Page last updated: 2024-11-06

fenofibric acid

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

Description

Fenofibric acid is a fibric acid derivative that is used as a medication to treat high cholesterol and triglycerides. It is a potent peroxisome proliferator-activated receptor alpha (PPARα) agonist. Fenofibric acid works by reducing the production of very low-density lipoproteins (VLDL) and increasing the breakdown of triglycerides. It is also known to increase the levels of high-density lipoproteins (HDL), the “good” cholesterol. Fenofibric acid is typically used in combination with lifestyle modifications, such as diet and exercise. Fenofibric acid is synthesized through several different pathways, starting with the precursor 2-chloro-4-trifluoromethylbenzoic acid. This compound is then reacted with different reagents to yield the final product. Fenofibric acid is studied due to its potential to treat dyslipidemia, a condition characterized by high levels of cholesterol and triglycerides in the blood. It is also being studied for its potential to treat other conditions, such as insulin resistance, metabolic syndrome, and inflammation. Fenofibric acid has several potential side effects, including muscle pain, liver problems, and gallstones. However, it is generally considered safe when used as directed.'

fenofibric acid: RN given refers to parent cpd without isomeric designation; structure [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

fenofibric acid : A monocarboxylic acid that is 2-methylpropanoic acid substituted by a 4-(4-chlorobenzoyl)phenoxy group at position 2. It is a metabolite of the drug fenofibrate. [Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Cross-References

ID SourceID
PubMed CID64929
CHEMBL ID981
CHEBI ID83469
SCHEMBL ID16377
MeSH IDM0047980

Synonyms (78)

Synonym
gtpl2662
fenofibric acid
2-(4-(4'-chlorophenoxy)phenoxy)propionic acid
42017-89-0
nsc-281318
nsc281318
einecs 255-626-9
ccris 7302
2-(4-(4-chlorobenzoyl)phenoxy)-2-methylpropanoic acid
brn 2058973
procetofenic acid
2-(4-(4-chlorobenzoyl)phenoxy)-2-methylpropionic acid
alpha 1081
nsc 281318
lf 153
propanoic acid, 2-(4-(4-chlorobenzoyl)phenoxy)-2-methyl-
lf 178 acid
chembl981 ,
bdbm28700
2-{4-[(4-chlorophenyl)carbonyl]phenoxy}-2-methylpropanoic acid
lf-153
chebi:83469 ,
fenofibrate free acid
2-[4-(4-chlorobenzoyl)phenoxy]-2-methylpropanoic acid
unii-bgf9mn2hu1
bgf9mn2hu1 ,
A825720
2-[4-(4-chloro-benzoyl)-phenoxy]-2-methyl-propionic acid
S4527
FT-0600402
fenofibric acid [vandf]
fenofibrate free acid [mi]
fenofibric acid [usp-rs]
fenofibric acid [orange book]
fenofibric acid [who-dd]
AKOS015889489
fnf acid
CCG-213311
HY-B0760
SCHEMBL16377
KS-1234
2-[4-(4-chlorobenzoyl)-phenoxy]-2-methylpropionic acid
MQOBSOSZFYZQOK-UHFFFAOYSA-N
feno-fibric acid
fenofibricacid
W-106287
AC-22277
F1011
2-[4-(4-chlorobenzoyl)phenoxy]-2-methylpropionic acid
AB01563028_01
DTXSID8041030 ,
fenofibric acid, analytical standard
2-[4-(4-chlorobenzoyl)phenoxy]-2-methylpropionic acid, 95%
mfcd00792461
propanoic acid, 2-[4-(4-chlorobenzoyl)phenoxy]-2-methyl-
fenofibrate impurity b, european pharmacopoeia (ep) reference standard
2-[4-(4-chlorobenzoyl)phenoxy]-2-methylpropanoic acid (fenofibric acid)
DB13873
fenofibric acid
BCP22437
Q27077290
fibricor (tn)
D11579
AMY25229
fenofibric-acid
D83849
SY052754
2-[4-(4-chlorobenzene-1-carbonyl)phenoxy]-2-methylpropanoic acid
f5a ,
2-[4-(p-chlorobenzoyl)phenoxy]-2-methylpropionicacid
EN300-1697940
fenofibric acid 100 microg/ml in acetonitrile
HY-B0760R
CS-0694864
fenofibric acid (standard)
fenofibric acid (usp-rs)
fenofibric acid delayed-release
dtxcid6021030

Research Excerpts

Overview

Fenofibric acid is a hypolipidemic drug that inhibits 3-hydroxy-3-methyl-glutaryl coenzyme A reductase. It is used as an active ingredient or is administered in the form of fenofibrate.

ExcerptReferenceRelevance
"Fenofibric acid is a hypolipidemic drug that is used as an active ingredient per se or is administered in the form of fenofibrate that releases fenofibric acid after absorption. "( In vitro metabolism of fenofibric acid by carbonyl reducing enzymes.
Kanavi, M; Malátková, P; Nobilis, M; Wsól, V, 2016
)
2.19
"Fenofibric acid is a hypolipidemic drug which inhibits 3-hydroxy-3-methyl-glutaryl coenzyme A reductase (HMGCoA reductase) activity, the limiting step of endogenous cholesterol synthesis."( Evidence for the inhibition of platelet-derived growth factor induced rat smooth muscle cells DNA synthesis by fenofibric acid at the Go/G1 cell cycle level.
Chazan, JB; Majoie, B; Pascal, M; Sepulchre, C, 1983
)
1.2

Effects

ExcerptReferenceRelevance
"Fenofibric acid (FA) has been studied in a large number of patients with mixed dyslipidemia, combined with a low- or moderate-dose statin."( A review of time courses and predictors of lipid changes with fenofibric acid-statin combination.
Filippatos, TD, 2012
)
1.34

Actions

Fenofibric acid gave lower plasma concentrations, AUC and Cmax values compared to choline fenofibrate in rats. The compound could inhibit this synthesis when it was present in the culture medium concomitantly with platelet extract.

ExcerptReferenceRelevance
"Fenofibric acid gave lower plasma concentrations, AUC and Cmax values compared to choline fenofibrate in rats."( Effect of magnesium carbonate on the solubility, dissolution and oral bioavailability of fenofibric acid powder as an alkalising solubilizer.
Cho, KH; Choi, HG; Jin, SG; Kim, DS; Kim, DW; Kim, JH; Kim, JO; Kim, KS; Li, DX; Woo, JS; Yong, CS, 2016
)
1.38
"Fenofibric acid could inhibit this synthesis when the compound was present in the culture medium concomitantly with platelet extract."( Evidence for the inhibition of platelet-derived growth factor induced rat smooth muscle cells DNA synthesis by fenofibric acid at the Go/G1 cell cycle level.
Chazan, JB; Majoie, B; Pascal, M; Sepulchre, C, 1983
)
1.2

Toxicity

The combination of rosuvastatin 5 mg + fenofibric acid 135 mg resulted in comprehensive improvements in the lipid profile of patients with mixed dyslipidemia. In conclusion, these data suggest that a combination of fenfibric Acid and a statin could be considered safe and efficacious.

ExcerptReferenceRelevance
" Due to the adverse effects provoked by hypolipidemic drugs on skeletal muscle function, we also investigated the blocking activity of S-2 and S-4 on skeletal muscle membrane chloride channel conductance and found that these ligands have a pharmacological profile more beneficial compared to fibrates currently used in therapy."( New 2-aryloxy-3-phenyl-propanoic acids as peroxisome proliferator-activated receptors alpha/gamma dual agonists with improved potency and reduced adverse effects on skeletal muscle function.
Conte Camerino, D; Fracchiolla, G; Laghezza, A; Lavecchia, A; Loiodice, F; Mazza, F; Montanari, R; Novellino, E; Piemontese, L; Pierno, S; Pochetti, G; Tortorella, P, 2009
)
0.35
"Mean percentage changes in lipid parameters, percentages of patients achieving optimal serum lipid/apolipoprotein levels, and incidence of adverse events."( Efficacy and safety of fenofibric acid co-administered with low- or moderate-dose statin in patients with mixed dyslipidemia and type 2 diabetes mellitus: results of a pooled subgroup analysis from three randomized, controlled, double-blind trials.
Cusi, K; Davidson, MH; Jones, PH; Kelly, MT; Setze, CM; Sleep, DJ; Stolzenbach, JC; Thakker, K, 2010
)
0.67
" Incidence of adverse events was generally similar among treatments."( Efficacy and safety of fenofibric acid co-administered with low- or moderate-dose statin in patients with mixed dyslipidemia and type 2 diabetes mellitus: results of a pooled subgroup analysis from three randomized, controlled, double-blind trials.
Cusi, K; Davidson, MH; Jones, PH; Kelly, MT; Setze, CM; Sleep, DJ; Stolzenbach, JC; Thakker, K, 2010
)
0.67
" Safety was evaluated based on data collected for adverse events (AEs), physical and electrocardiographic examinations, vital sign measurements, and clinical laboratory tests."( Efficacy and safety of rosuvastatin and fenofibric acid combination therapy versus simvastatin monotherapy in patients with hypercholesterolemia and hypertriglyceridemia: a randomized, double-blind study.
Carlson, DM; Gold, A; Jones, PH; Kelly, MT; McKenney, JM; Roth, EM; Setze, CM; Stolzenbach, JC; Williams, LA, 2010
)
0.63
" Overall, adverse events were similar in the 2 treatment groups."( Efficacy and safety of fenofibric acid in combination with atorvastatin and ezetimibe in patients with mixed dyslipidemia.
Goldberg, AC; Jones, PH; Kelly, MT; Knapp, HR; Setze, CM; Sleep, DJ; Stolzenbach, JC, 2010
)
0.67
"In conclusion, rosuvastatin 5 mg + fenofibric acid 135 mg resulted in comprehensive improvements in the lipid profile of patients with mixed dyslipidemia without unanticipated adverse events."( Efficacy and safety of rosuvastatin 5 mg in combination with fenofibric acid 135 mg in patients with mixed dyslipidemia - a phase 3 study.
Blasetto, JW; Carlson, DM; Fukumoto, SM; Khurmi, NS; Rosenson, RS; Roth, EM; Setze, CM; Stolzenbach, JC; Williams, LA, 2010
)
0.88
" In conclusion, these data suggest that a combination of fenofibric acid and a statin could be considered safe and efficacious for treating women with mixed dyslipidemia."( Efficacy of fenofibric acid plus statins on multiple lipid parameters and its safety in women with mixed dyslipidemia.
Bittner, V; Goldberg, AC; Kelly, MT; Lele, A; Pepine, CJ; Setze, CM; Sleep, DJ; Thakker, K, 2011
)
0.99
" Adverse events (AEs) and clinical laboratory values were also assessed."( One-year efficacy and safety of rosuvastatin + fenofibric acid combination therapy in patients with mixed dyslipidemia: evaluation of dose response.
Davidson, MH; Ferdinand, KC; Kelly, MT; Setze, CM, 2012
)
0.64
"Patients with mixed dyslipidemia can benefit from the combination of fenofibric acid (FA) with statins, but concerns about adverse events make physicians reluctant to prescribe the combination therapy."( Adverse events of statin-fenofibric acid versus statin monotherapy: a meta-analysis of randomized controlled trials.
Chen, H; Geng, Q; Lee, C; Liang, W; Ren, J, 2013
)
0.93
"In the present study, we performed a meta-analysis of randomized controlled trials (RCTs) to assess the efficacy and adverse events in patients taking statins and FA."( Adverse events of statin-fenofibric acid versus statin monotherapy: a meta-analysis of randomized controlled trials.
Chen, H; Geng, Q; Lee, C; Liang, W; Ren, J, 2013
)
0.69
"Medline, Embase and the Cochrane Library were searched to identify studies that reported adverse events."( Adverse events of statin-fenofibric acid versus statin monotherapy: a meta-analysis of randomized controlled trials.
Chen, H; Geng, Q; Lee, C; Liang, W; Ren, J, 2013
)
0.69
" The adverse events in the FA + moderate-dose statin group were almost identical to those in the FA + low-dose statin group."( Adverse events of statin-fenofibric acid versus statin monotherapy: a meta-analysis of randomized controlled trials.
Chen, H; Geng, Q; Lee, C; Liang, W; Ren, J, 2013
)
0.69
" However, we still need large-scale and long follow-up period RCTs to definitively confirm the adverse events of FA-statin therapy."( Adverse events of statin-fenofibric acid versus statin monotherapy: a meta-analysis of randomized controlled trials.
Chen, H; Geng, Q; Lee, C; Liang, W; Ren, J, 2013
)
0.69

Pharmacokinetics

Pharmacokinetic parameters of fenofibric acid was evaluated after oral (at doses of 20 mg/kg) administration of JW322. Fenofibrate was dosed alone and with rosuvastatin: AUC from 0 to 8 hours and Cmax decreased by 4% (90% CI, 0.5%)

ExcerptReferenceRelevance
"This study was conducted to evaluate the potential pharmacokinetic interaction between fenofibrate and pravastatin."( Lack of a clinically significant pharmacokinetic interaction between fenofibrate and pravastatin in healthy volunteers.
Achari, R; Gustavson, LE; Gutterman, C; Pan, WJ; Rieser, MJ; Wallin, BA; Ye, X, 2000
)
0.31
" Despite breaking all the "rule of five" criteria, the dimers had excellent oral bioavailability and pharmacokinetic properties, resulting in good in vivo efficacy in db/db mice."( Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
Albrektsen, T; Bury, PS; Deussen, HJ; Din, N; Fleckner, J; Frederiksen, KS; Jeppesen, L; Mogensen, JP; Nehlin, J; Pettersson, I; Sauerberg, P; Svensson, LA; Wulff, EM; Ynddal, L, 2003
)
0.32
"This study was conducted to evaluate the potential for pharmacokinetic interaction between fenofibrate and ezetimibe in healthy subjects."( Evaluation of the potential for pharmacokinetic interaction between fenofibrate and ezetimibe: A phase I, open-label, multiple-dose, three-period crossover study in healthy subjects.
Achari, R; Burt, DA; Chira, T; Edeki, T; Gustavson, LE; Kelly, MT; Rieser, MJ; Schweitzer, SM; Yannicelli, HD, 2006
)
0.33
" ABT-335 and rosuvastatin have different mechanisms of actions and exert complementary pharmacodynamic effects on lipids."( ABT-335, the choline salt of fenofibric acid, does not have a clinically significant pharmacokinetic interaction with rosuvastatin in humans.
Awni, WM; Chira, TO; Hosmane, B; Kelly, MT; Pradhan, RS; Sleep, DJ; Stolzenbach, JC; Wan, K; Zhu, T, 2009
)
0.64
"Management of a lipophilic-hydrophilic balance is a key element in drug design to achieve desirable pharmacokinetic characters."( A novel prodrug strategy for extremely hydrophobic agents: conjugation to symmetrically branched glycerol trimer improves pharmacological and pharmacokinetic properties of fenofibrate.
Abe, S; Hattori, H; Ishizawa, K; Kamiya, M; Kono, M; Matsushita, T; Miyamoto, L; Nemoto, H; Taoka, C; Tomida, Y; Tsuchiya, K; Watanabe, M, 2013
)
0.39
" The 90% CIs for the test/reference mean ratios of the ln-transformed pharmacokinetic variables Cmax, AUC0-t, and AUC0-∞ were within the conventional bioequivalence range of 80% to 125%."( Comparison of pharmacokinetics of two fenofibrate tablet formulations in healthy human subjects.
Chachad, SS; Gole, M; Malhotra, G; Naidu, R, 2014
)
0.4
"Open-label, single-sequence pharmacokinetic study."( Lack of an Effect of Ritonavir Alone and Lopinavir-Ritonavir on the Pharmacokinetics of Fenofibric Acid in Healthy Volunteers.
Alfaro, RM; Calderón, MM; Gordon, LA; Hadigan, C; Kovacs, JA; Malati, CY; McLaughlin, M; Penzak, SR, 2016
)
0.66
" Fenofibric acid pharmacokinetic parameter values were compared before and after concomitant ritonavir or lopinavir-ritonavir administration."( Lack of an Effect of Ritonavir Alone and Lopinavir-Ritonavir on the Pharmacokinetics of Fenofibric Acid in Healthy Volunteers.
Alfaro, RM; Calderón, MM; Gordon, LA; Hadigan, C; Kovacs, JA; Malati, CY; McLaughlin, M; Penzak, SR, 2016
)
1.57
" Pharmacokinetic parameters of fenofibric acid was evaluated after oral (at doses of 20 mg/kg) administration of JW322 and fenofibrate in rats."( Determination of Fenofibric Acid in Rat Plasma and its Application to a Comparative Pharmacokinetic Study of JW322 and Fenofibrate.
Kim, TK, 2017
)
1.08

Compound-Compound Interactions

A multicenter, double-blind, active-controlled study evaluated ABT-335 (fenofibric acid) in combination with 2 doses of atorvastatin in patients with mixed dyslipidemia.

