Page last updated: 2024-11-12

pevonedistat

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

Description

pevonedistat: a potent and selective inhibitor of NAE (NEDD8-activating enzyme) [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

pevonedistat : A pyrrolopyrimidine that is 7H-pyrrolo[2,3-d]pyrimidine which is substituted by a (1S)-2,3-dihydro-1H-inden-1-ylnitrilo group at position 4 and by a (1S,3S,4S)-3-hydroxy-4-[(sulfamoyloxy)methyl]cyclopentyl group at position 7. It is a potent and selective NEDD8-activating enzyme inhibitor with an IC50 of 4.7 nM, and currently under clinical investigation for the treatment of acute myeloid leukemia (AML) and myelodysplastic syndromes. [Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Cross-References

ID SourceID
PubMed CID16720766
CHEMBL ID1231160
CHEBI ID145535
CHEBI ID95028
SCHEMBL ID192875
MeSH IDM0534783

Synonyms (62)

Synonym
HY-70062
CHEMBL1231160
[(1s,2s,4r)-4-{4-[(1s)-2,3-dihydro-1h-inden-1-ylamino]-7h-pyrrolo[2,3-d]pyrimidin-7-yl}-2-hydroxycyclopentyl]methyl sulfamate
B39 ,
sulfamic acid [(1s,2s,4r)-4-[4-[[(1s)-2,3-dihydro-1h-inden-1-yl]amino]-7h-pyrrolo[2,3-d]pyrimidin-7-yl]-2-hydroxycyclopentyl]methyl ester
mln-4924
CHEBI:145535
pevonedistat
mln4924
pevonedistatum
mln 4924
905579-51-3
[(1s,2s,4r)-4-[4-[[(1s)-2,3-dihydro-1h-inden-1-yl]amino]pyrrolo[2,3-d]pyrimidin-7-yl]-2-hydroxy-cyclopentyl]methyl sulfamate
bdbm50285607
mln-4924003
sulfamic acid, ((1s,2s,4r)-4-(4-(((1s)-2,3-dihydro-1h-inden-1-yl)amino)-7h-pyrrolo(2,3-d)pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl ester
pevonedistat [usan:inn]
unii-s3azd8d215
s3azd8d215 ,
((1s,2s,4r)-4-(4-((1s)-2,3-dihydro-1h-inden-1-ylamino)-7h-pyrrolo(2,3-d)pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl sulfamate
((1s,2s,4r)-4-(4-((1s)-2,3-dihydro-1h-inden-1-ylamino)-7h-pyrrolo(2,3-d)pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl sulphamate
4-(4-((1s)-2,3-dihydro-1h-inden-1-ylamino)-7h-pyrrolo(2,3-d)pyrimidin-7-yl)-2-hydroxycyclopentyl methyl sulfamidate
pevonedistat (usan)
D10413
CS-0348
BRD-K67844266-001-01-3
MLS006010830
smr004701757
MPUQHZXIXSTTDU-QXGSTGNESA-N
((1s,2s,4r)-4-{4-[(1s)-2,3-dihydro-1h-inden-1-ylamino]-7h-pyrrolo[2,3-d]pyrimidin-7-yl}-2-hydroxycyclopentyl)methyl sulfamate
((1s,2s,4r)-4-{4-[(1s)-2,3-dihydro-1h-inden-1-ylamino]-7h-pyrrolo[2,3-d]-pyrimidin-7-yl}-2-hydroxycyclopentyl)methyl sulfamate
SCHEMBL192875
3GZN
((1s,2s,4r)-4-(4-(((1s)-2,3-dihydro-1h-inden-1-yl)amino(-7h-pyrrolo(2,3-d)pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl sulfamate
pevonedistat [who-dd]
pevonedistat [usan]
pevonedistat [inn]
((1s,2s,4r)-4-(4-(((s)-2,3-dihydro-1h-inden-1-yl)amino)-7h-pyrrolo[2,3-d]pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl sulfamate
((1s,2s,4r)-4-(4-((s)-2,3-dihydro-1h-inden-1-ylamino)-7h-pyrrolo[2,3-d]pyrimidin-7-yl)-2-hydroxycyclopentyl)methyl sulfamate
AC-27394
[(1s,2s,4r)-4-[4-[[(1s)-2,3-dihydro-1h-inden-1-yl]amino]pyrrolo[2,3-d]pyrimidin-7-yl]-2-hydroxycyclopentyl]methyl sulfamate
AKOS025401927
mln4924 (hydrochloride)
CHEBI:95028
NCGC00345794-10
NCGC00345794-07
DB11759
Q27166792
905579-51-3 (free base)
pevonedistat (mln-4924)
EX-A1472
[(1s,2s,4r)-4-{4-[(1s)-2,3-dihydro-1h-inden-1-ylamino]-7h-pyrrolo [2,3-d]pyrimidin-7-yl}-2-hydroxycyclopentyl]methyl sulfamate
nsc793435
nsc-793435
NCGC00345794-02
pevonedistat; mln4924
DTXSID701022549
gtpl12117
tak924
tak-924
mfcd17215201
Z2739897698

Research Excerpts

Overview

Pevonedistat is a first-in-class NEDD8-activating enzyme (NAE) inhibitor currently being tested in clinical trials for its antitumor potential. It blocks proteasomal degradation of cullin-RING ligase (CRL) involved in the degradation of short-lived regulatory proteins, including those involved with cell-cycle regulation.

ExcerptReferenceRelevance
"Pevonedistat (MLN4924) is a specific NEDD8-activating enzyme inhibitor that inactivates cullin-RING ligases involved in ubiquitylation and turnover of different signaling molecules. "( Pevonedistat attenuates cisplatin-induced nephrotoxicity in mice by downregulating the release of inflammatory mediators.
El-Far, YM; El-Mesery, M, 2021
)
3.51
"Pevonedistat is a first-in-class NEDD8-activating enzyme (NAE) inhibitor currently being tested in clinical trials for its antitumor potential."( Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, sensitizes cancer cells to VSVΔ51 oncolytic virotherapy.
Alwithenani, A; Arulanandam, R; Bergeron, A; Birdi, HK; Chen, A; Diallo, JS; Jirovec, A; Lorimer, I; Maznyi, G; Ng, K; Serrano, D; Spinelli, M; Taha, Z; Thomson, M; Vanderhyden, B; Wong, B, 2023
)
3.07
"Pevonedistat is a potent, selective, first-in-class NEDD8 activating enzyme inhibitor. "( Overexpression of ABCG2 confers resistance to pevonedistat, an NAE inhibitor.
Chen, ZS; Fan, YF; Li, JS; Ma, XY; Wei, LY; Wu, ZX; Yang, DH; Yang, Y; Zhao, M, 2020
)
2.26
"Pevonedistat is a neddylation inhibitor that blocks proteasomal degradation of cullin-RING ligase (CRL) proteins involved in the degradation of short-lived regulatory proteins, including those involved with cell-cycle regulation. "( The Anti-Tumor Activity of the NEDD8 Inhibitor Pevonedistat in Neuroblastoma.
Barbieri, E; Foster, JH; Horton, TM; Moreno-Smith, M; Scorsone, KA; Zage, P; Zhang, L, 2021
)
2.32
"Pevonedistat (MLN4924) is a potent and selective small molecule NEDD8-activating enzyme inhibitor."( Targeting NEDD8-activating enzyme is a new approach to treat canine diffuse large B-cell lymphoma.
Assumpção, ALFV; Lu, Z; Marlowe, KW; Pan, X; Shaffer, KS, 2018
)
1.2
"Pevonedistat (MLN4924) is a selective small molecule targeting the NEDD8-activating enzyme (NAE) and inhibits an early step in neddylation, resulting in DNA re-replication, cell cycle arrest and death."( Targeting the protein ubiquitination machinery in melanoma by the NEDD8-activating enzyme inhibitor pevonedistat (MLN4924).
Bagby, SM; Berger, A; Blakemore, SJ; Eckhardt, SG; Klauck, PJ; McDonald, A; Micel, LN; Pitts, TM; Selby, HM; Smith, PF; Spreafico, A; Tan, AC; Tentler, JJ; Wong, KM, 2017
)
1.39

Treatment

Pevonedistat treatment inhibits NF-B pathway activation and downregulates NF-κB target genes in canine DLBCL. Cotreatment with pevoningistat and cisplatin yielded dramatic HNSCC tumor regression and long-term animal survival in 100% of treated mice.

ExcerptReferenceRelevance
"Upon pevonedistat treatment alone or in combination with ruxolitinib, MPN mouse models exhibited reduced disease burden and improved survival."( Pevonedistat targets malignant cells in myeloproliferative neoplasms in vitro and in vivo via NFκB pathway inhibition.
Celik, H; Challen, GA; Collins, TB; De Togni, ES; Fisher, DAC; Fulbright, MC; Kong, T; Laranjeira, ABA; Oh, ST; Ruzinova, M; Wong, AJ, 2022
)
2.62
"Pevonedistat treatment inhibits NF-κB pathway activation and downregulates NF-κB target genes in canine DLBCL."( Targeting NEDD8-activating enzyme is a new approach to treat canine diffuse large B-cell lymphoma.
Assumpção, ALFV; Lu, Z; Marlowe, KW; Pan, X; Shaffer, KS, 2018
)
1.2
"Cotreatment with pevonedistat and cisplatin yielded dramatic HNSCC tumor regression and long-term animal survival in 100% of treated mice."( Targeting NEDDylation is a Novel Strategy to Attenuate Cisplatin-induced Nephrotoxicity.
Carew, JS; Chipollini, J; Espitia, CM; Jones, TM; Lee, BR; Nawrocki, ST; Wertheim, JA, 2023
)
1.24

Pharmacokinetics

ExcerptReferenceRelevance
"A population pharmacokinetic (PK) analysis was conducted to quantify the impact of patient-specific and concurrent medication factors on pevonedistat PK."( Population pharmacokinetics of pevonedistat alone or in combination with standard of care in patients with solid tumours or haematological malignancies.
Faessel, HM; Faller, DV; Mould, DR; Sedarati, F; Venkatakrishnan, K; Zhou, X, 2019
)
1

Compound-Compound Interactions

ExcerptReferenceRelevance
" In the reported study we aimed to provide evidence for this assumption and investigated the drug combination of cisplatin and the neddylation inhibitor MLN4924 in HCT-116 cells via cell biological analyses and mass spectrometry-based quantitative proteomics."( Systematic analysis of synergistic proteome modulations in a drug combination of cisplatin and MLN4924.
Abou-Eid, S; Megger, DA; Sitek, B; Zülch, B, 2018
)
0.48
"Data were pooled from 6 clinical studies consisting of 335 patients with solid tumours or haematological malignancies administered pevonedistat alone or in combination with azacitidine, docetaxel, carboplatin + paclitaxel, or gemcitabine."( Population pharmacokinetics of pevonedistat alone or in combination with standard of care in patients with solid tumours or haematological malignancies.
Faessel, HM; Faller, DV; Mould, DR; Sedarati, F; Venkatakrishnan, K; Zhou, X, 2019
)
1

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 (2)

RoleDescription
apoptosis inducerAny substance that induces the process of apoptosis (programmed cell death) in multi-celled organisms.
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
[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 (5)

ClassDescription
indanes
pyrrolopyrimidine
secondary amino compoundA compound formally derived from ammonia by replacing two hydrogen atoms by organyl groups.
cyclopentanolsAn alcohol in which one or more hydroxy groups are attached to a cyclopentane skeleton.
sulfamidateA compound where an oxygen atom and an amino group are bound to an SO2 unit.
indanes
[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]

Pathways (1)

PathwayProteinsCompounds
Orf10 Cul2 pathway (COVID-19 Disease Map)01

Protein Targets (17)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
PPM1D proteinHomo sapiens (human)Potency0.33000.00529.466132.9993AID1347411
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency3.58470.01237.983543.2770AID1645841
GVesicular stomatitis virusPotency22.61800.01238.964839.8107AID1645842
cytochrome P450 2D6Homo sapiens (human)Potency8.48660.00108.379861.1304AID1645840
Interferon betaHomo sapiens (human)Potency9.24520.00339.158239.8107AID1347411; AID1645842
HLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)Potency22.61800.01238.964839.8107AID1645842
Inositol hexakisphosphate kinase 1Homo sapiens (human)Potency22.61800.01238.964839.8107AID1645842
cytochrome P450 2C9, partialHomo sapiens (human)Potency22.61800.01238.964839.8107AID1645842
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Ubiquitin-like modifier-activating enzyme 6Homo sapiens (human)IC50 (µMol)1.40001.80001.80001.8000AID1371351; AID1872479
Ubiquitin-like modifier-activating enzyme 1 Mus musculus (house mouse)IC50 (µMol)1.50001.50001.50001.5000AID1872478
Potassium voltage-gated channel subfamily H member 2Homo sapiens (human)IC50 (µMol)40.00000.00091.901410.0000AID1894761
NEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)IC50 (µMol)0.04710.00030.04710.1800AID1728035; AID1872476; AID1894757; AID1894758
NEDD8Homo sapiens (human)IC50 (µMol)0.00470.00470.00470.0047AID1371347
NEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)IC50 (µMol)0.00330.00030.00330.0050AID1391002; AID1872476; AID1894757; AID1894758
SUMO-activating enzyme subunit 1Homo sapiens (human)IC50 (µMol)4.60002.20004.46678.2000AID1371349; AID1872477
SUMO-activating enzyme subunit 2Homo sapiens (human)IC50 (µMol)4.60008.20008.20008.2000AID1371349; AID1872477
[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)
NEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)EC50 (µMol)1.71500.03001.57673.4000AID1349085; AID1894759
NEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)EC50 (µMol)1.71500.03001.57673.4000AID1349085; AID1894759
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (112)

Processvia Protein(s)Taxonomy
ubiquitin-dependent protein catabolic processUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
learningUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
locomotory behaviorUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
protein ubiquitinationUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
amygdala developmentUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
hippocampus developmentUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
dendritic spine developmentUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
protein lipidationUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
autophagyUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
positive regulation of protein modification processUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
autophagosome assemblyUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
mitophagyUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
autophagyUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
cellular response to starvationUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
protein transportUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
macroautophagyUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
regulation of circadian rhythmUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
positive regulation of apoptotic processUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
positive regulation of protein catabolic processUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
rhythmic processUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
defense response to virusUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
cellular response to hyperoxiaUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
autophagy of mitochondrionUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
cellular response to nitrogen starvationUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
piecemeal microautophagy of the nucleusUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
C-terminal protein lipidationUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
protein modification by small protein conjugationUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell activation involved in immune responseInterferon betaHomo sapiens (human)
cell surface receptor signaling pathwayInterferon betaHomo sapiens (human)
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to virusInterferon betaHomo sapiens (human)
positive regulation of autophagyInterferon betaHomo sapiens (human)
cytokine-mediated signaling pathwayInterferon betaHomo sapiens (human)
natural killer cell activationInterferon betaHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylation of STAT proteinInterferon betaHomo sapiens (human)
cellular response to interferon-betaInterferon betaHomo sapiens (human)
B cell proliferationInterferon betaHomo sapiens (human)
negative regulation of viral genome replicationInterferon betaHomo sapiens (human)
innate immune responseInterferon betaHomo sapiens (human)
positive regulation of innate immune responseInterferon betaHomo sapiens (human)
regulation of MHC class I biosynthetic processInterferon betaHomo sapiens (human)
negative regulation of T cell differentiationInterferon betaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIInterferon betaHomo sapiens (human)
defense response to virusInterferon betaHomo sapiens (human)
type I interferon-mediated signaling pathwayInterferon betaHomo sapiens (human)
neuron cellular homeostasisInterferon betaHomo sapiens (human)
cellular response to exogenous dsRNAInterferon betaHomo sapiens (human)
cellular response to virusInterferon betaHomo sapiens (human)
negative regulation of Lewy body formationInterferon betaHomo sapiens (human)
negative regulation of T-helper 2 cell cytokine productionInterferon betaHomo sapiens (human)
positive regulation of apoptotic signaling pathwayInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell differentiationInterferon betaHomo sapiens (human)
natural killer cell activation involved in immune responseInterferon betaHomo sapiens (human)
adaptive immune responseInterferon betaHomo sapiens (human)
T cell activation involved in immune responseInterferon betaHomo sapiens (human)
humoral immune responseInterferon betaHomo sapiens (human)
positive regulation of T cell mediated cytotoxicityHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
adaptive immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
antigen processing and presentation of endogenous peptide antigen via MHC class I via ER pathway, TAP-independentHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of T cell anergyHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
defense responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
detection of bacteriumHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of interleukin-12 productionHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of interleukin-6 productionHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protection from natural killer cell mediated cytotoxicityHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
innate immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of dendritic cell differentiationHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
antigen processing and presentation of endogenous peptide antigen via MHC class IbHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by hormonePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane depolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
signal transductionNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
mitotic DNA replication checkpoint signalingNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
regulation of apoptotic processNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
regulation of neuron apoptotic processNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
protein neddylationNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
neuron apoptotic processNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
ubiquitin-dependent protein catabolic processNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
protein ubiquitinationNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
DNA damage responseNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
protein neddylationNEDD8Homo sapiens (human)
regulation of transcription by RNA polymerase IINEDD8Homo sapiens (human)
proteolysisNEDD8Homo sapiens (human)
ubiquitin-dependent protein catabolic processNEDD8Homo sapiens (human)
protein localizationNEDD8Homo sapiens (human)
anatomical structure morphogenesisNEDD8Homo sapiens (human)
protein modification processNEDD8Homo sapiens (human)
protein ubiquitinationNEDD8Homo sapiens (human)
protein neddylationNEDD8Homo sapiens (human)
regulation of proteolysisNEDD8Homo sapiens (human)
modification-dependent protein catabolic processNEDD8Homo sapiens (human)
proteolysisNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
endomitotic cell cycleNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
protein modification processNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
post-translational protein modificationNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
protein neddylationNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
regulation of cell cycleNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
inositol phosphate metabolic processInositol hexakisphosphate kinase 1Homo sapiens (human)
phosphatidylinositol phosphate biosynthetic processInositol hexakisphosphate kinase 1Homo sapiens (human)
negative regulation of cold-induced thermogenesisInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol phosphate biosynthetic processInositol hexakisphosphate kinase 1Homo sapiens (human)
protein ubiquitinationSUMO-activating enzyme subunit 1Homo sapiens (human)
protein sumoylationSUMO-activating enzyme subunit 1Homo sapiens (human)
positive regulation of protein sumoylationSUMO-activating enzyme subunit 1Homo sapiens (human)
positive regulation of protein targeting to mitochondrionSUMO-activating enzyme subunit 1Homo sapiens (human)
protein sumoylationSUMO-activating enzyme subunit 2Homo sapiens (human)
positive regulation of protein sumoylationSUMO-activating enzyme subunit 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (47)

Processvia Protein(s)Taxonomy
protein bindingUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
ATP bindingUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
FAT10 activating enzyme activityUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
thiosulfate sulfurtransferase activityUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
ubiquitin activating enzyme activityUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
nucleotidyltransferase activityUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
protein bindingUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
Atg12 activating enzyme activityUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
protein homodimerization activityUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
Atg8 activating enzyme activityUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
cytokine activityInterferon betaHomo sapiens (human)
cytokine receptor bindingInterferon betaHomo sapiens (human)
type I interferon receptor bindingInterferon betaHomo sapiens (human)
protein bindingInterferon betaHomo sapiens (human)
chloramphenicol O-acetyltransferase activityInterferon betaHomo sapiens (human)
TAP bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
signaling receptor bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protein bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
peptide antigen bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
TAP bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protein-folding chaperone bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
transcription cis-regulatory region bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ubiquitin protein ligase bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
identical protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein homodimerization activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
C3HC4-type RING finger domain bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
NEDD8 activating enzyme activityNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
protein bindingNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
ubiquitin protein ligase bindingNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
protein heterodimerization activityNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
ubiquitin activating enzyme activityNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
protein bindingNEDD8Homo sapiens (human)
ubiquitin protein ligase bindingNEDD8Homo sapiens (human)
protein tag activityNEDD8Homo sapiens (human)
NEDD8 activating enzyme activityNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
protein bindingNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
ATP bindingNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
NEDD8 activating enzyme activityNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
NEDD8 transferase activityNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
identical protein bindingNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
protein heterodimerization activityNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
inositol-1,3,4,5,6-pentakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol heptakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 5-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
protein bindingInositol hexakisphosphate kinase 1Homo sapiens (human)
ATP bindingInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 1-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 3-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol 5-diphosphate pentakisphosphate 5-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol diphosphate tetrakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
SUMO activating enzyme activitySUMO-activating enzyme subunit 1Homo sapiens (human)
ubiquitin activating enzyme activitySUMO-activating enzyme subunit 1Homo sapiens (human)
protein bindingSUMO-activating enzyme subunit 1Homo sapiens (human)
enzyme activator activitySUMO-activating enzyme subunit 1Homo sapiens (human)
ATP-dependent protein bindingSUMO-activating enzyme subunit 1Homo sapiens (human)
small protein activating enzyme bindingSUMO-activating enzyme subunit 1Homo sapiens (human)
protein heterodimerization activitySUMO-activating enzyme subunit 1Homo sapiens (human)
SUMO activating enzyme activitySUMO-activating enzyme subunit 2Homo sapiens (human)
magnesium ion bindingSUMO-activating enzyme subunit 2Homo sapiens (human)
protein bindingSUMO-activating enzyme subunit 2Homo sapiens (human)
ATP bindingSUMO-activating enzyme subunit 2Homo sapiens (human)
transferase activitySUMO-activating enzyme subunit 2Homo sapiens (human)
SUMO activating enzyme activitySUMO-activating enzyme subunit 2Homo sapiens (human)
SUMO bindingSUMO-activating enzyme subunit 2Homo sapiens (human)
small protein activating enzyme bindingSUMO-activating enzyme subunit 2Homo sapiens (human)
ubiquitin-like protein conjugating enzyme bindingSUMO-activating enzyme subunit 2Homo sapiens (human)
protein heterodimerization activitySUMO-activating enzyme subunit 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (31)

Processvia Protein(s)Taxonomy
cytoplasmUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
cytosolUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
cytoplasmUbiquitin-like modifier-activating enzyme 6Homo sapiens (human)
phagophore assembly siteUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
extracellular regionUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
cytoplasmUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
cytosolUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
axonemeUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
secretory granule lumenUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
ficolin-1-rich granule lumenUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
cytoplasmUbiquitin-like modifier-activating enzyme ATG7Homo sapiens (human)
extracellular spaceInterferon betaHomo sapiens (human)
extracellular regionInterferon betaHomo sapiens (human)
Golgi membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
endoplasmic reticulumHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
Golgi apparatusHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
plasma membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
cell surfaceHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
ER to Golgi transport vesicle membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
secretory granule membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
phagocytic vesicle membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
early endosome membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
recycling endosome membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular exosomeHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
lumenal side of endoplasmic reticulum membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
MHC class I protein complexHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular spaceHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
external side of plasma membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
perinuclear region of cytoplasmPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cytosolNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
plasma membraneNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
protein-containing complexNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
cytoplasmNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
nucleusNEDD8-activating enzyme E1 regulatory subunitHomo sapiens (human)
nucleusNEDD8Homo sapiens (human)
nucleoplasmNEDD8Homo sapiens (human)
cytosolNEDD8Homo sapiens (human)
extracellular exosomeNEDD8Homo sapiens (human)
nucleusNEDD8Homo sapiens (human)
cytoplasmNEDD8Homo sapiens (human)
nucleusNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
cytosolNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
protein-containing complexNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
cytoplasmNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
nucleusNEDD8-activating enzyme E1 catalytic subunitHomo sapiens (human)
fibrillar centerInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleoplasmInositol hexakisphosphate kinase 1Homo sapiens (human)
cytosolInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleusInositol hexakisphosphate kinase 1Homo sapiens (human)
cytoplasmInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleusSUMO-activating enzyme subunit 1Homo sapiens (human)
nucleoplasmSUMO-activating enzyme subunit 1Homo sapiens (human)
SUMO activating enzyme complexSUMO-activating enzyme subunit 1Homo sapiens (human)
cytoplasmSUMO-activating enzyme subunit 1Homo sapiens (human)
nucleoplasmSUMO-activating enzyme subunit 2Homo sapiens (human)
SUMO activating enzyme complexSUMO-activating enzyme subunit 2Homo sapiens (human)
cytoplasmSUMO-activating enzyme subunit 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (109)

