Page last updated: 2024-11-11

muromonab-cd3

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

Description

xestospongin C: was indexed to xestospongin A; from Xestospongia sponge; empties the ER calcium store but does not inhibit InsP3-induced Ca2+ release [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

xestospongin C : An organic heteropentacyclic compound that is isolated from the marine sponge Xestospongia exigua. [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]

cudraflavone B: antiproliferative from Cudrania tricuspidata [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

cudraflavone B : An extended flavonoid that consists of a pyranochromane skeleton that is 2H,6H-pyrano[3,2-g]chromen-6-one substituted by geminal methyl groups at position 2, a 2,4-dihydroxyphenyl group at position 8, a hydroxy group at position 5 and a prenyl group at position 7. Isolated from Morus alba and Morus species it exhibits anti-inflammatory activity. [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]

FloraRankFlora DefinitionFamilyFamily Definition
MorusgenusA plant genus of the family MORACEAE that is widely planted for shade.[MeSH]MoraceaeThe mulberry plant family of the order Urticales, subclass Hamamelidae, class Magnoliopsida. They have milky latex and small, petalless male or female flowers.[MeSH]
Morus albaspecies[no description available]MoraceaeThe mulberry plant family of the order Urticales, subclass Hamamelidae, class Magnoliopsida. They have milky latex and small, petalless male or female flowers.[MeSH]

Cross-References

ID SourceID
PubMed CID9846431
CHEMBL ID515501
CHEBI ID131784
SCHEMBL ID13820264
MeSH IDM0025653
PubMed CID5319925
CHEMBL ID221907
CHEBI ID67850
SCHEMBL ID23728289
MeSH IDM0025653

Synonyms (40)

Synonym
xestospongin c
xestospongin c, film
xec ,
CHEMBL515501
(-)-xestospongin c
araguspongin e
CHEBI:131784
araguspongine e
88903-69-9
(4ar,11r,12as,16as,23r,24as)-icosahydro-2h,5h,12ah,14h-1,23:11,13-diethano[1,11]dioxacycloicosino[10,9-b:20,19-b']dipyridine
(1r,4ar,11r,12as,13s,16as,23r,24as)-eicosahydro-5h,17h-1,23:11,13-diethano-2h,14h-[1,11]dioxacycloeicosino[2,3-b:12,13-b']dipyridine
SCHEMBL13820264
AKOS024456506
c28h50n2o2
NCGC00485590-01
DTXSID50893520
HB1241
(1s,8r,10s,15r,22r,29s)-9,30-dioxa-11,25-diazapentacyclo[20.6.2.28,11.010,15.025,29]dotriacontane
HY-103312
CS-0027594
MLS000697596
cudraflavone b
smr000470934
cid_5319925
2-(2,4-dihydroxyphenyl)-5-hydroxy-8,8-dimethyl-3-(3-methylbut-2-enyl)pyrano[3,2-g]chromen-4(8h)-one
bdbm50193718
2-(2,4-dihydroxy-phenyl)-5-hydroxy-8,8-dimethyl-3-(3-methyl-but-2-enyl)-8h-pyrano[3,2-g]chromen-4-one
chebi:67850 ,
CHEMBL221907 ,
8-(2,4-dihydroxyphenyl)-5-hydroxy-2,2-dimethyl-7-(3-methylbut-2-enyl)pyrano[3,2-g]chromen-6-one
HMS2268D05
8-(2,4-dihydroxyphenyl)-5-hydroxy-2,2-dimethyl-7-(3-methylbut-2-en-1-yl)-2h,6h-pyrano[3,2-g]chromen-6-one
19275-49-1
Q27136327
2-(2,4-dihydroxyphenyl)-3-(3-methyl-2-butenyl)-5-hydroxy-8,8-dimethyl-4h,8h-benzo[1,2-b:5,4-b/']dipy
SCHEMBL23728289
2-(2,4-dihydroxyphenyl)-3-(3-methyl-2-butenyl)-5-hydroxy-8,8-dimethyl-4h,8h-benzo[1,2-b
CS-0253680
HY-N10009
AKOS040761545

Research Excerpts

Bioavailability

ExcerptReferenceRelevance
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (6)

RoleDescription
marine metaboliteAny metabolite produced during a metabolic reaction in marine macro- and microorganisms.
IP3 receptor antagonistAn antagonist that binds to and deactivates IP3 receptors.
EC 2.7.10.1 (receptor protein-tyrosine kinase) inhibitorAn EC 2.7.10.* (protein-tyrosine kinase) inhibitor that interferes with the action of receptor protein-tyrosine kinase (EC 2.7.10.1).
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
anti-inflammatory agentAny compound that has anti-inflammatory effects.
plant metaboliteAny eukaryotic metabolite produced during a metabolic reaction in plants, the kingdom that include flowering plants, conifers and other gymnosperms.
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (9)

ClassDescription
organic heteropentacyclic compound
macrocycleA cyclic compound containing nine or more atoms as part of the cyclic system.
oxacycleAny organic heterocyclic compound containing at least one ring oxygen atom.
organonitrogen heterocyclic compoundAny organonitrogen compound containing a cyclic component with nitrogen and at least one other element as ring member atoms.
tertiary amino compoundA compound formally derived from ammonia by replacing three hydrogen atoms by organyl groups.
alkaloidAny of the naturally occurring, basic nitrogen compounds (mostly heterocyclic) occurring mostly in the plant kingdom, but also found in bacteria, fungi, and animals. By extension, certain neutral compounds biogenetically related to basic alkaloids are also classed as alkaloids. Amino acids, peptides, proteins, nucleotides, nucleic acids, amino sugars and antibiotics are not normally regarded as alkaloids. Compounds in which the nitrogen is exocyclic (dopamine, mescaline, serotonin, etc.) are usually classed as amines rather than alkaloids.
extended flavonoidAny flavonoid with one or more rings fused on to the phenyl substituted benzopyran framework.
pyranochromaneAn organic heterotricyclic compound that consists of a pyran ring fused on to a chromane skeleton.
trihydroxyflavoneAny hydroxyflavone carrying three hydroxy groups at unspecified positions.
[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 (16)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Chain A, JmjC domain-containing histone demethylation protein 3AHomo sapiens (human)Potency56.23410.631035.7641100.0000AID504339
glp-1 receptor, partialHomo sapiens (human)Potency1.25890.01846.806014.1254AID624417
TDP1 proteinHomo sapiens (human)Potency23.10930.000811.382244.6684AID686978; AID686979
Smad3Homo sapiens (human)Potency35.48130.00527.809829.0929AID588855
vitamin D3 receptor isoform VDRAHomo sapiens (human)Potency50.11870.354828.065989.1251AID504847
DNA polymerase iota isoform a (long)Homo sapiens (human)Potency100.00000.050127.073689.1251AID588590
gemininHomo sapiens (human)Potency29.09290.004611.374133.4983AID624296
VprHuman immunodeficiency virus 1Potency63.09571.584919.626463.0957AID651644
Alpha-synucleinHomo sapiens (human)Potency35.48130.56239.398525.1189AID652106
TAR DNA-binding protein 43Homo sapiens (human)Potency31.62281.778316.208135.4813AID652104
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Sterol O-acyltransferase 2Homo sapiens (human)IC50 (µMol)41.60000.11003.20369.2000AID276035
Tyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)IC50 (µMol)20.00000.00053.49849.7600AID1773062
Prostaglandin G/H synthase 2Homo sapiens (human)IC50 (µMol)2.50000.00010.995010.0000AID595370
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
streptokinase A precursorStreptococcus pyogenes M1 GASEC50 (µMol)75.79000.06008.9128130.5170AID1902; AID1914
Estrogen receptorRattus norvegicus (Norway rat)EC50 (µMol)150.00000.006022.3670130.5170AID1914
Estrogen receptor betaRattus norvegicus (Norway rat)EC50 (µMol)150.00000.006022.3670130.5170AID1914
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (183)

