Page last updated: 2024-12-05

2,4-thiazolidinedione

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

Description

thiazolidine-2,4-dione: structure in first source [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

1,3-thiazolidine-2,4-dione : A thiazolidenedione carrying oxo substituents at positions 2 and 4. [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 CID5437
CHEMBL ID85398
CHEBI ID50992
SCHEMBL ID5043
SCHEMBL ID8158011
MeSH IDM0237742

Synonyms (70)

Synonym
AC-3453
CHEMBL85398
BB 0255316
thiazolidinedione
usaf ek-5496
thiazolidinedione-2,4
nsc-6745
nsc6745
2,4-dioxothiazolidine
2,5h)-thiazoledione
2,4-thiazolidinedione ,
2295-31-0
wln: t5svmv ehj
1,3-thiazolidine-2,4-dione
thiazolidine-2,4-dione
CHEBI:50992 ,
2,4-thiazolidinedione, technical grade, 90%
nsc 6745
ai3-61185
einecs 218-941-2
brn 0110700
2,4(3h,5h)-thiazoledione
gw3478456
STK159523
T1990
AKOS000119892
A4870
thiazolidindione
unii-aa68lxk93c
4-27-00-03144 (beilstein handbook reference)
aa68lxk93c ,
1,3-thiazolane-2,4-dione
F1028-0003
FT-0610238
AM20090236
SCHEMBL5043
mfcd00005478
SY001546
thiazolidine-2,4 dione
2,4-thiazolidindione
2,4thiazolidinedione
2,4 thiazolidinedione
2,4-thiazolidine dione
thiazolidine-dione
thiazolidin-2,4-dione
SCHEMBL8158011
W-107446
J-610067
1,3-thiazolidine-2,4-dione #
2,4-dioxo-1,3-thiazolidine
thiazole-2,4-dione
2-thiazolin-4-one, 2-hydroxy-
DTXSID5040623
AKOS025397012
sr-01000945204
SR-01000945204-1
bdbm50502228
AC7504
Z57131032
CS-W019917
DB11898
AS-15441
BCP26341
Q1393556
BRD-K70526448-001-02-0
1,3-thiazole-2,4-dione
9ut ,
EN300-18067
HY-59272
OD3 ,

Research Excerpts

Toxicity

ExcerptReferenceRelevance
"The first thiazolidinedione derivative drug for diabetes, troglitazone, was found to cause fatal hepatotoxicity, although it was judged as safe during the clinical trial."( [Evaluation of thiazolidinedione derivative drugs for safety].
Iwata, M; Kobayashi, M, 2001
)
0.31
" Since that review, some new knowledge allows us to better predict that some structural moieties are more likely than others to form drug reactive metabolites that may be involved in causing toxic effects in humans."( Structure toxicity relationships--how useful are they in predicting toxicities of new drugs?
Nelson, SD, 2001
)
0.31
" While the exact mechanism of its toxicity remains unknown, it has been postulated that the formation of toxic reactive metabolites (RMs) may play an important role in the hepatotoxicity of TGZ."( Investigation of the role of the thiazolidinedione ring of troglitazone in inducing hepatotoxicity.
Chan, EC; Chui, WK; Ho, HK; New, LS; Saha, S, 2010
)
0.36
" Adverse events for the US cohort were consistent with previously reported data from the 3 trials."( Efficacy and safety of saxagliptin combination therapy in US patients with type 2 diabetes.
Allen, E; Chen, R; Donovan, M; Fleming, D; Karyekar, C; Ravichandran, S, 2011
)
0.37
"5 or 5 mg as add-on therapy to OADs results in improvement across key glycemic parameters compared with placebo add-on and was generally safe and well tolerated."( Efficacy and safety of saxagliptin combination therapy in US patients with type 2 diabetes.
Allen, E; Chen, R; Donovan, M; Fleming, D; Karyekar, C; Ravichandran, S, 2011
)
0.37
" Safety was assessed by adverse events, hypoglycemia, and body weight."( Saxagliptin efficacy and safety in patients with type 2 diabetes mellitus and cardiovascular disease history or cardiovascular risk factors: results of a pooled analysis of phase 3 clinical trials.
Allen, E; Bryzinski, B; Cook, W; Frederich, R; Slater, J, 2013
)
0.39
" Saxagliptin was well tolerated, with similar adverse event rates and types compared with placebo."( Saxagliptin efficacy and safety in patients with type 2 diabetes mellitus and cardiovascular disease history or cardiovascular risk factors: results of a pooled analysis of phase 3 clinical trials.
Allen, E; Bryzinski, B; Cook, W; Frederich, R; Slater, J, 2013
)
0.39
" However, their use has been curtailed by substantial adverse effects on weight, bone, heart, and hemodynamic balance."( PPARγ deacetylation dissociates thiazolidinedione's metabolic benefits from its adverse effects.
Area-Gomez, E; Chan, M; Creusot, RJ; Fan, L; Fu, W; Ji, R; Kon, N; Kraakman, MJ; Larrea, D; Liu, Q; Namwanje, M; Postigo-Fernandez, J; Qiang, L, 2018
)
0.48
"Little is known about gender-specific reporting of adverse events (AEs) associated with antidiabetic drugs."( Gender differences in adverse event reports associated with antidiabetic drugs.
Joung, KI; Jung, GW; Lee, H; Park, HH; Park, SH; Shin, JY, 2020
)
0.56
"Multicentre, randomized, clinical trials that included over 100 participants comparing antidiabetic agents with a placebo or a different antidiabetic agent and reporting major adverse cardiovascular events (MACEs), or primarily reporting heart failure, were searched in the PubMed, Embase and Cochrane databases."( Cardiovascular efficacy and safety of antidiabetic agents: A network meta-analysis of randomized controlled trials.
Frias, JP; Lim, S; Sohn, M, 2023
)
0.91
" Adverse events leading to drug discontinuation were higher with GLP-1 RAs and thiazolidinediones than placebo."( Cardiovascular efficacy and safety of antidiabetic agents: A network meta-analysis of randomized controlled trials.
Frias, JP; Lim, S; Sohn, M, 2023
)
0.91

Pharmacokinetics

ExcerptReferenceRelevance
" dosing and subjected to noncompartment pharmacokinetic analysis to obtain the values for the parameters."( Oral bioavailability and pharmacokinetics of PAT-5A, a new insulin sensitizer in rats.
Katneni, K; Mamidi, RN; Mullangi, R; Ramanujam, R,
)
0.13
" The method is suitable for pharmacokinetic (PK) studies of the parent drug NL-1 based on the preliminary serum results from dosed NL-1 mouse studies."( A quantitative LC-MS/MS method for determination of thiazolidinedione mitoNEET ligand NL-1 in mouse serum suitable for pharmacokinetic studies.
Anderson, DJ; Carroll, RT; Geldenhuys, WJ; Jogiraju, H; Lin, L; Pedada, KK; Zhou, X, 2014
)
0.4
" The determination of pharmacokinetic parameters for these two classes of compounds using a simultaneous chromatographic method with a low detection limit is a challenge."( Validation of a sensitive simultaneous LC-MS/MS method for the quantification of novel anti-cancer thiazolidinedione and quinazolin-4-one derivatives in rat plasma and its application in a rat pharmacokinetic study.
Sengottuvelan, S; Uppal, R; Vuppu, S, 2019
)
0.51

Compound-Compound Interactions

ExcerptReferenceRelevance
"The purpose of this study was to assess the efficacy and tolerability of the dipeptidyl peptidase-4 inhibitor vildagliptin in combination with the thiazolidinedione (TZD) pioglitazone in patients with type 2 diabetes (T2DM)."( Vildagliptin in combination with pioglitazone improves glycaemic control in patients with type 2 diabetes failing thiazolidinedione monotherapy: a randomized, placebo-controlled study.
Baron, MA; Dejager, S; Garber, AJ; Rochotte, E; Schweizer, A, 2007
)
0.34
"The present study analyses the effect of two plant phenolic compounds, namely chlorogenic acid and ferulic acid, and a plant alkaloid, berberine, alone and also in combination with two commercial oral hypoglycemic drugs (OHD), namely metformin and 2,4-thiazolodinedione (THZ), on the uptake of 2-deoxyglucose (2DG) by L6 myotubes."( Synergistic effect of phytochemicals in combination with hypoglycemic drugs on glucose uptake in myotubes.
Doble, M; Prabhakar, PK, 2009
)
0.35

Bioavailability

ExcerptReferenceRelevance
" In another study, a single intravenous bolus dose of PAT-5A was given to rats at 10 mg/kg dose following administration through the tail vein in order to obtain the absolute oral bioavailability and clearance parameters."( Oral bioavailability and pharmacokinetics of PAT-5A, a new insulin sensitizer in rats.
Katneni, K; Mamidi, RN; Mullangi, R; Ramanujam, R,
)
0.13
" In rats, LB appeared to be readily absorbed after an oral administration (an absolute bioavailability of ∼95%)."( Kinetics of the Absorption, Distribution, Metabolism, and Excretion of Lobeglitazone, a Novel Activator of Peroxisome Proliferator-Activated Receptor Gamma in Rats.
Ahn, SH; Chung, SJ; Jeong, YS; Kim, DD; Lee, JH; Lee, W; Noh, CK; Yim, CS, 2015
)
0.42