Assay IDTitleYearJournalArticle
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.
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.
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.
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.
AID1347412qHTS assay to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: Counter screen cell viability and HiBit confirmation2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1347411qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Mechanism Interrogation Plate v5.0 (MIPE) Libary2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1745854NCATS anti-infectives library activity on HEK293 viability as a counter-qHTS vs the C. elegans viability qHTS2023Disease models & mechanisms, 03-01, Volume: 16, Issue:3
In vivo quantitative high-throughput screening for drug discovery and comparative toxicology.
AID1519989Inhibition of Nedd8-cullin1 (unknown origin) adduct formation assessed as decrease in cullin1 neddylation at 10 uM preincubated for 10 mins followed by ATP addition and measured after 30 mins by immunoblot analysis2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1519986Antitumor activity against human A549 cells xenografted in nude mouse assessed as decrease in tumor weight at 60 mg/kg, ip qd measured after 22 days2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1758804Inhibition of DCN-1 in human NCI-H2170 cells assessed as increase in NRF2 expression at 5 to 10 uM incubated for 24 hrs by Western blot analysis2021European journal of medicinal chemistry, May-05, Volume: 217Development of phenyltriazole thiol-based derivatives as highly potent inhibitors of DCN1-UBC12 interaction.
AID1872476Inhibition of NAE1/UBA3 in human HCT-116 cells assessed as reduction in Ubc12-NEDD8 level incubated for 24 hrs by immunoblot analysis2022European journal of medicinal chemistry, Mar-05, Volume: 231NAE modulators: A potential therapy for gastric carcinoma.
AID1599611Inhibition of DCN1-UBE2M protein-protein interaction in human PC9 cells assessed as increase in NRF2 accumulation at 3 uM incubated for 24 hrs by Western blot analysis2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
Potent 5-Cyano-6-phenyl-pyrimidin-Based Derivatives Targeting DCN1-UBE2M Interaction.
AID1817932Antiproliferative activity against human HepG2 cells assessed as inhibition of cell proliferation measured after 72 hrs by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Discovery of a cinnamyl piperidine derivative as new neddylation inhibitor for gastric cancer treatment.
AID1894759Inhibition of NAE in human HCT-116 cells incubated for 1 hr by Western blot analysis2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1894758Inhibition of NAE (unknown origin)2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1371347Inhibition of NEDD8 (unknown origin) assessed as decrease in formation of E2-UBL thioester reaction product2018Journal of medicinal chemistry, 04-26, Volume: 61, Issue:8
Interrogating the Roles of Post-Translational Modifications of Non-Histone Proteins.
AID1894785Antiproliferative activity against human MV4-11 cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1817934Antiproliferative activity against human HGC-27 cells assessed as inhibition of cell proliferation measured after 72 hrs by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Discovery of a cinnamyl piperidine derivative as new neddylation inhibitor for gastric cancer treatment.
AID1599608Inhibition of DCN1-UBE2M protein-protein interaction in human NCI-H1975 cells assessed as increase in NRF2 accumulation at 3 uM incubated for 24 hrs by Western blot analysis2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
Potent 5-Cyano-6-phenyl-pyrimidin-Based Derivatives Targeting DCN1-UBE2M Interaction.
AID1894794Antiproliferative activity against human CAPAN-1 cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1894802Antitumor activity against human HCT-116 cells xenografted in nude mouse assessed as reduction in tumor growth at 60 mg/kg, sc bid administered at 5 on/5 off/8 for 18 days2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1728038Cytotoxicity against human 16H6E cells incubated for 48 hrs by CCK-8 assay2021European journal of medicinal chemistry, Jan-15, Volume: 210Development of novel benzimidazole-derived neddylation inhibitors for suppressing tumor growth invitro and invivo.
AID1882258Inhibition of proteasome (unknown origin)2022European journal of medicinal chemistry, Feb-05, Volume: 229A review on the treatment of multiple myeloma with small molecular agents in the past five years.
AID656451Inhibition of human recombinant UAE assessed as inhibition of UbcH6-Ub complex formation at 0.001 to 10 uM after 1.5 hrs by Western blot analysis2011ACS medicinal chemistry letters, Aug-11, Volume: 2, Issue:8
Identification of NAE Inhibitors Exhibiting Potent Activity in Leukemia Cells: Exploring the Structural Determinants of NAE Specificity.
AID1894788Antiproliferative activity against human HCT-116 cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1894789Antiproliferative activity against human HuTu80 cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1519934Antiproliferative activity against human H1299 cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1894765Clearance in Sprague-Dawley rat at 1 mg/kg, iv2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1894761Inhibition of hERG channel2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1519926Antiproliferative activity against human T47D cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1391002Inhibition of human recombinant His-tagged NAE1 expressed in baculovirus expression system after 90 mins in presence of FLAG-tagged NEDD8 by TR-FRET assay2018Journal of medicinal chemistry, 04-12, Volume: 61, Issue:7
Piperidinyl Ureas Chemically Control Defective in Cullin Neddylation 1 (DCN1)-Mediated Cullin Neddylation.
AID1519938Antiproliferative activity against human MCF7 cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1349112Induction of apoptosis in human Caco2 cells assessed as late apoptotic cells at 10 uM after 16 hrs by Annexin V-FITC/propidium iodide staining-based flow cytometric analysis (Rvb = 0.072%)2018European journal of medicinal chemistry, Jan-01, Volume: 143Structure-based identification of a NEDD8-activating enzyme inhibitor via drug repurposing.
AID1599613Inhibition of DCN1-UBE2M protein-protein interaction in human PC9 cells assessed as increase in HO-1 accumulation at 3 uM incubated for 24 hrs by Western blot analysis2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
Potent 5-Cyano-6-phenyl-pyrimidin-Based Derivatives Targeting DCN1-UBE2M Interaction.
AID1519937Antiproliferative activity against human T47D cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1519929Antiproliferative activity against human MGC803 cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1478769Antitumor activity against human HCT116 cells after 72 hrs by MTT assay2017European journal of medicinal chemistry, Jun-16, Volume: 133Discovery of benzothiazole derivatives as novel non-sulfamide NEDD8 activating enzyme inhibitors by target-based virtual screening.
AID1371350Inhibition of ATG7 (unknown origin) assessed as decrease in formation of E2-UBL thioester reaction product2018Journal of medicinal chemistry, 04-26, Volume: 61, Issue:8
Interrogating the Roles of Post-Translational Modifications of Non-Histone Proteins.
AID1728037Antiproliferative activity against human H1299 cells incubated for 48 hrs by CCK-8 assay2021European journal of medicinal chemistry, Jan-15, Volume: 210Development of novel benzimidazole-derived neddylation inhibitors for suppressing tumor growth invitro and invivo.
AID1478770Antitumor activity against human U2OS cells after 72 hrs by MTT assay2017European journal of medicinal chemistry, Jun-16, Volume: 133Discovery of benzothiazole derivatives as novel non-sulfamide NEDD8 activating enzyme inhibitors by target-based virtual screening.
AID1894768Inhibition of human partial length wild type TAOK1 at 1 uM by Kinomescan method relative to control2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1728033Inhibition of NAE (unknown origin)-mediated neddylation assessed as suppression of cullin1-Nedd8 adduct formation at 25 uM preincubated for 10 mins followed by ATP addition and measured after 30 mins by immunoblot analysis relative to control2021European journal of medicinal chemistry, Jan-15, Volume: 210Development of novel benzimidazole-derived neddylation inhibitors for suppressing tumor growth invitro and invivo.
AID1894770Inhibition of UAE (unknown origin) mediated neddylation assessed as decrease in UbcH10-Ub adduct formation incubated for 2 hrs in presence of L-glutathione and ATP by HTRF assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1758794Inhibition of NAE1 (unknown origin)-mediated Nedd8-Ubcl2 adduct formation at 10 uM incubated for 10 mins followed by Mg-ATP addition and measured after 60 mins by Western blot analysis2021European journal of medicinal chemistry, May-05, Volume: 217Development of phenyltriazole thiol-based derivatives as highly potent inhibitors of DCN1-UBC12 interaction.
AID1599610Inhibition of DCN1-UBE2M protein-protein interaction in human NCI-H1975 cells assessed as increase in HO-1 accumulation at 3 uM incubated for 24 hrs by Western blot analysis2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
Potent 5-Cyano-6-phenyl-pyrimidin-Based Derivatives Targeting DCN1-UBE2M Interaction.
AID1519935Antiproliferative activity against human HepG2 cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1817931Antiproliferative activity against human MCF7 cells assessed as inhibition of cell proliferation measured after 72 hrs by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Discovery of a cinnamyl piperidine derivative as new neddylation inhibitor for gastric cancer treatment.
AID1728039Cytotoxicity against human BEAS-2B cells incubated for 48 hrs by CCK-8 assay2021European journal of medicinal chemistry, Jan-15, Volume: 210Development of novel benzimidazole-derived neddylation inhibitors for suppressing tumor growth invitro and invivo.
AID1872480Inhibition of His-tagged ATG7 (unknown origin) expressed in sf9 insect cells2022European journal of medicinal chemistry, Mar-05, Volume: 231NAE modulators: A potential therapy for gastric carcinoma.
AID1519933Antiproliferative activity against human EKVX cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID656452Inhibition of human recombinant SAE1/2 assessed as inhibition of SUMO1-UbcH9 complex formation at 0.001 to 10 uM after 1.5 hrs by Western blot analysis2011ACS medicinal chemistry letters, Aug-11, Volume: 2, Issue:8
Identification of NAE Inhibitors Exhibiting Potent Activity in Leukemia Cells: Exploring the Structural Determinants of NAE Specificity.
AID1817929Antiproliferative activity against human MGC-803 cells assessed as inhibition of cell proliferation measured after 72 hrs by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Discovery of a cinnamyl piperidine derivative as new neddylation inhibitor for gastric cancer treatment.
AID1519995Toxicity in nude mouse xenografted with human A549 cells assessed as effect on body weight at 60 mg/kg, ip qd measured every 2 days2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1349085Inhibition of NAE-mediated Ubcl2-NEDD8 conjugation in human Caco2 cells after 16 hrs by Western blot analysis2018European journal of medicinal chemistry, Jan-01, Volume: 143Structure-based identification of a NEDD8-activating enzyme inhibitor via drug repurposing.
AID1894792Antiproliferative activity against human LNCaP-Clone-FGC cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1371349Inhibition of SAE (unknown origin) assessed as decrease in formation of E2-UBL thioester reaction product2018Journal of medicinal chemistry, 04-26, Volume: 61, Issue:8
Interrogating the Roles of Post-Translational Modifications of Non-Histone Proteins.
AID1349113Induction of apoptosis in human Caco2 cells assessed as necrotic cells at 10 uM after 16 hrs by Annexin V-FITC/propidium iodide staining-based flow cytometric analysis (Rvb = 0.12%)2018European journal of medicinal chemistry, Jan-01, Volume: 143Structure-based identification of a NEDD8-activating enzyme inhibitor via drug repurposing.
AID1817933Cytotoxicity against human GES1 cells assessed as inhibition of cell proliferation measured after 72 hrs by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Discovery of a cinnamyl piperidine derivative as new neddylation inhibitor for gastric cancer treatment.
AID1758841Inhibition of DCN-1 in human KYSE-70 cell line assessed as increase in NRF2 expression at 5 to 10 uM incubated for 24 hrs by Western blot analysis2021European journal of medicinal chemistry, May-05, Volume: 217Development of phenyltriazole thiol-based derivatives as highly potent inhibitors of DCN1-UBC12 interaction.
AID656453Inhibition of human recombinant UBA5 assessed as inhibition of UCF1-UFM1 complex formation at 10 uM after 1.5 hrs by Western blot analysis2011ACS medicinal chemistry letters, Aug-11, Volume: 2, Issue:8
Identification of NAE Inhibitors Exhibiting Potent Activity in Leukemia Cells: Exploring the Structural Determinants of NAE Specificity.
AID1519940Antiproliferative activity against human MGC803 cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1599578Inhibition of NEDD8-activating enzyme E1C (unknown origin) assessed as effect on NEDD8/UBE2M complex formation at 10 to 20 uM pre-incubated for 10 mins followed by Mg-ATP addition after 60 mins by SDS-PAGE analysis2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
Potent 5-Cyano-6-phenyl-pyrimidin-Based Derivatives Targeting DCN1-UBE2M Interaction.
AID1349089Competitive inhibition of NAE (unknown origin) assessed as decrease in NAE-mediated Ubc12-NEDD8 conjugation levels at 10 uM preincubated for 10 mins followed by Mg2+ and various concentrations of ATP addition measured after 60 mins by Lineweaver-Burk plot2018European journal of medicinal chemistry, Jan-01, Volume: 143Structure-based identification of a NEDD8-activating enzyme inhibitor via drug repurposing.
AID1519941Antiproliferative activity against human PLC cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1519939Antiproliferative activity against human MKN45 cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1728035Inhibition of NAE (unknown origin)-mediated neddylation assessed as suppression of cullin1-Nedd8 adduct formation preincubated for 10 mins followed by ATP addition and measured after 30 mins by immunoblot analysis2021European journal of medicinal chemistry, Jan-15, Volume: 210Development of novel benzimidazole-derived neddylation inhibitors for suppressing tumor growth invitro and invivo.
AID1758842Inhibition of DCN-1 in human KYSE-70 cell line assessed as increase in HO-1 expression at 5 to 10 uM incubated for 24 hrs by Western blot analysis2021European journal of medicinal chemistry, May-05, Volume: 217Development of phenyltriazole thiol-based derivatives as highly potent inhibitors of DCN1-UBC12 interaction.
AID1894760Antiproliferative activity against human HCT-116 cells assessed as cell growth inhibition by sulforhodamine B assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1646304Inhibition of NAE (unknown origin)-mediated neddylation assessed as decrease in Nedd8-Ubcl2 adduct formation at 10 uM by Western blot analysis2020Bioorganic & medicinal chemistry letters, 01-15, Volume: 30, Issue:2
Discovery of 1,2,4-triazine-based derivatives as novel neddylation inhibitors and anticancer activity studies against gastric cancer MGC-803 cells.
AID1872478Inhibition of mouse UAE1 in human HCT-116 cells assessed as reduction in Ubc10-Ub thioester level incubated for 24 hrs by immunoblot analysis2022European journal of medicinal chemistry, Mar-05, Volume: 231NAE modulators: A potential therapy for gastric carcinoma.
AID1728034Antiproliferative activity against human A549 cells assessed as inhibition of cell proliferation at 100 uM incubated for 48 hrs by CCK-8 assay relative to control2021European journal of medicinal chemistry, Jan-15, Volume: 210Development of novel benzimidazole-derived neddylation inhibitors for suppressing tumor growth invitro and invivo.
AID1872477Inhibition of SAE1/UBA2 in human HCT-116 cells assessed as reduction in Ubc9-SUMO level incubated for 24 hrs by immunoblot analysis2022European journal of medicinal chemistry, Mar-05, Volume: 231NAE modulators: A potential therapy for gastric carcinoma.
AID1894767Volume of distribution at steady state in Sprague-Dawley rat at 1 mg/kg, iv2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1894764Observed AUCinfinity in Sprague-Dawley rat at 1 mg/kg, iv2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1519930Antiproliferative activity against human PLC cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1894763AUClast in Sprague-Dawley rat at 1 mg/kg, iv2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1894766Observed MRTinfinity in Sprague-Dawley rat at 1 mg/kg, iv2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1349110Induction of apoptosis in human Caco2 cells assessed as viable cells at 10 uM after 16 hrs by Annexin V-FITC/propidium iodide staining-based flow cytometric analysis (Rvb = 98.4%)2018European journal of medicinal chemistry, Jan-01, Volume: 143Structure-based identification of a NEDD8-activating enzyme inhibitor via drug repurposing.
AID1894757Inhibition of NAE (unknown origin) mediated neddylation assessed as decrease in NEDD8-Ubcl2 adduct formation incubated for 2 hrs in presence of L-glutathione and ATP by HTRF assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1599609Inhibition of DCN1-UBE2M protein-protein interaction in human NCI-H1975 cells assessed as increase in NQO1 accumulation at 3 uM incubated for 24 hrs by Western blot analysis2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
Potent 5-Cyano-6-phenyl-pyrimidin-Based Derivatives Targeting DCN1-UBE2M Interaction.
AID1894787Antiproliferative activity against human Jurkat cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1371351Inhibition of UBA6 (unknown origin) assessed as decrease in formation of E2-UBL thioester reaction product2018Journal of medicinal chemistry, 04-26, Volume: 61, Issue:8
Interrogating the Roles of Post-Translational Modifications of Non-Histone Proteins.
AID1894791Antiproliferative activity against human HT-29 cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1519927Antiproliferative activity against human MCF7 cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1894786Antiproliferative activity against human THP-1 cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1519922Antiproliferative activity against human EKVX cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1894793Antiproliferative activity against human DU-145 cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1758805Inhibition of DCN-1 in human NCI-H2170 cells assessed as increase in HO-1 expression at 5 to 10 uM incubated for 24 hrs by Western blot analysis2021European journal of medicinal chemistry, May-05, Volume: 217Development of phenyltriazole thiol-based derivatives as highly potent inhibitors of DCN1-UBC12 interaction.
AID656471Antiproliferative activity against human K562 cells after 72 hrs by CellTiter-Glo assay2011ACS medicinal chemistry letters, Aug-11, Volume: 2, Issue:8
Identification of NAE Inhibitors Exhibiting Potent Activity in Leukemia Cells: Exploring the Structural Determinants of NAE Specificity.
AID1519936Antiproliferative activity against human HuH7 cells measured after 72 to 96 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1371348Inhibition of UAE (unknown origin) assessed as decrease in formation of E2-UBL thioester reaction product2018Journal of medicinal chemistry, 04-26, Volume: 61, Issue:8
Interrogating the Roles of Post-Translational Modifications of Non-Histone Proteins.
AID1872479Inhibition of His-tagged UBA6 (unknown origin) expressed in sf9 insect cells2022European journal of medicinal chemistry, Mar-05, Volume: 231NAE modulators: A potential therapy for gastric carcinoma.
AID1894790Antiproliferative activity against human RKO cells assessed as cell growth inhibition measured after 72 hrs by SRB assay2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1519925Antiproliferative activity against human HuH7 cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID656450Inhibition of human recombinant NAE assessed as inhibition of NEDD8-UbcH12 complex formation at 0.001 to 10 uM after 1.5 hrs by Western blot analysis2011ACS medicinal chemistry letters, Aug-11, Volume: 2, Issue:8
Identification of NAE Inhibitors Exhibiting Potent Activity in Leukemia Cells: Exploring the Structural Determinants of NAE Specificity.
AID1894769Inhibition of human partial length wild type TAOK1 by measuring selectivity score at 1 uM by Kinomescan method relative to control2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1599612Inhibition of DCN1-UBE2M protein-protein interaction in human PC9 cells assessed as increase in NQO1 accumulation at 3 uM incubated for 24 hrs by Western blot analysis2019Journal of medicinal chemistry, 06-13, Volume: 62, Issue:11
Potent 5-Cyano-6-phenyl-pyrimidin-Based Derivatives Targeting DCN1-UBE2M Interaction.
AID1519923Antiproliferative activity against human H1299 cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1894762Half life in Sprague-Dawley rat at 1 mg/kg, iv2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1728036Antiproliferative activity against human A549 cells incubated for 48 hrs by CCK-8 assay2021European journal of medicinal chemistry, Jan-15, Volume: 210Development of novel benzimidazole-derived neddylation inhibitors for suppressing tumor growth invitro and invivo.
AID1519928Antiproliferative activity against human MKN45 cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1519987Antitumor activity against human A549 cells xenografted in nude mouse assessed as decrease in tumor size at 60 mg/kg, ip qd measured after 22 days2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1519924Antiproliferative activity against human HepG2 cells measured after 48 hrs by CCK8 assay2020European journal of medicinal chemistry, Jan-01, Volume: 185Discovery of candesartan cilexetic as a novel neddylation inhibitor for suppressing tumor growth.
AID1349111Induction of apoptosis in human Caco2 cells assessed as early apoptotic cells at 10 uM after 16 hrs by Annexin V-FITC/propidium iodide staining-based flow cytometric analysis (Rvb = 1.40%)2018European journal of medicinal chemistry, Jan-01, Volume: 143Structure-based identification of a NEDD8-activating enzyme inhibitor via drug repurposing.
AID1894804Toxicity in nude mouse xenografted with human HCT-116 cells assessed as effect on body weight at 60 mg/kg, sc bid administered at 5 on/5 off/8 for 18 days2021Journal of medicinal chemistry, 05-13, Volume: 64, Issue:9
Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold.
AID1349114Cytotoxicity against human Caco2 cells after 72 hrs by MTT assay2018European journal of medicinal chemistry, Jan-01, Volume: 143Structure-based identification of a NEDD8-activating enzyme inhibitor via drug repurposing.
AID1817930Antiproliferative activity against human A549 cells assessed as inhibition of cell proliferation measured after 72 hrs by MTT assay2021European journal of medicinal chemistry, Dec-15, Volume: 226Discovery of a cinnamyl piperidine derivative as new neddylation inhibitor for gastric cancer treatment.
AID1347414qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: Secondary screen by immunofluorescence2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (298)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's2 (0.67)29.6817
2010's207 (69.46)24.3611
2020's89 (29.87)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 39.57

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 Index39.57 (24.57)
Research Supply Index5.74 (2.92)
Research Growth Index6.49 (4.65)
Search Engine Demand Index55.12 (26.88)
Search Engine Supply Index1.97 (0.95)

