Page last updated: 2024-11-12

cariprazine

Description Research Excerpts Clinical Trials Roles Classes Pathways Study Profile Bioassays Related Drugs Related Conditions Protein Interactions Research Growth Market Indicators

Description

cariprazine: Structure in first source [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

cariprazine : An N-alkylpiperazine that is N,N-dimethyl-N'-{trans-4-[2-(piperazin-1-yl)ethyl]cyclohexyl}urea substituted at position 4 on the piperazine ring by a 2,3-dichlorophenyl group. Used (as the hydrochloride salt) for treatment of schizophrenia and bipolar disorder. [Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Cross-References

ID SourceID
PubMed CID11154555
CHEMBL ID2028019
CHEMBL ID3085826
CHEBI ID90933
SCHEMBL ID184342
SCHEMBL ID184343
SCHEMBL ID7861573
SCHEMBL ID22946354
MeSH IDM0527151

Synonyms (76)

Synonym
FT-0664381
D09997
839712-12-8
cariprazine (usan/inn)
cariprazine
cariprazine [usan:inn]
rgh-188
unii-f6rjl8b278
mp-214
f6rjl8b278 ,
urea, n'-(trans-4-(2-(4-(2,3-dichlorophenyl)-1-piperazinyl)ethyl)cyclohexyl)-n,n-dimethyl-
rgh 188
hsdb 8310
mp 214
bdbm50382290
CHEMBL2028019
fri-7000188
ged-129
n'-[trans-4-[2-[4-(2,3-dichlorophenyl)-1-piperazinyl]ethyl]cyclohexyl]-n,n-dimethylurea
bdbm50443101
bdbm50443094
cariprazine [usan]
cariprazine [inn]
cariprazine [who-dd]
3-(trans-4-(2-(4-(2,3-dichlorophenyl)piperazin-1-yl)ethyl)cyclohexyl)-1,1-dimethylurea
cariprazine [mi]
KPWSJANDNDDRMB-QAQDUYKDSA-N
trans-1-{4-[2-[4-(2,3-dichlorophenyl)-piperazin-1-yl]-ethyl]-cyclohexyl}-3,3-dimethyl-urea
HY-14763
CS-1569
gtpl7671
3-[4-[2-[4-(2,3-dichlorophenyl)piperazin-1-yl]ethyl]cyclohexyl]-1,1-dimethylurea
reagila
SCHEMBL184342
SCHEMBL184343
CHEMBL3085826 ,
CHEBI:90933 ,
n'-(trans-4-{2-[4-(2,3-dichlorophenyl)piperazin-1-yl]ethyl}cyclohexyl)-n,n-dimethylurea
DTXSID80232867 ,
cariprazine(rgh188)
SCHEMBL7861573
AC-35342
trans-n-{4-[2-[4-(2,3-dichlorophenyl)piperazine-1-yl]ethyl]cyclohexyl}-n',n'-dimethylurea hydrochloride
DB06016
AKOS027250814
AS-74411
3,3-dimethyl-1-[(1r,4r)-4-{2-[4-(2,3-dichlorophenyl)piperazin-1-yl]ethyl}cyclohexyl]urea
C76682
3-((1r,4r)-4-(2-(4-(2,3-dichlorophenyl)piperazin-1-yl)ethyl)cyclohexyl)-1,1-dimethylurea
NCGC00379014-02
bdbm263449
us9550741, rgh-188
cariprazine (rgh-188)
A857938
Q2938837
trans-n-[4-[2-[4-(2,3-di-chlorophenyl)piperazin-1-yl]ethyl]cyclohexyl]-n',n'-dimethylurea
rgh-188;mp-214
rgh-188; rgh188; rgh 188; mp-214; mp 214; mp214
BCP14691
rgh-188; cariprazine
EX-A1644-1
urea, n'-[trans-4-[2-[4-(2,3-dichlorophenyl)-1-piperazinyl]ethyl]cyclohexyl]-n,n -dimethyl-
SB16839
3-(cis-4-(2-(4-(2,3-dichlorophenyl)piperazin-1-yl)ethyl)-cyclohexyl)-1,1-dimethylurea
cis-cariprazine
NCGC00379014-01
3-(trans-4-(2-(4-(2,3-dichlorophenyl)piperazin-1-yl)ethyl)-cyclohexyl)-1,1-dimethylurea
trans-cariprazine
NCGC00379014-03
SCHEMBL22946354
7ru ,
3-[4-[2-[4-[2,3-bis(chloranyl)phenyl]piperazin-1-yl]ethyl]cyclohexyl]-1,1-dimethyl-urea
EN300-7406880
GLXC-90442
urea, n'-[trans-4-[2-[4-(2,3-dichlorophenyl)-1-piperazinyl]ethyl]cyclohexyl]-n,n-dimethyl-; n'-[trans-4-[2-[4-(2,3-dichlorophenyl)-1-piperazinyl]ethyl]cyclohexyl]-n,n-dimethylurea; cariprazine; rgh 188; [trans-4-[[2-[4-(2,3-dichlorophenyl)piperazin-1-yl]e
mfcd19443701

Research Excerpts

Overview

Cariprazine hydrochloride is a second-generation antipsychotic drug. It has been approved by the US Food and Drug Administration and the European Medicines Agency. The drug is a dopamine-serotonin partial agonist, with a recent FDA approval as a monotherapy for BD type 1 (BD-I) depression.

ExcerptReferenceRelevance
"Cariprazine is a newer 3rd generation antipsychotic acting as partial agonist for dopamine receptors with unique higher affinity for D3 than D2 receptors. "( [The newer antipsychotic cariprazine (reagila): perspectives for use in different stages of schizophrenia therapy].
Ivanov, SV; Voronova, EI, 2021
)
2.37
"Cariprazine is a dopamine-serotonin partial agonist, with a recent FDA approval as a monotherapy for BD type 1 (BD-I) depression."( Cariprazine-induced mania: A case series report.
Anmella, G; Bioque, M; Catalan, R; Colomer, L; Druetta, M; Fernández-Plaza, T; Giménez-Palomo, A; Goikolea, JM; Madero, S; Marco-Estrada, O; Navarro-Cortés, L; Pacchiarotti, I; Palacios-Garrán, R; Pons-Cabrera, MT; Sagué-Vilavella, M; Sánchez-Sierra, C; Tardón-Senabre, L; Verdolini, N; Vieta, E, 2022
)
2.89
"Cariprazine is a partial agonist with high affinity at dopamine D"( Cariprazine: A new partial dopamine agonist with a familiar profile.
Hope, J; Keks, NA, 2022
)
3.61
"Cariprazine is a promising antipsychotic drug and may be a valuable resource when clinicians are in doubt if psychotic symptoms are due to schizophrenia or bipolar disorder."( Cariprazine for treating psychosis: an updated meta-analysis.
Cordeiro, Q; Generoso, MB; Kasper, S; Shiozawa, P; Taiar, I, 2023
)
3.07
"Cariprazine hydrochloride is a second-generation antipsychotic drug and has been approved by the US Food and Drug Administration and the European Medicines Agency. "( A comprehensive forced degradation studies of Cariprazine hydrochloride using LC-HRMS/MS and in silico toxicity predictions of its degradation products.
Jain, M; Khan, S, 2022
)
2.42
"Cariprazine is a novel antipsychotic conceived with the idea that high affinity for D3 receptors may elicit a favorable response in the management of cognitive deficits."( Evaluation of the Effect of Cariprazine on Memory and Cognition in Experimental Rodent Models.
Georgieva-Kotetarova, MT; Kandilarov, IK; Vilmosh, NB; Zlatanova, HI, 2022
)
1.74
"Cariprazine is an atypical antipsychotic that acts as a D3/D2 receptor partial agonist. "( Effects of the atypical antipsychotic and D3/D2 dopamine partial agonist cariprazine on effort-based choice behavior: implications for modeling avolition.
Conrad, RT; Correa, M; Ecevitoglu, A; Edelstein, GA; Kovach, A; Okifo, K; Presby, RE; Quiles, T; Rotolo, RA; Salamone, JD; Yang, JH, 2023
)
2.58
"Cariprazine is a dopamine D₃ preferring D₃/D₂ partial agonist with very similar dopamine receptor subtype selectivity as dopamine."( [Cariprazine, a new type - dopamine D₃ receptor preferring - partial agonist atypical antipsychotic for the treatment of schizophrenia and the primary negative symptoms].
Barabassy, A; Kapas, M; Kiss, B; Laszlovszky, I; Nemeth, G, 2019
)
2.15
"Cariprazine is a partial agonist at D2/D3 receptors that has been approved for the treatment of mania associated with bipolar disorder (BD). "( Cariprazine in the treatment of Bipolar Disorder: A systematic review and meta-analysis.
Chakrabarty, T; Keramatian, K; Pinto, JV; Saraf, G; Vigo, D; Yatham, LN, 2020
)
3.44
"Cariprazine is a new atypical antipsychotic drug approved for the treatment of schizophrenia and bipolar disorders."( Efficacy and Safety of Cariprazine in Acute Management of Psychiatric Disorders: a Meta-Analysis of Randomized Controlled Trials.
Cooper, H; Mishriky, R; Reyad, AA, 2020
)
2.31
"Cariprazine is a newer antipsychotic acting as dopamine D3- and in lesser extent D2-receptor partial agonist found to be effective in the treatment of negative symptoms in schizophrenia."( [Cariprazine in schizophrenia with predominantly negative symptoms: early effects of therapy (observational study)].
Beybalaeva, TZ; Ivanov, SV; Katok, AA; Kharkova, GS; Konohova, MV; Skurygina, EI; Smulevich, AB; Voronova, EI; Yakhin, KK, 2020
)
2.19
"Cariprazine is a new dopamine D2 and D3 receptor partial agonist antipsychotic. "( Cariprazine specificity profile in the treatment of acute schizophrenia: a meta-analysis and meta-regression of randomized-controlled trials.
Corponi, F; Fabbri, C; Montgomery, S; Serretti, A, 2017
)
3.34
"Cariprazine is a piprazine derivative approved by the FDA in 2015 for the treatment of schizophrenia and bipolar manic or mixed episodes in adults. "( Cariprazine in Bipolar Depression and Mania: State of the Art.
Carocci, V; Janiri, L; Marano, G; Mazza, M; Romano, B; Traversi, G, 2018
)
3.37
"Cariprazine is a D3-preferring dopamine D3/D2 receptor partial agonist and 5-HT1A partial agonist."( Long-term safety and tolerability of cariprazine as adjunctive therapy in major depressive disorder.
Barabássy, Á; Burgess, MV; Chen, C; Durgam, S; Earley, WR; Németh, G; Vieta, E; Zhong, Y, 2019
)
1.51
"Cariprazine is an atypical antipsychotic currently under investigation as an adjunctive to antidepressant treatment (ADT) for patients with major depressive disorder (MDD). "( Cariprazine Augmentation to Antidepressant Therapy in Major Depressive Disorder: Results of a Randomized, Double-Blind, Placebo-Controlled Trial.
Earley, WR; Guo, H; Harsányi, J; Németh, G; Thase, ME, 2018
)
3.37
"Cariprazine, is an emerging antipsychotic drug, D3/D2 receptor partial agonist, with affinity to several serotonin receptors."( Cariprazine - a novel antipsychotic drug and its place in the treatment of schizophrenia.
Bieńkowski, P; Misiak, B; Samochowiec, J, 2018
)
2.64
"Cariprazine is a new atypical antipsychotic drug (APD) with a unique pharmacodynamic profile, different from both typical and atypical APDs. "( The role of dopamine D
Calabrese, F; Racagni, G; Riva, MA; Tarazi, FI, 2020
)
2
"Cariprazine is an oral antipsychotic approved in the US for the treatment of schizophrenia, acute bipolar mania, and most recently, bipolar depression. "( Cariprazine for bipolar depression: What is the number needed to treat, number needed to harm and likelihood to be helped or harmed?
Citrome, L, 2019
)
3.4
"Cariprazine is a novel drug with partial agonist activity at dopamine D2/3 receptors and six- to eightfold higher affinity for human dopamine D3 over D2 receptors. "( Cariprazine, a new, orally active dopamine D2/3 receptor partial agonist for the treatment of schizophrenia, bipolar mania and depression.
Gründer, G; Paulzen, M; Veselinović, T, 2013
)
3.28
"Cariprazine is a dopamine D₃ and D₂ receptor partial agonist with preferential binding to D₃ receptors."( Long-term effects of cariprazine exposure on dopamine receptor subtypes.
Adham, N; Choi, YK; Gyertyán, I; Kiss, B; Tarazi, FI, 2014
)
1.44
"Cariprazine is an orally active and potent D3 and D2 partial agonist with preferential binding to D3 receptors in development for the treatment of schizophrenia and bipolar mania. "( An evaluation of the safety and efficacy of cariprazine in patients with acute exacerbation of schizophrenia: a phase II, randomized clinical trial.
Durgam, S; Laszlovszky, I; Li, D; Migliore, R; Németh, G; Ruth, A; Starace, A, 2014
)
2.11
"Cariprazine (Vraylar) is an oral atypical antipsychotic originated by Gedeon Richter. "( Cariprazine: First Global Approval.
McCormack, PL, 2015
)
3.3
"Cariprazine is a dopamine D3/D2 receptor partial agonist with preferential binding to D3 receptors, recently approved by the FDA for the treatment of schizophrenia and manic or mixed episodes associated with bipolar I disorder."( Effects of cariprazine, a novel antipsychotic, on cognitive deficit and negative symptoms in a rodent model of schizophrenia symptomatology.
Adham, N; Ferguson, P; Grayson, B; Gyertyán, I; Kiss, B; Neill, JC, 2016
)
1.55
"Cariprazine is a potent dopamine D3-preferring D3/D2 receptor partial agonist in development for the treatment of schizophrenia, bipolar mania, and depression. "( Clinical pharmacology study of cariprazine (MP-214) in patients with schizophrenia (12-week treatment).
Imada, M; Iwakaji, A; Kapás, M; Kubota, T; Morio, Y; Nakamura, T, 2016
)
2.16
"Cariprazine is a new therapeutic agent recently approved for the treatment of both schizophrenia and manic or mixed episodes associated with bipolar disorder, and is under investigation for the treatment of both bipolar depression and major depressive disorder. "( Mechanism of action of cariprazine.
Stahl, SM, 2016
)
2.19
"Cariprazine is an atypical antipsychotic currently under investigation as adjunctive therapy in patients with major depressive disorder (MDD) who have inadequate response to standard antidepressant therapy."( 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.
Durgam, S; Earley, W; Fava, M; Guo, H; Laszlovszky, I; Li, D; Montgomery, SA; Németh, G, 2016
)
2.16
"Cariprazine is a dopamine D3, D2 partial agonist, with greater affinity to D3. "( Cariprazine for the Treatment of Bipolar Disorder.
El-Mallakh, PL; El-Mallakh, RS; Findlay, LJ, 2017
)
3.34
"Cariprazine (RGH-188) is a novel antipsychotic drug that exerts partial agonism of dopamine D2/D3 receptors with preferential binding to D3 receptor, antagonism of 5HT2B receptors and partial agonism of 5HT1A. "( The Novel Antipsychotic Cariprazine (RGH-188): State-of-the-Art in the Treatment of Psychiatric Disorders.
Carano, A; Conti, C; de Bartolomeis, A; De Berardis, D; Di Giannantonio, M; Fornaro, M; Iasevoli, F; Martinotti, G; Matarazzo, I; Mazza, M; Orsolini, L; Perna, G; Pompili, M; Prinzivalli, E; Segura-García, C; Sepede, G; Serroni, N; Valchera, A; Vecchiotti, R; Vellante, F, 2016
)
2.18
"Cariprazine is an antipsychotic medication and received approval by the U.S. "( Cariprazine for the Treatment of Schizophrenia: A Review of this Dopamine D3-Preferring D3/D2 Receptor Partial Agonist.
Citrome, L, 2016
)
3.32
"Cariprazine is a novel antipsychotic agent recently approved for treating schizophrenia and bipolar mania in the USA. "( Tolerability and Safety Profile of Cariprazine in Treating Psychotic Disorders, Bipolar Disorder and Major Depressive Disorder: A Systematic Review with Meta-Analysis of Randomized Controlled Trials.
Besag, FM; Chan, EW; He, Y; Lao, KS; Wong, IC, 2016
)
2.15
"Cariprazine is a novel agent developed in Hungary, its activity on the dopamine D3 receptor might open up new dimensions in the pharmacotherapy of schizophrenia and affective disorders."( [The dopamin D3 receptor--the gray eminence of pharmacotherapy?].
Sumegi, A; Varga, E, 2010
)
1.08
"Cariprazine is a novel antipsychotic drug candidate that exhibits high selectivity and affinity to dopamine D(3) and D(2) receptors and moderate affinity to serotonin 5-HT(1A) receptors. "( 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.
Farkas, S; Finnema, SJ; Gulyás, B; Gyertyán, I; Halldin, C; Horváth, A; Innis, RB; Kapás, M; Kiss, B; Laszlovszky, I; Pásztor, G; Seneca, N, 2011
)
2.03
"Cariprazine (RGH-188) is a D₃-preferring dopamine D₃/D₂ receptor partial agonist antipsychotic candidate for the treatment of schizophrenia and bipolar mania. "( Cariprazine (RGH-188), a D₃-preferring dopamine D₃/D₂ receptor partial agonist antipsychotic candidate demonstrates anti-abuse potential in rats.
Gyertyán, I; Kiss, B; Román, V; Sághy, K; Szombathelyi, Z, 2013
)
3.28
"Cariprazine is a dopamine D(3)/D(2) receptor partial agonist antipsychotic candidate, which binds with high affinity to dopamine D(3) and D(2) receptors (with ∼10-fold higher in vitro affinity to D(3) vs. "( Brain uptake and distribution of the dopamine D3 /D2 receptor partial agonist [11 C]cariprazine: an in vivo positron emission tomography study in nonhuman primates.
Adham, N; Gulyás, B; Gyertyán, I; Halldin, C; Horváth, A; Kiss, B; Laszlovszky, I; Steiger, C; Tóth, M; Varrone, A, 2013
)
2.06
"Cariprazine is an atypical antipsychotic in clinical development for the treatment of schizophrenia and bipolar mania/mixed episodes."( Cariprazine: chemistry, pharmacodynamics, pharmacokinetics, and metabolism, clinical efficacy, safety, and tolerability.
Citrome, L, 2013
)
3.28
"Cariprazine is a dopamine D3-preferring D3/D2 receptor partial agonist. "( Cariprazine: chemistry, pharmacodynamics, pharmacokinetics, and metabolism, clinical efficacy, safety, and tolerability.
Citrome, L, 2013
)
3.28
"Cariprazine is a dopamine D3-preferring D3/D2 receptor partial agonist in late-stage clinical development for the treatment of bipolar disorder (manic/mixed and depressive episodes), as well as for schizophrenia, and as an adjunctive agent for the treatment of major depressive disorder. "( Cariprazine in bipolar disorder: clinical efficacy, tolerability, and place in therapy.
Citrome, L, 2013
)
3.28
"Cariprazine is a dopamine D3-preferring D3/D2 receptor partial agonist in late-stage clinical development for the treatment of schizophrenia, as well as for bipolar disorder (manic/mixed and depressive episodes), and as an adjunctive agent for the treatment of major depressive disorder. "( Cariprazine in schizophrenia: clinical efficacy, tolerability, and place in therapy.
Citrome, L, 2013
)
3.28

Effects

ExcerptReferenceRelevance
"Cariprazine has a higher affinity than dopamine for D3 receptors and has a 10-fold higher affinity for D3 than D2."( Cariprazine: an augmentation strategy for treatment-resistant schizophrenia with pro-cognitive and anti-hostility effects.
Boydstun, C; DiGenova, P; Lynch, S, 2023
)
3.07

Actions

Cariprazine displays higher affinity at dopamine D3 receptors and a similar affinity at D2 and 5-HT2B receptors. It had lower affinity at human and rat hippocampal 5- HT(1A) receptors.

ExcerptReferenceRelevance
"Cariprazine displays higher affinity at dopamine D3 receptors and a similar affinity at D2 and 5-HT2B receptors."( Cariprazine:New dopamine biased agonist for neuropsychiatric disorders.
De Deurwaerdère, P, 2016
)
2.6
"Cariprazine had lower affinity at human and rat hippocampal 5-HT(1A) receptors (pK(i) 8.59 and 8.34, respectively) and demonstrated low intrinsic efficacy."( Cariprazine (RGH-188), a dopamine D(3) receptor-preferring, D(3)/D(2) dopamine receptor antagonist-partial agonist antipsychotic candidate: in vitro and neurochemical profile.
Adham, N; Agai-Csongor, E; Bugovics, G; Domány, G; Fazekas, K; Gyertyán, I; Hornok, K; Horváth, A; Kiss, B; Laszlovszky, I; Némethy, Z; Orosz, S; Schmidt, E; Szombathelyi, Z; Tihanyi, K, 2010
)
2.52

Treatment

More cariprazine-treated than placebo-treated patients reported double-blind treatment-emergent AEs. Cariprazin- treated patients with bipolar I disorder attained clinically significant improvement in manic symptoms.