Dosage Studied

ExcerptRelevanceReference
" dosing and subjected to noncompartment pharmacokinetic analysis to obtain the values for the parameters."( Oral bioavailability and pharmacokinetics of PAT-5A, a new insulin sensitizer in rats.
Katneni, K; Mamidi, RN; Mullangi, R; Ramanujam, R,
)
0.13
" The hydroxylated M-VII was detected in incubations with rat, dog and human liver and kidney microsomes, and in plasma from rats and dogs dosed orally with [(3)H]pioglitazone."( Identification of novel metabolites of pioglitazone in rat and dog.
Bakhtiar, R; Creighton, M; Feeney, W; Franklin, RB; Hora, DF; Liu, DQ; Reed, JR; Shen, Z; Szewczyk, J; Tang, YS; Vincent, SH, 2003
)
0.32
" In this study, we examined the changes in both cancellous and cortical bone of 6-month-old male mice treated with darglitazone, a potent and selective PPARgamma agonist, at 10 mg/kg/day by dosing the compound in a food mixture for 2 or 8 weeks."( Surface-specific effects of a PPARgamma agonist, darglitazone, on bone in mice.
Brown, TA; Healy, DR; Ke, HZ; Li, M; Li, Y; Pan, LC; Robinson, BS; Simmons, HA, 2006
)
0.33
" As the prevalence of children diagnosed with type 2 diabetes continues to rise, the need for adequate information regarding the safety, efficacy, and appropriate dosing of oral diabetes medications in the pediatric population likewise increases."( The utility of oral diabetes medications in type 2 diabetes of the young.
Abu-Baker, A; Busch, RS; Kane, MP, 2005
)
0.33
" These data include information regarding drug safety and efficacy and/or drug pharmacokinetic and drug dosing information."( The utility of oral diabetes medications in type 2 diabetes of the young.
Abu-Baker, A; Busch, RS; Kane, MP, 2005
)
0.33
" The sitagliptin dosage recommended by the manufacturer is 100 mg once daily as monotherapy or in combination with metformin or a thiazolidinedione."( Sitagliptin: a novel agent for the management of type 2 diabetes mellitus.
Nogid, A; Pham, DQ; Plakogiannis, R, 2008
)
0.35
"446), the addition of pioglitazone could significantly lower serum RBP4 and HOMA-IR values, whereas an increased dosage of sulfonylurea agents did not alter HOMA-IR, RBP4, or adiponectin in type 2 diabetic patients who had been treated with metformin and/or sulfonylurea."( Thiazolidinedione addition reduces the serum retinol-binding protein 4 in type 2 diabetic patients treated with metformin and sulfonylurea.
Chang, YH; Hsiao, PJ; Li, TH; Lin, KD; Shin, SJ; Wang, CL; Yang, YH, 2008
)
0.35
" The method is suitable for pharmacokinetic (PK) studies of the parent drug NL-1 based on the preliminary serum results from dosed NL-1 mouse studies."( A quantitative LC-MS/MS method for determination of thiazolidinedione mitoNEET ligand NL-1 in mouse serum suitable for pharmacokinetic studies.
Anderson, DJ; Carroll, RT; Geldenhuys, WJ; Jogiraju, H; Lin, L; Pedada, KK; Zhou, X, 2014
)
0.4
" No adverse effects attributable to pioglitazone were evident in the otherwise healthy obese cats at this dosage and duration."( Effects of pioglitazone on insulin sensitivity and serum lipids in obese cats.
Clark, M; Dirikolu, L; Ferguson, DC; Hoenig, M; Thomaseth, K,
)
0.13
" Individuals newly prescribed an oral glucose-lowering agent who filled a second prescription for a medication in the same class and with a dosage at or above the World Health Organization's defined daily dose within 90 days of the end-of-day's supply of the first prescription were studied."( Initial choice of oral glucose-lowering medication for diabetes mellitus: a patient-centered comparative effectiveness study.
Avorn, J; Berkowitz, SA; Brennan, T; Brill, G; Choudhry, NK; Krumme, AA; Matlin, OS; Pezalla, EJ; Shrank, WH; Spettell, CM, 2014
)
0.4
", separate dosage forms) intended for a disease can be measured by different single estimators, termed as composite estimators of intra-disease multiple medication adherence: 80% days covered (a) by at least one medication ("at least one"); (b) by both medications ("both"); (c) by each medication measured separately ("all"); and (d) computing an average of the individual medication adherence estimates ("average")."( Comparing algorithms for composite measures of intra-disease multiple medication adherence: The case of diabetes.
Banahan, BF; Basak, R; Bentley, JP; McCaffrey, DJ; Przybyla, SM; West-Strum, D, 2019
)
0.51
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Drug Classes (1)

ClassDescription
thiazolidenedione
[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 (6)

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Maltase-glucoamylase, intestinalHomo sapiens (human)IC50 (µMol)0.54000.04003.46529.0000AID1509468
Lysosomal alpha-glucosidaseHomo sapiens (human)IC50 (µMol)0.54000.06002.28897.8000AID1509468
Sucrase-isomaltase, intestinalHomo sapiens (human)IC50 (µMol)0.54000.04902.72947.8000AID1509468
Glycogen synthase kinase-3 alphaHomo sapiens (human)IC50 (µMol)100.00000.00101.22499.1000AID282721
Glycogen synthase kinase-3 betaHomo sapiens (human)IC50 (µMol)100.00000.00060.801310.0000AID282721
Probable maltase-glucoamylase 2Homo sapiens (human)IC50 (µMol)0.54000.54004.02447.8000AID1509468
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (120)

Processvia Protein(s)Taxonomy
maltose catabolic processMaltase-glucoamylase, intestinalHomo sapiens (human)
starch catabolic processMaltase-glucoamylase, intestinalHomo sapiens (human)
dextrin catabolic processMaltase-glucoamylase, intestinalHomo sapiens (human)
maltose metabolic processLysosomal alpha-glucosidaseHomo sapiens (human)
regulation of the force of heart contractionLysosomal alpha-glucosidaseHomo sapiens (human)
diaphragm contractionLysosomal alpha-glucosidaseHomo sapiens (human)
heart morphogenesisLysosomal alpha-glucosidaseHomo sapiens (human)
glycogen catabolic processLysosomal alpha-glucosidaseHomo sapiens (human)
sucrose metabolic processLysosomal alpha-glucosidaseHomo sapiens (human)
glucose metabolic processLysosomal alpha-glucosidaseHomo sapiens (human)
lysosome organizationLysosomal alpha-glucosidaseHomo sapiens (human)
locomotory behaviorLysosomal alpha-glucosidaseHomo sapiens (human)
tissue developmentLysosomal alpha-glucosidaseHomo sapiens (human)
aorta developmentLysosomal alpha-glucosidaseHomo sapiens (human)
vacuolar sequesteringLysosomal alpha-glucosidaseHomo sapiens (human)
muscle cell cellular homeostasisLysosomal alpha-glucosidaseHomo sapiens (human)
neuromuscular process controlling postureLysosomal alpha-glucosidaseHomo sapiens (human)
neuromuscular process controlling balanceLysosomal alpha-glucosidaseHomo sapiens (human)
cardiac muscle contractionLysosomal alpha-glucosidaseHomo sapiens (human)
glycophagyLysosomal alpha-glucosidaseHomo sapiens (human)
sucrose catabolic processSucrase-isomaltase, intestinalHomo sapiens (human)
polysaccharide digestionSucrase-isomaltase, intestinalHomo sapiens (human)
regulation of systemic arterial blood pressureGlycogen synthase kinase-3 alphaHomo sapiens (human)
cardiac left ventricle morphogenesisGlycogen synthase kinase-3 alphaHomo sapiens (human)
glycogen metabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
protein phosphorylationGlycogen synthase kinase-3 alphaHomo sapiens (human)
dopamine receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
nervous system developmentGlycogen synthase kinase-3 alphaHomo sapiens (human)
insulin receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of autophagyGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of gene expressionGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of peptidyl-threonine phosphorylationGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of UDP-glucose catabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
Wnt signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
cell migrationGlycogen synthase kinase-3 alphaHomo sapiens (human)
peptidyl-threonine phosphorylationGlycogen synthase kinase-3 alphaHomo sapiens (human)
viral protein processingGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of protein ubiquitinationGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of TOR signalingGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
cellular response to insulin stimulusGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationGlycogen synthase kinase-3 alphaHomo sapiens (human)
cellular response to interleukin-3Glycogen synthase kinase-3 alphaHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of glycogen biosynthetic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of protein catabolic processGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of heart contractionGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of glucose importGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of insulin receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
excitatory postsynaptic potentialGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of cell growth involved in cardiac muscle cell developmentGlycogen synthase kinase-3 alphaHomo sapiens (human)
cellular response to lithium ionGlycogen synthase kinase-3 alphaHomo sapiens (human)
cellular response to glucocorticoid stimulusGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of adenylate cyclase-activating adrenergic receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of canonical Wnt signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
extrinsic apoptotic signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
extrinsic apoptotic signaling pathway in absence of ligandGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of adenylate cyclase-activating G protein-coupled receptor signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
autosome genomic imprintingGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of mitochondrial outer membrane permeabilization involved in apoptotic signaling pathwayGlycogen synthase kinase-3 alphaHomo sapiens (human)
regulation of mitophagyGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of amyloid-beta formationGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of protein targeting to mitochondrionGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of glycogen synthase activity, transferring glucose-1-phosphateGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of type B pancreatic cell developmentGlycogen synthase kinase-3 alphaHomo sapiens (human)
negative regulation of glycogen (starch) synthase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of glycogen (starch) synthase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
cell differentiationGlycogen synthase kinase-3 alphaHomo sapiens (human)
regulation of microtubule cytoskeleton organizationGlycogen synthase kinase-3 alphaHomo sapiens (human)
regulation of neuron projection developmentGlycogen synthase kinase-3 alphaHomo sapiens (human)
positive regulation of gene expressionGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of gene expressionGlycogen synthase kinase-3 betaHomo sapiens (human)
ER overload responseGlycogen synthase kinase-3 betaHomo sapiens (human)
peptidyl-serine phosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
intracellular signal transductionGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of apoptotic processGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein export from nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
epithelial to mesenchymal transitionGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of cell-matrix adhesionGlycogen synthase kinase-3 betaHomo sapiens (human)
glycogen metabolic processGlycogen synthase kinase-3 betaHomo sapiens (human)
protein phosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
mitochondrion organizationGlycogen synthase kinase-3 betaHomo sapiens (human)
dopamine receptor signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
circadian rhythmGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of autophagyGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of gene expressionGlycogen synthase kinase-3 betaHomo sapiens (human)
peptidyl-serine phosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
peptidyl-threonine phosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
viral protein processingGlycogen synthase kinase-3 betaHomo sapiens (human)
hippocampus developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
establishment of cell polarityGlycogen synthase kinase-3 betaHomo sapiens (human)
maintenance of cell polarityGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of cell migrationGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of axon extensionGlycogen synthase kinase-3 betaHomo sapiens (human)
neuron projection developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of protein-containing complex assemblyGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein-containing complex assemblyGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein ubiquitinationGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of phosphoprotein phosphatase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of microtubule-based processGlycogen synthase kinase-3 betaHomo sapiens (human)
intracellular signal transductionGlycogen synthase kinase-3 betaHomo sapiens (human)
cellular response to interleukin-3Glycogen synthase kinase-3 betaHomo sapiens (human)
regulation of circadian rhythmGlycogen synthase kinase-3 betaHomo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of GTPase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of cell differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of osteoblast differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of glycogen biosynthetic processGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of cilium assemblyGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein catabolic processGlycogen synthase kinase-3 betaHomo sapiens (human)
protein autophosphorylationGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of protein export from nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of dendrite morphogenesisGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of axonogenesisGlycogen synthase kinase-3 betaHomo sapiens (human)
canonical Wnt signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
excitatory postsynaptic potentialGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of microtubule cytoskeleton organizationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of calcineurin-NFAT signaling cascadeGlycogen synthase kinase-3 betaHomo sapiens (human)
superior temporal gyrus developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
cellular response to retinoic acidGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of canonical Wnt signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
extrinsic apoptotic signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
extrinsic apoptotic signaling pathway in absence of ligandGlycogen synthase kinase-3 betaHomo sapiens (human)
presynaptic modulation of chemical synaptic transmissionGlycogen synthase kinase-3 betaHomo sapiens (human)
neuron projection organizationGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of microtubule anchoring at centrosomeGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of cellular response to heatGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of protein localization to nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of long-term synaptic potentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of mitochondrial outer membrane permeabilization involved in apoptotic signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of protein acetylationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of extrinsic apoptotic signaling pathway via death domain receptorsGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein localization to ciliumGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of dopaminergic neuron differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
cellular response to amyloid-betaGlycogen synthase kinase-3 betaHomo sapiens (human)
positive regulation of protein localization to centrosomeGlycogen synthase kinase-3 betaHomo sapiens (human)
beta-catenin destruction complex disassemblyGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of type B pancreatic cell developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of glycogen (starch) synthase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of mesenchymal stem cell differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
negative regulation of TOR signalingGlycogen synthase kinase-3 betaHomo sapiens (human)
regulation of neuron projection developmentGlycogen synthase kinase-3 betaHomo sapiens (human)
cell differentiationGlycogen synthase kinase-3 betaHomo sapiens (human)
insulin receptor signaling pathwayGlycogen synthase kinase-3 betaHomo sapiens (human)
carbohydrate metabolic processProbable maltase-glucoamylase 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (27)