This Compound (39.57)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials11 (3.68%)5.53%
Reviews16 (5.35%)6.00%
Case Studies0 (0.00%)4.05%
Observational0 (0.00%)0.25%
Other272 (90.97%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (41)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A Phase 1 Study to Evaluate the Effects of Rifampin on Pharmacokinetics of Pevonedistat in Patients With Advanced Solid Tumors [NCT03486314]Phase 120 participants (Actual)Interventional2018-08-13Completed
A Feasibility Trial of MLN4924 (Pevonedistat, TAK 924) Given in Combination With Azacitidine, Fludarabine, and Cytarabine, in Children, Adolescents, and Young Adults With Relapsed or Refractory Acute Myeloid Leukemia or Myelodysplastic Syndrome [NCT03813147]Phase 112 participants (Actual)Interventional2019-05-17Active, not recruiting
A Phase II Study of Pevonedistat (MLN4924, TAK924) and Azacitidine as Maintenance Therapy After Allogeneic Stem Cell Transplantation for Non-Remission Acute Myelogenous Leukemia [NCT03709576]Phase 23 participants (Actual)Interventional2018-07-18Terminated(stopped due to Funding pulled)
A Phase I Study of Pevonedistat (TAK-924) and Decitabine Combination Therapy in High Risk Acute Myeloid Leukemia [NCT03009240]Phase 130 participants (Anticipated)Interventional2017-08-21Active, not recruiting
Pevonedistat as a Single Agent and in Combination With Chemotherapy in Patients With Malignant Mesothelioma [NCT03319537]Phase 1/Phase 29 participants (Actual)Interventional2017-10-05Completed
A Phase 2, Open-Label Study of Intravenous Pevonedistat in Combination With Oral Decitabine and Cedazuridine in Adult Patients With Higher-Risk Myelodysplastic Syndromes [NCT04985656]Phase 20 participants (Actual)Interventional2021-10-01Withdrawn(stopped due to Sponsor Decision to Terminate Trial)
A Randomized Phase II Trial of MLN4924 (Pevonedistat) With Azacitidine Versus Azacitidine in Adult Relapsed or Refractory Acute Myeloid Leukemia [NCT03745352]Phase 20 participants (Actual)Interventional2019-05-20Withdrawn(stopped due to Inadequate accrual rate)
A Phase II Trial of Pevonedistat and Azacitidine in MDS or MDS/MPN Patients Who Fail Primary Therapy With DNA Methyl Transferase Inhibitors [NCT03238248]Phase 271 participants (Actual)Interventional2017-08-07Active, not recruiting
A Phase I/II Trial of Pevonedistat in Combination With Pembrolizumab in Patients With dMMR/MSI-H Cancers [NCT04800627]Phase 1/Phase 22 participants (Actual)Interventional2021-03-29Terminated(stopped due to Per PI)
Pevonedistat in Combination With Ruxolitinib for Treatment of Patients With Myelofibrosis [NCT03386214]Phase 18 participants (Actual)Interventional2018-04-23Terminated(stopped due to Sponsor stopped development of pevonedistat)
A Master Protocol for Biomarker-Based Treatment of AML (The Beat AML Trial) [NCT03013998]Phase 1/Phase 22,000 participants (Anticipated)Interventional2016-11-30Recruiting
A Phase 2 Study of MLN4924 (Pevonedistat) in Combination With Carboplatin and Paclitaxel in Advanced NSCLC Previously Treated With Immunotherapy [NCT03965689]Phase 227 participants (Actual)Interventional2019-09-03Active, not recruiting
A Randomized, Crossover Phase 1 Study to Evaluate the Effects of Pevonedistat on the QTc Interval in Patients With Advanced Solid Tumors [NCT03330106]Phase 168 participants (Actual)Interventional2017-11-15Completed
Phase II Trial of Pevonedistat (TAK-924) Plus Docetaxel in Patients With Previously Treated Advanced Non-Small Cell Lung Cancer [NCT03228186]Phase 240 participants (Actual)Interventional2018-03-05Terminated(stopped due to Pharmaceutical company discontinued the study drug.)
A Multisite Phase Ib Study of Pevonedistat, Azacitidine (or Decitabine), and Venetoclax (PAVE) for the Treatment of Patients With Acute Myelogenous Leukemia (AML) [NCT04172844]Phase 124 participants (Anticipated)Interventional2020-01-13Active, not recruiting
A Phase I, Open-Label, Dose Escalation Study of MLN4924, a Novel Inhibitor of Nedd8-Activating Enzyme, in Adult Patients With Melanoma [NCT01011530]Phase 137 participants (Actual)Interventional2009-12-31Completed
A Phase I Study of Pevonedistat (MLN4924, TAK924) in Combination With Ibrutinib in Patients With Relapsed/Refractory Chronic Lymphocytic Leukemia and Non-Hodgkin Lymphoma [NCT03479268]Phase 118 participants (Actual)Interventional2018-03-22Active, not recruiting
A Phase 1/1b Study of Pevonedistat in Combination With Select Standard of Care Agents in Patients With Higher-Risk Myelodysplastic Syndromes, Chronic Myelomonocytic Leukemia, Acute Myelogenous Leukemia, or Advanced Solid Tumors With Severe Renal Impairmen [NCT03814005]Phase 117 participants (Actual)Interventional2019-07-10Completed
Phase 1B/II Study of Escalating Doses of Pevonedistat (TAK-924, Formerly MLN4924) Administered in Combination With Standard Induction Chemotherapy (Cytarabine and Idarubicin) in Newly Diagnosed High Risk Acute Myelogenous Leukemia (AML) [NCT03330821]Phase 1/Phase 253 participants (Anticipated)Interventional2018-04-18Active, not recruiting
A Phase I/II Study of Azacitidine, Venetoclax and Pevonedistat in Adults With Newly Diagnosed Secondary or Therapy-Related AML [NCT03862157]Phase 1/Phase 251 participants (Actual)Interventional2019-02-27Active, not recruiting
Expanded Access Program: Pevonedistat (in Combination With Azacitidine) for the First-line Treatment of Higher Risk Myelodysplastic Syndromes [NCT04484363]0 participants Expanded AccessNo longer available
An Open-Label, Dose Escalation, Phase I Study of MLN4924, a Novel Inhibitor of Nedd8 Activating Enzyme, in Adult Patients With Lymphoma or Multiple Myeloma [NCT00722488]Phase 156 participants (Actual)Interventional2008-06-30Completed
An Open-Label, Dose Escalation, Phase 1 Study of MLN4924, a Novel Inhibitor of Nedd8-Activating Enzyme, in Adult Patients With Acute Myelogenous Leukemia,Myelodysplastic Syndrome, and Acute Lymphoblastic Leukemia [NCT00911066]Phase 172 participants (Actual)Interventional2009-06-30Completed
An Open-Label, Dose Escalation, Phase I Study of MLN4924, a Novel Inhibitor of Nedd8-Activating Enzyme, in Adult Patients With Nonhematologic Malignancies [NCT00677170]Phase 162 participants (Actual)Interventional2008-04-30Completed
Phase I/II Study of MLN4924 Alone Followed by Dose-Adjusted EPOCH-Rituximab + MLN4924 With Gene Expression Profiling and Mutational Analysis in Relapsed/Refractory de Novo Diffuse Large B-Cell Lymphoma [NCT01415765]Phase 1/Phase 20 participants (Actual)Interventional2011-07-15Withdrawn
A Phase 1/1b, Open-label Study of Pevonedistat (MLN4924, TAK-924) as Single Agent and in Combination With Azacitidine in Adult East Asian Patients With Acute Myeloid Leukemia (AML) or Myelodysplastic Syndromes (MDS) [NCT02782468]Phase 123 participants (Actual)Interventional2016-05-16Completed
MLN9708 (Ixazomib) and MLN4924 (Pevonedistat) in Relapsed/Refractory Multiple Myeloma Patients: A Phase 1b Trial [NCT03770260]Phase 18 participants (Actual)Interventional2020-02-10Completed
A Phase 3, Randomized, Controlled, Open-label, Clinical Study of Pevonedistat Plus Azacitidine Versus Single-Agent Azacitidine as First-Line Treatment for Patients With Higher-Risk Myelodysplastic Syndromes, Chronic Myelomonocytic Leukemia, or Low-Blast A [NCT03268954]Phase 3454 participants (Actual)Interventional2017-11-28Active, not recruiting
A Phase 1 Study to Assess Mass Balance, Pharmacokinetics, and Metabolism of [14C]-Pevonedistat in Patients With Advanced Solid Tumors [NCT03057366]Phase 18 participants (Actual)Interventional2017-05-11Completed
Treatment of MDS/AML Patients With an Impending Hematological Relapse With Azacitidine Alone or in Combination With PEvonedistat - a Randomized Phase 2 Trial [NCT04712942]Phase 214 participants (Actual)Interventional2021-01-01Completed
An Open-Label, Dose Escalation, Phase 1 Study of PEVONEDISTAT, a Novel Inhibitor of the NEDD8-Activating Enzyme (NAE), in Combination With Low Dose Cytarabine (LDAC) in Adult Patients With Acute Myelogenous Leukemia (AML) and Advanced Myelodysplastic Synd [NCT03459859]Phase 112 participants (Actual)Interventional2018-05-21Completed
A Phase 1b, Open-Label, Dose-Escalation Study of MLN4924 Plus Azacitidine in Treatment-Naïve Patients With Acute Myelogenous Leukemia Who Are 60 Years or Older [NCT01814826]Phase 164 participants (Actual)Interventional2013-04-10Completed
A Phase I Trial of Pevonedistat in Combination With Induction Chemotherapy for Adolescent and Young Adults With Relapsed/Refractory Acute Lymphoblastic Leukemia or Lymphoblastic Non-Hodgkin Lymphoma [NCT03349281]Phase 16 participants (Actual)Interventional2019-03-25Completed
A Phase 2 Study of Pevonedistat in Combination With Carboplatin and Paclitaxel in Advanced Intrahepatic Cholangiocarcinoma [NCT04175912]Phase 252 participants (Anticipated)Interventional2020-03-16Active, not recruiting
A Randomized Phase III, Multicentre, Open Label Clinical Trial Comparing Azacitidine Plus Pevonedistat Versus Azacitidine in Older/Unfit Patients With Newly Diagnosed Acute Myeloid Leukemia Who Are Ineligible for Standard Induction Chemotherapy [NCT04090736]Phase 3302 participants (Actual)Interventional2019-09-24Active, not recruiting
A Phase 1 Study to Evaluate the Effects of Fluconazole and Itraconazole CYP3A-Mediated Inhibition on the Pharmacokinetics, Safety, and Tolerability of MLN4924 in Patients With Advanced Solid Tumors [NCT02122770]Phase 151 participants (Actual)Interventional2014-04-01Completed
A Phase 2, Randomized, Controlled, Open-Label, Clinical Study of the Efficacy and Safety of Pevonedistat Plus Azacitidine Versus Single-Agent Azacitidine in Patients With Higher-Risk Myelodysplastic Syndromes, Chronic Myelomonocytic Leukemia and Low-Blast [NCT02610777]Phase 2120 participants (Actual)Interventional2016-04-14Completed
A Randomized, Open-label, Controlled, Phase 2 Study of Pevonedistat, Venetoclax, and Azacitidine Versus Venetoclax Plus Azacitidine in Adults With Newly Diagnosed Acute Myeloid Leukemia Who Are Unfit for Intensive Chemotherapy [NCT04266795]Phase 2164 participants (Actual)Interventional2020-10-13Active, not recruiting
A Phase 1 Study of Pevonedistat (MLN4924), a NEDD8 Activating Enzyme (NAE) Inhibitor, in Combination With Temozolomide and Irinotecan in Pediatric Patients With Recurrent or Refractory Solid Tumors [NCT03323034]Phase 130 participants (Actual)Interventional2018-01-11Active, not recruiting
A Phase 1b, Open-Label, Dose Escalation, Multi-arm Study of MLN4924 Plus Docetaxel, Gemcitabine, or Combination of Carboplatin and Paclitaxel in Patients With Solid Tumors [NCT01862328]Phase 164 participants (Actual)Interventional2013-06-10Completed
A Phase 1 Study of MLN4924 (Pevonedistat) and Belinostat in Relapsed/Refractory Acute Myeloid Leukemia or Myelodysplastic Syndrome [NCT03772925]Phase 130 participants (Anticipated)Interventional2019-06-20Active, not recruiting
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT01814826 (29) [back to overview]MTD Expansion Phase, t1/2: Terminal Disposition Phase Half-life for MLN4924
NCT01814826 (29) [back to overview]MTD Expansion Phase, Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for MLN4924
NCT01814826 (29) [back to overview]MTD Expansion Phase, Vss: Volume of Distribution at Steady-state for MLN4924
NCT01814826 (29) [back to overview]Number of Participants With TEAEs Related to Clinically Significant Laboratory Evaluation Findings
NCT01814826 (29) [back to overview]Number of Participants With TEAEs Related to Clinically Significant Vital Sign Findings
NCT01814826 (29) [back to overview]Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)
NCT01814826 (29) [back to overview]Overall Survival
NCT01814826 (29) [back to overview]Dose-escalation Phase, t1/2: Terminal Disposition Phase Half-life for MLN4924
NCT01814826 (29) [back to overview]MTD Expansion Phase, CLp: Systemic Clearance for MLN4924
NCT01814826 (29) [back to overview]MTD Expansion Phase, Lambdaz: Terminal Disposition Phase Rate Constant for MLN4924
NCT01814826 (29) [back to overview]MTD Expansion Phase, Rac: Observed Accumulation Ratio for MLN4924
NCT01814826 (29) [back to overview]MTD Expansion Phase, AUCinf: Area Under the Plasma Concentration-time Curve Extrapolated to Infinity for MLN4924
NCT01814826 (29) [back to overview]Duration of Response
NCT01814826 (29) [back to overview]MTD Expansion Phase, AUC24hours: Area Under the Plasma Concentration-time Curve From Time Zero to 24 Hours Post-Dose for MLN4924
NCT01814826 (29) [back to overview]MTD Expansion Phase, AUC0-tau: Area Under the Plasma Concentration-time Curve From Time Zero to the End of the Dosing Interval (Tau) for MLN4924
NCT01814826 (29) [back to overview]Maximum Tolerated Dose (MTD) Expansion Phase, Cmax: Maximum Observed Plasma Concentration for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, Vss: Volume of Distribution at Steady-state for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, Rac: Observed Accumulation Ratio for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, Lambdaz: Terminal Disposition Phase Rate Constant for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, Ctrough: Observed Plasma Concentration at the End of the Dosing Interval for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, Cmax: Maximum Observed Plasma Concentration for MLN4924
NCT01814826 (29) [back to overview]MTD Expansion Phase, Ctrough: Observed Plasma Concentration at the End of the Dosing Interval for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, CLp: Systemic Clearance for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, AUCinf: Area Under the Plasma Concentration-time Curve Extrapolated to Infinity for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, AUC24hours: Area Under the Plasma Concentration-time Curve From Time Zero to 24 Hours Post-Dose for MLN4924
NCT01814826 (29) [back to overview]Dose-escalation Phase, AUC0-tau: Area Under the Plasma Concentration-time Curve From Time Zero to the End of the Dosing Interval (Tau) for MLN4924
NCT01814826 (29) [back to overview]Best Overall Response Rate
NCT01814826 (29) [back to overview]Thirty-day Mortality Rate
NCT01862328 (8) [back to overview]Duration of Response
NCT01862328 (8) [back to overview]Percentage of Participants With Objective Response
NCT01862328 (8) [back to overview]Dose-escalation Phase: Plasma Concentrations-time Data of MLN4924
NCT01862328 (8) [back to overview]MTD Expansion Phase: Plasma Concentrations-time Data of MLN4924
NCT01862328 (8) [back to overview]MTD Expansion Phase: Plasma Concentrations-time Data of MLN4924
NCT01862328 (8) [back to overview]Number of Participants Who Experience at Least 1 Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)
NCT01862328 (8) [back to overview]Number of Participants With TEAEs Related to Clinically Significant Laboratory Evaluation Findings
NCT01862328 (8) [back to overview]Number of Participants With TEAEs Related to Clinically Significant Vital Sign Findings
NCT02122770 (17) [back to overview]Part A AUC∞: Area Under the Plasma Concentration-time Curve From Time 0 to Infinity for MLN4924 and MLN4924 + Itraconazole
NCT02122770 (17) [back to overview]Part A AUC∞: Area Under the Plasma Concentration-time Curve From Time 0 to Infinity for MLN4924 and MLN4924 + Fluconazole
NCT02122770 (17) [back to overview]Part A: Plasma Clearance (CLp) for MLN4924
NCT02122770 (17) [back to overview]Part A: Blood to Plasma (B/P) Concentration Ratio for MLN4924
NCT02122770 (17) [back to overview]Part A Tmax: Time to Reach the Cmax for MLN4924
NCT02122770 (17) [back to overview]Number of Participants With TEAEs Related to Clinically Significant Vital Sign Findings
NCT02122770 (17) [back to overview]Part A: Volume of Distribution (Vz) for MLN4924
NCT02122770 (17) [back to overview]Number of Participants With Clinically Significant Change From Baseline in Body Weight Measurements
NCT02122770 (17) [back to overview]Number of Participants Who Experience at Least 1 Treatment-emergent Adverse Event (TEAE) and Serious Adverse Event (SAE)
NCT02122770 (17) [back to overview]Part B: Percentage of Participants With Objective Response
NCT02122770 (17) [back to overview]Part A: Terminal Phase Elimination Half-life (T1/2) for MLN4924
NCT02122770 (17) [back to overview]Part B: Duration of Response
NCT02122770 (17) [back to overview]Part A Cmax: Maximum Observed Plasma Concentration for MLN4924 and MLN4924 + Itraconazole
NCT02122770 (17) [back to overview]Number of Participants With TEAEs Related to Clinically Significant Laboratory Evaluation Findings
NCT02122770 (17) [back to overview]Part A AUClast: Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration for MLN4924 and MLN4924 + Itraconazole
NCT02122770 (17) [back to overview]Part A AUClast: Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration for MLN4924 and MLN4924 + Fluconazole
NCT02122770 (17) [back to overview]Part A Cmax: Maximum Observed Plasma Concentration for MLN4924 and MLN4924 + Fluconazole
NCT02610777 (27) [back to overview]Percentage of Participants With CR by Cycle 4
NCT02610777 (27) [back to overview]Percentage of Participants With CR and PR by Cycle 4
NCT02610777 (27) [back to overview]Percentage of Participants With CR and Partial Remission (PR)
NCT02610777 (27) [back to overview]Percentage of Participants With at Least 1 Inpatient Hospital Admissions Related to HR MDS, CMML or Low-blast AML
NCT02610777 (27) [back to overview]Overall Survival (OS)
NCT02610777 (27) [back to overview]One-year Survival Rate
NCT02610777 (27) [back to overview]Event-Free Survival (EFS)
NCT02610777 (27) [back to overview]Duration of Overall Response
NCT02610777 (27) [back to overview]Duration of Complete Remission (CR) in Low-blast AML
NCT02610777 (27) [back to overview]Duration of Complete Remission (CR) and Partial Remission (PR)
NCT02610777 (27) [back to overview]Duration of Complete Remission (CR)
NCT02610777 (27) [back to overview]Percentage of Participants With Complete Remission (CR)
NCT02610777 (27) [back to overview]Time to Progressive Disease (PD), Relapse, or Death
NCT02610777 (27) [back to overview]Time to Subsequent Therapy
NCT02610777 (27) [back to overview]Number of Participants in the Safety Analysis Population With Clinically Significant Change From Baseline Values in Vital Signs Reported as TEAEs
NCT02610777 (27) [back to overview]Number of Participants in the Safety Analysis Population With Clinically Significant Laboratory Abnormalities Reported as TEAEs
NCT02610777 (27) [back to overview]Number of Participants Reporting One or More Treatment-Emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)
NCT02610777 (27) [back to overview]Number of Participants With Change From Baseline Values in Eastern Cooperative Oncology Group (ECOG) Performance Status
NCT02610777 (27) [back to overview]Percentage of Participants With Red Blood Cells (RBCs) and Platelet-transfusion Independence
NCT02610777 (27) [back to overview]Time to First CR or PR
NCT02610777 (27) [back to overview]Time to AML Transformation in HR MDS or CMML Participants
NCT02610777 (27) [back to overview]Six-month Survival Rate
NCT02610777 (27) [back to overview]Percentage of Participants With Overall Response by Cycle 4
NCT02610777 (27) [back to overview]Percentage of Participants With Overall Response
NCT02610777 (27) [back to overview]Percentage of Participants With CR in Low-blast AML by Cycle 4
NCT02610777 (27) [back to overview]Percentage of Participants With CR in Low-blast AML
NCT02610777 (27) [back to overview]Number of Participants in the Safety Analysis Population With Clinically Significant Change From Baseline in Electrocardiogram (ECG) Values Reported as TEAEs
NCT02782468 (12) [back to overview]Percentage of Participants With CR for Participants With MDS
NCT02782468 (12) [back to overview]Overall Response Rate (ORR) for Participants With AML
NCT02782468 (12) [back to overview]Number of Participants Reporting One or More Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)
NCT02782468 (12) [back to overview]Percentage of Participants With CR for Participants With AML
NCT02782468 (12) [back to overview]ORR for Participants With MDS
NCT02782468 (12) [back to overview]Number of Participants With Dose Limiting Toxicities (DLTs) Based on National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) Version 4.03 During Cycle 1
NCT02782468 (12) [back to overview]Number of Participants With Clinically Significant Abnormal Laboratory Values
NCT02782468 (12) [back to overview]Cmax: Maximum Observed Plasma Concentration for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 5
NCT02782468 (12) [back to overview]Cmax: Maximum Observed Plasma Concentration for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 1
NCT02782468 (12) [back to overview]CL: Total Clearance for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 5
NCT02782468 (12) [back to overview]CL: Total Clearance for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 1
NCT02782468 (12) [back to overview]AUC(0-tau): Area Under the Plasma Concentration-time Curve From Time 0 to Time Tau Over the Dosing Interval for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 5
NCT03057366 (15) [back to overview]Part A: Aefeces,14C: Cumulative Amount of [14C]-Pevonedistat Excreted in Feces up to the Last Sampling Interval
NCT03057366 (15) [back to overview]Part A: Aetotal,14C: Total Cumulative Excretion of [14C]-Pevonedistat From the Body
NCT03057366 (15) [back to overview]Part A: Aeurine,14C: Cumulative Amount of [14C]-Pevonedistat Excreted in Urine up to the Last Sampling Interval
NCT03057366 (15) [back to overview]Part A: Aeurine: Cumulative Amount of Pevonedistat Dose Excreted in Urine at 144-168 Hours Post-dose
NCT03057366 (15) [back to overview]Part A: Feurine: Cumulative Percentage of Pevonedistat Dose Excreted in Urine at 144-168 Hours Post-dose
NCT03057366 (15) [back to overview]Part A: Renal Clearance (CLR) for Pevonedistat
NCT03057366 (15) [back to overview]Number of Participants Reporting One or More Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)
NCT03057366 (15) [back to overview]Part A: AUClast: Area Under the Plasma and Whole Blood Concentration-time Curve From Time 0 to Time of the Last Quantifiable Concentration for Pevonedistat
NCT03057366 (15) [back to overview]Part A: AUClast: Area Under the Plasma and Whole Blood TRA Concentration Curve From Time 0 to Time of the Last Quantifiable Concentration for [14C]-Pevonedistat Drug-related Material
NCT03057366 (15) [back to overview]Part A: Cmax: Maximum Observed Plasma and Whole Blood Concentration for Pevonedistat
NCT03057366 (15) [back to overview]Part A: Cmax: Maximum Observed Plasma and Whole Blood TRA Concentration for [14C]-Pevonedistat Drug-related Material
NCT03057366 (15) [back to overview]Part A: Percent Distribution of Total Radioactivity (TRA) for Pevonedistat and Its Metabolites in Plasma, Urine and Feces
NCT03057366 (15) [back to overview]Part A: Tmax: Time to Reach the Maximum Plasma and Whole Blood Concentration (Cmax) for Pevonedistat
NCT03057366 (15) [back to overview]Part A: Tmax: Time to Reach the Maximum Plasma and Whole Blood TRA Concentration (Cmax) for [14C]-Pevonedistat Drug-related Material
NCT03057366 (15) [back to overview]Part B: Number of Participants With Best Overall Response as Per Investigator's Assessment
NCT03228186 (5) [back to overview]The Number of Patients Who Achieve Stable Disease
NCT03228186 (5) [back to overview]Median Progression Free Survival Time
NCT03228186 (5) [back to overview]Median Overall Survival Time
NCT03228186 (5) [back to overview]The Number of Toxicities by System Organ Class
NCT03228186 (5) [back to overview]The Percentage of Patients That Respond to Treatment
NCT03238248 (5) [back to overview]Time to Progression
NCT03238248 (5) [back to overview]Rate of Marrow Complete Response (mCR)
NCT03238248 (5) [back to overview]Rate of Hematologic Response Per IWG
NCT03238248 (5) [back to overview]Overall Survial Time
NCT03238248 (5) [back to overview]Objective Response Rate
NCT03268954 (29) [back to overview]Number of Participants With CR and Marrow CR and PR
NCT03268954 (29) [back to overview]Number of Participants With CR and Marrow CR
NCT03268954 (29) [back to overview]Number of Participants With Complete Remission (CR) and CR+ Complete Remission With Incomplete Blood Count Recovery (CRi)
NCT03268954 (29) [back to overview]Number of Participants With at Least 1 Inpatient Hospital Admissions Related to HR MDS, CMML or Low-blast AML
NCT03268954 (29) [back to overview]Thirty-Day Mortality Reported as Number of Participants Who Died Up to Day 30
NCT03268954 (29) [back to overview]Kaplan-Meier Estimates of Six-Month Survival Rate
NCT03268954 (29) [back to overview]Kaplan-Meier Estimates of One-Year Survival Rate
NCT03268954 (29) [back to overview]Event-Free Survival in Participants Who Have TP53 Mutations, 17p Deletions, and/or Are Determined to be in an Adverse Cytogenetic Risk Group
NCT03268954 (29) [back to overview]Number of Participants With CR and Marrow CR, PR and Hematologic Improvement (HI)
NCT03268954 (29) [back to overview]Duration of Overall Response 2 (OR2)
NCT03268954 (29) [back to overview]Duration of Overall Response (OR)
NCT03268954 (29) [back to overview]Duration of Complete Remission + Complete Remission With Incomplete Blood Count Recovery (CRi)
NCT03268954 (29) [back to overview]Duration of Complete Remission (CR)
NCT03268954 (29) [back to overview]Percentage of Participants With Red Blood Cells (RBCs) and Platelet-transfusion Independence
NCT03268954 (29) [back to overview]Number of Participants With Overall Response in Participants Who Have TP53 Mutations, 17p Deletions, and/or Are Determined to be in an Adverse Cytogenetic Risk Group
NCT03268954 (29) [back to overview]Event-Free Survival (EFS)
NCT03268954 (29) [back to overview]Number of Participants With Overall Response by Cycle 6
NCT03268954 (29) [back to overview]Health-Related Quality of Life (HRQOL) Using European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire- Core 30
NCT03268954 (29) [back to overview]Duration of Red Blood Cells (RBCs) and Duration of Platelet-transfusion Independence and Duration of Red Blood Cells (RBCs) and Platelet-transfusion Independence
NCT03268954 (29) [back to overview]Time to Progressive Disease (PD), Relapse After CR (Low-blast AML), Relapse After CR or PR (HR MDS/CMML), or Death
NCT03268954 (29) [back to overview]Time to First Complete Remission (CR) or Partial Remission (PR) or Complete Remission With Incomplete Blood Count Recovery (CRi)
NCT03268954 (29) [back to overview]Sixty-Day Mortality Reported as Number of Participants Who Died Up to Day 60
NCT03268954 (29) [back to overview]Overall Survival in Participants Who Have TP53 Mutations, 17p Deletions, and/or Are Determined to be in an Adverse Cytogenetic Risk Group
NCT03268954 (29) [back to overview]Overall Survival (OS)
NCT03268954 (29) [back to overview]Number of Participants With Overall Response 2 (OR2)
NCT03268954 (29) [back to overview]Number of Participants With Overall Response (OR)
NCT03268954 (29) [back to overview]Number of Participants With Hematologic Improvement (HI)
NCT03268954 (29) [back to overview]Number of Participants With CR, Partial Remission (PR) and Hematologic Improvement (HI)
NCT03268954 (29) [back to overview]Time to Acute Myelogenous Leukemia (AML) Transformation in Higher-Risk Myelodysplastic Syndromes (HR MDS), Higher-Risk Chronic Myelomonocytic Leukemias (HR CMML) and HR MDS/CMML Participants
NCT03323034 (8) [back to overview]Number of Participants With Grade 3 or Greater Toxicities Associated With Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]Anti-tumor Activity of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]T Max of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]MTD/RP2D of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]Half-life of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]Clearance of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]C Max of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03323034 (8) [back to overview]AUC of Pevonedistat in Combination With Irinotecan and Temozolomide
NCT03330106 (9) [back to overview]Part A: Change From Time-matched Baseline in QRS After Pevonedistat Administration
NCT03330106 (9) [back to overview]Part A: Change From Time-matched Baseline in PR After Pevonedistat Administration
NCT03330106 (9) [back to overview]Part A: Change From Time-matched Baseline in Individual Corrected QT Interval (QTcI) After Pevonedistat Administration
NCT03330106 (9) [back to overview]Part A: Change From Time-matched Baseline in Heart Rate (HR) After Pevonedistat Administration
NCT03330106 (9) [back to overview]Part A: Change From Time-matched Baseline in Fridericia-corrected QT Interval (QTcF) After Pevonedistat Administration
NCT03330106 (9) [back to overview]Part B: Overall Response Rate (ORR)
NCT03330106 (9) [back to overview]Part A: Terminal Phase Elimination Half-life (t1/2) for Pevonedistat
NCT03330106 (9) [back to overview]Part A: Cmax: Maximum Observed Plasma Concentration for Pevonedistat
NCT03330106 (9) [back to overview]Part A: AUC(0-24): Area Under the Plasma Concentration-time Curve From Time 0 to 24 Hours Postdose for Pevonedistat
NCT03486314 (7) [back to overview]Part A: Ratio of Area Under the Plasma Concentration-time Curve From Time 0 to Infinity (AUC∞) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)
NCT03486314 (7) [back to overview]Part B: Number of Participants With Best Overall Response as Per Investigator's Assessment
NCT03486314 (7) [back to overview]Part A: Total Clearance After Intravenous Administration (CL) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)
NCT03486314 (7) [back to overview]Part A: Volume of Distribution at Steady State After Intravenous Administration (Vss) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)
NCT03486314 (7) [back to overview]Part A: Terminal Disposition Phase Half-life (T1/2z) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)
NCT03486314 (7) [back to overview]Part A: Ratio of Maximum Observed Plasma Concentration (Cmax) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)
NCT03486314 (7) [back to overview]Part A: Ratio of Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration (AUClast) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)
NCT03770260 (1) [back to overview]Number of Participants Experiencing a Dose Limiting Toxicity (Dose Escalation)
NCT03813147 (8) [back to overview]Total Plasma Clearance of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS
NCT03813147 (8) [back to overview]Area Under the Plasma Concentration Versus Time Curve of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS
NCT03813147 (8) [back to overview]Number of Participants With Dose Limiting Toxicities of MLN4924 (Pevonedistat)
NCT03813147 (8) [back to overview]Elimination Half-life of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS
NCT03813147 (8) [back to overview]Maximum Concentration of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS
NCT03813147 (8) [back to overview]Maximum Time to Concentration of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS
NCT03813147 (8) [back to overview]Number of Participants With Antitumor Activity of MLN4924 (Pevonedistat)
NCT03813147 (8) [back to overview]Number of Participants With Adverse Events Attributable to MLN4924 (Pevonedistat)
NCT03965689 (2) [back to overview]Duration of Progression Free Survival (PFS)
NCT03965689 (2) [back to overview]Overall Survival (OS)
NCT04266795 (1) [back to overview]Event-Free Survival (EFS)

