Page last updated: 2024-12-11

ganaxolone

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

Description

ganaxolone: a selective, high-affinity, steroid modulator of the GABA(A) receptor; structure given in first source; RN given refers to (3alpha,5alpha)-isomer [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

Cross-References

ID SourceID
PubMed CID6918305
CHEMBL ID1568698
CHEBI ID177658
SCHEMBL ID144522
MeSH IDM0274194

Synonyms (61)

Synonym
CHEBI:177658
1-[(3r,5s,8r,9s,10s,13s,14s,17s)-3-hydroxy-3,10,13-trimethyl-1,2,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydrocyclopenta[a]phenanthren-17-yl]ethanone
ganaxolone
ccd-1042
ganaxolone, solid
ganaxolone (usan/inn)
38398-32-2
D04300
3alpha-hydroxy-3beta-methyl-5alpha-pregnan-20-one
3alpha-hydroxy-3-methyl-5alpha-pregnan-20-one
c22h36o2
ccd 1042
ganaxolone [usan]
(3alpha,5alpha)-3-hydroxy-3-methylpregnan-20-one
pregnan-20-one, 3-hydroxy-3-methyl-, (3alpha,5alpha)-
NCGC00165802-02
NCGC00165802-03
HMS3261K19
ztalmy
dtxsid6046503 ,
tox21_112264
cas-38398-32-2
dtxcid4026503
bdbm50369240
ganaxolone [usan:inn]
98wi44ohiq ,
unii-98wi44ohiq
dea no. 2401
CHEMBL1568698
LP00379
ganaxolone [inn]
3.alpha.-hydroxy-3-methyl-5.alpha.-pregnan-20-one
pregnan-20-one, 3-hydroxy-3-methyl-, (3.alpha.,5.alpha.)-
ganaxolone [who-dd]
ganaxolone [mi]
DB05087
CCG-221683
SCHEMBL144522
PGTVWKLGGCQMBR-FLBATMFCSA-N
NCGC00263545-01
tox21_112264_1
NCGC00261064-01
tox21_500379
(3?,5?)-3-hydroxy-3-methyl-pregnan-20-one
pregnan-20-one, 3-hydroxy-3-methyl-, (3a,5a)-
(3alpha,5alpha)-3-hydroxy-3-methyl-pregnan-20-one
HB0938
AKOS027327586
mfcd09971088
Q3758034
1-((3r,5s,8r,9s,10s,13s,14s,17s)-3-hydroxy-3,10,13-trimethylhexadecahydro-1h-cyclopenta[a]phenanthren-17-yl)ethanone
AS-35253
SDCCGSBI-0633706.P001
AMY38916
1-[(3r,5s,8r,9s,10s,13s,14s,17s)-3-hydroxy-3,10,13-trimethyl-1,2,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydrocyclopenta[a]phenanthren-17-yl]ethanone.
1-((3r,5s,8r,9s,10s,13s,14s,17s)-3-hydroxy-3,10,13-trimethylhexadecahydro-1h-cyclopenta[a]phenanthren-17-yl)ethan-1-one
A901692
ccd1042
gtpl11942
ganaxolona
ganaxolonum

Research Excerpts

Overview

Ganaxolone is a 3beta-methyl-substituted analog of the endogenous neuroactive steroid 3alpha-hydroxy-5alpha-pregnan-20-one. It is an efficacious anticonvulsant agent in a variety of acute seizure models, as well as in electrical and chemical kindling models.

ExcerptReferenceRelevance
"Ganaxolone is an investigational neuroactive steroid in development for RSE treatment."( Intravenous ganaxolone for the treatment of refractory status epilepticus: Results from an open-label, dose-finding, phase 2 trial.
Aimetti, A; Gasior, M; Husain, AM; Ramsay, RE; Swisher, CB; Vaitkevicius, H, 2022
)
1.82
"Ganaxolone (CCD 1042) is a 3beta-methyl-substituted analog of the endogenous neuroactive steroid 3alpha-hydroxy-5alpha-pregnan-20-one. "( Characterization of the anticonvulsant properties of ganaxolone (CCD 1042; 3alpha-hydroxy-3beta-methyl-5alpha-pregnan-20-one), a selective, high-affinity, steroid modulator of the gamma-aminobutyric acid(A) receptor.
Belelli, D; Bolger, MB; Carter, RB; Gee, KW; Hawkinson, JE; Lambert, JJ; Lan, NC; Mirsadeghi, S; Tahir, SH; White, HS; Wieland, S; Wolf, HH; Wood, PL, 1997
)
1.99
"Ganaxolone is an efficacious anticonvulsant agent in a variety of acute seizure models, as well as in electrical and chemical kindling models, and is currently under Phase II clinical investigation for epilepsy."( Reversal of behavioral effects of pentylenetetrazol by the neuroactive steroid ganaxolone.
Beekman, M; Carter, RB; Dijkstra, D; Gasior, M; Goldberg, SR; Ungard, JT; Witkin, JM, 1998
)
1.25
"Ganaxolone is a synthetic neuroactive steroid that positively modulates GABA."( Effect of ganaxolone in a rodent model of cerebral hematoma.
Chong, A; Hassid, S; Jackson-Friedman, C; Lyden, P; Macdonald, RL; Shin, C, 2000
)
1.43

Actions

ExcerptReferenceRelevance
"Ganaxolone promotes remyelination in response to focal demyelination of the corpus callosum of ovariectomized rats. "( Ganaxolone enhances microglial clearance activity and promotes remyelination in focal demyelination in the corpus callosum of ovariectomized rats.
Kalakh, S; Mouihate, A, 2020
)
3.44

Treatment

Ganaxolone treatment was associated with an 86.4% reduction in the number of seizures compared to the asphyxia group. Treatment returned the hyperactive behavioural phenotype of preterm-born juvenile males to a term-born phenotype.

ExcerptReferenceRelevance
"Ganaxolone treatment was associated with an 86.4% reduction in the number of seizures compared to the asphyxia group."( Ganaxolone versus Phenobarbital for Neonatal Seizure Management.
Allison, BJ; Bennet, L; Boyd, BJ; Castillo-Melendez, M; Fahey, MC; Hirst, JJ; Hunt, RW; Jenkin, G; Malhotra, A; McDonald, C; Mihelakis, J; Miller, SL; Nitsos, I; Pham, Y; Sutherland, AE; Walker, DW; Wong, F; Yawno, T, 2022
)
2.89
"Ganaxolone treatment returned the hyperactive behavioural phenotype of preterm-born juvenile males to a term-born phenotype. "( Neurosteroid replacement therapy using the allopregnanolone-analogue ganaxolone following preterm birth in male guinea pigs.
Berry, MJ; Dyson, RM; Gray, C; Hirst, JJ; Palliser, HK; Shaw, JC, 2019
)
2.19
"Ganaxolone treatment of NP-C mice produced beneficial neurological effects, but these effects were not as robust as those obtained using allopregnanolone."( Endogenous and synthetic neurosteroids in treatment of Niemann-Pick Type C disease.
Gong, W; Mellon, SH; Schonemann, MD, 2008
)
1.07
"Ganaxolone pretreatment resulted in a significant prolongation of absence seizure in both the PTZ and GHB models."( Ganaxolone, a selective, high-affinity steroid modulator of the gamma-aminobutyric acid-A receptor, exacerbates seizures in animal models of absence.
Snead, OC, 1998
)
2.46

Toxicity

Treatment-emergent adverse events occurred in 43 (86%) of 50 patients in the ganaxolone group and in 45 (88%) of 51 patients in a placebo group.Based on our research, we have observed that ganAXolone is safe and has potential efficacy in the treatment of refractory epilepsy, waiting for further studies.

ExcerptReferenceRelevance
" Treatment-emergent adverse events occurred in 43 (86%) of 50 patients in the ganaxolone group and in 45 (88%) of 51 patients in the placebo group."( Safety and efficacy of ganaxolone in patients with CDKL5 deficiency disorder: results from the double-blind phase of a randomised, placebo-controlled, phase 3 trial.
Aimetti, AA; Amin, S; Bahi-Buisson, N; Benke, TA; Cross, JH; Demarest, ST; Devinsky, O; Fleming, TR; Gasior, M; Knight, EMP; Olson, HE; Specchio, N, 2022
)
1.26
"Based on our research, we have observed that ganaxolone is safe and has potential efficacy in the treatment of refractory epilepsy, waiting for further studies."( The efficacy and safety of ganaxolone for the treatment of refractory epilepsy: A meta-analysis from randomized controlled trials.
Liu, Y; Meng, J; Tao, X; Wang, F; Wang, W; Wang, Z; Xue, T; Yan, Z, 2023
)
1.47

Pharmacokinetics

ExcerptReferenceRelevance
" The method was used for the routine analysis of ganaxolone in rat, monkey, dog and human plasma and summary of the pharmacokinetic data are presented."( A high-performance liquid chromatography-tandem mass spectrometric method for the determination of pharmacokinetics of ganaxolone in rat, monkey, dog and human plasma.
Chien, B; Lam, GN; Ram, K, 2001
)
0.77

Bioavailability

ExcerptReferenceRelevance
" In animal studies, compound 6 (CCD 1042) is an orally active anticonvulsant, while the naturally occurring progesterone metabolites 1 and 2 are inactive when administered orally, suggesting that 3 beta-substitution slows metabolism of the 3-hydroxyl, resulting in orally bioavailable steroid modulators of the GABAA receptor."( Synthesis and in vitro activity of 3 beta-substituted-3 alpha-hydroxypregnan-20-ones: allosteric modulators of the GABAA receptor.
Acosta-Burruel, M; Alauddin, M; Bolger, MB; Gee, KW; Hawkinson, JE; Hogenkamp, DJ; Kimbrough, CL; Lan, NC; Tahir, SH; Upasani, RB; Whittemore, ER; Woodward, RM, 1997
)
0.3
" Neurosteroid replacement is a potential approach to therapy, but natural neurosteroids have poor bioavailability and may be converted to metabolites with undesired progestational activity."( Enhanced anticonvulsant activity of ganaxolone after neurosteroid withdrawal in a rat model of catamenial epilepsy.
Reddy, DS; Rogawski, MA, 2000
)
0.58
" Synthetic neurosteroids that exhibit better bioavailability and efficacy and drugs that enhance neurosteroid synthesis have therapeutic potential in anxiety, epilepsy, and other brain disorders."( Neurosteroids: endogenous role in the human brain and therapeutic potentials.
Reddy, DS, 2010
)
0.36
"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

Dosage Studied

This study's objective was to determine the appropriate dosing for IV ganaxolone in RSE and obtain a preliminary assessment of efficacy and safety. There were three ganAXolone dosing cohorts: low, 500 mg/day; medium, 650  mg/ day; and high, 713 MG/day.

ExcerptRelevanceReference
"Ninety-six healthy male and female volunteers received ganaxolone in a variety of formulations, doses, and dosing regimens."( Initial human experience with ganaxolone, a neuroactive steroid with antiepileptic activity.
Ashbrook, DW; Lee, DA; Monaghan, EP; Navalta, LA; Shum, L, 1997
)
0.83
" Pentobarbital had no statistically significant effect on the cocaine dose-response function."( Neuroactive steroids attenuate cocaine-induced sucrose intake in rats, but not cocaine-induced hyperactivity in mice.
Carter, RB; Huber, M; Suruki, M; Vanover, KE; Wilent, WB, 2000
)
0.31
" Dosing occurred three times per day: immediately after breakfast, lunch, and dinner."( Assessment of ganaxolone's anticonvulsant activity using a randomized, double-blind, presurgical trial design. Ganaxolone Presurgical Study Group.
Abou-Khalil, BW; Blum, D; Data, JL; Laxer, K; Lee, DA; Monaghan, EP; Morrell, MJ, 2000
)
0.67
" From the dose-response functions, ED50s (i."( Differential interactions engendered by benzodiazepine and neuroactive steroid combinations on schedule-controlled responding in rats.
Gunter, BW; Platt, DM; Rowlett, JK, 2015
)
0.42
" Future investigations of ganaxolone may benefit from higher dosing, rigorous monitoring of dosing adherence, a longer length of placebo-controlled testing, and targeting of treatment to PTSD subpopulations with demonstrably dysregulated pre-treatment neuroactive steroid levels."( A randomized controlled trial of ganaxolone in posttraumatic stress disorder.
Cusin, C; Farfel, GM; Geracioti, TD; Hamner, MB; Jain, S; Lang, AJ; Lohr, J; Marx, CE; Naylor, JC; Raman, R; Rasmusson, AM; Rosse, R; Stein, MB; Summerall, L; Sun, X; Tsai, J, 2017
)
1.04
" However, dosing studies are required to reduce the burden of unwanted side effects."( Neurosteroid replacement therapy using the allopregnanolone-analogue ganaxolone following preterm birth in male guinea pigs.
Berry, MJ; Dyson, RM; Gray, C; Hirst, JJ; Palliser, HK; Shaw, JC, 2019
)
0.75
"3 years; range, 53-69 years) with persistent depression despite adequate antidepressant treatment (current DSM-IV-TR major depressive episode per the Structured Clinical Interview for DSM-IV-TR, Montgomery-Asberg Depression Rating Scale [MADRS] score ≥ 16, and treated with an adequately dosed antidepressant for ≥ 6 weeks) were studied from December 2016 to April 2018."( Effects of Open-Label, Adjunctive Ganaxolone on Persistent Depression Despite Adequate Antidepressant Treatment in Postmenopausal Women: A Pilot Study.
Cusin, C; Dichtel, LE; Dording, C; Fava, M; Fisher, LB; Kimball, AS; Miller, KK; Mischoulon, D; Nyer, M; Pedrelli, P; Rao, EM; Shapero, BG, 2020
)
0.84
" Further studies optimising target mechanisms and dosing are warranted."( Neurosteroid-based intervention using Ganaxolone and Emapunil for improving stress-induced myelination deficits and neurobehavioural disorders.
Crombie, GK; Hirst, JJ; Hodgson, DM; Palliser, HK; Shaw, JC; Walker, DW, 2021
)
0.89
" This study's objective was to determine the appropriate dosing for IV ganaxolone in RSE and obtain a preliminary assessment of efficacy and safety."( Intravenous ganaxolone for the treatment of refractory status epilepticus: Results from an open-label, dose-finding, phase 2 trial.
Aimetti, A; Gasior, M; Husain, AM; Ramsay, RE; Swisher, CB; Vaitkevicius, H, 2022
)
1.33
" There were three ganaxolone dosing cohorts: low, 500 mg/day; medium, 650 mg/day; and high, 713 mg/day."( Intravenous ganaxolone for the treatment of refractory status epilepticus: Results from an open-label, dose-finding, phase 2 trial.
Aimetti, A; Gasior, M; Husain, AM; Ramsay, RE; Swisher, CB; Vaitkevicius, H, 2022
)
1.43
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Drug Classes (1)

ClassDescription
corticosteroid hormoneAny of a class of steroid hormones that are produced in the adrenal cortex.
[compound class information is derived from 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]

Protein Targets (30)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
thioredoxin reductaseRattus norvegicus (Norway rat)Potency6.30960.100020.879379.4328AID588453
RAR-related orphan receptor gammaMus musculus (house mouse)Potency4.21630.006038.004119,952.5996AID1159521; AID1159523
GLS proteinHomo sapiens (human)Potency0.63100.35487.935539.8107AID624146
AR proteinHomo sapiens (human)Potency7.97470.000221.22318,912.5098AID1259243; AID1259247
estrogen receptor 2 (ER beta)Homo sapiens (human)Potency2.11320.000657.913322,387.1992AID1259378
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency13.80290.01237.983543.2770AID1645841
cytochrome P450 2D6Homo sapiens (human)Potency1.23020.00108.379861.1304AID1645840
arylsulfatase AHomo sapiens (human)Potency3.01311.069113.955137.9330AID720538
euchromatic histone-lysine N-methyltransferase 2Homo sapiens (human)Potency7.94330.035520.977089.1251AID504332
v-jun sarcoma virus 17 oncogene homolog (avian)Homo sapiens (human)Potency9.52050.057821.109761.2679AID1159526
nuclear receptor subfamily 1, group I, member 2Rattus norvegicus (Norway rat)Potency3.98110.10009.191631.6228AID1346983
D(1A) dopamine receptorHomo sapiens (human)Potency0.46110.02245.944922.3872AID488982
atrial natriuretic peptide receptor 1 precursorHomo sapiens (human)Potency7.56860.134610.395030.1313AID1347049
Cellular tumor antigen p53Homo sapiens (human)Potency18.83360.002319.595674.0614AID651631
[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)
Gamma-aminobutyric acid receptor subunit piHomo sapiens (human)IC50 (µMol)0.04200.00011.02016.0000AID1205592
Gamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)IC50 (µMol)0.04200.00011.02016.0000AID1205592
Gamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)IC50 (µMol)0.04200.00011.14948.0000AID1205592
Gamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)IC50 (µMol)0.04200.00011.03936.0000AID1205592
Gamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)IC50 (µMol)0.04200.00011.29158.0000AID1205592
Gamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)IC50 (µMol)0.04200.00011.30188.0000AID1205592
Gamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)IC50 (µMol)0.04200.00010.98006.0000AID1205592
Gamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)IC50 (µMol)0.04200.00011.19936.0000AID1205592
Gamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)IC50 (µMol)0.04200.00011.02016.0000AID1205592
Gamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)IC50 (µMol)0.04200.00010.93746.0000AID1205592
Gamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)IC50 (µMol)0.04200.00011.01936.0000AID1205592
Gamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)IC50 (µMol)0.04200.00011.02016.0000AID1205592
Gamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)IC50 (µMol)0.04200.00011.02016.0000AID1205592
Gamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)IC50 (µMol)0.04200.00011.02016.0000AID1205592
Gamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)IC50 (µMol)0.04200.00011.02016.0000AID1205592
Gamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)IC50 (µMol)0.04200.00011.02016.0000AID1205592
[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)
Gamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)EC50 (µMol)0.20970.00112.000910.0000AID1205593; AID1205604; AID219787
Gamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)EC50 (µMol)0.18450.07552.12458.0000AID1205593; AID1205604
Gamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)EC50 (µMol)0.26000.00141.957810.0000AID219787
Gamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)EC50 (µMol)0.18450.00301.65329.8000AID1205593; AID1205604
Gamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)EC50 (µMol)0.18450.00301.58219.8000AID1205593; AID1205604
Gamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)EC50 (µMol)0.18450.01001.20095.6234AID1205593; AID1205604
Gamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)EC50 (µMol)0.18450.01201.17515.2000AID1205593; AID1205604
Gamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)EC50 (µMol)0.20970.00141.776810.0000AID1205593; AID1205604; AID219787
Gamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)EC50 (µMol)0.18450.07551.59585.2000AID1205593; AID1205604
Gamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)EC50 (µMol)0.18450.07551.19595.2000AID1205593; AID1205604
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (150)

Processvia Protein(s)Taxonomy
chloride transmembrane transportGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
regulation of membrane potentialGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
chemical synaptic transmissionGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
signal transductionGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
regulation of membrane potentialGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
chemical synaptic transmissionGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycle G2/M phase transitionCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
ER overload responseCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
mitophagyCellular tumor antigen p53Homo sapiens (human)
in utero embryonic developmentCellular tumor antigen p53Homo sapiens (human)
somitogenesisCellular tumor antigen p53Homo sapiens (human)
release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
hematopoietic progenitor cell differentiationCellular tumor antigen p53Homo sapiens (human)
T cell proliferation involved in immune responseCellular tumor antigen p53Homo sapiens (human)
B cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
T cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
response to ischemiaCellular tumor antigen p53Homo sapiens (human)
nucleotide-excision repairCellular tumor antigen p53Homo sapiens (human)
double-strand break repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
protein import into nucleusCellular tumor antigen p53Homo sapiens (human)
autophagyCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrestCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in transcription of p21 class mediatorCellular tumor antigen p53Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
Ras protein signal transductionCellular tumor antigen p53Homo sapiens (human)
gastrulationCellular tumor antigen p53Homo sapiens (human)
neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
protein localizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA replicationCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
determination of adult lifespanCellular tumor antigen p53Homo sapiens (human)
mRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
rRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
response to salt stressCellular tumor antigen p53Homo sapiens (human)
response to inorganic substanceCellular tumor antigen p53Homo sapiens (human)
response to X-rayCellular tumor antigen p53Homo sapiens (human)
response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
positive regulation of gene expressionCellular tumor antigen p53Homo sapiens (human)
cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
viral processCellular tumor antigen p53Homo sapiens (human)
glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
cerebellum developmentCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell growthCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
mitotic G1 DNA damage checkpoint signalingCellular tumor antigen p53Homo sapiens (human)
negative regulation of telomere maintenance via telomeraseCellular tumor antigen p53Homo sapiens (human)
T cell differentiation in thymusCellular tumor antigen p53Homo sapiens (human)
tumor necrosis factor-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
regulation of tissue remodelingCellular tumor antigen p53Homo sapiens (human)
cellular response to UVCellular tumor antigen p53Homo sapiens (human)
multicellular organism growthCellular tumor antigen p53Homo sapiens (human)
positive regulation of mitochondrial membrane permeabilityCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
entrainment of circadian clock by photoperiodCellular tumor antigen p53Homo sapiens (human)
mitochondrial DNA repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
transcription initiation-coupled chromatin remodelingCellular tumor antigen p53Homo sapiens (human)
negative regulation of proteolysisCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of RNA polymerase II transcription preinitiation complex assemblyCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
response to antibioticCellular tumor antigen p53Homo sapiens (human)
fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
circadian behaviorCellular tumor antigen p53Homo sapiens (human)
bone marrow developmentCellular tumor antigen p53Homo sapiens (human)
embryonic organ developmentCellular tumor antigen p53Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationCellular tumor antigen p53Homo sapiens (human)
protein stabilizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of helicase activityCellular tumor antigen p53Homo sapiens (human)
protein tetramerizationCellular tumor antigen p53Homo sapiens (human)
chromosome organizationCellular tumor antigen p53Homo sapiens (human)
neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
hematopoietic stem cell differentiationCellular tumor antigen p53Homo sapiens (human)
negative regulation of glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
type II interferon-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
cardiac septum morphogenesisCellular tumor antigen p53Homo sapiens (human)
positive regulation of programmed necrotic cell deathCellular tumor antigen p53Homo sapiens (human)
protein-containing complex assemblyCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressCellular tumor antigen p53Homo sapiens (human)
thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
necroptotic processCellular tumor antigen p53Homo sapiens (human)
cellular response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
cellular response to xenobiotic stimulusCellular tumor antigen p53Homo sapiens (human)
cellular response to ionizing radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to UV-CCellular tumor antigen p53Homo sapiens (human)
stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
cellular response to actinomycin DCellular tumor antigen p53Homo sapiens (human)
positive regulation of release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
cellular senescenceCellular tumor antigen p53Homo sapiens (human)
replicative senescenceCellular tumor antigen p53Homo sapiens (human)
oxidative stress-induced premature senescenceCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
oligodendrocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of execution phase of apoptosisCellular tumor antigen p53Homo sapiens (human)
negative regulation of mitophagyCellular tumor antigen p53Homo sapiens (human)
regulation of mitochondrial membrane permeability involved in apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of G1 to G0 transitionCellular tumor antigen p53Homo sapiens (human)
negative regulation of miRNA processingCellular tumor antigen p53Homo sapiens (human)
negative regulation of glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
negative regulation of pentose-phosphate shuntCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
regulation of fibroblast apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
positive regulation of cellular senescenceCellular tumor antigen p53Homo sapiens (human)
positive regulation of intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
monoatomic ion transportGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
signal transductionGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
response to toxic substanceGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
central nervous system neuron developmentGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
response to progesteroneGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
ovulation cycleGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
cellular response to histamineGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
regulation of presynaptic membrane potentialGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
chemical synaptic transmissionGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
regulation of membrane potentialGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
post-embryonic developmentGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
adult behaviorGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
cellular response to histamineGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
signal transductionGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
roof of mouth developmentGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
cellular response to histamineGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
chemical synaptic transmissionGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
regulation of membrane potentialGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
behavioral fear responseGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
signal transductionGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
associative learningGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
inner ear receptor cell developmentGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
innervationGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
cochlea developmentGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
regulation of presynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
regulation of presynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
chemical synaptic transmissionGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
inner ear receptor cell developmentGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
innervationGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
cellular response to histamineGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
cochlea developmentGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
regulation of membrane potentialGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
negative regulation of chloride transportGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
signal transductionGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
response to xenobiotic stimulusGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
synaptic transmission, GABAergicGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
inhibitory synapse assemblyGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
gamma-aminobutyric acid signaling pathwayGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
neurotransmitter transportGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
signal transductionGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
chemical synaptic transmissionGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
chloride transmembrane transportGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
regulation of membrane potentialGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (48)

