMDA receptor blockade. Neuropharmacology. 2003;45:174–181.
  • Kapur S, Mizrahi, Li M. From dopamine to salience to psychosis – linking biology, pharmacology and phenomenology of psychosis. Schizophr Res. 2005;79:59–68.
  • Natesan S, Reckless GE, Nobrega JN, et al. Dissociation between in vivo occupancy and functional antagonism of dopamine D2 receptors: comparing aripiprazole to other antipsychotics in animal models. Neuropsychopharmacology. 2006;31:1854–1863.
  • Neill JC, Barns S, Cook S, et al. Animal models of cognitive dysfunction and negative symptoms of schizophrenia: focus on NMDA receptor antagonism. Pharmacol Ther. 2010;128:419–432.
  • Hertzmann M, Reba RC, Kotlyarov EV. Single photon emission computed tomography in phencyclidine and related drug abuse. Am J Psychiatry. 1990;147:255–256.
  • Jentsch JD, Roth RH. The neuropsychopharmacology of phencyclidine: from NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia. Neuropsychopharmacology. 1999;20:201–225.
  • Neill JC, Graysoon B, Kiss B, et al. Effects of cariprazine, a novel antipsychotic, on cognitive and negative symptoms in a rodent model of schizophrenia symptomatology. Eur Neuropsychopharmacol. 2016;26:3–14.
  • Barnes SA, Sawiak SJ, Caprioli D, et al. Impaired limbic cortico-striatal structure and sustained visual attention in a rodent model of schizophrenia. Int J Neuropsychopharmacol. 2014;18:1–12.
  • Bubenikova-Valesova V, Horacek J, Vrajova M, et al. Models of schizophrenia in humans and animals based on inhibition of NMDA receptors. Neurosci Biobehav Rev. 2008;32:1014–1023.
  • Mouri A, Noda Y, Enomoto T, et al. Phencyclidine animal models of schizophrenia: approaches from abnormality of glutamatergic neurotransmission and neurodevelopment. Neurochem Int. 2007;51:173–184.
  • McIntosh AL, Ballard TM, Steward LJ, et al. Clozapine reverses recognition memory but not social interaction deficits in a “dual-hit” rat model of schizophrenia. Schizophr Res. 2012;136:S282.
  • Goosens KA, Maren S. Contextual and auditory fear conditioning ae mediated by the lateral, basal, and central amygdaloid nuclei in rats. Learn Mem. 2001;8:148–155.
  • Cools R, Brouwer WH, de Jong R, et al. Flexibility, inhibition, and planning: frontal dysfunctioning in schizophrenia. Brain Cogn. 2000;43:108–112.
  • Jacobs R, Anderson V. Planning and problem solving skills following focal frontal brain lesions in childhood: analysis using the Tower of London. Child Neuropsychol. 2002;8:93–106.
  • Shamay-Tsoory SG, Tomer R, Goldsher D, et al. Impairment in cognitive and affective empathy in patients with brain lesions: anatomical and cognitive correlates. J Clin Exp Neuropsychol. 2004;26:1113–1127.
  • Bowie CR, Harvey PD. Treatment of cognitive deficits in schizophrenia. Curr Opin Investig. 2006;7:608–613.
  • Elliott R, McKenna PJ, Robbins TW, et al. Neuropsychological evidence for frontostriatal dysfunction in schizophrenia. Psychol Med. 1995;25:619–630.
  • Leeson VC, Robbins TW, Matheson E, et al. Discrimination learning, reversal, and set-shifting in first-episode schizophrenia: stability over six years and specific associations with medication type and disorganization syndrome. Biol Psychiatry. 2009;66:586–593.
  • Pantelis C, Barber FZ, Barnes TR, et al. Comparison of set-shifting ability in patients with chronic schizophrenia and frontal lobe damage. Schizophr Res. 1999;37:251–270.
  • Bissonette GB, Powell EM. Reversal learning and attentional set-shifting in Mice. Neuropharmacology. 2012;62:1168–1174.
  • Zimnisky R, Chang G, Gyertyán I, et al. Cariprazine, a dopamine D3-receptor-preferring partial agonist, blocks phencyclidine-induced impairments of working memory, attention set-shifting, and recognition memory in the mouse. Psychopharmacology. 2013;226:91–100.