MTD Expansion Phase, t1/2: Terminal Disposition Phase Half-life for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionhour (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous7.458.07
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous7.307.89

[back to top]

MTD Expansion Phase, Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionhour (Median)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1.011.0
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous1.000.98

[back to top]

MTD Expansion Phase, Vss: Volume of Distribution at Steady-state for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionliter (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous370.53401.41
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous348.66370.97

[back to top] [back to top] [back to top]

Number of Participants With Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

(NCT01814826)
Timeframe: Baseline up to 30 days after the last dose of study drug (up to 5 years)

,,
InterventionParticipants (Count of Participants)
TEAEsSAEs
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous3221
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous2922
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous33

[back to top]

Overall Survival

Overall survival was defined as the time from the first dose of study drug to the date of death. The Kaplan-Meier method was used to estimate overall survival, along with the corresponding 95% confidence interval. (NCT01814826)
Timeframe: From the first dose of study drug up to date of death (up to 5 years)

Interventionmonths (Median)
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous12.25
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous4.93
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous5.22

[back to top]

Dose-escalation Phase, t1/2: Terminal Disposition Phase Half-life for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionhour (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous7.807.98
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous7.39NA

[back to top]

MTD Expansion Phase, CLp: Systemic Clearance for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
InterventionL/hr (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous41.3538.10
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous39.2034.02

[back to top]

MTD Expansion Phase, Lambdaz: Terminal Disposition Phase Rate Constant for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Intervention1/hr (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous0.100.09
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous0.100.09

[back to top]

MTD Expansion Phase, Rac: Observed Accumulation Ratio for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionratio (Mean)
Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 IntravenousNA
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 SubcutaneousNA

[back to top]

MTD Expansion Phase, AUCinf: Area Under the Plasma Concentration-time Curve Extrapolated to Infinity for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionhr*ng/mL (Mean)
Cycle 1 Day 1
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous990.58
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous1026.45

[back to top]

Duration of Response

The duration of response was defined in participants with disease response (CR, CRi, or PR) as the time between the first documentation of response and disease progression. Duration of response was determined by an investigator based on revised recommendations of the IWG Response Criteria for AML. CR: free of leukemia-related symptoms, absolute neutrophil count (ANC) greater than (>)1.0*10^9 per liter (/L), platelet count greater than or equal to (>=) 100*10^9/L, normal bone marrow with <5 percent (%) blasts and no Auer rods. CRi: As per CR but with residual thrombocytopenia (platelet count <100*10^9/L) or residual neutropenia (ANC <1.0*10^9/L). PR: >=50% decrease bone marrow blasts to 5 to 25% abnormal cells, or CR with less than or equal to (<=) 5% blasts if Auer rods present. (NCT01814826)
Timeframe: From the date of first documented CR, PR or CRi up to the date of first disease progression (Up to 5 years)

Interventionmonths (Median)
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous9.0
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous6.0

[back to top]

MTD Expansion Phase, AUC24hours: Area Under the Plasma Concentration-time Curve From Time Zero to 24 Hours Post-Dose for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 24 hours) post-dose (Cycle length = 28 days)

,
Interventionhr*ng/mL (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous891.89946.91
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous926.74954.07

[back to top]

MTD Expansion Phase, AUC0-tau: Area Under the Plasma Concentration-time Curve From Time Zero to the End of the Dosing Interval (Tau) for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionhr*ng/mL (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 IntravenousNA1125.90
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 SubcutaneousNA1122.91

[back to top]

Maximum Tolerated Dose (MTD) Expansion Phase, Cmax: Maximum Observed Plasma Concentration for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionng/mL (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous168.80176.65
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous159.78158.95

[back to top]

Dose-escalation Phase, Vss: Volume of Distribution at Steady-state for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionliter (L) (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous352.06351.82
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous257.32NA

[back to top]

Dose-escalation Phase, Tmax: Time to Reach the Maximum Plasma Concentration (Cmax) for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionhour (Median)
Cycle 1 Day 1Cycle Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1.061.0
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1.0NA

[back to top]

Dose-escalation Phase, Rac: Observed Accumulation Ratio for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionratio (Mean)
Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous0.99
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 IntravenousNA

[back to top]

Dose-escalation Phase, Lambdaz: Terminal Disposition Phase Rate Constant for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Intervention1 per hour (1/hr) (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous0.090.09
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous0.09NA

[back to top]

Dose-escalation Phase, Ctrough: Observed Plasma Concentration at the End of the Dosing Interval for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionng/mL (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous2.341.40
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1.68NA

[back to top]

Dose-escalation Phase, Cmax: Maximum Observed Plasma Concentration for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length is equal to [=] 28 days)

,
Interventionnanogram per milliliter (ng/mL) (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous173.60177.87
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous306.67NA

[back to top]

MTD Expansion Phase, Ctrough: Observed Plasma Concentration at the End of the Dosing Interval for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionng/mL (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous0.871.51
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous0.911.30

[back to top]

Dose-escalation Phase, CLp: Systemic Clearance for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionliter per hour (L/hr) (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous35.6935.27
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous29.78NA

[back to top]

Dose-escalation Phase, AUCinf: Area Under the Plasma Concentration-time Curve Extrapolated to Infinity for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionhr*ng/mL (Mean)
Cycle 1 Day 1
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1101.81
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1823.68

[back to top]

Dose-escalation Phase, AUC24hours: Area Under the Plasma Concentration-time Curve From Time Zero to 24 Hours Post-Dose for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 24 hours) post-dose (Cycle length = 28 days)

,
Interventionhr*ng/mL (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1020.321039.54
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1675.41NA

[back to top]

Dose-escalation Phase, AUC0-tau: Area Under the Plasma Concentration-time Curve From Time Zero to the End of the Dosing Interval (Tau) for MLN4924

(NCT01814826)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 24 hours) post-dose; Cycle 1 Day 5 pre-dose and at multiple time points (up to 48 hours) post-dose (Cycle length = 28 days)

,
Interventionhour*nanogram per milliliter (hr*ng/mL) (Mean)
Cycle 1 Day 1Cycle 1 Day 5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1151.201139.67
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous1805.47NA

[back to top]

Best Overall Response Rate

Disease response was based on best overall response as determined by an investigator based on revised recommendations of the International Working Group (IWG) Response Criteria for AML. Best overall response rate was defined as percentage of participants who had complete response (CR), partial response (PR), or CR/remission with incomplete blood count recovery (Cri). CR: free of leukemia-related symptoms, absolute neutrophil count (ANC) greater than (>)1.0*10^9 per liter (/L), platelet count greater than or equal to (>=) 100*10^9/L, normal bone marrow with <5 percent (%) blasts and no Auer rods. CRi: As per CR but with residual thrombocytopenia (platelet count <100*10^9/L) or residual neutropenia (ANC <1.0*10^9/L). PR: >=50% decrease bone marrow blasts to 5 to 25% abnormal cells, or CR with less than or equal to (<=) 5% blasts if Auer rods present. (NCT01814826)
Timeframe: Cycle(C)1Day(D)22 and at C2 between D20 and 28 and at C4 and beyond C4 after completion of every 3rd C between D15 and 28 up to 30 days after last dose of study drug/before start of subsequent antineoplastic therapy, if that occurred sooner(up to 5 years)

,,
Interventionpercentage of participants (Number)
CRPRCRi
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous43144
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous331313
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous5000

[back to top]

Thirty-day Mortality Rate

(NCT01814826)
Timeframe: 30 days after the first dose of study drug in Cycle 1 (Cycle Length=28 days)

Interventionpercentage of participants (Number)
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous5
MLN4924 20 mg/m^2 + Azacitidine 75 mg/m^2 Subcutaneous6
MLN4924 30 mg/m^2 + Azacitidine 75 mg/m^2 Intravenous0

[back to top]

Duration of Response

Duration of response: time from the date of first documented response per the investigator response assessment (CR or PR) to the date of PD or the date of last disease assessment if the participant discontinued the study before PD using RECIST 1.1 CR: complete disappearance of all target lesions and non-target disease, with exception of nodal disease; all nodes, both target and non-target, must decrease to normal (short axis <10 mm); no new lesions. PR: >=30% decrease under baseline of sum of diameters of all target lesions; short axis was used in sum for target nodes, while longest diameter was used in sum for all other target lesions; no unequivocal progression of non-target disease; no new lesions. PD: at least a 20% increase (including an absolute increase of at least 5 mm) in sum of diameters of target lesions, taking as reference smallest sum on study and/or unequivocal progression of existing non-target lesions and/or appearance of 1 or more new lesions. (NCT01862328)
Timeframe: From the date of first documented response (CR or PR) to the date of first documented PD or the date of last disease assessment if the participants discontinued the study before PD (up to 5 years)

Interventionmonths (Mean)
Arm 1: MLN4924 25 mg/m^2 + Docetaxel 75 mg/m^22.880
Arm 2a: MLN4924 15 mg/m^2 + Carboplatin AUC62.270
Arm 2:MLN4924 20 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC516.153
Arm 2:MLN4924 25 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC57.015

[back to top]

Percentage of Participants With Objective Response

Percentage of participants with objective response based on assessment of complete response (CR) or partial response (PR) according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. CR: complete disappearance of all target lesions and non-target disease, with exception of nodal disease; all nodes, both target and non-target, must decrease to normal (short axis less than [<] 10 millimeter [mm]); no new lesions. PR: greater than or equal to (>=) 30 percent (%) decrease under baseline of sum of diameters of all target lesions; short axis was used in sum for target nodes, while longest diameter was used in sum for all other target lesions; no unequivocal progression of non-target disease; no new lesions. (NCT01862328)
Timeframe: Screening, Cycle 2 Days 15 (Arm 1, 2a, and 2) and 22 (Arm 3) then every other Cycle thereafter up to 30 days after the last dose of study drug (up to 5 years) (Cycle Length = 21 days [Arm 1, 2a, and 2] and 28 days [Arm 3])

,,,,,,
Interventionpercentage of participants (Number)
CRPR
Arm 1: MLN4924 15 mg/m^2 + Docetaxel 75 mg/m^200
Arm 1: MLN4924 25 mg/m^2 + Docetaxel 75 mg/m^2019
Arm 2: MLN4924 15 mg/m^2+Paclitaxel 175mg/m^2+Carboplatin AUC500
Arm 2:MLN4924 20 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC51836
Arm 2:MLN4924 25 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC5040
Arm 2a: MLN4924 15 mg/m^2 + Carboplatin AUC6017
Arm 3: MLN4924 25 mg/m^2 + Gemcitabine 1000 mg/m^200

[back to top]

Dose-escalation Phase: Plasma Concentrations-time Data of MLN4924

(NCT01862328)
Timeframe: Cycle 1 Day 1 pre-dose and at multiple time points (up to 20 hours) post-dose (Cycle Length=21 days [Arm 1 and 2] and 28 days [Arm 3])

,,,,,
Interventionnanogram per milliliter (ng/ml) (Mean)
Cycle 1 Day 1: End-doseCycle 1 Day 1: 1.5 hours post doseCycle 1 Day 1: 3 hours post doseCycle 1 Day 1: 20 hours post dose
Arm 1: MLN4924 15 mg/m^2 + Docetaxel 75 mg/m^2163.882.048.07.5
Arm 1: MLN4924 25 mg/m^2 + Docetaxel 75 mg/m^2197.1130.695.814.6
Arm 2: MLN4924 15 mg/m^2+Paclitaxel 175mg/m^2+Carboplatin AUC5275.7156.8152.423.4
Arm 2:MLN4924 20 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC5257.3192.7138.034.6
Arm 2:MLN4924 25 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC5372.7246.7186.924.4
Arm 3: MLN4924 25 mg/m^2 + Gemcitabine 1000 mg/m^2222.0160.5103.014.2

[back to top]

MTD Expansion Phase: Plasma Concentrations-time Data of MLN4924

The number of participants analyzed includes only those participants who had data available for this measure. (NCT01862328)
Timeframe: Cycle 1 Days 1 and 5 pre-dose and at multiple time points (up to 20 hours) post-dose (Cycle Length=21 days [Arm 1 and 2])

Interventionng/ml (Mean)
Cycle 1 Day 1: End-doseCycle 1 Day 1: 1.5 hours post doseCycle 1 Day 1: 3 hours post doseCycle 1 Day 1: 6 hours post doseCycle 1 Day 1: 20 hours post doseCycle 1 Day 5: End-doseCycle 1 Day 5: 1.5 hours post doseCycle 1 Day 5: 3 hours post doseCycle 1 Day 5: 6 hours post doseCycle 1 Day 5: 20 hours post dose
Arm 2:MLN4924 20 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC5429.8253.6192.8136.330.2253.2171.0118.476.510.9

[back to top]

MTD Expansion Phase: Plasma Concentrations-time Data of MLN4924

The number of participants analyzed includes only those participants who had data available for this measure. (NCT01862328)
Timeframe: Cycle 1 Days 1 and 5 pre-dose and at multiple time points (up to 20 hours) post-dose (Cycle Length=21 days [Arm 1 and 2])

Interventionng/ml (Mean)
Cycle 1 Day 1: End-doseCycle 1 Day 1: 1.5 hours post doseCycle 1 Day 1: 3 hours post doseCycle 1 Day 1: 7 hours post doseCycle 1 Day 1: 20 hours post doseCycle 1 Day 5: End-doseCycle 1 Day 5: 1.5 hours post doseCycle 1 Day 5: 3 hours post doseCycle 1 Day 5: 7 hours post doseCycle 1 Day 5: 20 hours post dose
Arm 1: MLN4924 25 mg/m^2 + Docetaxel 75 mg/m^2152.8137.386.463.414.2211.1115.396.738.39.8

[back to top]

Number of Participants Who Experience at Least 1 Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

(NCT01862328)
Timeframe: Baseline up to 30 days after the last dose of study drug (up to 5 years)

,,,,,,
InterventionParticipants (Count of Participants)
TEAESAE
Arm 1: MLN4924 15 mg/m^2 + Docetaxel 75 mg/m^241
Arm 1: MLN4924 25 mg/m^2 + Docetaxel 75 mg/m^2189
Arm 2: MLN4924 15 mg/m^2+Paclitaxel 175mg/m^2+Carboplatin AUC572
Arm 2:MLN4924 20 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC5123
Arm 2:MLN4924 25 mg/m^2+Paclitaxel 175 mg/m^2+Carboplatin AUC571
Arm 2a: MLN4924 15 mg/m^2 + Carboplatin AUC663
Arm 3: MLN4924 25 mg/m^2 + Gemcitabine 1000 mg/m^2107

[back to top] [back to top] [back to top]

Part A AUC∞: Area Under the Plasma Concentration-time Curve From Time 0 to Infinity for MLN4924 and MLN4924 + Itraconazole

(NCT02122770)
Timeframe: Day 1 (MLN4924) and Day 8 (MLN4924 + Itraconazole): pre-dose and at multiple time points (up to 72 hours) post-dose

Interventionh*ng/mL (Geometric Mean)
Part A: MLN4924 8 mg/m^2465
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg585
Part A: MLN4924 15 mg/m^2798
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg1060
Part A: MLN4924 20 mg/m^21120
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg1130

[back to top]

Part A AUC∞: Area Under the Plasma Concentration-time Curve From Time 0 to Infinity for MLN4924 and MLN4924 + Fluconazole

(NCT02122770)
Timeframe: Day 1 (MLN4924) and Day 8 (MLN4924 + Fluconazole): pre-dose and at multiple time points (up to 72 hours) post-dose

Interventionh*ng/mL (Geometric Mean)
Part A: MLN4924 8 mg/m^2450
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg498

[back to top]

Part A: Plasma Clearance (CLp) for MLN4924

(NCT02122770)
Timeframe: Days 1 and 8: predose and at multiple time-points (up to 72 hours) postdose for Part A

,,,
Interventionliter per hour (L/h) (Geometric Mean)
Day 1Day 8
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg35.226.5
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg35.835.5
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg31.728.6
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg29.223.2

[back to top]

Part A: Blood to Plasma (B/P) Concentration Ratio for MLN4924

(NCT02122770)
Timeframe: Day 1 up to 24 hours post infusion

,,,
InterventionRatio (Geometric Mean)
End of infusion2 hours post infusion8 hours post infusion24 hours post infusion
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg55.266.570.766.3
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg35.148.856.758.0
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg51.169.271.171.9
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg43.754.773.767.6