Processvia Protein(s)Taxonomy
GABA-A receptor activityGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
neurotransmitter receptor activityGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
protein bindingGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
neurotransmitter receptor activityGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
transcription cis-regulatory region bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
core promoter sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
TFIID-class transcription factor complex bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
protease bindingCellular tumor antigen p53Homo sapiens (human)
p53 bindingCellular tumor antigen p53Homo sapiens (human)
DNA bindingCellular tumor antigen p53Homo sapiens (human)
chromatin bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activityCellular tumor antigen p53Homo sapiens (human)
mRNA 3'-UTR bindingCellular tumor antigen p53Homo sapiens (human)
copper ion bindingCellular tumor antigen p53Homo sapiens (human)
protein bindingCellular tumor antigen p53Homo sapiens (human)
zinc ion bindingCellular tumor antigen p53Homo sapiens (human)
enzyme bindingCellular tumor antigen p53Homo sapiens (human)
receptor tyrosine kinase bindingCellular tumor antigen p53Homo sapiens (human)
ubiquitin protein ligase bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase regulator activityCellular tumor antigen p53Homo sapiens (human)
ATP-dependent DNA/DNA annealing activityCellular tumor antigen p53Homo sapiens (human)
identical protein bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase bindingCellular tumor antigen p53Homo sapiens (human)
protein heterodimerization activityCellular tumor antigen p53Homo sapiens (human)
protein-folding chaperone bindingCellular tumor antigen p53Homo sapiens (human)
protein phosphatase 2A bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingCellular tumor antigen p53Homo sapiens (human)
14-3-3 protein bindingCellular tumor antigen p53Homo sapiens (human)
MDM2/MDM4 family protein bindingCellular tumor antigen p53Homo sapiens (human)
disordered domain specific bindingCellular tumor antigen p53Homo sapiens (human)
general transcription initiation factor bindingCellular tumor antigen p53Homo sapiens (human)
molecular function activator activityCellular tumor antigen p53Homo sapiens (human)
promoter-specific chromatin bindingCellular tumor antigen p53Homo sapiens (human)
GABA receptor activityGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
ligand-gated monoatomic ion channel activityGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
GABA receptor bindingGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
G protein-coupled neurotransmitter receptor activity involved in regulation of presynaptic membrane potentialGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
neurotransmitter receptor activityGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
protein bindingGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
neurotransmitter receptor activityGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
identical protein bindingGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
neurotransmitter receptor activityGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
signaling receptor activityGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
GABA receptor bindingGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
protein bindingGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
protein bindingGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
GABA receptor activityGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
neurotransmitter receptor activityGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
protein bindingGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
GABA receptor bindingGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
benzodiazepine receptor activityGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
transmembrane signaling receptor activityGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
GABA-A receptor activityGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
neurotransmitter transmembrane transporter activityGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
protein bindingGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
GABA-gated chloride ion channel activityGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
chloride channel activityGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
neurotransmitter receptor activityGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (47)

Processvia Protein(s)Taxonomy
plasma membraneGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
apical plasma membraneGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit piHomo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
axonGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
dendriteGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
neuronal cell bodyGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
postsynaptic membraneGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit deltaHomo sapiens (human)
nuclear bodyCellular tumor antigen p53Homo sapiens (human)
nucleusCellular tumor antigen p53Homo sapiens (human)
nucleoplasmCellular tumor antigen p53Homo sapiens (human)
replication forkCellular tumor antigen p53Homo sapiens (human)
nucleolusCellular tumor antigen p53Homo sapiens (human)
cytoplasmCellular tumor antigen p53Homo sapiens (human)
mitochondrionCellular tumor antigen p53Homo sapiens (human)
mitochondrial matrixCellular tumor antigen p53Homo sapiens (human)
endoplasmic reticulumCellular tumor antigen p53Homo sapiens (human)
centrosomeCellular tumor antigen p53Homo sapiens (human)
cytosolCellular tumor antigen p53Homo sapiens (human)
nuclear matrixCellular tumor antigen p53Homo sapiens (human)
PML bodyCellular tumor antigen p53Homo sapiens (human)
transcription repressor complexCellular tumor antigen p53Homo sapiens (human)
site of double-strand breakCellular tumor antigen p53Homo sapiens (human)
germ cell nucleusCellular tumor antigen p53Homo sapiens (human)
chromatinCellular tumor antigen p53Homo sapiens (human)
transcription regulator complexCellular tumor antigen p53Homo sapiens (human)
protein-containing complexCellular tumor antigen p53Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
cytoplasmic vesicle membraneGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
GABA receptor complexGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit alpha-1Homo sapiens (human)
nuclear envelopeGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
dendriteGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
presynaptic active zone membraneGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
Schaffer collateral - CA1 synapseGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit beta-1Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
axonGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
cytoplasmic vesicle membraneGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit gamma-2Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
cytoplasmic vesicle membraneGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit beta-3Homo sapiens (human)
nucleoplasmGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
cytosolGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
neuronal cell body membraneGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
presynaptic membraneGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-5Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
postsynaptic membraneGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit alpha-3Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
axonGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
synaptic vesicle membraneGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
neuronal cell bodyGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
inhibitory synapseGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit alpha-2Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
cytoplasmic vesicle membraneGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
extracellular exosomeGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit beta-2Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit alpha-4Homo sapiens (human)
postsynaptic membraneGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit epsilonHomo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
cerebellar Golgi cell to granule cell synapseGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
postsynaptic specialization membraneGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit alpha-6Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
postsynaptic membraneGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit gamma-1Homo sapiens (human)
nucleolusGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
microtubule cytoskeletonGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
postsynaptic membraneGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
GABA-ergic synapseGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
dendrite membraneGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
postsynapseGamma-aminobutyric acid receptor subunit gamma-3Homo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
postsynaptic membraneGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
chloride channel complexGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
receptor complexGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
plasma membraneGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
neuron projectionGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
transmembrane transporter complexGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
synapseGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
GABA-A receptor complexGamma-aminobutyric acid receptor subunit thetaHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (69)

Assay IDTitleYearJournalArticle
AID540230Dose normalised AUC in rat after po administration2005Xenobiotica; the fate of foreign compounds in biological systems, Feb, Volume: 35, Issue:2
Comparative evaluation of oral systemic exposure of 56 xenobiotics in rat, dog, monkey and human.
AID1205595Lipophilicity, log D of the compound at 20 uM at pH 7.4 by HPLC-MS/MS analysis2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID540233Dose normalised AUC in human after po administration2005Xenobiotica; the fate of foreign compounds in biological systems, Feb, Volume: 35, Issue:2
Comparative evaluation of oral systemic exposure of 56 xenobiotics in rat, dog, monkey and human.
AID1205602Ratio of drug level in brain to plasma in Sprague-Dawley rat at 5 mg/kg, iv after 30 mins by LC-MS/MS analysis2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID1205598Clearance in rat liver microsomes at 10 uM after 60 mins by HPLC-MS/MS analysis2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID1205597Clearance in human liver microsomes at 10 uM after 60 mins by HPLC-MS/MS analysis2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID219789Efficacy as fractional response (FR) relative to GABA maximal current elicited in Xenopus oocytes1997Journal of medicinal chemistry, Jan-03, Volume: 40, Issue:1
Synthesis and in vitro activity of 3 beta-substituted-3 alpha-hydroxypregnan-20-ones: allosteric modulators of the GABAA receptor.
AID1205599Clearance in mouse liver microsomes at 10 uM after 60 mins by HPLC-MS/MS analysis2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID71697Ability to allosterically modulate the binding of [35S]TBPS to gamma-aminobutyric-acid A receptor in rat brain cortical P2 membranes.1997Journal of medicinal chemistry, Jan-03, Volume: 40, Issue:1
Synthesis and in vitro activity of 3 beta-substituted-3 alpha-hydroxypregnan-20-ones: allosteric modulators of the GABAA receptor.
AID1205592Displacement of [3H]TBPS from GABAA receptor in Sprague-Dawley rat brain cortical membrane after 90 mins by liquid scintillation spectrometry2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID219787Potentiation of GABA-activated currents in Xenopus oocytes expressing cloned human alpha-1-beta-2-gamma-2L subunits of GABA-A receptor1997Journal of medicinal chemistry, Jan-03, Volume: 40, Issue:1
Synthesis and in vitro activity of 3 beta-substituted-3 alpha-hydroxypregnan-20-ones: allosteric modulators of the GABAA receptor.
AID1205596Aqueous solubility of the compound at pH 7.4 by HPLC-MS/MS analysis2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID540232Dose normalised AUC in monkey after po administration2005Xenobiotica; the fate of foreign compounds in biological systems, Feb, Volume: 35, Issue:2
Comparative evaluation of oral systemic exposure of 56 xenobiotics in rat, dog, monkey and human.
AID540231Dose normalised AUC in dog after po administration2005Xenobiotica; the fate of foreign compounds in biological systems, Feb, Volume: 35, Issue:2
Comparative evaluation of oral systemic exposure of 56 xenobiotics in rat, dog, monkey and human.
AID1205601Oral bioavailability in Sprague-Dawley rat at 20 mg/kg by LC-MS/MS analysis2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID1205600Clearance in Sprague-Dawley rat at 5 mg/kg, iv and 20 mg/kg, po by LC-MS/MS analysis2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID196544Percent of maximal extent of inhibition was calculated by fitting [35S]TBPS inhibition data to a sigmoid function (rat brain cortical P2 membranes).1997Journal of medicinal chemistry, Jan-03, Volume: 40, Issue:1
Synthesis and in vitro activity of 3 beta-substituted-3 alpha-hydroxypregnan-20-ones: allosteric modulators of the GABAA receptor.
AID1205604Positive allosteric modulation of recombinant alpha4beta3delta GABAA receptor (unknown origin) expressed in CHO cells assessed as potentiation of GABA-induced channel current treated for 30 s prior to GABA stimulation for 2s by manual patch assay2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID1205593Positive allosteric modulation of recombinant alpha1beta2gamma2 GABAA receptor (unknown origin) expressed in LTK cells assessed as potentiation of GABA-induced channel current treated for 30 s prior to GABA stimulation for 2s by Q-patch assay2015Journal of medicinal chemistry, Apr-23, Volume: 58, Issue:8
Neuroactive Steroids. 1. Positive Allosteric Modulators of the (γ-Aminobutyric Acid)A Receptor: Structure-Activity Relationships of Heterocyclic Substitution at C-21.
AID1347093qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
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.
AID1347095qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347154Primary screen GU AMC 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.
AID1347103qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1508630Primary qHTS for small molecule stabilizers of the endoplasmic reticulum resident proteome: Secreted ER Calcium Modulated Protein (SERCaMP) assay2021Cell reports, 04-27, Volume: 35, Issue:4
A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome.
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID1347407qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Pharmaceutical Collection2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1347092qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for A673 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347101qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347083qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: Viability assay - alamar blue signal for LASV Primary Screen2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347102qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347100qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID1347099qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347104qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347108qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347089qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347086qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lymphocytic Choriomeningitis Arenaviruses (LCMV): LCMV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347105qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347098qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347107qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347106qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347082qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: LASV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347091qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347094qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347425Rhodamine-PBP qHTS Assay for Modulators of WT P53-Induced Phosphatase 1 (WIP1)2019The Journal of biological chemistry, 11-15, Volume: 294, Issue:46
Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens.
AID1347097qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347096qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347090qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347424RapidFire Mass Spectrometry qHTS Assay for Modulators of WT P53-Induced Phosphatase 1 (WIP1)2019The Journal of biological chemistry, 11-15, Volume: 294, Issue:46
Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens.
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.
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.
AID504749qHTS profiling for inhibitors of Plasmodium falciparum proliferation2011Science (New York, N.Y.), Aug-05, Volume: 333, Issue:6043
Chemical genomic profiling for antimalarial therapies, response signatures, and molecular targets.
AID1347059CD47-SIRPalpha protein protein interaction - Alpha assay qHTS validation2019PloS one, , Volume: 14, Issue:7
Quantitative high-throughput screening assays for the discovery and development of SIRPα-CD47 interaction inhibitors.
AID588349qHTS for Inhibitors of ATXN expression: Validation of Cytotoxic Assay
AID504812Inverse Agonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID588378qHTS for Inhibitors of ATXN expression: Validation
AID504836Inducers of the Endoplasmic Reticulum Stress Response (ERSR) in human glioma: Validation2002The Journal of biological chemistry, Apr-19, Volume: 277, Issue:16
Sustained ER Ca2+ depletion suppresses protein synthesis and induces activation-enhanced cell death in mast cells.
AID504810Antagonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID1347045Natriuretic polypeptide receptor (hNpr1) antagonism - Pilot counterscreen GloSensor control cell line2019Science translational medicine, 07-10, Volume: 11, Issue:500
Inhibition of natriuretic peptide receptor 1 reduces itch in mice.
AID1347049Natriuretic polypeptide receptor (hNpr1) antagonism - Pilot screen2019Science translational medicine, 07-10, Volume: 11, Issue:500
Inhibition of natriuretic peptide receptor 1 reduces itch in mice.
AID1347050Natriuretic polypeptide receptor (hNpr2) antagonism - Pilot subtype selectivity assay2019Science translational medicine, 07-10, Volume: 11, Issue:500
Inhibition of natriuretic peptide receptor 1 reduces itch in mice.
AID1347057CD47-SIRPalpha protein protein interaction - LANCE assay qHTS validation2019PloS one, , Volume: 14, Issue:7
Quantitative high-throughput screening assays for the discovery and development of SIRPα-CD47 interaction inhibitors.
AID1347405qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS LOPAC collection2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1347151Optimization of GU AMC 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.
AID1347410qHTS for inhibitors of adenylyl cyclases using a fission yeast platform: a pilot screen against the NCATS LOPAC library2019Cellular signalling, 08, Volume: 60A fission yeast platform for heterologous expression of mammalian adenylyl cyclases and high throughput screening.
AID1347058CD47-SIRPalpha protein protein interaction - HTRF assay qHTS validation2019PloS one, , Volume: 14, Issue:7
Quantitative high-throughput screening assays for the discovery and development of SIRPα-CD47 interaction inhibitors.
AID1794808Fluorescence-based screening to identify small molecule inhibitors of Plasmodium falciparum apicoplast DNA polymerase (Pf-apPOL).2014Journal of biomolecular screening, Jul, Volume: 19, Issue:6
A High-Throughput Assay to Identify Inhibitors of the Apicoplast DNA Polymerase from Plasmodium falciparum.
AID1794808Fluorescence-based screening to identify small molecule inhibitors of Plasmodium falciparum apicoplast DNA polymerase (Pf-apPOL).
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (98)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's6 (6.12)18.2507
2000's26 (26.53)29.6817
2010's42 (42.86)24.3611
2020's24 (24.49)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 52.69