Processvia Protein(s)Taxonomy
cholesterol metabolic processSterol O-acyltransferase 2Homo sapiens (human)
macrophage derived foam cell differentiationSterol O-acyltransferase 2Homo sapiens (human)
cholesterol storageSterol O-acyltransferase 2Homo sapiens (human)
intestinal cholesterol absorptionSterol O-acyltransferase 2Homo sapiens (human)
cholesterol effluxSterol O-acyltransferase 2Homo sapiens (human)
very-low-density lipoprotein particle assemblySterol O-acyltransferase 2Homo sapiens (human)
low-density lipoprotein particle clearanceSterol O-acyltransferase 2Homo sapiens (human)
cholesterol homeostasisSterol O-acyltransferase 2Homo sapiens (human)
positive regulation of JUN kinase activityTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
protein dephosphorylationTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
insulin receptor signaling pathwayTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
regulation of signal transductionTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
negative regulation of signal transductionTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
actin cytoskeleton organizationTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
regulation of endocytosisTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
negative regulation of vascular endothelial growth factor receptor signaling pathwayTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
endoplasmic reticulum unfolded protein responseTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
regulation of intracellular protein transportTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
cellular response to unfolded proteinTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
peptidyl-tyrosine dephosphorylationTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
platelet-derived growth factor receptor-beta signaling pathwayTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
IRE1-mediated unfolded protein responseTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
insulin receptor recyclingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
negative regulation of MAP kinase activityTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
negative regulation of insulin receptor signaling pathwayTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
regulation of type I interferon-mediated signaling pathwayTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
growth hormone receptor signaling pathway via JAK-STATTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
positive regulation of protein tyrosine kinase activityTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
negative regulation of ERK1 and ERK2 cascadeTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
regulation of hepatocyte growth factor receptor signaling pathwayTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
negative regulation of endoplasmic reticulum stress-induced intrinsic apoptotic signaling pathwayTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
positive regulation of IRE1-mediated unfolded protein responseTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
negative regulation of PERK-mediated unfolded protein responseTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
peptidyl-tyrosine dephosphorylation involved in inactivation of protein kinase activityTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
positive regulation of receptor catabolic processTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
prostaglandin biosynthetic processProstaglandin G/H synthase 2Homo sapiens (human)
angiogenesisProstaglandin G/H synthase 2Homo sapiens (human)
response to oxidative stressProstaglandin G/H synthase 2Homo sapiens (human)
embryo implantationProstaglandin G/H synthase 2Homo sapiens (human)
learningProstaglandin G/H synthase 2Homo sapiens (human)
memoryProstaglandin G/H synthase 2Homo sapiens (human)
regulation of blood pressureProstaglandin G/H synthase 2Homo sapiens (human)
negative regulation of cell population proliferationProstaglandin G/H synthase 2Homo sapiens (human)
response to xenobiotic stimulusProstaglandin G/H synthase 2Homo sapiens (human)
response to nematodeProstaglandin G/H synthase 2Homo sapiens (human)
response to fructoseProstaglandin G/H synthase 2Homo sapiens (human)
response to manganese ionProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of vascular endothelial growth factor productionProstaglandin G/H synthase 2Homo sapiens (human)
cyclooxygenase pathwayProstaglandin G/H synthase 2Homo sapiens (human)
bone mineralizationProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of prostaglandin biosynthetic processProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of fever generationProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of synaptic plasticityProstaglandin G/H synthase 2Homo sapiens (human)
negative regulation of synaptic transmission, dopaminergicProstaglandin G/H synthase 2Homo sapiens (human)
prostaglandin secretionProstaglandin G/H synthase 2Homo sapiens (human)
response to estradiolProstaglandin G/H synthase 2Homo sapiens (human)
response to lipopolysaccharideProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of peptidyl-serine phosphorylationProstaglandin G/H synthase 2Homo sapiens (human)
response to vitamin DProstaglandin G/H synthase 2Homo sapiens (human)
cellular response to heatProstaglandin G/H synthase 2Homo sapiens (human)
response to tumor necrosis factorProstaglandin G/H synthase 2Homo sapiens (human)
maintenance of blood-brain barrierProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of protein import into nucleusProstaglandin G/H synthase 2Homo sapiens (human)
hair cycleProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of apoptotic processProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of nitric oxide biosynthetic processProstaglandin G/H synthase 2Homo sapiens (human)
negative regulation of cell cycleProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of vasoconstrictionProstaglandin G/H synthase 2Homo sapiens (human)
negative regulation of smooth muscle contractionProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of smooth muscle contractionProstaglandin G/H synthase 2Homo sapiens (human)
decidualizationProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of smooth muscle cell proliferationProstaglandin G/H synthase 2Homo sapiens (human)
regulation of inflammatory responseProstaglandin G/H synthase 2Homo sapiens (human)
brown fat cell differentiationProstaglandin G/H synthase 2Homo sapiens (human)
response to glucocorticoidProstaglandin G/H synthase 2Homo sapiens (human)
negative regulation of calcium ion transportProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of synaptic transmission, glutamatergicProstaglandin G/H synthase 2Homo sapiens (human)
response to fatty acidProstaglandin G/H synthase 2Homo sapiens (human)
cellular response to mechanical stimulusProstaglandin G/H synthase 2Homo sapiens (human)
cellular response to lead ionProstaglandin G/H synthase 2Homo sapiens (human)
cellular response to ATPProstaglandin G/H synthase 2Homo sapiens (human)
cellular response to hypoxiaProstaglandin G/H synthase 2Homo sapiens (human)
cellular response to non-ionic osmotic stressProstaglandin G/H synthase 2Homo sapiens (human)
cellular response to fluid shear stressProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of transforming growth factor beta productionProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of cell migration involved in sprouting angiogenesisProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of fibroblast growth factor productionProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of brown fat cell differentiationProstaglandin G/H synthase 2Homo sapiens (human)
positive regulation of platelet-derived growth factor productionProstaglandin G/H synthase 2Homo sapiens (human)
cellular oxidant detoxificationProstaglandin G/H synthase 2Homo sapiens (human)
regulation of neuroinflammatory responseProstaglandin G/H synthase 2Homo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathway in response to osmotic stressProstaglandin G/H synthase 2Homo sapiens (human)
cellular response to homocysteineProstaglandin G/H synthase 2Homo sapiens (human)
response to angiotensinProstaglandin G/H synthase 2Homo sapiens (human)
calcium ion homeostasisAlpha-synucleinHomo sapiens (human)
negative regulation of transcription by RNA polymerase IIAlpha-synucleinHomo sapiens (human)
microglial cell activationAlpha-synucleinHomo sapiens (human)
positive regulation of receptor recyclingAlpha-synucleinHomo sapiens (human)
positive regulation of neurotransmitter secretionAlpha-synucleinHomo sapiens (human)
negative regulation of protein kinase activityAlpha-synucleinHomo sapiens (human)
fatty acid metabolic processAlpha-synucleinHomo sapiens (human)
neutral lipid metabolic processAlpha-synucleinHomo sapiens (human)
phospholipid metabolic processAlpha-synucleinHomo sapiens (human)
activation of cysteine-type endopeptidase activity involved in apoptotic processAlpha-synucleinHomo sapiens (human)
mitochondrial membrane organizationAlpha-synucleinHomo sapiens (human)
adult locomotory behaviorAlpha-synucleinHomo sapiens (human)
response to xenobiotic stimulusAlpha-synucleinHomo sapiens (human)
response to iron(II) ionAlpha-synucleinHomo sapiens (human)
regulation of phospholipase activityAlpha-synucleinHomo sapiens (human)
negative regulation of platelet-derived growth factor receptor signaling pathwayAlpha-synucleinHomo sapiens (human)
regulation of glutamate secretionAlpha-synucleinHomo sapiens (human)
regulation of dopamine secretionAlpha-synucleinHomo sapiens (human)
synaptic vesicle exocytosisAlpha-synucleinHomo sapiens (human)
synaptic vesicle primingAlpha-synucleinHomo sapiens (human)
regulation of transmembrane transporter activityAlpha-synucleinHomo sapiens (human)
negative regulation of microtubule polymerizationAlpha-synucleinHomo sapiens (human)
receptor internalizationAlpha-synucleinHomo sapiens (human)
protein destabilizationAlpha-synucleinHomo sapiens (human)
response to magnesium ionAlpha-synucleinHomo sapiens (human)
negative regulation of transporter activityAlpha-synucleinHomo sapiens (human)
response to lipopolysaccharideAlpha-synucleinHomo sapiens (human)
negative regulation of monooxygenase activityAlpha-synucleinHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylationAlpha-synucleinHomo sapiens (human)
response to type II interferonAlpha-synucleinHomo sapiens (human)
cellular response to oxidative stressAlpha-synucleinHomo sapiens (human)
SNARE complex assemblyAlpha-synucleinHomo sapiens (human)
positive regulation of SNARE complex assemblyAlpha-synucleinHomo sapiens (human)
regulation of locomotionAlpha-synucleinHomo sapiens (human)
dopamine biosynthetic processAlpha-synucleinHomo sapiens (human)
mitochondrial ATP synthesis coupled electron transportAlpha-synucleinHomo sapiens (human)
regulation of macrophage activationAlpha-synucleinHomo sapiens (human)
positive regulation of apoptotic processAlpha-synucleinHomo sapiens (human)
negative regulation of apoptotic processAlpha-synucleinHomo sapiens (human)
negative regulation of cysteine-type endopeptidase activity involved in apoptotic processAlpha-synucleinHomo sapiens (human)
negative regulation of neuron apoptotic processAlpha-synucleinHomo sapiens (human)
positive regulation of endocytosisAlpha-synucleinHomo sapiens (human)
negative regulation of exocytosisAlpha-synucleinHomo sapiens (human)
positive regulation of exocytosisAlpha-synucleinHomo sapiens (human)
regulation of long-term neuronal synaptic plasticityAlpha-synucleinHomo sapiens (human)
synaptic vesicle endocytosisAlpha-synucleinHomo sapiens (human)
synaptic vesicle transportAlpha-synucleinHomo sapiens (human)
positive regulation of inflammatory responseAlpha-synucleinHomo sapiens (human)
regulation of acyl-CoA biosynthetic processAlpha-synucleinHomo sapiens (human)
protein tetramerizationAlpha-synucleinHomo sapiens (human)
positive regulation of release of sequestered calcium ion into cytosolAlpha-synucleinHomo sapiens (human)
neuron apoptotic processAlpha-synucleinHomo sapiens (human)
dopamine uptake involved in synaptic transmissionAlpha-synucleinHomo sapiens (human)
negative regulation of dopamine uptake involved in synaptic transmissionAlpha-synucleinHomo sapiens (human)
negative regulation of serotonin uptakeAlpha-synucleinHomo sapiens (human)
regulation of norepinephrine uptakeAlpha-synucleinHomo sapiens (human)
negative regulation of norepinephrine uptakeAlpha-synucleinHomo sapiens (human)
excitatory postsynaptic potentialAlpha-synucleinHomo sapiens (human)
long-term synaptic potentiationAlpha-synucleinHomo sapiens (human)
positive regulation of inositol phosphate biosynthetic processAlpha-synucleinHomo sapiens (human)
negative regulation of thrombin-activated receptor signaling pathwayAlpha-synucleinHomo sapiens (human)
response to interleukin-1Alpha-synucleinHomo sapiens (human)
cellular response to copper ionAlpha-synucleinHomo sapiens (human)
cellular response to epinephrine stimulusAlpha-synucleinHomo sapiens (human)
positive regulation of protein serine/threonine kinase activityAlpha-synucleinHomo sapiens (human)
supramolecular fiber organizationAlpha-synucleinHomo sapiens (human)
negative regulation of mitochondrial electron transport, NADH to ubiquinoneAlpha-synucleinHomo sapiens (human)
positive regulation of glutathione peroxidase activityAlpha-synucleinHomo sapiens (human)
positive regulation of hydrogen peroxide catabolic processAlpha-synucleinHomo sapiens (human)
regulation of synaptic vesicle recyclingAlpha-synucleinHomo sapiens (human)
regulation of reactive oxygen species biosynthetic processAlpha-synucleinHomo sapiens (human)
positive regulation of protein localization to cell peripheryAlpha-synucleinHomo sapiens (human)
negative regulation of chaperone-mediated autophagyAlpha-synucleinHomo sapiens (human)
regulation of presynapse assemblyAlpha-synucleinHomo sapiens (human)
amyloid fibril formationAlpha-synucleinHomo sapiens (human)
synapse organizationAlpha-synucleinHomo sapiens (human)
chemical synaptic transmissionAlpha-synucleinHomo sapiens (human)
negative regulation of protein phosphorylationTAR DNA-binding protein 43Homo sapiens (human)
mRNA processingTAR DNA-binding protein 43Homo sapiens (human)
RNA splicingTAR DNA-binding protein 43Homo sapiens (human)
negative regulation of gene expressionTAR DNA-binding protein 43Homo sapiens (human)
regulation of protein stabilityTAR DNA-binding protein 43Homo sapiens (human)
positive regulation of insulin secretionTAR DNA-binding protein 43Homo sapiens (human)
response to endoplasmic reticulum stressTAR DNA-binding protein 43Homo sapiens (human)
positive regulation of protein import into nucleusTAR DNA-binding protein 43Homo sapiens (human)
regulation of circadian rhythmTAR DNA-binding protein 43Homo sapiens (human)
regulation of apoptotic processTAR DNA-binding protein 43Homo sapiens (human)
negative regulation by host of viral transcriptionTAR DNA-binding protein 43Homo sapiens (human)
rhythmic processTAR DNA-binding protein 43Homo sapiens (human)
regulation of cell cycleTAR DNA-binding protein 43Homo sapiens (human)
3'-UTR-mediated mRNA destabilizationTAR DNA-binding protein 43Homo sapiens (human)
3'-UTR-mediated mRNA stabilizationTAR DNA-binding protein 43Homo sapiens (human)
nuclear inner membrane organizationTAR DNA-binding protein 43Homo sapiens (human)
amyloid fibril formationTAR DNA-binding protein 43Homo sapiens (human)
regulation of gene expressionTAR DNA-binding protein 43Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (53)

Processvia Protein(s)Taxonomy
fatty-acyl-CoA bindingSterol O-acyltransferase 2Homo sapiens (human)
sterol O-acyltransferase activitySterol O-acyltransferase 2Homo sapiens (human)
protein bindingSterol O-acyltransferase 2Homo sapiens (human)
cholesterol bindingSterol O-acyltransferase 2Homo sapiens (human)
acyltransferase activitySterol O-acyltransferase 2Homo sapiens (human)
cholesterol O-acyltransferase activitySterol O-acyltransferase 2Homo sapiens (human)
RNA bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
protein tyrosine phosphatase activityTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
insulin receptor bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
protein bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
zinc ion bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
enzyme bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
protein kinase bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
receptor tyrosine kinase bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
cadherin bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
ephrin receptor bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
protein phosphatase 2A bindingTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
non-membrane spanning protein tyrosine phosphatase activityTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
peroxidase activityProstaglandin G/H synthase 2Homo sapiens (human)
prostaglandin-endoperoxide synthase activityProstaglandin G/H synthase 2Homo sapiens (human)
protein bindingProstaglandin G/H synthase 2Homo sapiens (human)
enzyme bindingProstaglandin G/H synthase 2Homo sapiens (human)
heme bindingProstaglandin G/H synthase 2Homo sapiens (human)
protein homodimerization activityProstaglandin G/H synthase 2Homo sapiens (human)
metal ion bindingProstaglandin G/H synthase 2Homo sapiens (human)
oxidoreductase activity, acting on single donors with incorporation of molecular oxygen, incorporation of two atoms of oxygenProstaglandin G/H synthase 2Homo sapiens (human)
fatty acid bindingAlpha-synucleinHomo sapiens (human)
phospholipase D inhibitor activityAlpha-synucleinHomo sapiens (human)
SNARE bindingAlpha-synucleinHomo sapiens (human)
magnesium ion bindingAlpha-synucleinHomo sapiens (human)
transcription cis-regulatory region bindingAlpha-synucleinHomo sapiens (human)
actin bindingAlpha-synucleinHomo sapiens (human)
protein kinase inhibitor activityAlpha-synucleinHomo sapiens (human)
copper ion bindingAlpha-synucleinHomo sapiens (human)
calcium ion bindingAlpha-synucleinHomo sapiens (human)
protein bindingAlpha-synucleinHomo sapiens (human)
phospholipid bindingAlpha-synucleinHomo sapiens (human)
ferrous iron bindingAlpha-synucleinHomo sapiens (human)
zinc ion bindingAlpha-synucleinHomo sapiens (human)
lipid bindingAlpha-synucleinHomo sapiens (human)
oxidoreductase activityAlpha-synucleinHomo sapiens (human)
kinesin bindingAlpha-synucleinHomo sapiens (human)
Hsp70 protein bindingAlpha-synucleinHomo sapiens (human)
histone bindingAlpha-synucleinHomo sapiens (human)
identical protein bindingAlpha-synucleinHomo sapiens (human)
alpha-tubulin bindingAlpha-synucleinHomo sapiens (human)
cysteine-type endopeptidase inhibitor activity involved in apoptotic processAlpha-synucleinHomo sapiens (human)
tau protein bindingAlpha-synucleinHomo sapiens (human)
phosphoprotein bindingAlpha-synucleinHomo sapiens (human)
molecular adaptor activityAlpha-synucleinHomo sapiens (human)
dynein complex bindingAlpha-synucleinHomo sapiens (human)
cuprous ion bindingAlpha-synucleinHomo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingTAR DNA-binding protein 43Homo sapiens (human)
DNA bindingTAR DNA-binding protein 43Homo sapiens (human)
double-stranded DNA bindingTAR DNA-binding protein 43Homo sapiens (human)
RNA bindingTAR DNA-binding protein 43Homo sapiens (human)
mRNA 3'-UTR bindingTAR DNA-binding protein 43Homo sapiens (human)
protein bindingTAR DNA-binding protein 43Homo sapiens (human)
lipid bindingTAR DNA-binding protein 43Homo sapiens (human)
identical protein bindingTAR DNA-binding protein 43Homo sapiens (human)
pre-mRNA intronic bindingTAR DNA-binding protein 43Homo sapiens (human)
molecular condensate scaffold activityTAR DNA-binding protein 43Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (43)