[back to top]

Part A Tmax: Time to Reach the Cmax for MLN4924

(NCT02122770)
Timeframe: Days 1 and 8: predose and at multiple time-points (up to 72 hours) postdose for Part A

,,,
Interventionhour (Median)
Day 1Day 8
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg1.241.03
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg1.051.13
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg1.041.21
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg1.021.50

[back to top] [back to top]

Part A: Volume of Distribution (Vz) for MLN4924

(NCT02122770)
Timeframe: Days 1 and 8: predose and at multiple time-points (up to 72 hours) postdose for Part A

,,,
InterventionLiter (L) (Geometric Mean)
Day 1Day 8
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg494421
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg511550
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg503477
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg445445

[back to top]

Number of Participants With Clinically Significant Change From Baseline in Body Weight Measurements

(NCT02122770)
Timeframe: Part A: Baseline up to Day 40; Part B: Baseline up to approximately Cycle 29

,,,,,
Interventionparticipants (Number)
Grade 1 AE: 5 < 10% decrease from baselineGrade 2 AE: 10 - <20% decrease from baselineGrade 3 AE: >=20% decrease from baseline
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg000
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg110
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg000
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg000
Part B: MLN4924 + Carboplatin or Paclitaxel100
Part B: MLN4924 + Docetaxel100

[back to top]

Number of Participants Who Experience at Least 1 Treatment-emergent Adverse Event (TEAE) and Serious Adverse Event (SAE)

(NCT02122770)
Timeframe: Baseline up to 30 days after the last dose of study drug (Day 40 for Part A; approximately Cycle 29 for Part B)

,,,,,
InterventionParticipants (Count of Participants)
TEAESAE
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg62
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg185
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg133
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg133
Part B: MLN4924 + Carboplatin or Paclitaxel136
Part B: MLN4924 + Docetaxel2314

[back to top]

Part B: Percentage of Participants With Objective Response

Percentage of participants with objective response based on assessment of complete response (CR) or partial response (PR) according to Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. CR was defined as complete disappearance of all target lesions. All pathological lymph nodes, both target and non-target, must decrease to normal (short axis less than [<]10 millimeter [mm]). PR was defined as at least 30% decrease under baseline of the sum of diameters of all target lesions. (NCT02122770)
Timeframe: Baseline up to symptomatic deterioration, progressive disease (PD), treatment discontinuation, or until the study is stopped (approximately Cycle 29)

Interventionpercentage of participants (Number)
Part B: MLN4924 + Docetaxel10.5
Part B: MLN4924 + Carboplatin or Paclitaxel22.2

[back to top]

Part A: Terminal Phase Elimination Half-life (T1/2) for MLN4924

(NCT02122770)
Timeframe: Days 1 and 8: predose and at multiple time-points (up to 72 hours) postdose for Part A

,,,
Interventionhour (Geometric Mean)
Day 1Day 8
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg9.7411.0
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg9.8810.7
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg11.011.6
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg10.613.3

[back to top]

Part B: Duration of Response

The duration of response was defined in participants with disease response (CR or PR) as the time between the first documentation of response and progressive disease (PD). Responders without PD will be censored at the last clinical assessment of response. CR was defined as complete disappearance of all target lesions. All pathological lymph nodes, both target and non-target, must decrease to normal (short axis <10 mm). PR was defined as at least 30% decrease under baseline of the sum of diameters of all target lesions. PD was defined as at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). (NCT02122770)
Timeframe: Time from the date of first documentation of a response and PD (approximately up Cycle 29)

Interventionmonths (Mean)
Part B: MLN4924 + Docetaxel4.07
Part B: MLN4924 + Carboplatin or Paclitaxel8.46

[back to top]

Part A Cmax: Maximum Observed Plasma Concentration for MLN4924 and MLN4924 + Itraconazole

(NCT02122770)
Timeframe: Day 1 (MLN4924) and Day 8 (MLN4924 + Itraconazole): pre-dose and at multiple time points (up to 72 hours) post-dose

Interventionng/mL (Geometric Mean)
Part A: MLN4924 8 mg/m^259.1
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg66.8
Part A: MLN4924 15 mg/m^2121
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg193
Part A: MLN4924 20 mg/m^2178
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg137

[back to top] [back to top]

Part A AUClast: Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration for MLN4924 and MLN4924 + Itraconazole

(NCT02122770)
Timeframe: Day 1 (MLN4924) and Day 8 (MLN4924 + Itraconazole): pre-dose and at multiple time points (up to 72 hours) post-dose

Interventionh*ng/mL (Geometric Mean)
Part A: MLN4924 8 mg/m^2459
Part A: MLN4924 8 mg/m^2 + Itraconazole 200 mg571
Part A: MLN4924 15 mg/m^2793
Part A: MLN4924 15 mg/m^2 + Itraconazole 200 mg1030
Part A: MLN4924 20 mg/m^21110
Part A: MLN4924 20 mg/m^2 + Itraconazole 200 mg1140

[back to top]

Part A AUClast: Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration for MLN4924 and MLN4924 + Fluconazole

(NCT02122770)
Timeframe: Day 1 (MLN4924) and Day 8 (MLN4924 + Fluconazole): pre-dose and at multiple time points (up to 72 hours) post-dose

Interventionhour*nanogram per milliliter (h*ng/mL) (Geometric Mean)
Part A: MLN4924 8 mg/m^2445
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg491

[back to top]

Part A Cmax: Maximum Observed Plasma Concentration for MLN4924 and MLN4924 + Fluconazole

(NCT02122770)
Timeframe: Day 1 (MLN4924) and Day 8 (MLN4924 + Fluconazole): pre-dose and at multiple time points (up to 72 hours) post-dose

Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Part A: MLN4924 8 mg/m^251.6
Part A: MLN4924 8 mg/m^2 + Fluconazole 400 mg51.0

[back to top]

Percentage of Participants With CR by Cycle 4

Disease responses for HR MDS or CMML were based on modified IWG response criteria for MDS and for low-blast AML on revised IWG response criteria for AML. CR for HR MDS or CMML: <=5% myeloblasts with normal maturation of all cell lines in bone marrow, and >=11 g/dL Hb, >=100*10^9/L plt, ANC >=1.0*10^9/L and 0% blasts in peripheral blood. CR for low-blast AML: morphologic leukemia-free state, ANC of more than 1.0*10^9/L and plt of >=100*10^9/L, transfusion independence, no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery for low-blast AML: some participants fulfill all criteria for CR except for residual neutropenia (<1.0*10^9/L)/TTP (<100*10^9/L). The percentages are rounded off to report the nearest whole numbers. (NCT02610777)
Timeframe: From date of randomization until CR by Cycle 4 (cycle length is equal to [=] 28 days)

Interventionpercentage of participants (Number)
Azacitidine 75 mg/m^213.2
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^226.3

[back to top]

Percentage of Participants With CR and PR by Cycle 4

Disease responses for HR MDS/CMML per modified IWG response criteria for MDS and for low-blast AML on revised IWG response criteria for AML.CR for HR MDS/CMML: <=5% myeloblasts with normal maturation of all cell lines in bone marrow, >=11 g/dL Hb; >=100*10^9/L plt; >=1.0*10^9/L ANC and 0% blasts in peripheral blood.PR for HR MDS/CMML: considered achieved if all CR criteria is met except for bone marrow blasts decreased by>=50% over pretreatment but still>5%.CR for low-blast AML: morphologic leukemia-free state, ANC of>1.0*10^9/L and plt of>=100*10^9/L, transfusion independence, no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery for low-blast AML: fulfill all criteria for CR except for residual neutropenia (<100*10^9/L)/TTP (<100*10^9/L). PR for low-blast AML: all hematological values for CR but with decrease of >=50% in percentage of blasts to 5%-25% in bone marrow aspirate. The percentages are rounded off to report the nearest whole numbers. (NCT02610777)
Timeframe: From date of randomization until CR and PR, by Cycle 4 (cycle length=28 days)

Interventionpercentage of participants (Number)
Azacitidine 75 mg/m^221.1
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^231.6

[back to top]

Percentage of Participants With CR and Partial Remission (PR)

Disease responses for HR MDS/CMML per modified IWG response criteria for MDS and for low-blast AML on revised IWG response criteria for AML.CR for HR MDS/CMML: <=5% myeloblasts with normal maturation of all cell lines in bone marrow ,>=11 g/dL Hb;>=100*10^9/L plt; >=1.0*10^9/L ANC and 0% blasts in peripheral blood. PR for HR MDS/CMML:considered achieved if all CR criteria is met except for bone marrow blasts decreased by >=50%over pretreatment but still >5%. CR for low-blast AML: morphologic leukemia-free state, ANC of >1.0*10^9/L and plt of >=100*10^9/L, transfusion independence, no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery for low-blast AML:fulfill all criteria for CR except for residual neutropenia (<100*10^9/L)/TTP(<100*10^9/L). PR for low-blast AML:all hematological values for CR but with decrease of >=50% in percentage of blasts to 5%-25% in bone marrow aspirate. The percentages are rounded off to report the nearest whole numbers. (NCT02610777)
Timeframe: From date of randomization until CR and PR (up to approximately 5 years)

Interventionpercentage of participants (Number)
Azacitidine 75 mg/m^245
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^251

[back to top] [back to top]

Overall Survival (OS)

OS was defined as the time from the date of randomization to the date of death due to any cause. Participants without documented death at the time of the analysis were censored at the date the participant was last known to be alive. The Kaplan Meier estimates was used for the analysis. (NCT02610777)
Timeframe: From date of randomization until death (up to 3 years and 5 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^219.0
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^221.8

[back to top]

One-year Survival Rate

Kaplan-Meier estimate of probability of OS at the end of the first year from randomization. (NCT02610777)
Timeframe: Month 12

Interventionsurvival probability (Number)
Azacitidine 75 mg/m^20.677
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^20.845

[back to top]

Event-Free Survival (EFS)

EFS is defined as the time from the date of randomization to the date of the occurrence of an event. An event is defined as death or transformation to AML for HR MDS/CMML participants, whichever occurs first, or defined as death for low-blast AML participants. HR MDS/CMML participants without documented EFS event will be censored at the date of the last response assessment. HR MDS/CMML participants with no response assessment and no death will be censored at the date of randomization. Low-blast AML participants without documentation of death will be censored at the date the participant was last known to be alive. HR MDS/CMML participants who received alternative antineoplastic therapy before death or transformation to AML will be censored at the date of last adequate assessment prior to starting alternate antineoplastic therapy. The Kaplan-Meier estimate was used for the analysis. (NCT02610777)
Timeframe: From date of randomization until transformation to AML, or death due to any cause (up to approximately 5 years)

Interventionmonths (Median)
Azacitidine 75 mg/m^217.6
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^221.0

[back to top]

Duration of Overall Response

Duration of OR: response to first documentation of PD or relapse from CR for low-blast AML or relapse after CR or PR for HR MDS/CMML. Disease responses(HR MDS/CMML): modified IWG criteria for MDS;low-blast AML: revised IWG criteria for AML.Overall response(HR MDS/CMML)=CR,PR/HI,LB AML=CR+ Cri+PR.HR MDS/CMML-CR:≤5%myeloblasts with normal maturation of BM cell lines,≥11g/dL Hb,≥100*10^9/L plt,≥1.0*10^9/L ANC,0%blasts in peripheral blood; PR:CR criteria met except BM blasts≥50%less over pretreatment but still>5%; HI:hb increase(inc)≥1.5g/dL if baseline<11g/dL; plt inc≥30*10^9/L if baseline >20*10^9/L inc from<20*10^9/L->20*10^9/L,ANC inc by100%; absolute inc of >0.5*10^9/L if baseline <100*10^9/L.LB AML-CR: morphologic leukemia-freestate >1.0*10^9 ANC,≥100*1 ^9/Lplt, transfusion independence, no residual evidence of extramedullary leukemia; CRi:fulfill CR criteria except residual neutropenia<1.0*10^9/L/TTP<100*10^9/L; PR:all CR hematological values but>=50%less in BM aspirate. (NCT02610777)
Timeframe: From date of randomization until CR, PR or HI (up to approximately 5 years)

Interventionmonths (Median)
Azacitidine 75 mg/m^214.0
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^220.6

[back to top]

Duration of Complete Remission (CR) in Low-blast AML

Disease responses for low-blast AML is based on revised IWG response criteria for AML. CR for low-blast AML: morphologic leukemia-free state, ANC of more than 1.0*10^9/L and plt of >=100*10^9/L, transfusion independence, no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery for low-blast AML: some participants fulfill all criteria for CR except for residual neutropenia (<1.0*10^9/L)/TTP (<100*10^9/L). (NCT02610777)
Timeframe: From date of randomization until CR (up to approximately 5 years)

Interventionmonths (Median)
Azacitidine 75 mg/m^210.2
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^212.6

[back to top]

Duration of Complete Remission (CR) and Partial Remission (PR)

Duration of CR is first documented CR to the first documentation of PD or relapse from CR (participants with low-blast AML). Disease responses (HR MDS/CMML) are based on modified IWG response criteria for MDS and for low-blast AML on revised IWG response criteria for AML. For HR MDS/CMML-CR:<=5%myeloblasts with normal maturation of all bone marrow cell lines, >=11 g/dL Hb, >=100*10^9/L plt,>=1.0*10^9/L neutrophils, 0% blasts in peripheral blood;PR: all CR criteria met except bone marrow blasts >=50% decrease over pretreatment but still >5%; low-blast AML-CR: morphologic leukemia-free state, >1.0*10^9/L ANC ,plt >=100*10^9/L,transfusion independence, no residual evidence of extramedullary leukemia;CR with incomplete blood count recovery:fulfill CR criteria except residual neutropenia <1.0*10^9/L/TTP <100*10^9/L;PR:all CR hematological values but with a decrease of >=50% in blasts percentage to 5%-25% in bone marrow aspirate. (NCT02610777)
Timeframe: From date of randomization until CR or PR (up to approximately 5 years)

Interventionmonths (Median)
Azacitidine 75 mg/m^212.9
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^218.6

[back to top]

Duration of Complete Remission (CR)

Duration of CR is first documented CR to the first documentation of PD or relapse from CR (participants with low-blast AML) or relapse after CR or PR (participants with HR MDS/CMML). Disease responses for HR MDS or CMML are based on the Modified IWG Response Criteria for MDS and for low-blast AML on the Revised IWG Response Criteria for AML.CR for HR MDS or CMML≤5% myeloblasts with normal maturation of all cell lines in the bone marrow,≥11 g/dL Hgb,≥100*10^9/L pl,≥1.0*10^9/L neutrophils;0% blasts in peripheral blood.CR for low-blast AML:morphologic leukemia-free state,neutrophils of<1.0*10^9/L;pl of≥100*10^9/L,transfusion independence,no residual evidence of extramedullary leukemia.CRi for low-blast AML: participants fulfill all of the criteria for CR except for residual neutropenia (<1.0*10^9/L) or thrombocytopenia (pl<100*10^9/L). (NCT02610777)
Timeframe: From date of randomization until CR (up to approximately 5 years)

Interventionmonths (Median)
Azacitidine 75 mg/m^212.9
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^218.6

[back to top]

Percentage of Participants With Complete Remission (CR)

Disease responses for HR MDS or CMML is based on the modified International Working Group (IWG) response criteria for MDS and for low-blast AML on the revised IWG response criteria for AML. CR for HR MDS or CMML: <= 5% myeloblasts with normal maturation of all cell lines in the bone marrow, and >= 11 gram/deciliter (g/dL) hemoglobin (Hb), >=100*10^9/liter (/L) platelets (plt), >=1.0*10^9/L absolute neutrophil count (ANC) and 0% blasts in peripheral blood. CR for low-blast AML: morphologic leukemia-free state, ANC of more than 1.0*10^9/L and plt of >=1.0*10^9/L, transfusion independence, and no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery for low-blast AML: some participants fulfill all of the criteria for CR except for residual neutropenia (<1.0*10^9/L) or thrombocytopenia (TTP) (<100*10^9/L). The percentages are rounded off to report the nearest whole numbers. (NCT02610777)
Timeframe: From date of randomization until CR (up to approximately 5 years)

Interventionpercentage of participants (Number)
Azacitidine 75 mg/m^236
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^245

[back to top]

Time to Progressive Disease (PD), Relapse, or Death

Time from randomization until PD/transformation to AML/relapse/death due to any cause, whichever occurs first. Relapse after CR or PR in MDS/CMML: return to pretreatment bone marrow blast % or decrement of >=50% from maximum remission levels in ANC or plt, reduction in Hb concentration by >=1.5 g/dL or transfusion dependence. PD: at least 50% decrement from maximum remission in ANC or plt, or reduction in Hb by>=2g/dL or transfusion dependence;participants with <5% blasts: >=50% increase (inc) in blasts to >5%; 5%-10%: >=50% inc to >10%; 10%-20%: >=50% inc to>20%;20%-30%:>=50% inc to >30%. Relapse after CR in Low blast AML: reappearance of leukemic blasts in peripheral blood or >=5% blasts in bone marrow not attributable to any cause (example, bone marrow regeneration after consolidation therapy). If there are no circulating blasts, bone marrow contains 5%-20% blasts, a repeat analysis is performed a week later. (NCT02610777)
Timeframe: From date of randomization until PD, relapse or death (up to approximately 5 years)

Interventionmonths (Median)
Azacitidine 75 mg/m^213.6
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^215.2

[back to top]

Time to Subsequent Therapy

Time to subsequent therapy is defined as time from randomization to the date of the first subsequent therapy. Subsequent therapy is defined as agent(s) with antileukemic/anti-MDS activity. Participants who discontinue study treatment to receive single-agent azacitidine off study did not be counted as receiving subsequent therapy. (NCT02610777)
Timeframe: From date of randomization up to approximately 5 years

Interventionmonths (Median)
Azacitidine 75 mg/m^2NA
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^2NA

[back to top]

Number of Participants in the Safety Analysis Population With Clinically Significant Change From Baseline Values in Vital Signs Reported as TEAEs

Vital signs assessments included diastolic and systolic blood pressure, heart rate, and body temperature. (NCT02610777)
Timeframe: From date of first dose up to 30 days after administration of the last dose of any study drug (up to approximately 5 years)

,
InterventionParticipants (Count of Participants)
PyrexiaHypotension
Azacitidine 75 mg/m^2253
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^2226

[back to top]

Number of Participants in the Safety Analysis Population With Clinically Significant Laboratory Abnormalities Reported as TEAEs

Laboratory assessments included clinical chemistry, hematology, and urinalysis. (NCT02610777)
Timeframe: From date of first dose up to 30 days after administration of the last dose of any study drug (up to approximately 5 years)

,
InterventionParticipants (Count of Participants)
NeutropeniaAnaemiaNeutrophil count decreasedThrombocytopenia
Azacitidine 75 mg/m^22129615
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^221191216

[back to top]

Number of Participants Reporting One or More Treatment-Emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

An adverse event is any untoward medical occurrence in a participant administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. A SAE was defined as any untoward medical occurrence that: 1) results in death, 2) is life-threatening, 3) requires inpatient hospitalization or prolongation of existing hospitalization, 4) results in persistent or significant disability/incapacity, 5) leads to a congenital anomaly/birth defect in the offspring of the participant or 6) is an medically important event that satisfies any of the following: a) May require intervention to prevent items 1 through 5 above. b) May expose the participant to danger, even though the event is not immediately life threatening or fatal or does not result in hospitalization. A TEAE was defined as any adverse event occurring after the start of pevonedistat administration of the treatment period. (NCT02610777)
Timeframe: From date of first dose up to 30 days after administration of the last dose of any study drug (up to approximately 5 years)

,
InterventionParticipants (Count of Participants)
TEAEsSAEs
Azacitidine 75 mg/m^26240
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^25740

[back to top]

Number of Participants With Change From Baseline Values in Eastern Cooperative Oncology Group (ECOG) Performance Status

Number of participants with change from Baseline in ECOG performance status was measured on 6 point scale to assess participant's performance status, where: Grade 0(Normal activity. Fully active, able to carry on all pre-disease activities without restriction); Grade 1(Symptoms but ambulatory. Restricted in physically strenuous activity, but ambulatory and able to carry out light or sedentary work); Grade 2(In bed <50% of the time. Ambulatory and capable of all self-care, but unable to carry out any work activities. Up and about more than 50% of waking hours); Grade 3(In bed >50% of the time. Capable of only limited self-care, confined to bed or chair more than 50 percent of waking hours); Grade 4(100% bedridden. Completely disabled, cannot carry on any self-care, totally confined to bed or chair); Grade 5(Dead). (NCT02610777)
Timeframe: From date of first dose up to 30 days after administration of the last dose of any study drug (up to approximately 5 years)

,
InterventionParticipants (Count of Participants)
Baseline: 0; Overall: 0Baseline: 0; Overall: 1Baseline: 0; Overall: 2Baseline: 0; Overall: 3Baseline: 0; Overall: 4Baseline: 1; Overall: 1Baseline: 1; Overall: 2Baseline: 1; Overall: 3Baseline: 2; Overall: 2
Azacitidine 75 mg/m^2131530217552
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^291332018823

[back to top]

Percentage of Participants With Red Blood Cells (RBCs) and Platelet-transfusion Independence

A participant was defined as RBC or platelet-transfusion independent if he/she received no RBC or platelet transfusions for a period of at least 8 weeks before the first dose of study drug through 30 days after the last dose of any study drug. Rate of transfusion independence was defined as number of participants who became transfusion independent divided by the number of participants who were transfusion dependent at Baseline. (NCT02610777)
Timeframe: 8 weeks before randomization through 30 days after last dose of any study drug (up to approximately 5 years and 3 months)

,
Interventionpercentage of participants (Number)
RBCs-transfusion IndependencePlatelet-transfusion Independence
Azacitidine 75 mg/m^250.060.0
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^269.280.0

[back to top]

Time to First CR or PR

Time to first CR or PR: time from randomization to first documented CR or PR, whichever occurs first. Disease responses (HR MDS/CMML) based on modified IWG response criteria for MDS; low-blast AML on revised IWG response criteria for AML. HR MDS/CMML-CR: <=5% myeloblasts with normal maturation of all bone marrow cell lines, >=11 g/dL Hb, >=100*10^9/L plt,>=1.0*10^9/L ANC, 0% blasts in peripheral blood; PR: all CR criteria met except bone marrow blasts >=50% decrease over pretreatment but still >5%; For low-blast AML-CR: morphologic leukemia-free state, >1.0*10^9/L ANC, plt >=100*10^9/L, transfusion independence, no residual evidence of extramedullary leukemia; CR with incomplete blood count recovery: fulfill CR criteria except residual neutropenia <1.0*10^9/L/TTP <100*10^9/L; PR: all CR hematological values but with a decrease of >=50% in the percentage of blasts to 5% to 25% in the bone marrow aspirate. (NCT02610777)
Timeframe: From date of randomization until CR or PR (up to approximately 5 years)

Interventionmonths (Median)
Azacitidine 75 mg/m^213.2
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^28.3

[back to top]

Time to AML Transformation in HR MDS or CMML Participants

Time to AML transformation in HR MDS and CMML participants is defined as time from randomization to documented AML transformation. Participants without documented AML transformation at the time of the analysis are censored at the date of the last assessment. Participants who died before progression to AML are censored at the date of death. Transformation to AML is defined, according to World Health Organization (WHO) classification, as a participant having >20% blasts in the blood or marrow and increase of blast count by 50%. (NCT02610777)
Timeframe: From date of randomization until transformation to AML (up to approximately 5 years)

Interventionmonths (Median)
Azacitidine 75 mg/m^2NA
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^2NA