ExcerptReferenceRelevance
"In this study, a sensitive and selective method based on liquid chromatography combined with diode array and tandem mass spectrometry detection (LC-DAD-MS/MS) was developed for the simultaneous quantitative determination of fenofibric acid, pravastatin and its main metabolites in human plasma."( An automated method for the simultaneous determination of pravastatin, 3-hydroxy isomeric metabolite, pravalactone and fenofibric acid in human plasma by sensitive liquid chromatography combined with diode array and tandem mass spectrometry detection.
Cahay, B; Klinkenberg, R; Mertens, B; Streel, B, 2008
)
0.74
" A comprehensive, controlled clinical trial programme was thus designed to evaluate three separate statins in combination with ABT-335, a new formulation of fenofibric acid."( Evaluation of a new formulation of fenofibric acid, ABT-335, co-administered with statins : study design and rationale of a phase III clinical programme.
Bays, HE; Buttler, SM; Davidson, MH; Jones, PH; Kelly, MT; Setze, CM; Sleep, DJ; Stolzenbach, JC, 2008
)
0.82
" This multicenter, double-blind, active-controlled study evaluated ABT-335 (fenofibric acid) in combination with 2 doses of atorvastatin in patients with mixed dyslipidemia."( Efficacy and safety of ABT-335 (fenofibric acid) in combination with atorvastatin in patients with mixed dyslipidemia.
Ballantyne, CM; Bays, HE; Buttler, SM; Goldberg, AC; Kelly, MT; Setze, CM; Sleep, DJ; Stolzenbach, JC, 2009
)
0.87
" The fibrate class of drugs has proven efficacy in improving secondary targets; however, concerns regarding severe myopathy and rhabdomyolysis have limited their combination with statins."( Fibrate therapy in the management of dyslipidemias, alone and in combination with statins: role of delayed-release fenofibric acid.
Hilleman, DE; Maciejewski, SR; Mohiuddin, SM; Schima, SM; Williams, MA, 2010
)
0.57
" It has been extensively evaluated both as monotherapy and in combination with atorvastatin, rosuvastatin, and simvastatin in a large number of patients with mixed dyslipidemia for up to 2 years and appears to be a safe and effective option in the management of dyslipidemia."( Management of dyslipidemias with fibrates, alone and in combination with statins: role of delayed-release fenofibric acid.
Elisaf, MS; Kei, A; Milionis, HJ; Moutzouri, E, 2010
)
0.57

Bioavailability

Fenofibric acid is one of the newly approved forms of fenofibrate with enhanced bioavailability. It was recently approved by the Food and Drug Administation (FDA) for the treatment of various types of hyperlipidemia.

ExcerptReferenceRelevance
" Pharmacokinetic parameters of these three compounds were calculated using noncompartmental methods and compared by analyses of variance and bioavailability assessments."( Lack of a clinically significant pharmacokinetic interaction between fenofibrate and pravastatin in healthy volunteers.
Achari, R; Gustavson, LE; Gutterman, C; Pan, WJ; Rieser, MJ; Wallin, BA; Ye, X, 2000
)
0.31
" Despite breaking all the "rule of five" criteria, the dimers had excellent oral bioavailability and pharmacokinetic properties, resulting in good in vivo efficacy in db/db mice."( Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
Albrektsen, T; Bury, PS; Deussen, HJ; Din, N; Fleckner, J; Frederiksen, KS; Jeppesen, L; Mogensen, JP; Nehlin, J; Pettersson, I; Sauerberg, P; Svensson, LA; Wulff, EM; Ynddal, L, 2003
)
0.32
"This study compared the gastrointestinal (GI) absorption characteristics and absolute bioavailability of fenofibric acid and fenofibrate (which is converted to fenofibric acid in vivo) in healthy volunteers."( Comparison of the gastrointestinal absorption and bioavailability of fenofibrate and fenofibric acid in humans.
Ansquer, JC; Kelly, MT; Pradhan, RS; Sleep, DJ; Zhu, T, 2010
)
0.8
" Fenofibric acid is one of the newly approved forms of fenofibrate with enhanced bioavailability and was recently approved by the Food and Drug Administation (FDA) for the treatment of various types of hyperlipidemia, in conjunction with statins."( Fenofibric acid for hyperlipidemia.
Kaushik, M; Mohiuddin, SM; Saurav, A, 2012
)
2.73
" In summary, the present study demonstrated the particle size dependence of bioavailability of fenofibrate loaded LMP in rat model which correlates well with the in vitro drug release performed in the biorelevant medium."( Investigating the correlation between in vivo absorption and in vitro release of fenofibrate from lipid matrix particles in biorelevant medium.
Borkar, N; Gan, Y; Holm, R; Mu, H; Müllertz, A; Xia, D; Yang, M, 2014
)
0.4
" Most of these formulations show a higher bioavailability when taken with a high-fat meal."( The Lidose hard capsule formulation of fenofibrate is suprabioavailable compared to the nanoparticle tablet formulation under high-fat fed conditions.
Cahay, B; Coffiner, M; De Niet, S; Lebrun, S; Rennie, TW; Streel, B; Tremege, M; Verbeeck, RK, 2015
)
0.42
" The intestinal absorption rate for each drug was acquired by deconvolution, using historical intravenous data as reference, and used with the intestinal surface area and the dose remaining in the lumen to estimate the Peff."( Human in vivo regional intestinal permeability: quantitation using site-specific drug absorption data.
Dahlgren, D; Lennernäs, H; Roos, C; Sjögren, E, 2015
)
0.42
"The objective of this study was to develop a novel fenofibric acid-loaded controlled release pellet showing enhanced, or equivalent to, bioavailability compared with two commercially available products containing fenofibrate or choline fenofibrate."( Novel fenofibric acid-loaded controlled release pellet bioequivalent to choline fenofibrate-loaded commercial product in beagle dogs.
Choi, HG; Jin, SG; Kim, DW; Kim, JO; Kim, KS; Kim, YH; Mustapha, O; Woo, JS; Yong, CS; Yousaf, AM, 2015
)
1.15
"This research aimed to develop the omega-3 phospholipids based solid dispersion to improve the oral bioavailability of fenofibrate."( Development of omega-3 phospholipid-based solid dispersion of fenofibrate for the enhancement of oral bioavailability.
Han, HK; Shao, Y; Yang, L, 2015
)
0.42
" Furthermore, the effect of magnesium carbonate, an alkalising agent on the solubility, dissolution and oral bioavailability of fenofibric acid was assessed, a mixture of fenofibric acid and magnesium carbonate being prepared by simple blending at a weight ratio of 2/1."( Effect of magnesium carbonate on the solubility, dissolution and oral bioavailability of fenofibric acid powder as an alkalising solubilizer.
Cho, KH; Choi, HG; Jin, SG; Kim, DS; Kim, DW; Kim, JH; Kim, JO; Kim, KS; Li, DX; Woo, JS; Yong, CS, 2016
)
0.86
"This study aimed to prepare the aminoclay-lipid hybrid composite to enhance the drug release and improve the oral bioavailability of poorly water-soluble fenofibrate."( Aminoclay-lipid hybrid composite as a novel drug carrier of fenofibrate for the enhancement of drug release and oral absorption.
Han, HK; Shao, Y; Yang, L, 2016
)
0.43
" Monoacyl phosphatidylcholine (Lipoid S LPC 80 (LPC)) was incorporated into four different fenofibrate (FF)-loaded long-chain SEDDS to investigate the impact of LPC on the emulsion droplet size, extent of digestion, colloidal structure evolution and drug precipitation during in vitro lipolysis simulating human conditions and drug bioavailability in a rat model."( In vitro and in vivo performance of monoacyl phospholipid-based self-emulsifying drug delivery systems.
Amenitsch, H; Müllertz, A; Rades, T; Siqueira, SDVS; Tran, T, 2017
)
0.46
" After oral administration (20 mg/kg) of JW322, relative bioavailability was approximately 272."( Determination of Fenofibric Acid in Rat Plasma and its Application to a Comparative Pharmacokinetic Study of JW322 and Fenofibrate.
Kim, TK, 2017
)
0.79
"This study aimed to evaluate the effect of solid self-nanoemulsifying drug delivery system (S-SNEDDS) formation on the bioavailability of fenofibric acid."( Comparative Bioavailability Study of Solid Self-Nanoemulsifying Drug Delivery System of Fenofibric Acid in Healthy Male Subjects.
Mudhakir, D; Pamudji, JS; Suhery, WN; Sumirtapura, YC, 2022
)
1.15
" Bioavailability study was conducted in 12 Indonesian healthy male subjects after a single-dose administration of each formulation with three-way crossover design."( Comparative Bioavailability Study of Solid Self-Nanoemulsifying Drug Delivery System of Fenofibric Acid in Healthy Male Subjects.
Mudhakir, D; Pamudji, JS; Suhery, WN; Sumirtapura, YC, 2022
)
0.94
"S-SNEDDS increased the dissolution rate in acid medium and absorption rate of fenofibric acid but did not increase the extent of fenofibric acid absorption."( Comparative Bioavailability Study of Solid Self-Nanoemulsifying Drug Delivery System of Fenofibric Acid in Healthy Male Subjects.
Mudhakir, D; Pamudji, JS; Suhery, WN; Sumirtapura, YC, 2022
)
1.17

Dosage Studied

Once-daily fenofibric acid 135 mg plus a statin was generally as well tolerated as monotherapy. Blood samples were collected prior to dosing on multiple days and up to 120 hours after day 10 dosing.

ExcerptRelevanceReference
" At the same time the pharmacokinetics was studied to check whether this new dosage schedule would give a therapeutic plasma levels of fenofibrate."( Effect of a single daily dose treatment of fenofibrate on plasma lipoproteins in hyperlipoproteinaemia IIb.
Balestreri, R; Bertolini, S; Carozzi, A; Cotta Ramusino, AM; Daga, A; Degl'Innocenti, ML; Elicio, N; Fusi, MG, 1988
)
0.27
" At the end of this period, P was continued at the same dosage but F (300 mg daily) was added and both drugs were given simultaneously for a further eight-day period (period P + F)."( Changes in plasma activities of lipolytic enzymes and lipids of normolipidemic subjects given phenobarbital, a strong microsomal inducer, alone or in combination with fenofibrate.
Desager, JP; Harvengt, C; Heller, FR, 1988
)
0.27
" The study, followed-up as an open trial using higher doses (100 or 200 mg/day ciprofibrate, 400 mg/day fenofibrate) tried to demonstrate clearly the benefit of therapy after 9 months with the 2 drugs and to establish the dose-response effects."( Comparative evaluation of the effects of ciprofibrate and fenofibrate on lipids, lipoproteins and apoproteins A and B.
Bakir, R; Chanu, B; Djian, F; Goy-Loeper, J; Rouffy, J, 1985
)
0.27
" Compared with the control animals, activity of cyanide-insensitive fatty acyl-CoA (FA-CoA) oxidizing system was significantly increased by fenofibrate at the high dosage, carnitine acetyltransferase activity was markedly increased by both drugs at the high dosage and catalase activity remained unmodified."( Effect of fenofibrate and LF 2151 on hepatic peroxisomes in hamsters.
Harvengt, C; Heller, F; Pourbaix, S, 1984
)
0.27
" The drug was further evaluated in a study comprising 56 patients, which combined a dose-response trial with a subsequent comparison between the optimal fenofibrate dose and a clofibrate dose of 2 g/day."( Fenofibrate therapy of hyperlipoproteinaemia. A dose-response study and a comparison with clofibrate.
Orö, L; Rössner, S,
)
0.13
" The need for dosage titration of the micronized preparation from 67 mg/d upward to a final dose of 200 mg/d is also not supported by peer-reviewed literature (except in the case of renal impairment)."( Micronized fenofibrate: a new fibric acid hypolipidemic agent.
Guay, DR, 1999
)
0.3
" Healthy male volunteers received one of the following treatment regimens in each period: rosuvastatin 10 mg orally once daily; fenofibrate 67 mg orally TID; and rosuvastatin + fenofibrate dosed as above."( An open-label, randomized, three-way crossover trial of the effects of coadministration of rosuvastatin and fenofibrate on the pharmacokinetic properties of rosuvastatin and fenofibric acid in healthy male volunteers.
Dane, AL; Martin, PD; Schneck, DW; Warwick, MJ, 2003
)
0.51
" Blood samples were collected for up to 24 hours after dosing on study day 1 and up to 120 hours after dosing on study day 10 for determination of plasma concentrations of fenofibric acid, unconjugated (free) ezetimibe, and total (conjugated and unconjugated) ezetimibe using validated high-performance liquid chromatography methods with mass-spectrometric detection."( Evaluation of the potential for pharmacokinetic interaction between fenofibrate and ezetimibe: A phase I, open-label, multiple-dose, three-period crossover study in healthy subjects.
Achari, R; Burt, DA; Chira, T; Edeki, T; Gustavson, LE; Kelly, MT; Rieser, MJ; Schweitzer, SM; Yannicelli, HD, 2006
)
0.53
" Blood samples were collected prior to dosing on multiple days and up to 120 hours after day 10 dosing for the measurements of fenofibric acid and rosuvastatin plasma concentrations."( ABT-335, the choline salt of fenofibric acid, does not have a clinically significant pharmacokinetic interaction with rosuvastatin in humans.
Awni, WM; Chira, TO; Hosmane, B; Kelly, MT; Pradhan, RS; Sleep, DJ; Stolzenbach, JC; Wan, K; Zhu, T, 2009
)
0.85
" Once-daily fenofibric acid 135 mg plus a statin was generally as well tolerated as monotherapy with fenofibric acid 135 mg/day or the corresponding statin dosage in the three phase III trials in patients with mixed dyslipidemia."( Fenofibric acid: in combination therapy in the treatment of mixed dyslipidemia.
Keating, GM; Yang, LP, 2009
)
2.17
" Study 2 followed a similar dosing scheme and differed only in that volunteers received their single dose after being fed a standard meal (575 calories, of which 36% were contributed by fat)."( Single-dose bioequivalence of 105-mg fenofibric acid tablets versus 145-mg fenofibrate tablets under fasting and fed conditions: a report of two phase I, open-label, single-dose, randomized, crossover clinical trials.
Davis, MW; Digiacinto, J; Godfrey, AR, 2011
)
0.64
" A comprehensive, 12-concentration dose-response analysis using a cell-based assay showed aleglitazar to be highly potent, with EC(50) values of 5 nM and 9 nM for PPARα and PPARγ, respectively."( Comparative molecular profiling of the PPARα/γ activator aleglitazar: PPAR selectivity, activity and interaction with cofactors.
Benz, J; Dietz, M; Grether, U; Hartman, P; Kuhn, B; Maerki, HP; Mohr, P; Ruf, A; Wright, MB, 2012
)
0.38
" Replicate dosing was conducted for the reference formulation to determine its intrasubject variability."( Comparison of pharmacokinetics of two fenofibrate tablet formulations in healthy human subjects.
Chachad, SS; Gole, M; Malhotra, G; Naidu, R, 2014
)
0.4
"The therapeutic equivalence of multiple registered fenofibrate formulations, several of which are suprabioavailable and therefore marketed at lower dosage strengths than their reference products, is based on the results of bioequivalence studies."( The Lidose hard capsule formulation of fenofibrate is suprabioavailable compared to the nanoparticle tablet formulation under high-fat fed conditions.
Cahay, B; Coffiner, M; De Niet, S; Lebrun, S; Rennie, TW; Streel, B; Tremege, M; Verbeeck, RK, 2015
)
0.42
" Meanwhile, in vivo dosing of fenofibrate ameliorated the downregulated amplitudes of ERG c-wave in HFD-fed mice and suppressed the HFD-induced oxidative injury and inflammatory response in RPE tissues."( Fenofibrate Ameliorates Retinal Pigment Epithelium Injury Induced by Excessive Fat Through Upregulation of PI3K/AKT Signaling.
Feng, Y; Li, S; Li, W; Liu, X; Tzekov, R; Wang, X; Wu, Y; Xu, Y; Yang, J; Yu, C, 2023
)
0.91
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (2)

RoleDescription
marine xenobiotic metaboliteAny metabolite produced by metabolism of a xenobiotic compound in marine macro- and microorganisms.
drug metabolitenull
[role 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]

Drug Classes (3)

ClassDescription
chlorobenzophenoneA member of the class of benzophenones that is any substituted benzophenone in which at least one of the substituents is a chlorine.
monocarboxylic acidAn oxoacid containing a single carboxy group.
aromatic ketoneA ketone in which the carbonyl group is attached to an aromatic ring.
[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 (19)

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Fatty acid-binding protein, liverRattus norvegicus (Norway rat)Ki6.83880.01501.24876.9200AID1800412; AID407366; AID407369
Fatty acid-binding protein, liverHomo sapiens (human)Ki2.12000.01501.47708.1700AID1800412
Fatty acid-binding protein, intestinalHomo sapiens (human)Ki1.00000.30005.48149.4000AID407368
Prostaglandin G/H synthase 1Homo sapiens (human)IC50 (µMol)950.00000.00021.557410.0000AID1307967
Peroxisome proliferator-activated receptor gammaHomo sapiens (human)IC50 (µMol)141.50000.00501.205110.0000AID241843; AID242199; AID242406; AID276984; AID277004; AID354041
Peroxisome proliferator-activated receptor alphaHomo sapiens (human)IC50 (µMol)34.12500.00050.82696.3100AID156283; AID223553; AID241842; AID242198; AID242373; AID276983; AID277008; AID354040
Peroxisome proliferator-activated receptor alphaHomo sapiens (human)Ki35.00000.00011.01894.5000AID156292
[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)
Glutamate receptor ionotropic, NMDA 2DHomo sapiens (human)EC50 (µMol)300.00000.02101.27372.8000AID156801
Glutamate receptor ionotropic, NMDA 3BHomo sapiens (human)EC50 (µMol)300.00000.02100.51051.0000AID156801
Fatty acid-binding protein, liverRattus norvegicus (Norway rat)Kd12.64630.01801.78799.6000AID427199; AID427200; AID427203; AID427205; AID427206; AID427207; AID427208; AID427209; AID427210; AID427211; AID427212; AID427213; AID427214; AID427215; AID427216
Peroxisome proliferator-activated receptor alphaMus musculus (house mouse)EC50 (µMol)18.00000.00021.397110.0000AID156453
Peroxisome proliferator-activated receptor alphaRattus norvegicus (Norway rat)EC50 (µMol)131.30000.16600.86772.2600AID156459
Peroxisome proliferator-activated receptor gammaHomo sapiens (human)EC50 (µMol)171.88890.00000.992210.0000AID1546883; AID156801; AID1798916; AID240111; AID240253; AID318248; AID320687; AID354043; AID440654
Peroxisome proliferator-activated receptor gammaMus musculus (house mouse)EC50 (µMol)250.00000.00031.654210.0000AID157279
Peroxisome proliferator-activated receptor deltaHomo sapiens (human)EC50 (µMol)60.00000.00020.84609.1000AID1798916; AID280960; AID318249; AID320688
Glutamate receptor ionotropic, NMDA 1Homo sapiens (human)EC50 (µMol)300.00000.02100.35851.0000AID156801
Peroxisome proliferator-activated receptor alphaHomo sapiens (human)EC50 (µMol)24.17870.00061.607410.0000AID1546882; AID156123; AID156146; AID156464; AID1798916; AID240110; AID240227; AID240252; AID265706; AID276983; AID277009; AID280956; AID318247; AID320686; AID354042; AID440652; AID550055; AID590212; AID620697
Glutamate receptor ionotropic, NMDA 2AHomo sapiens (human)EC50 (µMol)300.00000.02100.32421.0000AID156801
Glutamate receptor ionotropic, NMDA 2BHomo sapiens (human)EC50 (µMol)300.00000.02100.51701.0000AID156801
Glutamate receptor ionotropic, NMDA 2CHomo sapiens (human)EC50 (µMol)300.00000.02101.24032.7000AID156801
Chloride channel isoform 1 Rattus norvegicus (Norway rat)EC50 (µMol)30.00000.19000.89501.6000AID440652
Glutamate receptor ionotropic, NMDA 3AHomo sapiens (human)EC50 (µMol)300.00000.02100.51051.0000AID156801
Sigma non-opioid intracellular receptor 1Rattus norvegicus (Norway rat)Kd20.00000.16000.16000.1600AID427214
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (206)