Processvia Protein(s)Taxonomy
catalytic activityMaltase-glucoamylase, intestinalHomo sapiens (human)
glucan 1,4-alpha-glucosidase activityMaltase-glucoamylase, intestinalHomo sapiens (human)
alpha-1,4-glucosidase activityMaltase-glucoamylase, intestinalHomo sapiens (human)
protein bindingMaltase-glucoamylase, intestinalHomo sapiens (human)
amylase activityMaltase-glucoamylase, intestinalHomo sapiens (human)
carbohydrate bindingMaltase-glucoamylase, intestinalHomo sapiens (human)
maltose alpha-glucosidase activityMaltase-glucoamylase, intestinalHomo sapiens (human)
alpha-1,4-glucosidase activityLysosomal alpha-glucosidaseHomo sapiens (human)
carbohydrate bindingLysosomal alpha-glucosidaseHomo sapiens (human)
maltose alpha-glucosidase activityLysosomal alpha-glucosidaseHomo sapiens (human)
alpha-glucosidase activityLysosomal alpha-glucosidaseHomo sapiens (human)
oligo-1,6-glucosidase activitySucrase-isomaltase, intestinalHomo sapiens (human)
sucrose alpha-glucosidase activitySucrase-isomaltase, intestinalHomo sapiens (human)
protein bindingSucrase-isomaltase, intestinalHomo sapiens (human)
carbohydrate bindingSucrase-isomaltase, intestinalHomo sapiens (human)
alpha-1,4-glucosidase activitySucrase-isomaltase, intestinalHomo sapiens (human)
protein serine/threonine kinase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
signaling receptor bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
protein bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
ATP bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
protein kinase A catalytic subunit bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
tau protein bindingGlycogen synthase kinase-3 alphaHomo sapiens (human)
tau-protein kinase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
protein serine kinase activityGlycogen synthase kinase-3 alphaHomo sapiens (human)
protease bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
p53 bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
protein kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
protein serine/threonine kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
protein bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
ATP bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
beta-catenin bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
protein kinase bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
ubiquitin protein ligase bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
protein kinase A catalytic subunit bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
dynactin bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
tau protein bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
tau-protein kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
NF-kappaB bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingGlycogen synthase kinase-3 betaHomo sapiens (human)
protein serine kinase activityGlycogen synthase kinase-3 betaHomo sapiens (human)
glucan 1,4-alpha-glucosidase activityProbable maltase-glucoamylase 2Homo sapiens (human)
carbohydrate bindingProbable maltase-glucoamylase 2Homo sapiens (human)
alpha-1,4-glucosidase activityProbable maltase-glucoamylase 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (30)

Processvia Protein(s)Taxonomy
plasma membraneMaltase-glucoamylase, intestinalHomo sapiens (human)
apical plasma membraneMaltase-glucoamylase, intestinalHomo sapiens (human)
extracellular exosomeMaltase-glucoamylase, intestinalHomo sapiens (human)
tertiary granule membraneMaltase-glucoamylase, intestinalHomo sapiens (human)
ficolin-1-rich granule membraneMaltase-glucoamylase, intestinalHomo sapiens (human)
lysosomeLysosomal alpha-glucosidaseHomo sapiens (human)
lysosomal membraneLysosomal alpha-glucosidaseHomo sapiens (human)
plasma membraneLysosomal alpha-glucosidaseHomo sapiens (human)
membraneLysosomal alpha-glucosidaseHomo sapiens (human)
azurophil granule membraneLysosomal alpha-glucosidaseHomo sapiens (human)
lysosomal lumenLysosomal alpha-glucosidaseHomo sapiens (human)
intracellular membrane-bounded organelleLysosomal alpha-glucosidaseHomo sapiens (human)
extracellular exosomeLysosomal alpha-glucosidaseHomo sapiens (human)
tertiary granule membraneLysosomal alpha-glucosidaseHomo sapiens (human)
ficolin-1-rich granule membraneLysosomal alpha-glucosidaseHomo sapiens (human)
autolysosome lumenLysosomal alpha-glucosidaseHomo sapiens (human)
Golgi apparatusSucrase-isomaltase, intestinalHomo sapiens (human)
plasma membraneSucrase-isomaltase, intestinalHomo sapiens (human)
brush borderSucrase-isomaltase, intestinalHomo sapiens (human)
apical plasma membraneSucrase-isomaltase, intestinalHomo sapiens (human)
extracellular exosomeSucrase-isomaltase, intestinalHomo sapiens (human)
mitochondrionGlycogen synthase kinase-3 alphaHomo sapiens (human)
cytosolGlycogen synthase kinase-3 alphaHomo sapiens (human)
beta-catenin destruction complexGlycogen synthase kinase-3 alphaHomo sapiens (human)
neuronal cell bodyGlycogen synthase kinase-3 alphaHomo sapiens (human)
apical dendriteGlycogen synthase kinase-3 alphaHomo sapiens (human)
postsynapseGlycogen synthase kinase-3 alphaHomo sapiens (human)
proximal dendriteGlycogen synthase kinase-3 alphaHomo sapiens (human)
cytoplasmGlycogen synthase kinase-3 alphaHomo sapiens (human)
nucleusGlycogen synthase kinase-3 alphaHomo sapiens (human)
axonGlycogen synthase kinase-3 alphaHomo sapiens (human)
cytosolGlycogen synthase kinase-3 alphaHomo sapiens (human)
glutamatergic synapseGlycogen synthase kinase-3 betaHomo sapiens (human)
nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
nucleoplasmGlycogen synthase kinase-3 betaHomo sapiens (human)
cytoplasmGlycogen synthase kinase-3 betaHomo sapiens (human)
mitochondrionGlycogen synthase kinase-3 betaHomo sapiens (human)
centrosomeGlycogen synthase kinase-3 betaHomo sapiens (human)
cytosolGlycogen synthase kinase-3 betaHomo sapiens (human)
plasma membraneGlycogen synthase kinase-3 betaHomo sapiens (human)
axonGlycogen synthase kinase-3 betaHomo sapiens (human)
dendriteGlycogen synthase kinase-3 betaHomo sapiens (human)
beta-catenin destruction complexGlycogen synthase kinase-3 betaHomo sapiens (human)
presynapseGlycogen synthase kinase-3 betaHomo sapiens (human)
postsynapseGlycogen synthase kinase-3 betaHomo sapiens (human)
Wnt signalosomeGlycogen synthase kinase-3 betaHomo sapiens (human)
cytosolGlycogen synthase kinase-3 betaHomo sapiens (human)
axonGlycogen synthase kinase-3 betaHomo sapiens (human)
nucleusGlycogen synthase kinase-3 betaHomo sapiens (human)
cytoplasmGlycogen synthase kinase-3 betaHomo sapiens (human)
membraneProbable maltase-glucoamylase 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (18)