Processvia Protein(s)Taxonomy
endoplasmic reticulumSterol O-acyltransferase 2Homo sapiens (human)
endoplasmic reticulum membraneSterol O-acyltransferase 2Homo sapiens (human)
brush borderSterol O-acyltransferase 2Homo sapiens (human)
endoplasmic reticulum membraneSterol O-acyltransferase 2Homo sapiens (human)
plasma membraneTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
cytoplasmTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
mitochondrial matrixTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
early endosomeTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
endoplasmic reticulumTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
cytosolTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
mitochondrial cristaTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
endosome lumenTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
sorting endosomeTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
cytoplasmic side of endoplasmic reticulum membraneTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
protein-containing complexTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
endoplasmic reticulumTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
cytoplasmTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
early endosomeTyrosine-protein phosphatase non-receptor type 1Homo sapiens (human)
nuclear inner membraneProstaglandin G/H synthase 2Homo sapiens (human)
nuclear outer membraneProstaglandin G/H synthase 2Homo sapiens (human)
cytoplasmProstaglandin G/H synthase 2Homo sapiens (human)
endoplasmic reticulumProstaglandin G/H synthase 2Homo sapiens (human)
endoplasmic reticulum lumenProstaglandin G/H synthase 2Homo sapiens (human)
endoplasmic reticulum membraneProstaglandin G/H synthase 2Homo sapiens (human)
caveolaProstaglandin G/H synthase 2Homo sapiens (human)
neuron projectionProstaglandin G/H synthase 2Homo sapiens (human)
protein-containing complexProstaglandin G/H synthase 2Homo sapiens (human)
neuron projectionProstaglandin G/H synthase 2Homo sapiens (human)
cytoplasmProstaglandin G/H synthase 2Homo sapiens (human)
platelet alpha granule membraneAlpha-synucleinHomo sapiens (human)
extracellular regionAlpha-synucleinHomo sapiens (human)
extracellular spaceAlpha-synucleinHomo sapiens (human)
nucleusAlpha-synucleinHomo sapiens (human)
cytoplasmAlpha-synucleinHomo sapiens (human)
mitochondrionAlpha-synucleinHomo sapiens (human)
lysosomeAlpha-synucleinHomo sapiens (human)
cytosolAlpha-synucleinHomo sapiens (human)
plasma membraneAlpha-synucleinHomo sapiens (human)
cell cortexAlpha-synucleinHomo sapiens (human)
actin cytoskeletonAlpha-synucleinHomo sapiens (human)
membraneAlpha-synucleinHomo sapiens (human)
inclusion bodyAlpha-synucleinHomo sapiens (human)
axonAlpha-synucleinHomo sapiens (human)
growth coneAlpha-synucleinHomo sapiens (human)
synaptic vesicle membraneAlpha-synucleinHomo sapiens (human)
perinuclear region of cytoplasmAlpha-synucleinHomo sapiens (human)
postsynapseAlpha-synucleinHomo sapiens (human)
supramolecular fiberAlpha-synucleinHomo sapiens (human)
protein-containing complexAlpha-synucleinHomo sapiens (human)
cytoplasmAlpha-synucleinHomo sapiens (human)
axon terminusAlpha-synucleinHomo sapiens (human)
neuronal cell bodyAlpha-synucleinHomo sapiens (human)
intracellular non-membrane-bounded organelleTAR DNA-binding protein 43Homo sapiens (human)
nucleusTAR DNA-binding protein 43Homo sapiens (human)
nucleoplasmTAR DNA-binding protein 43Homo sapiens (human)
perichromatin fibrilsTAR DNA-binding protein 43Homo sapiens (human)
mitochondrionTAR DNA-binding protein 43Homo sapiens (human)
cytoplasmic stress granuleTAR DNA-binding protein 43Homo sapiens (human)
nuclear speckTAR DNA-binding protein 43Homo sapiens (human)
interchromatin granuleTAR DNA-binding protein 43Homo sapiens (human)
nucleoplasmTAR DNA-binding protein 43Homo sapiens (human)
chromatinTAR DNA-binding protein 43Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (63)

Assay IDTitleYearJournalArticle
AID1347160Primary screen NINDS Rhodamine qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID1346986P-glycoprotein substrates identified in KB-3-1 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1346987P-glycoprotein substrates identified in KB-8-5-11 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1347159Primary screen GU Rhodamine qHTS for Zika virus inhibitors: Unlinked NS2B-NS3 protease assay2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID332284Antimicrobial activity against Candida albicans UCD FRI after 16 to 18 hrs by microbroth dilution assay2002Journal of natural products, Mar, Volume: 65, Issue:3
(+)-7S-Hydroxyxestospongin A from the marine sponge Xestospongia sp. and absolute configuration of (+)-xestospongin D.
AID332291Inhibition of IP3-sensitive Ca2+ channel activity in endoplasmic reticulum of rabbit cerebellum assessed as Ca2+ release2002Journal of natural products, Mar, Volume: 65, Issue:3
(+)-7S-Hydroxyxestospongin A from the marine sponge Xestospongia sp. and absolute configuration of (+)-xestospongin D.
AID332285Antimicrobial activity against Candida glabrata after 16 to 18 hrs by microbroth dilution assay2002Journal of natural products, Mar, Volume: 65, Issue:3
(+)-7S-Hydroxyxestospongin A from the marine sponge Xestospongia sp. and absolute configuration of (+)-xestospongin D.
AID332286Antimicrobial activity against Candida krusei after 16 to 18 hrs by microbroth dilution assay2002Journal of natural products, Mar, Volume: 65, Issue:3
(+)-7S-Hydroxyxestospongin A from the marine sponge Xestospongia sp. and absolute configuration of (+)-xestospongin D.
AID332283Antimicrobial activity against fluconazole-resistant Candida albicans ATCC 14503 after 16 to 18 hrs by microbroth dilution assay2002Journal of natural products, Mar, Volume: 65, Issue:3
(+)-7S-Hydroxyxestospongin A from the marine sponge Xestospongia sp. and absolute configuration of (+)-xestospongin D.
AID767579Induction of ROS production in mouse J774A1 cells at 0.25 uM after 30 mins by flow cytometry relative to control2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID595393Inhibition of ram seminal vesicle COX-1 assessed as PGE2 production from arachidonic acid after 20 mins enzyme immunoassay2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID595374Antiinflammatory activity against human THP1 cells assessed as inhibition of LPS-induced ZFP36 gene expression at 10 uM pretreated for 1 hr before LPS challenge measured after 2 hrs by RT-qPCR relative to control2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID767584Antioxidant activity assessed as hydrogen peroxide radical scavenging activity by chemiluminescence method2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID276041Inhibition of LPS-induced iNOS protein expression in mouse RAW 264.7 cells at 10 uM2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID595370Inhibition of human recombinant COX-2 assessed as PGE2 production from arachidonic acid after 20 mins enzyme immunoassay2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID1607865Antiinflammatory activity in mouse RAW264.7 cells assessed as inhibition of LPS-induced nitric oxide production2019European journal of medicinal chemistry, Oct-01, Volume: 179Human disorders associated with inflammation and the evolving role of natural products to overcome.
AID276033Antioxidant activity against human plasma LDL oxidation by TBARS assay2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID276039Effect on cell viability in mouse RAW 264.7 cells at 80 uM by MTT method2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID767573Induction of ROS production in LPS-stimulated mouse J774A1 cells at 0.25 to 1.25 uM after 24 hrs by flow cytometry2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID1155195Cytotoxicity against human THP1 cells after 24 hrs by WST1 assay2014Journal of natural products, Jun-27, Volume: 77, Issue:6
Evaluation of anti-inflammatory activity of prenylated substances isolated from Morus alba and Morus nigra.
AID276038Inhibition of LPS-induced iNOS protein expression in mouse RAW 264.7 cells at 5 uM2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID595369Antiinflammatory activity against human THP1 cells of assessed as inhibition of LPS-induced COX-2 gene expression at 10 uM pretreated for 1 hr before LPS challenge measured after 4 hrs by RT-qPCR2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID1887299Antibacterial activity against Bacillus subtilis ATCC 11774 assessed as inhibition of bacterial growth incubated for 24 hrs by optical density based analysis2022Journal of natural products, 09-23, Volume: 85, Issue:9
Total Syntheses and Antibacterial Evaluations of Neocyclomorusin and Related Flavones.
AID1887297Antibacterial activity against Staphylococcus aureus ATCC 25923 assessed as inhibition of bacterial growth incubated for 24 hrs by optical density based analysis2022Journal of natural products, 09-23, Volume: 85, Issue:9
Total Syntheses and Antibacterial Evaluations of Neocyclomorusin and Related Flavones.
AID595378Antiinflammatory activity against mouse RAW264.7 cells assessed as iNOS gene expression2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID276043Inhibition of LPS-induced iNOS protein expression in mouse RAW 264.7 cells at 50 uM2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID276034Inhibition of human ACAT1 expressed in Hi5 cells2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID595373Antiinflammatory activity against human THP1 cells assessed as inhibition of LPS-induced ZFP36 gene expression at 10 uM pretreated for 1 hr before LPS challenge measured after 4 hrs by RT-qPCR relative to control2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID1773061Inhibition of recombinant PTP1B catalytic domain (unknown origin) assessed as reduction in pNP formation using pNPP as substrate at 20 ug/ml by absorbance based analysis relative to control
AID276037Inhibition of LPS-induced NO production in mouse RAW 264.7 cells after 18 hrs2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID595367Antiinflammatory activity against human THP1 cells of assessed as inhibition of LPS-induced TNFalpha gene expression at 10 uM pretreated for 1 hr before LPS challenge measured after 2 to 10 hrs by RT-qPCR2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID595368Antiinflammatory activity against human THP1 cells of assessed as inhibition of LPS-induced TNFalpha protein synthesis at 10 uM pretreated for 1 hr before LPS challenge measured after 2 to 10 hrs relative to control2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID1887298Antibacterial activity against Staphylococcus epidermidis ATCC 51625 assessed as inhibition of bacterial growth incubated for 24 hrs by optical density based analysis2022Journal of natural products, 09-23, Volume: 85, Issue:9
Total Syntheses and Antibacterial Evaluations of Neocyclomorusin and Related Flavones.
AID1155197Antiinflammatory activity in human THP1 cells assessed as inhibition of IL-1beta secretion at 1 uM treated 1 hr before LPS challenge measured after 24 hrs by ELISA2014Journal of natural products, Jun-27, Volume: 77, Issue:6
Evaluation of anti-inflammatory activity of prenylated substances isolated from Morus alba and Morus nigra.
AID767582Antioxidant activity assessed as hypochlorous acid scavenging activity by fluorescence assay2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID595372Selectivity ratio of IC50 for human recombinant COX2 to IC50 for ram seminal vesicle COX12011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID1155194Antiinflammatory activity in human THP1 cells assessed as inhibition of TNFalpha secretion at 1 uM treated 1 hr before LPS challenge measured after 24 hrs by ELISA2014Journal of natural products, Jun-27, Volume: 77, Issue:6
Evaluation of anti-inflammatory activity of prenylated substances isolated from Morus alba and Morus nigra.
AID1155199Inhibition of NFkappaB p65 nuclear translocation in LPS-induced human THP1 cells at 1 uM treated 1 hr before LPS challenge measured after 3 hrs using propidium iodide staining by fluorescence microscopic analysis2014Journal of natural products, Jun-27, Volume: 77, Issue:6
Evaluation of anti-inflammatory activity of prenylated substances isolated from Morus alba and Morus nigra.
AID767586Antioxidant activity assessed as NADH/phenazine methosulfate generated superoxide anion scavenging after 2 mins by NBT reduction assay2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID595379Antiinflammatory activity against mouse RAW264.7 cells assessed as nitric oxide production2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID767587Reduction of LPS-induced COX2 expression in mouse J774A1 cells at 1.25 uM pretreated 1 hr before LPS challenge measured after 24 hrs2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID767572Inhibition of NFkappaB in mouse J774A1 cells assessed as LPS-stimulated IkappaB-alpha degradation at 0.25 to 1.25 uM pretreated 30 mins before LPS challenge measured after 30 mins2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID595375Antiinflammatory activity against human THP1 cells assessed as inhibition of LPS-induced NFKB p65 subunit translocation to nucleus at 10 uM pretreated for 1 hr before LPS challenge relative to control2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID1773062Inhibition of recombinant PTP1B catalytic domain (unknown origin) assessed as reduction in pNP formation using pNPP as substrate by absorbance based analysis
AID292582Inhibition at monoamine oxidase in mouse brain2007Journal of natural products, Jul, Volume: 70, Issue:7
Prenylated xanthones from the root bark of Cudrania tricuspidata.
AID448782Inhibition of Clostridium welchii neuraminidase2009Bioorganic & medicinal chemistry letters, Sep-01, Volume: 19, Issue:17
Structural characteristics of flavanones and flavones from Cudrania tricuspidata for neuraminidase inhibition.
AID276035Inhibition of human ACAT2 expressed in Hi5 cells2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID1190077Induction of apoptosis in TRAIL-resistant human AGS cells assessed as decrease in cell viability at 40 uM after 24 hrs in presence of TRAIL (Rvb = 9%)2015Journal of natural products, Jan-23, Volume: 78, Issue:1
Prenylated flavonoids and resveratrol derivatives isolated from Artocarpus communis with the ability to overcome TRAIL resistance.
AID767576Induction of ROS production in LPS-stimulated mouse J774A1 cells at 1.25 uM after 24 hrs by flow cytometry relative to control2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID276042Inhibition of LPS-induced iNOS protein expression in mouse RAW 264.7 cells at 20 uM2006Bioorganic & medicinal chemistry letters, Nov-01, Volume: 16, Issue:21
Anti-atherosclerotic and anti-inflammatory activities of catecholic xanthones and flavonoids isolated from Cudrania tricuspidata.
AID595365Antiinflammatory activity against human THP1 cells of assessed as inhibition of LPS-induced TNFalpha protein synthesis at 10 uM pretreated for 1 hr before LPS challenge measured after 2 to 10 hrs2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID595366Antiinflammatory activity against human THP1 cells of assessed as inhibition of LPS-induced TNFalpha gene expression at 10 uM pretreated for 1 hr before LPS challenge measured after 2 hrs by RT-qPCR relative to control2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID1887296Antibacterial activity against Escherichia coli ATCC 25922 assessed as inhibition of bacterial growth incubated for 24 hrs by optical density based analysis2022Journal of natural products, 09-23, Volume: 85, Issue:9
Total Syntheses and Antibacterial Evaluations of Neocyclomorusin and Related Flavones.
AID595364Cytotoxicity against human THP1 cells after 24 hrs by erythrosin B staining2011Journal of natural products, Apr-25, Volume: 74, Issue:4
Natural compound cudraflavone B shows promising anti-inflammatory properties in vitro.
AID767580Induction of ROS production in mouse J774A1 cells at 1.25 uM after 30 mins by flow cytometry relative to control2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID767575Induction of ROS production in LPS-stimulated mouse J774A1 cells at 1.25 uM after 30 mins by flow cytometry relative to control2013Journal of natural products, Sep-27, Volume: 76, Issue:9
Prenylated and geranylated flavonoids increase production of reactive oxygen species in mouse macrophages but inhibit the inflammatory response.
AID448783Noncompetitive inhibition of Clostridium welchii neuraminidase by Dixon plot2009Bioorganic & medicinal chemistry letters, Sep-01, Volume: 19, Issue:17
Structural characteristics of flavanones and flavones from Cudrania tricuspidata for neuraminidase inhibition.
AID1347083qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: Viability assay - alamar blue signal for LASV Primary Screen2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID1347082qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: LASV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347086qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lymphocytic Choriomeningitis Arenaviruses (LCMV): LCMV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (102)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's35 (34.31)29.6817
2010's56 (54.90)24.3611
2020's11 (10.78)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 38.68