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 Index52.69 (24.57)
Research Supply Index4.69 (2.92)
Research Growth Index4.87 (4.65)
Search Engine Demand Index83.43 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (52.69)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials10 (10.20%)5.53%
Reviews13 (13.27%)6.00%
Case Studies0 (0.00%)4.05%
Observational0 (0.00%)0.25%
Other75 (76.53%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (24)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Treatment-Resistant Depression Augmentation Therapy With An Analog of the Neuroactive Steroid Allopregnanolone: A Pilot Study [NCT02900092]10 participants (Actual)Interventional2016-11-30Completed
A Proof-of-Concept, Double-blind, Randomized, Placebo-controlled Study of Ganaxolone in Posttraumatic Stress Disorder [NCT01339689]Phase 2112 participants (Actual)Interventional2011-04-30Completed
A Multicenter, Double Blind, Randomized, Placebo-Controlled Trial to Determine the Efficacy and Safety of Ganaxolone as Adjunctive Therapy for Adults With Drug-Resistant Partial-Onset Seizures Followed by Long-term Open-Label Treatment [NCT01963208]Phase 3405 participants (Actual)Interventional2013-10-31Completed
A Phase 2 Open-label 12-Week Trial of Adjunctive Ganaxolone Treatment (Part A) in Tuberous Sclerosis Complex-related Epilepsy Followed by Long-term Treatment (Part B) [NCT04285346]Phase 223 participants (Actual)Interventional2020-04-08Completed
A Double-Blind Randomized, Placebo Controlled Study to Evaluate the Safety, Tolerability, Efficacy, and Pharmacokinetics of Intravenous Ganaxolone as Adjunctive Therapy to Treat Subjects With Status Epilepticus [NCT03350035]Phase 217 participants (Actual)Interventional2018-02-19Completed
Expanded Access With Adjunctive Ganaxolone Treatment in Children and Young Adults With Cyclin Dependent Kinase-like 5 Deficiency Disorder [NCT04678479]0 participants Expanded AccessAvailable
A Treatment Protocol for Ganaxolone as add-on Therapy in Adult Patients With Uncontrolled Partial-onset Seizures Deriving Benefit From Protocol 1042-0601 [NCT01002820]Phase 211 participants (Actual)Interventional2009-10-31Completed
A Double-blind, Randomized, Placebo-controlled Study to Evaluate the Efficacy and Safety of Intravenous Ganaxolone in Status Epilepticus [NCT04391569]Phase 3124 participants (Anticipated)Interventional2020-10-10Recruiting
A Study to Optimize the Dosing Regimen and Assess Safety and Efficacy of IV Ganaxolone as Adjuvant Therapy for Established Status Epilepticus [NCT05757544]Phase 2120 participants (Anticipated)Interventional2023-04-01Recruiting
A Double-blind, Randomized, Placebo-controlled Trial of Adjunctive Ganaxolone Treatment in Female Children With Protocadherin 19 (PCDH19)-Related Epilepsy Followed by Long-term Open-label Treatment. [NCT03865732]Phase 229 participants (Actual)Interventional2019-05-17Completed
A Multicenter, Open-Label Proof-of-Concept Trial of Ganaxolone in Children With PCDH19 Female Pediatric Epilepsy and Other Rare Genetic Epilepsies Followed by 52 Week Open-Label Treatment [NCT02358538]Phase 230 participants (Actual)Interventional2015-11-06Completed
A Phase 3, Open-label Study of Adjunctive Ganaxolone (GNX) Treatment in Children and Adults With Tuberous Sclerosis Complex (TSC)-Related Epilepsy (TrustTSC OLE) [NCT05604170]Phase 3132 participants (Anticipated)Interventional2022-05-16Enrolling by invitation
An Open-label Extension Study to Evaluate the Safety, Tolerability, and Efficacy of Ganaxolone as add-on Therapy in Adult Patients With Epilepsy Consisting of Uncontrolled Partial-onset Seizures. [NCT00512317]Phase 2123 participants (Actual)Interventional2007-06-30Completed
A Phase 2, Double-blind, Placebo-controlled, Multicenter Study to Evaluate Safety, Tolerability, and Efficacy of Oral Administration of Ganaxolone in Women With Postpartum Depression [NCT03460756]Phase 284 participants (Actual)Interventional2017-12-28Completed
A Phase 2A, Double-blind, Placebo-controlled, Multiple-dose Escalation Study to Evaluate Safety, Pharmacokinetics and Efficacy of Intravenously Administered Ganaxolone in Women With Postpartum Depression [NCT03228394]Phase 291 participants (Actual)Interventional2017-06-27Completed
A Double-blind, Randomized, Placebo-controlled Study to Evaluate the Efficacy, Safety, and Tolerability of Intravenous Ganaxolone Added to Standard of Care in Refractory Status Epilepticus [NCT05814523]Phase 370 participants (Anticipated)Interventional2023-09-30Not yet recruiting
A Double-blind, Randomized, Placebo-controlled Study to Evaluate the Safety, Tolerability, and Efficacy of Ganaxolone as add-on Therapy in Adult Subjects With Epilepsy Consisting of Uncontrolled Partial-onset Seizures. [NCT00465517]Phase 2147 participants (Actual)Interventional2007-02-28Completed
Double-blind, Randomized, Placebo-controlled Trial of Adjunctive Ganaxolone in the Treatment of Seizures Associated With Genetically Confirmed Cyclin-dependent Kinase-like 5 (CDKL5) Deficiency Disorder (CDD) in Pediatric Patients From 6 Months to Less Tha [NCT05249556]Phase 320 participants (Anticipated)Interventional2023-05-31Not yet recruiting
An Open-label Clinical Study to Evaluate the Safety and Antiepileptic Activity of Ganaxolone in Treatment of Patients Diagnosed With Infantile Spasms. [NCT00442104]Phase 254 participants (Actual)Interventional2007-01-31Terminated(stopped due to Sponsor's decision)
A follow-on, Two-year Open-label Extension Study of Ganaxolone as add-on Therapy in Adult Patients With Drug-resistant Partial-onset Seizures [NCT02519439]Phase 326 participants (Actual)Interventional2015-02-28Terminated(stopped due to Ganaxolone missed its primary endpoint in the double-blind portion of the 1042-0603 study. Due to this outcome Marinus discontinued this extension study.)
A Controlled, Double-blind, Crossover Trial of Ganaxolone in Children With Fragile X Syndrome [NCT01725152]Phase 259 participants (Actual)Interventional2012-11-30Completed
Proof-Of-Concept Investigation With a Neurosteroid Analog (Ganaxolone) as a Smoking Cessation Candidate [NCT01857531]Phase 236 participants (Actual)Interventional2013-06-30Completed
A Double-blind, Placebo-controlled, Dose-ranging Clinical Study to Evaluate the Safety, Tolerability, and Antiepileptic Activity of Ganaxolone in Treatment of Patients With Infantile Spasms [NCT00441896]Phase 257 participants (Actual)Interventional2007-01-31Completed
A Double-blind, Randomized, Placebo-controlled Trial of Adjunctive Ganaxolone Treatment in Children and Young Adults With Cyclin-dependent Kinase-like 5 (CDKL5) Deficiency Disorder (CDD) Followed by Long-term Open-label Treatment [NCT03572933]Phase 3101 participants (Actual)Interventional2018-06-30Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00441896 (9) [back to overview]Developmental Assessment Using Denver-II Developmental Test at Day 20
NCT00441896 (9) [back to overview]Change From Baseline in Frequency of Spasm Clusters at Day 10
NCT00441896 (9) [back to overview]Number of Participants With Absence of Hypsarrhythmia
NCT00441896 (9) [back to overview]Change From Baseline in Frequency of Spasm Clusters at Day 20
NCT00441896 (9) [back to overview]Number of Participants With Spasm-free Durations
NCT00441896 (9) [back to overview]Number of Participants With Seizure-free Days
NCT00441896 (9) [back to overview]Number of Responders
NCT00441896 (9) [back to overview]Number of Participants With Change in Clinical Status on the Investigator's Global Assessment
NCT00441896 (9) [back to overview]Number of Participants With Change in Clinical Status on Caregiver's Global Assessment
NCT00442104 (9) [back to overview]Percentage of Total Spasm-free Days and Cumulative Spasm-free Days as Determined From the Seizure Diary.
NCT00442104 (9) [back to overview]Number of Participants With Change in Clinical Status on Caregiver's Global Assessment
NCT00442104 (9) [back to overview]Number of Participants With Absence of Spasms
NCT00442104 (9) [back to overview]Developmental Assessment Using Denver-II Developmental Test
NCT00442104 (9) [back to overview]Number of Participants With Spasm-free Durations
NCT00442104 (9) [back to overview]Percentage of Participants Who Were Free of Spasms
NCT00442104 (9) [back to overview]Change From Baseline in Frequency of Individual Spasm
NCT00442104 (9) [back to overview]Change From Baseline in Frequency of Spasm Clusters
NCT00442104 (9) [back to overview]Number of Participants With Change in Clinical Status on the Investigator's Global Assessment
NCT00465517 (16) [back to overview]Percent Change From Baseline in Mean Weekly Seizure Frequency by Subtype During Weeks 1 Through 10
NCT00465517 (16) [back to overview]Percent Change From Baseline in Mean Weekly Seizure Frequency During Weeks 3 Through 10
NCT00465517 (16) [back to overview]Change From Baseline in Mean Weekly Seizure Frequency During Weeks 1 Through 10
NCT00465517 (16) [back to overview]Change From Baseline in Mean Weekly Seizure Frequency During Weeks 3 Through 10
NCT00465517 (16) [back to overview]Mean Weekly Seizure Frequency for Each Week After Dosing During Weeks 1 to 10
NCT00465517 (16) [back to overview]Number of Responders During Weeks 1 Through 10
NCT00465517 (16) [back to overview]Number of Responders During Weeks 3 Through 10
NCT00465517 (16) [back to overview]Number of Seizure-free Days During Week 1 Through 10
NCT00465517 (16) [back to overview]Number of Seizure-free Days During Week 3 Through 10
NCT00465517 (16) [back to overview]Number of Seizure-free Days Up to Week 2
NCT00465517 (16) [back to overview]Number of Seizure-free Participants During Weeks 1 Through 10
NCT00465517 (16) [back to overview]Number of Seizure-free Participants During Weeks 3 Through 10
NCT00465517 (16) [back to overview]Number of Seizure-free Participants Up to Week 2
NCT00465517 (16) [back to overview]Percent Change From Baseline in Mean Weekly Seizure Frequency During Weeks 1 Through 10
NCT00465517 (16) [back to overview]Mean Weekly Log-transformed Seizure Frequency During Weeks 1 Through 10
NCT00465517 (16) [back to overview]Mean Weekly Log-transformed Seizure Frequency During Weeks 3 Through 10
NCT00512317 (8) [back to overview]Number of Seizure-free Participants
NCT00512317 (8) [back to overview]Percent Change From Baseline in Weekly Seizure Frequency During Weeks 1 Through 117
NCT00512317 (8) [back to overview]Percent Change From Baseline in Weekly Seizure Frequency During Weeks 1 Through 117
NCT00512317 (8) [back to overview]Change From Baseline in Quality of Life in Epilepsy Inventory-31 (QOLIE-31) Questionnaire
NCT00512317 (8) [back to overview]Number of Responders During Weeks 1 Through 117
NCT00512317 (8) [back to overview]Number of Responders During Weeks 1 Through 117
NCT00512317 (8) [back to overview]Number of Seizure-free Days During Weeks 1 Through 117
NCT00512317 (8) [back to overview]Number of Seizure-free Days During Weeks 1 Through 117
NCT01002820 (1) [back to overview]Investigators Global Assessment
NCT01339689 (11) [back to overview]Number of Participants With Response to Clinical Global Impression - Improvement (Investigator) (CGI-II) Scale
NCT01339689 (11) [back to overview]Change From Baseline in Connor-Davidson Resilience Scale (CD-RISC) Score
NCT01339689 (11) [back to overview]Change From Baseline in Insomnia Severity Index (ISI) Score
NCT01339689 (11) [back to overview]Change From Baseline in Patient Health Questionnaire (PHQ-9) Score
NCT01339689 (11) [back to overview]Change From Baseline in Profile of Mood States (POMS) Total Score
NCT01339689 (11) [back to overview]Change From Baseline in Clinician-Administered Posttrautamtic Stress Disorder (PTSD) Scale (CAPS) to Week 6
NCT01339689 (11) [back to overview]Change From Baseline in PTSD Avoidance Score
NCT01339689 (11) [back to overview]Change From Baseline in PTSD Checklist (PCL) Scores
NCT01339689 (11) [back to overview]Change From Baseline in PTSD Hyperarousal Score
NCT01339689 (11) [back to overview]Change From Baseline in PTSD Re-Experience Score
NCT01339689 (11) [back to overview]Number of Participants With Response to CGI-I Subject Scale Clinical Global Impression - Improvement (Subject) (CGI-IS) Scale
NCT01725152 (6) [back to overview]Clinical Global Impression-Improvement (CGI-I) Scale
NCT01725152 (6) [back to overview]Pediatric Anxiety Rating Scale (PARS) Total Score
NCT01725152 (6) [back to overview]Visual Analogue Scale (VAS)
NCT01725152 (6) [back to overview]Swanson, Nolan, and Pelham-IV Questionnaire (SNAP-IV)
NCT01725152 (6) [back to overview]Anxiety, Depression, and Mood Scale (ADAMS)
NCT01725152 (6) [back to overview]Aberrant Behavior Checklist (ABC)
NCT01857531 (6) [back to overview]Percentage of Change in Expired Air Carbon Monoxide (CO) at End of Week 2
NCT01857531 (6) [back to overview]Number of Participants Completing Continuous 6-week Abstinence From Smoking
NCT01857531 (6) [back to overview]Number of Participants Completing Abstinence From Smoking During the Last Four Weeks of Treatment
NCT01857531 (6) [back to overview]Number of Participants Completing Continuous 2-week Abstinence From Smoking
NCT01857531 (6) [back to overview]Number of Participants Completing Point Abstinence From Smoking Two Weeks After Quitting
NCT01857531 (6) [back to overview]Percentage of Change in Expired Air Carbon Monoxide (CO) at End of Week 4
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Percentage of Responders Experiencing a ≥R% (80%, 60%, 40%, and 20%) Reduction From Baseline to the End of Treatment Period in 28-day Seizure Frequency During Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Percentage of Responders Experiencing a ≥R% (80%, 60%, 40%, and 20%) Reduction From Baseline to the End of Treatment Period in 28-day Seizure Frequency During Titration + Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Number of Participants With Clinical Global Impression of Change - Improvement (CGI-I) at Week 8
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Number of Participants With Clinical Global Impression of Change - Improvement (CGI-I) at Week 14
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Percent Change From Baseline in 28-day Seizure Frequency During Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Percentage of Seizure Free Participants During the Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Percentage of Participants Who Experienced at Least One 28-day Seizure Free Period During Titration + Maintenance Phase
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Percent Change From Baseline in 28-day Seizure Frequency During Titration + Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Percent Change From Baseline in 28-day Seizure Frequency for Different Subtypes of Seizures During Titration + Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Number of Participants With Patient Global Impression of Change - Improvement (PGI-I) at Week 8 and Week 14
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Longest Percent of Time Spent Seizure-free During Titration + Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Change From Baseline in the Number of Seizure Free Days Per 28-day Period During Titration + Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Change From Baseline in the Number of Seizure Free Days Per 28-day Period During Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Change From Baseline in 28-day Seizure Frequency During Titration + Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Change From Baseline in 28-day Seizure Frequency During Maintenance Period
NCT01963208 (16) [back to overview]Double Blind: Cohort 2: Number of Participants With ≥50% Responder Rate During Titration + Maintenance Period
NCT02358538 (6) [back to overview]Summary of CGII-P
NCT02358538 (6) [back to overview]Summary of CGII-C
NCT02358538 (6) [back to overview]Summary of 28-day Seizure Frequency for Sum of Individual Seizures and Clusters Through 52-week OLE (Median Percent Change)
NCT02358538 (6) [back to overview]Summary of 28-day Seizure Frequency for Sum of Individual Seizures and Clusters for 52-week OLE Period (Mean Percent Change & Standard Deviation)
NCT02358538 (6) [back to overview]Number of Participants With Responder Rate of Seizure Frequency
NCT02358538 (6) [back to overview]Mean Percentage Change of Individual Seizure-free Days
NCT02519439 (4) [back to overview]Number of Participants Who Showed Greater Than or Equal to 50% Reduction in 28-day Seizure Frequent From Baseline
NCT02519439 (4) [back to overview]Percent Change From Baseline in 28-day Seizure Frequency
NCT02519439 (4) [back to overview]Number of Participants With Patient/Caregiver Global Impression of Improvement (PGI-I) Scores
NCT02519439 (4) [back to overview]Number of Participants With Clinical Global Impression of Improvement (CGI-I) Scores
NCT02900092 (1) [back to overview]Montgomery-Asberg Depression Rating Scale (MADRS)
NCT03228394 (14) [back to overview]Change From Baseline in Spielberger State-Trait Anxiety Inventory 6-item Version (STAI6) Total Score
NCT03228394 (14) [back to overview]Change From Baseline in Spielberger State-Trait Anxiety Inventory 6-item Version (STAI6) Total Score
NCT03228394 (14) [back to overview]Number of Participants With HAMD17 Remission
NCT03228394 (14) [back to overview]Change From Baseline in HAMD17 Total Score at Indicated Time Points
NCT03228394 (14) [back to overview]Change From Baseline in Hamilton Depression Rating Scale 17-item Version (HAMD17) Total Score
NCT03228394 (14) [back to overview]Change From Baseline in Hamilton Depression Rating Scale 17-item Version (HAMD17) Total Score
NCT03228394 (14) [back to overview]Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score
NCT03228394 (14) [back to overview]Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score
NCT03228394 (14) [back to overview]Change From Baseline in HAMD17 Total Score at Indicated Time Points
NCT03228394 (14) [back to overview]Number of Participants With HAMD17 Remission
NCT03228394 (14) [back to overview]Number of Participants With HAMD17 Response
NCT03228394 (14) [back to overview]Number of Participants With HAMD17 Response
NCT03228394 (14) [back to overview]Number of Participants With Response to Clinical Global Impression-Improvement (CGI-I) Scale
NCT03228394 (14) [back to overview]Number of Participants With Response to Clinical Global Impression-Improvement (CGI-I) Scale
NCT03350035 (5) [back to overview]Seizure Burden
NCT03350035 (5) [back to overview]Number of Participants With No SE Recurrence Per Principal Investigator
NCT03350035 (5) [back to overview]Time to Cessation of SE
NCT03350035 (5) [back to overview]Number of Participants Who Required No Escalation of Treatment for Ongoing or Recurrent SE
NCT03350035 (5) [back to overview]Number of Participants Who Did Not Require an IV Anesthetic Drug for SE Treatment
NCT03460756 (20) [back to overview]Change From Baseline in Spielberger State-Trait Anxiety Inventory, 6-item Version
NCT03460756 (20) [back to overview]Change From Baseline in Hamilton Depression Rating Scale 17-item Version Post Treatment
NCT03460756 (20) [back to overview]Change From Baseline in Hamilton Depression Rating Scale 17-item Version Post Treatment
NCT03460756 (20) [back to overview]Change From Baseline in Hamilton Depression Rating Scale 17-item Version (HAMD17) Through Treatment
NCT03460756 (20) [back to overview]Change From Baseline in Hamilton Depression Rating Scale 17-item Version (HAMD17) Through Treatment
NCT03460756 (20) [back to overview]Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score
NCT03460756 (20) [back to overview]Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score
NCT03460756 (20) [back to overview]Change From Baseline in Hamilton Depression Rating Scale 17-item Version Post Treatment
NCT03460756 (20) [back to overview]Number of Participants With Clinical Global Impression-Improvement
NCT03460756 (20) [back to overview]Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score
NCT03460756 (20) [back to overview]Number of Participants With Clinical Global Impression-Improvement
NCT03460756 (20) [back to overview]Number of Participants With Clinical Global Impression-Improvement
NCT03460756 (20) [back to overview]Change From Baseline in Spielberger State-Trait Anxiety Inventory, 6-item Version
NCT03460756 (20) [back to overview]Change From Baseline in Spielberger State-Trait Anxiety Inventory, 6-item Version
NCT03460756 (20) [back to overview]Number of Participants With Response to Hamilton Depression Rating Scale 17 Item (HAMD17)
NCT03460756 (20) [back to overview]Number of Participants With Response to Hamilton Depression Rating Scale 17 Item (HAMD17)
NCT03460756 (20) [back to overview]Number of Participants With Hamilton Depression Rating Scale 17-item Remission
NCT03460756 (20) [back to overview]Number of Participants With Response to Hamilton Depression Rating Scale 17 Item (HAMD17)
NCT03460756 (20) [back to overview]Number of Participants With Hamilton Depression Rating Scale 17-item Remission
NCT03460756 (20) [back to overview]Number of Participants With Hamilton Depression Rating Scale 17-item Remission
NCT03572933 (8) [back to overview]Arithmetic Change in Longest Seizure Free Interval, Based on Primary Seizure Types
NCT03572933 (8) [back to overview]Caregiver Global Impression of Change in Attention
NCT03572933 (8) [back to overview]Caregiver Global Impression of Change in Seizure Intensity and Duration
NCT03572933 (8) [back to overview]Caregiver Global Impression of Change in Target Behavior
NCT03572933 (8) [back to overview]Clinical Global Impression of Improvement - Clinician
NCT03572933 (8) [back to overview]Clinical Global Impression of Improvement - Parent/Caregiver
NCT03572933 (8) [back to overview]Percentage of Seizure-free Days for Major Motor Seizure Types
NCT03572933 (8) [back to overview]Summary of 28-day Seizure Frequency for Major Motor Seizure Types
NCT03865732 (3) [back to overview]50% Primary Seizure Reduction
NCT03865732 (3) [back to overview]Summary of 28-day Seizure Frequency for Subjects in the Biomarker-positive Stratum (Median Percent Change)
NCT03865732 (3) [back to overview]Summary of 28-day Seizure Frequency Through 17 Week Post-Baseline Phase (Median Percent Change)
NCT04285346 (2) [back to overview]Percent Change From Baseline in 28-day Seizure Frequency Through the End of 12-Week Treatment Period
NCT04285346 (2) [back to overview]Percentage of Participants Experiencing a >=50 Percent Reduction in 28-day Primary Seizure Frequency Through the End of the 12-week Treatment Period Compared to the Baseline Period

Developmental Assessment Using Denver-II Developmental Test at Day 20

Denver-II Developmental Test measures a child's development in several areas:Personal-Social,Fine Motor-Adaptive,Language,and Gross Motor,from birth to 6 years old.It consists of 125 items that are organized into subscales and scored as pass,fail,or refused.To evaluate a child's progress,test compares their performance to a normative sample of children of same age.For each item,age at which 90% of children in normative sample pass it is determined.Derived score for each subscale is sum of item scores and represents difference between child's chronological age and age at which 90% of children in normative sample pass the items in that subscale.A higher derived score on a subscale indicates better performance on items in that subscale relative to other children of same age who have taken the test.Among subscales,Personal-Social subscale ranges from -16 months to 24 months;others range from - 12 months to 24 months.All subscales have a population mean of 0 and a standard deviation of 3. (NCT00441896)
Timeframe: At Day 20

,,,,,,,,,
InterventionScores on a scale (Mean)
Personal Social at Day 20Fine Motor Adaptive at Day 20Language at Day 20Gross Motor at Day 20
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone-5.48-4.38-1.36-3.96
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo-16.90-14.63-12.33-13.63
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone-8.79-7.49-7.23-7.16
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo-8.73-7.70-5.17-7.00
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone-7.76-6.40-5.82-5.58
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo-5.35-5.05-4.95-3.00
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone-5.99-6.16-6.09-6.89
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo-10.22-10.24-8.22-8.74
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone-7.72-8.32-7.44-8.76
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo-12.93-12.87-10.77-13.43

[back to top]

Change From Baseline in Frequency of Spasm Clusters at Day 10

Spasm clusters were determined by a 24-hour video-electroencephalography (vEEG) at Day 10. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Baseline was defined as the Day 0 assessment before study drug infusion. (NCT00441896)
Timeframe: Baseline (Day 0) and Day 10

InterventionSpasm clusters per day (Mean)
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone1.0
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo2.7
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone-5.4
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo-1.0
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone-1.4
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo-5.3
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone-3.7
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo0.2
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone-3.2
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo-1.7

[back to top]

Number of Participants With Absence of Hypsarrhythmia

Absence of hypsarrhythmia was determined by 24-hour vEEG at Day 10 and Day 20. (NCT00441896)
Timeframe: Day 10 and Day 20

,,,,,,,,,
InterventionParticipants (Count of Participants)
Day 10Day 20
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone11
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo00
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone22
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo00
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone22
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo00
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone22
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo22
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone13
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo00

[back to top]

Change From Baseline in Frequency of Spasm Clusters at Day 20

Spasm clusters were determined by a 24-hour vEEG at Day 20. Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Baseline was defined as the Day 0 assessment before study drug infusion. (NCT00441896)
Timeframe: Baseline (Day 0) and Day 20

InterventionSpasm clusters per day (Mean)
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone-3.2
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo6.0
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone-5.0
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo-1.5
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone-1.1
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo-12.7
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone-4.4
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo-1.2
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone-3.2
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo-5.3

[back to top]

Number of Participants With Spasm-free Durations

Clinical spasms were determined by vEEG for at least 24 hours at Day 10 and Day 20. The number of participants with spasm-free duration have been presented. (NCT00441896)
Timeframe: Day 10 and Day 20

,,,,,,,,,
InterventionParticipants (Count of Participants)
Spasm-free Durations at Day 10Spasm-free Durations at Day 20
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone33
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo11
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone32
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo22
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone22
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo01
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone31
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo01
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone10
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo00

[back to top]

Number of Participants With Seizure-free Days

Seizure-free days were measured using data obtained from participants' daily dairy. Participants without seizures have been reported. (NCT00441896)
Timeframe: From Day 8 to Day 10 and From Day 18 to Day 20

,,,,,,,,,
InterventionParticipants (Count of Participants)
From Day 8 to Day 10From Day 18 to Day 20
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone66
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo21
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone66
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo34
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone1010
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo33
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone66
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo55
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone66
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo33

[back to top]

Number of Responders

A responder is defined as a participant experiencing a greater than equal to (>=) 50 percent (%) decrease in spasm frequency. Test for responders was conducted by vEEG for up to 24 hours at Day 10 and Day 20 (NCT00441896)
Timeframe: Day 10 and Day 20

,,,,,,,,,
InterventionParticipants (Count of Participants)
Day 10Day 20
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone22
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo10
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone34
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo02
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone33
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo01
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone24
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo21
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone21
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo11

[back to top]

Number of Participants With Change in Clinical Status on the Investigator's Global Assessment

The investigators rated the participants' overall clinical status based on 7 clinical factors: seizure frequency, duration, and intensity; adverse experiences; social, intellectual, and motor functioning. Using a 7-point scale ([1] marked improvement, [2] moderate improvement, [3] slight improvement, [4] no change from Baseline, [5] slight worsening, [6] moderate worsening, or [7] marked worsening). Higher score indicated worse symptoms. The investigators assessed the participants' status compared to their condition prior to initiating study medication. (NCT00441896)
Timeframe: Baseline (Day 0), Day 10 and Day 20

,,,,,,,,,
InterventionParticipants (Count of Participants)
Marked improvement at Day 10Moderate improvement at Day 10Slight improvement at Day 10No change from Baseline at Day 10Slight worsening at Day 10Moderate worsening at Day 10Severe worsening at Day 10Marked improvement at Day 20Moderate improvement at Day 20Slight improvement at Day 20No change from Baseline at Day 20Slight worsening at Day 20Moderate worsening at Day 20Severe worsening at Day 20
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone10140002121000
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo00030001200000
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone02140001132000
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo00130000112000
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone10270001162000
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo00120000030000
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone01240000223000
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo00230000131000
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone01230000141000
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo00030000102000

[back to top]

Number of Participants With Change in Clinical Status on Caregiver's Global Assessment

Caregiver global assessment of seizure severity and response to treatment rated the participants' based on 7 clinical factors: seizure frequency, duration, and intensity; adverse experiences; social, intellectual, and motor functioning. Using a 7-point scale ([1] marked improvement, [2] moderate improvement, [3] slight improvement, [4] no change from Baseline, [5] slight worsening, [6] moderate worsening, or [7] marked worsening). Higher score indicated worse symptoms. The assessment compared the participants' current status to their condition prior to initiating study medication. (NCT00441896)
Timeframe: Baseline (Day 0), Day10 and Day 20

,,,,,,,,,
InterventionParticipants (Count of Participants)
Marked improvement at Day 10Moderate improvement at Day 10Slight improvement at Day 10No change from Baseline at Day 10Slight worsening at Day 10Moderate worsening at Day 10Severe worsening at Day 10Marked improvement at Day 20Moderate improvement at Day 20Slight improvement at Day 20No change from Baseline at Day 20Slight worsening at Day 20Moderate worsening at Day 20Severe worsening at Day 20
Cohort 1: Sequence A: Ganaxolone/Placebo/Ganaxolone10140002121000
Cohort 1: Sequence B: Placebo/Ganaxolone/Placebo00120001200000
Cohort 2: Sequence A: Ganaxolone/Placebo/Ganaxolone02140001123000
Cohort 2: Sequence B: Placebo/Ganaxolone/Placebo00220000112000
Cohort 3: Sequence A: Ganaxolone/Placebo/Ganaxolone10540001162000
Cohort 3: Sequence B: Placebo/Ganaxolone/Placebo00120000120000
Cohort 4: Sequence A: Ganaxolone/Placebo/Ganaxolone01231000232000
Cohort 4: Sequence B: Placebo/Ganaxolone/Placebo00320000130100
Cohort 5: Sequence A: Ganaxolone/Placebo/Ganaxolone01230000141000
Cohort 5: Sequence B: Placebo/Ganaxolone/Placebo00120000003000

[back to top]

Percentage of Total Spasm-free Days and Cumulative Spasm-free Days as Determined From the Seizure Diary.