ExcerptReferenceRelevance
"More cariprazine- than placebo-treated patients reported double-blind treatment-emergent AEs; the overall AE incidence was similar among cariprazine-dose groups."( Tolerability of cariprazine in the treatment of acute bipolar I mania: A pooled post hoc analysis of 3 phase II/III studies.
Debelle, M; Durgam, S; Earley, W; Laszlovszky, I; Lu, K; Vieta, E; Yatham, LN, 2017
)
1.26
"Cariprazine-treated patients with bipolar I disorder attained clinically significant improvement in manic symptoms as shown by significantly greater rates of response and remission versus placebo; improvement in manic symptoms did not induce depressive symptoms."( Clinically relevant response and remission outcomes in cariprazine-treated patients with bipolar I disorder.
Durgam, S; Earley, W; Laszlovszky, I; Lu, K; Németh, G; Ruth, A; Yatham, LN, 2018
)
2.17
"Cariprazine- and placebo-treated patients were categorised by baseline CGI-S scores; the proportion of patients who improved from more severe categories at baseline to less severe categories at end-point was evaluated using a logistic regression model."( Global improvement with cariprazine in the treatment of bipolar I disorder and schizophrenia: A pooled post hoc analysis.
Durgam, S; Earley, W; Laszlovszky, I; Litman, RE; Lu, K; Németh, G; Volk, S, 2017
)
1.48
"More cariprazine- vs placebo-treated patients shifted from the extremely/severely ill to mildly ill/better category (bipolar disorder = 55% vs 36%, odds ratio [OR] = 2.1; P = .09; schizophrenia = 42% vs 18%, OR = 3.4, P<.01)."( Global improvement with cariprazine in the treatment of bipolar I disorder and schizophrenia: A pooled post hoc analysis.
Durgam, S; Earley, W; Laszlovszky, I; Litman, RE; Lu, K; Németh, G; Volk, S, 2017
)
1.22
"More cariprazine-treated (41.6%) than placebo-treated (27.3%) patients sustained remission for any ≥ 6 consecutive month period (OR = 1.90, P = .0379; NNT = 7)."( Long-Term Remission With Cariprazine Treatment in Patients With Schizophrenia: A Post Hoc Analysis of a Randomized, Double-Blind, Placebo-Controlled, Relapse Prevention Trial.
Correll, CU; Earley, W; Harsányi, J; Potkin, SG; Szatmári, B; Zhong, Y, 2019
)
1.27
"More cariprazine- than placebo-treated patients met YMRS response and remission criteria, reaching significance for response in ≥2 DS (34% versus 47%; number-needed-to-treat [NNT] = 8, P = .0483) and ≥10 MADRS (31% versus 57%, NNT = 4, P < .0001) and for remission in ≥2 DS (27% versus 39%, NNT = 9, P = .0462), ≥10 MADRS (23% versus 44%, NNT = 5, P < .0001)."( Cariprazine for the treatment of bipolar mania with mixed features: A post hoc pooled analysis of 3 trials.
Earley, W; Masand, PS; McIntyre, RS; Patel, M, 2019
)
2.41
"Both cariprazine treatment groups showed statistically significant superiority to placebo on all 11 YMRS single items (all comparisons, P < .05)."( Efficacy and safety of low- and high-dose cariprazine in acute and mixed mania associated with bipolar I disorder: a double-blind, placebo-controlled study.
Calabrese, JR; Durgam, S; Keck, PE; Laszlovszky, I; Lu, K; Németh, G; Ruth, A; Starace, A, 2015
)
1.14
"The cariprazine treatment group was more likely to have clinically significant weight gain (RR 1.68, 95 % CI 1.12-2.52)."( Tolerability and Safety Profile of Cariprazine in Treating Psychotic Disorders, Bipolar Disorder and Major Depressive Disorder: A Systematic Review with Meta-Analysis of Randomized Controlled Trials.
Besag, FM; Chan, EW; He, Y; Lao, KS; Wong, IC, 2016
)
1.19
"Cariprazine pretreatment significantly attenuated the emergence of these cognitive deficits in PCP-treated wild-type mice, but not in PCP-treated D(3)-receptor knockout mice."( Cariprazine, a dopamine D(3)-receptor-preferring partial agonist, blocks phencyclidine-induced impairments of working memory, attention set-shifting, and recognition memory in the mouse.
Adham, N; Chang, G; Gyertyán, I; Kiss, B; Schmauss, C; Zimnisky, R, 2013
)
2.55

Toxicity

Long-term cariprazine treatment at doses up to 9 mg/d appeared to be generally safe and well tolerated in patients with schizophrenia. The most common treatment-related adverse events were akathisia (both groups) and nausea, constipation, and tremor (6-12mg/d only)

ExcerptReferenceRelevance
" There does not appear to be clinically relevant adverse effects of cariprazine on metabolic variables."( Cariprazine: chemistry, pharmacodynamics, pharmacokinetics, and metabolism, clinical efficacy, safety, and tolerability.
Citrome, L, 2013
)
2.07
" Safety parameters included adverse events (AEs), vital signs, laboratory measures, and extrapyramidal symptom (EPS) scales."( An evaluation of the safety and efficacy of cariprazine in patients with acute exacerbation of schizophrenia: a phase II, randomized clinical trial.
Durgam, S; Laszlovszky, I; Li, D; Migliore, R; Németh, G; Ruth, A; Starace, A, 2014
)
0.66
" The most common (≥ 5% and twice the rate of placebo) treatment-related adverse events for cariprazine were akathisia (both groups) and nausea, constipation, and tremor (6-12 mg/d only)."( Efficacy and safety of low- and high-dose cariprazine in acute and mixed mania associated with bipolar I disorder: a double-blind, placebo-controlled study.
Calabrese, JR; Durgam, S; Keck, PE; Laszlovszky, I; Lu, K; Németh, G; Ruth, A; Starace, A, 2015
)
0.9
" Safety included treatment-emergent adverse events, clinical laboratory values, vital signs, electrocardiograms, ophthalmologic examination, Columbia-Suicide Severity Rating Scale, and extrapyramidal symptom scales."( Efficacy and Safety of Cariprazine in Acute Exacerbation of Schizophrenia: Results From an International, Phase III Clinical Trial.
Durgam, S; Kane, JM; Laszlovszky, I; Lu, K; Nagy, K; Ruth, A; Wang, Y; Zukin, S, 2015
)
0.73
" The most common adverse events (≥10%) in cariprazine-treated patients were akathisia and insomnia; weight gain was slightly higher with cariprazine than with placebo."( An 8-Week Randomized, Double-Blind, Placebo-Controlled Evaluation of the Safety and Efficacy of Cariprazine in Patients With Bipolar I Depression.
Calabrese, JR; Durgam, S; Earley, W; Guo, H; Laszlovszky, I; Lipschitz, A; Németh, G; Vieta, E; Yatham, LN, 2016
)
0.92
" Safety assessments included adverse events, clinical laboratory tests, vital signs, electrocardiograms (ECGs), and suicidality."( 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.
Durgam, S; Earley, W; Fava, M; Guo, H; Laszlovszky, I; Li, D; Montgomery, SA; Németh, G, 2016
)
0.71
" Treatment-emergent adverse events reported in ≥ 10% of patients in either cariprazine dosage group were akathisia (22."( 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.
Durgam, S; Earley, W; Fava, M; Guo, H; Laszlovszky, I; Li, D; Montgomery, SA; Németh, G, 2016
)
0.94
" We conducted a meta-analysis to investigate outcomes, including risks of discontinuation due to adverse events (AEs), extrapyramidal side effects (EPS) or related events, metabolic syndrome and cardiovascular-related events."( Tolerability and Safety Profile of Cariprazine in Treating Psychotic Disorders, Bipolar Disorder and Major Depressive Disorder: A Systematic Review with Meta-Analysis of Randomized Controlled Trials.
Besag, FM; Chan, EW; He, Y; Lao, KS; Wong, IC, 2016
)
0.71
" EPSs were the main short-term adverse reactions reported in the limited number of patients studied."( Tolerability and Safety Profile of Cariprazine in Treating Psychotic Disorders, Bipolar Disorder and Major Depressive Disorder: A Systematic Review with Meta-Analysis of Randomized Controlled Trials.
Besag, FM; Chan, EW; He, Y; Lao, KS; Wong, IC, 2016
)
0.71
" The most common (≥10%) adverse events (AEs) observed were akathisia (16%), headache (13%), insomnia (13%), and weight gain (10%)."( Evaluation of the long-term safety and tolerability of cariprazine in patients with schizophrenia: results from a 1-year open-label study.
Cutler, AJ; Durgam, S; Laszlovszky, I; Lu, K; Migliore, R; Németh, G; Wang, Y, 2018
)
0.73
"Long-term cariprazine treatment at doses up to 9 mg/d appeared to be generally safe and well tolerated in patients with schizophrenia."( Evaluation of the long-term safety and tolerability of cariprazine in patients with schizophrenia: results from a 1-year open-label study.
Cutler, AJ; Durgam, S; Laszlovszky, I; Lu, K; Migliore, R; Németh, G; Wang, Y, 2018
)
1.13
" The only adverse events (AEs) leading to discontinuation of ≥2% of patients in any dose group were akathisia, worsening of schizophrenia, and psychotic disorder."( The safety and tolerability of cariprazine in long-term treatment of schizophrenia: a post hoc pooled analysis.
Cutler, AJ; Durgam, S; Earley, W; Laszlovszky, I; Lu, K; Nasrallah, HA; Németh, G; Wang, Y, 2017
)
0.74
"In these post hoc pooled analyses of data from 2 long-term open-label studies, treatment with cariprazine was generally safe and well tolerated."( The safety and tolerability of cariprazine in long-term treatment of schizophrenia: a post hoc pooled analysis.
Cutler, AJ; Durgam, S; Earley, W; Laszlovszky, I; Lu, K; Nasrallah, HA; Németh, G; Wang, Y, 2017
)
0.96
" Safety evaluations included adverse events (AEs), laboratory values, vital signs, and extrapyramidal symptom (EPS) scales."( The safety and tolerability of cariprazine in patients with manic or mixed episodes associated with bipolar I disorder: A 16-week open-label study.
Durgam, S; Ketter, TA; Laszlovszky, I; Lu, K; Németh, G; Sachs, GS; Starace, A, 2018
)
0.77
" The most common reason for discontinuation was adverse events (AEs; 13."( Long-term safety and tolerability of cariprazine as adjunctive therapy in major depressive disorder.
Barabássy, Á; Burgess, MV; Chen, C; Durgam, S; Earley, WR; Németh, G; Vieta, E; Zhong, Y, 2019
)
0.79
" Adverse events (AEs), laboratory results, vital signs, and suicide risk were monitored."( Efficacy and safety of cariprazine in bipolar I depression: A double-blind, placebo-controlled phase 3 study.
Burgess, MV; Calabrese, JR; Earley, WR; Khan, B; Rekeda, L; Suppes, T; Tohen, M, 2020
)
0.87
" The risk of discontinuation due to adverse effects and common side effects were examined."( Efficacy and Safety of Cariprazine in Acute Management of Psychiatric Disorders: a Meta-Analysis of Randomized Controlled Trials.
Cooper, H; Mishriky, R; Reyad, AA, 2020
)
0.87
"Sexual dysfunction is a common adverse effect in selective serotonin reuptake inhibitors users."( Resolution of Citalopram Sexual Adverse Effects With Low Dose of Cariprazine: A Case Report.
Dieci, M; Mansi, G; Trama, A,
)
0.37
" Safety parameters included spontaneous reports of adverse events and specific assessments of extrapyramidal side-effects."( The effectiveness and safety of cariprazine in schizophrenia patients with negative symptoms and insufficient effectiveness of previous antipsychotic therapy: an observational study.
Barabássy, Á; Dombi, ZB; Mátrai, P; Németh, G; Rancans, E; Sebe, B; Skrivele, I, 2021
)
0.9
" Descriptive statistics were used for safety analyses including treatment emergent adverse events (TEAEs) and discontinuation rates."( The efficacy and safety of cariprazine in the early and late stage of schizophrenia: a post hoc analysis of three randomized, placebo-controlled trials.
Acsai, K; Barabássy, Á; Dombi, ZB; Falkai, P; Németh, G; Schmitt, A, 2023
)
1.21

Pharmacokinetics

Cariprazine has a mean half-life of 2 - 5 d over a dose range of 1.5 d. The current study investigated whether the pharmacodynamic properties of caripazine fit into a previously developed model.

ExcerptReferenceRelevance
" The author provides the reader with knowledge of the fundamental pharmacokinetic characteristics and metabolic pathways of these new antipsychotics, emphasizing the clinically important common features and differences compared to other older agents."( Pharmacokinetics and metabolism update for some recent antipsychotics.
Caccia, S, 2011
)
0.37
"Aripiprazole, perospirone, lurasidone and cariprazine share some of the pharmacokinetic characteristics of older, lipophilic antipsychotics and, like these, each has some distinct pharmacokinetic features that are clinically beneficial and some that are not."( Pharmacokinetics and metabolism update for some recent antipsychotics.
Caccia, S, 2011
)
0.63
"The purpose of this review is to describe the chemistry, pharmacodynamic profile, pharmacokinetics, and clinical profile of cariprazine."( Cariprazine: chemistry, pharmacodynamics, pharmacokinetics, and metabolism, clinical efficacy, safety, and tolerability.
Citrome, L, 2013
)
2.04
" The current study investigated whether the pharmacodynamic properties of cariprazine fit into a previously developed model which was the first to be derived based on the strict combination of clinical and preclinical data."( Antidepressant efficacy of cariprazine in bipolar disorder and the role of its pharmacodynamic properties: A hypothesis based on data.
Fountoulakis, KN; Haarman, BCM; Ioannou, M; Tohen, M; Zarate, CA, 2023
)
1.44

Bioavailability

Oral bioavailability at a dose of 1mg/kg in rats was 52% with peak plasma concentrations of 91ng/mL. Cariprazine is rapidly absorbed, with high oral bioavailability and a long plasma elimination t1/2.

ExcerptReferenceRelevance
" Cariprazine is rapidly absorbed, with high oral bioavailability and a long plasma elimination t1/2."( Cariprazine, an orally active D2/D3 receptor antagonist, for the potential treatment of schizophrenia, bipolar mania and depression.
Gründer, G, 2010
)
2.71
" Oral bioavailability of cariprazine at a dose of 1mg/kg in rats was 52% with peak plasma concentrations of 91ng/mL."( Cariprazine (RGH-188), a potent D3/D2 dopamine receptor partial agonist, binds to dopamine D3 receptors in vivo and shows antipsychotic-like and procognitive effects in rodents.
Csongor, EÁ; Domány, G; Gémesi, LI; Gyertyán, I; Kapás, M; Kiss, B; Laszy, J; Pásztor, G; Sághy, K; Szabados, T; Szabó, G; Szombathelyi, Z; Tihanyi, K; Zájer-Balázs, M, 2011
)
2.12
"Cell membrane permeability is an important determinant for oral absorption and bioavailability of a drug molecule."( Highly predictive and interpretable models for PAMPA permeability.
Jadhav, A; Kerns, E; Nguyen, K; Shah, P; Sun, H; Xu, X; Yan, Z; Yu, KR, 2017
)
0.46
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51
" Additionally, an ∼85% correlation was obtained between PAMPA pH 5 permeability and in vivo oral bioavailability in mice and rats."( Using in vitro ADME data for lead compound selection: An emphasis on PAMPA pH 5 permeability and oral bioavailability.
Itkin, M; Kabir, M; Mathé, EA; Nguyễn, ÐT; Padilha, EC; Shah, P; Shinn, P; Siramshetty, V; Wang, AQ; Williams, J; Xu, X; Yu, KR; Zhao, T, 2022
)
0.72

Dosage Studied

Cariprazine was relatively well tolerated. Common treatment-emergent adverse events included headache, arthralgia, restlessness, fatigue, increased appetite, insomnia, dry mouth, and constipation.

ExcerptRelevanceReference
" These findings can be used to guide the selection of cariprazine dosing in humans."( 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.
Farkas, S; Finnema, SJ; Gulyás, B; Gyertyán, I; Halldin, C; Horváth, A; Innis, RB; Kapás, M; Kiss, B; Laszlovszky, I; Pásztor, G; Seneca, N, 2011
)
0.83
"75 mg/day dosage was similar to placebo."( An 8-Week Randomized, Double-Blind, Placebo-Controlled Evaluation of the Safety and Efficacy of Cariprazine in Patients With Bipolar I Depression.
Calabrese, JR; Durgam, S; Earley, W; Guo, H; Laszlovszky, I; Lipschitz, A; Németh, G; Vieta, E; Yatham, LN, 2016
)
0.65
" Treatment-emergent adverse events reported in ≥ 10% of patients in either cariprazine dosage group were akathisia (22."( 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.
Durgam, S; Earley, W; Fava, M; Guo, H; Laszlovszky, I; Li, D; Montgomery, SA; Németh, G, 2016
)
0.94
"5-6 and 9-12 mg/day; a dose-response relationship was observed for akathisia, extrapyramidal symptoms, and diastolic blood pressure."( 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.
Debelle, M; Durgam, S; Earley, W; Kane, JM; Laszlovszky, I; Lu, K, 2017
)
0.75
" Clinically insignificant changes in aminotransferase levels and alkaline phosphatase were observed; no dose-response relationship was observed across groups."( The safety and tolerability of cariprazine in long-term treatment of schizophrenia: a post hoc pooled analysis.
Cutler, AJ; Durgam, S; Earley, W; Laszlovszky, I; Lu, K; Nasrallah, HA; Németh, G; Wang, Y, 2017
)
0.74
" Cariprazine was relatively well tolerated, and common treatment-emergent adverse events (incidence ≥5% and twice the placebo group rate) in both dosage groups included headache, arthralgia, restlessness, fatigue, increased appetite, insomnia, dry mouth, and constipation."( Efficacy of adjunctive low-dose cariprazine in major depressive disorder: a randomized, double-blind, placebo-controlled trial.
Durgam, S; Earley, W; Fava, M; Hayes, R; Laszlovszky, I; Lu, K; Németh, G, 2018
)
1.67
"The dose-response relationships of antipsychotic drugs for schizophrenia are not well defined, but such information would be important for decision making by clinicians."( Dose-Response Meta-Analysis of Antipsychotic Drugs for Acute Schizophrenia.
Crippa, A; Davis, JM; Leucht, S; Orsini, N; Patel, MX; Siafis, S, 2020
)
0.56
" Dose-response curves were constructed with random-effects dose-response meta-analyses and a spline model."( Dose-Response Meta-Analysis of Antipsychotic Drugs for Acute Schizophrenia.
Crippa, A; Davis, JM; Leucht, S; Orsini, N; Patel, MX; Siafis, S, 2020
)
0.56
" For some drugs, higher than currently licensed doses might be tested in further trials, because their dose-response curves did not plateau."( Dose-Response Meta-Analysis of Antipsychotic Drugs for Acute Schizophrenia.
Crippa, A; Davis, JM; Leucht, S; Orsini, N; Patel, MX; Siafis, S, 2020
)
0.56
" Cariprazine-related akathisia and EPS can be minimized with conservative dosing and titration strategies."( Cariprazine and akathisia, restlessness, and extrapyramidal symptoms in patients with bipolar depression.
Barabássy, Á; Citrome, L; Earley, WR; Hankinson, A; Patel, MD; Yatham, LN, 2021
)
2.97
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Protein Targets (21)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
GVesicular stomatitis virusPotency21.31740.01238.964839.8107AID1645842
cytochrome P450 2D6Homo sapiens (human)Potency11.98770.00108.379861.1304AID1645840
Interferon betaHomo sapiens (human)Potency21.31740.00339.158239.8107AID1645842
HLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)Potency21.31740.01238.964839.8107AID1645842
Spike glycoproteinSevere acute respiratory syndrome-related coronavirusPotency17.78280.009610.525035.4813AID1479145
Inositol hexakisphosphate kinase 1Homo sapiens (human)Potency21.31740.01238.964839.8107AID1645842
cytochrome P450 2C9, partialHomo sapiens (human)Potency21.31740.01238.964839.8107AID1645842
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
5-hydroxytryptamine receptor 1AHomo sapiens (human)Ki0.00320.00010.532610.0000AID1332826; AID1517957; AID1682817; AID1782426; AID1907047
D(2) dopamine receptorHomo sapiens (human)IC50 (µMol)0.00170.00000.74728.0000AID1153309
D(2) dopamine receptorHomo sapiens (human)Ki0.00920.00000.651810.0000AID1054309; AID1153295; AID1153296; AID1332824; AID1444853; AID1444854; AID1485399; AID1485400; AID1517960; AID1682820; AID1782419; AID1889749
Alpha-1B adrenergic receptorRattus norvegicus (Norway rat)Ki0.15500.00010.949010.0000AID1682821
D(1A) dopamine receptorHomo sapiens (human)Ki2.75060.00010.836310.0000AID1153294; AID1485397; AID1782424
D(4) dopamine receptorHomo sapiens (human)Ki0.30700.00000.436210.0000AID1153298; AID1485402; AID1782429
D(1B) dopamine receptorHomo sapiens (human)Ki7.90000.00030.40177.9000AID1485398
Alpha-1D adrenergic receptorRattus norvegicus (Norway rat)Ki0.15500.00000.575110.0000AID1682821
5-hydroxytryptamine receptor 2AHomo sapiens (human)Ki0.07750.00000.385510.0000AID1153300; AID1332827; AID1485404; AID1517959; AID1682816; AID1782427
5-hydroxytryptamine receptor 2CHomo sapiens (human)Ki0.19890.00010.954910.0000AID1782428
5-hydroxytryptamine receptor 7Homo sapiens (human)Ki0.11100.00030.380610.0000AID1517962
Histamine H1 receptorHomo sapiens (human)Ki0.02320.00000.511010.0000AID1682819
D(3) dopamine receptorHomo sapiens (human)Ki0.00020.00000.602010.0000AID1153297; AID1332825; AID1444852; AID1485401; AID1682818; AID1782425
Alpha-1A adrenergic receptorRattus norvegicus (Norway rat)Ki0.15500.00000.965010.0000AID1682821
Potassium voltage-gated channel subfamily H member 2Homo sapiens (human)IC50 (µMol)20.72000.00091.901410.0000AID1332832
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
D(2) dopamine receptorHomo sapiens (human)EC50 (µMol)0.01760.00000.18743.9000AID1153305; AID1153311; AID1595585; AID1595588; AID1782420; AID1782422
D(3) dopamine receptorHomo sapiens (human)EC50 (µMol)0.00580.00010.02470.6690AID1153307
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (273)