Assay IDTitleYearJournalArticle
AID648279Inhibition of Escherichia coli MurA using PEP as substrate assessed as inorganic phosphate release at 25 uM preincubated for 10 mins measured after 60 mins by fluorimetric assay2012Journal of medicinal chemistry, Jan-26, Volume: 55, Issue:2
Privileged scaffolds or promiscuous binders: a comparative study on rhodanines and related heterocycles in medicinal chemistry.
AID282721Inhibition of rabbit GSK32005Journal of medicinal chemistry, Nov-17, Volume: 48, Issue:23
SAR and 3D-QSAR studies on thiadiazolidinone derivatives: exploration of structural requirements for glycogen synthase kinase 3 inhibitors.
AID420047Antimicrobial activity against Bacillus subtilis UFPEDA 16 at 300 ug/disk after 24 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID1509468Inhibition of alpha-glucosidase (unknown origin)2019European journal of medicinal chemistry, Aug-15, Volume: 176Synthetic heterocyclic candidates as promising α-glucosidase inhibitors: An overview.
AID420046Antimicrobial activity against Staphylococcus aureus UFPEDA 01 at 300 ug/disk after 24 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID420045Antimicrobial activity against Escherichia coli UFPEDA 224 at 300 ug/disk after 24 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID648277Inhibition of Dengue virus NS2B-NS3 protease using Abz-NleKRRS-3-(NO2)Y as substrate at 50 uM preincubated for 15 mins measured every sec for 15 mins by fluorimetric analysis2012Journal of medicinal chemistry, Jan-26, Volume: 55, Issue:2
Privileged scaffolds or promiscuous binders: a comparative study on rhodanines and related heterocycles in medicinal chemistry.
AID648278Inhibition of bovine plasma thrombin using Boc-Val-Pro-Arg-AMC as substrate at 25 uM preincubated for 15 mins measured after 10 mins by fluorimetric assay2012Journal of medicinal chemistry, Jan-26, Volume: 55, Issue:2
Privileged scaffolds or promiscuous binders: a comparative study on rhodanines and related heterocycles in medicinal chemistry.
AID39222In vitro inhibition of recombinant Mycobacterium smegmatis Arylamine N-Acetyltransferase (MSNAT) -catalyzed acetylation of isoniazid (INH); not active2003Bioorganic & medicinal chemistry letters, Aug-04, Volume: 13, Issue:15
Synthesis and in vitro evaluation of novel small molecule inhibitors of bacterial arylamine N-acetyltransferases (NATs).
AID420044Antimicrobial activity against Serratia marcescens UFPEDA 398 at 300 ug/disk after 24 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID420050Antimicrobial activity against Pseudomonas aeruginosa UFPEDA 39 at 300 ug/disk after 24 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID648280Inhibition of Escherichia coli MetAP at 10 uM after 15 mins by fluorescence analysis2012Journal of medicinal chemistry, Jan-26, Volume: 55, Issue:2
Privileged scaffolds or promiscuous binders: a comparative study on rhodanines and related heterocycles in medicinal chemistry.
AID420048Antimicrobial activity against Micrococcus luteus UFPEDA 06 at 300 ug/disk after 24 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID420052Antimicrobial activity against Candida albicans UFPEDA 1007 at 300 ug/disk after 48 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID420049Antimicrobial activity against Enterococcus faecalis UFPEDA 138 at 300 ug/disk after 24 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID420051Antimicrobial activity against Mycobacterium smegmatis UFPEDA 71 at 300 ug/disk after 24 hrs by disk diffusion method2009European journal of medicinal chemistry, May, Volume: 44, Issue:5
Synthesis, antimicrobial and cytotoxic activities of some 5-arylidene-4-thioxo-thiazolidine-2-ones.
AID738342Dissociation constant, pKa of the compound2013Bioorganic & medicinal chemistry, Apr-01, Volume: 21, Issue:7
Thiazolidine-2,4-diones: progress towards multifarious applications.
AID1159607Screen for inhibitors of RMI FANCM (MM2) intereaction2016Journal of biomolecular screening, Jul, Volume: 21, Issue:6
A High-Throughput Screening Strategy to Identify Protein-Protein Interaction Inhibitors That Block the Fanconi Anemia DNA Repair Pathway.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (765)

TimeframeStudies, This Drug (%)All Drugs %
pre-19903 (0.39)18.7374
1990's35 (4.58)18.2507
2000's397 (51.90)29.6817
2010's252 (32.94)24.3611
2020's78 (10.20)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 26.71

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 moderate demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index26.71 (24.57)
Research Supply Index6.72 (2.92)
Research Growth Index6.25 (4.65)
Search Engine Demand Index34.37 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (26.71)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials33 (4.14%)5.53%
Reviews221 (27.69%)6.00%
Case Studies12 (1.50%)4.05%
Observational3 (0.38%)0.25%
Other529 (66.29%)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
Incretin-based Drugs and the Risk of Heart Failure: A Multi-center Network Observational Study [NCT02456428]1,499,650 participants (Actual)Observational2014-03-31Completed
The Use of Incretin-based Drugs and the Risk of Acute Pancreatitis in Patients With Type 2 Diabetes [NCT02476760]1,417,914 participants (Actual)Observational2014-03-31Completed
Cardiovascular Outcomes in Patients With Type 2 Diabetes Mellitus [NCT03249506]25,358 participants (Actual)Observational2016-05-12Completed
Pathophysiology of Uric Acid Nephrolithiasis [NCT00904046]60 participants (Anticipated)Interventional2019-09-05Recruiting
Comparison of Canagliflozin vs. Alternative Antihyperglycemic Treatments on Risk of Heart Failure Hospitalization and Amputation for Patients With Type 2 Diabetes Mellitus and the Subpopulation With Established Cardiovascular Disease [NCT03492580]714,582 participants (Actual)Observational2018-02-22Completed
Bypass Angioplasty Revascularization Investigation in Type 2 Diabetes [NCT00006305]Phase 32,368 participants (Actual)Interventional2000-09-30Completed
Role of Pioglitazone and Berberine in Treatment of Non-alcoholic Fatty Liver Disease(NAFLD) Patients With Impaired Glucose Regulation or Type 2 Diabetes Mellitus [NCT00633282]Phase 2184 participants (Actual)Interventional2008-03-31Completed
Liraglutide Improve Cognitive Function in Patients With Type 2 Diabetes Mellitus [NCT05360147]Phase 330 participants (Actual)Interventional2021-01-20Completed
A Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel Group, Phase 3 Trial to Evaluate the Safety and Efficacy of Dapagliflozin in Combination With Thiazolidinedione Therapy in Subjects With Type 2 Diabetes Who Have Inadequate Glycemic Con [NCT00683878]Phase 3972 participants (Actual)Interventional2008-07-31Completed
Islet Transplantation in Type 1 Diabetic Patients Using a Steroid-free Immunosuppression Protocol and Thiazolidinedione Insulin Sensitizers [NCT00214253]Phase 114 participants (Actual)Interventional2002-02-28Completed
EXCEED - A Pan-European Post-Authorisation Safety Study: Risk of Pancreatic Cancer Among Type 2 Diabetes Patients Who Initiated Exenatide as Compared With Those Who Initiated Other Non-Glucagon-Like Peptide 1 Receptor Agonists Based Glucose Lowering Drugs [NCT05663515]2,400 participants (Anticipated)Observational2024-01-01Not yet recruiting
A Randomized, Placebo-controlled, 2-arm Parallel-group, Multicenter Study With a 24-week Double-blind Treatment Period Assessing the Efficacy and Safety of Lixisenatide in Patients With Type 2 Diabetes Insufficiently Controlled With Insulin Glargine and M [NCT00975286]Phase 3446 participants (Actual)Interventional2009-10-31Completed
The Echocardiographic Left Ventricular Functional Changes of Uncontrolled Diabetes by the Intervention of Dapagliflozin Treatment Trial (ELUCIDATE) [NCT03871621]Phase 476 participants (Actual)Interventional2019-04-01Completed
Integrating the Genetic and Metabolic Faces of Obesity [NCT00285844]Phase 188 participants (Actual)Interventional2005-10-31Completed
A National, Multicenter, Prospective, Interventional, Open-label, Single-arm, 24-Week Phase IV Study to Evaluate the Effectiveness and Safety of Initiation and Titration of Insulin Glargine U300 in Insulin-naïve Patients With T2DM Inadequately Controlled [NCT02954692]Phase 4112 participants (Actual)Interventional2016-11-30Completed
Substituting Lantus®(Insulin Glargine[rDNAorigin]Inj) for a Thiazolidinedione vs. a 3rd Oral Agent as add-on Therapy in Patients Failing a Thiazolidinedione & Sulfonylurea or Metformin Combination [NCT00046462]Phase 3240 participants Interventional2001-11-30Completed
A Multi-center, Randomized, Open-label, Active Controlled, Parallel Arm Study to Compare the Efficacy of 12 Weeks of Treatment With Vildagliptin 100 mg, qd to Thiazolidinedione (TZD) as add-on Therapy in Patients With Type 2 Diabetes Inadequately Controll [NCT00396227]Phase 32,665 participants (Actual)Interventional2006-10-31Completed
Effects of Elevated Free Fatty Acids on Endogenous Glucose Production in People With and Without Type 2 Diabetes Mellitus [NCT00308373]73 participants (Actual)Interventional2004-07-31Completed
Proteus Sustained Behavior Change Study [NCT01503008]0 participants (Actual)Interventional2012-02-29Withdrawn
Efficacy in Controlling Glycaemia With Victoza® (Liraglutide) as add-on to Metformin vs. OADs as add-on to Metformin After up to 104 Weeks of Treatment in Subjects With Type 2 Diabetes Inadequately Controlled With Metformin Monotherapy and Treated in a Pr [NCT02730377]Phase 41,991 participants (Actual)Interventional2016-03-28Completed
Ghrelin Regulation and Structure: Effect of Thiazolidinedione Therapy on Ghrelin [NCT00843791]6 participants (Actual)Interventional2009-02-28Terminated(stopped due to Insufficient funds to continue.)
A 52-Week, Open-Label, Multicenter Study to Determine the Long Term Safety and Efficacy of Albiglutide in Combination With Monotherapy of Oral Antihyperglycemic Medications in Japanese Patients With Type 2 Diabetes Mellitus [NCT01777282]Phase 3374 participants (Actual)Interventional2013-02-23Completed
A 52-Week, Open-Label, Long-Term Safety Study of LY2189265 in Combination With Monotherapy of Oral Antihyperglycemic Medications in Patients With Type 2 Diabetes Mellitus [NCT01468181]Phase 3394 participants (Actual)Interventional2011-11-30Completed
Dapagliflozin Effect in Cognitive Impairment in Stroke Trial [NCT05565976]Phase 2/Phase 3270 participants (Anticipated)Interventional2020-08-01Recruiting
The Use of Incretin-based Drugs and the Risk of Pancreatic Cancer in Patients With Type 2 Diabetes [NCT02475499]886,172 participants (Actual)Observational2014-03-31Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00006305 (2) [back to overview]Number of Participants With All-Cause Mortality
NCT00006305 (2) [back to overview]Number of Participants With Death, Myocardial Infarction, or Stroke
NCT00683878 (7) [back to overview]Adjusted Mean Change From Baseline in 120-minute Post-challenge Plasma Glucose (PPG) (mg/dL) at Week 24 (Last Observation Carried Forward [LOCF])
NCT00683878 (7) [back to overview]Adjusted Mean Change From Baseline in Fasting Plasma Glucose (FPG) at Week 24 (Last Observation Carried Forward [LOCF])
NCT00683878 (7) [back to overview]Adjusted Mean Change From Baseline in Hemoglobin A1C (HbA1c) at Week 24 (Last Observation Carried Forward [LOCF])
NCT00683878 (7) [back to overview]Adjusted Mean Change From Baseline in Total Body Weight (kg) Among Subjects With Baseline Body Mass Index (BMI) ≥ 27 kg/m^2 at Week 24 (Last Observation Carried Forward [LOCF])
NCT00683878 (7) [back to overview]Adjusted Mean Change From Baseline in Total Body Weight (kg) at Week 24 (Last Observation Carried Forward [LOCF])
NCT00683878 (7) [back to overview]Adjusted Mean Change From Baseline in Waist Circumference (cm) at Week 24 (Last Observation Carried Forward [LOCF])
NCT00683878 (7) [back to overview]Percentage of Participants Achieving a Therapeutic Glycemic Response (Hemoglobin A1c [HbA1C]) <7.0% at Week 24 (Last Observation Carried Forward [LOCF])
NCT00975286 (13) [back to overview]Absolute Change From Baseline in Glycosylated Hemoglobin (HbA1c) at Week 24
NCT00975286 (13) [back to overview]Change From Baseline in 2-Hour Postprandial Plasma Glucose (PPG) at Week 24
NCT00975286 (13) [back to overview]Change From Baseline in Average 7-Point Self Monitored Plasma Glucose (SMPG) Profile at Week 24
NCT00975286 (13) [back to overview]Change From Baseline in Average Insulin Glargine Daily Dose at Week 24
NCT00975286 (13) [back to overview]Change From Baseline in Body Weight at Week 24
NCT00975286 (13) [back to overview]Change From Baseline in Fasting Plasma Glucose (FPG) at Week 24
NCT00975286 (13) [back to overview]Change From Baseline in Glucose Excursion at Week 24
NCT00975286 (13) [back to overview]Change From Baseline in Treatment Satisfaction Score (Sum of Items 1, 4, 5, 6, 7 and 8 of DTSQ) at Week 24
NCT00975286 (13) [back to overview]Percentage of Patients Requiring Rescue Therapy During the Double-blind Period
NCT00975286 (13) [back to overview]Percentage of Patients With at Least 5% Weight Loss From Baseline at Week 24
NCT00975286 (13) [back to overview]Percentage of Patients With Glycosylated Hemoglobin (HbA1c) Level Less Than 7% at Week 24
NCT00975286 (13) [back to overview]Percentage of Patients With Glycosylated Hemoglobin (HbA1c) Level Less Than or Equal to 6.5% at Week 24
NCT00975286 (13) [back to overview]Number of Patients With Symptomatic Hypoglycemia and Severe Symptomatic Hypoglycemia
NCT01468181 (8) [back to overview]Percentage of Participants With Treatment-Emergent Adverse Events (TEAEs)
NCT01468181 (8) [back to overview]Change From Baseline in 7-Point Self-Monitored Blood Glucose (SMBG)
NCT01468181 (8) [back to overview]Change From Baseline in Body Weight
NCT01468181 (8) [back to overview]Change From Baseline in Fasting Blood Glucose (FBG)
NCT01468181 (8) [back to overview]Change From Baseline in Glycosylated Hemoglobin (HbA1c)
NCT01468181 (8) [back to overview]Change From Baseline in Updated Homeostasis Model Assessment (HOMA2)
NCT01468181 (8) [back to overview]Percentage of Participants Who Achieve HbA1c ≤6.5% or <7%
NCT01468181 (8) [back to overview]Percentage of Participants With Hypoglycemic Episodes
NCT01777282 (7) [back to overview]Change From Baseline in Fasting Plasma Glucose (FPG) at Week 52
NCT01777282 (7) [back to overview]Change From Baseline in Glycosylated Hemoglobin (HbA1c) at Week 52
NCT01777282 (7) [back to overview]Number of Participants With Any Hypoglycemic Event
NCT01777282 (7) [back to overview]Time to Study Withdrawal Due to Hyperglycemia
NCT01777282 (7) [back to overview]Number of Participants With Any Adverse Event (AE) or Any Serious Adverse Event (SAE)
NCT01777282 (7) [back to overview]Percentage of Participants Achieving Clinically Meaningful Levels of HbA1c (i.e., the Percentage of Participants Achieving Treatment Goal of <6.5% and <7.0% at Week 52)
NCT01777282 (7) [back to overview]Change From Baseline in Body Weight at Week 52
NCT02730377 (19) [back to overview]Number of AEs Leading to Permanent Discontinuation of Trial Product
NCT02730377 (19) [back to overview]Number of Documented Symptomatic Hypoglycaemic Episodes (ADA)
NCT02730377 (19) [back to overview]Number of Serious Adverse Events (SAEs)
NCT02730377 (19) [back to overview]Number of Severe Hypoglycaemic Episodes
NCT02730377 (19) [back to overview]Number of Severe or BG Confirmed Symptomatic Hypoglycaemic Episodes
NCT02730377 (19) [back to overview]Time to Inadequate Glycaemic Control
NCT02730377 (19) [back to overview]Change in Haemoglobin
NCT02730377 (19) [back to overview]Change in HbA1c
NCT02730377 (19) [back to overview]Change in Lipids: HDL Cholesterol, LDL-cholesterol, Total Cholesterol, Triglycerides
NCT02730377 (19) [back to overview]Change in Potassium
NCT02730377 (19) [back to overview]Change in Pulse
NCT02730377 (19) [back to overview]Change in Biochemistry- Alanine Aminotransferase (ALAT), Amylase, Aspartate Aminotransferase (ASAT), Lipase
NCT02730377 (19) [back to overview]Time to Premature Treatment Discontinuation (for Any Reason Including Inadequate Glycaemic Control)
NCT02730377 (19) [back to overview]Change in Biochemistry- Creatinine, Total Bilirubin
NCT02730377 (19) [back to overview]Change in Biochemistry- Estimated Glomerular Filtration Rate (eGFR) Serum
NCT02730377 (19) [back to overview]Change in Blood Pressure (Systolic and Diastolic Blood Pressure)
NCT02730377 (19) [back to overview]Change in Body Mass Index (BMI)
NCT02730377 (19) [back to overview]Change in Body Weight
NCT02730377 (19) [back to overview]Change in Fasting Plasma Glucose (FPG)