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

MetricThis Compound (vs All)
Research Demand Index38.68 (24.57)
Research Supply Index2.89 (2.92)
Research Growth Index4.83 (4.65)
Search Engine Demand Index53.49 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (38.68)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials1 (1.18%)5.53%
Trials0 (0.00%)5.53%
Reviews0 (0.00%)6.00%
Reviews1 (5.88%)6.00%
Case Studies0 (0.00%)4.05%
Case Studies0 (0.00%)4.05%
Observational0 (0.00%)0.25%
Observational0 (0.00%)0.25%
Other84 (98.82%)84.16%
Other16 (94.12%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (37)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Phase II Study of the Combination of Low-Intensity Chemotherapy and Blinatumomab in Patients With Philadelphia Chromosome Negative Relapsed/Refractory Acute Lymphoblastic Leukemia (ALL) [NCT03518112]Phase 26 participants (Actual)Interventional2018-04-18Terminated(stopped due to Due to Competing Studies)
A Phase 2 Study of Blinatumomab (NSC# 765986) in Combination With Nivolumab (NSC # 748726), a Checkpoint Inhibitor of PD-1, in B-ALL Patients Aged >/= 1 to < 31 Years Old With First Relapse [NCT04546399]Phase 2550 participants (Anticipated)Interventional2020-12-04Suspended(stopped due to Other - FDA Partial Clinical Hold)
A Phase III Randomized Trial of Blinatumomab for Newly Diagnosed BCR-ABL-Negative B Lineage Acute Lymphoblastic Leukemia in Adults [NCT02003222]Phase 3488 participants (Actual)Interventional2014-05-19Active, not recruiting
A Phase 2 Study of Ibrutinib and Blinatumomab in Relapsed and Refractory B-Cell Acute Lymphoblastic Leukemia [NCT02997761]Phase 220 participants (Anticipated)Interventional2017-06-27Recruiting
Risk-Stratified Randomized Phase III Testing of Blinatumomab (NSC#765986) in First Relapse of Childhood B-Lymphoblastic Leukemia (B-ALL) [NCT02101853]Phase 3669 participants (Actual)Interventional2014-12-17Active, not recruiting
Administration of Anti-CD3 (OKT3) in Combination With Beta-D-Glucosylceramide [NCT00619372]Phase 120 participants (Anticipated)Interventional2008-01-31Active, not recruiting
Phase II Study of the Sequential Combination of Low-Intensity Chemotherapy and Ponatinib Followed by Blinatumomab and Ponatinib in Patients With Philadelphia Chromosome (Ph)-Positive and/or BCR-ABL Positive Acute Lymphoblastic Leukemia (ALL) [NCT03147612]Phase 221 participants (Actual)Interventional2018-02-08Active, not recruiting
Oral Anti-CD3 for the Treatment of Active Ulcerative Colitis [NCT01287195]Phase 1/Phase 26 participants (Actual)Interventional2011-04-07Completed
A Phase II Study of Blinatumomab and POMP (Prednisone, Vincristine, Methotrexate, 6-Mercaptopurine) for Patients ≥ 65 Years of Age With Newly Diagnosed Philadelphia-Chromosome Negative (Ph-) Acute Lymphoblastic Leukemia (ALL) and of Dasatinib, Prednisone [NCT02143414]Phase 253 participants (Actual)Interventional2015-06-30Active, not recruiting
Phase II Study of the Combination of Blinatumomab and Ponatinib in Patients With Philadelphia Chromosome (Ph)-Positive and/or BCR-ABL Positive Acute Lymphoblastic Leukemia (ALL) [NCT03263572]Phase 290 participants (Anticipated)Interventional2017-11-29Recruiting
Phase II Study of the Hyper-CVAD Regimen in Sequential Combination With Blinatumomab With or Without Inotuzumab Ozogamicin as Frontline Therapy for Adults With B-Cell Lineage Acute Lymphocytic Leukemia [NCT02877303]Phase 280 participants (Anticipated)Interventional2016-11-01Recruiting
A Reduced Intensity Conditioning Regimen With CD3-Depleted Hematopoietic Stem Cells to Improve Survival for Patients With Hematologic Malignancies Undergoing Haploidentical Stem Cell Transplantation [NCT00566696]Phase 273 participants (Actual)Interventional2007-12-14Completed
Treatment of Psoriatic Arthritis With hOKT3γ1 (Ala-Ala) [NCT00239720]Phase 24 participants (Actual)Interventional2006-03-16Terminated(stopped due to Study team decision-impact(s) of change in hOKT3γ1 (Ala-Ala) manufacturer during study.)
Haploidentical Hematopoietic Stem Cell Transplantation Using A Novel Clofarabine Containing Conditioning Regimen For Patients With Refractory Hematologic Malignancies [NCT00824135]Phase 134 participants (Actual)Interventional2009-01-31Completed
Phase II Study of Blinatumomab in Patients With B-Cell Lineage Acute Lymphocytic Leukemia With Positive Minimal Residual Disease [NCT02458014]Phase 236 participants (Actual)Interventional2015-09-14Active, not recruiting
A Phase 3 Trial Investigating Blinatumomab (NSC# 765986) in Combination With Chemotherapy in Patients With Newly Diagnosed Standard Risk or Down Syndrome B-Lymphoblastic Leukemia (B-ALL) and the Treatment of Patients With Localized B-Lymphoblastic Lymphom [NCT03914625]Phase 36,720 participants (Anticipated)Interventional2019-07-03Recruiting
Durable-Response Therapy Evaluation For Early or New-Onset Type 1 Diabetes - DEFEND [NCT00678886]Phase 3272 participants (Actual)Interventional2008-07-29Completed
Allogeneic Bone Marrow Transplantation for Marrow Failure States [NCT00005852]Phase 20 participants Interventional1996-06-30Terminated(stopped due to low accrual)
Randomized, Multicenter, 2-Dose Level. Open-Label, Phase IIa Study With the Intraperitoneally Infused Trifunctional Bispecific Antibody Removab(TM) (Anti-EpCAM x Anti-CD3) to Select the Better Dose Level in Platinum Refractory Epithelial Ovarian Cancer Pa [NCT00189345]Phase 244 participants Interventional2004-05-31Completed
A Phase I/IIa, Double-Blind, Placebo-Controlled, Dose-Escalation Study of NI-0401 in Patients With Moderate to Severe Active Crohn´s Disease [NCT00630643]Phase 1/Phase 240 participants (Actual)Interventional2006-01-31Completed
[NCT00004482]Phase 240 participants Interventional1999-12-31Completed
A Dose-Finding Phase Ib Study of the Oral BCL-2 Inhibitor Venetoclax (ABT-199) in Combination With Standard Induction Therapy, Dasatinib, Prednisone, (and Rituximab in CD20+ Patients) in Adult Patients With Newly Diagnosed and Relapsed Philadelphia Chromo [NCT04872790]Phase 120 participants (Anticipated)Interventional2022-09-02Recruiting
A Phase III Randomized Trial of Steroids + Tyrosine Kinase Inhibitor (TKI) Induction With Chemotherapy or Blinatumomab for Newly Diagnosed BCR-ABL-Positive Acute Lymphoblastic Leukemia (ALL) in Adults [NCT04530565]Phase 3348 participants (Anticipated)Interventional2021-01-25Recruiting
Durable-Response Therapy Evaluation For Early- or New-Onset Type 1 Diabetes [NCT01123083]Phase 3179 participants (Actual)Interventional2010-05-17Completed
Giant Cell Myocarditis Treatment Trial Pilot Study [NCT00027443]40 participants (Anticipated)Interventional2001-08-31Completed
Transplantation of Haploidentical CD34+ Purified Peripheral Blood Stem Cells With NK-Cell Add-Back Following Conditioning With Total Body Irradiation, Thiotepa, Fludarabine and OKT3 [NCT00450983]Phase 21 participants (Actual)Interventional2006-12-31Terminated
A Phase IB Multiple Ascending Dose Study of Safety and Tolerability of Orally Administered Foralumab, a Fully Human Anti-CD3 Monoclonal Antibody, in Subjects With Moderate to Severely Active Crohn's Disease [NCT05028946]Phase 10 participants (Actual)Interventional2022-05-01Withdrawn(stopped due to For business reasons)
Adjuvant Autolymphocyte Therapy (ALT) For Patients With Non-Metastatic Renal Cell Carcinoma [NCT00002589]Phase 290 participants (Anticipated)Interventional1994-07-31Active, not recruiting
A PHASE I-II TRIAL OF MUROMONAB (OKT-3) WITH LOW-DOSE CYCLOPHOSPHAMIDE [NCT00002482]Phase 1/Phase 20 participants Interventional1991-06-30Active, not recruiting
Intracavitary Allogenic Cytotoxic T Lymphocytes and Human Recombinant Interleukin-2 Therapy for Recurrent Primary Brain Tumors [NCT00002572]Phase 110 participants (Anticipated)Interventional1994-11-30Completed
Immunotherapy for Malignant Glioma - Phase II Trial of Autologous Cancer Antigen Specific Immunotherapy [NCT00004024]Phase 260 participants (Anticipated)Interventional1997-06-30Completed
Immunotherapy for Malignant Melanoma - Phase II Trial of Autologous Cancer Antigen Specific Immunotherapy [NCT00004022]Phase 20 participants Interventional1997-06-30Completed
A Phase 1/2 Trial of Pembrolizumab in Combination With Blinatumomab in Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia [NCT03512405]Phase 1/Phase 236 participants (Anticipated)Interventional2019-08-02Recruiting
A Randomized International Phase 3 Trial of Imatinib and Chemotherapy With or Without Blinatumomab in Patients With Newly-Diagnosed Philadelphia Chromosome-Positive or Philadelphia Chromosome-Like ABL-Class B-Cell Acute Lymphoblastic Leukemia [NCT06124157]Phase 3680 participants (Anticipated)Interventional2024-01-22Not yet recruiting
A Phase II Study of Inotuzumab Ozogamicin Followed by Blinatumomab for Ph-Negative CD22-Positive B-Lineage Acute Lymphoblastic Leukemia in Newly Diagnosed Older Adults or Adults With Relapsed or Refractory Disease [NCT03739814]Phase 264 participants (Anticipated)Interventional2019-05-08Recruiting
A Phase 1 Study of Blinatumomab in Combination With Checkpoint Inhibitor(s) of PD-1 (Nivolumab) or Both PD-1 (Nivolumab) and CTLA-4 (Ipilimumab) in Patients With Poor-Risk, Relapsed or Refractory CD19+ Precursor B-Lymphoblastic Leukemia [NCT02879695]Phase 124 participants (Actual)Interventional2017-10-25Active, not recruiting
A Phase I Trial of the Combination of Lenalidomide and Blinatumomab in Patients With Relapsed or Refractory Non-Hodgkins Lymphoma (NHL) [NCT02568553]Phase 144 participants (Anticipated)Interventional2016-11-15Active, not recruiting
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00450983 (4) [back to overview]Risk of Developing Grades III-IV Acute Graft-vs-host Disease (GVHD)
NCT00450983 (4) [back to overview]Risk for Mortality From Infection Before Day 180
NCT00450983 (4) [back to overview]Risk for Life-threatening Infections
NCT00450983 (4) [back to overview]Risk for Graft Failure
NCT00566696 (7) [back to overview]Disease-Free Survival (DFS)
NCT00566696 (7) [back to overview]Event-free Survival (EFS)
NCT00566696 (7) [back to overview]Incidence of Regimen-related Mortality
NCT00566696 (7) [back to overview]Overall Survival (OS)
NCT00566696 (7) [back to overview]Incidence of Non-hematologic Regimen-related Toxicities
NCT00566696 (7) [back to overview]To Estimate the Cumulative Incidence of Relapse for Research Participants Who Receive This Study Treatment.
NCT00566696 (7) [back to overview]To Estimate the Rate of Overall Grade III-IV Acute GVHD, and the Rate and Severity of Chronic GVHD in Research Participants.
NCT00678886 (20) [back to overview]Change From Baseline in Average Daily Risk Range (ADRR) at Week 12 and Months 6 and 12.
NCT00678886 (20) [back to overview]Percent Change From Baseline in CD3/TCR Saturation on CD4+ T Cells and CD8+ T Cells at Day 1, Day 4, Day 8
NCT00678886 (20) [back to overview]Composite Rank Summary for C-Peptide AUC, HbA1c and Exogenous Insulin Use at Month 6 and Month 12
NCT00678886 (20) [back to overview]Composite Rank Summary for HbA1c and Exogenous Insulin Use at Month 6 and Month 12
NCT00678886 (20) [back to overview]HbA1c Level at Week 12 and Months 6 and 12
NCT00678886 (20) [back to overview]Magnitude of Greatest Hyperglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12.
NCT00678886 (20) [back to overview]Magnitude of Greatest Hypoglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12.
NCT00678886 (20) [back to overview]Mean Daily Insulin Use at Week 12 and Months 6 and 12.
NCT00678886 (20) [back to overview]Change From Baseline in Level of Cytokines Interleukin (IL-6), IL-10 and Tumor Necrosis Factor-alpha (TNF-a) at Day 1, Day 4, Day 8
NCT00678886 (20) [back to overview]Number of Hyperglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12.
NCT00678886 (20) [back to overview]Number of Hypoglycemic Events Defined by Hypoglycemic Event Categories From Baseline Upto Month 12
NCT00678886 (20) [back to overview]Number of Hypoglycemic Excursions (<=70 mg/dL) With Most Complete Glucose at Week 12 and Months 6 and 12.
NCT00678886 (20) [back to overview]Number of Participants Who Were Responders for (Glycosylated Hemoglobin) HbA1c/Insulin Use Response at Week 12 and Months 6 and 12
NCT00678886 (20) [back to overview]Number of Participants With Hyperglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12
NCT00678886 (20) [back to overview]Number of Participants With Hypoglycemic Events Defined by Hypoglycemic Event Categories From Baseline Upto Month 12
NCT00678886 (20) [back to overview]Number of Participants With Hypoglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12
NCT00678886 (20) [back to overview]Percent Change From Baseline in CD3/TCR Modulation on CD4+ T Cells and CD8+ T Cells at Day 1, Day 4, Day 8
NCT00678886 (20) [back to overview]Percent Change From Baseline in Cell-bound Otelixizumab on CD4+ T Cells at Day 1, Day 4, Day 8
NCT00678886 (20) [back to overview]Percent Change From Baseline in Circulating Peripheral Lymphocytes CD4+CD25+FoxP3+ T Cells and CD4+CD25hiFoxP3+ T Cells in Type 1 Diabetes Mellitus (TIDM) up to Month 12
NCT00678886 (20) [back to overview]Change From Baseline in 2-hour Mixed Meal Stimulated C-peptide Area Under Curve [AUC] (Normalized for 120-minute Time Interval) at Month 12
NCT01123083 (10) [back to overview]Change From Baseline in Stimulated C-Peptide Mean AUC at Week 12, Month 6, 12 and 18
NCT01123083 (10) [back to overview]Number of Participants With Responder Status
NCT01123083 (10) [back to overview]Composite Rank Sum: HbA1c and Exogenous Insulin Use at 6 and 12 Months
NCT01123083 (10) [back to overview]Composite Rank Sum: C-Peptide AUC, HbA1c and Exogenous Insulin Use at 6 and 12 Months
NCT01123083 (10) [back to overview]Change From Baseline in Mean Daily Insulin Use Over 7 Consecutive Days During the 2 Weeks Prior to the Assessment
NCT01123083 (10) [back to overview]Change From Baseline in HbA1c Levels Over 7 Consecutive Days During the 2 Weeks Prior to the Assessment
NCT01123083 (10) [back to overview]Change From Baseline in 2 Hour Mixed Meal-stimulated C-peptide AUC (Normalized for 120-minute Time Interval) at Week 12 and 6 Months
NCT01123083 (10) [back to overview]Average Number of Severe Hypoglycemic Events and Documented Symptomatic Hypoglycemic Events From Baseline to Month 12
NCT01123083 (10) [back to overview]Change From Baseline in 2 Hour Mixed Meal-stimulated C-peptide Area Under Curve (AUC) (Normalized for 120-minute Time Interval) at Month 12
NCT01123083 (10) [back to overview]Percentage of Participants With Change From Baseline in Severe Hypoglycemic Events and Documented Symptomatic Hypoglycemic Events at Month 12
NCT01287195 (8) [back to overview]Number of Participants With Anti-Drug Antibodies
NCT01287195 (8) [back to overview]Cytokine Production by PBMCs in Cell Culture
NCT01287195 (8) [back to overview]Mayo Score
NCT01287195 (8) [back to overview]Percentage of Biomarker-positive Immune Cells
NCT01287195 (8) [back to overview]Score in Histologic Evaluation of Flexible Sigmoidoscopy
NCT01287195 (8) [back to overview]Simple Clinical Colitis Activity Index (SCCAI) Score
NCT01287195 (8) [back to overview]T Cell Proliferation of PBMCs in Cell Culture
NCT01287195 (8) [back to overview]Number of Participants With Adverse Events
NCT02101853 (4) [back to overview]Overall Survival (OS) of HR and IR Relapse Patients
NCT02101853 (4) [back to overview]Disease Free Survival (DFS) of Low Risk (LR) Relapse Patients
NCT02101853 (4) [back to overview]Disease Free Survival (DFS) of High-risk (HR) and Intermediate-risk (IR) Relapse Patients
NCT02101853 (4) [back to overview]Overall Survival (OS) of LR Relapse Patients
NCT02143414 (6) [back to overview]Number of Participants With Grade 3 Through 5 Adverse Events That Are Related to Study Drugs
NCT02143414 (6) [back to overview]Incidence of Dose-limiting Toxicity (Cohort II)
NCT02143414 (6) [back to overview]Minimal Residual Disease Negativity
NCT02143414 (6) [back to overview]Complete Response Rate (Cohort I)
NCT02143414 (6) [back to overview]Overall Survival Rate (Cohort I)
NCT02143414 (6) [back to overview]Disease-free Survival (Cohort II)
NCT03518112 (5) [back to overview]Duration of Response
NCT03518112 (5) [back to overview]Participants With a Response
NCT03518112 (5) [back to overview]Overall Survival
NCT03518112 (5) [back to overview]Number of Participants Negative for Minimal Residual Disease (MRD)
NCT03518112 (5) [back to overview]Event Free Survival (EFS) Where Events Defined as no Response, Loss of Response, or Death

