Page last updated: 2024-09-19

cytarabine

Description

Guanosine Diphosphate Fucose: A nucleoside diphosphate sugar formed from GDPmannose, which provides fucose for lipopolysaccharides of bacterial cell walls, and for blood group substances and other glycoproteins. [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

Cross-References

ID SourceID
PubMed CID135412609
CHEBI ID17009
SCHEMBL ID21905367
MeSH IDM0005564
PubMed CID6253
CHEMBL ID803
CHEBI ID28680
SCHEMBL ID3140
SCHEMBL ID23152019
SCHEMBL ID22591193
MeSH IDM0005564

Synonyms (233)

Synonym
CHEBI:17009 ,
guanosine 5'-[3-(6-deoxy-l-galactopyranosyl) dihydrogen diphosphate]
guanosine diphosphate fucose
gtpl4578
[(2r,3s,4r,5r)-5-(2-amino-6-oxo-3h-purin-9-yl)-3,4-dihydroxyoxolan-2-yl]methyl [hydroxy-[(3s,4r,5s,6s)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxyphosphoryl] hydrogen phosphate
{[(2r,3s,4r,5r)-5-(2-amino-6-oxo-6,9-dihydro-1h-purin-9-yl)-3,4-dihydroxyoxolan-2-yl]methoxy}({[hydroxy({[(3s,4r,5s,6s)-3,4,5-trihydroxy-6-methyloxan-2-yl]oxy})phosphoryl]oxy})phosphinic acid
(6-deoxy-beta-l-galactopyranosyl) ester
SCHEMBL21905367
Q27077785
guanosine 5'-diphospho- beta -l-fucose sodium salt
MLS001066340
BIDD:PXR0139
BIDD:GT0371
BRD-K33106058-003-20-6
SRI-10828-20
SRI-10828-19
LOPAC0_000316
citarabina
cytarabinum
4-amino-1-beta-d-arabinofuranosylpyrimidin-2(1h)-one
CHEBI:28680 ,
arabinocytosine
D00168
cytarabine (jp17/usp/inn)
depocyt (tn)
cytonal
cytosine arabinoside (van)
cytosar-u
4-amino-1-beta-d-arabinofuranosyl-2(1h)-pyrimidinon [czech]
4-amino-1-arabinofuranosyl-2-oxo-1,2-dihydropyrimidin [czech]
nsc 287459
citarabina [inn-spanish]
cytarabinum [inn-latin]
beta-ara c
4-amino-1-b-d-arabinofuranosyl-2-(1h)-pyrimidinone
einecs 205-705-9
cytosine 1-beta-d-arabinofuranoside
1-beta-d-arabinofuranosylcytosine, cytosine arabinoside
arabinoside, cytosine
arabinofuranosyl cytidine
spongocytidine
u-19,920
arabinocytidine
aracytidine
nsc-287459
cytarabinoside
alexan
tarabine
1-.beta.-d-arabinofuranosyl-cytosine
4-amino-1-[(2r,3s,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl]pyrimidin-2-one
udicil
arac
2(1h)-pyrimidinone, 4-amino-1-.beta.-d-arabinofuranosyl-
arabinoside c
depocyt
depocyte
aracytine
1beta-d-arabinosylcytosine
1-beta-d-arabinofuranosyl-4-amino-2(1h)pyrimidinone
1-beta-d-arabinofuranosylcytosine
hsdb 3049
beta-d-arabinosylcytosine
ai3-52329
cytosine beta-d-arabinoside
cytarabina
arafcyt
aracytin
arabitin
cytosine beta-d-arabinofuranoside
1beta-d-arabinofuranosylcytosine
2(1h)-pyrimidinone, 4-amino-1-beta-d-arabinofuranosyl-
cytosinearabinoside
1-beta-d-arabinofaranosylcytosine
u 19920a
erpalfa
ar3 ,
cytosine, 1-beta-d-arabinosyl-
4-amino-1-beta-d-arabinofuranosyl-2(1h)-pyrimidinone
1beta-arabinofuranasylcytosine
chx 3311
ccris 913
depocyt (liposomal)
cytosine arabinofuranoside
cytosine, 1-beta-d-arabinofuranosyl-
cytosine-1-beta-d-arabinofuranoside
147-94-4
ara-c
C02961
cytosine arabinoside
CYTARABINE ,
cytosine beta-d-arabinofuranoside, crystalline, >=90% (hplc)
SR-01000075773-3
MLS000758310
smr000449317
2(1h)-pyrimidinone, 4-amino-1-y-d-arabinofuranosyl- [cas]
DB00987
NCGC00093356-03
NCGC00093356-04
HMS2090A18
HMS2051K19
C2035
NCGC00093356-05
bdbm50087289
ara-cytidine
arabinosyl cytosine
u-19920
CHEMBL803 ,
cytarabine liposome