Number of Participants With All-Cause Mortality

(NCT00006305)
Timeframe: five years

Interventionparticipants (Number)
Revascularization and Insulin Providing (IP)80
Revascularization and Insulin Sensitizing (IS)75
Medical Therapy and Insulin Providing (IP)80
Medical Therapy and Insulin Sensitizing (IS)81

[back to top]

Number of Participants With Death, Myocardial Infarction, or Stroke

(NCT00006305)
Timeframe: five years

Interventionparticipants (Number)
Revascularization and Insulin Providing (IP)145
Revascularization and Insulin Sensitizing (IS)121
Medical Therapy and Insulin Providing (IP)143
Medical Therapy and Insulin Sensitizing (IS)140

[back to top]

Adjusted Mean Change From Baseline in 120-minute Post-challenge Plasma Glucose (PPG) (mg/dL) at Week 24 (Last Observation Carried Forward [LOCF])

Secondary endpoints were tested using sequential testing procedure and are presented in hierarchical order. In post oral glucose tolerance test (OGTT), glucose was measured as milligrams per deciliter(mg/dL) by a central laboratory. Baseline was defined as the last assessment prior to the start date and time of the first dose of the double-blind study medication. In cases where time of the first dose or time of the assessment was not available, baseline was defined as the last assessment on or prior to the date of the first dose of the double-blind study medication. PPG measurements were obtained on Day 1 and week 24 in the double-blind period. (NCT00683878)
Timeframe: From Baseline to Week 24

Interventionmg/dL (Mean)
PLACEBO + Pioglitazone-14.1
Dapagliflozin 5MG + Pioglitazone-65.1
Dapagliflozin 10MG + Pioglitazone-67.5

[back to top]

Adjusted Mean Change From Baseline in Fasting Plasma Glucose (FPG) at Week 24 (Last Observation Carried Forward [LOCF])

Secondary endpoints were tested using sequential testing procedure and are presented in hierarchical order. Fasting plasma glucose was measured as milligrams per deciliter(mg/dL) by a central laboratory. Baseline was defined as the last assessment prior to the start date and time of the first dose of the double-blind study medication. In cases where time of the first dose or time of the assessment was not available, baseline was defined as the last assessment on or prior to the date of the first dose of the double-blind study medication. FPG measurements were obtained during the qualification and lead-in periods and on Day 1 and Weeks 1, 2, 4, 8, 12, 16, 20, and 24 in the double-blind period. (NCT00683878)
Timeframe: From Baseline to Week 24

Interventionmg/dL (Mean)
PLACEBO + Pioglitazone-5.5
Dapagliflozin 5MG + Pioglitazone-24.9
Dapagliflozin 10MG + Pioglitazone-29.6

[back to top]

Adjusted Mean Change From Baseline in Hemoglobin A1C (HbA1c) at Week 24 (Last Observation Carried Forward [LOCF])

HbA1c was measured as percent of hemoglobin by a central laboratory. Data after rescue medication was excluded from this analysis. Baseline was defined as the last assessment prior to the start date and time of the first dose of the double-blind study medication. In cases where time of the first dose or time of the assessment was not available, baseline was defined as the last assessment on or prior to the date of the first dose of the double-blind study medication. HbA1c measurements were obtained during the qualification and lead-in periods and on Day 1 and Weeks 4, 8, 12, 16, 20, and 24 in the double-blind period. (NCT00683878)
Timeframe: From Baseline to Week 24

Intervention% of hemoglobin (Mean)
PLACEBO + Pioglitazone-0.42
Dapagliflozin 5MG + Pioglitazone-0.82
Dapagliflozin 10MG + Pioglitazone-0.97

[back to top]

Adjusted Mean Change From Baseline in Total Body Weight (kg) Among Subjects With Baseline Body Mass Index (BMI) ≥ 27 kg/m^2 at Week 24 (Last Observation Carried Forward [LOCF])

Secondary endpoints were tested using sequential testing procedure and are presented in hierarchical order. Adjusted mean change from baseline in total body weight among subjects with baseline body mass index (BMI) ≥ 27 kg/m^2 at Week 24 (or the last postbaseline measurement prior to Week 24 if no Week 24 assessment was available was determined. Data after rescue medication was excluded from this analysis. Baseline was defined as the last assessment prior to the start date and time of the first dose of the double-blind study medication. In cases where time of the first dose or time of the assessment was not available, baseline was defined as the last assessment on or prior to the date of the first dose of the double-blind study medication. Body weight measurements were obtained during the qualification and lead-in periods and on Day 1 and Weeks 1, 2, 4, 8, 12, 16, 20, and 24 of the double-blind period. (NCT00683878)
Timeframe: From Baseline to Week 24

Interventionkg (Mean)
PLACEBO + Pioglitazone1.83
Dapagliflozin 5MG + Pioglitazone0.26
Dapagliflozin 10MG + Pioglitazone-0.07

[back to top]

Adjusted Mean Change From Baseline in Total Body Weight (kg) at Week 24 (Last Observation Carried Forward [LOCF])