Processvia Protein(s)Taxonomy
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell activation involved in immune responseInterferon betaHomo sapiens (human)
cell surface receptor signaling pathwayInterferon betaHomo sapiens (human)
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to virusInterferon betaHomo sapiens (human)
positive regulation of autophagyInterferon betaHomo sapiens (human)
cytokine-mediated signaling pathwayInterferon betaHomo sapiens (human)
natural killer cell activationInterferon betaHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylation of STAT proteinInterferon betaHomo sapiens (human)
cellular response to interferon-betaInterferon betaHomo sapiens (human)
B cell proliferationInterferon betaHomo sapiens (human)
negative regulation of viral genome replicationInterferon betaHomo sapiens (human)
innate immune responseInterferon betaHomo sapiens (human)
positive regulation of innate immune responseInterferon betaHomo sapiens (human)
regulation of MHC class I biosynthetic processInterferon betaHomo sapiens (human)
negative regulation of T cell differentiationInterferon betaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIInterferon betaHomo sapiens (human)
defense response to virusInterferon betaHomo sapiens (human)
type I interferon-mediated signaling pathwayInterferon betaHomo sapiens (human)
neuron cellular homeostasisInterferon betaHomo sapiens (human)
cellular response to exogenous dsRNAInterferon betaHomo sapiens (human)
cellular response to virusInterferon betaHomo sapiens (human)
negative regulation of Lewy body formationInterferon betaHomo sapiens (human)
negative regulation of T-helper 2 cell cytokine productionInterferon betaHomo sapiens (human)
positive regulation of apoptotic signaling pathwayInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell differentiationInterferon betaHomo sapiens (human)
natural killer cell activation involved in immune responseInterferon betaHomo sapiens (human)
adaptive immune responseInterferon betaHomo sapiens (human)
T cell activation involved in immune responseInterferon betaHomo sapiens (human)
humoral immune responseInterferon betaHomo sapiens (human)
positive regulation of T cell mediated cytotoxicityHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
adaptive immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
antigen processing and presentation of endogenous peptide antigen via MHC class I via ER pathway, TAP-independentHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of T cell anergyHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
defense responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
detection of bacteriumHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of interleukin-12 productionHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of interleukin-6 productionHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protection from natural killer cell mediated cytotoxicityHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
innate immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of dendritic cell differentiationHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
antigen processing and presentation of endogenous peptide antigen via MHC class IbHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
behavioral fear response5-hydroxytryptamine receptor 1AHomo sapiens (human)
G protein-coupled receptor signaling pathway5-hydroxytryptamine receptor 1AHomo sapiens (human)
adenylate cyclase-inhibiting serotonin receptor signaling pathway5-hydroxytryptamine receptor 1AHomo sapiens (human)
serotonin receptor signaling pathway5-hydroxytryptamine receptor 1AHomo sapiens (human)
gamma-aminobutyric acid signaling pathway5-hydroxytryptamine receptor 1AHomo sapiens (human)
positive regulation of cell population proliferation5-hydroxytryptamine receptor 1AHomo sapiens (human)
regulation of serotonin secretion5-hydroxytryptamine receptor 1AHomo sapiens (human)
regulation of vasoconstriction5-hydroxytryptamine receptor 1AHomo sapiens (human)
exploration behavior5-hydroxytryptamine receptor 1AHomo sapiens (human)
regulation of dopamine metabolic process5-hydroxytryptamine receptor 1AHomo sapiens (human)
serotonin metabolic process5-hydroxytryptamine receptor 1AHomo sapiens (human)
regulation of hormone secretion5-hydroxytryptamine receptor 1AHomo sapiens (human)
regulation of behavior5-hydroxytryptamine receptor 1AHomo sapiens (human)
chemical synaptic transmission5-hydroxytryptamine receptor 1AHomo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messenger5-hydroxytryptamine receptor 1AHomo sapiens (human)
phospholipase C-activating dopamine receptor signaling pathwayD(2) dopamine receptorHomo sapiens (human)
temperature homeostasisD(2) dopamine receptorHomo sapiens (human)
response to hypoxiaD(2) dopamine receptorHomo sapiens (human)
negative regulation of protein phosphorylationD(2) dopamine receptorHomo sapiens (human)
response to amphetamineD(2) dopamine receptorHomo sapiens (human)
nervous system process involved in regulation of systemic arterial blood pressureD(2) dopamine receptorHomo sapiens (human)
regulation of heart rateD(2) dopamine receptorHomo sapiens (human)
regulation of sodium ion transportD(2) dopamine receptorHomo sapiens (human)
G protein-coupled receptor internalizationD(2) dopamine receptorHomo sapiens (human)
positive regulation of neuroblast proliferationD(2) dopamine receptorHomo sapiens (human)
positive regulation of receptor internalizationD(2) dopamine receptorHomo sapiens (human)
autophagyD(2) dopamine receptorHomo sapiens (human)
adenylate cyclase-inhibiting dopamine receptor signaling pathwayD(2) dopamine receptorHomo sapiens (human)
neuron-neuron synaptic transmissionD(2) dopamine receptorHomo sapiens (human)
neuroblast proliferationD(2) dopamine receptorHomo sapiens (human)
axonogenesisD(2) dopamine receptorHomo sapiens (human)
synapse assemblyD(2) dopamine receptorHomo sapiens (human)
sensory perception of smellD(2) dopamine receptorHomo sapiens (human)
long-term memoryD(2) dopamine receptorHomo sapiens (human)
grooming behaviorD(2) dopamine receptorHomo sapiens (human)
locomotory behaviorD(2) dopamine receptorHomo sapiens (human)
adult walking behaviorD(2) dopamine receptorHomo sapiens (human)
protein localizationD(2) dopamine receptorHomo sapiens (human)
negative regulation of cell population proliferationD(2) dopamine receptorHomo sapiens (human)
associative learningD(2) dopamine receptorHomo sapiens (human)
visual learningD(2) dopamine receptorHomo sapiens (human)
response to xenobiotic stimulusD(2) dopamine receptorHomo sapiens (human)
response to light stimulusD(2) dopamine receptorHomo sapiens (human)
response to toxic substanceD(2) dopamine receptorHomo sapiens (human)
response to iron ionD(2) dopamine receptorHomo sapiens (human)
response to inactivityD(2) dopamine receptorHomo sapiens (human)
Wnt signaling pathwayD(2) dopamine receptorHomo sapiens (human)
striatum developmentD(2) dopamine receptorHomo sapiens (human)
orbitofrontal cortex developmentD(2) dopamine receptorHomo sapiens (human)
cerebral cortex GABAergic interneuron migrationD(2) dopamine receptorHomo sapiens (human)
adenohypophysis developmentD(2) dopamine receptorHomo sapiens (human)
negative regulation of cell migrationD(2) dopamine receptorHomo sapiens (human)
peristalsisD(2) dopamine receptorHomo sapiens (human)
auditory behaviorD(2) dopamine receptorHomo sapiens (human)
regulation of synaptic transmission, GABAergicD(2) dopamine receptorHomo sapiens (human)
positive regulation of cytokinesisD(2) dopamine receptorHomo sapiens (human)
circadian regulation of gene expressionD(2) dopamine receptorHomo sapiens (human)
negative regulation of dopamine secretionD(2) dopamine receptorHomo sapiens (human)
response to histamineD(2) dopamine receptorHomo sapiens (human)
response to nicotineD(2) dopamine receptorHomo sapiens (human)
positive regulation of urine volumeD(2) dopamine receptorHomo sapiens (human)
positive regulation of renal sodium excretionD(2) dopamine receptorHomo sapiens (human)
positive regulation of multicellular organism growthD(2) dopamine receptorHomo sapiens (human)
response to cocaineD(2) dopamine receptorHomo sapiens (human)
negative regulation of circadian sleep/wake cycle, sleepD(2) dopamine receptorHomo sapiens (human)
dopamine metabolic processD(2) dopamine receptorHomo sapiens (human)
drinking behaviorD(2) dopamine receptorHomo sapiens (human)
regulation of potassium ion transportD(2) dopamine receptorHomo sapiens (human)
response to morphineD(2) dopamine receptorHomo sapiens (human)
pigmentationD(2) dopamine receptorHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionD(2) dopamine receptorHomo sapiens (human)
positive regulation of G protein-coupled receptor signaling pathwayD(2) dopamine receptorHomo sapiens (human)
negative regulation of blood pressureD(2) dopamine receptorHomo sapiens (human)
negative regulation of innate immune responseD(2) dopamine receptorHomo sapiens (human)
positive regulation of transcription by RNA polymerase IID(2) dopamine receptorHomo sapiens (human)
negative regulation of insulin secretionD(2) dopamine receptorHomo sapiens (human)
acid secretionD(2) dopamine receptorHomo sapiens (human)
behavioral response to cocaineD(2) dopamine receptorHomo sapiens (human)
behavioral response to ethanolD(2) dopamine receptorHomo sapiens (human)
regulation of long-term neuronal synaptic plasticityD(2) dopamine receptorHomo sapiens (human)
response to axon injuryD(2) dopamine receptorHomo sapiens (human)
branching morphogenesis of a nerveD(2) dopamine receptorHomo sapiens (human)
arachidonic acid secretionD(2) dopamine receptorHomo sapiens (human)
epithelial cell proliferationD(2) dopamine receptorHomo sapiens (human)
negative regulation of epithelial cell proliferationD(2) dopamine receptorHomo sapiens (human)
negative regulation of protein secretionD(2) dopamine receptorHomo sapiens (human)
release of sequestered calcium ion into cytosolD(2) dopamine receptorHomo sapiens (human)
dopamine uptake involved in synaptic transmissionD(2) dopamine receptorHomo sapiens (human)
regulation of dopamine uptake involved in synaptic transmissionD(2) dopamine receptorHomo sapiens (human)
positive regulation of dopamine uptake involved in synaptic transmissionD(2) dopamine receptorHomo sapiens (human)
regulation of synapse structural plasticityD(2) dopamine receptorHomo sapiens (human)
negative regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionD(2) dopamine receptorHomo sapiens (human)
negative regulation of synaptic transmission, glutamatergicD(2) dopamine receptorHomo sapiens (human)
excitatory postsynaptic potentialD(2) dopamine receptorHomo sapiens (human)
positive regulation of growth hormone secretionD(2) dopamine receptorHomo sapiens (human)
prepulse inhibitionD(2) dopamine receptorHomo sapiens (human)
negative regulation of dopamine receptor signaling pathwayD(2) dopamine receptorHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascadeD(2) dopamine receptorHomo sapiens (human)
regulation of locomotion involved in locomotory behaviorD(2) dopamine receptorHomo sapiens (human)
postsynaptic modulation of chemical synaptic transmissionD(2) dopamine receptorHomo sapiens (human)
presynaptic modulation of chemical synaptic transmissionD(2) dopamine receptorHomo sapiens (human)
negative regulation of cellular response to hypoxiaD(2) dopamine receptorHomo sapiens (human)
positive regulation of glial cell-derived neurotrophic factor productionD(2) dopamine receptorHomo sapiens (human)
positive regulation of long-term synaptic potentiationD(2) dopamine receptorHomo sapiens (human)
hyaloid vascular plexus regressionD(2) dopamine receptorHomo sapiens (human)
negative regulation of neuron migrationD(2) dopamine receptorHomo sapiens (human)
negative regulation of cytosolic calcium ion concentrationD(2) dopamine receptorHomo sapiens (human)
regulation of dopamine secretionD(2) dopamine receptorHomo sapiens (human)
negative regulation of adenylate cyclase activityD(2) dopamine receptorHomo sapiens (human)
phospholipase C-activating dopamine receptor signaling pathwayD(2) dopamine receptorHomo sapiens (human)
negative regulation of voltage-gated calcium channel activityD(2) dopamine receptorHomo sapiens (human)
positive regulation of MAPK cascadeD(2) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayD(2) dopamine receptorHomo sapiens (human)
temperature homeostasisD(1A) dopamine receptorHomo sapiens (human)
conditioned taste aversionD(1A) dopamine receptorHomo sapiens (human)
behavioral fear responseD(1A) dopamine receptorHomo sapiens (human)
regulation of protein phosphorylationD(1A) dopamine receptorHomo sapiens (human)
synaptic transmission, dopaminergicD(1A) dopamine receptorHomo sapiens (human)
response to amphetamineD(1A) dopamine receptorHomo sapiens (human)
protein import into nucleusD(1A) dopamine receptorHomo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerD(1A) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating G protein-coupled receptor signaling pathwayD(1A) dopamine receptorHomo sapiens (human)
activation of adenylate cyclase activityD(1A) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating dopamine receptor signaling pathwayD(1A) dopamine receptorHomo sapiens (human)
synapse assemblyD(1A) dopamine receptorHomo sapiens (human)
memoryD(1A) dopamine receptorHomo sapiens (human)
mating behaviorD(1A) dopamine receptorHomo sapiens (human)
grooming behaviorD(1A) dopamine receptorHomo sapiens (human)
adult walking behaviorD(1A) dopamine receptorHomo sapiens (human)
visual learningD(1A) dopamine receptorHomo sapiens (human)
response to xenobiotic stimulusD(1A) dopamine receptorHomo sapiens (human)
astrocyte developmentD(1A) dopamine receptorHomo sapiens (human)
dopamine transportD(1A) dopamine receptorHomo sapiens (human)
transmission of nerve impulseD(1A) dopamine receptorHomo sapiens (human)
neuronal action potentialD(1A) dopamine receptorHomo sapiens (human)
dentate gyrus developmentD(1A) dopamine receptorHomo sapiens (human)
striatum developmentD(1A) dopamine receptorHomo sapiens (human)
cerebral cortex GABAergic interneuron migrationD(1A) dopamine receptorHomo sapiens (human)
positive regulation of cell migrationD(1A) dopamine receptorHomo sapiens (human)
peristalsisD(1A) dopamine receptorHomo sapiens (human)
operant conditioningD(1A) dopamine receptorHomo sapiens (human)
synaptic transmission, glutamatergicD(1A) dopamine receptorHomo sapiens (human)
regulation of dopamine metabolic processD(1A) dopamine receptorHomo sapiens (human)
vasodilationD(1A) dopamine receptorHomo sapiens (human)
dopamine metabolic processD(1A) dopamine receptorHomo sapiens (human)
maternal behaviorD(1A) dopamine receptorHomo sapiens (human)
positive regulation of potassium ion transportD(1A) dopamine receptorHomo sapiens (human)
glucose importD(1A) dopamine receptorHomo sapiens (human)
habituationD(1A) dopamine receptorHomo sapiens (human)
sensitizationD(1A) dopamine receptorHomo sapiens (human)
behavioral response to cocaineD(1A) dopamine receptorHomo sapiens (human)
positive regulation of release of sequestered calcium ion into cytosolD(1A) dopamine receptorHomo sapiens (human)
regulation of dopamine uptake involved in synaptic transmissionD(1A) dopamine receptorHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicD(1A) dopamine receptorHomo sapiens (human)
prepulse inhibitionD(1A) dopamine receptorHomo sapiens (human)
phospholipase C-activating dopamine receptor signaling pathwayD(1A) dopamine receptorHomo sapiens (human)
long-term synaptic potentiationD(1A) dopamine receptorHomo sapiens (human)
long-term synaptic depressionD(1A) dopamine receptorHomo sapiens (human)
cellular response to catecholamine stimulusD(1A) dopamine receptorHomo sapiens (human)
modification of postsynaptic structureD(1A) dopamine receptorHomo sapiens (human)
presynaptic modulation of chemical synaptic transmissionD(1A) dopamine receptorHomo sapiens (human)
positive regulation of neuron migrationD(1A) dopamine receptorHomo sapiens (human)
positive regulation of MAPK cascadeD(1A) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayD(1A) dopamine receptorHomo sapiens (human)
dopamine receptor signaling pathwayD(1A) dopamine receptorHomo sapiens (human)
positive regulation of MAP kinase activityD(4) dopamine receptorHomo sapiens (human)
behavioral fear responseD(4) dopamine receptorHomo sapiens (human)
synaptic transmission, dopaminergicD(4) dopamine receptorHomo sapiens (human)
response to amphetamineD(4) dopamine receptorHomo sapiens (human)
intracellular calcium ion homeostasisD(4) dopamine receptorHomo sapiens (human)
adenylate cyclase-inhibiting dopamine receptor signaling pathwayD(4) dopamine receptorHomo sapiens (human)
dopamine receptor signaling pathwayD(4) dopamine receptorHomo sapiens (human)
adult locomotory behaviorD(4) dopamine receptorHomo sapiens (human)
positive regulation of sodium:proton antiporter activityD(4) dopamine receptorHomo sapiens (human)
positive regulation of kinase activityD(4) dopamine receptorHomo sapiens (human)
response to histamineD(4) dopamine receptorHomo sapiens (human)
social behaviorD(4) dopamine receptorHomo sapiens (human)
regulation of dopamine metabolic processD(4) dopamine receptorHomo sapiens (human)
dopamine metabolic processD(4) dopamine receptorHomo sapiens (human)
fear responseD(4) dopamine receptorHomo sapiens (human)
regulation of circadian rhythmD(4) dopamine receptorHomo sapiens (human)
positive regulation of MAP kinase activityD(4) dopamine receptorHomo sapiens (human)
behavioral response to cocaineD(4) dopamine receptorHomo sapiens (human)
behavioral response to ethanolD(4) dopamine receptorHomo sapiens (human)
rhythmic processD(4) dopamine receptorHomo sapiens (human)
arachidonic acid secretionD(4) dopamine receptorHomo sapiens (human)
negative regulation of protein secretionD(4) dopamine receptorHomo sapiens (human)
positive regulation of dopamine uptake involved in synaptic transmissionD(4) dopamine receptorHomo sapiens (human)
inhibitory postsynaptic potentialD(4) dopamine receptorHomo sapiens (human)
regulation of postsynaptic neurotransmitter receptor internalizationD(4) dopamine receptorHomo sapiens (human)
negative regulation of voltage-gated calcium channel activityD(4) dopamine receptorHomo sapiens (human)
adenylate cyclase-inhibiting serotonin receptor signaling pathwayD(4) dopamine receptorHomo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerD(4) dopamine receptorHomo sapiens (human)
chemical synaptic transmissionD(4) dopamine receptorHomo sapiens (human)
synaptic transmission, dopaminergicD(1B) dopamine receptorHomo sapiens (human)
response to amphetamineD(1B) dopamine receptorHomo sapiens (human)
regulation of systemic arterial blood pressure by vasopressinD(1B) dopamine receptorHomo sapiens (human)
norepinephrine-epinephrine vasoconstriction involved in regulation of systemic arterial blood pressureD(1B) dopamine receptorHomo sapiens (human)
intracellular calcium ion homeostasisD(1B) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating G protein-coupled receptor signaling pathwayD(1B) dopamine receptorHomo sapiens (human)
activation of adenylate cyclase activityD(1B) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating dopamine receptor signaling pathwayD(1B) dopamine receptorHomo sapiens (human)
chemical synaptic transmissionD(1B) dopamine receptorHomo sapiens (human)
associative learningD(1B) dopamine receptorHomo sapiens (human)
transmission of nerve impulseD(1B) dopamine receptorHomo sapiens (human)
negative regulation of NAD(P)H oxidase activityD(1B) dopamine receptorHomo sapiens (human)
wound healingD(1B) dopamine receptorHomo sapiens (human)
response to cocaineD(1B) dopamine receptorHomo sapiens (human)
positive regulation of adenylate cyclase activityD(1B) dopamine receptorHomo sapiens (human)
negative regulation of blood pressureD(1B) dopamine receptorHomo sapiens (human)
regulation of female receptivityD(1B) dopamine receptorHomo sapiens (human)
sensitizationD(1B) dopamine receptorHomo sapiens (human)
phospholipase C-activating dopamine receptor signaling pathwayD(1B) dopamine receptorHomo sapiens (human)
long-term synaptic depressionD(1B) dopamine receptorHomo sapiens (human)
cellular response to catecholamine stimulusD(1B) dopamine receptorHomo sapiens (human)
reactive oxygen species metabolic processD(1B) dopamine receptorHomo sapiens (human)
positive regulation of MAPK cascadeD(1B) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayD(1B) dopamine receptorHomo sapiens (human)
dopamine receptor signaling pathwayD(1B) dopamine receptorHomo sapiens (human)
temperature homeostasis5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of cytokine production involved in immune response5-hydroxytryptamine receptor 2AHomo sapiens (human)
glycolytic process5-hydroxytryptamine receptor 2AHomo sapiens (human)
intracellular calcium ion homeostasis5-hydroxytryptamine receptor 2AHomo sapiens (human)
activation of phospholipase C activity5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of cytosolic calcium ion concentration5-hydroxytryptamine receptor 2AHomo sapiens (human)
memory5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of cell population proliferation5-hydroxytryptamine receptor 2AHomo sapiens (human)
response to xenobiotic stimulus5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of phosphatidylinositol biosynthetic process5-hydroxytryptamine receptor 2AHomo sapiens (human)
regulation of dopamine secretion5-hydroxytryptamine receptor 2AHomo sapiens (human)
artery smooth muscle contraction5-hydroxytryptamine receptor 2AHomo sapiens (human)
urinary bladder smooth muscle contraction5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of heat generation5-hydroxytryptamine receptor 2AHomo sapiens (human)
negative regulation of potassium ion transport5-hydroxytryptamine receptor 2AHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transduction5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of neuron apoptotic process5-hydroxytryptamine receptor 2AHomo sapiens (human)
protein localization to cytoskeleton5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of fat cell differentiation5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of glycolytic process5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of vasoconstriction5-hydroxytryptamine receptor 2AHomo sapiens (human)
symbiont entry into host cell5-hydroxytryptamine receptor 2AHomo sapiens (human)
sensitization5-hydroxytryptamine receptor 2AHomo sapiens (human)
behavioral response to cocaine5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of inflammatory response5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylation5-hydroxytryptamine receptor 2AHomo sapiens (human)
detection of temperature stimulus involved in sensory perception of pain5-hydroxytryptamine receptor 2AHomo sapiens (human)
detection of mechanical stimulus involved in sensory perception of pain5-hydroxytryptamine receptor 2AHomo sapiens (human)
release of sequestered calcium ion into cytosol5-hydroxytryptamine receptor 2AHomo sapiens (human)
negative regulation of synaptic transmission, glutamatergic5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascade5-hydroxytryptamine receptor 2AHomo sapiens (human)
G protein-coupled serotonin receptor signaling pathway5-hydroxytryptamine receptor 2AHomo sapiens (human)
presynaptic modulation of chemical synaptic transmission5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of execution phase of apoptosis5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of platelet aggregation5-hydroxytryptamine receptor 2AHomo sapiens (human)
positive regulation of DNA biosynthetic process5-hydroxytryptamine receptor 2AHomo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messenger5-hydroxytryptamine receptor 2AHomo sapiens (human)
phospholipase C-activating serotonin receptor signaling pathway5-hydroxytryptamine receptor 2AHomo sapiens (human)
serotonin receptor signaling pathway5-hydroxytryptamine receptor 2AHomo sapiens (human)
chemical synaptic transmission5-hydroxytryptamine receptor 2AHomo sapiens (human)
behavioral fear response5-hydroxytryptamine receptor 2CHomo sapiens (human)
intracellular calcium ion homeostasis5-hydroxytryptamine receptor 2CHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathway5-hydroxytryptamine receptor 2CHomo sapiens (human)
phospholipase C-activating serotonin receptor signaling pathway5-hydroxytryptamine receptor 2CHomo sapiens (human)
locomotory behavior5-hydroxytryptamine receptor 2CHomo sapiens (human)
feeding behavior5-hydroxytryptamine receptor 2CHomo sapiens (human)
positive regulation of phosphatidylinositol biosynthetic process5-hydroxytryptamine receptor 2CHomo sapiens (human)
cGMP-mediated signaling5-hydroxytryptamine receptor 2CHomo sapiens (human)
regulation of nervous system process5-hydroxytryptamine receptor 2CHomo sapiens (human)
regulation of appetite5-hydroxytryptamine receptor 2CHomo sapiens (human)
regulation of corticotropin-releasing hormone secretion5-hydroxytryptamine receptor 2CHomo sapiens (human)
positive regulation of fat cell differentiation5-hydroxytryptamine receptor 2CHomo sapiens (human)
positive regulation of calcium-mediated signaling5-hydroxytryptamine receptor 2CHomo sapiens (human)
release of sequestered calcium ion into cytosol5-hydroxytryptamine receptor 2CHomo sapiens (human)
positive regulation of ERK1 and ERK2 cascade5-hydroxytryptamine receptor 2CHomo sapiens (human)
G protein-coupled serotonin receptor signaling pathway5-hydroxytryptamine receptor 2CHomo sapiens (human)
serotonin receptor signaling pathway5-hydroxytryptamine receptor 2CHomo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messenger5-hydroxytryptamine receptor 2CHomo sapiens (human)
chemical synaptic transmission5-hydroxytryptamine receptor 2CHomo sapiens (human)
smooth muscle contraction5-hydroxytryptamine receptor 7Homo sapiens (human)
circadian rhythm5-hydroxytryptamine receptor 7Homo sapiens (human)
blood circulation5-hydroxytryptamine receptor 7Homo sapiens (human)
vasoconstriction5-hydroxytryptamine receptor 7Homo sapiens (human)
G protein-coupled serotonin receptor signaling pathway5-hydroxytryptamine receptor 7Homo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messenger5-hydroxytryptamine receptor 7Homo sapiens (human)
chemical synaptic transmission5-hydroxytryptamine receptor 7Homo sapiens (human)
inflammatory responseHistamine H1 receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayHistamine H1 receptorHomo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayHistamine H1 receptorHomo sapiens (human)
memoryHistamine H1 receptorHomo sapiens (human)
visual learningHistamine H1 receptorHomo sapiens (human)
regulation of vascular permeabilityHistamine H1 receptorHomo sapiens (human)
positive regulation of vasoconstrictionHistamine H1 receptorHomo sapiens (human)
regulation of synaptic plasticityHistamine H1 receptorHomo sapiens (human)
cellular response to histamineHistamine H1 receptorHomo sapiens (human)
G protein-coupled serotonin receptor signaling pathwayHistamine H1 receptorHomo sapiens (human)
G protein-coupled receptor signaling pathway, coupled to cyclic nucleotide second messengerHistamine H1 receptorHomo sapiens (human)
chemical synaptic transmissionHistamine H1 receptorHomo sapiens (human)
response to ethanolD(3) dopamine receptorHomo sapiens (human)
synaptic transmission, dopaminergicD(3) dopamine receptorHomo sapiens (human)
G protein-coupled receptor internalizationD(3) dopamine receptorHomo sapiens (human)
intracellular calcium ion homeostasisD(3) dopamine receptorHomo sapiens (human)
G protein-coupled receptor signaling pathwayD(3) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating dopamine receptor signaling pathwayD(3) dopamine receptorHomo sapiens (human)
adenylate cyclase-inhibiting dopamine receptor signaling pathwayD(3) dopamine receptorHomo sapiens (human)
learning or memoryD(3) dopamine receptorHomo sapiens (human)
learningD(3) dopamine receptorHomo sapiens (human)
locomotory behaviorD(3) dopamine receptorHomo sapiens (human)
visual learningD(3) dopamine receptorHomo sapiens (human)
response to xenobiotic stimulusD(3) dopamine receptorHomo sapiens (human)
regulation of dopamine secretionD(3) dopamine receptorHomo sapiens (human)
positive regulation of cytokinesisD(3) dopamine receptorHomo sapiens (human)
circadian regulation of gene expressionD(3) dopamine receptorHomo sapiens (human)
response to histamineD(3) dopamine receptorHomo sapiens (human)
social behaviorD(3) dopamine receptorHomo sapiens (human)
response to cocaineD(3) dopamine receptorHomo sapiens (human)
dopamine metabolic processD(3) dopamine receptorHomo sapiens (human)
response to morphineD(3) dopamine receptorHomo sapiens (human)
negative regulation of blood pressureD(3) dopamine receptorHomo sapiens (human)
positive regulation of mitotic nuclear divisionD(3) dopamine receptorHomo sapiens (human)
acid secretionD(3) dopamine receptorHomo sapiens (human)
behavioral response to cocaineD(3) dopamine receptorHomo sapiens (human)
negative regulation of oligodendrocyte differentiationD(3) dopamine receptorHomo sapiens (human)
arachidonic acid secretionD(3) dopamine receptorHomo sapiens (human)
negative regulation of protein secretionD(3) dopamine receptorHomo sapiens (human)
musculoskeletal movement, spinal reflex actionD(3) dopamine receptorHomo sapiens (human)
regulation of dopamine uptake involved in synaptic transmissionD(3) dopamine receptorHomo sapiens (human)
negative regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionD(3) dopamine receptorHomo sapiens (human)
prepulse inhibitionD(3) dopamine receptorHomo sapiens (human)
positive regulation of dopamine receptor signaling pathwayD(3) dopamine receptorHomo sapiens (human)
negative regulation of adenylate cyclase activityD(3) dopamine receptorHomo sapiens (human)
adenylate cyclase-activating adrenergic receptor signaling pathwayD(3) dopamine receptorHomo sapiens (human)
negative regulation of voltage-gated calcium channel activityD(3) dopamine receptorHomo sapiens (human)
regulation of potassium ion transportD(3) dopamine receptorHomo sapiens (human)
phospholipase C-activating dopamine receptor signaling pathwayD(3) dopamine receptorHomo sapiens (human)
positive regulation of MAPK cascadeD(3) dopamine receptorHomo sapiens (human)
negative regulation of cytosolic calcium ion concentrationD(3) dopamine receptorHomo sapiens (human)
negative regulation of synaptic transmission, glutamatergicD(3) dopamine receptorHomo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by hormonePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane depolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inositol phosphate metabolic processInositol hexakisphosphate kinase 1Homo sapiens (human)
phosphatidylinositol phosphate biosynthetic processInositol hexakisphosphate kinase 1Homo sapiens (human)
negative regulation of cold-induced thermogenesisInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol phosphate biosynthetic processInositol hexakisphosphate kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (54)