1-(arabinofuranosyl)cytosine
A808710
BRD-K33106058-001-07-7
NCGC00093356-06
AKOS007930145
4-amino-1-((2r,3s,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)tetrahydrofuran-2-yl)pyrimidin-2(1h)-one
dtxsid3022877 ,
dtxcid702877
tox21_301971
NCGC00255381-01
cas-147-94-4
MLS001424023
tox21_111203
HMS2230M16
CCG-51297
cytosine-beta-arabinoside
beta-cytosine arabinoside
cytosine, beta-d-arabinoside
cytarbel
cytarabine liposome injection
beta-arabinosylcytosine
mk 8242
4-amino-1-beta-d-arabinofuranosyl-2(1h)-pyrimidinon
cytarabine [usan:usp:inn:ban:jan]
unii-04079a1rdz
4-amino-1-arabinofuranosyl-2-oxo-1,2-dihydropyrimidin
04079a1rdz ,
cytartbine
SL-000002
NCGC00142483-02
AM84428
S1648
AKOS015896896
gtpl4827
4-amino-1-[(2r,3s,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-1,2-dihydropyrimidin-2-one
vyxeos component cytarabine
cytarabine [who-ip]
cytarabine [ema epar]
cytarabine [usp-rs]
cytarabine [inn]
cytarabine [ep monograph]
cytarabine [usp impurity]
cytarabine [usan]
cytarabinum [who-ip latin]
cytarabine [orange book]
cytarabine [mi]
cytarabine [usp monograph]
cytarabine liposome [vandf]
cytarabine [jan]
cytarabine [hsdb]
cytarabine [vandf]
cytarabine [mart.]
cytarabine [who-dd]
HY-13605
NC00070
SCHEMBL3140
KS-5063
J-700005
J-700166
ara-cell
1-(beta-d-arabinofuranosyl)cytosine
2(1h)-pyrimidinone, 4-amino-1beta-d-arabinofuranosyl-
1-beta-d-arabinofuranosyl cytosine
1-beta-d-arabinofuranosyl-cytosine
1-beta-arabinofuranosylcytosine
SRI-10828_24
J-520199
cytarabine, european pharmacopoeia (ep) reference standard
sr-01000721860
SR-01000721860-6
cytarabine, united states pharmacopeia (usp) reference standard
cytosine beta-d-arabinofuranoside, vetec(tm) reagent grade, 90%
cytarabine, pharmaceutical secondary standard; certified reference material
cytarabine; 4-amino-1-beta-d-arabinofuranosylpyrimidin-2(1h)-one
SR-01000075773-5
HMS3713N12
1-beta-d-arabinosyl-cytosine
cytosine, beta -d-arabinoside
beta -arabinosylcytosine
2(1h)-pyrimidinone, 4-amino-1beta -d-arabinofuranosyl-
cytosine-beta -d-arabinofuranoside
cytosine-beta -arabinoside
1-beta -d-arabinofaranosylcytosine
2(1h)-pyrimidinone, 4-amino-1- -d-arabinofuranosyl
beta -d-arabinosylcytosine
cytosine, 1-beta -d-arabinofuranosyl-
1beta -arabinofuranasylcytosine
1beta -d-arabinofuranosylcytosine
1-beta -d-arabinofuranosylcytosine
beta -cytosine arabinoside
2(1h)-pyrimidinone, 4-amino-1-beta -d-arabinofuranosyl-
1-beta -d-arabinofuranosyl-4-amino-2(1h)pyrimidinone
1beta -d-arabinosylcytosine
cytosine, 1-beta -d-arabinosyl-
SW197450-5
1-beta-d-arabinofuranosylcytosine; ara-c
Z1511499171
1-ss-d-arabinofuranosylcytosine
BCP02876
Q180983
SDCCGSBI-0050304.P002
NCGC00093356-19
SCHEMBL23152019
SCHEMBL22591193
cytosine -d-arabinofuranoside hydrochloride;cytosine arabinoside hydrochloride;ara-c hydrochloride
cytosine -d-arabinofuranoside;cytosine arabinoside;ara-c
BA164339
EN300-118320
l01bc01
arabine
citarabina (inn-spanish)
u 19,920a
cytarabine (usp impurity)
cytarabine (usp-rs)
beta-cytosine, arabinoside
1-beta-d-arabinosyl-4-amino-2(1h)pyrimidinone
1(beta-d-arabinofuranosyl)cytosine
cytarabinum (inn-latin)
u 19,920
cytarabine ocphosphate
cytarabine (usp monograph)
cytarabine (mart.)
cytarabine (usan:usp:inn:ban:jan)
wr-28453
cytarabine (ep monograph)