Secondary endpoints were tested using sequential testing procedure and are presented in hierarchical order. Adjusted mean change from baseline in total body weight at Week 24 (or the last postbaseline measurement prior to Week 24 if no Week 24 assessment was available was determined. Data after rescue medication was excluded from this analysis. Baseline was defined as the last assessment prior to the start date and time of the first dose of the double-blind study medication. In cases where time of the first dose or time of the assessment was not available, baseline was defined as the last assessment on or prior to the date of the first dose of the double-blind study medication. Body weight measurements were obtained during the qualification and lead-in periods and on Day 1 and Weeks 1, 2, 4, 8, 12, 16, 20, and 24 of the double-blind period. (NCT00683878)
Timeframe: From Baseline to Week 24

Interventionkg (Mean)
PLACEBO + Pioglitazone1.64
Dapagliflozin 5MG + Pioglitazone0.09
Dapagliflozin 10MG + Pioglitazone-0.14

[back to top]

Adjusted Mean Change From Baseline in Waist Circumference (cm) at Week 24 (Last Observation Carried Forward [LOCF])

Secondary endpoints were tested using sequential testing procedure and are presented in hierarchical order. Adjusted mean change from baseline in waist circumference at Week 24 (or the last postbaseline measurement prior to Week 24 if no Week 24 assessment was available was determined. Data after rescue medication was excluded from this analysis. Baseline was defined as the last assessment prior to the start date and time of the first dose of the double-blind study medication. In cases where time of the first dose or time of the assessment was not available, baseline was defined as the last assessment on or prior to the date of the first dose of the double-blind study medication. Waist circumference measurements were obtained during the qualification and lead-in periods and on Day 1 and Week 24 of the double-blind period. (NCT00683878)
Timeframe: From Baseline to Week 24

Interventioncm (Mean)
PLACEBO + Pioglitazone1.38
Dapagliflozin 5MG + Pioglitazone0.52
Dapagliflozin 10MG + Pioglitazone-0.17

[back to top]

Percentage of Participants Achieving a Therapeutic Glycemic Response (Hemoglobin A1c [HbA1C]) <7.0% at Week 24 (Last Observation Carried Forward [LOCF])

Secondary endpoints were tested using sequential testing procedure and are presented in hierarchical order. Percent adjusted for baseline HbA1c. Therapeutic glycemic response is defined as HbA1c <7.0%. Data after rescue medication was excluded from this analysis. HbA1c was measured as a percent of hemoglobin. Mean and standard error for percentage of participants were estimated by modified logistic regression model. (NCT00683878)
Timeframe: From Baseline to Week 24

InterventionPercentage of participants (Mean)
PLACEBO + Pioglitazone22.4
Dapagliflozin 5MG + Pioglitazone32.5
Dapagliflozin 10MG + Pioglitazone38.8

[back to top]

Absolute Change From Baseline in Glycosylated Hemoglobin (HbA1c) at Week 24

Absolute change = HbA1c value at Week 24 minus HbA1c value at baseline. The on-treatment period for this efficacy variable is the time from the first dose of study drug up to 14 days after the last dose of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionpercentage of hemoglobin (Least Squares Mean)
Placebo-0.40
Lixisenatide-0.71

[back to top]

Change From Baseline in 2-Hour Postprandial Plasma Glucose (PPG) at Week 24

The 2-hour PPG test measured blood glucose 2 hours after eating a standardized meal. Change was calculated by subtracting baseline value from Week 24 value. The on-treatment period for this efficacy variable is the time from the first dose of study drug up to the last dosing day of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionmmol/L (Least Squares Mean)
Placebo0.08
Lixisenatide-3.09

[back to top]

Change From Baseline in Average 7-Point Self Monitored Plasma Glucose (SMPG) Profile at Week 24

Patients recorded a 7-point plasma glucose profile measured before and 2 hours after each meal and at bedtime once in a week and the average value for the 7-time points was calculated. The on-treatment period for this efficacy variable is the time from the first dose of study drug up to the last dosing day of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionmmol/L (Least Squares Mean)
Placebo-0.08
Lixisenatide-0.47

[back to top]

Change From Baseline in Average Insulin Glargine Daily Dose at Week 24

Change was calculated by subtracting the baseline value from Week 24 value. The on-treatment period for this efficacy variable is the time from the first dose of study drug up to the last dosing day of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionunits per day (Least Squares Mean)
Placebo5.34
Lixisenatide3.10

[back to top]

Change From Baseline in Body Weight at Week 24

Change was calculated by subtracting baseline value from Week 24 value. The on-treatment period for this efficacy variable is the time from the first dose of study drug up to 3 days after the last dose of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionkilogram (Least Squares Mean)
Placebo1.16
Lixisenatide0.28

[back to top]

Change From Baseline in Fasting Plasma Glucose (FPG) at Week 24

Change was calculated by subtracting baseline value from Week 24 value. The on-treatment period for this efficacy variable is the time from the first dose of study drug up to 1 day after the last dose of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionmmol/L (Least Squares Mean)
Placebo0.46
Lixisenatide0.34

[back to top]

Change From Baseline in Glucose Excursion at Week 24

Glucose excursion = 2-hour PPG minus plasma glucose 30 minutes prior to the standardized meal test, before study drug administration. Change was calculated by subtracting baseline value from Week 24 value. The on-treatment period for this efficacy variable is the time from the first dose of study drug up to the last dosing day of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionmmol/L (Least Squares Mean)
Placebo-0.33
Lixisenatide-3.42

[back to top]

Change From Baseline in Treatment Satisfaction Score (Sum of Items 1, 4, 5, 6, 7 and 8 of DTSQ) at Week 24

Change was calculated by subtracting baseline value from Week 24 value. DTSQ: 8-item questionnaire to assess treatment satisfaction and patient perception of hyper and hypoglycemia. Each question (Q) scored on a Likert scale from 0 to 6. Six items (Q1 and 4-8; higher score = more satisfaction) measured treatment satisfaction and were summed to calculate treatment satisfaction score which ranged from 0 (very dissatisfied) to 36 (very satisfied). Two items (Q2 and 3), which were not included, measured perceived hyperglycemia and hypoglycemia, respectively and lower scores represented good perceived blood glucose control. The on-treatment period for this efficacy variable is the time from the first dose of study drug up to 3 days after the last dose of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionunits on a scale (Least Squares Mean)
Placebo0.65
Lixisenatide0.88

[back to top]

Percentage of Patients Requiring Rescue Therapy During the Double-blind Period

Routine fasting SMPG, central laboratory FPG and HbA1c values were used to determine the requirement of rescue medication. If fasting SMPG value exceeded the specified limit for 3 consecutive days, the central laboratory FPG and HbA1c were performed. Threshold values - from baseline to Week 8: fasting SMPG/FPG >200 milligram/deciliter (mg/dL) (11.1 mmol/L) or HbA1c >9%, from Week 8 to Week 24: fasting SMPG/FPG >180 mg/dL (10.0 mmol/L) or HbA1c >8.5%. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline up to Week 24

Interventionpercentage of participants (Number)
Placebo0.4
Lixisenatide0.4

[back to top]

Percentage of Patients With at Least 5% Weight Loss From Baseline at Week 24

The on-treatment period for this efficacy variable is the time from the first dose of study drug up to 3 days after the last dose of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Baseline, Week 24

Interventionpercentage of participants (Number)
Placebo3.2
Lixisenatide5.1

[back to top]

Percentage of Patients With Glycosylated Hemoglobin (HbA1c) Level Less Than 7% at Week 24

The on-treatment period for this efficacy variable is the time from the first dose of study drug up to 14 days after the last dose of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Week 24

Interventionpercentage of participants (Number)
Placebo38.5
Lixisenatide56.3

[back to top]

Percentage of Patients With Glycosylated Hemoglobin (HbA1c) Level Less Than or Equal to 6.5% at Week 24

The on-treatment period for this efficacy variable is the time from the first dose of study drug up to 14 days after the last dose of study drug or up to the introduction of rescue therapy, whichever is the earliest. For a patient to be included in mITT population, both baseline and at least 1 post baseline assessment for at least 1 efficacy variable, were required. (NCT00975286)
Timeframe: Week 24

Interventionpercentage of participants (Number)
Placebo16.3
Lixisenatide32.1

[back to top]

Number of Patients With Symptomatic Hypoglycemia and Severe Symptomatic Hypoglycemia

Symptomatic hypoglycemia was an event with clinical symptoms that were considered to result from a hypoglycemic episode with an accompanying plasma glucose less than 60 mg/dL (3.3 mmol/L) or associated with prompt recovery after oral carbohydrate, intravenous glucose, or glucagon administration if no plasma glucose measurement was available. Severe symptomatic hypoglycemia was symptomatic hypoglycemia event in which the patient required the assistance of another person and was associated with either a plasma glucose level below 36 mg/dL (2.0 mmol/L) or prompt recovery after oral carbohydrate, intravenous glucose, or glucagon administration, if no plasma glucose measurement was available. (NCT00975286)
Timeframe: First dose of study drug up to 3 days after the last dose administration

,
Interventionparticipants (Number)
Symptomatic hypoglycemiaSevere symptomatic hypoglycemia
Lixisenatide501
Placebo300

[back to top]

Percentage of Participants With Treatment-Emergent Adverse Events (TEAEs)

A TEAE was defined as an event that first occurs or worsens (increases in severity) after baseline, regardless of causality or severity. The percentage of participants with TEAEs was calculated by dividing the number of participants with at least 1 TEAE over the 52-week treatment period by the total number of participants analyzed, multiplied by 100%. A summary of serious and other non-serious adverse events, regardless of causality, is located in the Reported Adverse Events module. (NCT01468181)
Timeframe: Baseline through 52 Weeks

Interventionpercentage of participants (Number)
LY2189265 + SU85.5
LY2189265 + BG83.6
LY2189265 + a-GI70.8
LY2189265 + TZD80.3
LY2189265 + Glinides73.2

[back to top]

Change From Baseline in 7-Point Self-Monitored Blood Glucose (SMBG)

Participants were to test and record SMBG concentrations in their study diaries before each meal (breakfast, lunch, and dinner), approximately 2 hours after the start of each meal. For the mean of all 7-point blood glucose values, the daily mean was calculated as the average of 7 blood glucose values collected on a particular day. The mean of all 7-point blood glucose values at each visit was calculated as the average of 2 daily means. The change from baseline was calculated as the mean of all 7-point blood glucose values at endpoint minus the mean of all 7-point blood glucose values at baseline. (NCT01468181)
Timeframe: Baseline, up to 26 weeks and up to 52 weeks