Processvia Protein(s)Taxonomy
cytokine activityInterferon betaHomo sapiens (human)
cytokine receptor bindingInterferon betaHomo sapiens (human)
type I interferon receptor bindingInterferon betaHomo sapiens (human)
protein bindingInterferon betaHomo sapiens (human)
chloramphenicol O-acetyltransferase activityInterferon betaHomo sapiens (human)
TAP bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
signaling receptor bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protein bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
peptide antigen bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
TAP bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protein-folding chaperone bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
G protein-coupled serotonin receptor activity5-hydroxytryptamine receptor 1AHomo sapiens (human)
protein binding5-hydroxytryptamine receptor 1AHomo sapiens (human)
receptor-receptor interaction5-hydroxytryptamine receptor 1AHomo sapiens (human)
neurotransmitter receptor activity5-hydroxytryptamine receptor 1AHomo sapiens (human)
serotonin binding5-hydroxytryptamine receptor 1AHomo sapiens (human)
dopamine neurotransmitter receptor activity, coupled via Gi/GoD(2) dopamine receptorHomo sapiens (human)
G-protein alpha-subunit bindingD(2) dopamine receptorHomo sapiens (human)
protein bindingD(2) dopamine receptorHomo sapiens (human)
heterotrimeric G-protein bindingD(2) dopamine receptorHomo sapiens (human)
dopamine bindingD(2) dopamine receptorHomo sapiens (human)
ionotropic glutamate receptor bindingD(2) dopamine receptorHomo sapiens (human)
identical protein bindingD(2) dopamine receptorHomo sapiens (human)
heterocyclic compound bindingD(2) dopamine receptorHomo sapiens (human)
G protein-coupled receptor activityD(2) dopamine receptorHomo sapiens (human)
dopamine neurotransmitter receptor activity, coupled via GsD(1A) dopamine receptorHomo sapiens (human)
G-protein alpha-subunit bindingD(1A) dopamine receptorHomo sapiens (human)
dopamine neurotransmitter receptor activityD(1A) dopamine receptorHomo sapiens (human)
protein bindingD(1A) dopamine receptorHomo sapiens (human)
heterotrimeric G-protein bindingD(1A) dopamine receptorHomo sapiens (human)
dopamine bindingD(1A) dopamine receptorHomo sapiens (human)
arrestin family protein bindingD(1A) dopamine receptorHomo sapiens (human)
G protein-coupled receptor activityD(1A) dopamine receptorHomo sapiens (human)
dopamine neurotransmitter receptor activity, coupled via Gi/GoD(4) dopamine receptorHomo sapiens (human)
dopamine neurotransmitter receptor activityD(4) dopamine receptorHomo sapiens (human)
protein bindingD(4) dopamine receptorHomo sapiens (human)
potassium channel regulator activityD(4) dopamine receptorHomo sapiens (human)
SH3 domain bindingD(4) dopamine receptorHomo sapiens (human)
dopamine bindingD(4) dopamine receptorHomo sapiens (human)
identical protein bindingD(4) dopamine receptorHomo sapiens (human)
metal ion bindingD(4) dopamine receptorHomo sapiens (human)
epinephrine bindingD(4) dopamine receptorHomo sapiens (human)
norepinephrine bindingD(4) dopamine receptorHomo sapiens (human)
G protein-coupled serotonin receptor activityD(4) dopamine receptorHomo sapiens (human)
neurotransmitter receptor activityD(4) dopamine receptorHomo sapiens (human)
serotonin bindingD(4) dopamine receptorHomo sapiens (human)
dopamine neurotransmitter receptor activity, coupled via GsD(1B) dopamine receptorHomo sapiens (human)
dopamine neurotransmitter receptor activityD(1B) dopamine receptorHomo sapiens (human)
protein bindingD(1B) dopamine receptorHomo sapiens (human)
dopamine bindingD(1B) dopamine receptorHomo sapiens (human)
G protein-coupled receptor activityD(1B) dopamine receptorHomo sapiens (human)
Gq/11-coupled serotonin receptor activity5-hydroxytryptamine receptor 2AHomo sapiens (human)
virus receptor activity5-hydroxytryptamine receptor 2AHomo sapiens (human)
G protein-coupled serotonin receptor activity5-hydroxytryptamine receptor 2AHomo sapiens (human)
protein binding5-hydroxytryptamine receptor 2AHomo sapiens (human)
protein tyrosine kinase activator activity5-hydroxytryptamine receptor 2AHomo sapiens (human)
identical protein binding5-hydroxytryptamine receptor 2AHomo sapiens (human)
protein-containing complex binding5-hydroxytryptamine receptor 2AHomo sapiens (human)
serotonin binding5-hydroxytryptamine receptor 2AHomo sapiens (human)
1-(4-iodo-2,5-dimethoxyphenyl)propan-2-amine binding5-hydroxytryptamine receptor 2AHomo sapiens (human)
neurotransmitter receptor activity5-hydroxytryptamine receptor 2AHomo sapiens (human)
Gq/11-coupled serotonin receptor activity5-hydroxytryptamine receptor 2CHomo sapiens (human)
G protein-coupled serotonin receptor activity5-hydroxytryptamine receptor 2CHomo sapiens (human)
protein binding5-hydroxytryptamine receptor 2CHomo sapiens (human)
identical protein binding5-hydroxytryptamine receptor 2CHomo sapiens (human)
serotonin binding5-hydroxytryptamine receptor 2CHomo sapiens (human)
1-(4-iodo-2,5-dimethoxyphenyl)propan-2-amine binding5-hydroxytryptamine receptor 2CHomo sapiens (human)
neurotransmitter receptor activity5-hydroxytryptamine receptor 2CHomo sapiens (human)
protein binding5-hydroxytryptamine receptor 7Homo sapiens (human)
G protein-coupled serotonin receptor activity5-hydroxytryptamine receptor 7Homo sapiens (human)
neurotransmitter receptor activity5-hydroxytryptamine receptor 7Homo sapiens (human)
histamine receptor activityHistamine H1 receptorHomo sapiens (human)
G protein-coupled serotonin receptor activityHistamine H1 receptorHomo sapiens (human)
neurotransmitter receptor activityHistamine H1 receptorHomo sapiens (human)
dopamine neurotransmitter receptor activity, coupled via Gi/GoD(3) dopamine receptorHomo sapiens (human)
protein bindingD(3) dopamine receptorHomo sapiens (human)
G protein-coupled receptor activityD(3) dopamine receptorHomo sapiens (human)
transcription cis-regulatory region bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ubiquitin protein ligase bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
identical protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein homodimerization activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
C3HC4-type RING finger domain bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inositol-1,3,4,5,6-pentakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol heptakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 5-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
protein bindingInositol hexakisphosphate kinase 1Homo sapiens (human)
ATP bindingInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 1-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 3-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol 5-diphosphate pentakisphosphate 5-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol diphosphate tetrakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (58)

Processvia Protein(s)Taxonomy
extracellular spaceInterferon betaHomo sapiens (human)
extracellular regionInterferon betaHomo sapiens (human)
Golgi membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
endoplasmic reticulumHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
Golgi apparatusHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
plasma membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
cell surfaceHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
ER to Golgi transport vesicle membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
secretory granule membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
phagocytic vesicle membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
early endosome membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
recycling endosome membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular exosomeHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
lumenal side of endoplasmic reticulum membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
MHC class I protein complexHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular spaceHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
external side of plasma membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
plasma membrane5-hydroxytryptamine receptor 1AHomo sapiens (human)
synapse5-hydroxytryptamine receptor 1AHomo sapiens (human)
plasma membrane5-hydroxytryptamine receptor 1AHomo sapiens (human)
dendrite5-hydroxytryptamine receptor 1AHomo sapiens (human)
Golgi membraneD(2) dopamine receptorHomo sapiens (human)
acrosomal vesicleD(2) dopamine receptorHomo sapiens (human)
plasma membraneD(2) dopamine receptorHomo sapiens (human)
ciliumD(2) dopamine receptorHomo sapiens (human)
lateral plasma membraneD(2) dopamine receptorHomo sapiens (human)
endocytic vesicleD(2) dopamine receptorHomo sapiens (human)
axonD(2) dopamine receptorHomo sapiens (human)
dendriteD(2) dopamine receptorHomo sapiens (human)
synaptic vesicle membraneD(2) dopamine receptorHomo sapiens (human)
sperm flagellumD(2) dopamine receptorHomo sapiens (human)
dendritic spineD(2) dopamine receptorHomo sapiens (human)
perikaryonD(2) dopamine receptorHomo sapiens (human)
axon terminusD(2) dopamine receptorHomo sapiens (human)
postsynaptic membraneD(2) dopamine receptorHomo sapiens (human)
ciliary membraneD(2) dopamine receptorHomo sapiens (human)
non-motile ciliumD(2) dopamine receptorHomo sapiens (human)
dopaminergic synapseD(2) dopamine receptorHomo sapiens (human)
GABA-ergic synapseD(2) dopamine receptorHomo sapiens (human)
G protein-coupled receptor complexD(2) dopamine receptorHomo sapiens (human)
glutamatergic synapseD(2) dopamine receptorHomo sapiens (human)
presynaptic membraneD(2) dopamine receptorHomo sapiens (human)
plasma membraneD(2) dopamine receptorHomo sapiens (human)
nucleusD(1A) dopamine receptorHomo sapiens (human)
endoplasmic reticulum membraneD(1A) dopamine receptorHomo sapiens (human)
plasma membraneD(1A) dopamine receptorHomo sapiens (human)
ciliumD(1A) dopamine receptorHomo sapiens (human)
presynaptic membraneD(1A) dopamine receptorHomo sapiens (human)
dendritic spineD(1A) dopamine receptorHomo sapiens (human)
postsynaptic membraneD(1A) dopamine receptorHomo sapiens (human)
ciliary membraneD(1A) dopamine receptorHomo sapiens (human)
non-motile ciliumD(1A) dopamine receptorHomo sapiens (human)
glutamatergic synapseD(1A) dopamine receptorHomo sapiens (human)
GABA-ergic synapseD(1A) dopamine receptorHomo sapiens (human)
G protein-coupled receptor complexD(1A) dopamine receptorHomo sapiens (human)
plasma membraneD(1A) dopamine receptorHomo sapiens (human)
centrosomeD(4) dopamine receptorHomo sapiens (human)
plasma membraneD(4) dopamine receptorHomo sapiens (human)
membraneD(4) dopamine receptorHomo sapiens (human)
postsynapseD(4) dopamine receptorHomo sapiens (human)
glutamatergic synapseD(4) dopamine receptorHomo sapiens (human)
plasma membraneD(4) dopamine receptorHomo sapiens (human)
dendriteD(4) dopamine receptorHomo sapiens (human)
plasma membraneD(1B) dopamine receptorHomo sapiens (human)
ciliumD(1B) dopamine receptorHomo sapiens (human)
brush border membraneD(1B) dopamine receptorHomo sapiens (human)
synapseD(1B) dopamine receptorHomo sapiens (human)
ciliary membraneD(1B) dopamine receptorHomo sapiens (human)
non-motile ciliumD(1B) dopamine receptorHomo sapiens (human)
plasma membraneD(1B) dopamine receptorHomo sapiens (human)
neurofilament5-hydroxytryptamine receptor 2AHomo sapiens (human)
plasma membrane5-hydroxytryptamine receptor 2AHomo sapiens (human)
caveola5-hydroxytryptamine receptor 2AHomo sapiens (human)
axon5-hydroxytryptamine receptor 2AHomo sapiens (human)
cytoplasmic vesicle5-hydroxytryptamine receptor 2AHomo sapiens (human)
presynaptic membrane5-hydroxytryptamine receptor 2AHomo sapiens (human)
neuronal cell body5-hydroxytryptamine receptor 2AHomo sapiens (human)
dendritic shaft5-hydroxytryptamine receptor 2AHomo sapiens (human)
postsynaptic membrane5-hydroxytryptamine receptor 2AHomo sapiens (human)
cell body fiber5-hydroxytryptamine receptor 2AHomo sapiens (human)
glutamatergic synapse5-hydroxytryptamine receptor 2AHomo sapiens (human)
G protein-coupled serotonin receptor complex5-hydroxytryptamine receptor 2AHomo sapiens (human)
plasma membrane5-hydroxytryptamine receptor 2AHomo sapiens (human)
dendrite5-hydroxytryptamine receptor 2AHomo sapiens (human)
plasma membrane5-hydroxytryptamine receptor 2CHomo sapiens (human)
synapse5-hydroxytryptamine receptor 2CHomo sapiens (human)
G protein-coupled serotonin receptor complex5-hydroxytryptamine receptor 2CHomo sapiens (human)
plasma membrane5-hydroxytryptamine receptor 2CHomo sapiens (human)
dendrite5-hydroxytryptamine receptor 2CHomo sapiens (human)
plasma membrane5-hydroxytryptamine receptor 7Homo sapiens (human)
trans-Golgi network membrane5-hydroxytryptamine receptor 7Homo sapiens (human)
synapse5-hydroxytryptamine receptor 7Homo sapiens (human)
dendrite5-hydroxytryptamine receptor 7Homo sapiens (human)
plasma membrane5-hydroxytryptamine receptor 7Homo sapiens (human)
cytosolHistamine H1 receptorHomo sapiens (human)
plasma membraneHistamine H1 receptorHomo sapiens (human)
synapseHistamine H1 receptorHomo sapiens (human)
dendriteHistamine H1 receptorHomo sapiens (human)
plasma membraneHistamine H1 receptorHomo sapiens (human)
plasma membraneD(3) dopamine receptorHomo sapiens (human)
synapseD(3) dopamine receptorHomo sapiens (human)
plasma membraneD(3) dopamine receptorHomo sapiens (human)
virion membraneSpike glycoproteinSevere acute respiratory syndrome-related coronavirus
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
perinuclear region of cytoplasmPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
fibrillar centerInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleoplasmInositol hexakisphosphate kinase 1Homo sapiens (human)
cytosolInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleusInositol hexakisphosphate kinase 1Homo sapiens (human)
cytoplasmInositol hexakisphosphate kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (97)