Risk of Developing Grades III-IV Acute Graft-vs-host Disease (GVHD)

Count of participants with acute GVHD grades III-IV. (NCT00450983)
Timeframe: Up to day 100

InterventionParticipants (Count of Participants)
Treatment0

[back to top]

Risk for Mortality From Infection Before Day 180

Count of participant deaths from infection up to day 180. (NCT00450983)
Timeframe: Up to day 180

InterventionParticipants (Count of Participants)
Treatment0

[back to top]

Risk for Life-threatening Infections

Count of participants with life-threatening infections (NCT00450983)
Timeframe: Up to day 100

InterventionParticipants (Count of Participants)
Treatment1

[back to top]

Risk for Graft Failure

Count of participant that had graft failure. (NCT00450983)
Timeframe: Engraftment documented day +20

InterventionParticipants (Count of Participants)
Treatment0

[back to top]

Disease-Free Survival (DFS)

Estimate the one-year disease-free survival (DFS) for research participants who receive this study treatment. DFS is defined as time from transplantation to the occurrence of relapse or death due to relapse. Patients who are alive at the time of analysis or die due to other causes will be censored at the time of their events. The estimated percentage of participants with DFS at one-year post-transplantation is reported using Kaplan-Meier analysis. (NCT00566696)
Timeframe: One year post-transplant

InterventionPercentage of participants (Number)
High-Risk Hematologic Malignancies70.1

[back to top]

Event-free Survival (EFS)

To determine if one year event-free survival can be improved in pediatric patients undergoing a haploidentical transplant by using a reduced intensity conditioning regimen and a targeted dose T cell depleted donor product. EFS is defined as time from transplantation to the occurrence of relapse or death due to any cause. Patients who are alive at the time of analysis will be censored. The estimated percentage of participants with EFS at one-year post-transplantation is reported using Kaplan-Meier analysis. (NCT00566696)
Timeframe: one year post-transplant

InterventionPercentage of participants (Number)
High-Risk Hematologic Malignancies54.8

[back to top] [back to top]

Overall Survival (OS)

Estimate the one-year overall survival (OS) for research participants who receive this study treatment. OS is defined as time from transplantation to death due to any cause. Patient who are alive at the time of analysis will be censored. The estimated percentage of participants with OS at one-year post-transplantation is reported using Kaplan-Meier analysis. (NCT00566696)
Timeframe: one year post-transplant

InterventionPercentage of participants (Number)
High-Risk Hematologic Malignancies71.0

[back to top] [back to top]

To Estimate the Cumulative Incidence of Relapse for Research Participants Who Receive This Study Treatment.

The Cumulative Incidence of Relapse will be reported as the proportion of number of relapses since transplant to the total number of patients at risk at five years post-transplant. (NCT00566696)
Timeframe: five years post-transplant

InterventionPercentage of participants (Number)
The Cumulative Incidence of Relapse at five year pEstimate±SE
High-Risk Hematologic Malignancies30.08.6

[back to top]

To Estimate the Rate of Overall Grade III-IV Acute GVHD, and the Rate and Severity of Chronic GVHD in Research Participants.

The rate of Overall Grade III-IV Acute AVHD will be report as the proportion of patients who have Grade III-IV Acute GVHD to the total patients with transplant. The rate of Chronic GVHD will be report as the proportion of patients who have Chronic GVHD to the total patients with transplant. The Severity of Chronic GVHD will be reported using CTCAE grading system, as a number. (NCT00566696)
Timeframe: five years post-transplant

InterventionPercentage of participants (Number)
Rate of Overall Grade III-IV Acute AVHDRate of limited grade Chronic GVHD
High-Risk Hematologic Malignancies22.589.68

[back to top]

Change From Baseline in Average Daily Risk Range (ADRR) at Week 12 and Months 6 and 12.

Average daily risk range is a measure for evaluation of blood glucose variability that was designed to be equally sensitive to hypoglycemia and hyperglycemia. The ADRR was assessed over 30-day periods prior to Baseline and at key visits Week 12 and Months 6 and 12. Baseline assessments were carried out on the morning of Day 1, before the start of the first infusion of study drug. Change from Baseline was calculated by subtracting the Baseline value from the post-randomization value at Week 12 and Months 6 and 12. (NCT00678886)
Timeframe: Baseline (Day 1) and Week 12, Months 6 and 12.

,
InterventionRatio (Least Squares Mean)
ADRR at Week 12ADRR at Month 6ADRR at Month 12
Otelixizumab0.712.864.22
Placebo-0.412.033.29

[back to top]

Percent Change From Baseline in CD3/TCR Saturation on CD4+ T Cells and CD8+ T Cells at Day 1, Day 4, Day 8

The extent of saturation of CD3/TCR receptors on CD4+ and CD8+ lymphocytes was determined by flow cytometry. The extent of TCRαβ expression was determined using an antibody that bound to TCRαβ but did not compete with otelixizumab for binding sites at the expected range of concentrations. The MESF of the anti-TCRαβ antibody was used to quantify the number of CD3/TCR complexes present on T cells. The MESF of bound biotinylated otelixizumab was directly proportional to the availability of free otelixizumab binding sites. MESF of free CD3 sites on CD4+ T cells and CD8+ T cells was direct measurement of saturation of the CD3/TCR complex on CD4+ T cells and CD8+ T cells. Baseline assessments were carried out on the morning of Day 1, before the start of the first infusion of study drug. Change from Baseline was calculated by subtracting the Baseline value from the post-randomization value at Day 1, Day 4 and Day 8. (NCT00678886)
Timeframe: Baseline (Pre-dose Day 1), Day 4, Day 8

,
InterventionPercent change (Mean)
CD4+ T cells, Day 1, 2 hours after EOICD4+ T cells, Day 4, 2 hours after EOICD4+ T cells, Day 8, pre-doseCD4+ T cells, Day 8, 2 hours after EOICD8+ T cells, Day 1, 2 hours after EOICD8+ T cells, Day 4, 2 hours after EOICD8+ T cells, Day 8, pre-doseCD8+ T cells, Day 8, 2 hours after EOI
Otelixizumab90.950.259.021.791.451.660.823.0
Placebo93.4109.797.798.092.4116.797.498.2

[back to top]

Composite Rank Summary for C-Peptide AUC, HbA1c and Exogenous Insulin Use at Month 6 and Month 12

O'Brien analyses will be performed on a three-part composite of HbA1c level, C-peptide AUC, and mean daily Insulin use in the otelixizumab group compared with the placebo group at Months 6 and12. For the O'Brien mean rank analysis at a particular time point, HbA1c and insulin use will be ranked from smallest to largest, and C-peptide AUC will be ranked from largest to smallest. For each participant, the C-Peptide AUC, ranks for HbA1c and insulin use were added together, producing a composite rank. A treatment comparison test was then performed on the composite ranks. (NCT00678886)
Timeframe: Month 6 and 12

,
InterventionComposite rank score (Mean)
Month 6Month 12
Otelixizumab373366
Placebo365360

[back to top]

Composite Rank Summary for HbA1c and Exogenous Insulin Use at Month 6 and Month 12

O'Brien mean rank analyses was performed on a two-part composite of the baseline-adjusted HbA1c level and the baseline-adjusted mean total daily insulin use per kg body weight in the otelixizumab group compared with the placebo group at Months 6, 12. For the O'Brien mean rank analysis at a particular time point, adjusted HbA1c values (for both treatment groups together) was ranked from smallest to largest, and adjusted mean daily insulin use values were ranked from smallest to largest. For each participant, the HbA1c and insulin use ranks were added together, producing a composite rank. A treatment comparison test was then performed on the composite ranks. (NCT00678886)
Timeframe: Month 6 and 12

,
InterventionComposite rank score (Mean)
Composite rank summary, Month 6Composite rank summary, Month 12
Otelixizumab247244
Placebo238245

[back to top]

HbA1c Level at Week 12 and Months 6 and 12

HbA1c levels were recorded at Screening, Baseline (Day 1), Day 28, Week 8, Week 12, Months 4 to 12 and Month 24. Data has been presented for HbA1c levels at Week 12 and Months 6 and 12. (NCT00678886)
Timeframe: Week 12 and Months 6 and 12

,
InterventionPercentage (Least Squares Mean)
HbA1c levels at Month 12HbA1c levels at Month 6HbA1c levels at Week 12
Otelixizumab7.026.776.54
Placebo6.836.606.45

[back to top]

Magnitude of Greatest Hyperglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12.

The greatest hyperglycemic excursions during an interval was calculated as the largest recorded glucose level in the interval minus the hyperglycemic tolerance limit value (HGTLV). If a participant had data recorded during the interval but did not have a value above the HGTLV, the participants greatest hyperglycemic excursion for that interval was 0 mg/dL. Most complete glucose interval was the 7 day period with the maximum number of days with at least 4 recordings per day. If these results were in more than one 7 day period, then the 7 day period with the largest average number of daily recordings (out of those with the maximum number of days with at least 4 recordings per day) was selected. If there were 2 or more 7 day periods that have the same number of days with at least 4 recordings and the same maximum average number of glucose recordings, the period that ended closest to the day of the study was selected. (NCT00678886)
Timeframe: Week 12 and Months 6 and 12.