[back to top]

Six-month Survival Rate

Kaplan-Meier estimate of probability of OS at the end of the month 6 from randomization. (NCT02610777)
Timeframe: Month 6

Interventionsurvival probability (Number)
Azacitidine 75 mg/m^20.806
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^20.914

[back to top]

Percentage of Participants With Overall Response by Cycle 4

Disease responses (HR MDS/CMML): modified IWG criteria for MDS; low-blast (LB) AML: revised IWG criteria for AML. Overall response(HR MDS/CMML)=CR,PR/HI,LB AML=CR+ CR with incomplete blood count recovery(Cri)+PR.HR MDS/CMML-CR:<=5%myeloblasts with normal maturation of bone marrow (BM) cell lines,>=11g/dL Hb,>=100*10^9/L plt,>=1.0*10^9/L ANC,0% blasts in peripheral blood; PR:CR criteria met except BM blasts>=50%less over pretreatment but still>5%; HI:hb increase (inc)>=1.5g/dL if baseline<11g/dL;plt inc>=30*10^9/L if baseline>20*10^9/L/Inc. from <20*10^9/L->20*10^9/L, ANC inc. by 100%;absolute inc. of>0.5*10^9/L if baseline<100*10^9/L. LB AML-CR:morphologic leukemia-freestate>1.0*10^9ANC,>=100*10^9/L plt, transfusion independence, no residual evidence of extramedullary leukemia;CRi:fulfill CR criteria except residual neutropenia<1.0*10^9/L/TTP<100*10^9/L;PR:all CR hematological values but>=50%less in BM aspirate. The percentages are rounded off to report the nearest whole numbers. (NCT02610777)
Timeframe: From date of randomization until CR, PR or HI, by Cycle 4 (cycle length=28 days)

Interventionpercentage of participants (Number)
Azacitidine 75 mg/m^244.7
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^257.9

[back to top]

Percentage of Participants With Overall Response

Disease responses (HR MDS/CMML): modified IWG criteria for MDS; low-blast (LB) AML: revised IWG criteria for AML. Overall response(HR MDS/CMML)=CR,PR/HI,LB AML=CR+ CR with incomplete blood count recovery(Cri)+PR.HR MDS/CMML-CR:<=5%myeloblasts with normal maturation of bone marrow (BM) cell lines,>=11g/dL Hb,>=100*10^9/L plt,>=1.0*10^9/L ANC,0% blasts in peripheral blood; PR:CR criteria met except BM blasts>=50%less over pretreatment but still>5%; HI:hb increase (inc)>=1.5g/dL if baseline<11g/dL;plt inc>=30*10^9/L if baseline>20*10^9/L/Inc. from <20*10^9/L->20*10^9/L, ANC inc. by 100%;absolute inc. of>0.5*10^9/L if baseline<100*10^9/L. LB AML-CR:morphologic leukemia-freestate>1.0*10^9ANC,>=100*10^9/L plt, transfusion independence, no residual evidence of extramedullary leukemia;CRi:fulfill CR criteria except residual neutropenia<1.0*10^9/L/TTP<100*10^9/L;PR:all CR hematological values but>=50%less in BM aspirate. The percentages are rounded off to report the nearest whole numbers. (NCT02610777)
Timeframe: From date of randomization until CR, PR, or hematologic improvement (HI) (up to approximately 5 years)

Interventionpercentage of participants (Number)
Azacitidine 75 mg/m^262
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^271

[back to top]

Percentage of Participants With CR in Low-blast AML by Cycle 4

Disease response for low-blast AML is based on revised IWG response criteria for AML. CR for low-blast AML: morphologic leukemia-free state, ANC of more than 1.0*10^9/L and ptl of >=100*10^9/L, transfusion independence, no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery for low-blast AML: some participants fulfill all criteria for CR except for residual neutropenia (<1.0*10^9/L)/TTP (<100*10^9/L). The percentages are rounded off to report the nearest whole numbers. (NCT02610777)
Timeframe: From the date of randomization until CR by Cycle 4 (cycle length=28 days)

Interventionpercentage of participants (Number)
Azacitidine 75 mg/m^240.0
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^235.3

[back to top]

Percentage of Participants With CR in Low-blast AML

Disease response for low-blast AML is based on the revised IWG response criteria for AML. CR for low-blast AML: morphologic leukemia-free state, ANC of more than 1.0*10^9/L and plt of >=1.0*10^9/L, transfusion independence, and no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery for low-blast AML: some participants fulfill all of the criteria for CR except for residual neutropenia (<1.0*10^9/L) or thrombocytopenia (TTP) (<100*10^9/L). The percentages are rounded off to report the nearest whole numbers. (NCT02610777)
Timeframe: From date of randomization until CR (up to approximately 5 years)

Interventionpercentage of participants (Number)
Azacitidine 75 mg/m^260.0
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^241.2

[back to top]

Number of Participants in the Safety Analysis Population With Clinically Significant Change From Baseline in Electrocardiogram (ECG) Values Reported as TEAEs

ECG assessments included QT, QRS duration, PR interval, ventricular rate, QTcB, QTcF. (NCT02610777)
Timeframe: From date of first dose up to 30 days after administration of the last dose of any study drug (up to approximately 5 years)

,
InterventionParticipants (Count of Participants)
Atrial fibrillationTachycardia
Azacitidine 75 mg/m^241
Azacitidine 75 mg/m^2 + Pevonedistat 20 mg/m^244

[back to top]

Percentage of Participants With CR for Participants With MDS

CR was defined as participant with bone marrow: less than or equal to (<=) 5% myeloblasts with normal maturation of all cell lines; persistent dysplasia was noted peripheral blood: Hgb >=11 g/dL, platelets >=100*10^9/L, neutrophils >=1.0*10^9 per liter (/L), blasts 0%. CR assessment was based on IWG Response Criteria. (NCT02782468)
Timeframe: From first dose up to 30 days after last dose of study drug or before start of subsequent antineoplastic therapy, whichever comes earlier up to Cycle 19 (up to Day 429, single agent)(Cycle=21 days) and Cycle 65 (up to Day 1850, combination)(Cycle=28 days)

Interventionpercentage of participants (Number)
Single Agent Arm: Pevonedistat 44 mg/m^20
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^20
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^20

[back to top]

Overall Response Rate (ORR) for Participants With AML

ORR for AML was defined as the percentage of participants with a complete remission (CR), CR with incomplete blood count recovery (CRi), or partial remission (PR) based on Modified International Working Group (IWG) Response Criteria for AML. CR was defined as participant achieved the morphologic leukemia-free state and had an absolute neutrophil count (ANC) of more than 1,000 per microliter (/mcL) and platelets of greater than or equal to (>=) 100,000/mcL. A morphologic leukemia-free state requires less than (<) 5 percent (%) blasts in an aspirate sample with marrow spicules and with a count of at least 200 nucleated cells. CRi was, after chemotherapy, some participants who fulfilled all of the criteria for CR except for residual neutropenia (<1,000/mcL) or thrombocytopenia (<100,000/mcL). PR designation required all the hematologic values for a CR but with a decrease of at least 50% in the percentage of blasts to between 5% and 25% in the bone marrow aspirate. (NCT02782468)
Timeframe: From first dose up to 30 days after last dose of study drug or before start of subsequent antineoplastic therapy, whichever comes earlier up to Cycle 19 (up to Day 429, single agent)(Cycle=21 days) and Cycle 65 (up to Day 1850, combination)(Cycle=28 days)

Interventionpercentage of participants (Number)
Single Agent Arm: Pevonedistat 25 mg/m^20
Single Agent Arm: Pevonedistat 44 mg/m^20
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^250
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^280

[back to top]

Number of Participants Reporting One or More Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

(NCT02782468)
Timeframe: From first dose through 30 days after the last dose of study drug: single agent arms (up to Cycle 19 [up to Day 429]) (Cycle=21 days); combination arms (up to Cycle 65 [up to Day 1850]) (Cycle=28 days)

,,,
InterventionParticipants (Count of Participants)
TEAEsSAEs
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^233
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^2106
Single Agent Arm: Pevonedistat 25 mg/m^232
Single Agent Arm: Pevonedistat 44 mg/m^277

[back to top]

Percentage of Participants With CR for Participants With AML

CR was defined as participant achieved the morphologic leukemia-free state and had an ANC of more than 1,000/mcL and platelets of >=100,000/mcL. A morphologic leukemia-free state requires <5% blasts in an aspirate sample with marrow spicules and with a count of at least 200 nucleated cells. CR assessment was based on IWG Response Criteria. (NCT02782468)
Timeframe: From first dose up to 30 days after last dose of study drug or before start of subsequent antineoplastic therapy, whichever comes earlier up to Cycle 19 (up to Day 429, single agent)(Cycle=21 days) and Cycle 65 (up to Day 1850, combination)(Cycle=28 days)

Interventionpercentage of participants (Number)
Single Agent Arm: Pevonedistat 25 mg/m^20
Single Agent Arm: Pevonedistat 44 mg/m^20
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^250
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^20

[back to top]

ORR for Participants With MDS

ORR for MDS: Percentage of participants with CR,PR or hematologic improvement(HI) based on IWG Response Criteria. CR: BM:<=5% myeloblasts with normal maturation of all cell lines; persistent dysplasia noted peripheral blood: hemoglobin (Hgb)>=11 gram per deciliter (g/dL),platelets>=100*10^9/L,neutrophils >=1.0*10^9/L, blasts 0%. PR:CR criteria if abnormal before treatment except: BM blasts decreased by >=50% from pretreatment but still>5%; cellularity, morphology not relevant. HI criteria: Erythroid response:Hgb up by >=1.5 g/dL; transfused RBC reduced by at least 4 red blood cell (RBC) transfusions/8 weeks comparatively last 8 weeks; Platelet response: Increase of >=30*10^9/L for participants starting with >20*10^9/L platelets; increase from <20*10^9/L to >20*10^9/L and by 100%; Neutrophil response: At least 100% and absolute increase >0.5*10^9/L; Progression/relapse after HI: Granulocytes/platelets decreased by 50% from maximum; Hgb reduced by >=1.5 g/dL; transfusion dependence. (NCT02782468)
Timeframe: From first dose up to 30 days after last dose of study drug or before start of subsequent antineoplastic therapy, whichever comes earlier up to Cycle 19 (up to Day 429, single agent)(Cycle=21 days) and Cycle 65 (up to Day 1850, combination)(Cycle=28 days)

Interventionpercentage of participants (Number)
Single Agent Arm: Pevonedistat 44 mg/m^20
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^2100
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^20

[back to top]

Number of Participants With Dose Limiting Toxicities (DLTs) Based on National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) Version 4.03 During Cycle 1

DLT: Any of following events considered possibly related to study drug(s) by investigator: Grade (G) 3 or greater nausea and/or emesis despite use of optimal antiemetic prophylaxis; G3 diarrhea occurred despite maximal supportive therapy; G3 arthralgia/myalgia despite use of optimal analgesia; G3 nonhematologic toxicity with exceptions: 1) Brief fatigue, 2) hypophosphatemia; Persistent elevations of transaminases/bilirubin above G2 beyond 2 days between doses; Other study drug-related nonhematologic toxicities G2 or greater, required a dose reduction/discontinuation of pevonedistat. G3 or greater hematologic toxicities, including G3 or 4 febrile neutropenia, considered DLTs if: A delay in initiation of Cycle 2 due to lack of adequate recovery from treatment-related toxicity: 1) Of more than (>) 4 weeks due to hematologic toxicity believed not related to leukemic infiltration. Bone marrow (BM) evaluation may have been required. 2) Of >2 weeks due to nonhematologic toxicities. (NCT02782468)
Timeframe: Cycle 1 (Cycle length = 21 days [single agent arms]; 28 days [combination arms])

InterventionParticipants (Count of Participants)
Single Agent Arm: Pevonedistat 25 mg/m^20
Single Agent Arm: Pevonedistat 44 mg/m^20
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^20
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^21

[back to top]

Number of Participants With Clinically Significant Abnormal Laboratory Values

(NCT02782468)
Timeframe: Baseline up to Cycle 19 (up to Day 399) in single agent arms (Cycle=21days) and Cycle 65 (up to Day 1820) in combination arms (Cycle=28 days)

InterventionParticipants (Count of Participants)
Single Agent Arm: Pevonedistat 25 mg/m^20
Single Agent Arm: Pevonedistat 44 mg/m^20
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^20
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^20

[back to top]

Cmax: Maximum Observed Plasma Concentration for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 5

(NCT02782468)
Timeframe: Cycle 1 Day 5: pre-infusion and at multiple time points (up to 48 hours) post-infusion (Cycle length = 21 days [single agent arms]; 28 days [combination arms])

Interventionng/mL (Geometric Mean)
Single Agent Arm: Pevonedistat 25 mg/m^2252
Single Agent Arm: Pevonedistat 44 mg/m^2453
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^276.1
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^2170

[back to top]

Cmax: Maximum Observed Plasma Concentration for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 1

(NCT02782468)
Timeframe: Cycle 1 Day 1: pre-infusion and at multiple time points (up to 48 hours) post-infusion (Cycle length = 21 days [single agent arms]; 28 days [combination arms])

Interventionnanogram per milliliter (ng/mL) (Geometric Mean)
Single Agent Arm: Pevonedistat 25 mg/m^2253
Single Agent Arm: Pevonedistat 44 mg/m^2410
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^267.0
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^2160

[back to top]

CL: Total Clearance for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 5

(NCT02782468)
Timeframe: Cycle 1 Day 5: pre-infusion and at multiple time points (up to 48 hours) post-infusion (Cycle length = 21 days [single agent arms]; 28 days [combination arms])

Interventionliter per hour (Geometric Mean)
Single Agent Arm: Pevonedistat 25 mg/m^224.0
Single Agent Arm: Pevonedistat 44 mg/m^234.8
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^228.1
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^228.9

[back to top]

CL: Total Clearance for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 1

(NCT02782468)
Timeframe: Cycle 1 Day 1: pre-infusion and at multiple time points (up to 48 hours) post-infusion (Cycle length = 21 days [single agent arms]; 28 days [combination arms])

Interventionliter per hour (Geometric Mean)
Single Agent Arm: Pevonedistat 25 mg/m^224.1
Single Agent Arm: Pevonedistat 44 mg/m^232.6
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^222.8
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^228.4

[back to top]

AUC(0-tau): Area Under the Plasma Concentration-time Curve From Time 0 to Time Tau Over the Dosing Interval for Pevonedistat When Administered Alone and in Combination With Azacitidine on Cycle 1 Day 5

(NCT02782468)
Timeframe: Cycle 1 Day 5: pre-infusion and at multiple time points (up to 48 hours) post-infusion (Cycle length = 21 days [single agent arms]; 28 days [combination arms])

Interventionhour*nanogram per milliliter (h*ng/mL) (Geometric Mean)
Single Agent Arm: Pevonedistat 25 mg/m^21515
Single Agent Arm: Pevonedistat 44 mg/m^22163
Combination Arm: Pevonedistat 10 mg/m^2+ Azacitidine 75 mg/m^2647
Combination Arm: Pevonedistat 20 mg/m^2+ Azacitidine 75 mg/m^21224

[back to top]

Part A: Aefeces,14C: Cumulative Amount of [14C]-Pevonedistat Excreted in Feces up to the Last Sampling Interval

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionmcg eq (Mean)
Part A: [14C]-Pevonedistat 25 mg/m^225029.45

[back to top]

Part A: Aetotal,14C: Total Cumulative Excretion of [14C]-Pevonedistat From the Body

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionmcg eq (Mean)
Part A: [14C]-Pevonedistat 25 mg/m^244690.50

[back to top]

Part A: Aeurine,14C: Cumulative Amount of [14C]-Pevonedistat Excreted in Urine up to the Last Sampling Interval

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionmicrogram equivalent (mcg eq) (Mean)
Part A: [14C]-Pevonedistat 25 mg/m^219661.05

[back to top]

Part A: Aeurine: Cumulative Amount of Pevonedistat Dose Excreted in Urine at 144-168 Hours Post-dose

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionmicrogram (mcg) (Mean)
Part A: [14C]-Pevonedistat 25 mg/m^21161.47

[back to top]

Part A: Feurine: Cumulative Percentage of Pevonedistat Dose Excreted in Urine at 144-168 Hours Post-dose

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionpercentage of dose (Mean)
Part A: [14C]-Pevonedistat 25 mg/m^22.45

[back to top]

Part A: Renal Clearance (CLR) for Pevonedistat

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionliter per hour (L/hr) (Mean)
Part A: [14C]-Pevonedistat 25 mg/m^20.8343

[back to top]

Number of Participants Reporting One or More Treatment-emergent Adverse Events (TEAEs) and Serious Adverse Events (SAEs)

(NCT03057366)
Timeframe: Part A: From first dose of study drug in Part A up to Day 31; Part B: From first dose of study drug in Part B up to Cycle 11 Day 35 (Cycle length is equal to [=] 21 days)

,,
InterventionParticipants (Count of Participants)
TEAEsSAEs
Part A: [14C]-Pevonedistat 25 mg/m^241
Part B: Pevonedistat + Docetaxel21
Part B: Pevonedistat + Paclitaxel and Carboplatin52

[back to top]

Part A: AUClast: Area Under the Plasma and Whole Blood Concentration-time Curve From Time 0 to Time of the Last Quantifiable Concentration for Pevonedistat

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionhour*nanogram per milliliter (hr*ng/mL) (Mean)
PlasmaWhole Blood
Part A: [14C]-Pevonedistat 25 mg/m^21446.959284.0

[back to top] [back to top]

Part A: Cmax: Maximum Observed Plasma and Whole Blood Concentration for Pevonedistat

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionnanogram per milliliter (ng/mL) (Mean)
PlasmaWhole blood
Part A: [14C]-Pevonedistat 25 mg/m^2234.36862.9

[back to top] [back to top]

Part A: Percent Distribution of Total Radioactivity (TRA) for Pevonedistat and Its Metabolites in Plasma, Urine and Feces

(NCT03057366)
Timeframe: Up to 168 hours post-dose

Interventionpercentage distribution of TRA (Mean)
Pevonedistat in PlasmaMetabolite-1 in PlasmaMetabolite-2 in PlasmaMetabolite-3 in PlasmaMetabolite-7 in PlasmaMetabolite-10b in PlasmaMetabolite-16 in PlasmaMetabolite-22 in PlasmaPevonedistat in UrineMetabolite-1 in UrineMetabolite-2 in UrineMetabolite-3 in UrineMetabolite-7 in UrineMetabolite-10b in UrineMetabolite-16 in UrineMetabolite-22 in UrineMetabolite-23 in UrineMetabolite-24 in UrinePevonedistat in FecesMetabolite-1 in FecesMetabolite-2 in FecesMetabolite-3 in FecesMetabolite-7 in Feces
Part A: [14C]-Pevonedistat 25 mg/m^249.314.621.54.50.86.51.01.910.546.519.19.91.83.56.11.30.81.730.314.134.020.31.3

[back to top]

Part A: Tmax: Time to Reach the Maximum Plasma and Whole Blood Concentration (Cmax) for Pevonedistat

(NCT03057366)
Timeframe: Day 1 pre-dose and at multiple time points (up to 168 hours) post-dose

Interventionhour (Median)
PlasmaWhole Blood
Part A: [14C]-Pevonedistat 25 mg/m^20.9801.000

[back to top] [back to top]

Part B: Number of Participants With Best Overall Response as Per Investigator's Assessment

The best overall response was defined as the participants with best response among complete response (CR) or partial response (PR) or stable disease (SD), or progressive disease (PD). It was assessed by the investigator according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. CR: disappearance of all target lesions. Any pathological lymph nodes must have reduction in short axis to less than (<) 10 millimeter (mm). PR: at least a 30 percent (%) decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters as the best overall response after randomization. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study. PD: at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). (NCT03057366)
Timeframe: Up to Cycle 11 (Cycle length =21 days)

,
InterventionParticipants (Count of Participants)
CRPRSDPD
Part B: Pevonedistat + Docetaxel0002
Part B: Pevonedistat + Paclitaxel and Carboplatin0023

[back to top]

The Number of Patients Who Achieve Stable Disease

"Stable disease rate will be reported as the count and proportion of patients who achieve stable disease.~Stable Disease (SD) is defined as neither sufficient shrinkage to qualify for Partial Response (PR) nor sufficient increase to qualify for Progressive Disease (PD), taking as reference the smallest sum LD since the treatment started." (NCT03228186)
Timeframe: Up to 2 Years

InterventionParticipants (Count of Participants)
Pevonedistat Plus Docetaxel12

[back to top]

Median Progression Free Survival Time

"Progression-free survival is defined as the duration of time from start of treatment to time of progression.~Progressive Disease is defined as at least a 20% increase in the sum of the LD of target lesions, taking as reference the smallest sum LD recorded since the treatment started, or the appearance of one or more new lesions." (NCT03228186)
Timeframe: Up to 2 Years

Interventiondays (Median)
Pevonedistat Plus Docetaxel124

[back to top]

Median Overall Survival Time

Overall survival is defined as the duration of time from start of treatment to time of passing. (NCT03228186)
Timeframe: Up to 2 Years

Interventiondays (Median)
Pevonedistat Plus Docetaxel397

[back to top]

The Number of Toxicities by System Organ Class

All recorded toxicities will be listed and tabulated by system organ class. The NCI CTCAE version 4.03 will be utilized for AE reporting. (NCT03228186)
Timeframe: up to 30 days post last study drug dose, patients were allowed to stay on study drug treatment until progression. Data was collected over 3.5 years.

Interventionpercentage of Grade 3-5 Adverse Events (Number)
Blood and Lymphatic system disorders- grade 3Gastrointestinal disorders- grade 3General disorders and administration site conditions- grade 3hepatobiliary disorders- grade 3infections and infestations- grade 3infections and infestations- grade 4investigations- grade 3investigations- grade 4metabolism and nutrition disorders- grade 3nervous system disorders- grade 3respiratory, thoracic and mediastinal disorders- grade 3Skin and subcutaneous tissue disorders- grade 3
Pevonedistat Plus Docetaxel106.7103.33.33.316.713.33.33.33.33.3

[back to top]

The Percentage of Patients That Respond to Treatment

"Response is defined as either Partial Response or Complete Response. Partial Response is defined as at least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD. There can be no appearance of new lesions.~Complete Response is defined as the disappearance of all target lesions, determined by two separate observations conducted not less than 4 weeks apart. There can be no appearance of new lesions." (NCT03228186)
Timeframe: Up to 2 Years

Interventionpercent of participants (Number)
Pevonedistat Plus Docetaxel22

[back to top]

Time to Progression

Progression-free survival time is defined as time from day1 in cycle 1 to progression or death. Those alive without progression at the last follow up were censored. This survival will be summarized using the method of Kaplan and Meier. (NCT03238248)
Timeframe: Up to 24 months, or death

Interventionmonths (Median)
Pevonedistat and Azacitidine9.9

[back to top]

Rate of Marrow Complete Response (mCR)

Will assess morphologic features (e.g. presence of dysplastic features), presence of cytogenetic and/or molecular aberrancies, and myeloblast count. mCR is defined as the percentage of patients with only marrow CR among all patients evaluated. (NCT03238248)
Timeframe: Up to 24 months.

Interventionpercentage of participants (Number)
Pevonedistat and Azacitidine11.9

[back to top]

Rate of Hematologic Response Per IWG

Will assess complete and differential blood counts (peripheral blood), and transfusion requirements. The rate of hematologic improvement is defined as the percentage of patients with only hematologic improvement (defined in the protocol) among all patients evaluated for response. (NCT03238248)
Timeframe: Up to 24 months

Interventionpercentage of participants (Number)
Pevonedistat and Azacitidine7.5

[back to top]

Overall Survial Time

Overall survival time is defined as time from day 1 in cycle 1 to death for any reason. Patiens alive at last follow up were censored. Overall survival time will be summarized using the method of Kaplan and Meier. (NCT03238248)
Timeframe: up to 2 years after treatment, a maximum possible duration of 38 months.