Processvia Protein(s)Taxonomy
startle responseGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
adult locomotory behaviorGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of sensory perception of painGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
cellular response to L-glutamateGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
long-term synaptic potentiationGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
protein insertion into membraneGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
regulation of calcium ion transportGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
calcium ion transmembrane transportGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
modulation of chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
long-chain fatty acid transportFatty acid-binding protein, liverHomo sapiens (human)
positive regulation of fatty acid beta-oxidationFatty acid-binding protein, liverHomo sapiens (human)
response to vitamin B3Fatty acid-binding protein, liverHomo sapiens (human)
negative regulation of apoptotic processFatty acid-binding protein, liverHomo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in apoptotic processFatty acid-binding protein, liverHomo sapiens (human)
intestinal absorptionFatty acid-binding protein, liverHomo sapiens (human)
cellular response to hydrogen peroxideFatty acid-binding protein, liverHomo sapiens (human)
cellular response to hypoxiaFatty acid-binding protein, liverHomo sapiens (human)
cellular oxidant detoxificationFatty acid-binding protein, liverHomo sapiens (human)
fatty acid transportFatty acid-binding protein, liverHomo sapiens (human)
fatty acid metabolic processFatty acid-binding protein, intestinalHomo sapiens (human)
long-chain fatty acid transportFatty acid-binding protein, intestinalHomo sapiens (human)
intestinal lipid absorptionFatty acid-binding protein, intestinalHomo sapiens (human)
fatty acid transportFatty acid-binding protein, intestinalHomo sapiens (human)
prostaglandin biosynthetic processProstaglandin G/H synthase 1Homo sapiens (human)
response to oxidative stressProstaglandin G/H synthase 1Homo sapiens (human)
regulation of blood pressureProstaglandin G/H synthase 1Homo sapiens (human)
cyclooxygenase pathwayProstaglandin G/H synthase 1Homo sapiens (human)
regulation of cell population proliferationProstaglandin G/H synthase 1Homo sapiens (human)
cellular oxidant detoxificationProstaglandin G/H synthase 1Homo sapiens (human)
negative regulation of gene expressionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of cholesterol effluxPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
long-chain fatty acid transportPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of osteoblast differentiationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of smooth muscle cell proliferationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of receptor signaling pathway via STATPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of low-density lipoprotein receptor activityPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of signaling receptor activityPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of gene expressionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of BMP signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of MAP kinase activityPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of adiponectin secretionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of miRNA transcriptionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of cardiac muscle hypertrophy in response to stressPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of connective tissue replacement involved in inflammatory response wound healingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
placenta developmentPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
regulation of transcription by RNA polymerase IIPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
lipid metabolic processPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
activation of cysteine-type endopeptidase activity involved in apoptotic processPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
signal transductionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
G protein-coupled receptor signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
response to nutrientPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
regulation of blood pressurePeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of gene expressionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of gene expressionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
macrophage derived foam cell differentiationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of macrophage derived foam cell differentiationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of cholesterol storagePeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of lipid storagePeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of sequestering of triglyceridePeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of angiogenesisPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
monocyte differentiationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
BMP signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
epithelial cell differentiationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
cellular response to insulin stimulusPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
response to lipidPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
peroxisome proliferator activated receptor signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
glucose homeostasisPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
regulation of circadian rhythmPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
mRNA transcription by RNA polymerase IIPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
lipoprotein transportPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of blood vessel endothelial cell migrationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
innate immune responsePeroxisome proliferator-activated receptor gammaHomo sapiens (human)
cell fate commitmentPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of fat cell differentiationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of DNA-templated transcriptionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of DNA-templated transcriptionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
retinoic acid receptor signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
cell maturationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
rhythmic processPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
white fat cell differentiationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of DNA-binding transcription factor activityPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
lipid homeostasisPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of type II interferon-mediated signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of SMAD protein signal transductionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
regulation of cholesterol transporter activityPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
cellular response to low-density lipoprotein particle stimulusPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
cellular response to hypoxiaPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of mitochondrial fissionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
regulation of cellular response to insulin stimulusPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of extracellular matrix assemblyPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of miRNA transcriptionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of miRNA transcriptionPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of cellular response to transforming growth factor beta stimulusPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of adipose tissue developmentPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of vascular associated smooth muscle cell proliferationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of vascular associated smooth muscle cell apoptotic processPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of vascular endothelial cell proliferationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
positive regulation of fatty acid metabolic processPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
fatty acid metabolic processPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of inflammatory responsePeroxisome proliferator-activated receptor gammaHomo sapiens (human)
cell differentiationPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
hormone-mediated signaling pathwayPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
glucose metabolic processPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
generation of precursor metabolites and energyPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
regulation of transcription by RNA polymerase IIPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
lipid metabolic processPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
fatty acid beta-oxidationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
apoptotic processPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
embryo implantationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
cholesterol metabolic processPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
cell population proliferationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
axon ensheathmentPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
fatty acid catabolic processPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of gene expressionPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
regulation of skeletal muscle satellite cell proliferationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
fatty acid transportPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
intracellular receptor signaling pathwayPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
cell-substrate adhesionPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
cellular response to nutrient levelsPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
wound healingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
steroid hormone mediated signaling pathwayPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of skeletal muscle tissue regenerationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
keratinocyte proliferationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of fat cell differentiationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
negative regulation of myoblast differentiationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
negative regulation of DNA-templated transcriptionPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of DNA-templated transcriptionPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
decidualizationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
negative regulation of epithelial cell proliferationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
keratinocyte migrationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
adipose tissue developmentPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
fat cell proliferationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of fat cell proliferationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
cellular response to hypoxiaPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
energy homeostasisPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
apoptotic signaling pathwayPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
negative regulation of miRNA transcriptionPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
glucose transmembrane transportPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of myoblast proliferationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of fatty acid metabolic processPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
fatty acid metabolic processPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
negative regulation of cholesterol storagePeroxisome proliferator-activated receptor deltaHomo sapiens (human)
cell differentiationPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
negative regulation of inflammatory responsePeroxisome proliferator-activated receptor deltaHomo sapiens (human)
hormone-mediated signaling pathwayPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
cellular response to amyloid-betaGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
monoatomic cation transportGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
visual learningGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of calcium ion transport into cytosolGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
propylene metabolic processGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
regulation of membrane potentialGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
response to ethanolGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
protein heterotetramerizationGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium ion homeostasisGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of reactive oxygen species biosynthetic processGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
response to glycineGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
negative regulation of cytokine production involved in inflammatory responsePeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of reactive oxygen species biosynthetic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of hepatocyte apoptotic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of signaling receptor activityPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of ATP biosynthetic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of transformation of host cell by virusPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
response to hypoxiaPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
gluconeogenesisPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
heart developmentPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
response to nutrientPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
epidermis developmentPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
cellular response to starvationPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
regulation of cellular ketone metabolic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of macrophage derived foam cell differentiationPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of cholesterol storagePeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of sequestering of triglyceridePeroxisome proliferator-activated receptor alphaHomo sapiens (human)
regulation of fatty acid metabolic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
intracellular receptor signaling pathwayPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of fatty acid beta-oxidationPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of appetitePeroxisome proliferator-activated receptor alphaHomo sapiens (human)
response to insulinPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
circadian regulation of gene expressionPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
behavioral response to nicotinePeroxisome proliferator-activated receptor alphaHomo sapiens (human)
wound healingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
lipoprotein metabolic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
regulation of circadian rhythmPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
steroid hormone mediated signaling pathwayPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
response to ethanolPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of gluconeogenesisPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of blood pressurePeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of glycolytic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of DNA-templated transcriptionPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
nitric oxide metabolic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of fatty acid oxidationPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of lipid biosynthetic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of inflammatory responsePeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of cell growth involved in cardiac muscle cell developmentPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
enamel mineralizationPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
cellular response to fructose stimulusPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of miRNA transcriptionPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
negative regulation of leukocyte cell-cell adhesionPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
regulation of fatty acid transportPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
hormone-mediated signaling pathwayPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
fatty acid metabolic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
positive regulation of fatty acid metabolic processPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
cell differentiationPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
cellular response to amyloid-betaGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
startle responseGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
response to amphetamineGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
learning or memoryGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
memoryGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
visual learningGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
response to xenobiotic stimulusGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
response to woundingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
sensory perception of painGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
neurogenesisGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
protein catabolic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
sleepGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
directional locomotionGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
negative regulation of protein catabolic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
dopamine metabolic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
serotonin metabolic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
positive regulation of apoptotic processGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
response to ethanolGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
activation of cysteine-type endopeptidase activityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
protein localization to postsynaptic membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
long-term synaptic potentiationGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
learning or memoryGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
response to ethanolGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
protein heterotetramerizationGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
negative regulation of dendritic spine maintenanceGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
positive regulation of cysteine-type endopeptidase activityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
long-term synaptic potentiationGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
brain developmentGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
response to woundingGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
calcium-mediated signalingGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
directional locomotionGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
negative regulation of protein catabolic processGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
regulation of synaptic plasticityGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
regulation of neuronal synaptic plasticityGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
neuromuscular process controlling balanceGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
positive regulation of synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
calcium ion transmembrane import into cytosolGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
excitatory chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
protein localization to postsynaptic membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
regulation of monoatomic cation transmembrane transportGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
positive regulation of excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
long-term synaptic potentiationGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
excitatory postsynaptic potentialGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
calcium ion transportGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
dendrite developmentGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
response to ethanolGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
rhythmic processGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
prepulse inhibitionGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
negative regulation of dendritic spine developmentGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
calcium ion transmembrane transportGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
presynaptic modulation of chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
ionotropic glutamate receptor signaling pathwayGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
synaptic transmission, glutamatergicGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
modulation of chemical synaptic transmissionGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (73)

Processvia Protein(s)Taxonomy
glutamate-gated receptor activityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
glutamate bindingGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
voltage-gated monoatomic cation channel activityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
calcium channel activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
monoatomic cation channel activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
glycine bindingGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
neurotransmitter receptor activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
glutamate receptor activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
chromatin bindingFatty acid-binding protein, liverHomo sapiens (human)
long-chain fatty acid transmembrane transporter activityFatty acid-binding protein, liverHomo sapiens (human)
protein bindingFatty acid-binding protein, liverHomo sapiens (human)
phospholipid bindingFatty acid-binding protein, liverHomo sapiens (human)
antioxidant activityFatty acid-binding protein, liverHomo sapiens (human)
bile acid bindingFatty acid-binding protein, liverHomo sapiens (human)
oleic acid bindingFatty acid-binding protein, liverHomo sapiens (human)
heterocyclic compound bindingFatty acid-binding protein, liverHomo sapiens (human)
fatty acid bindingFatty acid-binding protein, liverHomo sapiens (human)
long-chain fatty acid transmembrane transporter activityFatty acid-binding protein, intestinalHomo sapiens (human)
fatty acid bindingFatty acid-binding protein, intestinalHomo sapiens (human)
protein bindingFatty acid-binding protein, intestinalHomo sapiens (human)
long-chain fatty acid bindingFatty acid-binding protein, intestinalHomo sapiens (human)
peroxidase activityProstaglandin G/H synthase 1Homo sapiens (human)
prostaglandin-endoperoxide synthase activityProstaglandin G/H synthase 1Homo sapiens (human)
protein bindingProstaglandin G/H synthase 1Homo sapiens (human)
heme bindingProstaglandin G/H synthase 1Homo sapiens (human)
metal ion bindingProstaglandin G/H synthase 1Homo sapiens (human)
oxidoreductase activity, acting on single donors with incorporation of molecular oxygen, incorporation of two atoms of oxygenProstaglandin G/H synthase 1Homo sapiens (human)
transcription cis-regulatory region bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
transcription coregulator bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
nucleic acid bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
DNA bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
chromatin bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
double-stranded DNA bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
DNA-binding transcription factor activityPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
nuclear receptor activityPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
prostaglandin receptor activityPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
protein bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
zinc ion bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
enzyme bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
peptide bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
identical protein bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
sequence-specific DNA bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
nuclear retinoid X receptor bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
arachidonic acid bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
DNA binding domain bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
LBD domain bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
alpha-actinin bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
R-SMAD bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
E-box bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
STAT family protein bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
DNA-binding transcription factor bindingPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
transcription coactivator bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
DNA bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
DNA-binding transcription factor activityPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
nuclear steroid receptor activityPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
nuclear receptor activityPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
protein bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
zinc ion bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
lipid bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
linoleic acid bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
DNA-binding transcription factor bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
sequence-specific double-stranded DNA bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium channel activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
amyloid-beta bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calcium ion bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
calmodulin bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
glycine bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
glutamate bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
protein-containing complex bindingGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
signaling receptor activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
ligand-gated monoatomic ion channel activityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
DNA-binding transcription activator activityPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
transcription coactivator bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
DNA bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
DNA-binding transcription factor activityPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
nuclear steroid receptor activityPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
nuclear receptor activityPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
protein bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
zinc ion bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
lipid bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
phosphatase bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
protein domain specific bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
mitogen-activated protein kinase kinase kinase bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
ubiquitin conjugating enzyme bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
sequence-specific DNA bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
protein-containing complex bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
NFAT protein bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
MDM2/MDM4 family protein bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
DNA-binding transcription factor bindingPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
amyloid-beta bindingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
zinc ion bindingGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
amyloid-beta bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
zinc ion bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
glycine bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
glutamate bindingGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
ligand-gated monoatomic ion channel activity involved in regulation of presynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
glutamate-gated calcium ion channel activityGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
NMDA glutamate receptor activityGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
calcium channel activityGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
protein bindingGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
glycine bindingGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
identical protein bindingGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
protein phosphatase 2A bindingGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
glutamate receptor activityGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
transmitter-gated monoatomic ion channel activity involved in regulation of postsynaptic membrane potentialGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (42)

Processvia Protein(s)Taxonomy
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
presynaptic active zone membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
hippocampal mossy fiber to CA3 synapseGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
glutamatergic synapseGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2DHomo sapiens (human)
neuronal cell bodyGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 3BHomo sapiens (human)
nucleoplasmFatty acid-binding protein, liverHomo sapiens (human)
peroxisomal matrixFatty acid-binding protein, liverHomo sapiens (human)
cytosolFatty acid-binding protein, liverHomo sapiens (human)
apical cortexFatty acid-binding protein, liverHomo sapiens (human)
extracellular exosomeFatty acid-binding protein, liverHomo sapiens (human)
protein-containing complexFatty acid-binding protein, liverHomo sapiens (human)
nucleusFatty acid-binding protein, liverHomo sapiens (human)
cytosolFatty acid-binding protein, liverHomo sapiens (human)
cytosolFatty acid-binding protein, intestinalHomo sapiens (human)
microvillusFatty acid-binding protein, intestinalHomo sapiens (human)
apical cortexFatty acid-binding protein, intestinalHomo sapiens (human)
cytosolFatty acid-binding protein, intestinalHomo sapiens (human)
nucleusFatty acid-binding protein, intestinalHomo sapiens (human)
nucleoplasmPeroxisome proliferator-activated receptor alphaMus musculus (house mouse)
photoreceptor outer segmentProstaglandin G/H synthase 1Homo sapiens (human)
cytoplasmProstaglandin G/H synthase 1Homo sapiens (human)
endoplasmic reticulum membraneProstaglandin G/H synthase 1Homo sapiens (human)
Golgi apparatusProstaglandin G/H synthase 1Homo sapiens (human)
intracellular membrane-bounded organelleProstaglandin G/H synthase 1Homo sapiens (human)
extracellular exosomeProstaglandin G/H synthase 1Homo sapiens (human)
cytoplasmProstaglandin G/H synthase 1Homo sapiens (human)
neuron projectionProstaglandin G/H synthase 1Homo sapiens (human)
nucleusPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
nucleusPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
nucleoplasmPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
cytosolPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
intracellular membrane-bounded organellePeroxisome proliferator-activated receptor gammaHomo sapiens (human)
RNA polymerase II transcription regulator complexPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
chromatinPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
receptor complexPeroxisome proliferator-activated receptor gammaHomo sapiens (human)
nucleoplasmPeroxisome proliferator-activated receptor gammaMus musculus (house mouse)
nucleusPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
nucleusPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
nucleoplasmPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
chromatinPeroxisome proliferator-activated receptor deltaHomo sapiens (human)
cytoplasmGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synaptic vesicleGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
cell surfaceGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
postsynaptic densityGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
dendriteGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
neuron projectionGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synaptic cleftGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
terminal boutonGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
dendritic spineGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synapseGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
excitatory synapseGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synaptic membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
synapseGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
neuron projectionGlutamate receptor ionotropic, NMDA 1Homo sapiens (human)
nucleusPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
nucleoplasmPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
chromatinPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
nucleusPeroxisome proliferator-activated receptor alphaHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
synaptic vesicleGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
cell surfaceGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
postsynaptic densityGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
cytoplasmic vesicle membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
presynaptic membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
dendritic spineGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
synaptic membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
glutamatergic synapseGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 2AHomo sapiens (human)
cytoplasmGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
lysosomeGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
late endosomeGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
cytoskeletonGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
cell surfaceGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
postsynaptic densityGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
neuron projectionGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
synaptic membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 2BHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
postsynaptic membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
glutamatergic synapseGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 2CHomo sapiens (human)
endoplasmic reticulum membraneGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
membraneGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
neuron projectionGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
neuronal cell bodyGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
synapseGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
presynapseGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
glutamatergic synapseGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
NMDA selective glutamate receptor complexGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
plasma membraneGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
postsynaptic density membraneGlutamate receptor ionotropic, NMDA 3AHomo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (150)