,
Interventionmilligrams per deciliter (Mean)
Threshold> 100 mg/dL at Week 12Threshold> 130 mg/dL at Week 12Threshold> 200 mg/dL at Week 12Threshold> 100 mg/dL at Month 6Threshold> 130 mg/dL at Month 6Threshold> 200 mg/dL at Month 6Threshold> 100 mg/dL at Month 12Threshold> 130 mg/dL at Month 12Threshold> 200 mg/dL at Month 12
Otelixizumab158.4128.465.7175.7145.780.1177.5147.582.6
Placebo158.3128.365.7154.6124.863.3176.7146.881.8

[back to top]

Magnitude of Greatest Hypoglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12.

"The greatest hypoglycemic excursions during an interval was calculated as 70 mg/dL minus the lowest recorded glucose level in the interval. If a participant had data recorded during the interval but did not have a value below 70 mg/dL, the participants greatest hypoglycemic excursion for that interval was 0 mg/dL.~Most complete glucose interval was the 7 day period with the maximum number of days with at least 4 recordings per day. If these results were in more than one 7 day period, then the 7 day period with the largest average number of daily recordings (out of those with the maximum number of days with at least 4 recordings per day) was selected. If there were 2 or more 7 day periods that have the same number of days with at least 4 recordings and the same maximum average number of glucose recordings, the period that ended closest to the day of the study was selected." (NCT00678886)
Timeframe: Week 12 and Months 6 and 12.

,
Interventionmilligrams per deciliter (Mean)
Magnitude of excursion, Week 12Magnitude of excursion, Month 6Magnitude of excursion, Month 12
Otelixizumab10.810.611.0
Placebo9.411.911.2

[back to top]

Mean Daily Insulin Use at Week 12 and Months 6 and 12.

Participants recorded their daily insulin use in their electronic diaries. In particular, insulin was recorded thoroughly and accurately for at least 7 consecutive days during the 2 weeks before the visits at Baseline, Week 12, and Months 6 and 12. During each of these visits, the investigator/designee accessed the invivodata DiaryPRO web site to review insulin-use data for the previous 2-week period to ensure completeness. If errors/gaps were identified (e.g., if the participant did not take insulin and not entered 0 units), the investigator/designee recorded the missing data from participant recall using a data clarification form (DCF). Paper diary to collect insulin use, were reviewed for completeness. Any missing data that could be recalled by the participant was entered. If the participant did not record any insulin use during the 2-week period before the visit, the site obtained an insulin use history for the previous 7 days and calculated the average daily insulin dose. (NCT00678886)
Timeframe: Week 12 and Months 6 and 12.

,
InterventionIU/kg (Least Squares Mean)
Mean daily insulin use at Week 12Mean daily insulin use at Month 6Mean daily insulin use at Month 12
Otelixizumab0.310.360.39
Placebo0.340.360.42

[back to top]

Change From Baseline in Level of Cytokines Interleukin (IL-6), IL-10 and Tumor Necrosis Factor-alpha (TNF-a) at Day 1, Day 4, Day 8

Levels of cytokine (TNFα, IL-6, IL-10) were measured at Baseline and at 2 hours after end of infusion (EOI) on Day 1, Day 4, Day 8 . Baseline assessments were carried out on the morning of Day 1, before the start of the first infusion of study drug. Change from Baseline was calculated by subtracting the Baseline value from the post-randomization value at Day 1, Day 4 and Day 8. (NCT00678886)
Timeframe: Day 1, Day 4, Day 8

,
Interventionpicograms per milliliter (Mean)
IL-6 levels, Day 1, 2 hours after EOIIL-6 levels, Day 4, 2 hours after EOIIL-6 levels, Day 8, 2 hours after EOIIL-10 levels, Day 1, 2 hours after EOIIL-10 levels, Day 4, 2 hours after EOIIL-10 levels, Day 8, 2 hours after EOITNF-a levels, Day 1, 2 hours after EOITNF-a levels, Day 4, 2 hours after EOITNF-a levels, Day 8, 2 hours after EOI
Otelixizumab2.783.0114.594.611.3525.572.3201.5093.614
Placebo0.76-0.130.450.150.551.84-0.1260.1110.135

[back to top]

Number of Hyperglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12.

The event frequency of glucose measurements that were hyperglycemic excursions were calculated on a per participant basis using the number of occurrences where blood glucose was greater than the hyperglycemic tolerance limit. There were 3 hyperglycemic tolerance limits considered: 200 mg/dL, 130 mg/dL and 100 mg/dL. Most complete glucose interval was the 7 day period with the maximum number of days with at least 4 recordings per day. If these results were in more than one 7 day period, then the 7 day period with the largest average number of daily recordings (out of those with the maximum number of days with at least 4 recordings per day) was selected. If there were 2 or more 7 day periods that have the same number of days with at least 4 recordings and the same maximum average number of glucose recordings, the period that ended closest to the day of the study was selected. (NCT00678886)
Timeframe: Week 12 and Months 6 and 12

,
InterventionHyperglycemic excursions (Mean)
Threshold > 100 mg/dL at Week 12Threshold > 130 mg/dL at Week 12Threshold > 200 mg/dL at Week 12Threshold > 100 mg/dL at Month 6Threshold > 130 mg/dL at Month 6Threshold > 200 mg/dL at Month 6Threshold > 100 mg/dL at Month 12Threshold > 130 mg/dL at Month 12Threshold > 200 mg/dL at Month 12
Otelixizumab32.919.95.635.122.77.933.422.26.9
Placebo34.020.55.733.520.86.535.624.08.6

[back to top]

Number of Hypoglycemic Events Defined by Hypoglycemic Event Categories From Baseline Upto Month 12

Hypoglycemic events reported by participants were classified as defined by the American Diabetes Association (ADA) Workgroup on Hypoglycemia as follows: Severe hypoglycemia: an event requiring assistance of another person to actively administer carbohydrate, glucagon/other resuscitative actions, documented symptomatic hypoglycemia: an event during which typical symptoms of hypoglycemia are accompanied by a measured plasma glucose concentration(PGC)<=70 mg/dL, asymptomatic hypoglycemia: an event not accompanied by typical symptoms of hypoglycemia but with a measured PGC<=70 mg/dL,probable symptomatic hypoglycemia: an event during which symptoms of hypoglycemia are not accompanied by a plasma glucose determination, but were presumably caused by a PGC<=70 mg/dL and relative hypoglycemia: an event during which the person with diabetes reports any of the typical symptoms of hypoglycemia, and interprets the symptoms as indicative of hypoglycemia, but with a measured PGC>70 mg/dL. (NCT00678886)
Timeframe: Upto Month 12

,
InterventionHypoglycemic events (Number)
Severe hypoglycemia upto Month 12Documented symptomatic hypoglycemia upto Month 12Asymptomatic hypoglycemia upto Month 12Probable symptomatic hypoglycemia upto Month 12Relative hypoglycemia upto Month 12
Otelixizumab56322996293211
Placebo2309247188485

[back to top]

Number of Hypoglycemic Excursions (<=70 mg/dL) With Most Complete Glucose at Week 12 and Months 6 and 12.

The event frequency of glucose measurements that were hypoglycemic excursions were calculated per participant basis, using the number of occurrences where blood glucose was less than or equal to 70 mg/dL. Most complete glucose interval was the 7 day period with the maximum number of days with at least 4 recordings per day. If these results were in more than one 7 day period, then the 7 day period with the largest average number of daily recordings (out of those with the maximum number of days with at least 4 recordings per day) was selected. If there were 2 or more 7 day periods that have the same number of days with at least 4 recordings and the same maximum average number of glucose recordings, the period that ended closest to the day of the study was selected. (NCT00678886)
Timeframe: Week 12 and Months 6 and 12.

,
InterventionHypoglycemic excursions (Mean)
Number of hypoglycemic excursions at Week 12Number of hypoglycemic excursions at Month 6Number of hypoglycemic excursions at Month 12
Otelixizumab2.82.93.2
Placebo2.33.02.9

[back to top]

Number of Participants Who Were Responders for (Glycosylated Hemoglobin) HbA1c/Insulin Use Response at Week 12 and Months 6 and 12

A participant was considered a responder if, at the given time point, the participant had HbA1c<= 6.5%, and mean daily insulin use over 7 consecutive days < 0.5 international units per kilogram per day (IU/kg/day) during the 2 weeks preceding the visit. Data has been presented from number of participants with their percentages who were responders at Week 12 and Months 6 and 12. (NCT00678886)
Timeframe: Week 12 and Months 6 and 12

,
InterventionParticipants (Count of Participants)
Responders at Week 12Responders at Month 6Responders at Month 12
Otelixizumab857456
Placebo454234

[back to top]

Number of Participants With Hyperglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12

Percentage of hyperglycemic excursions was calculated as the total number of observations that exceed the hyperglycemic excursion boundary (i.e. > HGTLV) divided by the total number of glucose measurements recorded in a time interval for the intervals: Baseline to Week 12, post-Week 12 to Month 6, post-Month 6 to Month 12. Most complete glucose interval was the 7 day period with the maximum number of days with at least 4 recordings per day. If these results were in more than one 7 day period, then the 7 day period with the largest average number of daily recordings (out of those with the maximum number of days with at least 4 recordings per day) was selected. If there were 2 or more 7 day periods that have the same number of days with at least 4 recordings and the same maximum average number of glucose recordings, the period that ended closest to the day of the study was selected. Data for number of participants with their percentages are presented. (NCT00678886)
Timeframe: Week 12 and Months 6 and 12

,
InterventionParticipants (Count of Participants)
Threshold> 100 mg/dL at Week 12Threshold> 130 mg/dL at Week 12Threshold> 200 mg/dL at Week 12Threshold> 100 mg/dL at Month 6Threshold> 130 mg/dL at Month 6Threshold> 200 mg/dL at Month 6Threshold> 100 mg/dL at Month 12Threshold> 130 mg/dL at Month 12Threshold> 200 mg/dL at Month 12
Otelixizumab177177130166166139161160131
Placebo909065807957787760

[back to top]

Number of Participants With Hypoglycemic Events Defined by Hypoglycemic Event Categories From Baseline Upto Month 12

Hypoglycemic events reported by participants were classified as defined by the ADA Workgroup on Hypoglycemia as Severe hypoglycemia: an event requiring assistance of another person to actively administer carbohydrate, glucagon/other resuscitative actions, documented symptomatic hypoglycemia: an event during which typical symptoms of hypoglycemia are accompanied by a measured plasma glucose concentration (PGC)<=70 mg/dL, asymptomatic hypoglycemia: an event not accompanied by typical symptoms of hypoglycemia but with a measured PGC<=70 mg/dL,probable symptomatic hypoglycemia: an event during which symptoms of hypoglycemia are not accompanied by a plasma glucose determination, but were presumably caused by a PGC<=70 mg/dL and relative hypoglycemia: an event during which the person with diabetes reports any of the typical symptoms of hypoglycemia, and interprets the symptoms as indicative of hypoglycemia, but with a measured PGC>70 mg/dL. Only categories with values are presented. (NCT00678886)
Timeframe: Upto Month 12

,
InterventionParticipants (Count of Participants)
Severe hypoglycemia upto Month 12Documented symptomatic hypoglycemia upto Month 12Asymptomatic hypoglycemia upto Month 12
Otelixizumab416313
Placebo27810

[back to top]

Number of Participants With Hypoglycemic Excursions With Most Complete Glucose at Week 12 and Months 6 and 12

Percentage of hypoglycemic excursions was calculated as the total number of observations that exceed the hypoglycemic excursion boundary (i.e.<= 70 mg/dL) divided by the total number of glucose measurements recorded in a time interval for the intervals: Baseline to Week 12, post-Week 12 to Month 6, post-Month 6 to Month 12. Most complete glucose interval was the 7 day period with the maximum number of days with at least 4 recordings per day. If these results were in more than one 7 day period, then the 7 day period with the largest average number of daily recordings (out of those with the maximum number of days with at least 4 recordings per day) was selected. If there were 2 or more 7 day periods that have the same number of days with at least 4 recordings and the same maximum average number of glucose recordings, the period that ended closest to the day of the study was selected. Data has been presented for number of participants with their percentages having hypoglycemic excursion. (NCT00678886)
Timeframe: Week 12 and Months 6 and 12

,
InterventionParticipants (Count of Participants)
Percentage of participants with excursion,Week 12Percentage of participants with excursion,Month 6Percentage of participants with excursion,Month 12
Otelixizumab127121111
Placebo725756

[back to top]

Percent Change From Baseline in CD3/TCR Modulation on CD4+ T Cells and CD8+ T Cells at Day 1, Day 4, Day 8

The extent of modulation of CD3/TCR receptors on CD4+ and CD8+ lymphocytes was determined by flow cytometry. The extent of TCRαβ expression was determined using an antibody that bound to TCRαβ but did not compete with otelixizumab for binding sites at the expected range of concentrations. The MESF of the anti-TCRαβ antibody was used to quantify the number of CD3/TCR complexes present on T cells. The MESF of bound biotinylated otelixizumab was directly proportional to the availability of free otelixizumab binding sites.CD3/TCR complexes on CD4+ and CD8+ T cells were detected with a non-competing antibody. Changes in the MESF of TCR expression was a direct measurement of TCR modulation. Baseline assessments were carried out on the morning of Day 1, before the start of the first infusion of study drug. Change from Baseline was calculated by subtracting the Baseline value from the post-randomization value at Day 1, Day 4 and Day 8. (NCT00678886)
Timeframe: Baseline (Pre-dose Day 1), Day 4, Day 8

,
InterventionPercent change (Mean)
CD4+ T cells, Day 1, 2 hours after EOICD4+ T cells, Day 4, 2 hours after EOICD4+ T cells, Day 8, pre-doseCD4+ T cells, Day 8, 2 hours after EOICD8+ T cells, Day 1, 2 hours after EOICD8+ T cells, Day 4, 2 hours after EOICD8+ T cells, Day 8, pre-doseCD8+ T cells, Day 8, 2 hours after EOI
Otelixizumab91.170.861.032.293.777.162.336.2
Placebo94.2103.8101.4103.495.2111.1101.8103.0

[back to top]

Percent Change From Baseline in Cell-bound Otelixizumab on CD4+ T Cells at Day 1, Day 4, Day 8