Interventionmonths (Median)
Pevonedistat and Azacitidine18.3

[back to top]

Objective Response Rate

Objective response rate is defined as the percentage of patients with best response of Complete remission (CR), Hematologic improvement (HI), Marrow CR (mCR), or Partial remission (PR) among all patients evaluated. The other categories of response are progressive disease, stable disease and inevaluable. The modified recommendations of the International Working Group (IWG) for Response Criteria for altering natural history of MDS , as well as the International Consortium of clinical experts in MDS/MPN devised uniform response criteria and criteria for disease progression in MDS/MPN overlap syndromes based on three independent academic MDS/MPN workshops (Marsh 2013, December 2013 and June 2014) were used for response assessment. (NCT03238248)
Timeframe: Up to 12 months

Interventionpercentage of participants (Number)
Pevonedistat and Azacitidine23.9

[back to top]

Number of Participants With CR and Marrow CR and PR

Disease responses for HR MDS/CMML based on Modified IWG Response Criteria for MDS. CR: <=5% myeloblasts with normal maturation of all bone marrow cell lines, >=11 g/dL Hgb, >=100*10^9/L pl, >=1.0*10^9/L neutrophils,0% blasts in peripheral blood. Marrow CR: Bone marrow: <=5% myeloblasts and decrease by >=50% over pretreatment. PR: all CR criteria met except bone marrow blasts >=50% decrease over pretreatment but still >5%. (NCT03268954)
Timeframe: From randomization until CR or Marrow CR and PR till data cut-off date: 28 May 2021 (up to approximately 42 months)

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^291
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^287

[back to top]

Number of Participants With CR and Marrow CR

Disease responses for HR MDS or CMML are based on the International Working Group (IWG) Response Criteria for MDS. CR for HR MDS or CMML is defined as <=5% myeloblasts with normal maturation of all cell lines in the bone marrow, and >=11 g/dL Hgb, >=100*10^9/L platelets (pl), >=1.0*10^9/L neutrophils and 0% blasts in peripheral blood. Marrow CR: Bone marrow: <=5% myeloblasts and decrease by >=50% over pretreatment. (NCT03268954)
Timeframe: From randomization until CR or marrow CR till data cut-off date: 28 May 2021 (up to approximately 42 months)

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^291
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^287

[back to top]

Number of Participants With Complete Remission (CR) and CR+ Complete Remission With Incomplete Blood Count Recovery (CRi)

CR for HR MDS or CMML is defined as <=5% myeloblasts with normal maturation of all cell lines in the bone marrow, and greater than or equal to >=11 gram per deciliter (g/dL) hemoglobin (Hgb),>=100*10^9/liter (/L) platelets (pl),>=1.0*10^9/L neutrophils and 0% blasts in peripheral blood. CR for low-blast AML: morphologic leukemia-free state, neutrophils of more than 1.0*10^9/L and pl of >=100*10^9/L, transfusion independence, and no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery (CRi) for low-blast AML: participants fulfill all of the criteria for CR except for residual neutropenia (<1.0*10^9/L) or thrombocytopenia (pl<100*10^9/L). (NCT03268954)
Timeframe: From randomization until CR till data cut-off date: 28 May 2021 (up to approximately 42 months)

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^271
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^263

[back to top] [back to top]

Thirty-Day Mortality Reported as Number of Participants Who Died Up to Day 30

30-day mortality was defined as number of participants who died within 30 days from the first dose of study drug. (NCT03268954)
Timeframe: Up to Day 30

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^26
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^25

[back to top]

Kaplan-Meier Estimates of Six-Month Survival Rate

Kaplan-Meier estimates for the probability (expressed as a percentage) of participants that survived at the end of Month 6 from randomization are presented. (NCT03268954)
Timeframe: Up to Month 6

Interventionpercentage probability (Number)
Azacitidine 75 mg/m^20.825
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^20.810

[back to top]

Kaplan-Meier Estimates of One-Year Survival Rate

Kaplan-Meier estimates for the probability (expressed as a percentage) of participants that survived at the end of the first year from randomization are presented. (NCT03268954)
Timeframe: Up to Year 1

Interventionpercentage probability (Number)
Azacitidine 75 mg/m^20.693
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^20.646

[back to top]

Event-Free Survival in Participants Who Have TP53 Mutations, 17p Deletions, and/or Are Determined to be in an Adverse Cytogenetic Risk Group

Event was defined as death or transformation to AML in participants with MDS or CMML, whichever occurred first. Transformation to AML was defined, according to World Health Organization (WHO) Classification as a participant having >20% blasts in the blood or marrow and increase of blast count by 50%. Event was defined as death in participants with low-blast AML. (NCT03268954)
Timeframe: From randomization until transformation to AML if eligible or death till data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^212.8
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^214.6

[back to top]

Number of Participants With CR and Marrow CR, PR and Hematologic Improvement (HI)

Disease responses for HR MDS/CMML based on Modified IWG Response Criteria for MDS. CR: <=5% myeloblasts with normal maturation of all bone marrow cell lines, >=11 g/dL Hgb, >=100*10^9/L pl, >=1.0*10^9/L neutrophils,0% blasts in peripheral blood. Marrow CR: Bone marrow: <=5% myeloblasts and decrease by >=50% over pretreatment. PR: all CR criteria met except bone marrow blasts >=50% decrease over pretreatment but still>5%. HI: Hgb increase >=1.5 g/dL if baseline <11 g/dL; pl increase >=30*10^9/L if baseline>20*10^9/L or increases from <20*10^9/L to >20*10^9/L and by at least 100%; neutrophil increases by 100% and absolute increases of >0.5*10^9/L if baseline <1.0*10^9/L. (NCT03268954)
Timeframe: From randomization until CR, marrow CR, PR or HI till data cut-off date: 28 May 2021 (up to approximately 42 months)

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^2112
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^2117

[back to top]

Duration of Overall Response 2 (OR2)

Duration of OR2: from date of first documentation of CR+PR+HI to first documentation of PD/relapse after CR/PR for responders of CR+PR+HI for HR MDS/CMML and CR,CRi,PR for low-blast AML. For HR MDS/CMML-CR:<=5% myeloblasts with normal maturation of all bone marrow cell lines,>=11 g/dL Hgb, >=100*10^9/L pl,>=1.0*10^9/L neutrophils, 0% blasts in peripheral blood; PR: all CR criteria met except bone marrow blasts>=50% decrease over pretreatment, still >5%; HI:Hgb inc >=1.5 g/dL if baseline <11 g/dL; pl inc>=30*10^9/L if baseline>20*10^9/L or inc from<20*10^9/L to>20*10^9/L by at least 100%; neutrophil inc by 100% and absolute inc of >0.5*10^9/L if baseline <1.0*10^9/L.For low-blast AML-CR: morphologic leukemia-free state,>1.0*10^9 neutrophils,>=100*10^9/L pl, transfusion independence, no residual evidence of extramedullary leukemia;CRi: fulfill CR criteria except residual neutropenia<1.0*10^9/L or pl <100*10^9/L;PR:all CR hematological values but>=50% decrease in bone marrow aspirate. (NCT03268954)
Timeframe: Up to data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^218.3
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^218.9

[back to top]

Duration of Overall Response (OR)

Duration of OR: response to first documentation of PD or relapse from CR for low-blast AML or relapse after CR or PR for HR MDS/CMML. Disease responses for HR MDS/CMML based on Modified IWG Response Criteria for MDS; for low-blast AML on Revised IWG Response Criteria for AML. Overall response=CR+PR for HR MDS/CMML and CR+Cri+ PR for low-blast AML.CR for HR MDS/CMML:<=5% myeloblasts with normal maturation of all bone marrow cell lines,>=11 g/dL Hgb,>=100*10^9/L pl,>=1.0*10^9/L neutrophils,0% blasts in peripheral blood and PR:all CR criteria met except bone marrow blasts>=50% decrease over pretreatment but still >5%. For low-blast AML-CR: morphologic leukemia-free state >1.0*10^9 neutrophils,>=100*10^9/L pl, transfusion independence, no residual evidence of extramedullary leukemia; CR with CRi: fulfill CR criteria except residual neutropenia <1.0*10^9/L or pl <100*10^9/L; PR: all CR hematological values but >=50% decrease in % of blasts in bone marrow aspirate. (NCT03268954)
Timeframe: Up to data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^218.3
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^217.1

[back to top]

Duration of Complete Remission + Complete Remission With Incomplete Blood Count Recovery (CRi)

Duration of CR is first documented CR to the first documentation of PD or relapse from CR (participants with low-blast AML). Disease responses for low-blast AML were based on the Revised IWG Response Criteria for AML. CR is defined as <=5% myeloblasts with normal maturation of all cell lines in the bone marrow, and greater than or equal to >=11 g/dL hemoglobin (Hgb),>=100*10^9/liter (/L) platelets (pl),>=1.0*10^9/L neutrophils and 0% blasts in peripheral blood. CR for low-blast AML: morphologic leukemia-free state, neutrophils of more than 1.0*10^9/L and pl of >=100*10^9/L, transfusion independence, and no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery (CRi) for low-blast AML: participants fulfill all of the criteria for CR except for residual neutropenia (<1.0*10^9/L) or thrombocytopenia (pl<100*10^9/L). (NCT03268954)
Timeframe: From CR until first documentation of PD or relapse from CR or relapse after CR or PR till data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^28.5
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^215.0

[back to top]

Duration of Complete Remission (CR)

Duration of CR is first documented CR to the first documentation of PD or relapse from CR (participants with low-blast AML) or relapse after CR or PR (participants with HR MDS/CMML). Disease responses for HR MDS or CMML are based on the Modified IWG Response Criteria for MDS and for low-blast AML on the Revised IWG Response Criteria for AML. CR for HR MDS or CMML is defined as <=5% myeloblasts with normal maturation of all cell lines in the bone marrow, and greater than or equal to >=11 g/dL Hgb,>=100*10^9/L pl,>=1.0*10^9/L neutrophils and 0% blasts in peripheral blood. CR for low-blast AML: morphologic leukemia-free state, neutrophils of more than 1.0*10^9/L and pl of >=100*10^9/L, transfusion independence, and no residual evidence of extramedullary leukemia. CR with incomplete blood count recovery (CRi) for low-blast AML: participants fulfill all of the criteria for CR except for residual neutropenia (<1.0*10^9/L) or thrombocytopenia (pl<100*10^9/L). (NCT03268954)
Timeframe: From CR until first documentation of PD or relapse from CR or relapse after CR or PR till data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^213.1
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^217.2

[back to top]

Percentage of Participants With Red Blood Cells (RBCs) and Platelet-transfusion Independence

A participant was defined as RBC or platelet-transfusion independent if he/she received no RBC or platelet transfusions for a period of at least 8 weeks before the first dose of study drug through 30 days after the last dose of any study drug. Rate of transfusion independence was defined as number of participants who became transfusion independent divided by the number of participants who were transfusion dependent at Baseline. (NCT03268954)
Timeframe: Up to data cut-off date: 28 May 2021 (up to approximately 42 months)

,
Interventionpercentage of participants (Number)
Percentage of Participants With RBCs-transfusion IndependencePercentage of Participants With Platelet-transfusion Independence
Azacitidine 75 mg/m^244.348.9
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^247.545.5

[back to top]

Number of Participants With Overall Response in Participants Who Have TP53 Mutations, 17p Deletions, and/or Are Determined to be in an Adverse Cytogenetic Risk Group

Disease responses for HR MDS/CMML based on Modified IWG Response Criteria for MDS; for low-blast AML on Revised IWG Response Criteria for AML. Overall response=CR + PR for HR MDS/CMML and CR + CRi + PR for low-blast AML. CR for HR MDS/CMML: <=5% myeloblasts with normal maturation of all bone marrow cell lines, >=11 g/dL Hgb, >=100*10^9/L pl, >=1.0*10^9/L neutrophils,0% blasts in peripheral blood and PR: all CR criteria met except bone marrow blasts>=50% decrease over pretreatment but still>5%. For low-blast AML-CR: morphologic leukemia-free state, >1.0*10^9 neutrophils,>=100*10^9/L pl, transfusion independence, no residual evidence of extramedullary leukemia; CR with incomplete blood count recovery (CRi): fulfill CR criteria except residual neutropenia<1.0*10^9/L or pl<100*10^9/L; PR: all CR hematological values but>=50% decrease in the percentage of blasts to 5% to 25% in bone marrow aspirate. (NCT03268954)
Timeframe: From randomization until CR, CRi and PR till data cut-off date: 28 May 2021 (up to approximately 42 months)

,
InterventionParticipants (Count of Participants)
OR: HR MDS/CMML ParticipantsOR: Low-blast AML Participants
Azacitidine 75 mg/m^21413
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^21312

[back to top]

Event-Free Survival (EFS)

EFS was defined as the time from randomization to the date of an EFS event. An EFS event was defined as death or transformation to acute myelogenous leukemia (AML) (World Health Organization [WHO] classification as a participant having greater than 20 % blasts in the blood or marrow and an increase of blast count by 50%), whichever event occurred first, in participants with myelodysplastic syndromes (MDS) or chronic myelomonocytic leukemias (CMML). An EFS event was defined as death in participants with low-blast AML. (NCT03268954)
Timeframe: From randomization until transformation to acute myeloid leukemia, or death due to any cause up to data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^215.7
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^217.7

[back to top]

Number of Participants With Overall Response by Cycle 6

Responses for HR MDS/CMML are based on Modified International Working Group (IWG) Response Criteria for MDS and for low-blast AML on Revised IWG Response Criteria for AML. Overall response=CR and PR for HR MDS/CMML and CR+CR with incomplete blood count recovery(CRi)+PR for low-blast AML. CR for HR MDS/CMML:<=5% myeloblasts with normal maturation of all bone marrow cell lines,>=11g/dL hemoglobin (Hgb),>=100*10^9/L platelet (pl),>=1.0*10^9/L neutrophils, 0% blasts in peripheral blood, and PR:all CR criteria met except bone marrow blasts >=50% decrease over pretreatment but still >5%. For low-blast AML-CR:morphologic leukemia-free state,>1.0*10^9 neutrophils, >=100*10^9/L pl, transfusion independence, no residual evidence of extramedullary leukemia;CRi:fulfill CR criteria except residual neutropenia <1.0*10^9/L/thrombocytopenia (pl<100*10^9/L); PR:all CR hematological values but>=50% decrease in percentage of blasts to 5%-25% in bone marrow aspirate. (NCT03268954)
Timeframe: Up to Cycle 6 (up to approximately Day 168)

,
InterventionParticipants (Count of Participants)
Overall Response for HR MDS/CMMLOverall Response for Low-blast AML
Azacitidine 75 mg/m^23613
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^22922

[back to top] [back to top]

Duration of Red Blood Cells (RBCs) and Duration of Platelet-transfusion Independence and Duration of Red Blood Cells (RBCs) and Platelet-transfusion Independence

Duration of RBC and platelet transfusion independence was defined as the longest time between the last RBC and/or platelet transfusion before the start of the RBC and/or platelet transfusion-independent period and the first RBC and/or platelet transfusion after the start of the transfusion-independent period, which occurs >= 8 weeks later. (NCT03268954)
Timeframe: Up to data cut-off date: 28 May 2021 (up to approximately 42 months)

,
Interventionmonths (Median)
Duration of RBC Transfusion IndependenceDuration of Platelet Transfusion IndependenceDuration of Transfusion Independence
Azacitidine 75 mg/m^215.614.912.0
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^213.511.613.5

[back to top]

Time to Progressive Disease (PD), Relapse After CR (Low-blast AML), Relapse After CR or PR (HR MDS/CMML), or Death

Time to PD, relapse after CR(low-blast AML), relapse after CR or PR(HR MDS/CMML), or death,defined as time from date of randomization until date of first documentation of PD,relapse after CR(low-blast AML),relapse after CR or PR(HR MDS/CMML),or death due to any cause, whichever occurs first. In HR MDS/CMML,PD: Participants with<5% blasts:>=50% inc >5% blasts, with 5%-9% blasts:>=50% inc>10% blasts, with 10%-19% blasts:>=50% inc >20% blasts,with 20%-30% blasts, at least 50% decrement from maximum remission/response in granulocytes or pl or reduction in Hgb by>=2 g/dL/new transfusion dependence.Relapse after CR or PR: return to pretreatment bone marrow blast %/Decrement of >=50% from maximum remission/response levels in granulocytes/pl/reduction in Hgb conc.>=1.5 g/dL/transfusion dependence. In AML,PD:>50% inc in bone marrow blasts to >30% blasts,>50% inc in circulating blasts to>30% blasts in peripheral blood, Development of extramedullary disease/new sites of extramedullary leukemia. (NCT03268954)
Timeframe: From randomization until PD, relapse after CR, or relapse after CR or PR, or death due to any cause, whichever occurs first till data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^211.8
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^213.1

[back to top]

Time to First Complete Remission (CR) or Partial Remission (PR) or Complete Remission With Incomplete Blood Count Recovery (CRi)

Time to first CR or PR is defined as the time from randomization to first documented CR or PR, whichever occurs first. Disease responses for HR MDS or CMML or low-blast AML cycle 6 are based on Modified IWG Response Criteria for MDS and for low-blast AML on Revised IWG Response Criteria for AML. For HR MDS or CMML-CR:<=5% myeloblasts with normal maturation of all bone marrow cell lines,>=11 g/dL Hgb,>=100*10^9/L pl,>=1.0*10^9/L neutrophils, 0% blasts in peripheral blood; PR: all CR criteria met except bone marrow blasts>=50% decrease over pretreatment but still>5%; For low-blast AML-CR: morphologic leukemia-free state,>1.0*10^9/L neutrophils, pl>=100*10^9/L, transfusion independence, no residual evidence of extramedullary leukemia; CR with incomplete blood count recovery: fulfill CR criteria except residual neutropenia<1.0*10^9/L or pl <100*10^9/L; PR: all CR hematological values but with a decrease of at least 50% in the percentage of blasts to 5% to 25% in the bone marrow aspirate. (NCT03268954)
Timeframe: From randomization until CR or PR till data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^2NA
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^2NA

[back to top]

Sixty-Day Mortality Reported as Number of Participants Who Died Up to Day 60

60-day mortality was defined as number of participants who died within 60 days from the first dose of study drug. (NCT03268954)
Timeframe: Up to Day 60

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^215
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^214

[back to top]

Overall Survival in Participants Who Have TP53 Mutations, 17p Deletions, and/or Are Determined to be in an Adverse Cytogenetic Risk Group

OS was calculated from date of randomization to the date of death due to any cause. Participants without documented death at the time of the analysis were censored as of the date the participant was last known to be alive. (NCT03268954)
Timeframe: From randomization until death till data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^212.8
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^216.1

[back to top]

Overall Survival (OS)

Overall survival was defined as the time from randomization to death from any cause. (NCT03268954)
Timeframe: Up to data cut-off date: 28 May 2021 (up to approximately 42 months)

Interventionmonths (Median)
Azacitidine 75 mg/m^216.8
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^220.3

[back to top]

Number of Participants With Overall Response 2 (OR2)

OR2=participant with best overall response of CR+PR+HI in HR MDS/CMML participants,or of CR+CRi+PR in low-blast AML participants.CR:≤5% myeloblasts with normal maturation of all bone marrow(BM)cell lines,≥11g/dL Hgb,≥100*10^9/L pl,≥1.0*10^9/L neutrophils,0% blasts in peripheral blood;PR:all CR criteria met except BM blasts ≥50% decrease over pretreatment but still >5%;HI:Hgb increase(inc) ≥1.5g/dL if baseline(BL)<11 g/dL;pl inc≥30*10^9/L if BL>20*10^9/L or inc from <20*10^9/L to >20*10^9/L and by 100%;neutrophil inc by 100%;absolute inc of >0.5*10^9/L if BL<100*10^9/L.For low-blast AML-CR:morphologic leukemia-free state >1.0*10^9 neutrophils,≥100*10^9/L pl,transfusion independence,no residual evidence of extramedullary leukemia;CR with incomplete blood count recovery:fulfill CR criteria except residual neutropenia <1.0*10^9/L or pl<100*10^9/L;PR:all CR hematological values but ≥50% decrease in BM aspirate.Number of responders determined by independent review committee(IRC) assessment. (NCT03268954)
Timeframe: From randomization until, CR, PR or HI or CR, CRi or PR till data cut-off date: 28 May 2021 (up to approximately 42 months)

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^294
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^294

[back to top]

Number of Participants With Overall Response (OR)

Disease responses for HR MDS/CMML based on Modified IWG Response Criteria for MDS; for low-blast AML on Revised IWG Response Criteria for AML. Overall response=CR or PR for HR MDS/CMML and CR + CRi + PR for low-blast AML. CR for HR MDS/CMML: <=5% myeloblasts with normal maturation of all bone marrow cell lines, >=11 g/dL Hgb, >=100*10^9/L pl, >=1.0*10^9/L neutrophils, 0% blasts in peripheral blood and PR: all CR criteria met except bone marrow blasts >=50% decrease over pretreatment but still>5%. For low-blast AML-CR:morphologic leukemia-free state>1.0*10^9 neutrophils, >=100*10^9/L pl, transfusion independence, no residual evidence of extramedullary leukemia; CR with incomplete blood count recovery (CRi): fulfill CR criteria except residual neutropenia <1.0*10^9/L or pl <100*10^9/L; PR: all CR hematological values but >=50% decrease in bone marrow aspirate. (NCT03268954)
Timeframe: From randomization until CR and PR or CR, CRi and PR till data cut-off date: 28 May 2021 (up to approximately 42 months)

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^273
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^264

[back to top]

Number of Participants With Hematologic Improvement (HI)

Disease responses for HR MDS/CMML based on Modified IWG Response Criteria for MDS. HI: Hgb increase >=1.5 g/dL if baseline <11 g/dL; pl increase >=30*10^9/L if baseline >20*10^9/L or increase from <20*10^9/L to >20*10^9/L by at least 100%; neutrophil increase by 100% and absolute increase of >0.5*10^9/L if baseline <1.0*10^9/L. (NCT03268954)
Timeframe: From randomization until HI till data cut-off date: 28 May 2021 (up to approximately 42 months)

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^276
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^270

[back to top]

Number of Participants With CR, Partial Remission (PR) and Hematologic Improvement (HI)

Disease responses for HR MDS/CMML based on Modified IWG Response Criteria for MDS. CR: <=5% myeloblasts with normal maturation of all bone marrow cell lines, >=11 g/dL Hgb, >=100*10^9/L pl, >=1.0*10^9/L neutrophils,0% blasts in peripheral blood. Marrow CR: Bone marrow: <=5% myeloblasts and decrease by >=50% over pretreatment. PR: all CR criteria met except bone marrow blasts >=50% decrease over pretreatment but still >5%. HI: Hgb increase >=1.5 g/dL if <11 g/dL; pl increase >=30*10^9/L if baseline>20*10^9/L or increase from <20*10^9/L to >20*10^9/L and by at least 100%; neutrophil increase by 100% and absolute increase of >0.5*10^9/L if baseline <1.0*10^9/L. (NCT03268954)
Timeframe: From randomization until, CR, PR or HI till data cut-off date: 28 May 2021 (up to approximately 42 months)

InterventionParticipants (Count of Participants)
Azacitidine 75 mg/m^277
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^276

[back to top]