Assay IDTitleYearJournalArticle
AID407369Displacement of 1-anilinonaphthalene-8-sulphonic acid from rat recombinant L-FABP low binding affinity site expressed in Escherichia coli BL21 by competitive fluorescence displacement assay2008Journal of medicinal chemistry, Jul-10, Volume: 51, Issue:13
Characterization of the drug binding specificity of rat liver fatty acid binding protein.
AID280959Activity at human adipose tissue PPAR gamma expressed in HEK293 cells by PPAR-GAL4 transactivation assay relative to rosiglitazone2007Journal of medicinal chemistry, Apr-05, Volume: 50, Issue:7
Identification and synthesis of a novel selective partial PPARdelta agonist with full efficacy on lipid metabolism in vitro and in vivo.
AID280956Activity at human liver PPAR alpha expressed in HEK293 cells by PPAR-GAL4 transactivation assay2007Journal of medicinal chemistry, Apr-05, Volume: 50, Issue:7
Identification and synthesis of a novel selective partial PPARdelta agonist with full efficacy on lipid metabolism in vitro and in vivo.
AID223556Binding affinity at human PPAR gamma2001Journal of medicinal chemistry, Jun-21, Volume: 44, Issue:13
Design and synthesis of 2-methyl-2-[4-(2-[5-methyl-2-aryloxazol-4-yl]ethoxy)phenoxy]propionic acids: a new class of dual PPARalpha/gamma agonists.
AID242198Inhibition of human Peroxisome proliferator activated receptor alpha binding2005Journal of medicinal chemistry, Apr-07, Volume: 48, Issue:7
Discovery of a novel series of peroxisome proliferator-activated receptor alpha/gamma dual agonists for the treatment of type 2 diabetes and dyslipidemia.
AID156944In vitro transactivation of human Peroxisome proliferator activated receptor gamma; Not calculated for transactivation <25% at 30 uM2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID590212Partial agonist activity at human PPARalpha expressed in CHO cells co-transfected with Gal4-responsive luciferase reporter plasmid after 24 hrs by transactivation assay2011Bioorganic & medicinal chemistry letters, Apr-01, Volume: 21, Issue:7
Synthesis and pharmacological evaluation of novel benzoylazole-based PPAR α/γ activators.
AID280963Effect on fatty acid oxidation in rat L6 cells2007Journal of medicinal chemistry, Apr-05, Volume: 50, Issue:7
Identification and synthesis of a novel selective partial PPARdelta agonist with full efficacy on lipid metabolism in vitro and in vivo.
AID156123Compound was tested for agonist activity on human Peroxisome proliferator activated receptor alpha-Gal4 chimeric receptor in transfected CV-1 cells1999Journal of medicinal chemistry, Sep-23, Volume: 42, Issue:19
A ureido-thioisobutyric acid (GW9578) is a subtype-selective PPARalpha agonist with potent lipid-lowering activity.
AID318248Agonist activity at human PPARgamma expressed in monkey CV1 cells by transactivation assay2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Discovery of a novel class of 1,3-dioxane-2-carboxylic acid derivatives as subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID354042Agonist activity at human PPARalpha by luciferase reporter transactivation assay2009Bioorganic & medicinal chemistry letters, May-01, Volume: 19, Issue:9
Aleglitazar, a new, potent, and balanced dual PPARalpha/gamma agonist for the treatment of type II diabetes.
AID280957Activity at human liver PPAR alpha expressed in HEK293 cells by GAL4 transactivation assay relative to NNC61-46552007Journal of medicinal chemistry, Apr-05, Volume: 50, Issue:7
Identification and synthesis of a novel selective partial PPARdelta agonist with full efficacy on lipid metabolism in vitro and in vivo.
AID280960Activity at human placenta PPAR delta expressed in HEK293 cells by PPAR-GAL4 transactivation assay2007Journal of medicinal chemistry, Apr-05, Volume: 50, Issue:7
Identification and synthesis of a novel selective partial PPARdelta agonist with full efficacy on lipid metabolism in vitro and in vivo.
AID112794Area under blood glucose time curve after oral glucose test in mice2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID241843Inhibition of human Peroxisome proliferator activated receptor gamma2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design and synthesis of N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5- methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]glycine [Muraglitazar/BMS-298585], a novel peroxisome proliferator-activated receptor alpha/gamma dual agonist with efficacious glucose and
AID240252Effective concentration against human Peroxisome proliferator activated receptor alpha in Gal4 transactivation assay2005Journal of medicinal chemistry, Apr-07, Volume: 48, Issue:7
Discovery of a novel series of peroxisome proliferator-activated receptor alpha/gamma dual agonists for the treatment of type 2 diabetes and dyslipidemia.
AID1774078Stabilization of TTR V3OM mutant (unknown origin) assessed as acid-mediated protein aggregation inhibition ratio at 4 uM incubated for 1 week by absorbance method2021Journal of medicinal chemistry, 10-14, Volume: 64, Issue:19
Repositioning of the Anthelmintic Drugs Bithionol and Triclabendazole as Transthyretin Amyloidogenesis Inhibitors.
AID241842Inhibition of human Peroxisome proliferator activated receptor alpha2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design and synthesis of N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5- methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]glycine [Muraglitazar/BMS-298585], a novel peroxisome proliferator-activated receptor alpha/gamma dual agonist with efficacious glucose and
AID242199Inhibition of human Peroxisome proliferator activated receptor gamma binding2005Journal of medicinal chemistry, Apr-07, Volume: 48, Issue:7
Discovery of a novel series of peroxisome proliferator-activated receptor alpha/gamma dual agonists for the treatment of type 2 diabetes and dyslipidemia.
AID354043Agonist activity at human PPARgamma expressed in BHK21 cells assessed as SEAP activity by luciferase reporter transactivation assay2009Bioorganic & medicinal chemistry letters, May-01, Volume: 19, Issue:9
Aleglitazar, a new, potent, and balanced dual PPARalpha/gamma agonist for the treatment of type II diabetes.
AID277006Activity at human PPARgamma in CV1 cells at 10 uM relative to 2-methyl-2-(4-{3-propyl-(5-pyridin-2yl-thiophene-2-sulphonyl)-amino]-pro-pyl}-phenoxy)-propionic acid2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Synthesis and evaluation of aminomethyl dihydrocinnamates as a new class of PPAR ligands.
AID156464Cotransfection activity of compound against human Peroxisome proliferator activated receptor alpha was determined2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of a potent and selective triazolone-based peroxisome proliferator-activated receptor alpha agonist.
AID320703Hypolipidemic effect on diabetic KK-Ay mouse assessed as reduction of plasma VLDL cholesterol level at 300 mg/kg/day, po after 4 days2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Structure-activity studies on 1,3-dioxane-2-carboxylic acid derivatives, a novel class of subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID223551Binding affinity at human PPAR alpha2001Journal of medicinal chemistry, Jun-21, Volume: 44, Issue:13
Design and synthesis of 2-methyl-2-[4-(2-[5-methyl-2-aryloxazol-4-yl]ethoxy)phenoxy]propionic acids: a new class of dual PPARalpha/gamma agonists.
AID746618Agonist activity at PPARgamma (unknown origin) transfected in HEK293 cells at 10 uM by luciferase reporter gene assay2013Bioorganic & medicinal chemistry letters, May-15, Volume: 23, Issue:10
Synthesis and evaluation of 2,3-dinorprostaglandins: Dinor-PGD1 and 13-epi-dinor-PGD1 are peroxisome proliferator-activated receptor α/γ dual agonists.
AID1307967Inhibition of human recombinant COX1 expressed in Sf9 cell microsomes assessed as reduction in conversion of arachidonic acid to PGE2 incubated for 5 mins by HTRF assay2016Journal of medicinal chemistry, 05-12, Volume: 59, Issue:9
Impact of Binding Site Comparisons on Medicinal Chemistry and Rational Molecular Design.
AID276986Activity at human PPARalpha expressed in CV1 cells using GAL4 chimeric system relative to control2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Tetrahydroisoquinoline PPARgamma agonists: design of novel, highly selective non-TZD antihyperglycemic agents.
AID427207Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) at 30 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID156615In vitro transactivation of human Peroxisome proliferator activated receptor delta (hPPARdelta); Not calculated for transactivation <25% at 30 uM2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID427210Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) at 25 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID128559Brown adipose tissue weight in db/db mouse after 150 mg/kg peroral administration2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID156460In vitro transactivation of rat Peroxisome proliferator activated receptor alpha2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID1307966Inhibition of human recombinant COX1 expressed in Sf9 cell microsomes assessed as reduction in conversion of arachidonic acid to PGE2 at 250 uM incubated for 5 mins by HTRF assay2016Journal of medicinal chemistry, 05-12, Volume: 59, Issue:9
Impact of Binding Site Comparisons on Medicinal Chemistry and Rational Molecular Design.
AID277007Activity at human PPARgamma in CV1 cells by CTF assay relative to 2-methyl-2-(4-{3-propyl-(5-pyridin-2yl-thiophene-2-sulphonyl)-amino]-pro-pyl}-phenoxy)-propionic acid2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Synthesis and evaluation of aminomethyl dihydrocinnamates as a new class of PPAR ligands.
AID427212Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) at 20 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID185029Minimum effective dose producing a 40-60% decrease in serum TLDL cholesterol in rats relative to vehicle -treated controls1999Journal of medicinal chemistry, Sep-23, Volume: 42, Issue:19
A ureido-thioisobutyric acid (GW9578) is a subtype-selective PPARalpha agonist with potent lipid-lowering activity.
AID115313Area under blood glucose time curve after oral glucose test in mice2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID427208Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) at 30 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID156786Compound was tested for its agonist activity against murine Peroxisome proliferator activated receptor delta-GAL4 chimeric receptor in transfected CV-1 cells; IA = Inactive1999Journal of medicinal chemistry, Sep-23, Volume: 42, Issue:19
A ureido-thioisobutyric acid (GW9578) is a subtype-selective PPARalpha agonist with potent lipid-lowering activity.
AID1774079Stabilization of TTR V3OM mutant (unknown origin) assessed as acid-mediated protein aggregation inhibition ratio at 10 uM incubated for 1 week by absorbance method2021Journal of medicinal chemistry, 10-14, Volume: 64, Issue:19
Repositioning of the Anthelmintic Drugs Bithionol and Triclabendazole as Transthyretin Amyloidogenesis Inhibitors.
AID27582Partition coefficient (logP)2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID276983Displacement of tritium labeled ligand from human PPARalpha by SPA assay2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Tetrahydroisoquinoline PPARgamma agonists: design of novel, highly selective non-TZD antihyperglycemic agents.
AID156292Binding affinity towards peroxisome proliferator activated receptor alpha (PPAR alpha)2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID223557Transcriptional activation in CV- cells expressing hPPARgamma2001Journal of medicinal chemistry, Jun-21, Volume: 44, Issue:13
Design and synthesis of 2-methyl-2-[4-(2-[5-methyl-2-aryloxazol-4-yl]ethoxy)phenoxy]propionic acids: a new class of dual PPARalpha/gamma agonists.
AID320702Hypoglycemic effect on diabetic KK-Ay mouse assessed as reduction of plasma glucose level at 300 mg/kg/day, po after 4 days2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Structure-activity studies on 1,3-dioxane-2-carboxylic acid derivatives, a novel class of subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID1474166Liver toxicity in human assessed as induction of drug-induced liver injury by measuring severity class index2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID320701Hypolipidemic effect on diabetic KK-Ay mouse assessed as reduction of plasma triglyceride level at 300 mg/kg/day, po after 4 days2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Structure-activity studies on 1,3-dioxane-2-carboxylic acid derivatives, a novel class of subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID354040Displacement of radio labeled 2(S)-(2-benzoyl-phenylamino)-3-{4-[1,1-ditritio-2-(5-methyl-2-phenyl-oxazol-4-yl)-ethoxy]-phenyl}-propionic acid from GST-fused human PPARalpha expressed in Escherichia coli BL21 cells2009Bioorganic & medicinal chemistry letters, May-01, Volume: 19, Issue:9
Aleglitazar, a new, potent, and balanced dual PPARalpha/gamma agonist for the treatment of type II diabetes.
AID115315In vivo nonfasting blood glucose in db/db mice after oral treatment2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID440654Agonist activity at GAL4-tagged human PPARgamma ligand binding domain expressed in human HepG2 cells assessed as receptor transactivation by luciferase reporter gene assay2009Journal of medicinal chemistry, Oct-22, Volume: 52, Issue:20
New 2-aryloxy-3-phenyl-propanoic acids as peroxisome proliferator-activated receptors alpha/gamma dual agonists with improved potency and reduced adverse effects on skeletal muscle function.
AID240253Effective concentration against human Peroxisome proliferator activated receptor gamma in Gal4 transactivation assay2005Journal of medicinal chemistry, Apr-07, Volume: 48, Issue:7
Discovery of a novel series of peroxisome proliferator-activated receptor alpha/gamma dual agonists for the treatment of type 2 diabetes and dyslipidemia.
AID354045Agonist activity at human PPARgamma expressed in BHK21 cells assessed as SEAP activity by luciferase reporter transactivation assay relative to edaglitazone2009Bioorganic & medicinal chemistry letters, May-01, Volume: 19, Issue:9
Aleglitazar, a new, potent, and balanced dual PPARalpha/gamma agonist for the treatment of type II diabetes.
AID115314In vivo insulin effect in db/db mice after oral treatment2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID427214Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) at 15 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID590213Partial agonist activity at human PPARalpha expressed in CHO cells co-transfected with Gal4-responsive luciferase reporter plasmid after 24 hrs by transactivation assay relative to fenofibric acid2011Bioorganic & medicinal chemistry letters, Apr-01, Volume: 21, Issue:7
Synthesis and pharmacological evaluation of novel benzoylazole-based PPAR α/γ activators.
AID620698Binding affinity to histidine-tagged human PPARalpha-LBD by SPR analysis2011Bioorganic & medicinal chemistry letters, Oct-01, Volume: 21, Issue:19
Ursolic acid is a PPAR-α agonist that regulates hepatic lipid metabolism.
AID320704Hypolipidemic effect on diabetic KK-Ay mouse assessed as increase in plasma HDL cholesterol level at 300 mg/kg/day, po after 4 days2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Structure-activity studies on 1,3-dioxane-2-carboxylic acid derivatives, a novel class of subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID132139Heart weight of db/db mouse after 150 mg/kg peroral administration2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID242373Mean inhibitory concentration against human peroxisome proliferator activated receptor alpha2005Bioorganic & medicinal chemistry letters, Jan-03, Volume: 15, Issue:1
2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.
AID277008Displacement of [3H]2-(4-{2-[3-(2,4-difluoro-phenyl)-1-heptyl-ureido]-ethyl}-phenoxy)-2-methyl-butyric acid from hPPARalpha2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Synthesis and evaluation of aminomethyl dihydrocinnamates as a new class of PPAR ligands.
AID427205Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) at 37 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID427202Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) assessed as occupancy at a drug-protein molar ratio of 3:1 by NMR chemical shift perturbation method2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID550054Agonist activity at human PPARalpha ligand binding domain expressed in COS-1 cells co-transfected with Gal4 at 10 uM by luciferase reporter gene assay relative to control2011Bioorganic & medicinal chemistry letters, Jan-01, Volume: 21, Issue:1
Development of a new class of benzoylpyrrole-based PPARα/γ activators.
AID277009Activity at human PPARalpha in CV1 cells2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Synthesis and evaluation of aminomethyl dihydrocinnamates as a new class of PPAR ligands.
AID320688Agonist activity at human PPARdelta expressed in CV1 cells by transactivation assay2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Structure-activity studies on 1,3-dioxane-2-carboxylic acid derivatives, a novel class of subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID1774076Inhibition of 8-anilinonaphthalene-l-sulfonic acid binding to TTR V3OM mutant (unknown origin) expressed in Escherichia coli at 400 uM incubated for 1 hr in presence of 75 uM ANS by fluorescence method (Rvb = 91 +/- 0.92%)2021Journal of medicinal chemistry, 10-14, Volume: 64, Issue:19
Repositioning of the Anthelmintic Drugs Bithionol and Triclabendazole as Transthyretin Amyloidogenesis Inhibitors.
AID277004Displacement of [3H]2-methyl-2-(4-{3-propyl-(5-pyridin-2yl-thiophene-2-sulfonyl)-amino]-pro-pyl}-phenoxy)-propionic acid from human PPARgamma2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Synthesis and evaluation of aminomethyl dihydrocinnamates as a new class of PPAR ligands.
AID440655Inhibition of CIC1 in Wistar rat extensor digitorum longus muscle assessed as decrease in resting chloride channel conductance at 100 uM by electrophysiology method2009Journal of medicinal chemistry, Oct-22, Volume: 52, Issue:20
New 2-aryloxy-3-phenyl-propanoic acids as peroxisome proliferator-activated receptors alpha/gamma dual agonists with improved potency and reduced adverse effects on skeletal muscle function.
AID113350In vivo nonfasting triglyceride in db/db mice after oral treatment2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID156449Inhibitory activity of compound against the binding of human Peroxisome proliferator activated receptor delta was determined; NB means no binding2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of a potent and selective triazolone-based peroxisome proliferator-activated receptor alpha agonist.
AID157112Inhibitory activity of compound against the binding of human Peroxisome proliferator activated receptor gamma was determined; NB means no binding2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of a potent and selective triazolone-based peroxisome proliferator-activated receptor alpha agonist.
AID1546883Agonist activity at PPARgamma (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
The Race to Bash NASH: Emerging Targets and Drug Development in a Complex Liver Disease.
AID127565Total Cholesterol change in db/db mouse after 150 mg/kg peroral administration2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID1547176Protection against STZ-induced diabetic retinopathy Brown Norway rat model assessed as reduction in vascular leakage by measuring retinal vascular permeability at 23 mg/kg, ip administered daily for 4 weeks starting at 2 weeks post STZ challenge measured 2020Journal of medicinal chemistry, 03-26, Volume: 63, Issue:6
Evolution of a 4-Benzyloxy-benzylamino Chemotype to Provide Efficacious, Potent, and Isoform Selective PPARα Agonists as Leads for Retinal Disorders.