"Percentage of total spasm-free days during a visit period is defined as total number of spasm free days as recorded in the Seizure Diary/ total number of days during that period, multiplied by 100.~Percentage of cumulative total spasm-free days during a visit period is defined as sum of total number of spasm-free days during this period as recorded in the Seizure Diary/ total number of days in the treatment period, multiplied by 100." (NCT00442104)
Timeframe: Weeks 4 through Week 32

InterventionPercentage of Days (Mean)
Total Spasm-free days during Week 4 visitTotal Spasm-free days during Week 8 visitTotal Spasm-free days during Week 14 visitTotal Spasm-free days during Week 20 visitTotal Spasm-free days during Week 26 visitTotal Spasm-free days during week 32 visitCumulative Spasm-free days during Week 4 visitCumulative Spasm-free days during Week 8 visitCumulative Spasm-free days during Week 14 visitCumulative Spasm-free days during Week 20 visitCumulative Spasm-free days during Week 26 visitCumulative Spasm-free days during Week 32 visit
Ganaxolone11.1018.7420.7428.7928.7327.9910.8613.8215.7019.9722.0722.99

[back to top]

Number of Participants With Change in Clinical Status on Caregiver's Global Assessment

Caregiver global assessment of seizure severity and response to treatment rated the participants' based on 7 clinical factors: seizure frequency, duration, and intensity; adverse experiences; social, intellectual, and motor functioning. Using a 7-point scale ([1] marked improvement, [2] moderate improvement, [3] slight improvement, [4] no change from baseline, [5] slight worsening, [6] moderate worsening, or [7] marked worsening). Higher score indicated worse symptoms. The assessment compared the participants' current status to their condition prior to initiating study medication. (NCT00442104)
Timeframe: Week 4 through Week 32

Interventionparticipants (Number)
Caregiver's- Week 4 - Marked ImprovementCaregiver's - Week 4 - Moderate ImprovementCaregiver's -Week 4 - Slight ImprovementCaregiver's- Week 4 - No change from BaselineCaregiver's - Week 4 - Moderate WorseningCaregiver's - Week 4 - Slight WorseningCaregiver's - Week 4 - Marked WorseningCaregiver's - Week 8 - Marked ImprovementCaregiver's- Week 8 - Moderate ImprovementCaregiver's -Week 8 - Slight ImprovementCaregiver's- Week 8 - No change from BaselineCaregiver's - Week 8 - Slight WorseningCaregiver's - Week 8 - Moderate WorseningCaregiver's - Week 8 - Marked WorseningCaregiver's- Week 14 - Marked ImprovementCaregiver's - Week 14 - Moderate ImprovementCaregiver's -Week 14 - Slight ImprovementCaregiver's - Week 14 - No change from BaselineCaregiver's - Week 14 - Slight WorseningCaregiver's - Week 14 - Moderate WorseningCaregiver's - Week 14 - Marked WorseningCaregiver's - Week 20 - Marked ImprovementCaregiver's - Week 20 - Moderate ImprovementCaregiver's -Week 20 - Slight ImprovementCaregiver's- Week 20 - No change from BaselineCaregiver's- Week 20 - Slight WorseningCaregiver's - Week 20 - Moderate WorseningCaregiver's - Week 20 - Marked WorseningCaregiver's- Week 26 - Marked ImprovementCaregiver's- Week 26 - Moderate ImprovementCaregiver's -Week 26 - Slight ImprovementCaregiver's - Week 26 - No change from BaselineCaregiver's - Week 26 - Slight WorseningCaregiver's - Week 26 - Moderate WorseningCaregiver's - Week 26 - Marked WorseningCaregiver's - Week 32 - Marked ImprovementCaregiver's - Week 32 - Moderate ImprovementCaregiver's-Week 32 - Slight ImprovementCaregiver's - Week 32 - No change from BaselineCaregiver's - Week 32 - Slight WorseningCaregiver's - Week 32 - Moderate WorseningCaregiver's - Week 32 - Marked Worsening
Ganaxolone1111166010712181200813142100913103100912730008782010

[back to top]

Number of Participants With Absence of Spasms

Absence of spasms is defined as percentage of total spasm-free days during a visit period=100%. Parents/Guardians or nursing staff maintained a spasm/seizure diary to record the number and type of seizures each day, including spasms, throughout the entire study. The participants achieving absence of spasms have been presented. (NCT00442104)
Timeframe: Week 4 through Week 32

InterventionParticipants (Count of Participants)
Achieved absence of spasms at Week 4Achieved absence of spasms at Week 8Achieved absence of spasms at Week 14Achieved absence of spasms at Week 20Achieved absence of spasms at Week 26Achieved absence of spasms at Week 32
Ganaxolone233451

[back to top]

Developmental Assessment Using Denver-II Developmental Test

Denver-II Developmental Test measures a child's development in several areas:Personal-Social,Fine Motor-Adaptive,Language,and Gross Motor,from birth to 6 years old.It consists of 125 items that are organized into subscales and scored as pass,fail,or refused.To evaluate a child's progress,test compares their performance to a normative sample of children of same age.For each item,age at which 90% of children in normative sample pass it is determined.Derived score for each subscale is sum of item scores and represents difference between child's chronological age and age at which 90% of children in normative sample pass the items in that subscale.A higher derived score on a subscale indicates better performance on items in that subscale relative to other children of same age who have taken the test.Among subscales,Personal-Social subscale ranges from -16 months to 24 months;others range from - 12 months to 24 months.All subscales have a population mean of 0 and a standard deviation of 3. (NCT00442104)
Timeframe: Week 8 through Week 32

InterventionScores on a scale (Mean)
Week 8 - Personal SocialWeek 8 - Fine Motor-AdaptiveWeek 8 - LanguageWeek 8 - Gross MotorWeek 20- Personal SocialWeek 20- Fine Motor-AdaptiveWeek 20- LanguageWeek 20 - Gross MotorWeek 32-Personal SocialWeek 32-Fine Motor-AdaptiveWeek 32-LanguageWeek 32-Gross Motor
Ganaxolone-0.72-1.56-1.45-1.05-2.82-3.73-2.39-2.69-4.86-6.80-6.58-6.13

[back to top]

Number of Participants With Spasm-free Durations

Spasm-free duration is defined as total number of spasm-free days recorded in the spasm/seizure diary. Parents/Guardians or nursing staff maintained a spasm/seizure diary to record the number and type of seizures each day, including spasms, throughout the entire study. The number of participants with spasm-free duration of at least 24 hours have been presented. (NCT00442104)
Timeframe: Week 4 through Week 32

InterventionParticipants (Count of Participants)
Spasm Free at Week 4Spasm Free at Week 8Spasm Free at Week 14Spasm Free at Week 20Spasm Free at Week 26Spasm Free at Week 32
Ganaxolone191818181713

[back to top]

Percentage of Participants Who Were Free of Spasms

Clinical spasms were determined by video-electroencephalography (VEEG). The number of participants with spasm-free duration have been presented. (NCT00442104)
Timeframe: Weeks 4 through Week 96

Interventionpercentage of participants (Number)
Ganaxolone100

[back to top]

Change From Baseline in Frequency of Individual Spasm

Individual Spasm are seizures that may last only a second or two and were determined by a 24-hour video-electroencephalography (vEEG). Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Baseline was defined as the Day 0 assessment prior to ganaxolone dosing in Protocol 1042-0500. (NCT00442104)
Timeframe: Baseline and Week 4 through Week 32

InterventionIndividual spasms per day (Mean)
Individual Spasms - Week 4Individual Spasms - Week 8Individual Spasms - Week 14Individual Spasms - Week 20Individual Spasms - Week 26Individual Spasms - Week 32
Ganaxolone-6.8-17.4-20.8-15.8-19.9-17.7

[back to top]

Change From Baseline in Frequency of Spasm Clusters

Infantile spasms that come one after another in a cluster and lasts several minutes are called Spasm Clusters. Spasm clusters were determined by a 24-hour video-electroencephalography (vEEG). Change from Baseline was calculated as the post-Baseline value minus the Baseline value. Baseline was defined as the Day 0 assessment prior to ganaxolone dosing in Protocol 1042-0500. (NCT00442104)
Timeframe: Baseline and Week 4 through Week 32

InterventionSpasm clusters per day (Mean)
Spasm Clusters - Week 4Spasm Clusters - Week 8Spasm Clusters - Week 14Spasm Clusters - Week 20Spasm Clusters - Week 26Spasm Clusters - Week 32
Ganaxolone-2.9-4.1-4.1-5.8-5.6-5.3

[back to top]

Number of Participants With Change in Clinical Status on the Investigator's Global Assessment

The investigators rated the participants' overall clinical status based on 7 clinical factors: seizure frequency, duration, and intensity; adverse experiences; social, intellectual, and motor functioning. Using a 7-point scale ([1] marked improvement, [2] moderate improvement, [3] slight improvement, [4] no change from baseline, [5] slight worsening, [6] moderate worsening, or [7] marked worsening). Higher score indicated worse symptoms. The investigators assessed the participants' status compared to their condition prior to initiating study medication. (NCT00442104)
Timeframe: Week 4 through Week 32

Interventionparticipants (Number)
Investigator's- Week 4 - Marked ImprovementInvestigator's - Week 4 - Moderate ImprovementInvestigator's -Week 4 - Slight ImprovementInvestigator's- Week 4 - No change from BaselineInvestigator's - Week 4 - Moderate WorseningInvestigator's - Week 4 - Slight WorseningInvestigator's - Week 4 - Marked WorseningInvestigator's - Week 8 - Marked ImprovementInvestigator's- Week 8 - Moderate ImprovementInvestigator's -Week 8 - Slight ImprovementInvestigator's- Week 8 - No change from BaselineInvestigator's - Week 8 - Slight WorseningInvestigator's - Week 8 - Moderate WorseningInvestigator's - Week 8 - Marked WorseningInvestigator's- Week 14 - Marked ImprovementInvestigator's - Week 14 - Moderate ImprovementInvestigator's -Week 14 - Slight ImprovementInvestigator's - Week 14 - No change from BaselineInvestigator's - Week 14 - Slight WorseningInvestigator's - Week 14 - Moderate WorseningInvestigator's - Week 14 - Marked WorseningInvestigator's - Week 20 - Marked ImprovementInvestigator's - Week 20 - Moderate ImprovementInvestigator's -Week 20 - Slight ImprovementInvestigator's- Week 20 - No change from BaselineInvestigator's- Week 20 - Slight WorseningInvestigator's - Week 20 - Moderate WorseningInvestigator's - Week 20 - Marked WorseningInvestigator's- Week 26 - Marked ImprovementInvestigator's- Week 26 - Moderate ImprovementInvestigator's -Week 26 - Slight ImprovementInvestigator's - Week 26 - No change from BaselineInvestigator's - Week 26 - Slight WorseningInvestigator's - Week 26 - Moderate WorseningInvestigator's - Week 26 - Marked WorseningInvestigator's - Week 32 - Marked ImprovementInvestigator's - Week 32 - Moderate ImprovementInvestigator's -Week 32 - Slight ImprovementInvestigator's - Week 32 - No change from BaselineInvestigator's - Week 32 - Slight WorseningInvestigator's - Week 32 - Moderate WorseningInvestigator's - Week 32 - Marked Worsening
Ganaxolone715184100612193000616151000714111100514940006983100

[back to top]

Percent Change From Baseline in Mean Weekly Seizure Frequency by Subtype During Weeks 1 Through 10

Seizure subtypes included Complex partial seizures (CPS), Generalized tonic-clonic seizure (GTCS), and Simple partial seizure (SPS)-motor. Percent Change from Baseline in Mean Weekly Seizure frequency by seizure subtype is presented. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT00465517)
Timeframe: Baseline and at Week 1 through Week 10

,
InterventionPercent change (Mean)
CPSGTCSPS-motor
Ganaxolone-17.47-30.65-30.63
Placebo3.88-37.7519.70

[back to top]

Percent Change From Baseline in Mean Weekly Seizure Frequency During Weeks 3 Through 10

Summary of percent change from Baseline in mean weekly seizure frequency is presented. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT00465517)
Timeframe: Baseline and at Week 3 through Week 10

InterventionPercent change (Mean)
Ganaxolone-12.08
Placebo4.57

[back to top]

Change From Baseline in Mean Weekly Seizure Frequency During Weeks 1 Through 10

Summary of change from Baseline in mean weekly seizure frequency is presented. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT00465517)
Timeframe: Baseline and at Week 1 through Week 10

InterventionSeizures per week (Mean)
Ganaxolone-1.27
Placebo1.41

[back to top]

Change From Baseline in Mean Weekly Seizure Frequency During Weeks 3 Through 10

Summary of change from Baseline in mean weekly seizure frequency is presented. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT00465517)
Timeframe: Baseline and at Week 3 through Week 10

InterventionSeizures per week (Mean)
Ganaxolone-1.07
Placebo2.52

[back to top]

Mean Weekly Seizure Frequency for Each Week After Dosing During Weeks 1 to 10

Mean weekly Seizure Frequency for each week post-dosing During Weeks 1 to 10 is presented. (NCT00465517)
Timeframe: Weeks 1, 2, 3, 4, 5, 6, 7, 8, 9 and 10

,
InterventionSeizures per week (Mean)
Week 1Week 2Week 3Week 4Week 5Week 6Week 7Week 8Week 9Week 10
Ganaxolone4.535.095.025.695.125.656.684.635.074.83
Placebo7.258.797.809.1311.6410.4313.1513.4711.6413.60

[back to top]

Number of Responders During Weeks 1 Through 10

Responders were defined as participants experiencing ≥50% of reduction in mean weekly seizure frequency from the Baseline. Baseline was defined as the Day 0 assessment before study drug infusion. (NCT00465517)
Timeframe: Baseline and at Week 1 through Week 10

InterventionParticipants (Count of Participants)
Ganaxolone23
Placebo7

[back to top]

Number of Responders During Weeks 3 Through 10

Responders were defined as participants experiencing ≥50% of reduction in mean weekly seizure frequency from the Baseline. Baseline was defined as the Day 0 assessment before study drug infusion. (NCT00465517)
Timeframe: Baseline and at Week 3 through Week 10

InterventionParticipants (Count of Participants)
Ganaxolone25
Placebo6

[back to top]

Number of Seizure-free Days During Week 1 Through 10

Summary of Seizure-Free days is presented. (NCT00465517)
Timeframe: Week 1 through Week 10

InterventionSeizure free days (Mean)
Ganaxolone45.9
Placebo46.8

[back to top]

Number of Seizure-free Days During Week 3 Through 10

Summary of Seizure-Free days is presented. (NCT00465517)
Timeframe: Week 3 through Week 10

InterventionSeizure free days (Mean)
Ganaxolone37.1
Placebo38.5

[back to top]

Number of Seizure-free Days Up to Week 2

Summary of Seizure-Free days is presented. (NCT00465517)
Timeframe: Up to Week 2

InterventionSeizure free days (Mean)
Ganaxolone10.3
Placebo10.0

[back to top]

Number of Seizure-free Participants During Weeks 1 Through 10

Number of seizure-free participants is presented. (NCT00465517)
Timeframe: Week 1 through Week 10

InterventionParticipants (Count of Participants)
Ganaxolone1
Placebo0

[back to top]

Number of Seizure-free Participants During Weeks 3 Through 10

Number of seizure-free participants is presented. (NCT00465517)
Timeframe: Week 3 through Week 10

InterventionParticipants (Count of Participants)
Ganaxolone0
Placebo1

[back to top]

Number of Seizure-free Participants Up to Week 2

Number of seizure-free participants is presented. (NCT00465517)
Timeframe: Up to Week 2

InterventionParticipants (Count of Participants)
Ganaxolone16
Placebo4

[back to top]

Percent Change From Baseline in Mean Weekly Seizure Frequency During Weeks 1 Through 10

Summary of percent change from Baseline in mean weekly seizure frequency is presented. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT00465517)
Timeframe: Baseline and at Week 1 through Week 10

InterventionPercent change (Mean)
Ganaxolone-17.59
Placebo1.99

[back to top]

Mean Weekly Log-transformed Seizure Frequency During Weeks 1 Through 10

Weekly seizure frequency, analyzed as mean weekly log-transformed seizure frequency [including partial-onset seizures (POS) with or without secondary generalization, but not non-motor simple partial seizure (SPS)] during Weeks 1 through 10. (NCT00465517)
Timeframe: Week 1 through Week 10

Interventionlog[seizures per week] (Mean)
Ganaxolone5.20
Placebo10.77

[back to top]

Mean Weekly Log-transformed Seizure Frequency During Weeks 3 Through 10

Weekly seizure frequency, analyzed as mean weekly log-transformed seizure frequency [including POS with or without secondary generalization, but not non-motor SPS] during the Weeks 3 through 10 (NCT00465517)
Timeframe: Week 3 through Week 10

Interventionlog[seizures per week] (Mean)
Ganaxolone5.44
Placebo11.90

[back to top]

Number of Seizure-free Participants

Number of Seizure-free participants is presented. (NCT00512317)
Timeframe: Day 1 through Day 224 (Week 32)

,
InterventionParticipants (Count of Participants)
>=1 day>= 28 days (4 weeks)>= 56 days (8 weeks)>=70 days (10 weeks)>=91 days (13 weeks)>=119 days (17 weeks)>=133 days (19 weeks)>=154 days (22 weeks)>=182 days (26 weeks)>=224 days (32 weeks)
GNX/GNX7520644433300
PBO/GNX41383331292727252018

[back to top]

Percent Change From Baseline in Weekly Seizure Frequency During Weeks 1 Through 117

Percent Change in weekly seizure frequency by treatment group compared to Baseline at the beginning of the double-blind study 1042-0600 is presented. Weekly seizure frequency included partial-onset seizures (POS) with or without secondary generalization, but not non-motor simple partial seizure (SPS) during Weeks 1 through Week 117. Baseline was defined as the Day 0 assessment before study drug infusion of the double-blind study 1042-0600. (NCT00512317)
Timeframe: Baseline (Day 0) and Week 1 through Week 117

InterventionPercent Change (Mean)
Weeks 1-10Weeks 1-13Weeks >13-26Weeks >26-39Weeks >39-52Weeks >52-65Weeks >65-78Weeks >78-91Weeks >91-104Weeks >104-117
GNX/GNX-14.8-14.0-19.6-37.7-29.1-29.9-41.4-38.8-69.4-93.3

[back to top]

Percent Change From Baseline in Weekly Seizure Frequency During Weeks 1 Through 117

Percent Change in weekly seizure frequency by treatment group compared to Baseline at the beginning of the double-blind study 1042-0600 is presented. Weekly seizure frequency included partial-onset seizures (POS) with or without secondary generalization, but not non-motor simple partial seizure (SPS) during Weeks 1 through Week 117. Baseline was defined as the Day 0 assessment before study drug infusion of the double-blind study 1042-0600. (NCT00512317)
Timeframe: Baseline (Day 0) and Week 1 through Week 117

InterventionPercent Change (Mean)
Weeks 1-10Weeks 1-13Weeks >13-26Weeks >26-39Weeks >39-52Weeks >52-65Weeks >65-78Weeks >78-91Weeks >91-104
PBO/GNX-12.3-13.7-19.8-48.6-42.9-43.1-67.7-43.6-100

[back to top]

Change From Baseline in Quality of Life in Epilepsy Inventory-31 (QOLIE-31) Questionnaire

QOLIE-31 was a survey of health-related QOL for adults with epilepsy and evaluated how much distress the participant feels about problems and worries related to epilepsy. It included 38 items grouped into eight multi-item subscales - Energy/Fatigue, Emotional Well-Being, Daily Activities/Social Functioning, Cognitive Functioning, Medication Effect, Seizure Worry, Overall Quality of Life (QoL) and Distress. The subscale scores and the total score were calculated according to the scoring algorithm defined by the author with scores ranging from 0 to 100; higher scores indicated better function. Baseline was defined as the last non-missing observation prior to the first dose in double-blind study 1042-0600. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT00512317)
Timeframe: Baseline (Day 0) and up to Week 104

,
InterventionScores on a Scale (Mean)
Energy/FatigueEmotional Well-BeingSocial FunctionCognitiveMedication EffectSeizure WorryOverall QoLDistress
GNX/GNX-5.7-4.7-3.62.1-3.93.6-3.8-5.5
PBO/GNX3.93.38.42.90.9-3.10.00.7

[back to top]

Number of Responders During Weeks 1 Through 117

Responders were defined as participants experiencing ≥50% of reduction in mean weekly seizure frequency from the Baseline. Baseline was defined as the Day 0 assessment before study drug infusion of the double-blind study 1042-0600. (NCT00512317)
Timeframe: Baseline (Day 0) and Week 1 through Week 117

InterventionParticipants (Count of Participants)
Weeks 1-10Weeks 1-13Weeks >13-26Weeks >26-39Weeks >39-52Weeks >52-65Weeks >65-78Weeks >78-91Weeks >91-104Weeks >104-117
GNX/GNX221912111086431

[back to top]

Number of Responders During Weeks 1 Through 117

Responders were defined as participants experiencing ≥50% of reduction in mean weekly seizure frequency from the Baseline. Baseline was defined as the Day 0 assessment before study drug infusion of the double-blind study 1042-0600. (NCT00512317)
Timeframe: Baseline (Day 0) and Week 1 through Week 117

InterventionParticipants (Count of Participants)
Weeks 1-10Weeks 1-13Weeks >13-26Weeks >26-39Weeks >39-52Weeks >52-65Weeks >65-78Weeks >78-91Weeks >91-104
PBO/GNX11128121081141

[back to top]

Number of Seizure-free Days During Weeks 1 Through 117

Average number of seizure-free days per week for a given period was calculated as follows: (Total number of days with no seizures of any type during that period / number of days with seizure diary in that period) multiplied by 7. (NCT00512317)
Timeframe: Week 1 through Week 117

InterventionSeizure free days (Mean)
Weeks 1-10Weeks 1-13Weeks >13-26Weeks >26-39Weeks >39-52Weeks >52-65Weeks >65-78Weeks >78-91Weeks >91-104Weeks >104-117
GNX/GNX4.84.85.15.25.45.15.35.45.66.9

[back to top]

Number of Seizure-free Days During Weeks 1 Through 117

Average number of seizure-free days per week for a given period was calculated as follows: (Total number of days with no seizures of any type during that period / number of days with seizure diary in that period) multiplied by 7. (NCT00512317)
Timeframe: Week 1 through Week 117

InterventionSeizure free days (Mean)
Weeks 1-10Weeks 1-13Weeks >13-26Weeks >26-39Weeks >39-52Weeks >52-65Weeks >65-78Weeks >78-91Weeks >91-104
PBO/GNX5.05.05.35.45.45.25.74.76.8