Assay IDTitleYearJournalArticle
AID1782425Displacement of [3H]-SCH23390 from D3 receptor (unknown origin) expressed in HEK293T cell membranes measured after 2 hrs by microbeta scintillation counting method2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1595588Partial agonist activity at D2L receptor (unknown origin) expressed in human HTLA cells assessed as beta-arrestin2 recruitment after 20 to 22 hrs by brightglo-luciferase reporter gene assay
AID1054307Agonist activity at human dopamine D2L receptor expressed in CHO cell membranes assessed as inhibition of forskolin-stimulated cAMP production after 30 mins by Alphascreen assay2013Journal of medicinal chemistry, Nov-27, Volume: 56, Issue:22
A structure-activity analysis of biased agonism at the dopamine D2 receptor.
AID1153304Agonist activity at human dopamine D2 short receptor transiently expressed in HEK293 cells co-expressing Galphao1 after 30 mins by [35S]GTPgammaS binding assay relative to quinpirole2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1682820Displacement of [3H]-spiperone from recombinant human D2L receptor expressed in CHO cells
AID1517957Displacement of [3H]-8-OH-DPAT from human 5HT1A receptor expressed in CHO-K1 cell membranes incubated for 60 mins by microbeta scintillation counting analysis
AID1595586Agonist activity at human D2L receptor in HEK293T cells assessed as inhibition of isoproterenol-induced cAMP accumulation preincubated for 15 mins followed by isoproterenol-stimulation and measured by Glosensor-based luminescence assay
AID1153306Agonist activity at human dopamine D2 short receptor transiently expressed in HEK293 cells co-expressing Galphai2 after 30 mins by [35S]GTPgammaS binding assay relative to quinpirole2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1332835Selectivity ratio of IC50 for recombinant human ERG expressed in CHOK1 cells at -80 mV holding potential to Ki for displacement of [3H]8-OH-DPAT from recombinant human 5-HT1A receptor expressed in HEK293 cell membranes2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1517959Displacement of [3H]-Ketanserin from human 5-HT2A receptor expressed in rat cortex tissue incubated for 30 mins by liquid scintillation counting method
AID1153308Agonist activity at human dopamine D3 receptor transiently expressed in HEK293 cells co-expressing Galphao1 after 30 mins by [35S]GTPgammaS binding assay relative to quinpirole2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1782423Partial agonist activity at D2 receptor (unknown origin) expressed in HEK293T cells co-expressing GFP2-beta-arrestin2 assessed as beta-arrestin2 recruitment preincubated for 2 mins with coelenterazine followed by compound addition and measured after 2 min2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1682818Displacement of [3H]methyl-spiperone from recombinant human D3 receptor expressed in CHO cells
AID1332834Selectivity ratio of IC50 for recombinant human ERG expressed in CHOK1 cells at -80 mV holding potential to Ki for displacement of [3H]methyl-spiperone from recombinant human D3 receptor expressed in CHO cell membranes2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1153301Displacement of [3Hprazosin from adrenergic alpha1 receptor in pig cerebral cortex homogenates by competitive binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1153316Partial agonist activity at human dopamine D2 short receptor transiently expressed in HEK293 cells assessed as beta-arrestin recruitment after 6 hrs by chemiluminescence assay relative to control2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1682810Antipsychotic activity in orally dosed Sprague-Dawley rat assessed as inhibition of PCP-induced hyperactivity administered 20 mins before PCP stimulation and measured after 1 hr
AID1054309Displacement of [3H]-spiperone from human dopamine D2L receptor expressed in CHO cell membranes after 3 hrs by liquid scintillation counting analysis2013Journal of medicinal chemistry, Nov-27, Volume: 56, Issue:22
A structure-activity analysis of biased agonism at the dopamine D2 receptor.
AID1153295Displacement of [3H]Spiperone from human dopamine D2 long receptor expressed in CHO cells by competitive binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1782420Partial agonist activity at D2 receptor (unknown origin) expressed in HEK293T cells co-expressing Rluc8-tagged Galpahi1 assessed as Galphai1 dissociation preincubated for 2 mins with coelenterazine followed by compound addition and measured after 2 mins b2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1332833Selectivity ratio of IC50 for recombinant human ERG expressed in CHOK1 cells at -80 mV holding potential to Ki for displacement of [3H]spiperone from recombinant human D2L receptor expressed in CHO cell membranes2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1474767Agonist activity at D2 long receptor (unknown origin) expressed in Flp-In-CHO cells assessed as change in transduction coefficient by measuring cellular impedance measured immediately at 15 secs interval for 90 mins by xCELLigence assay2017Bioorganic & medicinal chemistry letters, 06-01, Volume: 27, Issue:11
Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective.
AID1907047Binding affinity to 5-HT1AR (unknown origin) assessed as inhibition constant
AID1595585Partial agonist activity at human Gi/o-coupled D2R expressed in HEK293T cells assessed as inhibition of isoproterenol-induced cAMP accumulation preincubated for 15 mins followed by isoproterenol-stimulation and measured by Glosensor-based luminescence ass
AID1485399Displacement of [3H]spiperone from human D2-long receptor expressed in CHO cell membranes2017Bioorganic & medicinal chemistry, 07-01, Volume: 25, Issue:13
Development of molecular tools based on the dopamine D
AID1332827Displacement of [3H]ketanserin from recombinant human 5-HT2A receptor expressed in HEK293 cell membranes after 60 mins by scintillation counting method2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1332831Displacement of [3H]pirenzepine from recombinant human M1 receptor expressed in CHO cell membranes at 1 uM after 60 mins by scintillation counting method relative to control2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1153299Displacement of [3H]WAY-100635 from 5-HT1A receptor in pig cerebral cortex homogenates by competitive binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1889749Modulation of human D2L receptor2022European journal of medicinal chemistry, Mar-15, Volume: 232Novel D
AID1782427Displacement of [3H]-LSD from 5HT2A receptor (unknown origin) expressed in HEK293T cell membranes measured after 2 hrs by microbeta scintillation counting method2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1332853Toxicity in Sprague-Dawley rat assessed as cataleptogenic effect by measuring time spent in cataleptic position at 1 to 10 mg/kg, po measured after 0.5 to 2 hrs2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1153309Antagonist activity at human dopamine D2 short receptor transiently expressed in HEK293 cells assessed as inhibition of beta-arrestin recruitment after 6 hrs by chemiluminescence assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1782438Metabolic stability in mouse liver microsomes assessed as half life preincubated for 5 mins followed by NADPH addition measured after 10 mins by LC-MS analysis2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1444853Displacement of [3H]Spiperone from human dopamine D2L receptor expressed in CHO cells2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Synthetic Approaches to the New Drugs Approved During 2015.
AID1332836Selectivity ratio of IC50 for recombinant human ERG expressed in CHOK1 cells at -80 mV holding potential to Ki for displacement of [3H]ketanserin from recombinant human 5-HT2A receptor expressed in HEK293 cell membranes2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1054308Agonist activity at human dopamine D2L receptor expressed in CHO cell membranes assessed as phosphorylation of ERK1/2 by Alphascreen assay2013Journal of medicinal chemistry, Nov-27, Volume: 56, Issue:22
A structure-activity analysis of biased agonism at the dopamine D2 receptor.
AID1332829Antipsychotic activity in Sprague-Dawley rat assessed as inhibition of conditioned avoidance behavior at 1.0 to 2.0 mg/kg, ip administered as single dose measured after 1 hr2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1153307Agonist activity at human dopamine D3 receptor transiently expressed in HEK293 cells co-expressing Galphao1 after 30 mins by [35S]GTPgammaS binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1682816Displacement of [3H]ketanserin from recombinant human 5-HT2A receptor expressed in human HEK293 cells
AID1153310Antagonist activity at human dopamine D2 short receptor transiently expressed in HEK293 cells assessed as inhibition of beta-arrestin recruitment after 6 hrs by chemiluminescence assay relative to qunipirole2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1153296Displacement of [3H]Spiperone from human dopamine D2 short receptor expressed in CHO cells by competitive binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1782422Partial agonist activity at D2 receptor (unknown origin) expressed in HEK293T cells co-expressing GFP2-beta-arrestin2 assessed as beta-arrestin2 recruitment preincubated for 2 mins with coelenterazine followed by compound addition and measured after 2 min2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1782419Displacement of [3H]-N-methylspiperone from D2 receptor (unknown origin) expressed in HEK293T cell membranes measured after 2 hrs by microbeta scintillation counting method2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1474756Agonist activity at D2 long receptor (unknown origin) expressed in Flp-In-CHO cells co-expressing Rluc8-tagged GalphaoB assessed as change in transduction coefficient by measuring GalphaoB activation in presence of coelenterazine by BRET assay2017Bioorganic & medicinal chemistry letters, 06-01, Volume: 27, Issue:11
Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective.
AID1444854Displacement of [3H]Spiperone from human D2S receptor expressed in CHO cells2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Synthetic Approaches to the New Drugs Approved During 2015.
AID1332876Cognitive enhancement effect in Sprague-Dawley rat assessed as reversal of PCP-impaired performance by measuring reduction in time spent exploring novel object in comparison to familiar object at 0.1 mg/kg, po administered 60 mins prior to test by novel o2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1782424Displacement of [3H]-SCH23390 from D1 receptor (unknown origin) expressed in HEK293T cell membranes measured after 2 hrs by microbeta scintillation counting method2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1782426Displacement of [3H]-LSD from 5HT1A receptor (unknown origin) expressed in HEK293T cell membranes measured after 2 hrs by microbeta scintillation counting method2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1332823Selectivity ratio of Ki for displacement of [3H]spiperone from recombinant human D2L receptor expressed in CHO cell membranes to Ki for displacement of [3H]methyl-spiperone from recombinant human D3 receptor expressed in CHO cell membranes2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1517960Displacement of [3H]-raclopride from human D2L receptor expressed in HEK293 cells incubated for 1 hr by liquid scintillation counting method
AID1153305Agonist activity at human dopamine D2 short receptor transiently expressed in HEK293 cells co-expressing Galphai2 after 30 mins by [35S]GTPgammaS binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1604605Half life in schizophrenic patient at 1.5 to 12.5 mg/day2020Journal of medicinal chemistry, 03-26, Volume: 63, Issue:6
Urea Derivatives in Modern Drug Discovery and Medicinal Chemistry.
AID1474759Agonist activity at D2 long receptor (unknown origin) expressed in Flp-In-CHO cells assessed as change in transduction coefficient by measuring inhibition of forskolin-induced cAMP production measured after 5 mins in presence of coelenterazine by BRET ass2017Bioorganic & medicinal chemistry letters, 06-01, Volume: 27, Issue:11
Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective.
AID1485400Displacement of [3H]spiperone from human D2-short receptor expressed in CHO cell membranes2017Bioorganic & medicinal chemistry, 07-01, Volume: 25, Issue:13
Development of molecular tools based on the dopamine D
AID1682817Displacement of [3H]OH-DPAT from recombinant human 5-HT1A receptor expressed in human HEK293 cells
AID1782439Metabolic stability in human liver microsomes assessed as half life preincubated for 5 mins followed by NADPH addition measured after 10 mins by LC-MS analysis2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1517962Displacement of [3H]-5-CT from human 5HT7 receptor expressed in HEK293 cells incubated for 1 hr by liquid scintillation counting method
AID1782429Displacement of [3H]-SCH23390 from D4 receptor (unknown origin) expressed in HEK293T cell membranes measured after 2 hrs by microbeta scintillation counting method2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1332824Displacement of [3H]spiperone from recombinant human D2L receptor expressed in CHO cell membranes after 60 mins by scintillation counting method2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1595587Partial agonist activity at human Gi/o-coupled D2R expressed in HEK293T cells assessed as inhibition of isoproterenol-induced cAMP accumulation preincubated for 15 mins followed by isoproterenol-stimulation and measured by Glosensor-based luminescence ass
AID1332825Displacement of [3H]methyl-spiperone from recombinant human D3 receptor expressed in CHO cell membranes after 60 mins by scintillation counting method2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1682821Displacement of [3H]-prazosin from adrenergic alpha1 in rat cerebral cortex
AID1485398Displacement of [3H]SCH23990 from human D5 receptor expressed in HEK293T cell membranes2017Bioorganic & medicinal chemistry, 07-01, Volume: 25, Issue:13
Development of molecular tools based on the dopamine D
AID1485397Displacement of [3H]SCH23990 from human D1 receptor expressed in HEK293T cell membranes2017Bioorganic & medicinal chemistry, 07-01, Volume: 25, Issue:13
Development of molecular tools based on the dopamine D
AID1332826Displacement of [3H]8-OH-DPAT from recombinant human 5-HT1A receptor expressed in HEK293 cell membranes after 60 mins by scintillation counting method2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1444852Displacement of [3H]Spiperone from dopamine D3 receptor (unknown origin)2017Journal of medicinal chemistry, 08-10, Volume: 60, Issue:15
Synthetic Approaches to the New Drugs Approved During 2015.
AID1485403Displacement of [3H]WAY600135 from porcine cerebral cortex 5-HT1A receptor2017Bioorganic & medicinal chemistry, 07-01, Volume: 25, Issue:13
Development of molecular tools based on the dopamine D
AID1485402Displacement of [3H]spiperone from human D4 receptor expressed in CHO cell membranes2017Bioorganic & medicinal chemistry, 07-01, Volume: 25, Issue:13
Development of molecular tools based on the dopamine D
AID1474757Agonist activity at D2 long receptor (unknown origin) expressed in Flp-In-CHO cells co-expressing Rluc8-tagged Galphai1 assessed as change in transduction coefficient by measuring Galphai1 activation after 5 mins in presence of coelenterazine by BRET assa2017Bioorganic & medicinal chemistry letters, 06-01, Volume: 27, Issue:11
Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective.
AID1332828Reversal of PCP-induced hyperlocomotion activity in ip dosed Sprague-Dawley rat pretreated for 10 mins followed by PCP challenge measured after 1.5 hrs2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1153297Displacement of [3H]Spiperone from human dopamine D3 receptor expressed in CHO cells by competitive binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1153300Displacement of [3H]-ketanserin from human 5HT2A receptor expressed in HEK293 cells by competitive binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1153294Displacement of [3H]SCH23390 from human dopamine D1 receptor expressed in HEK293 cells by competitive binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1153298Displacement of [3H]Spiperone from human dopamine D4.4 receptor expressed in CHO cells by competitive binding assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1332832Inhibition of recombinant human ERG expressed in CHOK1 cells at -80 mV holding potential after 2 mins by patch clamp assay2016European journal of medicinal chemistry, Nov-10, Volume: 123Synthesis and pharmacological characterization of novel N-(trans-4-(2-(4-(benzo[d]isothiazol-3-yl)piperazin-1-yl)ethyl)cyclohexyl)amides as potential multireceptor atypical antipsychotics.
AID1485401Displacement of [3H]spiperone from human D3 receptor expressed in CHO cell membranes2017Bioorganic & medicinal chemistry, 07-01, Volume: 25, Issue:13
Development of molecular tools based on the dopamine D
AID1485404Displacement of [3H]ketanserin from human 5-HT2A receptor expressed in HEK293T cell membranes2017Bioorganic & medicinal chemistry, 07-01, Volume: 25, Issue:13
Development of molecular tools based on the dopamine D
AID1153311Partial agonist activity at human dopamine D2 short receptor transiently expressed in HEK293 cells assessed as inhibition of beta-arrestin recruitment after 6 hrs by chemiluminescence assay2014Journal of medicinal chemistry, Jun-12, Volume: 57, Issue:11
Functionally selective dopamine D₂, D₃ receptor partial agonists.
AID1595590Bias factor, transduction co-efficient for partial agonist activity at human Gi/o-coupled D2R expressed in HEK293T cells assessed as inhibition of isoproterenol-induced cAMP accumulation to transduction co-efficient for partial agonist activity at D2L rec
AID1595589Partial agonist activity at D2L receptor (unknown origin) expressed in human HTLA cells assessed as beta-arrestin2 recruitment after 20 to 22 hrs by brightglo-luciferase reporter gene assay relative to control
AID1474754Displacement of [3H]spiperone from D2 long receptor (unknown origin) expressed in Flp-In-CHO cell membranes assessed as transduction coefficient by measuring dissociation half life after 5 mins by liquid scintillation counter method2017Bioorganic & medicinal chemistry letters, 06-01, Volume: 27, Issue:11
Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective.
AID1782428Displacement of [3H]-LSD from 5HT2C receptor (unknown origin) expressed in HEK293T cell membranes measured after 2 hrs by microbeta scintillation counting method2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1474755Agonist activity at Rluc8-tagged D2 long receptor (unknown origin) expressed in Flp-In-CHO cells co-expressing YFP-tagged beta-arrestin2 assessed as change in transduction coefficient by measuring beta-arrestin2 recruitment measured after 5 mins by BRET a2017Bioorganic & medicinal chemistry letters, 06-01, Volume: 27, Issue:11
Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective.
AID1682819Displacement of [3H]-pyrilamine from human H1 histamine receptor expressed in human HEK293 cells
AID1474758Agonist activity at D2 long receptor (unknown origin) expressed in Flp-In-CHO cells assessed as change in transduction coefficient by measuring induction of ERK1/2 phosphorylation after 5 mins by Alphascreen assay2017Bioorganic & medicinal chemistry letters, 06-01, Volume: 27, Issue:11
Pharmacological property optimization for allosteric ligands: A medicinal chemistry perspective.
AID1782421Partial agonist activity at D2 receptor (unknown origin) expressed in HEK293T cells co-expressing Rluc8-tagged Galpahi1 assessed as Galphai1 dissociation preincubated for 2 mins with coelenterazine followed by compound addition and measured after 2 mins b2021Journal of medicinal chemistry, 12-09, Volume: 64, Issue:23
2-Phenylcyclopropylmethylamine Derivatives as Dopamine D
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID1645871NCATS Parallel Artificial Membrane Permeability Assay (PAMPA) Profiling in pH 5 buffer2022Bioorganic & medicinal chemistry, 02-15, Volume: 56Using in vitro ADME data for lead compound selection: An emphasis on PAMPA pH 5 permeability and oral bioavailability.
AID1347159Primary screen GU Rhodamine qHTS for Zika virus inhibitors: Unlinked NS2B-NS3 protease assay2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1346987P-glycoprotein substrates identified in KB-8-5-11 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1508591NCATS Rat Liver Microsome Stability Profiling2020Scientific reports, 11-26, Volume: 10, Issue:1
Retrospective assessment of rat liver microsomal stability at NCATS: data and QSAR models.
AID1508612NCATS Parallel Artificial Membrane Permeability Assay (PAMPA) Profiling2017Bioorganic & medicinal chemistry, 02-01, Volume: 25, Issue:3
Highly predictive and interpretable models for PAMPA permeability.
AID1346986P-glycoprotein substrates identified in KB-3-1 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1347160Primary screen NINDS Rhodamine qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1645848NCATS Kinetic Aqueous Solubility Profiling2019Bioorganic & medicinal chemistry, 07-15, Volume: 27, Issue:14
Predictive models of aqueous solubility of organic compounds built on A large dataset of high integrity.
AID1345833Human D3 receptor (Dopamine receptors)2012Bioorganic & medicinal chemistry letters, May-15, Volume: 22, Issue:10
Discovery of cariprazine (RGH-188): a novel antipsychotic acting on dopamine D3/D2 receptors.
AID1345788Human D2 receptor (Dopamine receptors)2012Bioorganic & medicinal chemistry letters, May-15, Volume: 22, Issue:10
Discovery of cariprazine (RGH-188): a novel antipsychotic acting on dopamine D3/D2 receptors.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (160)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's1 (0.63)29.6817
2010's101 (63.13)24.3611
2020's58 (36.25)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 104.42