,,,,
Interventionmg/dL (Mean)
Pre-morning mean at 26 Weeks (n=131,61,64,66,71)Pre-morning mean at 52 Weeks (n=131,61,64,66,71)2hr postmorning meal at 26Weeks(n=131,61,65,66,71)2hr postmorning meal at 52Weeks(n=131,61,65,66,71)Pre-midday meal at 26 Weeks (n=131,61,65,66,71)Pre-midday meal at 52 Weeks (n=131,61,65,66,71)2hr postmidday meal at 26 Weeks(n=131,60,65,66,71)2hr postmidday meal at 52 Weeks(n=131,60,65,66,71)Pre-evening meal at 26 Weeks (n=130,61,65,66,70)Pre-evening meal at 52 Weeks (n=130,61,65,66,70)2hr postevening meal at 26Weeks(n=128,61,65,66,69)2hr postevening meal at 52Weeks(n=128,61,65,66,69)Bedtime at 26 Weeks (n=128, 61, 65, 63, 67)Bedtime at 52 Weeks (n=128, 61, 65, 63, 67)
LY2189265 + a-GI-45.97-45.69-73.43-73.62-54.75-55.96-67.70-63.47-57.82-54.66-71.15-63.82-63.98-66.74
LY2189265 + BG-34.60-37.19-64.23-63.81-26.26-33.34-50.03-52.70-25.66-25.34-45.41-48.61-49.98-52.70
LY2189265 + Glinides-48.26-46.45-67.99-73.78-54.51-56.07-68.49-65.46-46.85-51.64-68.75-64.22-64.01-64.41
LY2189265 + SU-44.70-42.93-69.66-58.50-49.95-43.90-67.24-57.27-44.57-43.90-62.24-58.98-61.07-54.07
LY2189265 + TZD-35.40-39.83-65.73-67.51-51.80-50.21-80.02-81.64-43.22-42.15-66.14-69.30-66.19-68.42

[back to top]

Change From Baseline in Body Weight

(NCT01468181)
Timeframe: Baseline, up to 26 weeks and up to 52 weeks

,,,,
Interventionkilograms (kg) (Mean)
26 Weeks52 Weeks
LY2189265 + a-GI-1.22-1.24
LY2189265 + BG-0.74-0.87
LY2189265 + Glinides0.190.04
LY2189265 + SU0.020.10
LY2189265 + TZD0.781.02

[back to top]

Change From Baseline in Fasting Blood Glucose (FBG)

(NCT01468181)
Timeframe: Baseline, up to 26 weeks and up to 52 weeks

,,,,
Interventionmilligrams/deciliters (mg/dL) (Mean)
26 Weeks52 Weeks
LY2189265 + a-GI-46.8-47.0
LY2189265 + BG-37.9-36.0
LY2189265 + Glinides-42.7-45.8
LY2189265 + SU-46.8-43.2
LY2189265 + TZD-42.1-39.6

[back to top]

Change From Baseline in Glycosylated Hemoglobin (HbA1c)

(NCT01468181)
Timeframe: Baseline, up to 26 Weeks and up to 52 Weeks

,,,,
Interventionpercentage of HbA1c (Mean)
26 Weeks52 Weeks
LY2189265 + a-GI-1.67-1.65
LY2189265 + BG-1.58-1.57
LY2189265 + Glinides-1.80-1.65
LY2189265 + SU-1.93-1.67
LY2189265 + TZD-1.71-1.69

[back to top]

Change From Baseline in Updated Homeostasis Model Assessment (HOMA2)

The HOMA2 is a computer model that uses fasting plasma insulin and glucose concentrations to estimate steady state pancreatic beta cell function (%B) and to estimate insulin sensitivity (%S) as a percentage of a normal reference population (normal young adults). The normal reference population was set at 100%. The change from baseline for fasting insulin concentrations are presented as insulin secretion (HOMA2-%B) and insulin sensitivity (HOMA2-%S). (NCT01468181)
Timeframe: Baseline, up to 26 weeks and up to 52 weeks

,,,,
Interventionpercentage of HOMA2 (Mean)
HOMA2-%B, 26 WeeksHOMA2-%B, 52 WeeksHOMA2-%S, 26 WeeksHOMA2-%S, 52 Weeks
LY2189265 + a-GI30.8828.067.576.86
LY2189265 + BG28.1526.05-3.48-3.83
LY2189265 + Glinides26.9333.51-4.41-5.51
LY2189265 + SU29.1026.06-4.80-3.99
LY2189265 + TZD27.3425.38-5.500.47

[back to top]

Percentage of Participants Who Achieve HbA1c ≤6.5% or <7%

(NCT01468181)
Timeframe: 26 weeks and 52 weeks

,,,,
Interventionpercentage of participants (Number)
HbA1c ≤6.5% at Week 26HbA1c ≤6.5% at Week 52HbA1c <7.0% at Week 26HbA1c <7.0% at Week 52
LY2189265 + a-GI72.370.881.583.1
LY2189265 + BG54.157.473.873.8
LY2189265 + Glinides53.547.969.062.0
LY2189265 + SU35.131.361.848.9
LY2189265 + TZD60.657.672.777.3

[back to top]

Percentage of Participants With Hypoglycemic Episodes

The percentage of participants with hypoglycemic episodes was calculated by dividing the number of participants with at least 1 hypoglycemic episode over the 52-week treatment period by the total number of participants analyzed, multiplied by 100%. All classifications of hypoglycemia (documented symptomatic, asymptomatic, severe, nocturnal, non-nocturnal, probable symptomatic, relative, and unspecified) were included, except for episodes of relative hypoglycemia that were not severe. A summary of serious and other non-serious adverse events, regardless of causality, is located in the Reported Adverse Events module. (NCT01468181)
Timeframe: Baseline through 52 Weeks

Interventionpercentage of participants (Number)
LY2189265 + SU33.6
LY2189265 + BG3.3
LY2189265 + a-GI6.2
LY2189265 + TZD6.1
LY2189265 + Glinides9.9

[back to top]

Change From Baseline in Fasting Plasma Glucose (FPG) at Week 52

FPG is an indicator of efficacy. The Baseline FPG value is defined as the last non-missing value before the start of treatment. Change from Baseline was calculated as the FPG value at Week 52 minus the FPG value at Baseline. Participants who discontinued from study treatment before Week 52 had their last on-treatment, post-Baseline FPG observation carried forward for the analysis. (NCT01777282)
Timeframe: Baseline and Week 52

InterventionMilligrams per deciliter (mg/dL) (Mean)
Albiglutide Plus (+) Background OAD (Sulfonylurea)-16.4
Albiglutide Plus (+) Background OAD (Biguanide)-24.3
Albiglutide Plus (+) Background OAD (Glinide)-16.4
Albiglutide Plus (+) Background OAD (Thiazolidinedione)-32.1
Albiglutide Plus (+) Background OAD (α-Glucosidase Inhibitor)-33.2

[back to top]

Change From Baseline in Glycosylated Hemoglobin (HbA1c) at Week 52

HbA1c is a form of hemoglobin that is measured primarily to identify the average plasma glucose concentration over a 2- to 3-month period. The Baseline HbA1c value is defined as the last non-missing value before the start of treatment. Change from Baseline was calculated as the value at Week 52 minus the value at Baseline. Participants who discontinued from study treatment before Week 52 had their last on-treatment, post-Baseline HbA1c observation carried forward for the analysis. (NCT01777282)
Timeframe: Baseline and Week 52

InterventionPercentage of HbA1c in the blood (Mean)
Albiglutide Plus (+) Background OAD (Sulfonylurea)-1.04
Albiglutide Plus (+) Background OAD (Biguanide)-0.94
Albiglutide Plus (+) Background OAD (Glinide)-0.95
Albiglutide Plus (+) Background OAD (Thiazolidinedione)-1.42
Albiglutide Plus (+) Background OAD (α-Glucosidase Inhibitor)-1.39

[back to top]

Number of Participants With Any Hypoglycemic Event

Hypoglycemia events are defined with respect to low plasma glucose level, mostly accompanied by typical symptoms and/or assistance needed from third party with glucose administration. These events were reported by the investigators upon verification of the plasma glucose levels, symptoms and assistance recorded by the participants, and/or plasma glucose values obtained from laboratory evaluations. (NCT01777282)
Timeframe: From Baseline through Week 52

InterventionParticipants (Number)
Albiglutide Plus (+) Background OAD (Sulfonylurea)17
Albiglutide Plus (+) Background OAD (Biguanide)1
Albiglutide Plus (+) Background OAD (Glinide)4
Albiglutide Plus (+) Background OAD (Thiazolidinedione)2
Albiglutide Plus (+) Background OAD (α-Glucosidase Inhibitor)0

[back to top]

Time to Study Withdrawal Due to Hyperglycemia

Participants who experienced persistent hyperglycemia after uptitration were to be withdrawn from the study. Hyperglycemia is defined as a fasting plasma glucose >=280 mg/dL (>=15.5 mmol/L) from >=Week 2 to =230 mg/dL (>=12.8 mmol/L) from >=Week 12 to NCT01777282)
Timeframe: Week 52

InterventionWeeks (Mean)
Albiglutide Plus (+) Background OAD (Sulfonylurea)13.0
Albiglutide Plus (+) Background OAD (Glinide)16.5

[back to top]

Number of Participants With Any Adverse Event (AE) or Any Serious Adverse Event (SAE)

An AE is defined as any untoward medical occurrence in a clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a medicinal product. An SAE is defined as any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, is a congenital anomaly/birth defect, may jeopardize the participant or may require medical or surgical intervention to prevent one of the other outcomes listed in this definition, or is an event of possible drug-induced liver injury. Refer to the general AE/SAE module for a list of non-serious AEs and SAEs. Non-serious hypoglycemia events are not included. (NCT01777282)
Timeframe: From Baseline through Week 52

,,,,
InterventionParticipants (Number)
Any AEAny SAE
Albiglutide Plus (+) Background OAD (Biguanide)470
Albiglutide Plus (+) Background OAD (Glinide)581
Albiglutide Plus (+) Background OAD (Sulfonylurea)972
Albiglutide Plus (+) Background OAD (Thiazolidinedione)492
Albiglutide Plus (+) Background OAD (α-Glucosidase Inhibitor)433

[back to top]

Percentage of Participants Achieving Clinically Meaningful Levels of HbA1c (i.e., the Percentage of Participants Achieving Treatment Goal of <6.5% and <7.0% at Week 52)

HbA1c is a form of hemoglobin that is measured primarily to identify the average plasma glucose concentration over a 2- to 3 month period. The Baseline HbA1c value is defined as the last non-missing value before the start of treatment. Participants who discontinued from study treatment before Week 52 had their last on-treatment, post-Baseline HbA1c observation carried forward for the analysis. Clinically meaningful levels of response in HbA1c are defined as <6.5% and <7.0%. (NCT01777282)
Timeframe: Week 52