[back to top]

Investigators Global Assessment

Investigator's global assessment of the patient's response to ganaxolone treatment was collected during the study to support continued trial participation. The investigator's global assessment of the patient's response to ganaxolone treatment was collected during the study to support continued trial participation. Responses categories were Improved Markedly, Improved Moderately, Improved Slightly and No change from baseline. (NCT01002820)
Timeframe: Screening through 52 weeks

InterventionParticipants (Count of Participants)
Improved Markedly - Visit 1 - ScreeningImproved Moderately - Visit 1 - ScreeningImproved Slightly - Visit 1 - ScreeningNo change from baseline - Visit 1 - ScreeningImproved Markedly - Visit 2 - Week 26Improved Moderately - Visit 2 - Week 26Improved Slightly - Visit 2 - Week 26No change from baseline - Visit 2 - Week 26Improved Markedly - Visit 3 - Week 52 or Taper VisitImproved Moderately - Visit 3 - Week 52 or Taper VisitImproved Slightly - Visit 3 - Week 52 or Taper VisitNo change from baseline - Visit 3 - Week 52 or Taper Visit
Ganaxolone640134002411

[back to top]

Number of Participants With Response to Clinical Global Impression - Improvement (Investigator) (CGI-II) Scale

The CGI-II scale is a 7-point scale where the investigator was required to assess how much the participant's illness has improved or worsened relative to a Baseline state at the beginning of the intervention, and rated as 1, very much improved; 2, much improved; 3, minimally improved; 4, no change; 5, minimally worse; 6, much worse; or 7, very much worse. Higher score indicated worse symptoms. The score was grouped into two categories for analysis: Responders=1 or 2, Non-responders=3-7. (NCT01339689)
Timeframe: Week 2, Week 4 and Week 6

,
InterventionParticipants (Count of Participants)
Week 2 - Non-ResponderWeek 2 - ResponderWeek 4 - Non-ResponderWeek 4 - ResponderWeek 6 - Non-ResponderWeek 6 - Responder
Ganaxolone4533852714
Placebo4273793212

[back to top]

Change From Baseline in Connor-Davidson Resilience Scale (CD-RISC) Score

The CD-RISC comprises 25 items, each rated on a 5-point scale (0-4), with higher scores reflecting greater resilience that may be a target for treatment in stress disorders. The total score is the sum of 25 item scores and ranged from 0: no symptoms to 100: worse symptoms. Higher score indicates worse symptoms. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value (NCT01339689)
Timeframe: Baseline (Week 0) and Week 6

InterventionScores on a scale (Mean)
Placebo60.84
Ganaxolone61.01

[back to top]

Change From Baseline in Insomnia Severity Index (ISI) Score

The ISI is a self-rated questionnaire assessing the difficulty over the past two weeks with falling to sleep, awakening in the middle of the night, and early morning awakening, as well as the degree to which insomnia impairs daily function. It included 5 items (rst item has 3 sub-questions), so a total of 7 questions. Each question is scored 0-4 (0=none, 1=mild, 2=moderate, 3=severe, 4=very severe). The total score is the sum of 7 item scores and ranged from 0: no symptoms to 28: severe symptoms. Higher score indicated worse symptoms. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01339689)
Timeframe: Baseline (Week 0) and Week 6

InterventionScores on a scale (Mean)
Placebo17.04
Ganaxolone14.36

[back to top]

Change From Baseline in Patient Health Questionnaire (PHQ-9) Score

The PHQ-9 is the nine-item depression scale of the Patient Health Questionnaire. The nine items of the PHQ-9 are based directly on the nine diagnostic criteria for major depressive disorder in the DSM-IV. Each item is scored 0-3 (0=not at all; 1=several days; 2=more than half the days; 3= nearly every day). The total score is the sum of 9 item scores and ranged 0: no depression to 27: severe depression. The higher the total score, the worse the depression. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01339689)
Timeframe: Baseline (Week 0) and Week 6

InterventionScores on a scale (Mean)
Placebo12.13
Ganaxolone10.71

[back to top]

Change From Baseline in Profile of Mood States (POMS) Total Score

The POMS is a factor-analytically derived 65 item self-report inventory with 5-point measures of five dimensions of negative mood: anger/irritability, anxiety/tension, depression/dejection, confusion/bewilderment, and fatigue/inertia, as well as vigor/activity. Total score was calculated as Fatigue + Mood (sum [Confusion, anger, Anxiety/Tension, Depression])- Vigor. Score ranged from 0: no symptoms to 200: severe symptoms; higher score indicated worse symptoms. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01339689)
Timeframe: Baseline (Week 0) and Week 6

InterventionScores on a scale (Mean)
Placebo80.07
Ganaxolone66.98

[back to top]

Change From Baseline in Clinician-Administered Posttrautamtic Stress Disorder (PTSD) Scale (CAPS) to Week 6

The CAPS was a structured interview that queries participants about each of the 17 Diagnostic and Statistical manual of Mental Disorders IV (DSM-IV) criteria B, C, and D symptoms of PTSD. Each item has a frequency score (0-4) and intensity score (0-4). The CAPS total score is the sum of frequency and intensity ratings for each item and the score range was 0: no symptoms to 136: severe symptoms. Higher score indicates worse symptoms. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01339689)
Timeframe: Baseline (Week 0) and at Week 2, Week 4 and Week 6

,
InterventionScores on a scale (Mean)
Week 2Week 4Week 6
Ganaxolone62.6761.3054.05
Placebo65.7863.260.64

[back to top]

Change From Baseline in PTSD Avoidance Score

The CAPS was a structured interview that queries participants about each of the 17 Diagnostic and Statistical manual of Mental Disorders IV (DSM-IV) criteria B, C, and D symptoms of PTSD. Each item has a frequency score (0-4) and intensity score (0-4). The Avoidance score is the sum of frequency and intensity ratings for items from cluster C.and the score range was 0: no symptoms to 56: severe symptoms. Higher score indicates worse symptoms. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01339689)
Timeframe: Baseline (Week 0) and at Week 2, Week 4 and Week 6

,
InterventionScores on a scale (Mean)
Week 2Week 4Week 6
Ganaxolone26.2926.0923.52
Placebo27.0625.8724.82

[back to top]

Change From Baseline in PTSD Checklist (PCL) Scores

The PCL is a 17-item self-report scale representing all DSM-IV criteria for PTSD, including re-experiencing, avoidance and numbing, and hyperarousal criteria. Each item is scored 1-5 (1=not at all; 2=a little bit; 3=moderately; 4=quite a bit; 5=extremely). The total score is the sum of 17 item scores and ranged from 17: no symptoms to 85: severe symptoms. Higher score indicated worse symptoms. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01339689)
Timeframe: Baseline (Week 0) and at Week 2, Week 4 and Week 6

,
InterventionScores on a scale (Mean)
Week 2Week 4Week 6
Ganaxolone50.7948.7446.33
Placebo54.452.9351.75

[back to top]

Change From Baseline in PTSD Hyperarousal Score

The CAPS was a structured interview that queries participants about each of the 17 Diagnostic and Statistical manual of Mental Disorders IV (DSM-IV) criteria B, C, and D symptoms of PTSD. Each item has a frequency score (0-4) and intensity score (0-4). The Hyperarousal score is the sum of frequency and intensity ratings for items from cluster D and the score range was 0: no symptoms to 40: severe symptoms. Higher score indicates worse symptoms. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01339689)
Timeframe: Baseline (Week 0) and at Week 2, Week 4 and Week 6

,
InterventionScores on a scale (Mean)
Week 2Week 4Week 6
Ganaxolone21.9421.0518.21
Placebo22.1620.7820.32

[back to top]

Change From Baseline in PTSD Re-Experience Score

The CAPS was a structured interview that queries participants about each of the 17 Diagnostic and Statistical manual of Mental Disorders IV (DSM-IV) criteria B, C, and D symptoms of PTSD. Each item has a frequency score (0-4) and intensity score (0-4). The Re-experience score is the sum of frequency and intensity ratings for items from cluster B and the score range was 0: no symptoms to 40: severe symptoms. Higher score indicates worse symptoms. Baseline was defined as the Day 0 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01339689)
Timeframe: Baseline (Week 0) and at Week 2, Week 4 and Week 6

,
InterventionScores on a scale (Mean)
Week 2Week 4Week 6
Ganaxolone14.4514.1612.31
Placebo16.5616.5415.5

[back to top]

Number of Participants With Response to CGI-I Subject Scale Clinical Global Impression - Improvement (Subject) (CGI-IS) Scale

The CGI-IS scale is a 7-point scale where the participant assesses how much the illness has improved or worsened relative to a Baseline state at the beginning of the intervention, and rated as 1, very much improved; 2, much improved; 3, minimally improved; 4, no change; 5, minimally worse; 6, much worse; or 7, very much worse. Higher score indicated worse symptoms. The score was grouped into two categories for analysis: Responders=1 or 2, Non-responders=3-7. (NCT01339689)
Timeframe: Week 2, Week 4 and Week 6

,
InterventionParticipants (Count of Participants)
Week 2 - Non-ResponderWeek 2 - ResponderWeek 4 - Non-ResponderWeek 4 - ResponderWeek 6 - Non-ResponderWeek 6 - Responder
Ganaxolone462376375
Placebo455397377

[back to top]

Clinical Global Impression-Improvement (CGI-I) Scale

"The CGI-I scale is a clinician-rated 7-point scale used to assess how much the participant's illness had improved or worsened relative to a Baseline state at the beginning of the intervention. It was rated as: 1. very much improved; 2. much improved; 3. minimally improved; 4. no change; 5. minimally worse; 6. much worse; 7. very much worse. Higher scores indicated worse condition." (NCT01725152)
Timeframe: Week 14 (End of Treatment)

InterventionScores on a scale (Least Squares Mean)
Ganaxolone3.4
Placebo3.5

[back to top]

Pediatric Anxiety Rating Scale (PARS) Total Score

Pediatric Anxiety Rating Scale (PARS) is a clinician administered measure of anxiety in children and adolescents. The PARS is comprised of a 50-item symptom checklist used to determine the presence or absence of specific anxiety symptoms during the prior week and 7 severity/impairment items, each scored from 0 to 5. The score on the 7 items allows the clinician to rate symptom severity and associated impairment on a range from 0 to 35, with higher scores reflecting greater symptom severity and associated impairment. (NCT01725152)
Timeframe: Week 14 (End of Treatment)

InterventionScores on a scale (Least Squares Mean)
Ganaxolone8.3
Placebo9.2

[back to top]

Visual Analogue Scale (VAS)

"A semi-structured VAS design was employed, providing two of the three behaviors to be assessed as anxiety and attention, chosen by caregiver(s) of one additional behavior from a bank of five. This bank included sociability, attention, aggression, language, and hyperactivity/impulsivity. Parents mark on a visual line measuring 10 centimeters (cm) with worst behavior at 0 cm and best behavior at 10 cm. For each behavior the caregiver is instructed to mark their impression of behavior at Baseline visit and again at each visit. The calculated distance in cm between the baseline and each visit marks thereby demonstrates whether each behavior improved, worsened, or stayed the same during the study, and by how much. Shown here is the least square mean distance and its corresponding standard error in cm from the worst behavior side, at baseline. Smaller the value, worser the behavior." (NCT01725152)
Timeframe: Week 14 (End of Treatment)

,
InterventionCentimeters (cm) (Least Squares Mean)
Severity of AnxietySeverity of AttentionSeverity of Target Behavior 3
Ganaxolone5.64.44.4
Placebo5.03.93.9

[back to top]

Swanson, Nolan, and Pelham-IV Questionnaire (SNAP-IV)

The SNAP-IV is a revision of the Swanson, Nolan, and Pelham (SNAP) Questionnaire. The SNAP-IV: ADHD Inattention Subscale (items 1-9) scores the intensity of each item during the last seven days on a 0 to 3 scale (0=not at all, 1=just a little, 2=pretty much, 3=very much). Possible scores ranged from 0-27; higher scores indicated a greater intensity. The SNAP-IV ADHD Hyperactivity/Impulsivity Subscale (items 10-18) scores the intensity of each item in the last seven days on a 0 to 3 scale (0=not at all, 1=just a little, 2=pretty much, 3=very much). Possible scores ranged from 0-27; higher scores indicated a greater intensity. SNAP-IV ADHD Combined Scale score (inattention + hyperactivity/impulsivity) ranged from 0-54. A low score of 0 indicates less inattention + hyperactivity/impulsivity. A high score of 54 indicates more inattention + hyperactivity. (NCT01725152)
Timeframe: Week 14 (End of Treatment)

,
InterventionScores on a scale (Least Squares Mean)
ADHD InattentionADHD Hyperactivity/ImpulsiveADHD Combined
Ganaxolone15.513.929.3
Placebo14.612.627.1

[back to top]

Anxiety, Depression, and Mood Scale (ADAMS)

The ADAMS is a 28-item behavior-based informant instrument rated by the parent(s), legal authorized guardian(s), or consistent caregiver(s). The scale is composed of 5 factors, which addresses Manic/Hyperactive Behavior, Depressed Mood, Social Avoidance, General Anxiety, and Obsessive/Compulsive Behavior. Items are scored on a 0-3 Likert scale that combines frequency and severity ratings (where 0=behavior has not occurred or is not a problem, 3=behavior occurs a lot or is a severe problem). Each subscale score is calculated separately; the scale range for Manic/Hyperactive Behavior is 0-15; for Depressed Mood, 0-21; for Social Avoidance, 0-21; for General Anxiety, 0-21; for Obsessive Behavior, 0-9. There is an overlapped item between Manic/Hypertension Behavior and General Anxiety; hence, the subscale ranges appear to reflect scores for 29 items instead of 28 items. (NCT01725152)
Timeframe: Week 14 (End of Treatment)

,
InterventionScores on a scale (Least Squares Mean)
Manic/Hyperactive Behavior TotalDepress Mood TotalSocial Avoidance TotalGeneral Anxiety TotalObsessive/Compulsive Behavior Total
Ganaxolone7.43.55.86.22.7
Placebo7.82.66.37.02.8

[back to top]

Aberrant Behavior Checklist (ABC)

The ABC is a 58-item parent rated from 0 (not at all a problem) to 3 (the problem is severe in degree with 6 subscales: Irritability (includes agitation, aggression, and self-injury, 15 items) with range of scores from 0-45; Social Withdrawal/Lethargy (16 items) with range of scores from 0-48; Stereotypy (7 items) with range of scores from 0-21; Hyperactivity (16 items) with range of scores from 0-48; Inappropriate Speech (4 items) with range of scores from 0-12 and Social avoidance (4 items) with range of scores from 0-12. Higher scores indicated greater severity. Social Withdrawal and Lethargy are reporting the same scale and Social Avoidance is a subscale of Social Withdrawal; hence, the subscale ranges appear to reflect scores for 62 items instead of 58 items. (NCT01725152)
Timeframe: Week 14 (End of Treatment)

,
InterventionScores on a scale (Least Squares Mean)
Total - Subscale I (Irritability)Total - Subscale II (Social Withdrawal/Lethargy)Total - Subscale III - (Stereotypy)Total - Subscale IV (Hyperactivity)Total - Subscale V (Inappropriate Speech)Total - Subscale VI (Social Avoidance)
Ganaxolone15.75.65.711.35.12.4
Placebo16.15.76.412.35.32.8

[back to top]

Percentage of Change in Expired Air Carbon Monoxide (CO) at End of Week 2

To evaluate the effects of ganaxolone on ad lib smoking by looking at the percent change in expired air CO at the end of week two (relative to baseline). (NCT01857531)
Timeframe: Baseline and 2 Weeks

Interventionpercentage change (Mean)
Ganaxolone -- Nicotine Patch-0.52

[back to top]

Number of Participants Completing Continuous 6-week Abstinence From Smoking

Continuous six week abstinence from smoking at final study visit (approximately 6 weeks post quit date), based on self-reported abstinence confirmed by expired air CO (NCT01857531)
Timeframe: 6 Weeks post quit

Interventionparticipants (Number)
Ganaxolone -- Nicotine Patch4

[back to top]

Number of Participants Completing Abstinence From Smoking During the Last Four Weeks of Treatment

End of treatment abstinence from smoking during the last four weeks of treatment, based on self-reported abstinence during last four weeks confirmed by expired air CO (NCT01857531)
Timeframe: 4 Week abstinence from smoking at 6 weeks post quit

Interventionparticipants (Number)
Ganaxolone -- Nicotine Patch5

[back to top]

Number of Participants Completing Continuous 2-week Abstinence From Smoking

Continuous two week abstinence from smoking at the first post-quit visit (approximately 2 weeks post quit date), based on self-reported abstinence confirmed by expired air CO. (NCT01857531)
Timeframe: 2 Weeks post quit

Interventionparticipants (Number)
Ganaxolone -- Nicotine Patch3

[back to top]

Number of Participants Completing Point Abstinence From Smoking Two Weeks After Quitting

Point abstinence from smoking two weeks post quit, based on self-reported abstinence during last seven days confirmed by expired air CO at first post-quit visit. (NCT01857531)
Timeframe: 7 day point abstinence from smoking at 2 weeks post quit

Interventionparticipants (Number)
Ganaxolone -- Nicotine Patch4

[back to top]

Percentage of Change in Expired Air Carbon Monoxide (CO) at End of Week 4

To evaluate the effects of ganaxolone as an augmentation treatment in conjunction with nicotine patch by looking at the percent change in expired air CO at the end of week four (relative to baseline). (NCT01857531)
Timeframe: Baseline and 4 Weeks

Interventionpercentage change (Mean)
Ganaxolone -- Nicotine Patch-39.40

[back to top]

Double Blind: Cohort 2: Percentage of Responders Experiencing a ≥R% (80%, 60%, 40%, and 20%) Reduction From Baseline to the End of Treatment Period in 28-day Seizure Frequency During Maintenance Period

Percentage of participants who had reductions of ≥ 80%, ≥ 60%, ≥ 40%, and ≥ 20% in 28-day seizure frequency from Baseline is presented. A responder is an individual whose reduction of percent change from Baseline in 28-day seizure frequency was ≥ 50%. Baseline was defined as non-missing value of last assessment before first dose. (NCT01963208)
Timeframe: Week 2 to Week 14

,
InterventionPercentage of participants (Number)
Reduction ≥ 80%Reduction ≥ 60%Reduction ≥ 40%Reduction ≥ 20%
Double Blind: Cohort 2 - Ganaxolone8.4324.1633.7147.19
Double Blind: Cohort 2 - Placebo5.8118.0231.442.44

[back to top]

Double Blind: Cohort 2: Percentage of Responders Experiencing a ≥R% (80%, 60%, 40%, and 20%) Reduction From Baseline to the End of Treatment Period in 28-day Seizure Frequency During Titration + Maintenance Period

Percentage of participants who had reductions of ≥ 80%, ≥ 60%, ≥ 40%, and ≥ 20% in 28-day seizure frequency from Baseline is presented. A responder is an individual whose reduction of percent change from Baseline in 28-day seizure frequency was ≥ 50%. Baseline was defined as non-missing value of last assessment before first dose. (NCT01963208)
Timeframe: Up to Week 14

,
InterventionPercentage of participants (Number)
Reduction ≥ 80%Reduction ≥ 60%Reduction ≥ 40%Reduction ≥ 20%
Double Blind: Cohort 2 - Ganaxolone7.320.7933.1551.69
Double Blind: Cohort 2 - Placebo2.3316.8629.6543.6

[back to top]

Double Blind: Cohort 2: Number of Participants With Clinical Global Impression of Change - Improvement (CGI-I) at Week 8

"The CGI-I scale is a clinician-rated 7-point Likert scale used to assess the degree to which the participant's epilepsy symptoms have changed relative to Baseline. It was rated as 1. very much improved; 2. much improved; 3. minimally improved; 4. no change; 5. minimally worse; 6. much worse; 7. very much worse. Higher scores indicated worse condition. Baseline was defined as non-missing value of last assessment before first dose." (NCT01963208)
Timeframe: At Week 8

,
InterventionParticipants (Count of Participants)
Very much improvedMuch improvedMinimally improvedNo changeMinimally worseMuch worseVery much worse
Double Blind: Cohort 2 - Ganaxolone3275068891
Double Blind: Cohort 2 - Placebo42049711251

[back to top]

Double Blind: Cohort 2: Number of Participants With Clinical Global Impression of Change - Improvement (CGI-I) at Week 14

"The CGI-I scale is a clinician-rated 7-point Likert scale used to assess the degree to which the participant's epilepsy symptoms have changed relative to Baseline. It was rated as 1. very much improved; 2. much improved; 3. minimally improved; 4. no change; 5. minimally worse; 6. much worse; 7. very much worse. Higher scores indicated worse condition. Baseline was defined as non-missing value of last assessment before first dose." (NCT01963208)
Timeframe: At Week 14

,
InterventionParticipants (Count of Participants)
Very much improvedMuch improvedMinimally improvedNo changeMinimally worseMuch worseVery much worse
Double Blind: Cohort 2 - Ganaxolone7284156620
Double Blind: Cohort 2 - Placebo5304767820

[back to top]

Double Blind: Cohort 2: Percent Change From Baseline in 28-day Seizure Frequency During Maintenance Period

Seizure frequency was based on the number of seizures per 28 days, calculated as the number of seizures over the time interval multiplied by 28 and divided by the number of days in the interval. Baseline 28-day seizure frequency was calculated as the number of seizures in the Baseline period (≤ 56 days) divided by the number of days with available seizure data in the Baseline period and multiplied by 28. Baseline was defined as non-missing value of last assessment before first dose. (NCT01963208)
Timeframe: Baseline and Week 2 to Week 14

InterventionPercent change (Median)
Double Blind: Cohort 2 - Ganaxolone-20.56
Double Blind: Cohort 2 - Placebo-12.50

[back to top]

Double Blind: Cohort 2: Percentage of Seizure Free Participants During the Maintenance Period

Percentage of participants who completed the study without any seizures is presented (NCT01963208)
Timeframe: Week 2 to Week 14

InterventionPercentage of participants (Number)
Double Blind: Cohort 2 - Ganaxolone1.12
Double Blind: Cohort 2 - Placebo0

[back to top]

Double Blind: Cohort 2: Percentage of Participants Who Experienced at Least One 28-day Seizure Free Period During Titration + Maintenance Phase

Percentage of participants who experienced at least one 28-day seizure free period is presented (NCT01963208)
Timeframe: Up to Week 14

InterventionPercentage of participants (Number)
Double Blind: Cohort 2 - Ganaxolone17.98
Double Blind: Cohort 2 - Placebo18.02

[back to top]

Double Blind: Cohort 2: Percent Change From Baseline in 28-day Seizure Frequency During Titration + Maintenance Period

Seizure frequency was based on the number of seizures per 28 days, calculated as the number of seizures over the time interval multiplied by 28 and divided by the number of days in the interval. Baseline 28-day seizure frequency was calculated as the number of seizures in the Baseline period (≤ 56 days) divided by the number of days with available seizure data in the Baseline period and multiplied by 28. Baseline was defined as non-missing value of last assessment before first dose. Primary analysis was performed using a rank analysis of covariance (ANCOVA). (NCT01963208)
Timeframe: Baseline and Week 14