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be very strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index104.42 (24.57)
Research Supply Index5.34 (2.92)
Research Growth Index6.62 (4.65)
Search Engine Demand Index185.40 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (104.42)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials31 (17.51%)5.53%
Reviews49 (27.68%)6.00%
Case Studies11 (6.21%)4.05%
Observational2 (1.13%)0.25%
Other84 (47.46%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (43)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A Double-Blind, Placebo-Controlled Study of Cariprazine as an Adjunct to Antidepressants in the Treatment of Patients With Major Depressive Disorder Who Have Had an Inadequate Response to Antidepressants Alone [NCT03739203]Phase 3752 participants (Actual)Interventional2018-11-10Completed
Evaluation of the Effects of Sequential Multiple-Dose Regimens of Cariprazine on Cardiac Repolarization in Patients With Schizophrenia [NCT01376076]Phase 1129 participants (Actual)Interventional2011-06-30Completed
A Phase 3, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group, Multicenter, Fixed-Dose Clinical Trial Evaluating The Efficacy, Safety And Tolerability Of Cariprazine In Patients With Bipolar I Depression [NCT02670538]Phase 3493 participants (Actual)Interventional2016-03-31Completed
Vraylar® (Cariprazine) in the Treatment of Social Anxiety Disorder: A Double-Blind Study [NCT05384483]Phase 440 participants (Anticipated)Interventional2022-07-15Not yet recruiting
A Randomized, Double-Blind, Placebo-Controlled, Parallel-Group, Multi-Center, Fixed-Dose, Phase 3 Clinical Trial Evaluating the Efficacy and Safety of WID-RGC20 3 mg/Day and 6 mg/Day in Patients With Acute Psychotic Episode of Schizophrenia [NCT05168007]Phase 3342 participants (Anticipated)Interventional2021-11-19Enrolling by invitation
A Double-blind, Placebo-controlled, Randomized Withdrawal, Multicenter Clinical Trial Evaluating the Efficacy, Safety and Tolerability of Cariprazine in a Dose-reduction Paradigm in the Prevention of Relapse in Bipolar I Disorder Patients Whose Current Ep [NCT03573297]Phase 3901 participants (Actual)Interventional2018-06-15Completed
A Randomized, Single-blind, Placebo-controlled Phase II Study to Assess the Effects of Cariprazine on Brain and Behavior in Subjects With Cocaine Use Disorder [NCT03430544]Phase 214 participants (Actual)Interventional2018-04-04Terminated(stopped due to Low recruitment in final grant year, halted on-site research due to COVID-19 during NCE year.)
A Double-Blind, Placebo-Controlled Study of Cariprazine as an Adjunct to Antidepressants in the Treatment of Patients With Major Depressive Disorder Who Have Had an Inadequate Response to Antidepressants Alone [NCT03738215]Phase 3759 participants (Actual)Interventional2018-11-09Completed
A Double-Blind, Placebo-Controlled, Evaluation of the Safety and Efficacy of Cariprazine in Patients With Acute Mania Associated With Bipolar I Disorder [NCT01058668]Phase 3497 participants (Actual)Interventional2010-02-28Completed
A Long-term Open-label Study of the Safety and Tolerability of Cariprazine in Patients With Bipolar I Disorder [NCT01059539]Phase 3403 participants (Actual)Interventional2010-02-28Completed
Effectiveness and Safety of Cariprazine Monotherapy Versus Treatment as Usual for Major Depressive Disorder: A Pragmatic Open Label Randomized Controlled Trial at Sultan Qaboos University Hospital Department of Behavioral Medicine [NCT05933538]Phase 4110 participants (Anticipated)Interventional2024-08-31Not yet recruiting
A 6-week, Multicenter, Randomized, Double-blind, Parallel-group, Placebo-controlled Study of the Efficacy and Safety of Cariprazine in Pediatric Participants (10 to 17 Years of Age) in the Treatment of Depressive Episodes Associated With Bipolar I Disorde [NCT04777357]Phase 3380 participants (Anticipated)Interventional2021-04-28Recruiting
A Double-Blind, Randomized, Placebo-Controlled, Parallel-Group, Fixed-Dose, Phase 2 Study Evaluating the Safety and Efficacy of Cariprazine as an Adjunctive Therapy to Antidepressant Therapies (ADTs), in the Treatment of Subjects With Generalized Anxiety [NCT04965272]Phase 20 participants (Actual)Interventional2021-08-18Withdrawn(stopped due to Strategic Decision)
A Double-Blind, Placebo-Controlled Evaluation of the Safety and Efficacy of RGH-188 in Patients With Acute Mania Associated With Bipolar I Disorder [NCT00488618]Phase 2238 participants (Actual)Interventional2007-06-30Completed
A Single-Dose Study to Evaluate the Pharmacokinetic, Safety, Tolerability Profile and the Effects of Food on the Pharmacokinetics of Different Formulations of Cariprazine in Healthy Male Subjects [NCT02165098]Phase 1160 participants (Actual)Interventional2014-06-30Completed
Phase II Study of MP-214 in Patients With Schizophrenia (Exploratory Study) [NCT00862992]Phase 234 participants (Actual)Interventional2008-04-30Completed
A Double-Blind, Placebo-Controlled Study of Cariprazine (RGH-188) As Adjunctive Therapy In Major Depressive Disorder [NCT00854100]Phase 2231 participants (Actual)Interventional2009-06-30Completed
Pharmacokinetics, Safety, and Tolerability of Cariprazine in Pediatric Participants With Autism Spectrum Disorder Aged 5-17 Years [NCT04382885]Phase 124 participants (Actual)Interventional2020-06-26Completed
A Long-term, Open-label Extension Study of the Safety and Tolerability of RGH-188 (Cariprazine) in Patients With Schizophrenia [NCT00839852]Phase 297 participants (Actual)Interventional2009-05-31Completed
Evaluation of the Safety and Efficacy of RGH-188 in the Acute Exacerbation of Schizophrenia [NCT00694707]Phase 2732 participants (Actual)Interventional2008-06-30Completed
Evaluation of the Long-term Safety, Tolerability, and Pharmacokinetics of Cariprazine in Patients With Schizophrenia [NCT01104792]Phase 3752 participants (Actual)Interventional2010-05-31Completed
A Double-Blind, Placebo-Controlled Evaluation of the Safety and Efficacy of Cariprazine in the Acute Exacerbation of Schizophrenia [NCT01104779]Phase 3446 participants (Actual)Interventional2010-04-27Completed
A Double-blind, Placebo-controlled Study of RGH-188 (Cariprazine) in Bipolar Depression [NCT00852202]Phase 2234 participants (Actual)Interventional2009-06-30Completed
A Double-Blind, Placebo-Controlled, Evaluation of the Safety and Efficacy of Cariprazine in Patients With Acute Mania Associated With Bipolar I Disorder [NCT01058096]Phase 3323 participants (Actual)Interventional2010-02-28Completed
An Exploratory Analysis of Immune and Inflammatory Response Associated With Clozapine Versus Non-Clozapine Antipsychotics in Individuals With Treatment-resistant Schizophrenia [NCT05741502]Phase 460 participants (Anticipated)Interventional2023-08-16Recruiting
A Double-Blind, Placebo and Active-Controlled Evaluation of the Safety and Efficacy of Cariprazine in the Acute Exacerbation of Schizophrenia [NCT01104766]Phase 3617 participants (Actual)Interventional2010-04-23Completed
A 6-week, International, Multicenter, Randomized, Double-blind, Parallel-group, Placebo-controlled Study of the Efficacy and Safety of Cariprazine in the Treatment of Adolescent Participants (13 to 17 Years of Age) With Schizophrenia [NCT03817502]Phase 3330 participants (Anticipated)Interventional2019-06-06Recruiting
A 52-Week, Multicenter, Open-Label, Flexible-dose Study to Evaluate the Long-term Safety and Tolerability of Cariprazine in the Treatment of Pediatric Subjects With Schizophrenia, Bipolar I Disorder, or Autism Spectrum Disorder [NCT04578756]Phase 3280 participants (Anticipated)Interventional2021-02-01Recruiting
A Double-Blind, Placebo-Controlled, Randomized Withdrawal, Multicenter Clinical Trial Evaluating the Efficacy, Safety, and Tolerability of Cariprazine in a Dose-Reduction Paradigm in the Prevention of Relapse in Patients With Schizophrenia [NCT03593213]Phase 3587 participants (Actual)Interventional2018-07-30Terminated(stopped due to FDA Released Allergan from this post marketing requirement)
A Double-Blind, Placebo-Controlled Evaluation of the Safety and Efficacy of RGH-188 in the Acute Exacerbation of Schizophrenia [NCT00404573]Phase 2375 participants (Anticipated)Interventional2006-11-30Completed
An 8-Week, Multicenter, Randomized, Double-Blind, Parallel-Group, Placebo-Controlled, Phase 3 Study of the Efficacy and Safety of Cariprazine in the Treatment of Pediatric Subjects (5 to 17 Years of Age) With Autism Spectrum Disorder [NCT05439616]Phase 3152 participants (Anticipated)Interventional2022-07-07Recruiting
A 6-Week, Double-Blind, Placebo-Controlled Study Evaluating the Efficacy and Safety of Cariprazine in the Acute Exacerbation of Schizophrenia, With an Additional 18-Week Blinded Extension Period [NCT05368558]Phase 3250 participants (Anticipated)Interventional2022-08-18Recruiting
A Randomized, Double-blind, Placebo-controlled, Parallel-group Study of Cariprazine (RGH-188) in the Prevention of Relapse in Patients With Schizophrenia [NCT01412060]Phase 3765 participants (Actual)Interventional2011-09-27Completed
A Phase 2a Randomized, Single-blind, Placebo-controlled Pilot Study to Evaluate the Impact of Cariprazine (1.5mg) on Cocaine Use in OUD-CocUD Patients on Buprenorphine-naloxone. [NCT05063201]Phase 248 participants (Anticipated)Interventional2022-08-05Recruiting
Efficacy of Cariprazine in Improving Cognitive Functioning in Euthymic Patients With Bipolar I Disorder: A Proof of Concept Randomized, Double Blind Placebo Controlled Trial [NCT04771299]Phase 330 participants (Anticipated)Interventional2021-07-07Recruiting
An Open-label, Flexible Dose, Single Site Study Evaluating the Safety, Efficacy and Tolerability of Cariprazine as an Adjunct to Psychostimulants in Adult Patients With ADHD Who Have Had an Inadequate Response to Psychostimulants Alone [NCT04843423]Phase 415 participants (Anticipated)Interventional2021-12-01Recruiting
A Double-Blind, Placebo-Controlled Study of Cariprazine (RGH-188) as Adjunctive Therapy in Major Depressive Disorder [NCT01469377]Phase 2819 participants (Actual)Interventional2011-12-15Completed
A Phase 3, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group, Multicenter, Fixed-Dose Clinical Trial Evaluating The Efficacy, Safety And Tolerability Of Cariprazine In Patients With Bipolar I Depression [NCT02670551]Phase 3488 participants (Actual)Interventional2016-03-17Completed
Lithium Versus Cariprazine in the Acute Phase Treatment of Bipolar Depression: a Pragmatic Head-to-head Open, Randomized Multicenter Study: The 9th Study of the Danish University Antidepressant Group (DUAG 9) [NCT05913947]Phase 4122 participants (Anticipated)Interventional2022-12-13Recruiting
A Double-Blind, Placebo-Controlled Evaluation of the Safety and Efficacy of Cariprazine in Patients With Bipolar Depression [NCT01396447]Phase 2584 participants (Actual)Interventional2011-07-26Completed
A Phase 3, Double-Blind, Placebo-Controlled Study of Cariprazine as Adjunctive Therapy in Major Depressive Disorder [NCT01715805]Phase 31,022 participants (Actual)Interventional2012-11-15Completed
A Phase 3, Long-term, Open-label Study of Safety and Tolerability of Cariprazine as Adjunctive Therapy in Major Depressive Disorder [NCT01838876]Phase 3442 participants (Actual)Interventional2013-04-29Completed
Dopamine D3 Receptor Occupancy in Bipolar Depression by Cariprazine (Vraylar): Evaluating Its Antidepressant Benefit [NCT05060549]Phase 48 participants (Anticipated)Interventional2024-01-07Not yet recruiting
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00488618 (2) [back to overview]Change From Baseline in Clinical Global Impression-Severity (CGI-S) Total Score at Week 3
NCT00488618 (2) [back to overview]Change From Baseline in the Young Mania Rating Scale (YMRS) Total Score at Week 3
NCT00694707 (2) [back to overview]Change From Baseline to Week 6 in the CGI-S Score
NCT00694707 (2) [back to overview]Change From Baseline to Week 6 in the PANSS Total Score
NCT00839852 (2) [back to overview]Change From Baseline to Week 48 in the PANSS Total Score
NCT00839852 (2) [back to overview]Change From Baseline to Week 48 in the CGI-S Score
NCT00852202 (2) [back to overview]Change in Baseline in Clinical Global Impressions-Improvement ( CGI-I )
NCT00852202 (2) [back to overview]Change From Baseline in Montgomery-Asberg Depression Rating Scale (MADRS)
NCT00854100 (2) [back to overview]Clinical Global Impression - Improvement (CGI-I)
NCT00854100 (2) [back to overview]Montgomery-Asberg Depression Rating Scale (MADRS)
NCT00862992 (7) [back to overview]Change From Baseline in the Clinical Global Impression-Severity (CGI-S) Score at Day 14 (Last Observation Carried Forward; LOCF)
NCT00862992 (7) [back to overview]Change From Baseline in Positive and Negative Syndrome Scale (PANSS) Total Score at Day 14 (Last Observation Carried Forward; LOCF)
NCT00862992 (7) [back to overview]The Clinical Global Impression-Improvement (CGI-I) Score at Day 14 (Last Observation Carried Forward; LOCF)
NCT00862992 (7) [back to overview]Area Under the Plasma Concentration-Time Curve From Time Zero to Last of Unchanged MP-214, M7 (Desmethyl Cariprazine) and M6 (Didesmethyl Cariprazine) at Day 14
NCT00862992 (7) [back to overview]Maximum Plasma Concentration of Unchanged MP-214, M7 (Desmethyl Cariprazine) and M6 (Didesmethyl Cariprazine) at Day 14
NCT00862992 (7) [back to overview]Number of Participants With Adverse Event and Adverse Drug Reaction
NCT00862992 (7) [back to overview]Time to Maximum Plasma Concentration of Unchanged MP-214, M7 (Desmethyl Cariprazine) and M6 (Didesmethyl Cariprazine) at Day 14
NCT01058096 (2) [back to overview]Change From Baseline in the Young Mania Rating Scale (YMRS) Total Score at Week 3
NCT01058096 (2) [back to overview]Change From Baseline in Clinical Global Impression-Severity (CGI-S) Total Score at Week 3
NCT01058668 (2) [back to overview]Change From Baseline in Clinical Global Impression-Severity (CGI-S) Total Score at Week 3
NCT01058668 (2) [back to overview]Change From Baseline in the Young Mania Rating Scale (YMRS) Total Score at Week 3
NCT01059539 (2) [back to overview]Change From Baseline in the MADRS Total Score at Week 16
NCT01059539 (2) [back to overview]Change From Baseline in the YMRS Total Score at Week 16
NCT01104766 (2) [back to overview]Measurement of Schizophrenia Symptoms: Change From Baseline in Positive and Negative Syndrome Scale (PANSS) Total Score.
NCT01104766 (2) [back to overview]Measurement of the Overall Severity of Illness: Change From Baseline in Clinical Global Impression-Severity (CGI-S)
NCT01104779 (2) [back to overview]Measurement of Schizophrenia Symptoms: Change From Baseline in Positive and Negative Syndrome Scale (PANSS) Total Score
NCT01104779 (2) [back to overview]Measurement of Schizophrenia Symptoms: Change From Baseline in Clinical Global Impression-Severity (CGI-S)
NCT01104792 (2) [back to overview]Change From Baseline to Week 48 in the PANSS Total Score
NCT01104792 (2) [back to overview]Change From Baseline to Week 48 in the CGI-S Score
NCT01396447 (2) [back to overview]Change From Baseline in the Clinical Global Impressions-Severity Total Score at Week 6
NCT01396447 (2) [back to overview]Change From Baseline in the Montgomery-Åsberg Depression Rating Scale Total Score at Week 6
NCT01412060 (1) [back to overview]Time From Baseline to the First Symptom Relapse During the Double-blind Phase
NCT01469377 (2) [back to overview]Change From Baseline in the Montgomery-Åsberg Depression Rating Scale (MADRS) Total Score at Week 8
NCT01469377 (2) [back to overview]Change From Baseline in the Sheehan Disability Scale (SDS) Total Score at Week 8
NCT01715805 (3) [back to overview]Change From Baseline in Montgomery-Asberg Depression Rating Scale (MADRS) in the Double-Blind Period
NCT01715805 (3) [back to overview]Change From Baseline in Sheehan Disability Scale (SDS) Score in the Double-Blind Period
NCT01715805 (3) [back to overview]Montgomery-Asberg Depression Rating Scale (MADRS) at Baseline in the Double-Blind Period
NCT01838876 (9) [back to overview]Number of Participants With Clinically Significant Changes From Baseline in Electrocardiograms (ECG)
NCT01838876 (9) [back to overview]Number of Participants With Clinically Significant Changes From Baseline in Clinical Laboratory Parameters
NCT01838876 (9) [back to overview]Number of Participants in the Most Severe Suicidal Ideation and Suicidal Behavior Recorded on the C-SSRS During the Treatment Period
NCT01838876 (9) [back to overview]Number of Participants With Clinically Significant Changes From Baseline in Vital Sign Parameters
NCT01838876 (9) [back to overview]Number of Participants With Extrapyramidal Symptom (EPS)-Related TEAEs
NCT01838876 (9) [back to overview]Number of Participants With Newly Emergent Adverse Events (NEAEs) in the Safety Follow-up Period
NCT01838876 (9) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs) in the Treatment Period
NCT01838876 (9) [back to overview]Number of Participants With Treatment-Emergent Ocular Events
NCT01838876 (9) [back to overview]Change From Baseline in the Arizona Sexual Experiences Scale (ASEX) Score
NCT02670538 (2) [back to overview]Change From Baseline in Clinical Global Impressions-Severity (CGI-S) Score
NCT02670538 (2) [back to overview]Change From Baseline in Montgomery-Åsberg Depression Rating Scale (MADRS)
NCT02670551 (2) [back to overview]Change From Baseline in Clinical Global Impressions-Severity (CGI-S) Score at Week 6
NCT02670551 (2) [back to overview]Change From Baseline in the Montgomery-Åsberg Depression Rating Scale (MADRS) Score at Week 6
NCT03573297 (1) [back to overview]Time to First Relapse of Any Mood Episode During the Double-Blind Treatment Period
NCT03593213 (1) [back to overview]Time to First Relapse During Double-blind Treatment Period
NCT03738215 (2) [back to overview]Change From Baseline to Week 6 in the Clinical Global Impressions-Severity (CGI-S) Score
NCT03738215 (2) [back to overview]Total Score Change From Baseline to Week 6 in the MADRS (Montgomery-Åsberg Depression Rating Scale)
NCT03739203 (2) [back to overview]Change From Baseline to Week 6 in the MADRS (Montgomery-Åsberg Depression Rating Scale) Total Score
NCT03739203 (2) [back to overview]Change From Baseline to Week 6 in the Clinical Global Impressions-Severity (CGI-S) Score

Change From Baseline in Clinical Global Impression-Severity (CGI-S) Total Score at Week 3

The CGI-S measures the investigator's assessment of overall severity of the participant's illness compared with the severity of illness in other patients the physician has observed using a 7-point scale (1=Normal, not ill at all to 7=Among the most extremely ill participants). A negative change from Baseline indicates improvement. Analyses are based on ANCOVA model for change from Baseline with treatment group and study center as factors and Baseline CGI-S score as covariate. (NCT00488618)
Timeframe: Baseline, 3 Weeks

Interventionscore on a scale (Least Squares Mean)
Placebo-0.93
Cariprazine-1.57

[back to top]

Change From Baseline in the Young Mania Rating Scale (YMRS) Total Score at Week 3