The amount of cell-bound otelixizumab was determined by flow cytometry. The extent of T cell receptor alpha beta (TCRαβ) expression was determined using an antibody that bound to TCRαβ but did not compete with otelixizumab for binding sites at the expected range of concentrations. The MESF of the anti-TCRαβ antibody was used to quantify the number of CD3/TCR complexes present on T cells. Free otelixizumab binding sites (sites not occupied by otelixizumab) were detected by staining with biotinylated otelixizumab. The MESF of bound biotinylated otelixizumab was directly proportional to the availability of free otelixizumab binding sites. MESF of bound antibody on CD4+ T cells was a direct measurement of cell-bound otelixizumab on CD4+ T cells. Baseline assessments were carried out on the morning of Day 1, before the start of the first infusion of study drug. Change from Baseline was calculated by subtracting the Baseline value from the post-randomization value at Day 1, Day 4 and Day 8. (NCT00678886)
Timeframe: Baseline (pre-dose on Day 1), Day 4 and Day 8

,
InterventionPercent change (Mean)
CD4+ T cells, Day 1, 2 hours after EOICD4+ T cells, Day 4, 2 hours after EOICD4+ T cells, Day 8, pre-doseCD4+ T cells, Day 8, 2 hours after EOI
Otelixizumab914.82719.8471.51387.5
Placebo125.1174.3151.5167.9

[back to top]

Percent Change From Baseline in Circulating Peripheral Lymphocytes CD4+CD25+FoxP3+ T Cells and CD4+CD25hiFoxP3+ T Cells in Type 1 Diabetes Mellitus (TIDM) up to Month 12

Blood samples were drawn for lymphocyte subset evaluations at Baseline and at 2hours after EOI on Day 4, pre-dose and after E0I on Day 8, Day 14, Day 21, Day 28, Week 6, Week 8, Week 10, Week 12, Month 6 and Month 12. Percentages of relevant lymphocyte subsets were determined by flow cytometry. The lymphocyte subsets assessed included CD8+CD25+ T lymphocytes, as well as the subsets of lymphocytes of these type that were positive for FoxP3, a protein that was expressed at high levels in the cytoplasm of regulatory T cells. CD4+CD25hiFoxP3+ T lymphocytes, a cell type was of interest because it played a regulatory role in T1DM. Baseline assessments were carried out on the morning of Day 1, before the start of the first infusion of study drug. Change from Baseline was calculated by subtracting the Baseline value from the post-randomization value at the time of assessment. (NCT00678886)
Timeframe: Baseline (pre-dose on Day 1) and up to 12 Months

,
InterventionPercent change (Median)
CD4+CD25+FoxP3+ T, Day 4, 2 hours after EOICD4+CD25+FoxP3+ T, Day 8 pre-doseCD4+CD25+FoxP3+ T, Day 8, 2 hours after EOICD4+CD25+FoxP3+ T, Day 14CD4+CD25+FoxP3+ T, Day 21CD4+CD25+FoxP3+ T, Day 28CD4+CD25+FoxP3+ T, Week 6CD4+CD25+FoxP3+ T, Week 8CD4+CD25+FoxP3+ T, Week 10CD4+CD25+FoxP3+ T, Week 12CD4+CD25+FoxP3+ T, Month 6CD4+CD25+FoxP3+ T, Month 12CD4+CD25hiFoxP3+ T Cells, Day 4, 2 hours after EOICD4+CD25hiFoxP3+ T Cells, Day 8 pre-doseCD4+CD25hiFoxP3+ T Cells, Day 8, 2 hours after EOICD4+CD25hiFoxP3+ T Cells, Day 14CD4+CD25hiFoxP3+ T Cells, Day 21CD4+CD25hiFoxP3+ T Cells, Day 28CD4+CD25hiFoxP3+ T Cells, Week 6CD4+CD25hiFoxP3+ T Cells, Week 8CD4+CD25hiFoxP3+ T Cells, Week 10CD4+CD25hiFoxP3+ T Cells, Week 12CD4+CD25hiFoxP3+ T Cells, Month 6CD4+CD25hiFoxP3+ T Cells, Month 12
Otelixizumab53.449.740.1101.3101.9101.7106.6107.1102.785.5133.3106.960.843.535.9109.798.8108.7110.7120.3107.090.9153.2160.6
Placebo118.894.4113.2114.5114.695.1104.1110.1109.9102.5137.9122.0151.993.8110.7118.4110.0101.1106.9131.797.7113.6150.9166.4

[back to top]

Change From Baseline in 2-hour Mixed Meal Stimulated C-peptide Area Under Curve [AUC] (Normalized for 120-minute Time Interval) at Month 12

Mixed meal-stimulated C-peptide AUC was the area under the C-peptide/time curve from Time 0 to 120 minutes, calculated using the trapezoidal rule. This reported AUC was normalized for time interval by dividing it by 120 minutes. This normalized AUC was calculated for each participant at Baseline, Week 12, and at Months 6, 12, 18, and 24. Data has been presented for meal stimulated C-peptide Area under assessment performed at Month 12. Baseline assessments were carried out on the morning of Day 1, before the start of the first infusion of study drug. Change from Baseline was calculated by subtracting the Baseline value from the post-randomization value at Month 12. (NCT00678886)
Timeframe: Baseline (0-120 minutes on Day 1) and Month 12 (0-120 minutes)

Interventionnanomoles per liter (Least Squares Mean)
Placebo-0.21
Otelixizumab-0.21

[back to top]

Change From Baseline in Stimulated C-Peptide Mean AUC at Week 12, Month 6, 12 and 18

C-peptide is a protein that shows how much insulin the body is producing. For 3 days before the mixed meal tolerance test participants were asked to eat a balanced diet. The test was performed only when the finger-stick blood glucose level was above 70 mg/dL and no higher than 200 mg/dL. Mixed meal-stimulated C-peptide AUC was the area under the C-peptide/time curve from time 0 to 120-minutes, calculated using the trapezoidal rule. This AUC was normalized for time interval by dividing it by 120-minutes (the number of minutes over which it was determined), and was adjusted by inclusion of Baseline C-peptide AUC as a covariate in the analysis. Baseline was defined at Day 1. Change from Baseline was calculated by subtracting the post Baseline value from the Baseline value. (NCT01123083)
Timeframe: Baseline (Day 1) and Week 12, Month 6, 12, 18

,
Interventionnanomoles per liter (Mean)
Week 12Month 6Month 12Month 18
Otelixizumab-0.0472-0.0945-0.23310.0502
Placebo0.0519-0.0455-0.1236-0.4125

[back to top]

Number of Participants With Responder Status

A participant was considered a responder when, at the given time point, the participant had: glycosylated hemoglobin (HbA1c) less than or equal to 6.5 percent and mean daily insulin use less than 0.5 international unit per kilogram per day (IU/kg/day) over 7 consecutive days during the 2 weeks preceding the visit. (NCT01123083)
Timeframe: Month 3, 6 and 12

,
InterventionParticipants (Count of Participants)
Week 12Month 6Month 12
Otelixizumab422623
Placebo191110

[back to top]

Composite Rank Sum: HbA1c and Exogenous Insulin Use at 6 and 12 Months

O'Brien mean rank analyses was performed on a two-part composite of the Baseline-adjusted HbA1c level and the Baseline-adjusted mean total daily insulin use per kg body weight in the otelixizumab group compared with the placebo group at Months 6 and 12. Baseline adjustment was used to reduce the potential impact of imbalances in Baseline levels between treatment groups on the treatment comparisons at later time points. For the O'Brien mean rank analysis at a particular time point, adjusted HbA1c values (for both treatment groups together) and adjusted mean daily insulin use values was ranked from smallest to largest. For each participant, the HbA1c and insulin use ranks were added together, producing a composite rank. A treatment comparison test was then performed on the composite ranks. If otelixizumab treatment was effective on this composite endpoint, then the mean of the ranks sum in the otelixizumab group was smaller than the mean of the ranks sum in the placebo group. (NCT01123083)
Timeframe: Month 6 and 12

,
InterventionRank (Mean)
Month 6Month 12
Otelixizumab110110
Placebo11996

[back to top]

Composite Rank Sum: C-Peptide AUC, HbA1c and Exogenous Insulin Use at 6 and 12 Months

O'Brien analyses was performed on a three-part composite of HbA1c level, C-peptide AUC, and mean daily insulin use in the otelixizumab group compared with the placebo group at Months 6 and 12. The three variables was adjusted for Baseline values. Baseline adjustment was used to reduce the potential impact of imbalances in Baseline levels between treatment groups on the treatment comparisons at later time points. HbA1c and insulin use was ranked from smallest to largest, and C-peptide AUC was ranked from largest to smallest. If otelixizumab treatment was effective on the composite endpoint, then the mean of the ranks sum in the otelixizumab group was smaller than the mean of the ranks sum in the placebo group. (NCT01123083)
Timeframe: Month 6 and 12

,
InterventionRank (Mean)
Month 6Month 12
Otelixizumab186170
Placebo206180

[back to top]

Change From Baseline in Mean Daily Insulin Use Over 7 Consecutive Days During the 2 Weeks Prior to the Assessment

Mean daily insulin use over 7 consecutive days during the 2 weeks preceding each key visit was calculated as the mean of the values of amount of insulin used per day on each of the 7 consecutive days. Baseline was defined at Day 1. Change from Baseline was calculated by subtracting the post Baseline value from the Baseline value. (NCT01123083)
Timeframe: Baseline (Day 1) and Month 3, 6, 12

,
InterventionInternational unit per kilogram (IU/kg) (Least Squares Mean)
Week 12Month 6Month 12
Otelixizumab-0.08-0.040.05
Placebo-0.040.010.01

[back to top]

Change From Baseline in HbA1c Levels Over 7 Consecutive Days During the 2 Weeks Prior to the Assessment

HbA1c level was recorded at Baseline, Month 3, 6 and 12. Baseline was defined at Day 1. Change from Baseline was calculated by subtracting the post Baseline value from the Baseline value. (NCT01123083)
Timeframe: Baseline (Day 1) and Month 3, 6, 12

,
InterventionPercentage of HbA1c (Least Squares Mean)
Week 12Month 6Month 12
Otelixizumab-1.01-0.65-0.47
Placebo-0.82-0.65-0.61

[back to top]

Change From Baseline in 2 Hour Mixed Meal-stimulated C-peptide AUC (Normalized for 120-minute Time Interval) at Week 12 and 6 Months

C-peptide is a protein that shows how much insulin the body is producing. For 3 days before the mixed meal tolerance test participants were asked to eat a balanced diet. The test was performed only when the finger-stick blood glucose level was above 70 mg/dL and no higher than 200 mg/dL. Mixed meal-stimulated C-peptide AUC was the area under the C-peptide/time curve from time 0 to 120-minutes, calculated using the trapezoidal rule. This AUC was normalized for time interval by dividing it by 120-minutes (the number of minutes over which it was determined), and was adjusted by inclusion of Baseline C-peptide AUC as a covariate in the analysis. Baseline was defined at Day 1. Change from Baseline was calculated by subtracting the post Baseline value from the Baseline value. (NCT01123083)
Timeframe: Baseline (Day 1) and Week 12, Month 6

,
Interventionnanomoles per liter (Least Squares Mean)
Week 12Month 6
Otelixizumab-0.05-0.10
Placebo0.05-0.04

[back to top]

Average Number of Severe Hypoglycemic Events and Documented Symptomatic Hypoglycemic Events From Baseline to Month 12

Severe hypoglycemia was considered as an event that required assistance of another person to actively administer carbohydrate, glucagon, or other resuscitative actions. These episodes may be associated with sufficient neuroglycopenia to induce seizure or coma. Plasma glucose measurements may not be available during such an event, but neurological recovery attributable to the restoration of plasma glucose to normal is considered sufficient evidence that the event was induced by a low plasma glucose concentration. Documented symptomatic hypoglycemia was considered as an event during which typical symptoms of hypoglycemia are accompanied by a measured plasma glucose concentration less than or equal to 70 mg/dL and it was collected in (1) eCRF using two forms - for single event and for mutiple events, and (2) the IVRS system. The numbers of participant-reported hypoglycemic events per participant of severe hypoglycemia and documented symptomatic hypoglycemia have been reported. (NCT01123083)
Timeframe: Baseline (Day 1) and Month 12

,
InterventionEvents (Number)
Severe hypoglycemiaDocumented symptomatic hypoglycemia from IVRSDocumented symptomatic hypoglycemia from eCRF
Otelixizumab116901300
Placebo33429822

[back to top]

Change From Baseline in 2 Hour Mixed Meal-stimulated C-peptide Area Under Curve (AUC) (Normalized for 120-minute Time Interval) at Month 12

C-peptide is a protein that shows how much insulin the body is producing. For 3 days before the mixed meal tolerance test participants were asked to eat a balanced diet. The test was performed only when the finger-stick blood glucose level was above 70 mg per deciliter (mg/dL) and no higher than 200 mg/dL. Mixed meal-stimulated C-peptide AUC was the area under the C-peptide/time curve from time 0 to 120-minutes, calculated using the trapezoidal rule. This AUC was normalized for time interval by dividing it by 120-minutes (the number of minutes over which it was determined), and was adjusted by inclusion of Baseline C-peptide AUC as a covariate in the analysis. Baseline was defined at Day 1. Change from Baseline was calculated by subtracting the value at Month 12 from the Baseline value. (NCT01123083)
Timeframe: Baseline (Day 1) and Month 12

Interventionnanomoles per liter (Least Squares Mean)
Placebo-0.14
Otelixizumab-0.23

[back to top]

Percentage of Participants With Change From Baseline in Severe Hypoglycemic Events and Documented Symptomatic Hypoglycemic Events at Month 12