,,,,
InterventionPercentage of participants (Number)
HbA1c <6.5%HbA1c <7.0%
Albiglutide Plus (+) Background OAD (Biguanide)26.959.7
Albiglutide Plus (+) Background OAD (Glinide)24.652.3
Albiglutide Plus (+) Background OAD (Sulfonylurea)20.054.2
Albiglutide Plus (+) Background OAD (Thiazolidinedione)45.980.3
Albiglutide Plus (+) Background OAD (α-Glucosidase Inhibitor)26.267.2

[back to top]

Change From Baseline in Body Weight at Week 52

The Baseline body weight value is defined as the last non-missing value before the start of treatment. Change from Baseline was calculated as the body weight value at Week 52 minus the value at Baseline. Participants who discontinued from study treatment before Week 52 had their last on-treatment, post-Baseline weight observation carried forward for the analysis. (NCT01777282)
Timeframe: Baseline and Week 52

InterventionKilograms (kg) (Mean)
Albiglutide Plus (+) Background OAD (Sulfonylurea)0.25
Albiglutide Plus (+) Background OAD (Biguanide)-0.33
Albiglutide Plus (+) Background OAD (Glinide)-0.04
Albiglutide Plus (+) Background OAD (Thiazolidinedione)0.52
Albiglutide Plus (+) Background OAD (α-Glucosidase Inhibitor)-0.13

[back to top]

Number of AEs Leading to Permanent Discontinuation of Trial Product

An adverse event (AE) was any untoward medical occurrence in a participant who administered a product, and which did not necessarily had a causal relationship with this treatment. An AE was considered as treatment emergent if it had an onset or increase in severity on or after the time of first trial product administration and no later than 7 days after the time of last trial product administration. Number of treatment emergent AEs that led to permanent discontinuation of trial product are presented. (NCT02730377)
Timeframe: Weeks 0-105

InterventionEvents (Number)
Liraglutide 1.8 mg188
Oral Antidiabetic Drug98

[back to top]

Number of Documented Symptomatic Hypoglycaemic Episodes (ADA)

Documented symptomatic hypoglycaemic were defined as episodes with typical symptoms of hypoglycaemia accompanied by measure plasma glucose concentration <= 3.9 mmol/L. Number of documented symptomatic hypoglycaemic episodes that occured during the weeks 0-104 are presented. (NCT02730377)
Timeframe: Weeks 0-104

InterventionEpisodes (Number)
Liraglutide 1.8 mg98
Oral Antidiabetic Drug155

[back to top]

Number of Serious Adverse Events (SAEs)

A serious adverse event (SAE) was defined as any event that resulted in any of the following: death, life-threatening experience, in-patient hospitalisation or prolongation of existing hospitalisation, persistent or significant disability or incapacity, congenital anomaly or birth defect or suspicion of transmission of infectious agents via the trial product. An SAE was considered as treatment emergent if it had an onset or increase in severity on or after the time of first trial product administration and no later than 7 days after the time of last trial product administration. Number of treatment emergent serious adverse events are presented. (NCT02730377)
Timeframe: Weeks 0-105

InterventionEvents (Number)
Liraglutide 1.8 mg145
Oral Antidiabetic Drug140

[back to top]

Number of Severe Hypoglycaemic Episodes

Severe hypoglycaemic episodes were defined as episodes that required assistance of another person to actively administer carbohydrate, glucagon, or take other corrective actions. Number of severe hypoglycaemic episodes that occured during weeks 0-104 are presented. (NCT02730377)
Timeframe: Weeks 0-104

InterventionEpisodes (Number)
Liraglutide 1.8 mg32
Oral Antidiabetic Drug52

[back to top]

Number of Severe or BG Confirmed Symptomatic Hypoglycaemic Episodes

Severe or BG confirmed symptomatic hypoglycaemic episodes were defined as episodes that were severe according to the ADA classification (required assistance of another person to actively administer carbohydrate, glucagon, or take other corrective actions) or BG confirmed by a plasma glucose value <3.1 mmol/L with symptoms consistent with hypoglycaemia. Number of severe or BG confirmed symptomatic hypoglycaemic episodes that occured during weeks 0-104 are presented. (NCT02730377)
Timeframe: Weeks 0-104

InterventionEpisodes (Number)
Liraglutide 1.8 mg24
Oral Antidiabetic Drug44

[back to top]

Time to Inadequate Glycaemic Control

Inadequate glycaemic control was defined as glycosylated haemoglobin (HbA1c) of 7.0% (53 mmol/mol) or greater at two consecutive visits after the first 26 weeks of treatment and up to 104 weeks. 25%, median (50%) and 75% percentiles for the cumulative distribution function, are obtained from the Kaplan-Meier survival function. HbA1c was recorded at weeks 38, 52, 65, 78, 91 and 104. (NCT02730377)
Timeframe: Weeks 26-104

InterventionWeeks (Median)
Liraglutide 1.8 mg108.9
Oral Antidiabetic Drug64.9

[back to top]

Change in Haemoglobin

Change from baseline (week 0) in haemoglobin at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionGrams per deciliter (g/dL) (Mean)
Change at week 104Change at premature treatment discontinuation
Liraglutide 1.8 mg-0.4-0.3
Oral Antidiabetic Drug-0.0-0.3

[back to top]

Change in HbA1c

Change from baseline (week 0) in HbA1c at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionPercentage point of HbA1c (Mean)
Change at week 104Change at premature treatment discontinuation
Liraglutide 1.8 mg-1.4-0.6
Oral Antidiabetic Drug-1.1-0.2

[back to top]

Change in Lipids: HDL Cholesterol, LDL-cholesterol, Total Cholesterol, Triglycerides

Change from baseline (week 0) in high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, total cholesterol (TC) and triglycerides (TG) at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionMillimoles per liter (mmol/L) (Mean)
HDL: Change at week 104HDL: Change at premature treatment discontinuationLDL: Change at week 104LDL: Change at premature treatment discontinuationTC: Change at week 104TC: Change at premature treatment discontinuationTG: Change at week 104TG: Change at premature treatment discontinuation
Liraglutide 1.8 mg0.1-0.0-0.1-0.1-0.2-0.0-0.3-0.0
Oral Antidiabetic Drug0.10.00.0-0.10.1-0.1-0.1-0.0

[back to top]

Change in Potassium

Change from baseline (week 0) in potassium at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionMillimoles per liter (mmol/L) (Mean)
Change at week 104Change at premature treatment discontinuation
Liraglutide 1.8 mg-0.1-0.0
Oral Antidiabetic Drug-0.0-0.2

[back to top]

Change in Pulse

Change from baseline (week 0) in pulse at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionBeats per minute (beats/min) (Mean)
Change at week 104Change at premature treatment discontinuation
Liraglutide 1.8 mg1.00.7
Oral Antidiabetic Drug-0.60.9

[back to top]

Change in Biochemistry- Alanine Aminotransferase (ALAT), Amylase, Aspartate Aminotransferase (ASAT), Lipase

Change from baseline (week 0) in alanine aminotransferase (ALAT), amylase, aspartate aminotransferase (ASAT) and lipase at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionUnits per liter (U/L) (Mean)
ALAT: Week 104ALAT: Premature treatment discontinuationAmylase: Week 104Amylase: Premature treatment discontinuationASAT: Week 104ASAT: Premature treatment discontinuationLipase: Week 104Lipase: Premature treatment discontinuation
Liraglutide 1.8 mg-4.6-3.28.90.6-2.0-1.915.110.4
Oral Antidiabetic Drug-5.4-3.35.12.1-2.3-0.4-0.5-2.2

[back to top]

Time to Premature Treatment Discontinuation (for Any Reason Including Inadequate Glycaemic Control)

The time to premature treatment discontinuation (for any reason including inadequate glycaemic control) was analysed and presented using the generalised log rank test. 25%, median (50%) and 75% percentiles for the cumulative distribution function, are obtained from the Kaplan-Meier survival function. (NCT02730377)
Timeframe: Weeks 0-104

InterventionWeeks (Median)
Liraglutide 1.8 mg80.4
Oral Antidiabetic Drug52.3

[back to top]

Change in Biochemistry- Creatinine, Total Bilirubin

Change from baseline (week 0) in creatinine and total bilirubin (TB) at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionMicromoles per liter (umol/L) (Mean)
Creatinine: week 104Creatinine: premature treatment discontinuationTB: week 104TB: premature treatment discontinuation
Liraglutide 1.8 mg3.32.90.4-0.0
Oral Antidiabetic Drug1.02.60.7-0.6

[back to top]

Change in Biochemistry- Estimated Glomerular Filtration Rate (eGFR) Serum

The estimated GFR was derived from serum creatinine using the MDRD (Modification of diet in renal disease) formula. eGFR was measured as milliliter per min per specific surface area (mL/min/SSA). (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionmL/min/SSA (Mean)
Change at week 104Change at premature treatment discontinuation
Liraglutide 1.8 mg-5.1-3.0
Oral Antidiabetic Drug-1.6-1.7

[back to top]

Change in Blood Pressure (Systolic and Diastolic Blood Pressure)

Change from baseline (week 0) in systolic and diastolic blood pressure at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionMillimeters of mercury (mmHg) (Mean)
SBP: Change at week 104SBP: Change at premature treatment discontinuationDBP: Change at week 104DBP: Change at premature treatment discontinuation
Liraglutide 1.8 mg-2.4-2.8-1.3-1.0
Oral Antidiabetic Drug-1.1-2.9-0.60.2

[back to top]

Change in Body Mass Index (BMI)

Change from baseline (week 0) in BMI at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionKilograms per square meter (kg/m^2) (Mean)
Change at week 104Change at premature treatment discontinuation
Liraglutide 1.8 mg-1.3-1.1
Oral Antidiabetic Drug-1.2-0.8

[back to top]

Change in Body Weight

Change from baseline (week 0) in body weight at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionKilogram (Kg) (Mean)
Change at week 104Change at premature treatment discontinuation
Liraglutide 1.8 mg-3.8-2.9
Oral Antidiabetic Drug-3.5-2.2

[back to top]

Change in Fasting Plasma Glucose (FPG)

Change from baseline (week 0) in FPG at week 104 or at premature treatment discontinuation is presented. (NCT02730377)
Timeframe: Week 0, week 104/premature treatment discontinuation

,
InterventionMillimoles per liter (mmol/L) (Mean)
Change at week 104Change at premature treatment discontinuation
Liraglutide 1.8 mg-2.2-0.6
Oral Antidiabetic Drug-1.2-0.6

[back to top]