The YMRS is an 11-item scale that assesses manic symptoms based on the participant's perception of his or her condition over the previous 48 hours, as well as the physician's clinical observations during the interview. The 11-items are elevated mood, increased motor activity-energy, sexual interest, sleep, irritability, rate and amount of speech, language-thought disorder, content, disruptive-aggressive behavior, appearance, and insight. The severity of the abnormality for 7-items are rated on a five-point scale (0-4) and 4-items on a nine-point scale (0-8). The individual scores are summed for a total possible score of 0 (best) to 60 (worst). A negative change from Baseline indicates improvement. Analyses are based on an Analysis of Covariance (ANCOVA) model for change from Baseline with treatment group and study center as factors and Baseline value as covariate. (NCT00488618)
Timeframe: Baseline, Week 3

Interventionscore on a scale (Least Squares Mean)
Placebo-8.91
Cariprazine-15.02

[back to top]

Change From Baseline to Week 6 in the CGI-S Score

The Clinical Global Impressions-Severity (CGI-S) scale is a 7-point scale that measures the overall severity of the illness compared with the severity of illness in other patients the Investigator has observed. The Investigator assesses the severity of the patient's illness as one of the following: 1 = Normal, not at all ill; 2 = Borderline ill; 3 = Mildly ill; 4 = Moderately ill; 5 = Markedly ill; 6 = Severely ill; 7 = Among the most extremely ill patients. The CGI-S score can range from 1 to 7. A higher score indicates more severe illness. A negative change score indicates improvement. (NCT00694707)
Timeframe: Baseline to Week 6

,,,,
InterventionUnits on a scale (Mean)
BaselineChange at Week 6
Cariprazine 1.5 mg4.7-0.9
Cariprazine 3.0 mg4.9-1.1
Cariprazine 4.5 mg4.8-1.2
Placebo4.9-0.6
Risperidone 4.0 mg4.8-1.4

[back to top]

Change From Baseline to Week 6 in the PANSS Total Score

The Positive and Negative Syndrome Scale (PANSS) is a 30-item rating scale that assesses the positive and negative symptoms of individuals with schizophrenia. Responses to the 30 items are based on a structured clinical interview with the patient and on supporting clinical information obtained from family, hospital staff, or other reliable informants. Of the 30 psychiatric parameters measured by the scale, 7 assess positive symptoms (eg, delusions, grandiosity); 7 assess negative symptoms (eg, blunted affect, emotional withdrawal); and 16 assess general psychopathology (eg, poor attention, active social avoidance). Each item is scored on a 7-point scale (1 = absent, 2 = minimal, 3 = mild, 4 = moderate, 5 = moderately severe, 6 = severe, and 7 = extreme). The PANSS total score can range from 30 to 210. A higher score indicates worse symptoms. A negative change score indicates improvement. (NCT00694707)
Timeframe: Baseline to Week 6

,,,,
InterventionUnits on a scale (Mean)
BaselineChange at Week 6
Cariprazine 1.5 mg97.1-17.3
Cariprazine 3.0 mg97.2-18.7
Cariprazine 4.5 mg96.7-20.2
Placebo97.3-9.5
Risperidone 4.0 mg98.1-25.3

[back to top]

Change From Baseline to Week 48 in the PANSS Total Score

The Positive and Negative Syndrome Scale (PANSS) is a 30-item rating scale that assesses the positive and negative symptoms of individuals with schizophrenia. Responses to the 30 items are based on a structured clinical interview with the patient and on supporting clinical information obtained from family, hospital staff, or other reliable informants. Of the 30 psychiatric parameters measured by the scale, 7 assess positive symptoms (eg, delusions, grandiosity); 7 assess negative symptoms (eg, blunted affect, emotional withdrawal); and 16 assess general psychopathology (eg, poor attention, active social avoidance). Each item is scored on a 7-point scale (1 = absent, 2 = minimal, 3 = mild, 4 = moderate, 5 = moderately severe, 6 = severe, and 7 = extreme). The PANSS total score can range from 30 to 210. A higher score indicates worse symptoms. A negative change score indicates improvement. (NCT00839852)
Timeframe: Baseline to Week 48

InterventionUnits on a scale (Mean)
Cariprazine-38.5

[back to top]

Change From Baseline to Week 48 in the CGI-S Score

The Clinical Global Impressions-Severity (CGI-S) scale is a 7-point scale that measures the overall severity of the illness compared with the severity of illness in other patients the Investigator has observed. The Investigator assesses the severity of the patient's illness as one of the following: 1 = Normal, not at all ill; 2 = Borderline ill; 3 = Mildly ill; 4 = Moderately ill; 5 = Markedly ill; 6 = Severely ill; 7 = Among the most extremely ill patients. The CGI-S score can range from 1 to 7. A higher score indicates more severe illness. A negative change score indicates improvement. (NCT00839852)
Timeframe: Baseline to Week 48

InterventionUnits on a scale (Mean)
Cariprazine-2.0

[back to top]

Change in Baseline in Clinical Global Impressions-Improvement ( CGI-I )

The patient was rated on a scale from 1 to 7, with 1 indicating the patient was very much improved and 7 indicating that the patient was very much worse. (NCT00852202)
Timeframe: Baseline to Week 8

InterventionScore on Scale (Mean)
Placebo2.1
0.25 to 0.75 mg Cariprazine2.1
1.5 to 3.0mg Cariprazine2.1

[back to top]

Change From Baseline in Montgomery-Asberg Depression Rating Scale (MADRS)

The patient is rated on a scale from 0-6 on 10 items. Apparent sadness, reported sadness, lassitude, pessimistic thoughts, inner tension, suicidal thoughts, reduced sleep and appetite, concentration difficulties, inability to feel. The overall MADRS score ranges from 0-60, with 0 meaning no symptoms and score of 60 meaning maximum severity. (NCT00852202)
Timeframe: Baseline to Week 8

InterventionScore on scale (Mean)
Placebo29.9
0.25 to 0.75 mg Cariprazine30.2
1.5 to 3.0 mg Cariprazine30.9

[back to top]

Clinical Global Impression - Improvement (CGI-I)

The Clinical Global Impression-Improvement (CGI-I) scale is a clinician rated scale that, in this study, will be used to rate total improvement or worsening of mental illness starting at Visit 2 (Week 2) and taken at every visit through Visit 11 (Week 16). The patient will be rated on a scale from 1 to 7, 1 indicating that the patient is very much improved and 7 indicating that the patient is very much worse. The secondary efficacy parameter was the CGI-I total score at Week 16. (NCT00854100)
Timeframe: Week 16

InterventionUnits on a Scale (Least Squares Mean)
Placebo2.5
Cariprazine 0.1 - 0.3 mg2.5
Cariprazine 1.0 - 2.0 mg2.3

[back to top]

Montgomery-Asberg Depression Rating Scale (MADRS)

The patient is rated on a scale from 0-6 on 10 items. Apparent sadness, reported sadness, lassitude, pessimistic thoughts, inner tension, suicidal thoughts, reduced sleep and appetite, concentration difficulties, inability to feel. The overall MADRS score ranges from 0-60, with 0 meaning no symptoms and score of 60 meaning maximum severity. The primary efficacy parameter was the change in MADRS score totals from the scores taken at Baseline (Week 8) and during at least one more time point up to and including Week 16. (NCT00854100)
Timeframe: Baseline (Week 8) to Week 16

InterventionUnits on a Scale (Least Squares Mean)
Placebo-8.0
Cariprazine 0.1 - 0.3 mg-7.5
Cariprazine 1.0 - 2.0 mg-9.8

[back to top]

Change From Baseline in the Clinical Global Impression-Severity (CGI-S) Score at Day 14 (Last Observation Carried Forward; LOCF)

CGI-S is a 7-point scale to assess the global severity of the participant's illness. CGI-S scores range from 1 (normal, not ill at all) to 7 (extremely ill). (NCT00862992)
Timeframe: at baseline, and on Day 14 or a last observation carried forward (LOCF)

Interventionunits on a scale (Mean)
Cariprazine 3 mg0.3
Cariprazine 6 mg-0.1
Cariprazine 12.5 mg-0.1

[back to top]

Change From Baseline in Positive and Negative Syndrome Scale (PANSS) Total Score at Day 14 (Last Observation Carried Forward; LOCF)

PANSS score is a 30-item rating scale to assess both the positive and negative symptom syndromes of patients with schizophrenia. Each item is scored on a 7-point scale, from 1 (absent) to 7 (extreme). The PANSS total score of the 30 PANSS items ranges from 30 to 210. High scores indicate greater severity of symptoms. (NCT00862992)
Timeframe: at baseline, and on Day 14 or a last observation carried forward (LOCF)

Interventionunits on a scale (Mean)
Cariprazine 3 mg-0.2
Cariprazine 6 mg-0.6
Cariprazine 12.5 mg-3.9

[back to top]

The Clinical Global Impression-Improvement (CGI-I) Score at Day 14 (Last Observation Carried Forward; LOCF)

CGI-I is a 7-point scale to assess the global improvement of the participant's illness relative to baseline. CGI-I scores range from 1 (very much improved) to 7 (very much worse). (NCT00862992)
Timeframe: on Day 14 or a last observation carried forward (LOCF)

Interventionunits on a scale (Mean)
Cariprazine 3 mg4.1
Cariprazine 6 mg3.7
Cariprazine 12.5 mg3.8

[back to top]

Area Under the Plasma Concentration-Time Curve From Time Zero to Last of Unchanged MP-214, M7 (Desmethyl Cariprazine) and M6 (Didesmethyl Cariprazine) at Day 14

(NCT00862992)
Timeframe: Pre-dose, 3, 4, 6, 8, 24, 48, 72, 96 and 168 hours post-dose of Day 14.

,,
Interventionng*hr/mL (Mean)
Unchanged MP-214M7 (desmethyl cariprazine)M6 (didesmethyl cariprazine)
Cariprazine 12.5 mg1017.96437.645950.73
Cariprazine 3 mg298.36135.251884.24
Cariprazine 6 mg522.38232.383397.12

[back to top]

Maximum Plasma Concentration of Unchanged MP-214, M7 (Desmethyl Cariprazine) and M6 (Didesmethyl Cariprazine) at Day 14

(NCT00862992)
Timeframe: Pre-dose, 3, 4, 6, 8, 24, 48, 72, 96 and 168 hours post-dose of Day 14.

,,
Interventionng/mL (Mean)
Unchanged MP-214M7 (desmethyl cariprazine)M6 (didesmethyl cariprazine)
Cariprazine 12.5 mg36.3911.3754.99
Cariprazine 3 mg9.193.1413.79
Cariprazine 6 mg19.435.8328.08

[back to top]

Number of Participants With Adverse Event and Adverse Drug Reaction

(NCT00862992)
Timeframe: Up to 7 weeks

,,
InterventionParticipants (Count of Participants)
Number of Participants With Adverse eventNumber of Participants With Adverse drug Reaction
Cariprazine 12.5 mg1110
Cariprazine 3 mg95
Cariprazine 6 mg96

[back to top]

Time to Maximum Plasma Concentration of Unchanged MP-214, M7 (Desmethyl Cariprazine) and M6 (Didesmethyl Cariprazine) at Day 14

(NCT00862992)
Timeframe: Pre-dose, 3, 4, 6, 8, 24, 48, 72, 96 and 168 hours post-dose of Day 14.

,,
Interventionhour (Mean)
Unchanged MP-214M7 (desmethyl cariprazine)M6 (didesmethyl cariprazine)
Cariprazine 12.5 mg3.224.365.22
Cariprazine 3 mg3.715.254.79
Cariprazine 6 mg3.485.105.37

[back to top]

Change From Baseline in the Young Mania Rating Scale (YMRS) Total Score at Week 3

The YMRS is an 11-item scale that assesses manic symptoms based on the participant's perception of his or her condition over the previous 48 hours, as well as the physician's clinical observations during the interview. The 11-items are elevated mood, increased motor activity-energy, sexual interest, sleep, irritability, rate and amount of speech, language-thought disorder, content, disruptive-aggressive behavior, appearance, and insight. The severity of the abnormality for 7-items are rated on a 5-point scale (0-4) and 4-items on a 9-point scale (0-8). The individual scores are summed for a total possible score of 0 (best) to 60 (worst). A negative change from Baseline indicates improvement. Analysis was a mixed model for repeated measurements (MMRM) observed cases (OC), with treatment group, pooled study center, visit, treatment group-by-visit interaction as factors, baseline value and baseline-by-visit interaction as covariates and an unstructured covariance matrix. (NCT01058096)
Timeframe: Baseline, Week 3

Interventionscore on a scale (Least Squares Mean)
Placebo-15.3
Cariprazine-19.6

[back to top]

Change From Baseline in Clinical Global Impression-Severity (CGI-S) Total Score at Week 3

The CGI-S measures the investigator's assessment of overall severity of the participant's illness compared with the severity of illness in other patients the physician has observed using a 7-point scale (1=Normal, not ill at all to 7=Among the most extremely ill participants). A negative change from Baseline indicates improvement. Analysis was based on a MMRM using the observed cases (OC) data, with treatment group, pooled study center, visit, treatment group-by-visit interaction as factors, baseline value and baseline-by-visit interaction as covariates and an unstructured covariance matrix. (NCT01058096)
Timeframe: Baseline, Week 3

Interventionscore on a scale (Least Squares Mean)
Placebo-1.3
Cariprazine-1.6

[back to top]

Change From Baseline in Clinical Global Impression-Severity (CGI-S) Total Score at Week 3

The CGI-S measures the investigator's assessment of overall severity of the participant's illness compared with the severity of illness in other patients the physician has observed using a 7-point scale (1=Normal, not ill at all to 7= Among the most extremely ill participants). A negative change from Baseline indicates improvement. Analysis is based on a MMRM using the observed cases data, with treatment group, pooled study center, visit, treatment group-by-visit interaction as factors, baseline value and baseline-by-visit interaction as covariates and an unstructured covariance matrix. (NCT01058668)
Timeframe: Baseline, Week 3

Interventionscore on a scale (Least Squares Mean)
Placebo-1.3
Cariprazine (3-6 mg/Day)-1.9
Cariprazine (6-12 mg/Day)-1.9

[back to top]

Change From Baseline in the Young Mania Rating Scale (YMRS) Total Score at Week 3

The YMRS is an 11-item scale that assesses manic symptoms based on the participant's perception of his or her condition over the previous 48 hours, as well as the physician's clinical observations during the interview. The 11-items are elevated mood, increased motor activity-energy, sexual interest, sleep, irritability, rate and amount of speech, language-thought disorder, content, disruptive-aggressive behavior, appearance, and insight. The severity of the abnormality for 7-items are rated on a five-point scale (0-4) and 4-items on a nine-point scale (0-8). The individual scores are summed for a total possible score of 0 (best) to 60 (worst). A negative change from Baseline indicates improvement. Analysis is a mixed model for repeated measurements (MMRM) using observed cases, with treatment group, pooled study center, visit, treatment group-by-visit interaction as factors, baseline value and baseline-by-visit interaction as covariates and an unstructured covariance matrix. (NCT01058668)
Timeframe: Baseline, Week 3

Interventionscore on a scale (Least Squares Mean)
Placebo-12.5
Cariprazine (3-6 mg/Day)-18.6
Cariprazine (6-12 mg/Day)-18.5

[back to top]

Change From Baseline in the MADRS Total Score at Week 16

The Montgomery-Asberg Depression Rating Scale (MADRS) is a clinician-rated scale that evaluates the patient's depressive symptomatology during the previous week. Patients are rated on 10 items assessing feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty in concentration, and lack of interest. Each item is scored on a 7-point (0-6) scale. The total score can range from 0 to 42. A higher score indicates greater depressive symptomatology. A negative change score indicates improvement. (NCT01059539)
Timeframe: Baseline to Week 16

InterventionUnits on a scale (Mean)
Cariprazine 3-12 mg/Day for 16 Weeks-1.6

[back to top]

Change From Baseline in the YMRS Total Score at Week 16

The Young Mania Rating Scale (YMRS) is an 11-item scale that assesses manic symptoms based on the patient's perception of his or her condition over the previous 48 hours, as well as the physician's clinical observations during the interview. The 11 items are elevated mood, increased motor activity-energy, sexual interest, sleep, irritability, rate and amount of speech, language-thought disorder, content, disruptive-aggressive behavior, appearance, and insight. The severity of each item is rated on a 5-point (0-4) or a 9-point (0-8) scale. The total score of all 11 items can range from 0 to 60. A higher score indicates worse manic symptoms. A negative change score indicates improvement. (NCT01059539)
Timeframe: Baseline to Week 16

InterventionUnits on a scale (Mean)
Cariprazine 3-12 mg/Day for 16 Weeks-15.2

[back to top]

Measurement of Schizophrenia Symptoms: Change From Baseline in Positive and Negative Syndrome Scale (PANSS) Total Score.

The Positive and Negative Syndrome Scale is a 30-item rating scale specifically developed to asses both the positive and negative symptom syndromes of patients with schizophrenia. The PANSS total score is rated based on a structured clinical interview with the patient and supporting clinical information obtained from family, hospital staff, or other reliable informants. This assessment provides scores in 9 clinical domains, including a positive syndrome, a negative syndrome, depression, a composite index, and general psychopathology. Each item is scored on a 7-point (1 to 7) scale, with 1 being minimal impact, and 7 being highest impact. The cumulative score ranges from 30 to 210. A negative change from baseline score indicates improvement. (NCT01104766)
Timeframe: Baseline to Week 6

InterventionUnits on a Scale (Least Squares Mean)
Placebo-14.3
Cariprazine 3.0 mg-20.2
Cariprazine 6.0mg-23.0
Aripiprazole 10.0 mg-21.2

[back to top]

Measurement of the Overall Severity of Illness: Change From Baseline in Clinical Global Impression-Severity (CGI-S)

"The Clinical Global Impressions-Severity scale is a clinician-rated scale that measures the overall severity of a participant's illness in comparison with the severity of illness in other participants the physician has observed. The participant is rated on a scale from 1 to 7 with 1 indicating a normal state and 7 indicating among the most extremely ill participants. A higher score indicates greater illness. A negative change from baseline score indicates improvement." (NCT01104766)
Timeframe: Baseline to Week 6

InterventionUnits on a Scale (Least Squares Mean)
Placebo-1.0
Cariprazine 3.0 mg-1.4
Cariprazine 6.0mg-1.5
Aripiprazole 10.0 mg-1.4

[back to top]

Measurement of Schizophrenia Symptoms: Change From Baseline in Positive and Negative Syndrome Scale (PANSS) Total Score

The Positive and Negative Syndrome Scale is a 30-item rating scale specifically developed to asses both the positive and negative symptom syndromes of patients with schizophrenia. The PANSS total score is rated based on a structured clinical interview with the patient and supporting clinical information obtained from family, hospital staff, or other reliable informants. This assessment provides scores in 9 clinical domains, including a positive syndrome, a negative syndrome, depression, a composite index, and general psychopathology. Each item is scored on a 7-point (1 to 7) scale, with 1 being minimal impact, and 7 being highest impact. The cumulative score ranges from 30 to 210. A negative change score indicates improvement. (NCT01104779)
Timeframe: Baseline to Week 6

InterventionUnits on a Scale (Least Squares Mean)
Placebo-16.0
Cariprazine (3-6 mg/Day)-22.8
Cariprazine (6-9 mg/Day)-25.9

[back to top]

Measurement of Schizophrenia Symptoms: Change From Baseline in Clinical Global Impression-Severity (CGI-S)

"The Clinical Global Impressions-Severity scale is a clinician-rated scale that measures the overall severity of a participant's illness in comparison with the severity of illness in other participants the physician has observed. The participant is rated on a scale from 1 to 7 with 1 indicating a normal state and 7 indicating among the most extremely ill participants. A higher score indicates greater illness. A negative change score indicates improvement." (NCT01104779)
Timeframe: Baseline to Week 6

Interventionunits on a scale (Least Squares Mean)
Placebo-1.0
Cariprazine (3-6 mg/Day)-1.4
Cariprazine (6-9 mg/Day)-1.6

[back to top]

Change From Baseline to Week 48 in the PANSS Total Score

The Positive and Negative Syndrome Scale (PANSS) is a 30-item rating scale that assesses the positive and negative symptoms of individuals with schizophrenia. Responses to the 30 items are based on a structured clinical interview with the patient and on supporting clinical information obtained from family, hospital staff, or other reliable informants. Of the 30 psychiatric parameters measured by the scale, 7 assess positive symptoms (eg, delusions, grandiosity); 7 assess negative symptoms (eg, blunted affect, emotional withdrawal); and 16 assess general psychopathology (eg, poor attention, active social avoidance). Each item is scored on a 7-point scale (1 = absent, 2 = minimal, 3 = mild, 4 = moderate, 5 = moderately severe, 6 = severe, and 7 = extreme). The PANSS total score can range from 30 to 210. A higher score indicates worse symptoms. A negative change score indicates improvement. (NCT01104792)
Timeframe: Baseline to Week 48

InterventionUnits on a scale (Mean)
BaselineChange From Baseline
Cariprazine66.5-5.0

[back to top]

Change From Baseline to Week 48 in the CGI-S Score

The Clinical Global Impressions-Severity (CGI-S) scale is a 7-point scale that measures the overall severity of the illness compared with the severity of illness in other patients the Investigator has observed. The Investigator assesses the severity of the patient's illness as one of the following: 1 = Normal, not at all ill; 2 = Borderline ill; 3 = Mildly ill; 4 = Moderately ill; 5 = Markedly ill; 6 = Severely ill; 7 = Among the most extremely ill patients. The CGI-S score can range from 1 to 7. A higher score indicates more severe illness. A negative change score indicates improvement. (NCT01104792)
Timeframe: Baseline to Week 48