Severe hypoglycemia was considered as an event that required assistance of another person to actively administer carbohydrate, glucagon, or other resuscitative actions. These episodes may be associated with sufficient neuroglycopenia to induce seizure or coma. Plasma glucose measurements may not be available during such an event, but neurological recovery attributable to the restoration of plasma glucose to normal is considered sufficient evidence that the event was induced by a low plasma glucose concentration. Documented symptomatic hypoglycemia was considered as an event during which typical symptoms of hypoglycemia are accompanied by a measured plasma glucose concentration less than or equal to 70 mg/dL and it was collected in (1) eCRF using two forms - for single event and for mutiple events, and (2) the IVRS system. The percentage of participant with incidence of severe hypoglycemia and documented symptomatic hypoglycemia have been reported. (NCT01123083)
Timeframe: Baseline (Day 1) and Month 12

,
InterventionPercentage of participants (Number)
Severe hypoglycemiaDocumented symptomatic hypoglycemia from IVRSDocumented symptomatic hypoglycemia from eCRF
Otelixizumab5.928.843.2
Placebo11.536.145.9

[back to top]

Number of Participants With Anti-Drug Antibodies

Serum samples were obtained to measure anti-drug antibodies during the study. (NCT01287195)
Timeframe: From baseline to Week 10

InterventionParticipants (Count of Participants)
Oral OKT30

[back to top]

Cytokine Production by PBMCs in Cell Culture

Cytokine production was assessed in cell cultures of PBMCs obtained from participants at baseline, Weeks 1, 3 and 5. The following cytokines were detected and are reported here: interferon gamma (IFN-gamma), interleukin (IL)-17A, IL-6, IL-1 beta, tumor necrosis factor (TNF) and IL-10. (NCT01287195)
Timeframe: Baseline, Weeks 1, 3 and 5

Interventionpicograms/milliliter (pg/mL) (Mean)
IFN gamma, BaselineIFN gamma, Week 1IFN gamma, Week 3IFN gamma, Week 5IL-17A, BaselineIL-17A, Week 1IL-17A, Week 3IL-17A, Week 5IL-6, BaselineIL-6, Week 1IL-6, Week 3IL-6, Week 5IL-1 beta, BaselineIL-1 beta, Week 1IL-1 beta, Week 3IL-1 beta, Week 5TNF, BaselineTNF, Week 1TNF, Week 3TNF, Week 5IL-10, BaselineIL-10, Week 1IL-10, Week 3IL-10, Week 5
Oral OKT34935115211017828326911418174147026781978567110155185383615857151956311512015

[back to top]

Mayo Score

The Mayo Score is determined by the investigator by assigning a score to the following four assessments: stool frequency, rectal bleeding, physician's global assessment and endoscopy. Total range for Mayo score is 0-12 with a higher score indicating a worse outcome. (NCT01287195)
Timeframe: Baseline, Week 5

Interventionscore on a scale (Mean)
BaselineWeek 5
Oral OKT38.87.5

[back to top]

Percentage of Biomarker-positive Immune Cells

Peripheral blood mononuclear cells were (PBMCs) were isolated from blood samples taken from each participant at baseline and Week 5. PBMCs were stained with a panel of fluorochrome-conjugated antibodies against multiple surface and intracellular biomarkers, including Cluster of Differentiation (CD) 3, CD4, CD8, Forkhead box P3 protein (FOXP3) and latency-associated peptide (LAP). The percentage of T cells positive for each of these biomarkers was determined by fluorescence activated cell sorting (FACS) and the mean percentage of positive T cells for each biomarker for all analyzed participants is reported. (NCT01287195)
Timeframe: Baseline, Week 5

Interventionpercentage of biomarker-positive T cells (Mean)
CD3, BaselineCD3, Week 5CD4, BaselineCD4, Week 5CD8, BaselineCD8, Week 5FOXP3, BaselineFOXP3, Week 5LAP, BaselineLAP, Week 5
Oral OKT3383059753124540.90.5

[back to top]

Score in Histologic Evaluation of Flexible Sigmoidoscopy

Mucosal biopsies were obtained from the most inflamed area seen during flexible sigmoidoscopy and blindly scored by a single pathologist with scores ranging 0-3 with a higher score indicating a worse outcome. (NCT01287195)
Timeframe: Baseline, Week 5

Interventionscore on a scale (Mean)
BaselineWeek 5
Oral OKT32.21.75

[back to top]

Simple Clinical Colitis Activity Index (SCCAI) Score

SCCAI is a symptom based questionnaire addressing five assessments, including bowel frequency day, bowel frequency night, urgency of defecation, blood in stool and general well-being. Score ranges from 0-15 with one additional point added for each manifestation of extracolonic features. A higher score indicates a worse outcome. (NCT01287195)
Timeframe: Baseline, Week 5

Interventionscore on a scale (Mean)
BaselineWeek 5
Oral OKT37.76

[back to top]

T Cell Proliferation of PBMCs in Cell Culture

PBMCs isolated from whole blood obtained from participants at baseline, Weeks 1, 3, and 5 were assessed for proliferation in cell culture using a radioactive thymidine incorporation assay. A higher number indicates a higher level of proliferation. (NCT01287195)
Timeframe: Baseline, Weeks 1, 3 and 5

Interventionradioactive counts of cells per minute (Mean)
BaselineWeek 1Week 3Week 5
Oral OKT327470496174657520993

[back to top]

Number of Participants With Adverse Events

An adverse event is any untoward medical occurrence in a participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with the treatment. An adverse event can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a pharmaceutical product, whether or not considered related to the pharmaceutical product. Preexisting conditions which worsen during a study are also considered as adverse events. (NCT01287195)
Timeframe: From baseline to Week 10

InterventionParticipants (Count of Participants)
Oral OKT33

[back to top]

Overall Survival (OS) of HR and IR Relapse Patients

OS rates of HR and IR relapse B-ALL patients who are randomized following Induction Block 1 chemotherapy to receive either two intensive chemotherapy blocks or two 5-week blocks of blinatumomab (HR/IR Randomization). OS is calculated as the time from randomization to date of death or date of last contact. Two-year OS estimates will be calculated from date of randomization for both Arm A and Arm B. Two-sided 95% confidence intervals will be calculated. (NCT02101853)
Timeframe: Up to 2 years from date of randomization

Interventionpercentage of participants (Number)
Arm A (HR and IR Control)58.40
Arm B (HR and IR Blinatumomab)71.33

[back to top]

Disease Free Survival (DFS) of Low Risk (LR) Relapse Patients

DFS rates of LR relapse B-ALL patients who are randomized following Block 1 chemotherapy to receive either chemotherapy alone or chemotherapy plus blinatumomab (LR Randomization). DFS is calculated as the time from randomization to date of first event (relapse, second malignancy, remission death) or date of last contact. Three-year DFS estimates will be calculated from date of randomization for both Arm C and Arm D. Two-sided 95% confidence intervals will be calculated. (NCT02101853)
Timeframe: Up to 3 years from date of randomization

Interventionpercentage of participants (Number)
Arm C (LR Control)58.94
Arm D (LR Blinatumomab)67.00

[back to top]

Disease Free Survival (DFS) of High-risk (HR) and Intermediate-risk (IR) Relapse Patients

DFS rates of HR and IR relapse B-ALL patients who are randomized following Induction Block 1 chemotherapy to receive either two intensive chemotherapy blocks or two 5-week blocks of blinatumomab (HR/IR Randomization). DFS is calculated as the time from randomization to date of first event (treatment failure, relapse, second malignancy, remission death) or date of last contact. Two-year DFS estimates will be calculated from date of randomization for both Arm A and Arm B. Two-sided 95% confidence intervals will be calculated. (NCT02101853)
Timeframe: Up to 2 years from date of randomization

Interventionpercentage of participants (Number)
Arm A (HR and IR Control)39.04
Arm B (HR and IR Blinatumomab)54.44

[back to top]

Overall Survival (OS) of LR Relapse Patients

OS rates of LR relapse B-ALL patients who are randomized following Block 1 chemotherapy to receive either chemotherapy alone or chemotherapy plus blinatumomab (LR Randomization). OS is calculated as the time from randomization to date of death or date of last contact. Three-year OS estimates will be calculated from date of randomization for both Arm C and Arm D. Two-sided 95% confidence intervals will be calculated. (NCT02101853)
Timeframe: Up to 3 years from date of randomization

Interventionpercentage of participants (Number)
Arm C (LR Control)88.29
Arm D (LR Blinatumomab)90.37

[back to top] [back to top]

Incidence of Dose-limiting Toxicity (Cohort II)

Defined as any grade 4 or higher treatment-related, non-hematologic toxicity in the first cycle of post-remission therapy (blinatumomab/dasatinib) graded by National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Only participants with Ph-positive ALL or Ph-like DSMKF ALL were evaluated. (NCT02143414)
Timeframe: Up to day 42 of post-remission therapy

InterventionParticipants (Count of Participants)
Cohort II (Ph+/Ph-like)0

[back to top]

Minimal Residual Disease Negativity

To estimate in each cohort the rate of minimal residual disease (MRD) negativity. (NCT02143414)
Timeframe: Participants are assessed after induction treatment and again after re-induction treatment, if re-induction treatment is received (i.e. up to 85 days after registration)

,
InterventionParticipants (Count of Participants)
MRD-MRD+
Cohort I (Ph-)121
Cohort II (Ph+/Ph-like)79

[back to top]

Complete Response Rate (Cohort I)

Complete response rate is measured by the number of participants achieving complete remission (CR) or complete remission with incomplete platelet recovery (CRi) rate. CR is defined as having <5% marrow aspirate blasts, ANC >1,000/mcL, platelets > 100,000/mcL, no blasts in peripheral blood, and C1 extramedullary disease status. CRi is the same as CR but platelet count may be <= 100,000/mcL and/or ANC <=1,000/mcL. (NCT02143414)
Timeframe: Participants are assessed after induction treatment and again after re-induction treatment, if re-induction treatment is received (i.e. up to 85 days after registration)

InterventionParticipants (Count of Participants)
CR or CRiNo CR or CRi
Cohort I (Ph-)1910

[back to top]

Overall Survival Rate (Cohort I)

To evaluate the 3-year overall survival rate in elderly participants with newly diagnosed Ph-negative ALL treated with blinatumomab followed by POMP maintenance. Overall (NCT02143414)
Timeframe: From the day of registration on study until death from any cause, assessed at 3 years

Interventionpercentage of participants (Number)
Cohort I (Ph-)34

[back to top]

Disease-free Survival (Cohort II)

An estimate of disease free survival in Ph-positive ALL and Ph-like DSMKF ALL (Cohort II). Disease free survival is measured by the number of years between the date the patient first achieves complete remission (CR) or complete remission with incomplete platelet recovery (CRi) until relapse from CR/CRi or death from any cause. CR is defined as having <5% marrow aspirate blasts, ANC >1,000/mcL, platelets > 100,000/mcL, no blasts in peripheral blood, and C1 extramedullary disease status. CRi is the same as CR but platelet count may be <= 100,000/mcL and/or ANC <=1,000/mcL. (NCT02143414)
Timeframe: Duration of treatment and follow up until death or date of primary analysis (about 7.5 years)

Interventionyears (Median)
Cohort II (Ph+/Ph-like)5.3

[back to top]

Duration of Response

"Response date to loss of response or last follow up. Complete Remission (CR): Normalization of the peripheral blood and bone marrow with 5% or less blasts in normocellular or hypercellular marrow with a granulocyte count of 1 x 10^9/L or above, and platelet count of 100 x 10^9/L. Complete resolution of all sites of extramedullary disease is required for CR.~Complete remission without recovery of counts (CRi): Peripheral blood and marrow results as for CR, but with incomplete recover of counts (platelets < 100 x 10^9/L; neutrophils < 1 x 10^9/L).~Partial Response (PR): As above for CR except for the presence of 6-25% marrow blasts." (NCT03518112)
Timeframe: Time from the first day of treatment assessed up to 3 years, 1 month

InterventionMonths (Median)
Treatment (Blinatumomab, Combination Chemotherapy)4.4

[back to top]

Participants With a Response

"defined as the percentage of patients achieving complete response (CR) or CR with inadequate count recovery (CRi). Complete Remission (CR): Normalization of the peripheral blood and bone marrow with 5% or less blasts in normocellular or hypercellular marrow with a granulocyte count of 1 x 10^9/L or above, and platelet count of 100 x 10^9/L. Complete resolution of all sites of extramedullary disease is required for CR.~Complete remission without recovery of counts (CRi): Peripheral blood and marrow results as for CR, but with incomplete recover of counts (platelets < 100 x 10^9/L; neutrophils < 1 x 10^9/L)." (NCT03518112)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
Treatment (Blinatumomab, Combination Chemotherapy)4

[back to top]

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT03518112)
Timeframe: Time from the first day of treatment assessed up to 3 years, 1 month

InterventionMonths (Median)
Treatment (Blinatumomab, Combination Chemotherapy)16.7

[back to top]

Number of Participants Negative for Minimal Residual Disease (MRD)

Minimal Residual Disease (MRD) was assessed by flow cytometry. MRD negativity: Absence of detectable leukemia using multiparameter flow cytometry with a sensitivity of NCT03518112)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
Treatment (Blinatumomab, Combination Chemotherapy)4

[back to top]

Event Free Survival (EFS) Where Events Defined as no Response, Loss of Response, or Death

"Time from date of treatment start until the date of first objective documentation of disease-relapse. Relapse and resistant disease will be defined based on morphological assessment of bone marrow and peripheral blood.~Complete Remission (CR): Normalization of the peripheral blood and bone marrow with 5% or less blasts in normocellular or hypercellular marrow with a granulocyte count of 1 x 10^9/L or above, and platelet count of 100 x 10^9/L. Complete resolution of all sites of extramedullary disease is required for CR.~Complete remission without recovery of counts (CRi): Peripheral blood and marrow results as for CR, but with incomplete recover of counts (platelets < 100 x 10^9/L; neutrophils < 1 x 10^9/L).~Partial Response (PR): As above for CR except for the presence of 6-25% marrow blasts." (NCT03518112)
Timeframe: Time from the first day of treatment assessed up to 3 years, 1 month

InterventionMonths (Median)
Treatment (Blinatumomab, Combination Chemotherapy)5.7

[back to top]