InterventionPercent change (Median)
Double Blind: Cohort 2 - Ganaxolone-21.28
Double Blind: Cohort 2 - Placebo-10.25

[back to top]

Double Blind: Cohort 2: Percent Change From Baseline in 28-day Seizure Frequency for Different Subtypes of Seizures During Titration + Maintenance Period

Seizure frequency was based on the number of seizures per 28 days, calculated as the number of seizures over the time interval multiplied by 28 and divided by the number of days in the interval. The analysis was conducted for Partial-Onset Seizure (POS) only which included seizure subtypes: Complex partial seizures (CPS), secondarily generalized tonic-clonic (SGTC) seizures, simple partial seizure with motor/observable component (SPS-Motor) and Simple partial seizure (SPS) without motor/observable component. Baseline 28-day seizure frequency was calculated as the number of seizures in the Baseline period (≤ 56 days) divided by the number of days with available seizure data in the Baseline period and multiplied by 28. Baseline was defined as non-missing value of last assessment before first dose. (NCT01963208)
Timeframe: Baseline and Week 14

,
InterventionPercent change (Mean)
CPSSGTCSPS-MotorSPS
Double Blind: Cohort 2 - Ganaxolone-4.70-27.42-5.5212.57
Double Blind: Cohort 2 - Placebo-6.521.02-21.97-3.53

[back to top]

Double Blind: Cohort 2: Number of Participants With Patient Global Impression of Change - Improvement (PGI-I) at Week 8 and Week 14

"The PGI-I scale was a 7-point Likert scale completed by the Patient or Caregiver representing the degree to which the participant's epilepsy symptoms had changed relative to Baseline. It was rated as 1. very much improved; 2. much improved; 3. minimally improved; 4. no change; 5. minimally worse; 6. much worse; 7. very much worse. Higher score indicated worse condition. Baseline was defined as non-missing value of last assessment before first dose." (NCT01963208)
Timeframe: Week 8 and Week 14

,
InterventionParticipants (Count of Participants)
Week 8: Very Much ImprovedWeek 8: Much ImprovedWeek 8: Minimally ImprovedWeek 8: No ChangeWeek 8: Minimally WorseWeek 8: Much WorseWeek 8: Very Much WorseWeek 14: Very Much ImprovedWeek 14: Much ImprovedWeek 14: Minimally ImprovedWeek 14: No ChangeWeek 14: Minimally WorseWeek 14: Much WorseWeek 14: Very Much Worse
Double Blind: Cohort 2 - Ganaxolone730445912867334346731
Double Blind: Cohort 2 - Placebo52151651144102943601241

[back to top]

Double Blind: Cohort 2: Longest Percent of Time Spent Seizure-free During Titration + Maintenance Period

The longest period of time seizure-free was defined as the percent of the longest seizure-free period (days) divided by the days with available seizure data, and then multiplied by 100%. (NCT01963208)
Timeframe: Up to Week 14

InterventionPercentage of time spent (Mean)
Double Blind: Cohort 2 - Ganaxolone24.00
Double Blind: Cohort 2 - Placebo17.58

[back to top]

Double Blind: Cohort 2: Change From Baseline in the Number of Seizure Free Days Per 28-day Period During Titration + Maintenance Period

Baseline number of seizure free days per 28-day period was calculated as: the number of seizure free days in the entire Baseline period (<=56 days) divided by the number of days with available seizure data in the baseline period and multiplied by 28. Post-Baseline number of seizure free days per 28-day period was calculated as: the number of seizure free days in the entire treatment period divided by the number of days with available seizure data in the treatment period and multiplied by 28. Change from Baseline in number of seizure free days per 28-day period from Baseline was calculated as: Post-Baseline number of seizure free days per 28-day period minus Baseline number of seizure free days per 28-day period. Baseline was defined as non-missing value of last assessment before first dose. (NCT01963208)
Timeframe: Baseline and Week 14

InterventionSeizure free days (Mean)
Double Blind: Cohort 2 - Ganaxolone1.47
Double Blind: Cohort 2 - Placebo1.01

[back to top]

Double Blind: Cohort 2: Change From Baseline in the Number of Seizure Free Days Per 28-day Period During Maintenance Period

Baseline number of seizure free days per 28-day period was calculated as: the number of seizure free days in the entire Baseline period (≤ 56 days) divided by the number of days with available seizure data in the Baseline period and multiplied by 28. Post-Baseline number of seizure free days per 28-day period was calculated as: the number of seizure free days in the entire treatment period divided by the number of days with available seizure data in the treatment period and multiplied by 28. Change from Baseline in number of seizure free days per 28-day period from Baseline was calculated as: Post-Baseline number of seizure free days per 28-day period minus Baseline number of seizure free days per 28-day period. Baseline was defined as non-missing value of last assessment before first dose. (NCT01963208)
Timeframe: Baseline and Week 2 to Week 14

InterventionSeizure free days (Mean)
Double Blind: Cohort 2 - Ganaxolone1.63
Double Blind: Cohort 2 - Placebo1.20

[back to top]

Double Blind: Cohort 2: Change From Baseline in 28-day Seizure Frequency During Titration + Maintenance Period

Seizure frequency was based on the number of seizures per 28 days, calculated as the number of seizures over the time interval multiplied by 28 and divided by the number of days in the interval. Baseline 28-day seizure frequency was calculated as the number of seizures in the Baseline period (≤ 56 days) divided by the number of days with available seizure data in the Baseline period and multiplied by 28. Baseline was defined as non-missing value of last assessment before first dose. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01963208)
Timeframe: Baseline and Week 14

InterventionSeizures per 28 days (Mean)
Double Blind: Cohort 2 - Ganaxolone-1.46
Double Blind: Cohort 2 - Placebo-0.33

[back to top]

Double Blind: Cohort 2: Change From Baseline in 28-day Seizure Frequency During Maintenance Period

Seizure frequency was based on the number of seizures per 28 days, calculated as the number of seizures over the time interval multiplied by 28 and divided by the number of days in the interval. Baseline 28-day seizure frequency was calculated as the number of seizures in the Baseline period (≤ 56 days) divided by the number of days with available seizure data in the Baseline period and multiplied by 28. Baseline was defined as non-missing value of last assessment before first dose. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT01963208)
Timeframe: Baseline and Week 2 to Week 14

InterventionSeizures per 28 days (Mean)
Double Blind: Cohort 2 - Ganaxolone-1.67
Double Blind: Cohort 2 - Placebo-0.64

[back to top]

Double Blind: Cohort 2: Number of Participants With ≥50% Responder Rate During Titration + Maintenance Period

A 50% responder was a participant who experienced at least a 50% decrease in 28-day seizure frequency compared to Baseline. (NCT01963208)
Timeframe: Up to Week 14

InterventionParticipants (Count of Participants)
Double Blind: Cohort 2 - Ganaxolone50
Double Blind: Cohort 2 - Placebo39

[back to top]

Summary of CGII-P

Patient Global Impression of Change score as assessed by questionnaire. [ Time Frame: 78 Weeks ] CGII-P scale is qualitative values and not quantitative. (NCT02358538)
Timeframe: Patient Global Impression of Change score as assessed by questionnaire. [ Time Frame: 78 Weeks ]

,,,
InterventionParticipants (Count of Participants)
Visit 4 (End of Week 4) - Very Much ImprovedVisit 4 (End of Week 4) - Much ImprovedVisit 4 (End of Week 4) - Minimally ImprovedVisit 4 (End of Week 4) - No ChangeVisit 4 (End of Week 4) - Minimally WorseVisit 4 (End of Week 4) - Much WorseVisit 4 (End of Week 4) - Very Much WorseVisit 5 (End of Week 8) - Very Much ImprovedVisit 5 (End of Week 8) - Much ImprovedVisit 5 (End of Week 8) - Minimally ImprovedVisit 5 (End of Week 8) - No ChangeVisit 5 (End of Week 8) - Minimally WorseVisit 5 (End of Week 8) - Much WorseVisit 5 (End of Week 8) - Very Much WorseVisit 6 (End of Week 17) - Very Much ImprovedVisit 6 (End of Week 17) - Much ImprovedVisit 6 (End of Week 17) - Minimally ImprovedVisit 6 (End of Week 17) - No ChangeVisit 6 (End of Week 17) - Minimally WorseVisit 6 (End of Week 17) - Much WorseVisit 6 (End of Week 17) - Very Much WorseVisit 7 (End of Week 26) - Very Much ImprovedVisit 7 (End of Week 26) - Much ImprovedVisit 7 (End of Week 26) - Minimally ImprovedVisit 7 (End of Week 26) - No ChangeVisit 7 (End of Week 26) - Minimally WorseVisit 7 (End of Week 26) - Much WorseVisit 7 (End of Week 26) - Very Much WorseVisit 8 (End of Week 44) - Very Much ImprovedVisit 8 (End of Week 44) - Much ImprovedVisit 8 (End of Week 44) - Minimally ImprovedVisit 8 (End of Week 44) - No ChangeVisit 8 (End of Week 44) - Minimally WorseVisit 8 (End of Week 44) - Much WorseVisit 8 (End of Week 44)- Very Much WorseVisit 9 (End of Week 62) - Very Much ImprovedVisit 9 (End of Week 62) - Much ImprovedVisit 9 (End of Week 62)- Minimally ImprovedVisit 9 (End of Week 62) - No ChangeVisit 9 (End of Week 62) - Minimally WorseVisit 9 (End of Week 62) - Much WorseVisit 9 (End of Week 62) - Very Much WorseVisit 10 (End of Week 78) - Very Much ImprovedVisit 10 (End of Week 78) - Much ImprovedVisit 10 (End of Week 78)- Minimally ImprovedVisit 10 (End of Week 78) - No ChangeVisit 10 (End of Week 78) - Minimally WorseVisit 10 (End of Week 78) - Much WorseVisit 10 (End of Week 78) - Very Much Worse
CDKL50142000015010012200000401110020200022000001200000
CSWSNANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANA
Lennox-Gastaut3230000223100011301001131010000110000100000010000
PCDH191253000022111011400002230110200000020000002010010

[back to top]

Summary of CGII-C

Clinician Global Impression of Change score as assessed by questionnaire. [ Time Frame: 78 Weeks ] CGII-C scale is qualitative values and not quantitative. (NCT02358538)
Timeframe: End of Week 4, End of Week 8, End of Week 17, End of Week 26, Week 44, Week 62, Week 78

,,,
InterventionParticipants (Count of Participants)
Visit 4 (End of Week 4) - Very Much ImprovedVisit 4 (End of Week 4) - Much ImprovedVisit 4 (End of Week 4) - Minimally ImprovedVisit 4 (End of Week 4) - No ChangeVisit 4 (End of Week 4) - Minimally WorseVisit 4 (End of Week 4) - Much WorseVisit 4 (End of Week 4) - Very Much WorseVisit 5 (End of Week 8) - Very Much ImprovedVisit 5 (End of Week 8) - Much ImprovedVisit 5 (End of Week 8) - Minimally ImprovedVisit 5 (End of Week 8) - No ChangeVisit 5 (End of Week 8) - Minimally WorseVisit 5 (End of Week 8) - Much WorseVisit 5 (End of Week 8) - Very Much WorseVisit 6 (End of Week 17) - Very Much ImprovedVisit 6 (End of Week 17) - Much ImprovedVisit 6 (End of Week 17) - Minimally ImprovedVisit 6 (End of Week 17) - No ChangeVisit 6 (End of Week 17) - Minimally WorseVisit 6 (End of Week 17) - Much WorseVisit 6 (End of Week 17) - Very Much WorseVisit 7 (End of Week 26) - Very Much ImprovedVisit 7 (End of Week 26) - Much ImprovedVisit 7 (End of Week 26) - Minimally ImprovedVisit 7 (End of Week 26) - No ChangeVisit 7 (End of Week 26) - Minimally WorseVisit 7 (End of Week 26) - Much WorseVisit 7 (End of Week 26) - Very Much WorseVisit 8 (End of Week 44) - Very Much ImprovedVisit 8 (End of Week 44) - Much ImprovedVisit 8 (End of Week 44) - Minimally ImprovedVisit 8 (End of Week 44) - No ChangeVisit 8 (End of Week 44) - Minimally WorseVisit 8 (End of Week 44) - Much WorseVisit 8 (End of Week 44) - Very Much WorseVisit 9 (End of Week 62) - Very Much ImprovedVisit 9 (End of Week 62) - Much ImprovedVisit 9 (End of Week 62) - Minimally ImprovedVisit 9 (End of Week 62) - No ChangeVisit 9 (End of Week 62) - Minimally WorseVisit 9 (End of Week 62) - Much WorseVisit 9 (End of Week 62) - Very Much WorseVisit 10 (End of Week 78) - Very Much ImprovedVisit 10 (End of Week 78) - Much ImprovedVisit 10 (End of Week 78) - Minimally ImprovedVisit 10 (End of Week 78) - No ChangeVisit 10 (End of Week 78) - Minimally WorseVisit 10 (End of Week 78) - Much WorseVisit 10 (End of Week 78) - Very Much Worse
CDKL50322000042100012200000312010031000004000000300000
CSWSNANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANANA
Lennox-Gastaut2420000142100023010001112110000110000100000010000
PCDH191433000022120011400002222010200000020000002000110

[back to top]

Summary of 28-day Seizure Frequency for Sum of Individual Seizures and Clusters Through 52-week OLE (Median Percent Change)

Percentage change from baseline in 28-day seizure frequency at 3 months (day 91), 26 weeks, 52 week OLE (Median Percent Change) (NCT02358538)
Timeframe: Baseline through 52-week open- label period

,,,
InterventionMedian Percent Change in Frequency (Median)
Percent change from baseline (at Day 91)Percent change from baseline (at week 26)Percent change from baseline (at week 26) - with 6 duplicate diary entries removedPercent change from baseline (52 week OLE through month 6)Percent change from baseline (52 week OLE period)
CDKL5-47.34-37.70-44.4-58.94-61.93
CSWSNANANANANA
Lennox-Gastaut-10.22-9.19NA-38.74-37.75
PCDH19-25.98-24.59NA-13.48-13.48

[back to top]

Summary of 28-day Seizure Frequency for Sum of Individual Seizures and Clusters for 52-week OLE Period (Mean Percent Change & Standard Deviation)

Percentage change from baseline in 28-day seizure frequency at 3 months (day 91), 26 weeks, 52 week OLE (Mean Percent Change & Standard Deviation) (NCT02358538)
Timeframe: Baseline through 52 week open label period

,,,
Interventionpercentage of change of frequency (Mean)
Percent Change from baseline (at day 91)Percent Change from baseline (at week 26)Percent change from baseline (52 week OLE through month 6)Percent change from baseline (52 week OLE period)
CDKL5-31.23-20.55-54.41-49.20
CSWSNANANANA
Lennox- Gastaut122.10125.38-38.74-37.75
PCDH1952.8346.36-19.98-19.95

[back to top]

Number of Participants With Responder Rate of Seizure Frequency

Responder Rate in Terms of 28-day Seizure Frequency Based on the Sum of Individual Seizures and Clusters (NCT02358538)
Timeframe: Month 3 and Week 26

,,,
InterventionParticipants (Count of Participants)
Post-baseline through Month 3 (Day 91) - 25% ResponderPost-baseline through Month 3 (Day 91) - 50% ResponderPost-baseline through Month 3 (Day 91) - 75% ResponderPost-baseline 26-week open-label Period - 25% ResponderPost-baseline 26-week open-label Period - 50% ResponderPost-baseline 26-week open-label Period - 75% Responder
CDKL5431421
CSWSNANANANANANA
Lennox-Gastaut420310
PCDH19641531

[back to top]

Mean Percentage Change of Individual Seizure-free Days

Mean Percentage Change of Individual Seizure-free days per 28-day period (through 52-week OLE) period relative to baseline (NCT02358538)
Timeframe: Baseline, Day 91, Week 26, 52-week OLE through month 6, 52-week OLE Period

,,,
InterventionPercentage Change (Mean)
Mean Percentage Change of individual seizure-free days from baseline to Day 91Mean Percentage Change of individual seizure-free days from baseline to Week 26Mean Percentage Change of individual seizure-free days from baseline to 52-week OLE (181 days)Mean Percentage Change of individual seizure-free days from baseline to 52-week OLE Period
CDKL511.8411.802120.44
CSWSNANANANA
Lennox-Gastaut-1.12-2.1316.3514.95
PCDH197.877.9417.1217.02

[back to top]

Number of Participants Who Showed Greater Than or Equal to 50% Reduction in 28-day Seizure Frequent From Baseline

A 50% responder is an individual whose reduction of percent change from Baseline to the end of the open label extension period in 28-day partial-onset seizure (POS) seizure frequency is greater than or equal to 50%. (NCT02519439)
Timeframe: Baseline and at Day 28

InterventionParticipants (Count of Participants)
Ganaxolone14

[back to top]

Percent Change From Baseline in 28-day Seizure Frequency

Baseline 28-day seizure frequency was calculated as the number of seizures in the Baseline period of Study 1042-0603 (less than or equal to 56 days) divided by the number of days with available seizure data in the Baseline period and multiplied by 28. Post-baseline 28-day seizure frequency was calculated as the number of seizures in the entire treatment period divided by the number of days with available seizure data in the treatment period and multiplied by 28. Baseline was defined as the last non-missing value obtained before the first treatment in the preceding Study 1042-0603. The calculation for percent change from Baseline in 28-day seizure frequency was done as follows for each participant: post-Baseline 28-day seizure frequency minus Baseline 28-day seizure frequency whole divided by Baseline 28-day seizure frequency multiplied by 100 percent. (NCT02519439)
Timeframe: Baseline and at Day 28

InterventionPercent change (Mean)
Ganaxolone-41.86

[back to top]

Number of Participants With Patient/Caregiver Global Impression of Improvement (PGI-I) Scores

"The participant is asked to rate the total improvement of their partial-onset seizures whether or not in the participant's judgment it is due entirely to drug treatment based on a 7-point scale using the markers very much improved, much improved, slightly improved, no change, slightly worse, much worse, or very much worse (1 = very much improved; 7 = very much worse). Higher scores indicated worse condition. Participants who showed PGI improvement at Week 104 (End of treatment) has been presented." (NCT02519439)
Timeframe: At Week 104

InterventionParticipants (Count of Participants)
Very much improvedMuch improvedMinimally improvedNo changeMinimally worseMuch worseVery much worse
Ganaxolone48101000

[back to top]

Number of Participants With Clinical Global Impression of Improvement (CGI-I) Scores

"The CGI-I scale is a clinician-rated 7-point scale used to assess how much the participant's illness had improved or worsened relative to a Baseline state at the beginning of the intervention. It was rated as: 1. very much improved 2. much improved 3. minimally improved 4. no change 5. minimally worse 6. much worse 7. very much worse. Higher scores indicated worse condition. Participants who showed CGI improvement at Week 104 (End of treatment) has been presented." (NCT02519439)
Timeframe: At Week 104

InterventionParticipants (Count of Participants)
Very much improvedMuch improvedMinimally improvedNo changeMinimally worseMuch worseVery much worse
Ganaxolone19112000

[back to top]

Montgomery-Asberg Depression Rating Scale (MADRS)

The MADRS is a diagnostic questionnaire that is used to measure the severity of depressive episodes in patients with mood disorders. The minimum and maximum values are 0 and 60, respectively, (higher scores are more severe). (NCT02900092)
Timeframe: Week 8

Interventionunits on a scale (Mean)
Ganaxolone Treatment Arm12.8

[back to top]

Change From Baseline in Spielberger State-Trait Anxiety Inventory 6-item Version (STAI6) Total Score

"The STAI6 is a 6-item self-rated instrument used to assess anxiety state. The STAI6 questions included: 1. I feel calm; 2. I am tense; 3. I feel upset; 4. I am relaxed; 5. I feel content; 6. I am worried. Each of these questions were rated as: a) not at all; b) somewhat; c) moderately; d) very much. For questions 2, 3, and 6, the scoring was a = 1, b = 2, c = 3, and d = 4. For the other 3 questions, the scoring was a = 4, b = 3, c = 2, and d = 1. The STAI6 score was the result of first totaling the 6 individual item scores, and then prorating by a multiplication factor of 20/6 to acquire a score range of 20 (low anxiety) to 80 (high anxiety). Higher score indicated higher level of anxiety. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value." (NCT03228394)
Timeframe: Baseline, Day 1 (12 hours post start of infusion), Day 2, Day 3 post-infusion (60 hours post start of infusion), Day 4, Day 8, Day 11, Day 15, Day 22, Day 29, Day 34, Day 36, Day 57 and Day 71

,
InterventionScores on a scale (Mean)
Day 1 (12 hours post start of infusion)Day 2Day 8Day 15Day 22Day 29Day 36Day 57Day 71
Ganaxolone Cohort 6-17.7-19.0-15.6-17.4-21.8-18.9-21.7-13.3-21.0
Placebo Cohort 6-9.4-12.9-10.2-12.3-15.9-20.8-21.9-21.7-31.7

[back to top]

Change From Baseline in Spielberger State-Trait Anxiety Inventory 6-item Version (STAI6) Total Score

"The STAI6 is a 6-item self-rated instrument used to assess anxiety state. The STAI6 questions included: 1. I feel calm; 2. I am tense; 3. I feel upset; 4. I am relaxed; 5. I feel content; 6. I am worried. Each of these questions were rated as: a) not at all; b) somewhat; c) moderately; d) very much. For questions 2, 3, and 6, the scoring was a = 1, b = 2, c = 3, and d = 4. For the other 3 questions, the scoring was a = 4, b = 3, c = 2, and d = 1. The STAI6 score was the result of first totaling the 6 individual item scores, and then prorating by a multiplication factor of 20/6 to acquire a score range of 20 (low anxiety) to 80 (high anxiety). Higher score indicated higher level of anxiety. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value." (NCT03228394)
Timeframe: Baseline, Day 1 (12 hours post start of infusion), Day 2, Day 3 post-infusion (60 hours post start of infusion), Day 4, Day 8, Day 11, Day 15, Day 22, Day 29, Day 34, Day 36, Day 57 and Day 71

,,,,,
InterventionScores on a scale (Mean)
Day 1 (12 hours post start of infusion)Day 2Day 3 post-infusion (60 hours post start of infusion)Day 4Day 11Day 34
Ganaxolone Cohort 1-4.2-8.7-22.0-20.7-18.7-16.0
Ganaxolone Cohort 2-12.01-14.2-21.0-22.9-20.8-24.4
Ganaxolone Cohort 3-11.3-12.0-24.0-22.3-16.0-17.7
Placebo Cohort 3-10.7-12.7-24.3-25.0-17.0-18.7
Placebo Cohort 1-8.9-9.2-11.7-20.0-21.7-20.8
Placebo Cohort 2-14.5-15.0-18.2-21.0-16.9-16.1

[back to top]