AID354041Displacement of radio labeled 2(S)-(2-benzoyl-phenylamino)-3-{4-[1,1-ditritio-2-(5-methyl-2-phenyl-oxazol-4-yl)-ethoxy]-phenyl}-propionic acid from GST-fused human PPARgamma expressed in Escherichia coli BL21 cells by scintillation proximity assay2009Bioorganic & medicinal chemistry letters, May-01, Volume: 19, Issue:9
Aleglitazar, a new, potent, and balanced dual PPARalpha/gamma agonist for the treatment of type II diabetes.
AID715914Transactivation of PPAR-alpha transfected in HEK293 cells at 10 uM after 24 hrs by luciferase reporter gene assay2012Bioorganic & medicinal chemistry letters, Jun-15, Volume: 22, Issue:12
Ethyl 2,4,6-trihydroxybenzoate is an agonistic ligand for liver X receptor that induces cholesterol efflux from macrophages without affecting lipid accumulation in HepG2 cells.
AID427216Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) at 10 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID240111Effective concentration against human Peroxisome proliferator activated receptor gamma2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design and synthesis of N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5- methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]glycine [Muraglitazar/BMS-298585], a novel peroxisome proliferator-activated receptor alpha/gamma dual agonist with efficacious glucose and
AID156459In vitro transactivation of rat Peroxisome proliferator activated receptor alpha2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID715913Transactivation of PPAR-gamma transfected in HEK293 cells at 10 uM after 24 hrs by luciferase reporter gene assay2012Bioorganic & medicinal chemistry letters, Jun-15, Volume: 22, Issue:12
Ethyl 2,4,6-trihydroxybenzoate is an agonistic ligand for liver X receptor that induces cholesterol efflux from macrophages without affecting lipid accumulation in HepG2 cells.
AID129088Body weight of db/db mouse after 150 mg/kg peroral administration2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID620705Induction of BODIPY-labeled 4,4-difluoro-5-methyl-4-bora-3a,4a-diaza-s-indacene-3-dodecanoic acid uptake in human HepG2 cells after 1 min by FACS analysis2011Bioorganic & medicinal chemistry letters, Oct-01, Volume: 21, Issue:19
Ursolic acid is a PPAR-α agonist that regulates hepatic lipid metabolism.
AID113348In vivo insulin effect in db/db mice after oral treatment2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID156618Cotransfection activity of compound against human Peroxisome proliferator activated receptor delta was determined at 10 uM2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of a potent and selective triazolone-based peroxisome proliferator-activated receptor alpha agonist.
AID156619In vitro transactivation of human Peroxisome proliferator activated receptor delta (hPPARdelta)2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID239802Mean percent of maximum efficacy against human peroxisome proliferator-activated receptor alpha2005Bioorganic & medicinal chemistry letters, Jan-03, Volume: 15, Issue:1
2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.
AID156948Cotransfection activity of compound against human Peroxisome proliferator activated receptor gamma was determined at 10 uM2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of a potent and selective triazolone-based peroxisome proliferator-activated receptor alpha agonist.
AID427211Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) at 20 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID242406Mean inhibitory concentration against human peroxisome proliferator-activated receptor gamma2005Bioorganic & medicinal chemistry letters, Jan-03, Volume: 15, Issue:1
2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.
AID1546882Agonist activity at PPARalpha (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
The Race to Bash NASH: Emerging Targets and Drug Development in a Complex Liver Disease.
AID620697Binding affinity to histidine-tagged human PPARalpha-LBD assessed as recruitment of co-activator peptide fluorescein-labeled PGC1alpha after 2 hrs by TR-FRET assay2011Bioorganic & medicinal chemistry letters, Oct-01, Volume: 21, Issue:19
Ursolic acid is a PPAR-α agonist that regulates hepatic lipid metabolism.
AID427204Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) at 42 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID280964Effect on increase in fatty acid oxidation in rat L6 cells relative to GW-5015162007Journal of medicinal chemistry, Apr-05, Volume: 50, Issue:7
Identification and synthesis of a novel selective partial PPARdelta agonist with full efficacy on lipid metabolism in vitro and in vivo.
AID115316In vivo nonfasting triglyceride in db/db mice after oral treatment2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID265706Agonist activity at human recombinant PPARalpha by transactivation of TK-MH100x4-LUC reporter gene in HEK293 cells2006Bioorganic & medicinal chemistry letters, Jun-15, Volume: 16, Issue:12
Diphenylmethane skeleton as a multi-template for nuclear receptor ligands: preparation of FXR and PPAR ligands.
AID246948In vivo effective dose value in NIDDM animal model (ZDF rat); (not tested)2005Bioorganic & medicinal chemistry letters, Jan-03, Volume: 15, Issue:1
2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.
AID407366Displacement of 1-anilinonaphthalene-8-sulphonic acid from rat recombinant L-FABP high binding affinity site expressed in Escherichia coli BL21 by competitive fluorescence displacement assay2008Journal of medicinal chemistry, Jul-10, Volume: 51, Issue:13
Characterization of the drug binding specificity of rat liver fatty acid binding protein.
AID280961Activity at human placenta PPAR delta expressed in HEK293 cells by PPAR-GAL4 transactivation assay relative to carbacyclin2007Journal of medicinal chemistry, Apr-05, Volume: 50, Issue:7
Identification and synthesis of a novel selective partial PPARdelta agonist with full efficacy on lipid metabolism in vitro and in vivo.
AID130223Triglyceride change in db/db mouse after 150 mg/kg peroral administration2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID156149In vitro transactivation of human Peroxisome proliferator activated receptor alpha (hPPARalpha)2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID276987Activity at human PPARgamma expressed in CV1 cells by cotransfection assay relative to control2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Tetrahydroisoquinoline PPARgamma agonists: design of novel, highly selective non-TZD antihyperglycemic agents.
AID131593Fasting plasma glucose levels after 150 mg/kg peroral administration in db/db Mouse2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID427206Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) at 37 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID427203Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) at 42 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID320686Agonist activity at human PPARalpha expressed in CV1 cells by transactivation assay2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Structure-activity studies on 1,3-dioxane-2-carboxylic acid derivatives, a novel class of subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID318249Agonist activity at human PPARdelta expressed in monkey CV1 cells by transactivation assay2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Discovery of a novel class of 1,3-dioxane-2-carboxylic acid derivatives as subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID354070Increase in plasma HDLC level in human ApoA1 transgenic mouse model at 300 mg/kg/day after 12 days by FPLC method2009Bioorganic & medicinal chemistry letters, May-01, Volume: 19, Issue:9
Aleglitazar, a new, potent, and balanced dual PPARalpha/gamma agonist for the treatment of type II diabetes.
AID1474167Liver toxicity in human assessed as induction of drug-induced liver injury by measuring verified drug-induced liver injury concern status2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID244407Intrinsic activity against Peroxisome proliferator activated receptors at 1 uM relative to 1 uM GW-2331 for PPARalpha or rosiglitazone for PPARgamma2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design and synthesis of N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5- methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]glycine [Muraglitazar/BMS-298585], a novel peroxisome proliferator-activated receptor alpha/gamma dual agonist with efficacious glucose and
AID590196Partial agonist activity at human PPARgamma expressed in CHO cells co-transfected with Gal4-responsive luciferase reporter plasmid after 24 hrs by transactivation assay2011Bioorganic & medicinal chemistry letters, Apr-01, Volume: 21, Issue:7
Synthesis and pharmacological evaluation of novel benzoylazole-based PPAR α/γ activators.
AID156146In vitro transactivation of human Peroxisome proliferator activated receptor alpha (hPPARalpha)2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID427199Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) at 5 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID427201Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) assessed as occupancy at a drug-protein molar ratio of 3:1 by NMR chemical shift perturbation method2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID550055Agonist activity at human PPARalpha ligand binding domain expressed in COS-1 cells co-transfected with Gal4 by luciferase reporter gene assay2011Bioorganic & medicinal chemistry letters, Jan-01, Volume: 21, Issue:1
Development of a new class of benzoylpyrrole-based PPARα/γ activators.
AID427213Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) at 15 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID427209Binding affinity to rat recombinant L-FABP low affinity site expressed in Escherichia coli BL21(DE3) at 25 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID427200Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) at 5 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID223559Binding affinity at human peroxidase proliferator activated receptor gamma (hPPARgamma); No binding2001Journal of medicinal chemistry, Jun-21, Volume: 44, Issue:13
Design and synthesis of 2-methyl-2-[4-(2-[5-methyl-2-aryloxazol-4-yl]ethoxy)phenoxy]propionic acids: a new class of dual PPARalpha/gamma agonists.
AID223552Transcriptional activation in CV-1 cells expressing hPPARalpha2001Journal of medicinal chemistry, Jun-21, Volume: 44, Issue:13
Design and synthesis of 2-methyl-2-[4-(2-[5-methyl-2-aryloxazol-4-yl]ethoxy)phenoxy]propionic acids: a new class of dual PPARalpha/gamma agonists.
AID440652Agonist activity at GAL4-tagged human PPARalpha ligand binding domain expressed in human HepG2 cells assessed as receptor transactivation by luciferase reporter gene assay2009Journal of medicinal chemistry, Oct-22, Volume: 52, Issue:20
New 2-aryloxy-3-phenyl-propanoic acids as peroxisome proliferator-activated receptors alpha/gamma dual agonists with improved potency and reduced adverse effects on skeletal muscle function.
AID354044Agonist activity at human PPARalpha by luciferase reporter transactivation assay relative to GW-2625702009Bioorganic & medicinal chemistry letters, May-01, Volume: 19, Issue:9
Aleglitazar, a new, potent, and balanced dual PPARalpha/gamma agonist for the treatment of type II diabetes.
AID1307968Selectivity index, ratio of IC50 for human recombinant COX2 to IC50 for human recombinant COX12016Journal of medicinal chemistry, 05-12, Volume: 59, Issue:9
Impact of Binding Site Comparisons on Medicinal Chemistry and Rational Molecular Design.
AID746617Agonist activity at PPARdelta (unknown origin) transfected in HEK293 cells at 10 uM by luciferase reporter gene assay2013Bioorganic & medicinal chemistry letters, May-15, Volume: 23, Issue:10
Synthesis and evaluation of 2,3-dinorprostaglandins: Dinor-PGD1 and 13-epi-dinor-PGD1 are peroxisome proliferator-activated receptor α/γ dual agonists.
AID156949In vitro transactivation of human Peroxisome proliferator activated receptor gamma2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID407368Displacement of 1-anilinonaphthalene-8-sulphonic acid from I-FABP2008Journal of medicinal chemistry, Jul-10, Volume: 51, Issue:13
Characterization of the drug binding specificity of rat liver fatty acid binding protein.
AID156453Compound was tested for its agonist activity against murine Peroxisome proliferator activated receptor alpha-Gal4 chimeric receptor transfected CV-1 cells1999Journal of medicinal chemistry, Sep-23, Volume: 42, Issue:19
A ureido-thioisobutyric acid (GW9578) is a subtype-selective PPARalpha agonist with potent lipid-lowering activity.
AID240227Mean effective concentration against human peroxisome proliferator activated receptor alpha2005Bioorganic & medicinal chemistry letters, Jan-03, Volume: 15, Issue:1
2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.
AID157279Compound was tested for its agonist activity against murine Peroxisome proliferator activated receptor gamma-Gal4 chimeric receptor in transfected CV-1 cells1999Journal of medicinal chemistry, Sep-23, Volume: 42, Issue:19
A ureido-thioisobutyric acid (GW9578) is a subtype-selective PPARalpha agonist with potent lipid-lowering activity.
AID156801Compound was tested for agonist activity on human Peroxisome proliferator activated receptor gamma-Gal4 chimeric receptor in transfected CV-1 cells1999Journal of medicinal chemistry, Sep-23, Volume: 42, Issue:19
A ureido-thioisobutyric acid (GW9578) is a subtype-selective PPARalpha agonist with potent lipid-lowering activity.
AID1774075Inhibition of 8-anilinonaphthalene-l-sulfonic acid binding to TTR V3OM mutant (unknown origin) expressed in Escherichia coli assessed as ANS saturation ratio at 400 uM incubated for 1 hr in presence of 7.5 uM ANS by fluorescence method (Rvb = 56 +/- 2.3%)2021Journal of medicinal chemistry, 10-14, Volume: 64, Issue:19
Repositioning of the Anthelmintic Drugs Bithionol and Triclabendazole as Transthyretin Amyloidogenesis Inhibitors.
AID1547179Toxicity in STZ-induced diabetic retinopathy Brown Norway rat model assessed as liver weight at 23 mg/kg, ip administered daily for 4 weeks starting at 2 weeks post STZ challenge relative to body weight (Rvb = 4.36 +/- 0.37%)2020Journal of medicinal chemistry, 03-26, Volume: 63, Issue:6
Evolution of a 4-Benzyloxy-benzylamino Chemotype to Provide Efficacious, Potent, and Isoform Selective PPARα Agonists as Leads for Retinal Disorders.
AID157104Binding affinity towards peroxisome proliferator activated receptor gamma (PPAR gamma); Not active2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID113349In vivo nonfasting blood glucose in db/db mice after oral treatment2003Journal of medicinal chemistry, Nov-06, Volume: 46, Issue:23
Large dimeric ligands with favorable pharmacokinetic properties and peroxisome proliferator-activated receptor agonist activity in vitro and in vivo.
AID239803Mean percent of maximum efficacy against human peroxisome proliferator-activated receptor gamma2005Bioorganic & medicinal chemistry letters, Jan-03, Volume: 15, Issue:1
2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.
AID277011Activity at human PPARalpha in CV1 cells by CTF assay relative to 2-(4-{2-[3-(2,4-difluoro-phenyl)-1-heptyl-ureido]-ethyl}-phenoxy)-2-methyl-butyric acid2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Synthesis and evaluation of aminomethyl dihydrocinnamates as a new class of PPAR ligands.
AID427215Binding affinity to rat recombinant L-FABP high affinity site expressed in Escherichia coli BL21(DE3) at 10 deg C by fluorimetric assay2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Probing the fibrate binding specificity of rat liver fatty acid binding protein.
AID223553Binding affinity towards human peroxidase proliferator activated receptor alpha (hPPARalpha)2001Journal of medicinal chemistry, Jun-21, Volume: 44, Issue:13
Design and synthesis of 2-methyl-2-[4-(2-[5-methyl-2-aryloxazol-4-yl]ethoxy)phenoxy]propionic acids: a new class of dual PPARalpha/gamma agonists.
AID156283Inhibitory activity against human Peroxisome proliferator activated receptor alpha2003Journal of medicinal chemistry, Nov-20, Volume: 46, Issue:24
Design and synthesis of a potent and selective triazolone-based peroxisome proliferator-activated receptor alpha agonist.
AID156291Binding affinity towards human peroxisome proliferator activated receptor alpha (PPAR alpha) by HTRF assay; Weakly active2003Bioorganic & medicinal chemistry letters, Oct-06, Volume: 13, Issue:19
O-arylmandelic acids as highly selective human PPAR alpha/gamma agonists.
AID276984Displacement of tritium labeled ligand from human PPARgamma by SPA assay2006Bioorganic & medicinal chemistry letters, Dec-15, Volume: 16, Issue:24
Tetrahydroisoquinoline PPARgamma agonists: design of novel, highly selective non-TZD antihyperglycemic agents.
AID318247Agonist activity at human PPARalpha expressed in monkey CV1 cells by transactivation assay2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Discovery of a novel class of 1,3-dioxane-2-carboxylic acid derivatives as subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID156601Compound was tested for agonist activity on human Peroxisome proliferator activated receptor delta-GAL4 chimeric receptor in transfected CV-1 cells; IA = Inactive1999Journal of medicinal chemistry, Sep-23, Volume: 42, Issue:19
A ureido-thioisobutyric acid (GW9578) is a subtype-selective PPARalpha agonist with potent lipid-lowering activity.
AID240110Effective concentration against human Peroxisome proliferator activated receptor alpha2005Journal of medicinal chemistry, Mar-24, Volume: 48, Issue:6
Design and synthesis of N-[(4-methoxyphenoxy)carbonyl]-N-[[4-[2-(5- methyl-2-phenyl-4-oxazolyl)ethoxy]phenyl]methyl]glycine [Muraglitazar/BMS-298585], a novel peroxisome proliferator-activated receptor alpha/gamma dual agonist with efficacious glucose and
AID320687Agonist activity at human PPARgamma expressed in CV1 cells by transactivation assay2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Structure-activity studies on 1,3-dioxane-2-carboxylic acid derivatives, a novel class of subtype-selective peroxisome proliferator-activated receptor alpha (PPARalpha) agonists.
AID746619Agonist activity at PPARalpha (unknown origin) transfected in HEK293 cells at 10 uM by luciferase reporter gene assay2013Bioorganic & medicinal chemistry letters, May-15, Volume: 23, Issue:10
Synthesis and evaluation of 2,3-dinorprostaglandins: Dinor-PGD1 and 13-epi-dinor-PGD1 are peroxisome proliferator-activated receptor α/γ dual agonists.
AID242552Mean inhibitory concentration against human peroxisome proliferator-activated receptor delta; (not tested)2005Bioorganic & medicinal chemistry letters, Jan-03, Volume: 15, Issue:1
2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.
AID1346004Rat fatty acid binding protein 1 (Fatty acid-binding proteins)2008Journal of medicinal chemistry, Jul-10, Volume: 51, Issue:13
Characterization of the drug binding specificity of rat liver fatty acid binding protein.
AID1800412ANS Fluorescence Displacement Assay from Article 10.1021/bi401014k: \\The human liver fatty acid binding protein T94A variant alters the structure, stability, and interaction with fibrates.\\2013Biochemistry, Dec-23, Volume: 52, Issue:51
The human liver fatty acid binding protein T94A variant alters the structure, stability, and interaction with fibrates.
AID1798916Cell-Based Transcription Assay from Article 10.1021/jm058056x: \\Substituted 2-[(4-aminomethyl)phenoxy]-2-methylpropionic acid PPARalpha agonists. 1. Discovery of a novel series of potent HDLc raising agents.\\2007Journal of medicinal chemistry, Feb-22, Volume: 50, Issue:4
Substituted 2-[(4-aminomethyl)phenoxy]-2-methylpropionic acid PPARalpha agonists. 1. Discovery of a novel series of potent HDLc raising agents.
AID493017Wombat Data for BeliefDocking2005Bioorganic & medicinal chemistry letters, Jan-03, Volume: 15, Issue:1
2-Alkoxydihydrocinnamates as PPAR agonists. Activity modulation by the incorporation of phenoxy substituents.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (239)