  • Richmond MA, Yee BK, Pouzet B, et al. Dissociating context and space within the hippocampus: effects of complete, dorsal, and ventral excitotoxic hippocampal lesions on conditioned freezing and spatial learning. Behav Neurosci. 1999;113:1189–1203.
  • Der-Avakian A, Markou A. The neurobiology of anhedonia and other reward-related deficits. Trends Neurosci. 2012;35:68–77.
  • Willner P. The chronic mild stress model of depression: history, evaluation and usage. Neurobiol Stress. 2017;6:7–93.
  • Papp M. Models of affective illness: chronic mild stress in the rat. Curr Protoc Pharmacol. 2012.;57:5.9.1–5.9.11.
  • Papp M, Gruca P, Lasoń-Tyburkiewicz, et al. Attenuation of anhedonia by cariprazine in the chronic mild stress model of depression. Behav Pharmacol. 2014;25:567–574.
  • Duric V, Banasr M, Franklin T, et al. Cariprazine exhibits anxiolytic and dopamine D3 receptor-dependent antidepressant effects in the chronic stress model. Int J Neuropsychopharmacol. 2017;20:78–96.
  • Bersani G, Pesaresi L, Orlandi V, et al. Atypical antipsychotics and polydipsia: a cause or a treatment? Hum Psychopharmacol. 2007;22:103–107.
  • Meulendijks D, Mannesse CK, Jansen PA, et al. Antipsychotic-induced hyponatraemia: a systematic review of the published evidence. Drug Saf. 2010;33:101–114.
  • Herman L, Hougland T, El-Mallakh RS. Mimicking human bipolar ion dysregulation models mania in rats. Neurosci Biobehav Rev. 2007;31:874–881.
  • Gao Y, Peterson S, Masri B, et al. Cariprazine exerts antimanic properties and interferes with dopamine D2 receptor β-arrestin interactions. Pharmacol Res Perspect. 2014;3:1–10.
  • Yildiz A, Vieta E, Leucht S, et al. Efficacy of antimanic treatments: a meta-analysis of randomized, controlled trials. Neuropsychopharmacology. 2011;36:375–389.
  • Román V, Gyertyán I, Sághy K, et al. Cariprazine (RGH-188), a D3-preferring dopamine D3/D2 receptor partial agonist antipsychotic candidate demonstrates anti-abuse potential in rats. Psychopharmacology. 2013;226:285–293.
  • Citrome L. Cariprazine for the treatment of schizophrenia: a review of this dopamine D3-preferring D3/D2 receptor partial agonist. Clin Schizophr Relat Psychoses. 2016;10(2):109–119.
  • Stahl SM. Mechanism of action of cariprazine. CNS Spectrums. 2016;21(2):123–127.
  • Auclair AL, Cathala A, Sarrazin F, et al. The central serotonin 2B receptor: a new pharmacological target to modulate the mesoaccumbens dopaminergic pathway activity. J Neurochem. 2010;114(5):1323–1332.
  • Pitychoutis P, Belmer A, Moutkine I, et al. Mice lacking the serotonin Htr2B receptor gene present an antipsychotic-sensitive schizophrenic-like phenotype. Neuropsychopharmacology. 2015;40(12):2764–2773.
  • Celada P, Puig M, Amargós-Bosch M, et al. The therapeutic role of 5-HT1A and 5-HT2A receptors in depression. J Psychiatry Neurosci. 2004;29:252–265.
  • Németh G, Laszlovszky I, Czobor P, et al. Cariprazine versus risperidone monotherapy for treatment of predominant negative symptoms in patients with schizophrenia: a randomised, double-blind, controlled trial. Lancet. 2017;389:1103–1113.
  • Corponi F, Serretti A, Montgomery S, et al. Cariprazine specificity profile in the treatment of acute schizophrenia: a meta-analysis and meta-regression of randomized-controlled trials. Int Clin Psychopharmacol. 2017;32:309–318.
  • Scarff JR. The prospects of cariprazine in the treatment of schizophrenia. Ther Adv Psychopharmacol. 2017;7:237–239.
  • Durgam S, Earley W, Guo H, et al. Efficacy and safety of adjunctive cariprazine in inadequate responders to antidepressants: a randomized, double-blind, placebo-controlled study in adult patients with major depressive disorder. J Clin Psychiatry. 2016;77:371–378.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.