InterventionUnits on a scale (Mean)
BaselineChange From Baseline
Cariprazine3.0-0.1

[back to top]

Change From Baseline in the Clinical Global Impressions-Severity Total Score at Week 6

"The Clinical Global Impressions-Severity scale is a clinician-rated scale that measures the overall severity of a participant's illness in comparison with the severity of illness in other participants the physician has observed. The participant is rated on a scale from 1 to 7 with 1 indicating a normal state and 7 indicating among the most extremely ill participants. A higher score indicates greater illness. A negative change score indicates improvement." (NCT01396447)
Timeframe: Baseline to Week 6

Interventionunits on a scale (Least Squares Mean)
Placebo-1.0
Cariprazine 0.75 mg-1.1
Cariprazine 1.5 mg-1.4
Cariprazine 3.0 mg-1.3

[back to top]

Change From Baseline in the Montgomery-Åsberg Depression Rating Scale Total Score at Week 6

The Montgomery-Åsberg Depression Rating Scale is a 10-item, clinician-rated scale that evaluates the participant's depressive symptomatology during the past week. Participants are rated on items assessing feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty in concentration, and lack of interest. Each item is scored on a 7-point scale with a score of 0 reflecting no symptoms and a score of 6 reflecting symptoms of maximum severity. The scores on the 10 items are summed for a total score that can range from 0 to 60. A higher score indicates greater depression. A negative change score indicates improvement. (NCT01396447)
Timeframe: Baseline to Week 6

Interventionunits on a scale (Least Squares Mean)
Placebo-11.1
Cariprazine 0.75 mg-13.0
Cariprazine 1.5 mg-15.1
Cariprazine 3.0 mg-13.7

[back to top]

Time From Baseline to the First Symptom Relapse During the Double-blind Phase

"Relapse was defined as meeting ≥1 of the following criteria:1-Hospitalization due to worsening of condition;2-increase in Positive and Negative Syndrome Scale(PANSS) total score by ≥30% for participants,scored ≥50 or a ≥10-point increase for participants,scored <50 at randomization;3-increase in Clinical Global Impressions-Severity(CGI-S) score by ≥2 points at Week 20;4-deliberate self-injury or aggressive behaviour;5-suicidal/homicidal ideation judged clinically significant by Investigator;6-score of >4 on 1 or more of following PANSS items:P1,P2,P3,P6,P7,G8 or G14. Second assessment not performed based on Investigator discretion.~PANSS is 30-item rating scale. Each item scored on 7-point scale. Total score ranges from 30 to 210. Lower score indicates fewer schizophrenic symptoms. CGI-S is 7-point scale,measures severity of participant's illness in comparison with others with same diagnosis. Lower score indicates less severe illness. 25th percentile for time to relapse was reported. (NCT01412060)
Timeframe: Up to 34 Weeks and Bi-Weekly thereafter until Week 92

InterventionDays (Number)
Placebo92
Cariprazine224

[back to top]

Change From Baseline in the Montgomery-Åsberg Depression Rating Scale (MADRS) Total Score at Week 8

The MADRS is a clinician-rated scale to assess depressive symptomatology during the preceding week. Participants are rated on 10 items (feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty concentrating, and lack of interest) each on a 7-point scale from 0 (no symptoms) to 6 (symptoms of maximum severity). The total score ranges from 0 to 60 with a higher score indicating more depression. A negative change score indicates improvement. (NCT01469377)
Timeframe: Baseline to Week 8

InterventionUnits on a scale (Least Squares Mean)
Placebo-12.5
Cariprazine 1-2 mg-13.4
Cariprazine 2-4.5 mg-14.6

[back to top]

Change From Baseline in the Sheehan Disability Scale (SDS) Total Score at Week 8

The SDS measures an individual's perception of the extent to which his or her emotional symptoms are disrupting his or her functioning in 3 domains, work/school, social life/leisure activities, and family life/home responsibilities. The participant is asked to rate the degree to which their functioning is impaired on an 11-point scale, ranging from 0 (not at all) to 10 (extremely). Scores of 0 to 3 indicate mild functional impairment, 4 to 6 indicate moderate functional impairment, and 7 to 9 indicate marked functional impairment. The scores for the 3 domains are summed into a total score that ranges from 0 (unimpaired) to 30 (highly impaired). A higher score indicates greater impairment. A negative change score indicates improvement. (NCT01469377)
Timeframe: Baseline to Week 8

InterventionUnits on a scale (Least Squares Mean)
Placebo-6.6
Cariprazine 1-2 mg-7.7
Cariprazine 2-4.5 mg-8.0

[back to top]

Change From Baseline in Montgomery-Asberg Depression Rating Scale (MADRS) in the Double-Blind Period

The MADRS is a clinician-rated scale to assess depressive symptomatology during the past week. Participants are rated on 10 items to assess feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty concentrating and lack of interest. Each item is scored on a 7-point scale. A score of 0 indicates the absence of symptoms, and a score of 6 indicates symptoms of maximum severity for a total possible score of 0 to 60. A negative change from Baseline indicates improvement. Mixed-effects model for repeated measures (MMRM) with treatment group, study center, visit, and treatment group-by-visit interaction as fixed effects, and the baseline value and baseline by-visit interaction as the covariates was used for analyses. (NCT01715805)
Timeframe: Baseline (Week 8) to Week 16

Interventionscore on a scale (Least Squares Mean)
Placebo + ADT (Double-Blind)-7.5
Cariprazine + ADT (Double-Blind)-7.7

[back to top]

Change From Baseline in Sheehan Disability Scale (SDS) Score in the Double-Blind Period

The Sheehan Disability Scale (SDS) is a 3-item patient-rated questionnaire used to evaluate impairments in the domains of work, social life/leisure, and family life/home responsibility. All items are rated on an 11-point continuum from 0 (no impairment) to 10 (most severe). The 3 individual scores are summed for a total possible score of 0 (unimpaired) to 30 (highly impaired). A negative change from Baseline indicates improvement. MMRM with treatment group, study center, visit, and treatment group-by-visit interaction as fixed effects, and the baseline value and baseline by-visit interaction as the covariates was used for analyses. (NCT01715805)
Timeframe: Baseline (Week 8) to Week 16

Interventionscore on a scale (Least Squares Mean)
Placebo + ADT (Double-Blind)-3.1
Cariprazine + ADT (Double-Blind)-3.7

[back to top]

Montgomery-Asberg Depression Rating Scale (MADRS) at Baseline in the Double-Blind Period

The MADRS is a clinician-rated scale to assess depressive symptomatology during the past week. Patients are rated on 10 items to assess feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty concentrating and lack of interest. Each item is scored on a 7-point scale. A score of 0 indicates the absence of symptoms, and a score of 6 indicates symptoms of maximum severity for a total possible score of 0 to 60. (NCT01715805)
Timeframe: Baseline (Week 8)

Interventionscore on a scale (Mean)
Placebo + ADT (Double-Blind)25.2
Cariprazine + ADT (Double-Blind)25.4

[back to top]

Number of Participants With Clinically Significant Changes From Baseline in Electrocardiograms (ECG)

A standard 12-lead ECG was performed. The investigator determined the clinical significance of the ECG findings using the central ECG interpretation laboratory report. (NCT01838876)
Timeframe: Baseline (Week 0) to up to 26 weeks

InterventionParticipants (Count of Participants)
Cariprazine + ADT1

[back to top]

Number of Participants With Clinically Significant Changes From Baseline in Clinical Laboratory Parameters

Clinical laboratory parameters included tests of hematology, chemistry, urinalysis and prolactin. The investigator assessed the results for clinical significance. (NCT01838876)
Timeframe: Baseline (Week 0) to up to 26 weeks in the Treatment Period

InterventionParticipants (Count of Participants)
Cariprazine + ADT0

[back to top]

Number of Participants in the Most Severe Suicidal Ideation and Suicidal Behavior Recorded on the C-SSRS During the Treatment Period

The Columbia-Suicide Severity Rating Scale (C-SSRS) is a clinician-rated instrument that reports the severity of both suicidal ideation and behavior. Suicidal ideation is classified on a 5-item scale: 1 (wish to be dead) to 5 (active suicidal ideation with specific plan and intent). The C-SSRS also captures information about the intensity of ideation, specifically the frequency, duration, controllability, deterrents, and reasons for the most severe types of ideation. Suicidal behavior is classified on a 5-item scale: 0 (no suicidal behavior to 4 (actual attempt). More than 1 classification can be selected provided they represent separate episodes. (NCT01838876)
Timeframe: Baseline (Lead-in study Baseline for roll-over participants and prior to first dose in this study for new participants) to Week 26 in this study

Interventionparticipants (Number)
No Suicidal IdeationSuicidal IdeationSuicidal Ideation with Specific Plan and IntentIdeation,Some Intent to Act,without Specific PlanIdeation,any Methods,without Intent to ActNon-specific Active Suicidal ThoughtsWish to be DeadNo Suicidal BehaviorSuicidal BehaviorCompleted SuicideActual Suicide Attempt
Cariprazine + ADT30837106327344101

[back to top]

Number of Participants With Clinically Significant Changes From Baseline in Vital Sign Parameters

Vital sign parameters included blood pressure, pulse rate, body mass index (BMI), weight, and waist circumference. The investigator assessed the results for clinical significance. (NCT01838876)
Timeframe: Baseline (Week 0) to up to 26 weeks in the Treatment Period plus a 2-week Safety Follow-up Period (Up to 28 weeks)

InterventionParticipants (Count of Participants)
Cariprazine + ADT0

[back to top] [back to top]

Number of Participants With Newly Emergent Adverse Events (NEAEs) in the Safety Follow-up Period

An AE is any untoward medical occurrence in a patient or clinical investigation participant administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. An adverse event can therefore be any unfavorable and unintended sign (i.e. laboratory value), symptom, or disease temporally associated with the use of a medicinal product, whether or not related to the medicinal product. A NEAE is a new AE that occurred during the 2-week Safety Follow-up Period. (NCT01838876)
Timeframe: 2 weeks following the 26-week Treatment Period

InterventionParticipants (Count of Participants)
Cariprazine + ADT20

[back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs) in the Treatment Period

An adverse event (AE) is any untoward medical occurrence in a patient or clinical investigation participant administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. An adverse event can therefore be any unfavorable and unintended sign (i.e. laboratory value), symptom, or disease temporally associated with the use of a medicinal product, whether or not related to the medicinal product. A TEAE is an AE that occurs or worsens after receiving study drug. (NCT01838876)
Timeframe: First dose of study drug to last dose of study drug in the 26-week Treatment Period and within 30 days of last dose of study drug for participants who did not participate in the 2-week Safety Follow-up Period (Up to 30 weeks)

InterventionParticipants (Count of Participants)
Cariprazine + ADT274

[back to top]

Number of Participants With Treatment-Emergent Ocular Events

A TEAE is an AE that occurs or worsens after receiving study drug. Ocular events are adverse events related to the eye. (NCT01838876)
Timeframe: First dose of study drug to last dose of study drug in the 26-week Treatment Period plus a 2-week Safety Follow-up Period or within 30 days of last dose of study drug for participants who did not participate in the Safety Follow-up Period (Up to 30 weeks)

InterventionParticipants (Count of Participants)
Cariprazine + ADT1

[back to top]

Change From Baseline in the Arizona Sexual Experiences Scale (ASEX) Score

The ASEX is a participant-completed scale to evaluate overall sexual experiences over the previous 7 days consisting of 5 questions answered on a scale of 1 (best) to 6 (worst) for a total possible score of 3 to 30 (2 questions were only answered if the participant was sexually active in the past week), higher score indicates greater sexual dysfunction. There are different forms for males and females. A negative change from Baseline indicates improvement. (NCT01838876)
Timeframe: Baseline (Lead-in study Baseline for roll-over participants and prior to first dose of this study for new participants) to End of Treatment (Up to Week 26) in this study

,
Interventionscore on a scale (Mean)
BaselineChange from Baseline to the End of Treatment
Cariprazine + ADT (Female)20.1-0.9
Cariprazine + ADT (Male)17.2-0.1

[back to top]

Change From Baseline in Clinical Global Impressions-Severity (CGI-S) Score

"CGI-S is a clinician-rated scale that measures the overall severity of a participant's illness in comparison with the severity of other patients the physician has observed. The participant was rated on a scale from 1 to 7, with 1 indicating a normal state and 7 indicating among the most extremely ill patients. A negative change from Baseline indicates improvement. MMRM with fixed factors (treatment group, pooled study center, and visit), baseline (a covariate), and interactions (treatment group by visit, baseline by visit)." (NCT02670538)
Timeframe: Baseline (Week 0) to Week 6

Interventionscore on a scale (Least Squares Mean)
Placebo-1.2
Cariprazine 1.5 mg-1.5
Cariprazine 3.0 mg-1.4

[back to top]

Change From Baseline in Montgomery-Åsberg Depression Rating Scale (MADRS)

MADRS is a 10-item, clinician-rated scale that evaluates the participants depressive symptomatology during the past week. Participants were rated on items assessing feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty in concentration, and lack of interest. Each of the 10 items was scored on a 7-point scale with a score of 0 reflecting no symptoms and a score of 6 reflecting symptoms of maximum severity for a total possible score of 0 (best) to 60 (worst). A negative change from Baseline indicates improvement. Mixed-effects Model for Repeated Measures (MMRM) with fixed factors (treatment group, pooled study center, and visit), baseline (a covariate), and interactions (treatment group by visit, baseline by visit). (NCT02670538)
Timeframe: Baseline (Week 0) to Week 6

Interventionscore on a scale (Least Squares Mean)
Placebo-12.4
Cariprazine 1.5 mg-14.8
Cariprazine 3.0 mg-14.1

[back to top]

Change From Baseline in Clinical Global Impressions-Severity (CGI-S) Score at Week 6

"The Clinical Global Impressions-Severity (CGI-S) is a clinician-rated scale that measures the overall severity of a participant's illness in comparison with the severity of other participants the physician has observed. The participant was rated on a scale from 1 to 7, with 1 indicating a normal state and 7 indicating among the most extremely ill participants. A negative change from Baseline indicates improvement." (NCT02670551)
Timeframe: Baseline (Week 0) to Week 6

Interventionscore on a scale (Least Squares Mean)
Placebo-1.3
Cariprazine 1.5 mg-1.6
Cariprazine 3.0 mg-1.6

[back to top]

Change From Baseline in the Montgomery-Åsberg Depression Rating Scale (MADRS) Score at Week 6

The Montgomery-Åsberg Depression Rating Scale (MADRS) is a 10-item, clinician-rated scale that evaluates the participant's depressive symptomatology during the past week. Participants were rated on items assessing feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty in concentration, and lack of interest. Each of the 10 items was scored on a 7-point scale with a score of 0 reflecting no symptoms and a score of 6 reflecting symptoms of maximum severity for a total possible score of 0 (best) to 60 (worst). A negative change from Baseline indicates improvement. (NCT02670551)
Timeframe: Baseline (Week 0) to Week 6

Interventionscore on a scale (Least Squares Mean)
Placebo-12.6
Cariprazine 1.5 mg-15.1
Cariprazine 3.0 mg-15.6

[back to top]

Time to First Relapse of Any Mood Episode During the Double-Blind Treatment Period

"Relapse was defined as the occurrence of any 1 of the following:~Young Mania Rating Score (YMRS) total score ≥ 17 (range 0-60; higher score indicates a worse outcome);~Montgomery Asberg Depression Rating Scale; (MADRS) total score ≥ 20 (range 0-60; higher score indicates more depressive symptoms);~Clinical Global Impression-Improvement scale (CGI-S) ≥ 4 (range from 1 [normal, not at all ill] to 7 [extremely ill]);~Initiation of additional psychiatric medication;~Psychiatric hospitalization;~Exacerbation of illness as judged by clinical impression of the Investigator.~Time to first relapse (days) was calculated as the date of the first relapse - the date of randomization + 1. Participants who did not meet the relapse criteria were considered censored at the time of completion or discontinuation from the Double-Blind Treatment Period (DBTP) of the study. Percentiles (95% Confidence Intervals [CI]) are based on Kaplan-Meier estimates." (NCT03573297)
Timeframe: From Week 16 to Week 55

,,
Interventiondays (Median)
25% percentile50% percentile75% percentile
Double-Blind Cariprazine 1.5 mg QDNANANA
Double-Blind Cariprazine 3.0 mg QDNANANA
Double-Blind Placebo QDNANANA

[back to top]

Time to First Relapse During Double-blind Treatment Period

"Time to Relapse is the number of days from randomization to first relapse.~Relapse is defined as any 1 of the following:~Increase in Positive and Negative Syndrome Scale(PANSS) by ≥30% for participants who had total score of ≥50 at randomization or ≥10-point increased score with total score <50 at randomization [PANSS=30 questions where 1=absence of symptoms to 7=extremely severe symptom;total score=30 to 210;higher score more severe symptoms]~Increase in Clinical Global Impression-Severity (CGI-S) score by 2 or more points [1=normal to 7=among most extremely ill]~Score of >4 on 1 or more of 7 PANSS items:P1-delusions,P2-conceptual disorganization,P3-hallucinatory behavior,P6-suspiciousness,P7-hostility,G8-uncooperativeness,G14-poor impulse control~Deliberate self-injury~Initiation of treatment with mood stabilizer,antidepressant,antipsychotics or benzodiazepine that exceeds specified allowance~Psychiatric hospitalization~Exacerbation of psychiatric illness" (NCT03593213)
Timeframe: Randomization (Week 18) to End of Treatment (Week 44)

Interventiondays (Median)
Placebo (Double-blind Treatment Period)NA
Cariprazine 3.0 mg/Day (Double-blind Treatment Period)NA
Cariprazine 4.5 mg/Day (Double-blind Treatment Period)NA

[back to top]

Change From Baseline to Week 6 in the Clinical Global Impressions-Severity (CGI-S) Score

The CGI-S is a clinician-rated scale used to rate the severity of the participant's current state of mental illness compared with MDD population. The participant was rated on a scale from 1 to 7, where 1=normal, not at all ill and 7=among the most extremely ill participants. Higher scores indicate worsening of mental illness. A negative change from Baseline indicates improvement. MMRM was used for analyses. mITT Population included all randomized participants who had ≥1 postbaseline assessment of the MADRS total score. Number of subjects analyzed are the number of participants with data available for analyses. (NCT03738215)
Timeframe: Baseline and Week 6

Interventionscore on a scale (Least Squares Mean)
Placebo + ADT-1.1
Cariprazine 1.5 mg/Day + ADT-1.4
Cariprazine 3 mg/Day + ADT-1.3

[back to top]

Total Score Change From Baseline to Week 6 in the MADRS (Montgomery-Åsberg Depression Rating Scale)

The MADRS is a 10-item, clinician-rated scale that evaluates the patient's depressive symptomatology during the past week. Patients are rated on items assessing feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty in concentration, and lack of interest. Each item is scored on a 7-point scale with a score of 0 reflecting no symptoms and a score of 6 reflecting symptoms of maximum severity. mITT Population included all randomized participants who had ≥1 postbaseline assessment of the MADRS total score. Number of subjects analyzed are the number of participants with data available for analyses. (NCT03738215)
Timeframe: Baseline and Week 6

Interventionscore on a scale (Least Squares Mean)
Placebo + ADT-11.5
Cariprazine 1.5 mg/Day + ADT-14.1
Cariprazine 3 mg/Day + ADT-13.1

[back to top]

Change From Baseline to Week 6 in the MADRS (Montgomery-Åsberg Depression Rating Scale) Total Score

The MADRS is a 10-item, clinician-rated scale that evaluates the participant's depressive symptomatology during the past week. Participants were rated on items assessing feelings of sadness, lassitude, pessimism, inner tension, suicidality, reduced sleep or appetite, difficulty in concentration, and lack of interest. Each item was scored on a 7-point scale with a score of 0 reflecting no symptoms and a score of 6 reflecting symptoms of maximum severity. The total score ranges from 0 to 60 with a higher score indicating more depression. A negative change from Baseline indicates improvement. Mixed-effects Model for Repeated Measures (MMRM) was used for analyses. (NCT03739203)
Timeframe: Baseline and Week 6

Interventionscore on a scale (Least Squares Mean)
Placebo + ADT-13.4
Cariprazine 1.5 mg/Day + ADT-13.8
Cariprazine 3 mg/Day + ADT-14.8

[back to top]

Change From Baseline to Week 6 in the Clinical Global Impressions-Severity (CGI-S) Score

The CGI-S is a clinician-rated scale used to rate the severity of the participant's current state of mental illness compared with MDD population. The participant was rated on a scale from 1 to 7, where 1=normal, not at all ill and 7=among the most extremely ill participants. Higher score indicates worsening of mental illness. A negative change from Baseline indicates improvement. MMRM was used for analyses. (NCT03739203)
Timeframe: Baseline and Week 6

Interventionscore on a scale (Least Squares Mean)
Placebo + ADT-1.4
Cariprazine 1.5 mg/Day + ADT-1.4
Cariprazine 3 mg/Day + ADT-1.6

[back to top]