TimeframeStudies, This Drug (%)All Drugs %
pre-199050 (20.92)18.7374
1990's25 (10.46)18.2507
2000's65 (27.20)29.6817
2010's91 (38.08)24.3611
2020's8 (3.35)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 74.00

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 Index74.00 (24.57)
Research Supply Index5.73 (2.92)
Research Growth Index4.75 (4.65)
Search Engine Demand Index128.76 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (74.00)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials51 (20.00%)5.53%
Reviews19 (7.45%)6.00%
Case Studies6 (2.35%)4.05%
Observational0 (0.00%)0.25%
Other179 (70.20%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (25)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A Randomized, Open-label, Multiple Dosing, 3-way Crossover Study to Evaluate the Drug-drug Interaction Between Atorvastatin and Fenofibric Acid in Healthy Male Volunteers [NCT02422030]Phase 130 participants (Actual)Interventional2015-03-17Completed
FEnofibRate as a Metabolic INtervention for Coronavirus Disease 2019 [NCT04517396]Phase 2701 participants (Actual)Interventional2020-08-18Completed
A 30-Week, Multicenter, Randomized, Double-Blind, Parallel-Group Study of the Combination of ABT-335 and Rosuvastatin Compared to Rosuvastatin Monotherapy in Dyslipidemic Subjects With Stage 3 Chronic Kidney Disease [NCT00680017]Phase 3280 participants (Actual)Interventional2008-06-30Completed
Comparison of Fenofibric Acid Bioavailability From 45 mg and 135 mg Strength ABT-335 Capsules [NCT00839293]Phase 132 participants (Anticipated)Interventional2009-02-28Completed
Open-Label, Randomized, Single-Dose, 3-Arm, Crossover Pharmacokinetic and Bioequivalence Study of One 35 mg Fenofibric Acid Tablet and Three 35 mg Fenofibric Acid Tablets Versus One 105 mg Fenofibric Acid Tablet Under Fasting Conditions [NCT00961259]Phase 154 participants (Actual)Interventional2008-02-29Completed
A 12-week, Multicenter, Randomized, Double-Blind, Parallel-Group Study of the Combination of ABT-335 and Rosuvastatin Compared to ABT-335 and Rosuvastatin Monotherapy in Subjects With Type IIa and IIb Dyslipidemia [NCT00463606]Phase 3760 participants (Actual)Interventional2007-04-30Completed
[NCT00719693]Phase 1115 participants (Actual)Interventional2008-07-31Completed
[NCT00681525]Phase 118 participants (Actual)Interventional2008-04-30Completed
A Multi-Center, Randomized, Double-Blind, Placebo- and Active-Controlled Study Assessing the Efficacy, Safety and Tolerability of Diazoxide Choline Controlled-Release Tablet (DCCR) in Subjects Without Diabetes Mellitus Having Very High Fasting Triglycerid [NCT00973271]Phase 30 participants (Actual)Interventional2011-03-31Withdrawn(stopped due to Did not continue with development of DCCR in Very High Triglycerides)
A Single-Dose, Bioequivalence Study of 105 mg Fenofibric Acid Tablets Versus 145 mg TriCor® (Fenofibrate) Tablets Under Fed Conditions(Standard Meal) [NCT00960687]Phase 154 participants (Actual)Interventional2007-10-31Completed
A Phase II Study of Metronomic and Targeted Anti-angiogenesis Therapy for Children With Recurrent/Progressive Medulloblastoma, Ependymoma and ATRT [NCT01356290]Phase 2100 participants (Anticipated)Interventional2014-04-30Recruiting
A Year 2, Long-Term, Open-Label, Safety Extension Study of the Combination of ABT-335 and Statin Therapy for Subjects With Mixed Dyslipidemia [NCT00491530]Phase 3310 participants (Actual)Interventional2007-06-30Completed
An Open-label, Randomized, 6-Sequence, 3-Period Crossover Study to Evaluate a Pharmacokinetic Drug Interaction Between Rosuvastatin and Fenofibric Acid in Healthy Adult Subjects [NCT01964365]Phase 130 participants (Actual)Interventional2013-09-30Completed
Comparison of the Efficacy and AtorVastatin 20mg mOnotherapy Versus Combination Atorvastatin/Fenofibric Acid 10/135mg in the Mixed hyperlipiDemia Who Were Not at Lipid gOals With Atorvastatin 10mg Monotherapy. [NCT01974297]194 participants (Anticipated)Interventional2013-07-31Recruiting
A Single-Dose, Bioequivalence Study of 105 mg Fenofibric Acid Tablets Versus 145 mg Tricor® (Fenofibrate) Tablets Under Fasting Conditions. [NCT00961116]Phase 154 participants (Actual)Interventional2007-10-31Completed
An Open-Label Drug Interaction Study to Investigate the Effects of Steady State Fenofibric Acid on the Single-Dose Pharmacokinetics of Efavirenz in Healthy Volunteers [NCT00960570]Phase 130 participants (Actual)Interventional2008-02-29Completed
A Four Arm, Single-Dose, Food Effect Evaluation With 105 mg Fenofibric Acid Tablets Administered in a Fasted State and Three Different Fed Conditions, Low-Fat/Low Calorie Meal, Standard Meal, and High-Fat/High Calorie Meal [NCT00960856]Phase 140 participants (Actual)Interventional2007-11-30Completed
Multiple-Dose Pharmacokinetic and Pharmacodynamic Interaction Between ABT-335, Rosuvastatin and Warfarin [NCT00487136]Phase 145 participants (Actual)Interventional2007-06-30Completed
Open-label, Randomized Study to Evaluate the Effect of Food on the Pharmacokinetic Characteristics of Fenofibric Acid for HIP0901 Capsule in Healthy Male Subjects [NCT01555398]Phase 148 participants (Actual)Interventional2012-02-29Completed
A Randomized, Open Label, Three-Treatment, Three-Period, Six-Sequence Crossover Study to Compare the Pharmacokinetics of CJ-30039 and Lipidil Supra and to Investigate Food-effect on Pharmacokinetics of CJ-30039 [NCT01501435]Phase 148 participants (Actual)Interventional2011-12-31Completed
Phase II Study to Evaluate Fenofibrate Therapy in Patients With Smoldering or Symptomatic Multiple Myeloma [NCT01965834]Phase 26 participants (Actual)Interventional2012-11-19Terminated(stopped due to Lack of adequate accrual.)
Mechanisms of Atheroprotection by Fenofibric Acid (ABT 335) Added to a Statin in Subjects With Insulin Resistance (Hypertriglyceridemia and Low HDL-C) [NCT01025492]Phase 424 participants (Actual)Interventional2009-11-30Terminated(stopped due to Collaborator/corporate sponsor withdrew funding and permission to continue.)
Evaluation of Choline Fenofibrate (ABT-335) on Carotid Intima-Media Thickness (cIMT) in Subjects With Type IIb Dyslipidemia With Residual Risk in Addition to Atorvastatin Therapy (FIRST) Trial [NCT00616772]Phase 3682 participants (Actual)Interventional2008-02-29Completed
A Multicenter, Randomized, Double-Blind, Prospective Study Comparing the Safety and Efficacy of ABT-335 in Combination With Atorvastatin and Ezetimibe to Atorvastatin in Combination With Ezetimibe in Subjects With Combined (Atherogenic) Dyslipidemia [NCT00639158]Phase 3543 participants (Actual)Interventional2008-02-29Completed
An Open-label, Drug Interaction Study to Investigate the Effects of Steady-State Fenofibric Acid on the Single-Dose Pharmacokinetics of Efavirenz in Healthy Subjects [NCT01472380]Phase 130 participants (Actual)Interventional2011-11-30Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00463606 (9) [back to overview]Mean Percent Change From Baseline to the Final Visit in Low-density Lipoprotein Cholesterol (LDL-C) (Full Analysis Set)
NCT00463606 (9) [back to overview]Mean Percent Change From Baseline to the Final Visit in Apolipoprotein B (ApoB) (Full Analysis Set)
NCT00463606 (9) [back to overview]Mean Percent Change From Baseline to the Final Visit in High-density Lipoprotein Cholesterol (HDL-C) (Full Analysis Set)
NCT00463606 (9) [back to overview]Mean Percent Change From Baseline to the Final Visit in Non-high-density Lipoprotein Cholesterol (Non-HDL-C), With ABT-335 135 mg in Combination With Rosuvastatin 5 mg Versus ABT-335 135 mg Monotherapy (Full Analysis Set)
NCT00463606 (9) [back to overview]Mean Percent Change From Baseline to the Final Visit in Non-high-density Lipoprotein Cholesterol (Non-HDL-C), With ABT-335 135 mg in Combination With Rosuvastatin 5 mg Versus Rosuvastatin 5 mg Monotherapy (Full Analysis Set)
NCT00463606 (9) [back to overview]Mean Percent Change From Baseline to the Final Visit in Total Cholesterol (Full Analysis Set)
NCT00463606 (9) [back to overview]Mean Percent Change From Baseline to the Final Visit in Triglycerides (Full Analysis Set)
NCT00463606 (9) [back to overview]Mean Percent Change From Baseline to the Final Visit in Very-low-density Lipoprotein Cholesterol (VLDL-C) (Full Analysis Set)
NCT00463606 (9) [back to overview]Median Percent Change From Baseline to the Final Visit in High Sensitivity C-reactive Protein (hsCRP) (Full Analysis Set)
NCT00491530 (7) [back to overview]Mean Percent Change in Very Low-Density Lipoprotein Cholesterol (VLDL-C) From Baseline to Week 104 of This Open-Label Year 2 Study
NCT00491530 (7) [back to overview]Median Percent Change in Triglycerides From Baseline to Week 104 of This Open-Label Year 2 Study
NCT00491530 (7) [back to overview]Percentage of Subjects Reporting Adverse Events During Combination Therapy in the Preceding Double-Blind Studies or in the Preceding Open-Label Year 1 Study or in This Open-Label Year 2 Study
NCT00491530 (7) [back to overview]Mean Percent Change in Direct Low-Density Lipoprotein Cholesterol (LDL-C) From Baseline to Week 104 of This Open-Label Year 2 Study
NCT00491530 (7) [back to overview]Mean Percent Change in High-Density Lipoprotein Cholesterol (HDL-C) From Baseline to Week 104 of This Open-Label Year 2 Study
NCT00491530 (7) [back to overview]Mean Percent Change in Non-High-Density Lipoprotein Cholesterol (Non-HDL-C) From Baseline to Week 104 of This Open-Label Year 2 Study
NCT00491530 (7) [back to overview]Mean Percent Change in Total Cholesterol (Total-C) From Baseline to Week 104 of This Open-Label Year 2 Study
NCT00616772 (5) [back to overview]Rate of Change in Mean Posterior-wall Carotid Intima-media Thickness (cIMT)
NCT00616772 (5) [back to overview]Rate of Change in Mean of Maximal Posterior-wall Carotid Intima-media Thickness (cIMT)
NCT00616772 (5) [back to overview]Rate of Change in Composite of Mean of the Mean Posterior-wall Intima-media Thickness (IMT)
NCT00616772 (5) [back to overview]Rate of Change in Composite of Mean of Maximal Posterior-wall Intima-media Thickness (IMT)
NCT00616772 (5) [back to overview]Rate of Change in Composite of Mean of Maximal Posterior-wall and Anterior-wall Intima-media Thickness (IMT)
NCT00639158 (8) [back to overview]Median Percent Change in High-Sensitivity C-Reactive Protein (hsCRP) From Baseline to Final Visit
NCT00639158 (8) [back to overview]Mean Percent Change in Very Low-Density Lipoprotein Cholesterol (VLDL-C) From Baseline to Final Visit
NCT00639158 (8) [back to overview]Mean Percent Change in Non-High-Density Lipoprotein Cholesterol (Non-HDL-C) From Baseline to Final Visit
NCT00639158 (8) [back to overview]Mean Percent Change in Apolipoprotein CIII (apoCIII) From Baseline to Final Visit
NCT00639158 (8) [back to overview]Mean Percent Change in Apolipoprotein B (apoB) From Baseline to Final Visit
NCT00639158 (8) [back to overview]Mean Percent Change in Apolipoprotein AI (apoAI) From Baseline to Final Visit
NCT00639158 (8) [back to overview]Mean Percent Change in High-Density Lipoprotein Cholesterol (HDL-C) From Baseline to Final Visit
NCT00639158 (8) [back to overview]Median Percent Change in Triglycerides From Baseline to Final Visit
NCT00680017 (2) [back to overview]Mean Percent Change in High-Density Lipoprotein Cholesterol From Baseline to Week 8.
NCT00680017 (2) [back to overview]Median Percent Change in Triglycerides From Baseline to Week 8.
NCT00960570 (3) [back to overview]Area Under the Concentration Versus Time Curve From Time 0 Extrapolated to Infinity [AUC(0-∞)]
NCT00960570 (3) [back to overview]Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]
NCT00960570 (3) [back to overview]Maximum Plasma Concentration (Cmax) of Efavirenz
NCT00960687 (3) [back to overview]Maximum Plasma Concentration (Cmax)
NCT00960687 (3) [back to overview]Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]
NCT00960687 (3) [back to overview]The Area Under the Plasma Concentration Versus Time Curve From Time 0 to Infinity AUC(0-∞)
NCT00960856 (3) [back to overview]Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]
NCT00960856 (3) [back to overview]Maximum Plasma Concentration (Cmax)
NCT00960856 (3) [back to overview]The Area Under the Plasma Concentration Versus Time Curve From Time 0 to Infinity AUC(0-∞)
NCT00961116 (3) [back to overview]Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]
NCT00961116 (3) [back to overview]Maximum Plasma Concentration (Cmax)
NCT00961116 (3) [back to overview]The Area Under the Plasma Concentration Versus Time Curve From Time 0 to Infinity AUC(0-∞)
NCT00961259 (3) [back to overview]The Area Under the Plasma Concentration Versus Time Curve From Time 0 to Infinity AUC(0-∞)
NCT00961259 (3) [back to overview]Maximum Plasma Concentration (Cmax)
NCT00961259 (3) [back to overview]Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]
NCT01472380 (3) [back to overview]Pharmacokinetics: Area Under the Concentration Versus Time Curve From Time 0 Extrapolated to Infinity [AUC(0-infinity]
NCT01472380 (3) [back to overview]Pharmacokinetics: Area Under the Concentration Versus Time Curve From Time 0 to Time t[AUC(0-t)]
NCT01472380 (3) [back to overview]Pharmacokinetics: Maximum Plasma Concentration (Cmax)
NCT01965834 (3) [back to overview]Number of Subjects Experiencing Adverse Events
NCT01965834 (3) [back to overview]Proportion of Participants Achieving Progression-Free Survival
NCT01965834 (3) [back to overview]Rate of Response in Participants Receiving Fenofibrate Therapy
NCT04517396 (7) [back to overview]All-Cause Death
NCT04517396 (7) [back to overview]Seven-category Ordinal Scale
NCT04517396 (7) [back to overview]Secondary Hierarchical Composite Endpoint
NCT04517396 (7) [back to overview]Primary Hierarchical Composite Endpoint
NCT04517396 (7) [back to overview]Number of Days Alive, Out of the Intensive Care Unit, Free of Mechanical Ventilation/Extracorporeal Membrane Oxygenation, or Maximal Available Respiratory Support in the 30 Days Following Randomization
NCT04517396 (7) [back to overview]Number of Days Alive and Out of the Hospital During the 30 Days Following Randomization
NCT04517396 (7) [back to overview]Exploratory Hierarchical Composite Endpoint