Time to Acute Myelogenous Leukemia (AML) Transformation in Higher-Risk Myelodysplastic Syndromes (HR MDS), Higher-Risk Chronic Myelomonocytic Leukemias (HR CMML) and HR MDS/CMML Participants

Time to AML transformation in HR MDS and CMML participants was defined as time from randomization to documented AML transformation as determined by the independent review committee (IRC) assessment. Participants who died before progression to AML were censored. Transformation to AML was defined, according to WHO classification, as a participant having 20% blasts in the blood or marrow and increase of blast count by 50%. (NCT03268954)
Timeframe: From randomization until transformation to AML till data cut-off date: 28 May 2021 (up to approximately 42 months)

,
Interventionmonths (Median)
HR MDS ParticipantsHR CMML ParticipantsHR MDS/CMML Participants
Azacitidine 75 mg/m^235.6NA35.6
Pevonedistat 20 mg/m^2 + Azacitidine 75 mg/m^2NANANA

[back to top]

Number of Participants With Grade 3 or Greater Toxicities Associated With Pevonedistat in Combination With Irinotecan and Temozolomide

Frequency of patients who experience at least one grade 3 or higher toxicity that is at least possibly attributable to pevonedistat using the Common Terminology Criteria for Adverse Events version 5.0 by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 3 years 8 months

InterventionParticipants (Count of Participants)
Part A Dose Level 1: 15 mg/m^25
Part A Dose Level 2: 20 mg/m^24
Part A Dose Level 3: 25 mg/m^26
Part A Dose Level 4: 35 mg/m^22
Part A PK Dose Level 4: 35 mg/m^24

[back to top]

Anti-tumor Activity of Pevonedistat in Combination With Irinotecan and Temozolomide

Frequency of disease response (best overall response of partial or complete response) assessed per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR by dose level. (NCT03323034)
Timeframe: Up to 4 years

InterventionParticipants (Count of Participants)
Part A Dose Level 1: 15 mg/m^21
Part A Dose Level 2: 20 mg/m^21
Part A Dose Level 3: 25 mg/m^20
Part A Dose Level 4: 35 mg/m^20
Part A PK Dose Level 4: 35 mg/m^20

[back to top]

T Max of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (Range) for the time at which the maximum (peak) serum concentration occurs by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

Interventionhours (Median)
Part A Dose Level 1: 15 mg/m^21.
Part A Dose Level 2: 20 mg/m^21
Part A Dose Level 3: 25 mg/m^21
Part A Dose Level 4: 35 mg/m^21.1
Part A PK Dose Level 4: 35 mg/m^21.1

[back to top]

MTD/RP2D of Pevonedistat in Combination With Irinotecan and Temozolomide

The maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) will be the maximum dose at which fewer than one-third of patients experience dose limiting toxicities. (NCT03323034)
Timeframe: Up to 28 days

Interventionmg/m^2 (Number)
Treatment (Pevonedistat, Temozolomide, Irinotecan)35

[back to top]

Half-life of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (Range) for the time required for the serum concentration to fall to 50% of its starting dose by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

Interventionhours (Median)
Part A Dose Level 1: 15 mg/m^25.3
Part A Dose Level 2: 20 mg/m^25
Part A Dose Level 3: 25 mg/m^25.2
Part A Dose Level 4: 35 mg/m^24.7
Part A PK Dose Level 4: 35 mg/m^26.1

[back to top]

Clearance of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (Range) for the rate of elimination of the drug by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

InterventionL/hr/m^2 (Median)
Part A Dose Level 1: 15 mg/m^218.8
Part A Dose Level 2: 20 mg/m^220.8
Part A Dose Level 3: 25 mg/m^218.9
Part A Dose Level 4: 35 mg/m^217.5
Part A PK Dose Level 4: 35 mg/m^217.9

[back to top]

C Max of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (Range) for the maximum (peak) serum concentration by dose level stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

Interventionng/mL (Median)
Part A Dose Level 1: 15 mg/m^2156
Part A Dose Level 2: 20 mg/m^2218.5
Part A Dose Level 3: 25 mg/m^2303.5
Part A Dose Level 4: 35 mg/m^2419
Part A PK Dose Level 4: 35 mg/m^2363.8

[back to top]

AUC of Pevonedistat in Combination With Irinotecan and Temozolomide

Median (range) of the area under the drug concentration over time (pre-dose and then 0, 1, 2, 4, 6-8, and 24-hours post-dose infusion) curve stratified by study part and dose level. (NCT03323034)
Timeframe: Up to 10 days

Interventionhr*ng/mL (Median)
Part A Dose Level 1: 15 mg/m^2803
Part A Dose Level 2: 20 mg/m^2973.5
Part A Dose Level 3: 25 mg/m^21340
Part A Dose Level 4: 35 mg/m^22018.5
Part A PK Dose Level 4: 35 mg/m^21957

[back to top]

Part A: Change From Time-matched Baseline in QRS After Pevonedistat Administration

Change from time-matched baseline in QRS was assessed following a single intravenous dose administration of pevonedistat at 25 and 50 mg/m^2 and was analysed by dose. Some participants were treated with pevonedistat 25 mg/m^2 or 50 mg/m^2 on Day 1 while others received treatment on Day 8. ANOVA was used for the analysis. (NCT03330106)
Timeframe: Predose, and at multiple time points post dose up to 24 hours on Day 1 or Day 8 in Part A

,
Interventionmilliseconds (Least Squares Mean)
Predose1 Hour Postdose2 Hours Postdose3 Hours Postdose4 Hours Postdose6 Hours Postdose9 Hours Postdose11 Hours Postdose24 Hours Postdose
Pevonedistat 25 mg/m^2-0.4591.447-0.2120.138-1.068-0.192-0.791-0.551-0.946
Pevonedistat 50 mg/m^2-0.4820.850-0.0430.184-0.241-0.319-0.221-0.769-0.470

[back to top]

Part A: Change From Time-matched Baseline in PR After Pevonedistat Administration

Change from time-matched baseline in PR was assessed following a single intravenous dose administration of pevonedistat at 25 and 50 mg/m^2 and was analysed by dose. Some participants were treated with pevonedistat 25 mg/m^2 or 50 mg/m^2 on Day 1 while others received treatment on Day 8. ANOVA was used for the analysis. (NCT03330106)
Timeframe: Predose, and at multiple time points post dose up to 24 hours on Day 1 or Day 8 in Part A

,
Interventionmilliseconds (Least Squares Mean)
Predose1 Hour Postdose2 Hours Postdose3 Hours Postdose4 Hours Postdose6 Hours Postdose9 Hours Postdose11 Hours Postdose24 Hours Postdose
Pevonedistat 25 mg/m^2-2.8941.152-0.090-1.557-3.406-4.665-4.499-3.018-5.245
Pevonedistat 50 mg/m^2-1.2203.3601.514-0.167-1.802-4.463-3.608-2.333-6.499

[back to top]

Part A: Change From Time-matched Baseline in Individual Corrected QT Interval (QTcI) After Pevonedistat Administration

Change from time-matched baseline in QTcI was assessed following a single intravenous dose administration of pevonedistat at 25 and 50 mg/m^2 and was analysed by dose. Some participants were treated with pevonedistat 25 mg/m^2 or 50 mg/m^2 on Day 1 while others received treatment on Day 8. ANOVA was used for the analysis. (NCT03330106)
Timeframe: Predose, and at multiple time points up to 24 hours post dose on Day 1 or Day 8 in Part A

,
Interventionmilliseconds (Least Squares Mean)
Predose1 Hour Postdose2 Hours Postdose3 Hours Postdose4 Hours Postdose6 Hours Postdose9 Hours Postdose11 Hours Postdose24 Hours Postdose
Pevonedistat 25 mg/m^22.0993.5801.4773.0743.8162.143-1.4190.020-1.897
Pevonedistat 50 mg/m^2-1.884-0.206-0.0991.8771.3050.198-2.482-5.250-3.105

[back to top]

Part A: Change From Time-matched Baseline in Heart Rate (HR) After Pevonedistat Administration

Change from time-matched baseline in HR was assessed following a single intravenous dose administration of pevonedistat at 25 and 50 mg/m^2 and was analysed by dose. Some participants were treated with pevonedistat 25 mg/m^2 or 50 mg/m^2 on Day 1 while others received treatment on Day 8. ANOVA was used for the analysis. (NCT03330106)
Timeframe: Predose, and at multiple time points post dose up to 24 hours on Day 1 or Day 8 in Part A

,
Interventionbeats per minute (bpm) (Least Squares Mean)
Predose1 Hour Postdose2 Hours Postdose3 Hours Postdose4 Hours Postdose6 Hours Postdose9 Hours Postdose11 Hours Postdose24 Hours Postdose
Pevonedistat 25 mg/m^20.602-2.068-1.812-0.993-0.2792.7122.5452.7944.432
Pevonedistat 50 mg/m^20.981-1.595-0.5561.9292.7366.4117.1347.5117.609

[back to top]

Part A: Change From Time-matched Baseline in Fridericia-corrected QT Interval (QTcF) After Pevonedistat Administration

Change from time-matched baseline in QTcF was assessed following a single intravenous dose administration of pevonedistat at 25 and 50 mg/m^2 and was analysed by dose. Some participants were treated with pevonedistat 25 mg/m^2 or 50 mg/m^2 on Day 1 while others received treatment on Day 8. Data is reported at pre-dose and at multiple timepoints (1, 2, 3, 4, 6, 9, 11 and 24 hours) postdose up to Day 8 in Part A. Analysis of variance (ANOVA) was used for the analysis. (NCT03330106)
Timeframe: Baseline up to Day 8

,
Interventionmilliseconds (Least Squares Mean)
Predose1 Hour Postdose2 Hours Postdose3 Hours Postdose4 Hours Postdose6 Hours Postdose9 Hours Postdose11 Hours Postdose24 Hours Postdose
Pevonedistat 25 mg/m^21.3103.1800.8401.7542.5670.598-2.998-2.478-2.930
Pevonedistat 50 mg/m^2-2.231-0.639-2.127-1.184-1.127-3.465-6.898-8.638-7.653

[back to top]

Part B: Overall Response Rate (ORR)

Percentage of participants who achieve an overall response per investigator's assessment at end of treatment,according to Response Evaluation Criteria in Solid Tumor(RECIST),version 1.1 guideline. Complete response(CR):Disappearance of all target lesions.Any pathological lymph nodes(whether target and non target)must have reduction in short axis to <10 millimeter(mm).Partial Response(PR):atleast 30% decrease in sum of diameter of target lesions,taking as reference baseline sum of diameter.Stable Disease(SD):Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for disease progression(PD),taking as reference smallest sum of diameter; PD:atleast 20% increase in sum of diameter of target lesions,taking as reference,smallest sum on study(this includes baseline sum if that is smallest on study).In addition to relative increase of 20%,sum must also demonstrate an absolute increase of atleast 5 mm.Appearance of 1 or more new lesions is also considered progression. (NCT03330106)
Timeframe: Up to Cycle 45 (end of treatment) (Cycle length=21 days)

Interventionpercentage of participants (Number)
Part B: Pevonedistat 25 mg/m^2 + Docetaxel9.1
Part B: Pevonedistat 20 mg/m^2 + Carboplatin + Paclitaxel8.3

[back to top]

Part A: Terminal Phase Elimination Half-life (t1/2) for Pevonedistat

(NCT03330106)
Timeframe: Days 1 or 8 predose and at multiple time points (up to 24 hours) post dose in Part A

Interventionhours (h) (Mean)
Pevonedistat 25 mg/m^27.21
Pevonedistat 50 mg/m^26.73

[back to top]

Part A: Cmax: Maximum Observed Plasma Concentration for Pevonedistat

(NCT03330106)
Timeframe: Days 1 or 8 predose and at multiple time points (up to 24 hours) post dose in Part A

Interventionnanograms per milliliter (ng/mL) (Geometric Mean)
Pevonedistat 25 mg/m^2197
Pevonedistat 50 mg/m^2509

[back to top]

Part A: AUC(0-24): Area Under the Plasma Concentration-time Curve From Time 0 to 24 Hours Postdose for Pevonedistat

(NCT03330106)
Timeframe: Days 1 or 8 predose and at multiple time points (up to 24 hours) post dose in Part A

Interventionhour*nanogram per milliliter (h*ng/mL) (Geometric Mean)
Pevonedistat 25 mg/m^21190
Pevonedistat 50 mg/m^22510

[back to top]

Part A: Ratio of Area Under the Plasma Concentration-time Curve From Time 0 to Infinity (AUC∞) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)

The LS means ratio of Day 10 over Day 1 were calculated from a mixed-model analysis of variance model. (NCT03486314)
Timeframe: Days 1 and 10 pre-dose and at multiple time points (up to 48 hours) post-dose

Interventionratio (Least Squares Mean)
Part A: Pevonedistat 50 mg/m^2 + Rifampin 600 mg0.790

[back to top]

Part B: Number of Participants With Best Overall Response as Per Investigator's Assessment

Best overall response was defined as participants with best response among complete response (CR) or partial response (PR) or stable disease (SD), or progressive disease (PD). It was assessed by investigator according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. CR: disappearance of all target lesions. Any pathological lymph nodes (whether target and non-target) must have reduction in short axis to less than (<) 10 millimeter (mm). PR: at least 30 percent (%) decrease in sum of diameter of target lesions, taking as reference baseline sum of diameter. SD: neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference smallest sum of diameter. PD: at least 20% increase in sum of diameter of target lesions, taking as reference, smallest sum on study. (NCT03486314)
Timeframe: Up to Cycle 17 (end of treatment) (Cycle length =21 days)

,
InterventionParticipants (Count of Participants)
CRPRSDPD
Part B: Pevonedistat 20 mg/m^2 + Carboplatin AUC5 + Paclitaxel 175 mg/m^20133
Part B: Pevonedistat 25 mg/m^2 + Docetaxel 75 mg/m^20222

[back to top]

Part A: Total Clearance After Intravenous Administration (CL) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)

(NCT03486314)
Timeframe: Days 1 and 10 pre-dose and at multiple time points (up to 48 hours) post-dose

Interventionliter per hour (L/h) (Mean)
Pevonedistat without rifampin (Day 1)Pevonedistat with rifampin (Day 10)
Part A: Pevonedistat 50 mg/m^2 + Rifampin 600 mg35.2243.77

[back to top]

Part A: Volume of Distribution at Steady State After Intravenous Administration (Vss) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)

(NCT03486314)
Timeframe: Days 1 and 10 pre-dose and at multiple time points (up to 48 hours) post-dose

Interventionliter (Mean)
Pevonedistat without rifampin (Day 1)Pevonedistat with rifampin (Day 10)
Part A: Pevonedistat 50 mg/m^2 + Rifampin 600 mg312.82296.10

[back to top]

Part A: Terminal Disposition Phase Half-life (T1/2z) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)

(NCT03486314)
Timeframe: Days 1 and 10 pre-dose and at multiple time points (up to 48 hours) post-dose

Interventionhour (Mean)
Pevonedistat without rifampin (Day 1)Pevonedistat with rifampin (Day 10)
Part A: Pevonedistat 50 mg/m^2 + Rifampin 600 mg7.4425.708

[back to top]

Part A: Ratio of Maximum Observed Plasma Concentration (Cmax) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)

The Least square (LS) means ratio of Day 10 over Day 1 were calculated from a mixed-model analysis of variance model. (NCT03486314)
Timeframe: Days 1 and 10 pre-dose and at multiple time points (up to 48 hours) post-dose

Interventionratio (Least Squares Mean)
Part A: Pevonedistat 50 mg/m^2 + Rifampin 600 mg0.962

[back to top]

Part A: Ratio of Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration (AUClast) for Pevonedistat Without Rifampin (Day 1) and With Rifampin (Day 10)

The LS means ratio of Day 10 over Day 1 were calculated from a mixed-model analysis of variance model. (NCT03486314)
Timeframe: Days 1 and 10 pre-dose and at multiple time points (up to 48 hours) post-dose

Interventionratio (Least Squares Mean)
Part A: Pevonedistat 50 mg/m^2 + Rifampin 600 mg0.785

[back to top]

Number of Participants Experiencing a Dose Limiting Toxicity (Dose Escalation)

Number of participants experiencing a dose limiting toxicity to determine the maximum tolerated dose (MTD) (NCT03770260)
Timeframe: At Day 28

,,
InterventionParticipants (Count of Participants)
Experienced a Dose Limiting ToxicityDid Not Experience a Dose Limiting ToxicityWithdrew from Treatment
Dose Level 1030
Dose Level 2031
Dose Level 3010

[back to top]

Total Plasma Clearance of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS

Median (Range) of the total plasma clearance of MLN4924 (pevonedistat) added to the 3-drug backbone of azacitidine (aza), fludarabine, and cytarabine re-induction for pediatric patients with recurrent/refractory AML and MDS by dose level measured pre-dose, end of infusion, 4-6 hours, and 24 hours post-dose infusion on Days 1 and 5. (NCT03813147)
Timeframe: Up to 5 days

InterventionL/h/m^2 (Median)
Stratum 1 With 20 mg/m^220.1
PK With 20 mg/m^219.2

[back to top]

Area Under the Plasma Concentration Versus Time Curve of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS

Median (Range) of the area under the plasma concentration versus time curve of MLN4924 (pevonedistat) added to the 3-drug backbone of azacitidine (aza), fludarabine, and cytarabine re-induction for pediatric patients with recurrent/refractory AML and MDS by dose level measured pre-dose, end of infusion, 4-6 hours, and 24 hours post-dose infusion on Days 1 and 5. (NCT03813147)
Timeframe: Up to 5 days

Interventionhr*ng/mL (Median)
Stratum 1 With 20 mg/m^21004.9
PK With 20 mg/m^21047.4

[back to top]

Number of Participants With Dose Limiting Toxicities of MLN4924 (Pevonedistat)

Frequency of patients with dose limiting toxicity in the first cycle of MLN4924 (pevonedistat) added to the 3-drug backbone of azacitidine (aza), fludarabine, and cytarabine re-induction for pediatric patients with recurrent/refractory AML and MDS by dose level among patients in the dose escalation cohort. (NCT03813147)
Timeframe: Up to 35 days

InterventionParticipants (Count of Participants)
Stratum 1 With 20 mg/m^21
PK With 20 mg/m^22

[back to top]

Elimination Half-life of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS

Median (Range) of the elimination half-life of MLN4924 (pevonedistat) added to the 3-drug backbone of azacitidine (aza), fludarabine, and cytarabine re-induction for pediatric patients with recurrent/refractory AML and MDS by dose level measured pre-dose, end of infusion, 4-6 hours, and 24 hours post-dose infusion on Days 1 and 5. (NCT03813147)
Timeframe: Up to 5 days

InterventionHour (Median)
Stratum 1 With 20 mg/m^24.7
PK With 20 mg/m^25.5

[back to top]

Maximum Concentration of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS

Median (Range) of the maximum concentration of MLN4924 (pevonedistat) added to the 3-drug backbone of azacitidine (aza), fludarabine, and cytarabine re-induction for pediatric patients with recurrent/refractory AML and MDS by dose level measured pre-dose, end of infusion, 4-6 hours, and 24 hours post-dose infusion on Days 1 and 5. (NCT03813147)
Timeframe: Up to 5 days

Interventionng/mL (Median)
Stratum 1 With 20 mg/m^2248.5
PK With 20 mg/m^2213

[back to top]

Maximum Time to Concentration of MLN4924 (Pevonedistat) Added to the 3-drug Backbone of Azacitidine (Aza), Fludarabine, and Cytarabine Re-induction for Pediatric Patients With Recurrent/Refractory AML and MDS

Median (Range) of the maximum time to concentration of MLN4924 (pevonedistat) added to the 3-drug backbone of azacitidine (aza), fludarabine, and cytarabine re-induction for pediatric patients with recurrent/refractory AML and MDS by dose level measured pre-dose, end of infusion, 4-6 hours, and 24 hours post-dose infusion on Days 1 and 5. (NCT03813147)
Timeframe: Up to 5 days

InterventionHour (Median)
Stratum 1 With 20 mg/m^21.2
PK With 20 mg/m^21.1

[back to top]

Number of Participants With Antitumor Activity of MLN4924 (Pevonedistat)

Frequency of participants with best overall response by dose level of PR or CR for MLN4924 (pevonedistat) added to the 3-drug backbone of azacitidine (aza), fludarabine, and cytarabine re-induction Per Response Evaluation Criteria for CR (M1 bone marrow (< 5% blasts) with no evidence of circulating blasts or extramedullary disease and with recovery of peripheral blood counts (ANC > 1000/uL and platelet count > 100,000/uL), CRp (M1 bone marrow (< 5% blasts) and no evidence of circulating blasts or extramedullary disease and with recovery of ANC > 1000/uL and platelet transfusion independence), CRi (M1 bone marrow (<5% blasts) and no evidence of circulating blasts or extramedullary disease and with ANC < 1000/uL or platelet count < 100,000/uL without platelet transfusion independence), or PR (M2 marrow status (> 5% or < 25% blasts cells) and at least 50% decrease in bone marrow blast percent from baseline. (NCT03813147)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Stratum 1 With 20 mg/m^21
PK With 20 mg/m^22

[back to top]

Number of Participants With Adverse Events Attributable to MLN4924 (Pevonedistat)

Frequency of patients with at least one grade 3 adverse event at least possibly attributable to MLN4924 (pevonedistat) added to the 3-drug backbone of azacitidine (aza), fludarabine, and cytarabine re-induction for pediatric patients with recurrent/refractory AML and MDS by dose level. (NCT03813147)
Timeframe: Up to 70 days

InterventionParticipants (Count of Participants)
Stratum 1 With 20 mg/m^26
PK With 20 mg/m^26

[back to top]

Duration of Progression Free Survival (PFS)

Progression free survival (PFS) is defined at the time all participants completed treatment and using Response Evaluation Criteria in Solid Tumors (RECIST v1.1) as >= 20% increase in the sum of target lesions, a measurable increase in a non-target lesion, or the appearance of a new lesion. The duration is measured from the start of treatment to the time of progression or death, whichever occurs first. (NCT03965689)
Timeframe: Up to 2.5 years

Interventionmonths (Median)
Treatment (Paclitaxel, Carboplatin, Pevonedistat)3.68

[back to top]

Overall Survival (OS)

Defined as the duration of time from start of treatment to time of death or last known date alive at the time all participants completed treatment. (NCT03965689)
Timeframe: Up to 2.5 years

Interventionmonths (Median)
Treatment (Paclitaxel, Carboplatin, Pevonedistat)7.73

[back to top]

Event-Free Survival (EFS)

EFS was defined as time from study randomization to date of failure to achieve complete remission (CR)/CR with incomplete blood count recovery (CRi), relapse from CR/CRi, or death. Assessments of disease response based on criteria: European Leukemia Net (ELN) 2017 guidelines. CR was defined as bone marrow blasts <5%; absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; absolute neutrophil count (ANC)≥1.0×10^9/L (1000/µL); platelet count≥100×10^9/L (100,000/µL). CRi was defined as all CR criteria except for residual neutropenia (<1.0×10^9/L [1000/µL]) or thrombocytopenia (<100×10^9/L [100,000/µL]). For participants who achieved CR/CRi, if relapse is not observed by time of analysis, was censored at the date of last disease assessment. If failed to achieve CR/CRi, date of treatment failure was set on day of randomization. (NCT04266795)
Timeframe: Up to 22 months

Interventionmonths (Median)
Venetoclax 100/200/400 mg + Azacitidine 75 mg/m^211.24
Pevonedistat 20 mg/m^2 + Venetoclax 100/200/400 mg + Azacitidine 75 mg/m^27.72

[back to top]