Number of Participants With HAMD17 Remission

HAMD17 Remission was defined as total score ≤7. The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Number of participants with HAMD17 remission is presented (NCT03228394)
Timeframe: Day1 post-infusion (6 hours post start of infusion [SOI]), Day 1 (12 hours post SOI), Day2 (24 hours post SOI), Day 3 post-infusion (48 hours post SOI), Day 3 post-infusion (60 hours post SOI), Day 4 (72 hours post SOI), Days 8,11,15,22,29,34,57 and 71

,,,,,
InterventionParticipants (Count of Participants)
Day 1 (12 hours post start of infusion): RemissionDay 2 (24 hours post start of infusion): RemissionDay 3 post-infusion (48 hours post start of infusion): RemissionDay 3 post-infusion (60 hours post start of infusion): RemissionDay 4 (72 hours post start of infusion): RemissionDay 11: RemissionDay 34: Remission
Ganaxolone Cohort 10001101
Ganaxolone Cohort 20134755
Ganaxolone Cohort 30022314
Placebo Cohort 10011213
Placebo Cohort 20034434
Placebo Cohort 30112322

[back to top]

Change From Baseline in HAMD17 Total Score at Indicated Time Points

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT03228394)
Timeframe: Baseline, Day1 post-infusion(6hours post start of infusion), Day1(12hours post start of infusion), Day2(24hours post start of infusion),Day3 post-infusion(48hours post start of infusion),Day4(72hours post start of infusion),Days 8,11,15,22,36,57 and 71

,,,,,
InterventionScores on a scale (Mean)
Day 1 (12 hours post start of infusion)Day 2 (24 hours post start of infusion)Day 3 post-infusion (48 hours post start of infusion)Day 4 (72 hours post start of infusion)Day 11Day 36
Ganaxolone Cohort 1-2.4-3.8-11.0-10.2-6.2-11.0
Ganaxolone Cohort 2-2.7-10.1-13.7-17.3-14.0-16.5
Ganaxolone Cohort 30.3-3.4-11.2-10.5-8.9-11.1
Placebo Cohort 1-3.8-8.0-10.8-12.8-13.3-17.0
Placebo Cohort 2-3.2-6.4-10.5-14.5-9.7-13.3
Placebo Cohort 3-1.9-5.2-8.7-14.0-8.2-9.1

[back to top]

Change From Baseline in Hamilton Depression Rating Scale 17-item Version (HAMD17) Total Score

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT03228394)
Timeframe: Baseline and Day 3 post-infusion (60 hours post start of infusion) for Cohorts 1 to 3 and Baseline and Day 29 for Cohort 6

,,,,,
InterventionScores on a scale (Mean)
Day 3 post-infusion (60 hours post start of infusion)
Ganaxolone Cohort 1-12.0
Ganaxolone Cohort 2-14.3
Ganaxolone Cohort 3-11.3
Placebo Cohort 1-13.0
Placebo Cohort 2-14.0
Placebo Cohort 3-11.0

[back to top]

Change From Baseline in Hamilton Depression Rating Scale 17-item Version (HAMD17) Total Score

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT03228394)
Timeframe: Baseline and Day 3 post-infusion (60 hours post start of infusion) for Cohorts 1 to 3 and Baseline and Day 29 for Cohort 6

,
InterventionScores on a scale (Mean)
Day 29
Ganaxolone Cohort 6-11.5
Placebo Cohort 6-13.6

[back to top]

Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score

The EPDS is a validated 10-question depression assessment for postpartum women, which includes anxiety symptoms and excludes constitutional symptoms associated with depression common in the postpartum period, such as changes in sleeping patterns. It consists of 10 multiple choice questions with scores for each question ranging from 0-3. Questions 1, 2, & 4 are scored 0, 1, 2 or 3 with top box scored as 0 and the bottom box scored as 3. Questions 3 and 5 to 10 are reverse scored, with the top box scored as a 3 and the bottom box scored as 0. Scores are then summed for a total score, which ranges from 0: no depression to 30: severe depression. Higher score indicates severe depression. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT03228394)
Timeframe: Baseline, Day 1, Day 2, Day 3 post-infusion (60 hours post start of infusion), Day 8, Day 11, Day 15, Day 22, Day 29, Day 34, Day 36, Day 57 and Day 71

,,,,,
InterventionScores on a scale (Mean)
Day 3 post-infusion (60 hours post start of infusion)Day 11Day 34
Ganaxolone Cohort 1-8.0-8.0-8.6
Ganaxolone Cohort 2-10.8-11.5-13.4
Ganaxolone Cohort 3-10.9-8.2-8.8
Placebo Cohort 1-8.8-13.5-14.5
Placebo Cohort 2-10.8-9.6-9.8
Placebo Cohort 3-12.2-8.8-9.1

[back to top]

Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score

The EPDS is a validated 10-question depression assessment for postpartum women, which includes anxiety symptoms and excludes constitutional symptoms associated with depression common in the postpartum period, such as changes in sleeping patterns. It consists of 10 multiple choice questions with scores for each question ranging from 0-3. Questions 1, 2, & 4 are scored 0, 1, 2 or 3 with top box scored as 0 and the bottom box scored as 3. Questions 3 and 5 to 10 are reverse scored, with the top box scored as a 3 and the bottom box scored as 0. Scores are then summed for a total score, which ranges from 0: no depression to 30: severe depression. Higher score indicates severe depression. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT03228394)
Timeframe: Baseline, Day 1, Day 2, Day 3 post-infusion (60 hours post start of infusion), Day 8, Day 11, Day 15, Day 22, Day 29, Day 34, Day 36, Day 57 and Day 71

,
InterventionScores on a scale (Mean)
Day 1Day 2Day 8Day 15Day 22Day 29Day 36Day 57Day 71
Ganaxolone Cohort 6-5.4-7.5-9.6-11.3-11.2-10.0-12.4-9.4-11.5
Placebo Cohort 6-1.9-4.9-6.7-8.0-8.2-10.9-11.8-11.8-14.0

[back to top]

Change From Baseline in HAMD17 Total Score at Indicated Time Points

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline was calculated by subtracting Baseline value from the post-dose visit value. (NCT03228394)
Timeframe: Baseline, Day1 post-infusion(6hours post start of infusion), Day1(12hours post start of infusion), Day2(24hours post start of infusion),Day3 post-infusion(48hours post start of infusion),Day4(72hours post start of infusion),Days 8,11,15,22,36,57 and 71

,
InterventionScores on a scale (Mean)
Day 1 post-infusion (6 hours post start of infusion)Day 2 (24 hours post start of infusion)Day 4 (72 hours post start of infusion)Day 8Day 11Day 15Day 22Day 36Day 57Day 71
Ganaxolone Cohort 6-6.1-7.8-7.0-8.7-13.6-12.7-12.7-13.0-12.0-14.8
Placebo Cohort 6-3.2-5.1-7.8-8.7-13.6-11.6-12.3-13.5-16.0-19.0

[back to top]

Number of Participants With HAMD17 Remission

HAMD17 Remission was defined as total score ≤7. The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Number of participants with HAMD17 remission is presented (NCT03228394)
Timeframe: Day1 post-infusion (6 hours post start of infusion [SOI]), Day 1 (12 hours post SOI), Day2 (24 hours post SOI), Day 3 post-infusion (48 hours post SOI), Day 3 post-infusion (60 hours post SOI), Day 4 (72 hours post SOI), Days 8,11,15,22,29,34,57 and 71

,
InterventionParticipants (Count of Participants)
Day 1 post-infusion (6 hours post start of infusion): RemissionDay 2 (24 hours post start of infusion): RemissionDay 4 (72 hours post start of infusion): RemissionDay 8: RemissionDay 11: RemissionDay 15: RemissionDay 22: RemissionDay 29: RemissionDay 34: RemissionDay 57: RemissionDay 71: Remission
Ganaxolone Cohort 631033354446
Placebo Cohort 600224446567

[back to top]

Number of Participants With HAMD17 Response

HAMD17 Response was defined as ≥50% reduction from Baseline in total score. The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Number of participants with HAMD17 response is presented. (NCT03228394)
Timeframe: Day1 post-infusion (6 hours post start of infusion [SOI]), Day 1 (12 hours post SOI), Day 2 (24 hours post SOI),Day 3 post-infusion (48 hours post SOI), Day 3 post-infusion (60 hours post SOI), Day 4 (72 hours post SOI), Days 8,11,15,22,29,36,57 and 71

,
InterventionParticipants (Count of Participants)
Day 1 post-infusion (6 hours post start of infusion): ResponderDay 2 (24 hours post start of infusion): ResponderDay 4 (72 hours post start of infusion): ResponderDay 8: ResponderDay 11: ResponderDay 15: ResponderDay 22: ResponderDay 29: ResponderDay 36: ResponderDay 57: ResponderDay 71: Responder
Ganaxolone Cohort 656356687869
Placebo Cohort 621346878689

[back to top]

Number of Participants With HAMD17 Response

HAMD17 Response was defined as ≥50% reduction from Baseline in total score. The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Number of participants with HAMD17 response is presented. (NCT03228394)
Timeframe: Day1 post-infusion (6 hours post start of infusion [SOI]), Day 1 (12 hours post SOI), Day 2 (24 hours post SOI),Day 3 post-infusion (48 hours post SOI), Day 3 post-infusion (60 hours post SOI), Day 4 (72 hours post SOI), Days 8,11,15,22,29,36,57 and 71

,,,,,
InterventionParticipants (Count of Participants)
Day 1 (12 hours post start of infusion): ResponderDay 2 (24 hours post start of infusion): ResponderDay 3 post-infusion (48 hours post start of infusion): ResponderDay 3 post-infusion (60 hours post start of infusion): ResponderDay 4 (72 hours post start of infusion): ResponderDay 11: ResponderDay 36: Responder
Ganaxolone Cohort 10012313
Ganaxolone Cohort 204771157
Ganaxolone Cohort 30045535
Placebo Cohort 10122223
Placebo Cohort 21467755
Placebo Cohort 30243634

[back to top]

Number of Participants With Response to Clinical Global Impression-Improvement (CGI-I) Scale

"The CGI-I scale is a clinician-rated 7-point scale used to assess how much the participant's illness had improved or worsened relative to a Baseline state at the beginning of the intervention. It was rated as: 1. very much improved; 2. much improved; 3. minimally improved; 4. no change; 5. minimally worse; 6. much worse; 7. very much worse. Higher scores indicated worse condition. Response was defined as >50% decrease from Baseline in CGI-I total score." (NCT03228394)
Timeframe: Day 1 (12 hours post start of infusion), Day 2, Day 3 post-infusion (60 hours post start of infusion), Day 4, Day 8, Day 11, Day 15, Day 22, Day 29, Day 34, Day 36, Day 57 and Day 71

,,,,,
InterventionParticipants (Count of Participants)
Day 1 (12 hours post start of infusion): Very much or much improvedDay 1 (12 hours post start of infusion): Minimally or not improvedDay 2: Very much or much improvedDay 2: Minimally or not improvedDay 3 post-infusion (60 hours post start of infusion): Very much or much improvedDay 3 post-infusion (60 hours post start of infusion): Minimally or not improvedDay 4: Very much or much improvedDay 4: Minimally or not improvedDay 11: Very much or much improvedDay 11: Minimally or not improvedDay 34: Very much or much improvedDay 34: Minimally or not improved
Ganaxolone Cohort 1051441320532
Ganaxolone Cohort 231287941239484
Ganaxolone Cohort 30103773735555
Placebo Cohort 1041331312231
Placebo Cohort 221231076677557
Placebo Cohort 3191955554655

[back to top]

Number of Participants With Response to Clinical Global Impression-Improvement (CGI-I) Scale

"The CGI-I scale is a clinician-rated 7-point scale used to assess how much the participant's illness had improved or worsened relative to a Baseline state at the beginning of the intervention. It was rated as: 1. very much improved; 2. much improved; 3. minimally improved; 4. no change; 5. minimally worse; 6. much worse; 7. very much worse. Higher scores indicated worse condition. Response was defined as >50% decrease from Baseline in CGI-I total score." (NCT03228394)
Timeframe: Day 1 (12 hours post start of infusion), Day 2, Day 3 post-infusion (60 hours post start of infusion), Day 4, Day 8, Day 11, Day 15, Day 22, Day 29, Day 34, Day 36, Day 57 and Day 71

,
InterventionParticipants (Count of Participants)
Day 1 (12 hours post start of infusion): Very much or much improvedDay 1 (12 hours post start of infusion): Minimally or not improvedDay 2: Very much or much improvedDay 2: Minimally or not improvedDay 8: Very much or much improvedDay 8: Minimally or not improvedDay 15: Very much or much improvedDay 15: Minimally or not improvedDay 22: Very much or much improvedDay 22: Minimally or not improvedDay 29: Very much or much improvedDay 29: Minimally or not improvedDay 36: Very much or much improvedDay 36: Minimally or not improvedDay 57: Very much or much improvedDay 57: Minimally or not improvedDay 71: Very much or much improvedDay 71: Minimally or not improved
Ganaxolone Cohort 679979613210512411384103
Placebo Cohort 63143146961087610576675

[back to top]

Seizure Burden

Seizure Burden (%) Baseline and Percentage Change from Baseline by Time Point (NCT03350035)
Timeframe: Baseline (Pre-dose) to <-24hrs (Post Dose)

,,
InterventionPercentage change from baseline (Mean)
Change from Baseline at 0 to <- 1 Hour Post-DoseChange from Baseline at 0 to <- 4 Hour Post-DoseChange from Baseline at >4 to <- 8 Hours Post-DoseChange from Baseline at >8 to <- 16 Hours Post-DoseChange from Baseline at >16 to <- 24 Hours Post-DoseChange from Baseline at 0 to <- 24 Hours Post-Dose
High - GNX-84.01-87.99-96.58-93.75-81.16-89.06
Low - GNX-94.80-98.59-77.53-47.98-99.53-78.64
Medium - GNX-74.63-71.75-59.80-53.29-48.25-56.10

[back to top]

Number of Participants With No SE Recurrence Per Principal Investigator

Number of participants with No SE Recurrence per Principal Investigator within 24hrs of starting treatment, during treatment period (excluding taper), during taper, during 24-hr follow-up period, and during follow-up period. (NCT03350035)
Timeframe: Baseline (within 24hrs of treatment) through follow up period, up to approximately 4 weeks.

,,
InterventionParticipants (Count of Participants)
Within 24hrs of starting treatmentDuring treatment period (excluding taper)During taperDuring 24-hr follow-up periodDuring follow-up period
High - GNX77787
Low - GNX53331
Medium - GNX44233

[back to top]

Time to Cessation of SE

Summary of Time to SE Cessation (NCT03350035)
Timeframe: Time to SE Cessation, assessed up to 24 hours

InterventionMinutes (Median)
Low - GNX5.000
Medium - GNX5.580
High - GNX10.16

[back to top]

Number of Participants Who Required No Escalation of Treatment for Ongoing or Recurrent SE

Number of participants who Required No Escalation of Treatment for Ongoing or Recurrent SE (NCT03350035)
Timeframe: Drug initiation through follow-up period, up to approximately 4 weeks

InterventionParticipants (Count of Participants)
Low - GNX3
Medium - GNX3
High - GNX8

[back to top]

Number of Participants Who Did Not Require an IV Anesthetic Drug for SE Treatment

Number of participants who did not require an intravenous (IV) Anesthetic Drug (a third-line Treatment) for Status Epilepticus (SE) within the first 24 hours after Study Drug Initiation. (NCT03350035)
Timeframe: 24 hours post study drug initiation

InterventionParticipants (Count of Participants)
Low - GNX5
Medium - GNX4
High - GNX8

[back to top]

Change From Baseline in Spielberger State-Trait Anxiety Inventory, 6-item Version

"The Spielberger State-Trait Anxiety Inventory was a 6-item questionnaire designed and validated. It is a reliable and sensitive measure of anxiety. The 6-item version has been shown to have acceptable reliability, producing similar scores as the full 20-item inventory for those with normal and raised levels of anxiety. Participants are prompted to respond to indicate how [the participant] feel right now, at this moment, followed by a list of common experiences like I feel calm, I am tense, I feel upset, I am relaxed I feel content and I am worried. Responses range from Not at all (1) to Very much (4), with overall scores ranging from 20-80, where higher scores reflected higher states of anxiety. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value." (NCT03460756)
Timeframe: Baseline (Day 1) through Day 119

,
InterventionScores on a scale (Mean)
Day 2/4Day 7Day 8/10Day 15/17Day 36/38
QHS (2-week)-6.4-9.0-5.7-14.8-11.3
TID (2-week)-11.7-11.7-20.0-33.3-31.7

[back to top]

Change From Baseline in Hamilton Depression Rating Scale 17-item Version Post Treatment

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value. (NCT03460756)
Timeframe: Baseline (Day 1) and Through Day 38 for 2 week treatment group and Day 71 and 119 for 4 week treatment groups

Interventionscore on a scale (Mean)
Day 36/38Day 57/59Day 71
1125 mg (4-week)-12.4-12.3-12.2

[back to top]

Change From Baseline in Hamilton Depression Rating Scale 17-item Version Post Treatment

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value. (NCT03460756)
Timeframe: Baseline (Day 1) and Through Day 38 for 2 week treatment group and Day 71 and 119 for 4 week treatment groups

,
Interventionscore on a scale (Mean)
Day 36/38
QHS (2-week)-7.1
TID (2-week)-19.0

[back to top]

Change From Baseline in Hamilton Depression Rating Scale 17-item Version (HAMD17) Through Treatment

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value. (NCT03460756)
Timeframe: Baseline (Day 1) and through Day 10 for 2 week treatment group and Day 29 for 4 week treatment groups

,
Interventionscore on a scale (Mean)
Day 2/4Day 8/10Day 15/17Day 22Day 29
QHS (4-week)-0.8-6.8-9.7-10.3-12.2
1125 mg (4-week)-2.6-10.2-9.4-12.0-14.0

[back to top]

Change From Baseline in Hamilton Depression Rating Scale 17-item Version (HAMD17) Through Treatment

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value. (NCT03460756)
Timeframe: Baseline (Day 1) and through Day 10 for 2 week treatment group and Day 29 for 4 week treatment groups

,
Interventionscore on a scale (Mean)
Day 2/4Day 7Day 8/10Day 15/17
QHS (2-week)-5.7-6.7-10.5-8.8
TID (2-week)-8.0-15.0-3.0-9.5

[back to top]

Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score

The EPDS is a validated 10-question depression assessment for postpartum women, which includes anxiety symptoms and excludes constitutional symptoms associated with depression common in the postpartum period, such as changes in sleeping patterns. It consists of 10 multiple choice questions with scores for each question ranging from 0-3. Questions 1, 2, & 4 are scored 0, 1, 2 or 3 with top box scored as 0 and the bottom box scored as 3. Questions 3 and 5 to 10 are reverse scored, with the top box scored as a 3 and the bottom box scored as 0. Scores are then summed for a total score, which ranges from 0: no depression to 30: severe depression. Higher score indicates severe depression. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value. (NCT03460756)
Timeframe: Baseline (Day 1) through Day 119

InterventionScore on a scale (Mean)
Day 2/4Day 8/10Day 15/17Day 22Day 29Day 36/38Day 57/59Day 89Day 119
QHS (4-week)-0.8-1.5-1.8-2.7-2.9-3.1-2.5-2.6-3.3

[back to top]

Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score

The EPDS is a validated 10-question depression assessment for postpartum women, which includes anxiety symptoms and excludes constitutional symptoms associated with depression common in the postpartum period, such as changes in sleeping patterns. It consists of 10 multiple choice questions with scores for each question ranging from 0-3. Questions 1, 2, & 4 are scored 0, 1, 2 or 3 with top box scored as 0 and the bottom box scored as 3. Questions 3 and 5 to 10 are reverse scored, with the top box scored as a 3 and the bottom box scored as 0. Scores are then summed for a total score, which ranges from 0: no depression to 30: severe depression. Higher score indicates severe depression. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value. (NCT03460756)
Timeframe: Baseline (Day 1) through Day 119

,
InterventionScore on a scale (Mean)
Day 2/4Day 7Day 8/10Day 15/17Day 36/38
QHS (2-week)-1.4-2.3-1.3-2.6-2.6
TID (2-week)-0.5-2.0-3.0-5.0-2.0

[back to top]

Change From Baseline in Hamilton Depression Rating Scale 17-item Version Post Treatment

The HAMD17 was a 17-item clinician-rated instrument used to assess the range of symptoms that were most frequently observed in participants with major depression. All items were scored on an ordinal scale between 0 and 4 (8 items) or 0 and 2 (9 items) of increasing severity. Each of 17 items was rated by clinician with rating of 0: absent, 1: doubtful to mild, 2: mild to moderate, 3: moderate to severe, and 4: very severe. A total score (0 to 52) was calculated by adding scores of all 17 items, where 0 indicated no depression and 52 indicated severe depression. Higher score represented more severe condition. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value. (NCT03460756)
Timeframe: Baseline (Day 1) and Through Day 38 for 2 week treatment group and Day 71 and 119 for 4 week treatment groups

Interventionscore on a scale (Mean)
Day 36/38Day 57/59Day 89Day 119
QHS (4-week)-14.6-13.2-14.2-20.5

[back to top]

Number of Participants With Clinical Global Impression-Improvement

"The CGI-I scale is a clinician-rated 7-point scale used to assess how much the participant's illness had improved or worsened relative to a Baseline state at the beginning of the intervention. It was rated as: 1. very much improved 2. much improved 3. minimally improved 4. no change 5. minimally worse 6. much worse 7. very much worse. Higher scores indicated worse condition. Only those participants with CGI- improvement have been presented." (NCT03460756)
Timeframe: Baseline through Day 119

InterventionParticipants (Count of Participants)
Day 2/4 - Much ImprovedDay 2/4 - Minimally ImprovedDay 8/10 - Much ImprovedDay 8/10 - Minimally ImprovedDay 15/17 - Very much ImprovedDay 15/17 - Much ImprovedDay 15/17 - Minimally ImprovedDay 22 - Very much ImprovedDay 22 - Much ImprovedDay 22 - Minimally ImprovedDay 29 - Very much ImprovedDay 29 - Much ImprovedDay 29 - Minimally ImprovedDay 36/38 - Very much ImprovedDay 36/38 - Much ImprovedDay 36/38 - Minimally ImprovedDay 57/59 - Very Much ImprovedDay 57/59 - Much ImprovedDay 57/59 - Minimally ImprovedDay 89 - Very much ImprovedDay 89 - Much ImprovedDay 89 - Minimally ImprovedDay 119 - Very Much ImprovedDay 119 - Much ImprovedDay 119 - Minimally Improved
QHS (4-week)1478195196478847492646533

[back to top]

Change From Baseline in Edinburgh Postnatal Depression Scale (EPDS) Total Score

The EPDS is a validated 10-question depression assessment for postpartum women, which includes anxiety symptoms and excludes constitutional symptoms associated with depression common in the postpartum period, such as changes in sleeping patterns. It consists of 10 multiple choice questions with scores for each question ranging from 0-3. Questions 1, 2, & 4 are scored 0, 1, 2 or 3 with top box scored as 0 and the bottom box scored as 3. Questions 3 and 5 to 10 are reverse scored, with the top box scored as a 3 and the bottom box scored as 0. Scores are then summed for a total score, which ranges from 0: no depression to 30: severe depression. Higher score indicates severe depression. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value. (NCT03460756)
Timeframe: Baseline (Day 1) through Day 119