Mean Percent Change From Baseline to the Final Visit in Low-density Lipoprotein Cholesterol (LDL-C) (Full Analysis Set)

The mean percent change from baseline to the final visit in low-density lipoprotein cholesterol (LDL-C), with ABT-335 135 mg in combination with rosuvastatin 5 mg versus ABT-335 135 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Mean)
ABT-335 and Rosuvastatin Calcium-28.7
ABT-335-4.1

[back to top]

Mean Percent Change From Baseline to the Final Visit in Apolipoprotein B (ApoB) (Full Analysis Set)

The mean percent change from baseline to the final visit in apolipoprotein B (ApoB), with ABT-335 135 mg in combination with rosuvastatin 5 mg versus rosuvastatin 5 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Mean)
ABT-335 and Rosuvastatin Calcium-30.9
Rosuvastatin Calcium-26.4

[back to top]

Mean Percent Change From Baseline to the Final Visit in High-density Lipoprotein Cholesterol (HDL-C) (Full Analysis Set)

The mean percent change from baseline to the final visit in High-density lipoprotein cholesterol (HDL-C), with ABT-335 135 mg in combination with rosuvastatin 5 mg versus rosuvastatin 5 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Mean)
ABT-335 and Rosuvastatin Calcium23.0
Rosuvastatin Calcium12.4

[back to top]

Mean Percent Change From Baseline to the Final Visit in Non-high-density Lipoprotein Cholesterol (Non-HDL-C), With ABT-335 135 mg in Combination With Rosuvastatin 5 mg Versus ABT-335 135 mg Monotherapy (Full Analysis Set)

The mean percent change from baseline to the final visit in non-high-density lipoprotein cholesterol (non-HDL-C), with ABT-335 135 mg in combination with rosuvastatin 5 mg versus ABT-335 135 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Mean)
ABT-335 and Rosuvastatin Calcium-37.4
ABT-335-16.0

[back to top]

Mean Percent Change From Baseline to the Final Visit in Non-high-density Lipoprotein Cholesterol (Non-HDL-C), With ABT-335 135 mg in Combination With Rosuvastatin 5 mg Versus Rosuvastatin 5 mg Monotherapy (Full Analysis Set)

The mean percent change from baseline to the final visit in non-high-density lipoprotein cholesterol (non-HDL-C), with ABT-335 135 mg in combination with rosuvastatin 5 mg versus rosuvastatin 5 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Mean)
ABT-335 and Rosuvastatin Calcium-37.4
Rosuvastatin Calcium-31.8

[back to top]

Mean Percent Change From Baseline to the Final Visit in Total Cholesterol (Full Analysis Set)

The mean percent change from baseline to the final visit in total cholesterol, with ABT-335 135 mg in combination with rosuvastatin 5 mg versus rosuvastatin 5 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Mean)
ABT-335 and Rosuvastatin Calcium-28.1
Rosuvastatin Calcium-25.0

[back to top]

Mean Percent Change From Baseline to the Final Visit in Triglycerides (Full Analysis Set)

The mean percent change from baseline to the final visit in triglycerides, with ABT-335 135 mg in combination with rosuvastatin 5 mg versus rosuvastatin 5 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Mean)
ABT-335 and Rosuvastatin Calcium-40.3
Rosuvastatin Calcium-17.5

[back to top]

Mean Percent Change From Baseline to the Final Visit in Very-low-density Lipoprotein Cholesterol (VLDL-C) (Full Analysis Set)

The mean percent change from baseline to the final visit in very-low-density lipoprotein cholesterol (VLDL-C), with ABT-335 135 mg in combination with rosuvastatin 5 mg versus rosuvastatin 5 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Mean)
ABT-335 and Rosuvastatin Calcium-41.3
Rosuvastatin Calcium-22.2

[back to top]

Median Percent Change From Baseline to the Final Visit in High Sensitivity C-reactive Protein (hsCRP) (Full Analysis Set)

The median percent change from baseline to the final visit in high sensitivity C-reactive protein (hsCRP), with ABT-335 135 mg in combination with rosuvastatin 5 mg versus rosuvastatin 5 mg monotherapy. (NCT00463606)
Timeframe: Baseline to 12 Weeks

Interventionpercent change (Median)
ABT-335 and Rosuvastatin Calcium-28.0
Rosuvastatin Calcium-11.4

[back to top]

Mean Percent Change in Very Low-Density Lipoprotein Cholesterol (VLDL-C) From Baseline to Week 104 of This Open-Label Year 2 Study

[(Week 104 VLDL-C minus baseline VLDL-C)/baseline VLDL-C] X 100. Baseline is the last value prior to the first dose of combination therapy. (NCT00491530)
Timeframe: Baseline to Week 104 (may include weeks in preceding double-blind studies [combination treatment arms], plus 52 weeks in preceding open-label year 1 study, and open-label year 2 study, up to 104 weeks)

Interventionpercent change (Mean)
ABT-335 + 20 mg Rosuvastatin-33.7
ABT-335 + 40 mg Simvastatin-18.7
ABT-335 + 40 mg Atorvastatin-26.6

[back to top]

Median Percent Change in Triglycerides From Baseline to Week 104 of This Open-Label Year 2 Study

[(Week 104 triglycerides minus baseline triglycerides)/baseline triglycerides] X 100. Baseline is the last value prior to the first dose of combination therapy. (NCT00491530)
Timeframe: Baseline to Week 104 (may include weeks in preceding double-blind studies [combination treatment arms], plus 52 weeks in preceding open-label year 1 study, and open-label year 2 study, up to 104 weeks)

Interventionpercent change (Median)
ABT-335 + 20 mg Rosuvastatin-36.9
ABT-335 + 40 mg Simvastatin-29.6
ABT-335 + 40 mg Atorvastatin-38.7

[back to top]

Percentage of Subjects Reporting Adverse Events During Combination Therapy in the Preceding Double-Blind Studies or in the Preceding Open-Label Year 1 Study or in This Open-Label Year 2 Study

All serious and non-serious adverse events are reported from the time of combination study drug initiation until 30 days after discontinuation of study drug. Adverse events are unfavorable changes in health that occur in subjects during a clinical trial or within a specified period following a trial. Serious adverse events are those that result in death, require inpatient hospitalization or the prolongation of hospitalization, result in congenital anomaly/birth defect, or significant disability/incapacity or are life-threatening. (NCT00491530)
Timeframe: Anytime after initiation of combination therapy (in the preceding 12-week double-blind studies or in the preceding open-label year 1 study) up to 116 weeks, to within 30 days after the last dose of combination therapy.

Interventionpercentage of participants (Number)
ABT-335 + 20 mg Rosuvastatin94.8
ABT-335 + 40 mg Simvastatin90.0
ABT-335 + 40 mg Atorvastatin97.7

[back to top]

Mean Percent Change in Direct Low-Density Lipoprotein Cholesterol (LDL-C) From Baseline to Week 104 of This Open-Label Year 2 Study

[(Week 104 LDL-C minus baseline LDL-C)/baseline LDL-C] X 100. Baseline is the last value prior to the first dose of combination therapy. (NCT00491530)
Timeframe: Baseline to Week 104 (may include weeks in preceding double-blind studies [combination treatment arms], plus 52 weeks in preceding open-label year 1 study, and open-label year 2 study, up to 104 weeks)

Interventionpercent change (Mean)
ABT-335 + 20 mg Rosuvastatin-19.2
ABT-335 + 40 mg Simvastatin-20.2
ABT-335 + 40 mg Atorvastatin-20.5

[back to top]

Mean Percent Change in High-Density Lipoprotein Cholesterol (HDL-C) From Baseline to Week 104 of This Open-Label Year 2 Study

[(Week 104 HDL-C minus baseline HDL-C)/baseline HDL-C] X 100. Baseline is the last value prior to the first dose of combination therapy. (NCT00491530)
Timeframe: Baseline to Week 104 (may include weeks in preceding double-blind studies [combination treatment arms], plus 52 weeks in preceding open-label year 1 study, and open-label year 2 study, up to 104 weeks)

Interventionpercent change (Mean)
ABT-335 + 20 mg Rosuvastatin13.7
ABT-335 + 40 mg Simvastatin11.2
ABT-335 + 40 mg Atorvastatin5.2

[back to top]

Mean Percent Change in Non-High-Density Lipoprotein Cholesterol (Non-HDL-C) From Baseline to Week 104 of This Open-Label Year 2 Study

[(Week 104 Non-HDL-C minus baseline Non-HDL-C)/baseline Non-HDL-C] X 100. Baseline is the last value prior to the first dose of combination therapy. (NCT00491530)
Timeframe: Baseline to Week 104 (may include weeks in preceding double-blind studies [combination treatment arms], plus 52 weeks in preceding open-label year 1 study, and open-label year 2 study, up to 104 weeks)

Interventionpercent change (Mean)
ABT-335 + 20 mg Rosuvastatin-26.9
ABT-335 + 40 mg Simvastatin-23.8
ABT-335 + 40 mg Atorvastatin-25.1

[back to top]

Mean Percent Change in Total Cholesterol (Total-C) From Baseline to Week 104 of This Open-Label Year 2 Study

[(Week 104 Total-C minus baseline Total-C)/baseline Total-C] X 100. Baseline is the last value prior to the first dose of combination therapy. (NCT00491530)
Timeframe: Baseline to Week 104 (may include weeks in preceding double-blind studies [combination treatment arms], plus 52 weeks in preceding open-label year 1 study, and open-label year 2 study, up to 104 weeks)

Interventionpercent change (Mean)
ABT-335 + 20 mg Rosuvastatin-20.1
ABT-335 + 40 mg Simvastatin-17.9
ABT-335 + 40 mg Atorvastatin-20.4

[back to top]

Rate of Change in Mean Posterior-wall Carotid Intima-media Thickness (cIMT)

Rate of change (mm/year) from baseline in mean of posterior-wall carotid intima-media thickness (cIMT) of the left and right common carotid artery. The statistical model used change from baseline as the dependent variable, with time of cIMT assessment (in years) as one of the factors in the model. The between-group difference in the rate of change was based on the parameter coefficient for the time-by-treatment interaction. The within-group rate of change was obtained from estimate statements within the repeated measures analysis. cIMT was measured using non-invasive ultrasound. (NCT00616772)
Timeframe: Baseline, 6 months, 12 months, 18 months, and 24 months

Interventionmm/year (Mean)
ABT-335 + Atorvastatin-0.006
Placebo + Atorvastatin0.000

[back to top]

Rate of Change in Mean of Maximal Posterior-wall Carotid Intima-media Thickness (cIMT)

Rate of change (mm/year) from baseline in mean of maximal posterior-wall carotid intima-media thickness (cIMT) of the left and right common carotid artery. The statistical model used change from baseline as the dependent variable, with time of cIMT assessment (in years) as one of the factors in the model. The between-group difference in the rate of change was based on the parameter coefficient for the time-by-treatment interaction. The within-group rate of change was obtained from estimate statements within the repeated measures analysis. cIMT was measured using non-invasive ultrasound. (NCT00616772)
Timeframe: Baseline, 6 months, 12 months, 18 months, and 24 months

Interventionmm/year (Mean)
ABT-335 + Atorvastatin-0.005
Placebo + Atorvastatin-0.003

[back to top]

Rate of Change in Composite of Mean of the Mean Posterior-wall Intima-media Thickness (IMT)

Rate of change (mm/year) from baseline in composite of mean of the mean posterior-wall intima-media thickness (IMT) of the left and right common carotid artery, internal carotid artery, and carotid bifurcation. The statistical model used change from baseline as the dependent variable, with time of IMT assessment (in years) as one of the factors in the model. The between-group difference in the rate of change was based on the parameter coefficient for the time-by-treatment interaction. The within-group rate of change was obtained from estimate statements within the repeated measures analysis. IMT was measured using non-invasive ultrasound. (NCT00616772)
Timeframe: Baseline, 6 months, 12 months, 18 months, and 24 months

Interventionmm/year (Mean)
ABT-335 + Atorvastatin-0.010
Placebo + Atorvastatin-0.004

[back to top]

Rate of Change in Composite of Mean of Maximal Posterior-wall Intima-media Thickness (IMT)

Rate of change (mm/year) from baseline in composite of mean of maximal posterior-wall intima-media thickness (IMT) of the left and right common carotid artery, internal carotid artery, and carotid bifurcation. The statistical model used change from baseline as the dependent variable, with time of IMT assessment (in years) as one of the factors in the model. The between-group difference in the rate of change was based on the parameter coefficient for the time-by-treatment interaction. The within-group rate of change was obtained from estimate statements within the repeated measures analysis. IMT was measured using non-invasive ultrasound. (NCT00616772)
Timeframe: Baseline, 6 months, 12 months, 18 months, and 24 months

Interventionmm/year (Mean)
ABT-335 + Atorvastatin-0.014
Placebo + Atorvastatin-0.008

[back to top]

Rate of Change in Composite of Mean of Maximal Posterior-wall and Anterior-wall Intima-media Thickness (IMT)

Rate of change (mm/year) from baseline in composite of mean of maximal posterior-wall and anterior-wall intima-media thickness (IMT) of the left and right common carotid artery, internal carotid artery, and carotid bifurcation. The statistical model used change from baseline as the dependent variable, with time of IMT assessment (in years) as one of the factors in the model. The between-group difference in the rate of change was based on the parameter coefficient for the time-by-treatment interaction. The within-group rate of change was obtained from estimate statements within the repeated measures analysis. IMT was measured using non-invasive ultrasound. (NCT00616772)
Timeframe: Baseline, 6 months, 12 months, 18 months, and 24 months

Interventionmm/year (Mean)
ABT-335 + Atorvastatin-0.003
Placebo + Atorvastatin-0.019

[back to top]

Median Percent Change in High-Sensitivity C-Reactive Protein (hsCRP) From Baseline to Final Visit

[(Week 12 hsCRP minus baseline hsCRP)/baseline hSCRP] x 100 (NCT00639158)
Timeframe: Baseline to 12 weeks (Final Visit)

InterventionPercent change (Median)
ABT-335 + 40 mg Atorvastatin + 10 mg Ezetimibe-52.1
Placebo + 40 mg Atorvastatin + 10 mg Ezetimibe-40.3

[back to top]

Mean Percent Change in Very Low-Density Lipoprotein Cholesterol (VLDL-C) From Baseline to Final Visit

[(Week 12 VLDL-C minus baseline VLDL-C)/baseline VLDL-C] x 100 (NCT00639158)
Timeframe: Baseline to 12 weeks (final visit)

InterventionPercent change (Mean)
ABT-335 + 40 mg Atorvastatin + 10 mg Ezetimibe-57.8
Placebo + 40 mg Atorvastatin + 10 mg Ezetimibe-41.1

[back to top]

Mean Percent Change in Non-High-Density Lipoprotein Cholesterol (Non-HDL-C) From Baseline to Final Visit

[(Week 12 non-HDL-C minus baseline non-HDL-C)/baseline non-HDL-C] x 100 (NCT00639158)
Timeframe: Baseline to 12 weeks (Final Visit)

InterventionPercent change (Mean)
ABT-335 + 40 mg Atorvastatin + 10 mg Ezetimibe-55.6
Placebo + 40 mg Atorvastatin + 10 mg Ezetimibe-51.0

[back to top]

Mean Percent Change in Apolipoprotein CIII (apoCIII) From Baseline to Final Visit

[(Week 12 apoCIII minus baseline apoCIII)/baseline apoCIII] x 100 (NCT00639158)
Timeframe: Baseline to 12 weeks (Final Visit)

InterventionPercent change (Mean)
ABT-335 + 40 mg Atorvastatin + 10 mg Ezetimibe-42.5
Placebo + 40 mg Atorvastatin + 10 mg Ezetimibe-25.3

[back to top]

Mean Percent Change in Apolipoprotein B (apoB) From Baseline to Final Visit

[(Week 12 apoB minus baseline apoB)/baseline apoB] x 100 (NCT00639158)
Timeframe: Baseline to 12 weeks (Final Visit)

InterventionPercent change (Mean)
ABT-335 + 40 mg Atorvastatin + 10 mg Ezetimibe-49.1
Placebo + 40 mg Atorvastatin + 10 mg Ezetimibe-44.7

[back to top]

Mean Percent Change in Apolipoprotein AI (apoAI) From Baseline to Final Visit

[(Week 12 apoAI minus baseline apoAI)/baseline apoAI] x 100 (NCT00639158)
Timeframe: Baseline to 12 weeks (Final Visit)

InterventionPercent change (Mean)
ABT-335 + 40 mg Atorvastatin + 10 mg Ezetimibe1.8
Placebo + 40 mg Atorvastatin + 10 mg Ezetimibe-1.3

[back to top]

Mean Percent Change in High-Density Lipoprotein Cholesterol (HDL-C) From Baseline to Final Visit

[(Week 12 HDL-C minus baseline HDL-C)/baseline HDL-C] x 100 (NCT00639158)
Timeframe: Baseline to 12 weeks (Final Visit)

InterventionPercent change (Mean)
ABT-335 + 40 mg Atorvastatin + 10 mg Ezetimibe13.0
Placebo + 40 mg Atorvastatin + 10 mg Ezetimibe4.2

[back to top]

Median Percent Change in Triglycerides From Baseline to Final Visit

[(Week 12 triglycerides minus baseline triglycerides)/baseline triglycerides] x 100 (NCT00639158)
Timeframe: Baseline to 12 Weeks (Final Visit)

InterventionPercent change (Median)
ABT-335 + 40 mg Atorvastatin + 10 mg Ezetimibe-57.3
Placebo + 40 mg Atorvastatin + 10 mg Ezetimibe-39.7

[back to top]

Mean Percent Change in High-Density Lipoprotein Cholesterol From Baseline to Week 8.

High-density lipoprotein cholesterol (HDL-C) was measured in milligrams/deciliter (mg/dL). (NCT00680017)
Timeframe: Baseline to 8 weeks

Interventionpercent change (Least Squares Mean)
ABT-335 Plus Rosuvastatin16.9
Rosuvastatin7.8

[back to top]

Median Percent Change in Triglycerides From Baseline to Week 8.

Triglycerides were measured in milligrams/deciliter. (NCT00680017)
Timeframe: Baseline to 8 weeks

Interventionpercent change (Median)
ABT-335 Plus Rosuvastatin-38.0
Rosuvastatin-22.4

[back to top]

Area Under the Concentration Versus Time Curve From Time 0 Extrapolated to Infinity [AUC(0-∞)]

The area under the plasma concentration versus time curve from time 0 to infinity. [AUC(0-∞)] was calculated as the sum of AUC(0-t) plus the ratio of the last measurable plasma concentration to the elimination rate constant for efavirenz. (NCT00960570)
Timeframe: serial pharmacokinetic blood samples drawn immediately prior to dosing on Days 1 and 31 and then 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12, 16, 20, 24, 48, 72, 96 and 120 hours after dose administration

Interventionng-hr/mL (Mean)
Efavirenz Alone106,689.60
Efavirenz With Fenofibric Acid96,382.08

[back to top]

Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]

The area under the plasma concentration versus time curve from time 0 to the time of the last measurable concentration (t), as calculated by the linear trapezoidal rule for efavirenz. (NCT00960570)
Timeframe: serial pharmacokinetic blood samples drawn prior to dosing on Days 1 and 31 and then 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12, 16, 20, 24, 48, 72, 96 and 120 hours after dose administration

Interventionng-hr/mL (Mean)
Efavirenz Alone70,144.35
Efavirenz With Fenofibric Acid63,313.23

[back to top]

Maximum Plasma Concentration (Cmax) of Efavirenz

The maximum or peak concentration that efavirenz reaches in the plasma. (NCT00960570)
Timeframe: serial pharmacokinetic blood samples drawn prior to dosing on Days 1 and 31 and then 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12, 16, 20, 24, 48, 72, 96 and 120 hours after dose administration.

Interventionng/mL (Mean)
Efavirenz Alone2,461.58
Efavirenz With Fenofibric Acid2,384.79

[back to top]

Maximum Plasma Concentration (Cmax)

The maximum or peak concentration that the drug reaches in the plasma. (NCT00960687)
Timeframe: serial pharmacokinetic plasma concentrations were drawn prior to dose administration (0 hour) and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours after drug administration.

Interventionng/mL (Mean)
Fenofibric Acid 105 mg Tablets8,545.50
Fenofibrate 145 mg Tablets9,456.66

[back to top]

Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]

The area under the plasma concentration versus time curve, from time 0 to the time of the last measurable concentration (t), as calculated by the linear trapezoidal rule. (NCT00960687)
Timeframe: serial pharmacokinetic plasma concentrations were drawn prior to dose administration (0 hour) and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours after drug administration.

Interventionng-hr/mL (Mean)
Fenofibric Acid 105 mg Tablets121,974.07
Fenofibrate 145 mg Tablets132,463.37

[back to top]

The Area Under the Plasma Concentration Versus Time Curve From Time 0 to Infinity AUC(0-∞)

The area under the plasma concentration versus time curve from time 0 to infinity. AUC(0-∞) was calculated as the sum of AUC(0-t) plus the ratio of the last measurable plasma concentration to the elimination rate constant. (NCT00960687)
Timeframe: serial pharmacokinetic plasma concentrations were drawn prior to dose administration (0 hour) and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours after drug administration.

Interventionng-hr/mL (Mean)
Fenofibric Acid 105 mg Tablets134,576.33
Fenofibrate 145 mg Tablets146,847.53

[back to top]

Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]

The area under the plasma concentration versus time curve, from time 0 to the time of the last measurable concentration (t), as calculated by the linear trapezoidal rule. (NCT00960856)
Timeframe: serial pharmacokinetic blood samples drawn immediately prior to dosing and then 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48, and 72 hours after dose administration

Interventionng-hr/mL (Mean)
Low-fat Meal - Treatment A132,636.84
Standard Meal - Treatment B134,928.11
High-Fat, High-Calorie Meal - Treatment C134,814.42
Fasted - Treatment D139,975.28

[back to top]

Maximum Plasma Concentration (Cmax)

The maximum or peak concentration that the drug reaches in the plasma. (NCT00960856)
Timeframe: serial pharmacokinetic blood samples drawn immediately prior to dosing and then 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48, and 72 hours after dose administration

Interventionng/mL (Mean)
Low-fat Meal - Treatment A9,806.74
Standard Meal - Treatment B9,854.39
High-Fat, High-Calorie Meal - Treatment C7,950.75
Fasted - Treatment D12,061.67

[back to top]

The Area Under the Plasma Concentration Versus Time Curve From Time 0 to Infinity AUC(0-∞)

The area under the plasma concentration versus time curve from time 0 to infinity. AUC(0-∞) was calculated as the sum of AUC(0-t) plus the ratio of the last measurable plasma concentration to the elimination rate constant. (NCT00960856)
Timeframe: serial pharmacokinetic plasma concentrations were drawn prior to dose administration (0 hour) and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours after drug administration.

Interventionng-hr/mL (Mean)
Low-fat Meal - Treatment A145,827.10
Standard Meal - Treatment B148,367.21
High-Fat, High-Calorie Meal - Treatment C148,488.53
Fasted - Treatment D152,571.61

[back to top]

Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]

The area under the plasma concentration versus time curve, from time 0 to the time of the last measurable concentration (t), as calculated by the linear trapezoidal rule. (NCT00961116)
Timeframe: serial pharmacokinetic plasma concentrations were drawn prior to dose administration (0 hour) and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours after drug administration.

Interventionng-hr/mL (Mean)
Fenofibric Acid 105 mg Tablets159,866.23
Fenofibrate 145 mg Tablets170,727.10

[back to top]

Maximum Plasma Concentration (Cmax)

The maximum or peak concentration that the drug reaches in the plasma. (NCT00961116)
Timeframe: serial pharmacokinetic plasma concentrations were drawn prior to dose administration (0 hour) and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours after drug administration.

Interventionng/mL (Mean)
Fenofibric Acid 105 mg Tablets12,344.00
Fenofibrate 145 mg Tablets10,940.05

[back to top]

The Area Under the Plasma Concentration Versus Time Curve From Time 0 to Infinity AUC(0-∞)

The area under the plasma concentration versus time curve from time 0 to infinity. AUC(0-∞) was calculated as the sum of AUC(0-t) plus the ratio of the last measurable plasma concentration to the elimination rate constant. (NCT00961116)
Timeframe: serial pharmacokinetic plasma concentrations were drawn prior to dose administration (0 hour) and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours after drug administration.

Interventionng-hr/mL (Mean)
Fenofibric Acid 105 mg Tablets176,816.78
Fenofibrate 145 mg Tablets188,331.49

[back to top]

The Area Under the Plasma Concentration Versus Time Curve From Time 0 to Infinity AUC(0-∞)

The area under the plasma concentration versus time curve from time 0 to infinity. AUC(0-∞) was calculated as the sum of AUC(0-t) plus the ratio of the last measurable plasma concentration to the elimination rate constant. For the dosing group, Fenofibric Acid 35 mg (35 mg Dose-adjusted to 105 mg), the AUC(0-∞) values were dose-adjusted in order to assess pharmacokinetic linearity. (NCT00961259)
Timeframe: serial pharmacokinetic plasma concentrations were drawn prior to dose administration (0 hour) and at 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48 and 72 hours after drug administration.

Interventionng-hr/mL (Mean)
Fenofibric Acid 35 mg (1 x 35 mg Tablet) - Treatment A58,304.35
Fenofibric Acid 35 mg (35 mg Dose-adjusted to 105 mg)174,913.04
Fenofibric Acid 105 mg (3 x 35 mg Tablet) - Treatment B142,858.88
Fenofibric Acid 105 mg (1 x 105 mg Tablet) - Treatment C147,555.58

[back to top]

Maximum Plasma Concentration (Cmax)

The maximum or peak concentration that the drug reaches in the plasma. For the dosing group, Fenofibric Acid 35 mg (35 mg Dose-adjusted to 105 mg), the Cmax values were dose-adjusted in order to assess pharmacokinetic linearity. (NCT00961259)
Timeframe: serial pharmacokinetic blood samples drawn immediately prior to dosing and then 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48, and 72 hours after dose administration

Interventionng/mL (Mean)
Fenofibric Acid 35 mg (1 x 35 mg Tablet) - Treatment A3,569.47
Fenofibric Acid 35 mg (35 mg Dose-adjusted to 105 mg)10,708.40
Fenofibric Acid 105 mg (3 x 35 mg Tablet) - Treatment B11,014.93
Fenofibric Acid 105 mg (1 x 105 mg Tablet) - Treatment C11,202.87

[back to top]

Area Under the Concentration Versus Time Curve From Time 0 to Time t [AUC(0-t)]

The area under the plasma concentration versus time curve, from time 0 to the time of the last measurable concentration (t), as calculated by the linear trapezoidal rule. For the dosing group, Fenofibric Acid 35 mg (35 mg Dose-adjusted to 105 mg), the [AUC(0-t)] values were dose-adjusted in order to assess pharmacokinetic linearity. (NCT00961259)
Timeframe: serial pharmacokinetic blood samples drawn immediately prior to dosing and then 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 5, 6, 8, 10, 12, 16, 24, 36, 48, and 72 hours after dose administration

Interventionng-hr/mL (Mean)
Fenofibric Acid 35 mg (1 x 35 mg Tablet) - Treatment A49,419.89
Fenofibric Acid 35 mg (35 mg Dose-adjusted to 105 mg)148,259.66
Fenofibric Acid 105 mg (3 x 35 mg Tablet) - Treatment B131,976.95
Fenofibric Acid 105 mg (1 x 105 mg Tablet) - Treatment C135,119.10

[back to top]

Pharmacokinetics: Area Under the Concentration Versus Time Curve From Time 0 Extrapolated to Infinity [AUC(0-infinity]

The area under the plasma concentration versus time curve from time 0 to infinity. [AUC(0 to infinity)] was calculated as the sum of AUC (0-t) plus the ratio of the last measurable plasma concentration to the elimination rate constant for efavirenz. (NCT01472380)
Timeframe: serial pharmacokinetic blood samples drawn immediately prior to dosing on Days 1 and 31 and then 0.5, 1, 2, 3, 4, 5, 6, 8, 10, 12, 16, 20, 24, 48, 72, 96, and 120 hours after dose administration

Interventionh*ug/mL (Mean)
Efavirenz Alone121
Efavirenz With Fenofibric Acid112

[back to top]

Pharmacokinetics: Area Under the Concentration Versus Time Curve From Time 0 to Time t[AUC(0-t)]

The area under the plasma concentration versus time curve from time 0 to the time of the last measurable concentration (t), as calculated by the linear trapezoidal rule for efavirenz (NCT01472380)
Timeframe: serial pharmacokinetic blood samples drawn immediately prior to dosing on Days 1 and 31 and then 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 16, 20, 24, 48, 72, 96, and 120 hours after dose administration

Interventionh*ug/mL (Mean)
Efavirenz Alone70.58
Efavirenz With Fenofibric Acid64.62

[back to top]

Pharmacokinetics: Maximum Plasma Concentration (Cmax)

The maximum or peak concentration that the drug reaches in the plasma for efavirenz (NCT01472380)
Timeframe: serial pharmacokinetic blood samples drawn immediately prior to dosing on Days 1 and 31 and then 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 16, 20, 24, 48, 72, 96, and 120 hours after dose administration

Interventionug/mL (Mean)
Efavirenz Alone2.62
Efavirenz With Fenofibric Acid2.63

[back to top]

Number of Subjects Experiencing Adverse Events

To evaluate safety and tolerability of fenofibrate therapy in patients with multiple myeloma. (NCT01965834)
Timeframe: Up to 8 months

Interventionparticipants (Number)
Fenofibrate Therapy5

[back to top]

Proportion of Participants Achieving Progression-Free Survival

Proportion of participants achieving progression-free survival. Measured from date of initiation of treatment (Day 1) to the earliest occurrence of any of the following events: documented disease progression, or death from any cause. Patients who are alive and progression-free will be censored at the date of last documented progression-free status. (NCT01965834)
Timeframe: 6 months, 12 months

Interventionparticipants (Number)
6 months12 months
Fenofibrate Therapy00

[back to top]

Rate of Response in Participants Receiving Fenofibrate Therapy

To determine response rate (Strict Complete Response (sCR), Complete response (CR), Very Good Partial Response (VgPR), and Partial Response (PR)) in multiple myeloma patients receiving oral fenofibrate therapy. Response will be measured by serum and urine protein electrophoresis and immunofixation, as well as by percentage of plasma cells present on bone marrow biopsy. (NCT01965834)
Timeframe: After two cycles, about 2 months

Interventionparticipants (Number)
Strict Complete Response (sCR)Complete response (CR)Very Good Partial Response (VgPR)Partial Response (PR)
Fenofibrate Therapy0000

[back to top]

All-Cause Death

Death from any cause during the observation period (NCT04517396)
Timeframe: Up to 30 days

InterventionParticipants (Count of Participants)
Fenofibrate + Usual Care19
Placebo + Usual Care22

[back to top]

Seven-category Ordinal Scale

A seven-category ordinal scale consisting of the following categories: 1, not hospitalized with resumption of normal activities; 2, not hospitalized, but unable to resume normal activities; 3, hospitalized, not requiring supplemental oxygen; 4, hospitalized, requiring supplemental oxygen; 5, hospitalized, requiring nasal high-flow oxygen therapy, noninvasive mechanical ventilation, or both; 6, hospitalized, requiring extracorporeal membrane oxygenation (ECMO), invasive mechanical ventilation, or both; and 7, death. (NCT04517396)
Timeframe: At 15 days

Interventionscore on a scale (Median)
Fenofibrate + Usual Care1
Placebo + Usual Care1

[back to top]

Secondary Hierarchical Composite Endpoint

The secondary global rank score, or global severity score, is a nonparametric, hierarchically ranked outcome. The global rank score was generated by ranking all 701 participants on a scale of 1 to 701, from worst to best clinical outcomes. Participants were ranked by (1) time to death; (2) the number of days supported by invasive mechanical ventilation or extracorporeal membrane oxygenation (ECMO); (3) The inspired concentration of oxygen/percent oxygen saturation (FiO2/SpO2) ratio area under the curve; (4) For participants enrolled as outpatients who are subsequently hospitalized, the number of days out of the hospital during the 30 day-period following randomization; (5) For participants enrolled as outpatients who don't get hospitalized during the 30-day observation period, a COVID-19 symptom scale rating fever, cough, dyspnea, muscle aches, sore throat, loss of smell or taste, headache, diarrhea, fatigue, nausea/vomiting, chest pain (each are rated from 0-10 then summed). (NCT04517396)
Timeframe: Up to 30 days

Interventionscore on a scale (Median)
Fenofibrate + Usual Care5.05
Placebo + Usual Care5.05

[back to top]

Primary Hierarchical Composite Endpoint

The primary endpoint of the trial is a global rank score that ranks patient outcomes according to 5 factors. The global rank score, or global severity score, is a nonparametric, hierarchically ranked outcome. The global rank score was generated by ranking all 701 participants on a scale of 1 to 701, from worst to best clinical outcomes. Participants were ranked by (1) time to death; (2) the number of days supported by invasive mechanical ventilation or extracorporeal membrane oxygenation (ECMO); (3) The inspired concentration of oxygen/percent oxygen saturation (FiO2/SpO2) ratio area under the curve; (4) For participants enrolled as outpatients who are subsequently hospitalized, the number of days out of the hospital during the 30 day-period following randomization; (5) For participants enrolled as outpatients who don't get hospitalized during the 30-day observation period, the modified Borg dyspnea scale (NCT04517396)
Timeframe: 30 days

InterventionRanked Severity Score (Median)
Fenofibrate + Usual Care5.32
Placebo + Usual Care5.33

[back to top]

Number of Days Alive, Out of the Intensive Care Unit, Free of Mechanical Ventilation/Extracorporeal Membrane Oxygenation, or Maximal Available Respiratory Support in the 30 Days Following Randomization

Number of days participants were alive, out of the intensive care unit, free of mechanical ventilation/extracorporeal membrane oxygenation, or maximal available respiratory support during the 30 days that followed randomization (NCT04517396)
Timeframe: Up to 30 days

Interventiondays (Mean)
Fenofibrate + Usual Care28.8
Placebo + Usual Care28.3

[back to top]

Number of Days Alive and Out of the Hospital During the 30 Days Following Randomization

Number of days that participants were alive and out of the hospital during the 30 days following randomization (NCT04517396)
Timeframe: Up to 30 days

Interventiondays (Median)
Fenofibrate + Usual Care30
Placebo + Usual Care30

[back to top]

Exploratory Hierarchical Composite Endpoint

The exploratory global rank score, or global severity score, is a nonparametric, hierarchically ranked outcome. The global rank score was generated by ranking all 701 participants on a scale of 1 to 701, from worst to best clinical outcomes. Participants were ranked by (1) time to death; (2) the number of days supported by invasive mechanical ventilation or extracorporeal membrane oxygenation (ECMO); (3) The inspired concentration of oxygen/percent oxygen saturation (FiO2/SpO2) ratio area under the curve; (4) The number of days out of the hospital during the 30 day-period following randomization. (NCT04517396)
Timeframe: Up to 30 days

Interventionscore on a scale (Median)
Fenofibrate + Usual Care5.03
Placebo + Usual Care5.03

[back to top]