InterventionScore on a scale (Mean)
Day 2/4Day 8/10Day 15/17Day 22Day 29Day 36/38Day 57/59Day 71
1125 mg (4-week)-0.6-2.3-2.2-3.0-3.1-3.7-3.4-3.3

[back to top]

Number of Participants With Clinical Global Impression-Improvement

"The CGI-I scale is a clinician-rated 7-point scale used to assess how much the participant's illness had improved or worsened relative to a Baseline state at the beginning of the intervention. It was rated as: 1. very much improved 2. much improved 3. minimally improved 4. no change 5. minimally worse 6. much worse 7. very much worse. Higher scores indicated worse condition. Only those participants with CGI- improvement have been presented." (NCT03460756)
Timeframe: Baseline through Day 119

InterventionParticipants (Count of Participants)
Day 2/4 - Much ImprovedDay 2/4 - Minimally ImprovedDay 8/10 - Very much ImprovedDay 8/10 - Much ImprovedDay 8/10 - Minimally ImprovedDay 15/17 - Very much ImprovedDay 15/17 - Much ImprovedDay 15/17 - Minimally ImprovedDay 22 - Very much ImprovedDay 22 - Much ImprovedDay 22 - Minimally ImprovedDay 29 - Very much ImprovedDay 29 - Much ImprovedDay 29 - Minimally ImprovedDay 36/38 - Very much ImprovedDay 36/38 - Much ImprovedDay 36/38 - Minimally ImprovedDay 57/59 - Very Much ImprovedDay 57/59 - Much ImprovedDay 57/59 - Minimally ImprovedDay 71 - Very Much ImprovedDay 71 - Much ImprovedDay 71 - Minimally Improved
1125 mg (4-week)41531411615971661116512101091478146

[back to top]

Number of Participants With Clinical Global Impression-Improvement

"The CGI-I scale is a clinician-rated 7-point scale used to assess how much the participant's illness had improved or worsened relative to a Baseline state at the beginning of the intervention. It was rated as: 1. very much improved 2. much improved 3. minimally improved 4. no change 5. minimally worse 6. much worse 7. very much worse. Higher scores indicated worse condition. Only those participants with CGI- improvement have been presented." (NCT03460756)
Timeframe: Baseline through Day 119

,
InterventionParticipants (Count of Participants)
Day 2/4 - Much ImprovedDay 2/4 - Minimally ImprovedDay 7 - Very much ImprovedDay 7 - Much ImprovedDay 7 - Minimally ImprovedDay 8/10 - Very much ImprovedDay 8/10 - Much ImprovedDay 8/10 - Minimally ImprovedDay 15/17 - Very much ImprovedDay 15/17 - Much ImprovedDay 15/17 - Minimally ImprovedDay 36/38 - Very much ImprovedDay 36/38 - Much ImprovedDay 36/38 - Minimally Improved
QHS (2-week)16152252253131
TID (2-week)01011010020020

[back to top]

Change From Baseline in Spielberger State-Trait Anxiety Inventory, 6-item Version

"The Spielberger State-Trait Anxiety Inventory was a 6-item questionnaire designed and validated. It is a reliable and sensitive measure of anxiety. The 6-item version has been shown to have acceptable reliability, producing similar scores as the full 20-item inventory for those with normal and raised levels of anxiety. Participants are prompted to respond to indicate how [the participant] feel right now, at this moment, followed by a list of common experiences like I feel calm, I am tense, I feel upset, I am relaxed I feel content and I am worried. Responses range from Not at all (1) to Very much (4), with overall scores ranging from 20-80, where higher scores reflected higher states of anxiety. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value." (NCT03460756)
Timeframe: Baseline (Day 1) through Day 119

InterventionScores on a scale (Mean)
Day 2/4Day 8/10Day 15/17Day 22Day 29Day 36/38Day 57/59Day 89Day 119
QHS (4-week)-1.8-11.8-14.5-13.0-17.13-20.5-16.3-14.7-16.7

[back to top]

Change From Baseline in Spielberger State-Trait Anxiety Inventory, 6-item Version

"The Spielberger State-Trait Anxiety Inventory was a 6-item questionnaire designed and validated. It is a reliable and sensitive measure of anxiety. The 6-item version has been shown to have acceptable reliability, producing similar scores as the full 20-item inventory for those with normal and raised levels of anxiety. Participants are prompted to respond to indicate how [the participant] feel right now, at this moment, followed by a list of common experiences like I feel calm, I am tense, I feel upset, I am relaxed I feel content and I am worried. Responses range from Not at all (1) to Very much (4), with overall scores ranging from 20-80, where higher scores reflected higher states of anxiety. Baseline was defined as the Day 1 assessment before study drug infusion. Change from Baseline is defined as post dose visit value minus Baseline value." (NCT03460756)
Timeframe: Baseline (Day 1) through Day 119

InterventionScores on a scale (Mean)
Day 2/4Day 8/10Day 15/17Day 22Day 29Day 36/38Day 57/59Day 71
1125 mg (4-week)-9.9-16.8-15.2-21.8-21.4-23.2-22.8-19.2

[back to top]

Number of Participants With Response to Hamilton Depression Rating Scale 17 Item (HAMD17)

HAMD17 Response was defined as ≥50% reduction from Baseline in total score. The HAMD-17 is a 17-item assessment used to assess the severity of depression and its improvement during the course of therapy. Each item was evaluated and scored using either a 5-point scale of 0 (not present/absent) to 4 (very severe) or a 3-point scale of 0 (not present/absent) to 2 (marked). Items rated on the 3-point scale include: insomnia early, middle, late; somatic symptoms gastrointestinal and general; genital symptoms; loss of weight; insight. Items rated on 5-point scale include: depressed mood; feelings of guilt; suicide; work and activities; retardation; agitation; anxiety psychic and somatic; hypochondriasis. The total score was the sum of the scores from HAMD-17 Items 1 through 17 and ranged from 0 (not at all depressed) to 52 (severely depressed). Higher scores indicate greater symptom severity. Number of participants with response to HAMD-17 has been presented. (NCT03460756)
Timeframe: Day 2 (all treatment groups), Day 7 (TID & QHS), Day 8/10 (all treatment groups), Day 15/17 (all treatment groups), Day 22 and 29 (QHS, 1125mg), Day 36/38 (all treatment groups), Day 57/59 (QHS, 1125mg), Day 71 (1125mg), Day 89 (QHS), Day 119 (QHS)

InterventionParticipants (Count of Participants)
Day 2/4 - ResponseDay 8/10 - ResponseDay 15/17 - ResponseDay 22 - ResponseDay 29 - ResponseDay 36/38 - ResponseDay 57/59 - ResponseDay 71 - Response
1125 mg (4-week)312162022171813

[back to top]

Number of Participants With Response to Hamilton Depression Rating Scale 17 Item (HAMD17)

HAMD17 Response was defined as ≥50% reduction from Baseline in total score. The HAMD-17 is a 17-item assessment used to assess the severity of depression and its improvement during the course of therapy. Each item was evaluated and scored using either a 5-point scale of 0 (not present/absent) to 4 (very severe) or a 3-point scale of 0 (not present/absent) to 2 (marked). Items rated on the 3-point scale include: insomnia early, middle, late; somatic symptoms gastrointestinal and general; genital symptoms; loss of weight; insight. Items rated on 5-point scale include: depressed mood; feelings of guilt; suicide; work and activities; retardation; agitation; anxiety psychic and somatic; hypochondriasis. The total score was the sum of the scores from HAMD-17 Items 1 through 17 and ranged from 0 (not at all depressed) to 52 (severely depressed). Higher scores indicate greater symptom severity. Number of participants with response to HAMD-17 has been presented. (NCT03460756)
Timeframe: Day 2 (all treatment groups), Day 7 (TID & QHS), Day 8/10 (all treatment groups), Day 15/17 (all treatment groups), Day 22 and 29 (QHS, 1125mg), Day 36/38 (all treatment groups), Day 57/59 (QHS, 1125mg), Day 71 (1125mg), Day 89 (QHS), Day 119 (QHS)

,
InterventionParticipants (Count of Participants)
Day 2/4 - ResponseDay 7 - ResponseDay 8/10 - ResponseDay 15/17 - ResponseDay 36/38 - Response
QHS (2-week)24443
TID (2-week)01012

[back to top]

Number of Participants With Hamilton Depression Rating Scale 17-item Remission

Remission was defined as an observed total score of 0 to 7. The HAMD-17 is a 17-item assessment used to assess the severity of depression and its improvement during the course of therapy. Each item was evaluated and scored using either a 5-point scale of 0 (not present/absent) to 4 (very severe) or a 3-point scale of 0 (not present/absent) to 2 (marked). Items rated on the 3-point scale include: insomnia early, middle, late; somatic symptoms gastrointestinal and general; genital symptoms; loss of weight; insight. Items rated on 5-point scale include: depressed mood; feelings of guilt; suicide; work and activities; retardation; agitation; anxiety psychic and somatic; hypochondriasis. The total score was the sum of the scores from HAMD-17 Items 1 through 17 and ranged from 0 (not at all depressed) to 52 (severely depressed). Higher scores indicate greater symptom severity. Number of participants with HAMD-17 remission has been presented. (NCT03460756)
Timeframe: Day 8/10 (all treatment arms), Day 15/17 (all treatment arms), Day 22 and 29 (QHS & 1125mg), Day 36/38 (all treatment arms), Day 57/59 (QHS & 1125mg), Day 71 (1125mg), Day 89 & 119 (QHS)

InterventionParticipants (Count of Participants)
Day 8/10 - RemissionDay 15/17 - RemissionDay 22 - RemissionDay 29 - RemissionDay 36/38 - RemissionDay 57/59 - RemissionDay 89 - RemissionDay 119 - Remission
QHS (4-week)14379578

[back to top]

Number of Participants With Response to Hamilton Depression Rating Scale 17 Item (HAMD17)

HAMD17 Response was defined as ≥50% reduction from Baseline in total score. The HAMD-17 is a 17-item assessment used to assess the severity of depression and its improvement during the course of therapy. Each item was evaluated and scored using either a 5-point scale of 0 (not present/absent) to 4 (very severe) or a 3-point scale of 0 (not present/absent) to 2 (marked). Items rated on the 3-point scale include: insomnia early, middle, late; somatic symptoms gastrointestinal and general; genital symptoms; loss of weight; insight. Items rated on 5-point scale include: depressed mood; feelings of guilt; suicide; work and activities; retardation; agitation; anxiety psychic and somatic; hypochondriasis. The total score was the sum of the scores from HAMD-17 Items 1 through 17 and ranged from 0 (not at all depressed) to 52 (severely depressed). Higher scores indicate greater symptom severity. Number of participants with response to HAMD-17 has been presented. (NCT03460756)
Timeframe: Day 2 (all treatment groups), Day 7 (TID & QHS), Day 8/10 (all treatment groups), Day 15/17 (all treatment groups), Day 22 and 29 (QHS, 1125mg), Day 36/38 (all treatment groups), Day 57/59 (QHS, 1125mg), Day 71 (1125mg), Day 89 (QHS), Day 119 (QHS)

InterventionParticipants (Count of Participants)
Day 2/4 - ResponseDay 8/10 - ResponseDay 15/17 - ResponseDay 22 - ResponseDay 29 - ResponseDay 36/38 - ResponseDay 57/59 - ResponseDay 89 - ResponseDay 119 - Response
QHS (4-week)077810128910

[back to top]

Number of Participants With Hamilton Depression Rating Scale 17-item Remission

Remission was defined as an observed total score of 0 to 7. The HAMD-17 is a 17-item assessment used to assess the severity of depression and its improvement during the course of therapy. Each item was evaluated and scored using either a 5-point scale of 0 (not present/absent) to 4 (very severe) or a 3-point scale of 0 (not present/absent) to 2 (marked). Items rated on the 3-point scale include: insomnia early, middle, late; somatic symptoms gastrointestinal and general; genital symptoms; loss of weight; insight. Items rated on 5-point scale include: depressed mood; feelings of guilt; suicide; work and activities; retardation; agitation; anxiety psychic and somatic; hypochondriasis. The total score was the sum of the scores from HAMD-17 Items 1 through 17 and ranged from 0 (not at all depressed) to 52 (severely depressed). Higher scores indicate greater symptom severity. Number of participants with HAMD-17 remission has been presented. (NCT03460756)
Timeframe: Day 8/10 (all treatment arms), Day 15/17 (all treatment arms), Day 22 and 29 (QHS & 1125mg), Day 36/38 (all treatment arms), Day 57/59 (QHS & 1125mg), Day 71 (1125mg), Day 89 & 119 (QHS)

InterventionParticipants (Count of Participants)
Day 8/10 - RemissionDay 15/17 - RemissionDay 22 - RemissionDay 29 - RemissionDay 36/38 - RemissionDay 57/59 - RemissionDay 71 - Remission
1125 mg (4-week)88101411129

[back to top]

Number of Participants With Hamilton Depression Rating Scale 17-item Remission

Remission was defined as an observed total score of 0 to 7. The HAMD-17 is a 17-item assessment used to assess the severity of depression and its improvement during the course of therapy. Each item was evaluated and scored using either a 5-point scale of 0 (not present/absent) to 4 (very severe) or a 3-point scale of 0 (not present/absent) to 2 (marked). Items rated on the 3-point scale include: insomnia early, middle, late; somatic symptoms gastrointestinal and general; genital symptoms; loss of weight; insight. Items rated on 5-point scale include: depressed mood; feelings of guilt; suicide; work and activities; retardation; agitation; anxiety psychic and somatic; hypochondriasis. The total score was the sum of the scores from HAMD-17 Items 1 through 17 and ranged from 0 (not at all depressed) to 52 (severely depressed). Higher scores indicate greater symptom severity. Number of participants with HAMD-17 remission has been presented. (NCT03460756)
Timeframe: Day 8/10 (all treatment arms), Day 15/17 (all treatment arms), Day 22 and 29 (QHS & 1125mg), Day 36/38 (all treatment arms), Day 57/59 (QHS & 1125mg), Day 71 (1125mg), Day 89 & 119 (QHS)

,
InterventionParticipants (Count of Participants)
Day 8/10 - RemissionDay 15/17 - RemissionDay 36/38 - Remission
QHS (2-week)121
TID (2-week)001

[back to top]

Arithmetic Change in Longest Seizure Free Interval, Based on Primary Seizure Types

Arithmetic change in longest seizure free interval, based on primary seizure types during the double-blind treatment period of ganaxolone compared to placebo (NCT03572933)
Timeframe: End of the double-blind 17 week treatment period

Interventiondays (Mean)
Placebo-4.63
Ganaxolone-0.02

[back to top]

Caregiver Global Impression of Change in Attention

Caregiver global impression of change in attention during the double-blind treatment period of ganaxolone compared to placebo. Investigators and caregivers reported improvements in attention, mood, behavior and sleep via investigator narratives (NCT03572933)
Timeframe: End of the double-blind 17 week treatment period

,
InterventionParticipants (Count of Participants)
Very Much Improved - Visit 5 (End of Week 17)Much Improved - Visit 5 (End of Week 17)Minimally Improved - Visit 5 (End of Week 17)No Change - Visit 5 (End of Week 17)Minimally Worse - Visit 5 (End of Week 17)Much Worse - Visit 5 (End of Week 17)Very Much Worse - Visit 5 (End of Week 17)
Ganaxolone122118111
Placebo171423110

[back to top]

Caregiver Global Impression of Change in Seizure Intensity and Duration

Caregiver global impression of change in seizure intensity and duration during the double-blind treatment period of ganaxolone compared to placebo. CGI-C is a 7 point scale that requires the clinician to assess how much the patient's illness has improved or worsened relative to a baseline state at the beginning of the intervention. (NCT03572933)
Timeframe: End of the double-blind 17 week treatment period

,
InterventionParticipants (Count of Participants)
Very Much Improved - Visit 5 (End of Week 17)Much Improved - Visit 5 (End of Week 17)Minimally Improved - Visit 5 (End of Week 17)No Change - Visit 5 (End of Week 17)4Minimally Worse - Visit 5 (End of Week 17)Much Worse - Visit 5 (End of Week 17)Very Much Worse - Visit 5 (End of Week 17)
Ganaxolone2151110322
Placebo151121540

[back to top]

Caregiver Global Impression of Change in Target Behavior

Caregiver global impression of change in target behavior during the double-blind treatment period of ganaxolone compared to placebo. Investigators and caregivers reported improvements in attention, mood, behavior and sleep via investigator narratives. (NCT03572933)
Timeframe: End of the double-blind 17 week treatment period

,
InterventionParticipants (Count of Participants)
Very Much Improved - Visit 5 (End of Week 17)Much Improved - Visit 5 (End of Week 17)Minimally Improved - Visit 5 (End of Week 17)No Change - Visit 5 (End of Week 17)Minimally Worse - Visit 5 (End of Week 17)Much Worse - Visit 5 (End of Week 17)Very Much Worse - Visit 5 (End of Week 17)
Ganaxolone042019200
Placebo061422121

[back to top]

Clinical Global Impression of Improvement - Clinician

Clinical global impression of improvement during the double-blind treatment period of ganaxolone compared to placebo (NCT03572933)
Timeframe: [Time Frame: End of the double-blind 17 week treatment period]

,
InterventionParticipants (Count of Participants)
Very Much Improved - Visit 5 (End of Week 17) - ClinicianMuch Improved - Visit 5 (End of Week 17) - ClinicianMinimally Improved - Visit 5 (End of Week 17) - ClinicianNo Change - Visit 5 (End of Week 17) - ClinicianMinimally Worse - Visit 5 (End of Week 17) - ClinicianMuch Worse - Visit 5 (End of Week 17) - ClinicianVery Much Worse - Visit 5 (End of Week 17) - Clinician
Ganaxolone071916231
Placebo071319900

[back to top]

Clinical Global Impression of Improvement - Parent/Caregiver

Clinical global impression of improvement during the double-blind treatment period of ganaxolone compared to placebo. The CGI is rated on a 7-point scale, with the severity of illness scale using a range of responses. (NCT03572933)
Timeframe: End of the double-blind 17 week treatment period

,
InterventionParticipants (Count of Participants)
Very Much Improved - Visit 5 (End of Week 17) - Parent/CaregiverMuch Improved - Visit 5 (End of Week 17) - Parent/CaregiverMinimally Improved - Visit 5 (End of Week 17) - Parent/CaregiverNo Change - Visit 5 (End of Week 17) - Parent/CaregiverMinimally Worse - Visit 5 (End of Week 17) - Parent/CaregiverMuch Worse - Visit 5 (End of Week 17) - Parent/CaregiverVery Much Worse - Visit 5 (End of Week 17) - Parent/Caregiver
Ganaxolone0131714220
Placebo171322410

[back to top]

Percentage of Seizure-free Days for Major Motor Seizure Types

Percentage of Seizure-free Days for Major Motor Seizure types during the double-blind treatment period of ganaxolone compared to placebo. The major motor seizure types include bilateral tonic (sustained motor activity = 3 seconds), generalized tonic-clonic, atonic/drop, bilateral clonic, and focal to bilateral tonic-clonic. (NCT03572933)
Timeframe: End of the double-blind 17 week treatment period

,
Interventionpercent of seizure-free days (Mean)
Baseline17-week-Post-Baseline PhaseArithmetic Change from Baseline
Ganaxolone22.5732.299.62
Placebo30.3236.175.86

[back to top]

Summary of 28-day Seizure Frequency for Major Motor Seizure Types

"Summary of 28-day seizure frequency for Major Motor Seizure Types during the double-blind treatment period relative to the 6-week prospective baseline period~Note: Summaries are based on the sum of the individual seizures, the countable seizures, and the clusters with uncountable seizures (each cluster with uncountable seizures counts as 1 seizure). Within the baseline and post baseline intervals, 28-day seizure frequency was calculated as the total number of seizures in the interval divided by the number of days with available seizure data in the interval, multiplied by 28." (NCT03572933)
Timeframe: End of the double-blind 17 week treatment period

,
InterventionSeizures per day (Median)
Baseline (Median)17 week-post baseline phase (Median)
Ganaxolone54.0045.03
Placebo49.1755.50

[back to top]

50% Primary Seizure Reduction

Percent of subjects experiencing a greater than or equal to 50% reduction in 28-day primary seizure frequency relative to the 12-week baseline (NCT03865732)
Timeframe: End of the double-blind 17 week treatment period

InterventionParticipants (Count of Participants)
Placebo4
Ganaxolone5

[back to top]

Summary of 28-day Seizure Frequency for Subjects in the Biomarker-positive Stratum (Median Percent Change)

Summary of 28-day Seizure Frequency for Seizure Types for Subjects in the Biomarker-positive Stratum through 17 weeks (Median Percent Change) (NCT03865732)
Timeframe: [Time Frame: End of the double-blind 17 week treatment period]

InterventionMedian % Change in Number of Seizures (Median)
Placebo-18.71
Ganaxolone-35.90

[back to top]

Summary of 28-day Seizure Frequency Through 17 Week Post-Baseline Phase (Median Percent Change)

Summary of 28-Day Seizure Frequency for Seizure Types through 17 week Post-Baseline Phase (Median Percent Change) (NCT03865732)
Timeframe: End of the double-blind 17 week treatment period

InterventionMedian % Change in Number of Seizures (Median)
Placebo-23.97
Ganaxolone-61.52

[back to top]

Percent Change From Baseline in 28-day Seizure Frequency Through the End of 12-Week Treatment Period

Primary seizures include atonic/drop, bilateral clonic, bilateral tonic, focal motor without impairment of consciousness or awareness, focal (motor or non-motor) with impairment of consciousness or awareness, focal to bilateral tonic-clonic, generalized tonic-clonic. Baseline 28-day seizure frequency was calculated as the total number of primary seizures in the Baseline period divided by the number of days with non-missing seizure data in the Baseline period, multiplied by 28. The Baseline Visit was defined as Week 0. Percent change from Baseline in 28-day seizure frequent was calculated as the difference in post-Baseline 28-day seizure frequency and Baseline 28-day seizure frequency, divided by Baseline 28-day seizure frequency, multiplied by 100. (NCT04285346)
Timeframe: Baseline and Up to Week 12

InterventionPercent change (Median)
Treatment Period: Ganaxolone-16.61

[back to top]

Percentage of Participants Experiencing a >=50 Percent Reduction in 28-day Primary Seizure Frequency Through the End of the 12-week Treatment Period Compared to the Baseline Period

Primary seizures include atonic/drop, bilateral clonic, bilateral tonic, focal motor without impairment of consciousness or awareness, focal (motor or non-motor) with impairment of consciousness or awareness, focal to bilateral tonic-clonic, generalized tonic-clonic. Percentage of participants reporting >=50 percent reduction in seizure frequency has been presented. (NCT04285346)
Timeframe: Baseline and up to 12 Weeks

InterventionPercentage of participants (Number)
Treatment Period: Ganaxolone30.4

[back to top]