Roles (4)

RoleDescription
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
antimetaboliteA substance which is structurally similar to a metabolite but which competes with it or replaces it, and so prevents or reduces its normal utilization.
antiviral agentA substance that destroys or inhibits replication of viruses.
immunosuppressive agentAn agent that suppresses immune function by one of several mechanisms of action. Classical cytotoxic immunosuppressants act by inhibiting DNA synthesis. Others may act through activation of T-cells or by inhibiting the activation of helper cells. In addition, an immunosuppressive agent is a role played by a compound which is exhibited by a capability to diminish the extent and/or voracity of an immune response.
[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 (4)

ClassDescription
GDP-fucoseA GDP-hexose in which the hexose component is a fucosyl residue.
beta-D-arabinoside
pyrimidine nucleoside
monosaccharide derivativeA carbohydrate derivative that is formally obtained from a monosaccharide.
[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 (26)

cytarabine is involved in 26 pathway(s), involving a total of 5415 unique proteins and 1302 unique compounds

PathwayProteinsCompounds
Transport of small molecules39295
SLC-mediated transmembrane transport13567
Transport of vitamins, nucleosides, and related molecules2718
Transport of nucleotide sugars712
Signaling Pathways1269117
Signaling by NOTCH11314
Pre-NOTCH Expression and Processing2413
Pre-NOTCH Processing in the Endoplasmic Reticulum24
Metabolism of proteins1058144
Post-translational protein modification666112
Asparagine N-linked glycosylation16478
Biosynthesis of the N-glycan precursor (dolichol lipid-linked oligosaccharide, LLO) and transfer to a nascent protein5068
Synthesis of substrates in N-glycan biosythesis3756
GDP-fucose biosynthesis616
Disease1278231
Disorders of transmembrane transporters10243
SLC transporter disorders4537
Defective SLC35C1 causes congenital disorder of glycosylation 2C (CDG2C)01
colanic acid building blocks biosynthesis949
GDP-L-fucose biosynthesis I (from GDP-D-mannose)213
superpathway of GDP-mannose-derived O-antigen building blocks biosynthesis1239
GDP-L-galactose biosynthesis212
L-ascorbate biosynthesis I (L-galactose pathway)923
VTC2 cycle213
extended VTC2 cycle213
GDP-L-fucose biosynthesis I (from GDP-D-mannose)16
Xyloglucan biosynthesis18

Protein Targets (60)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Chain A, MAJOR APURINIC/APYRIMIDINIC ENDONUCLEASEHomo sapiens (human)Potency3.16230.003245.467312,589.2998AID2517
endonuclease IVEscherichia coliPotency7.94330.707912.432431.6228AID2565
glp-1 receptor, partialHomo sapiens (human)Potency1.25890.01846.806014.1254AID624417
thioredoxin reductaseRattus norvegicus (Norway rat)Potency0.89130.100020.879379.4328AID588453
phosphopantetheinyl transferaseBacillus subtilisPotency100.00000.141337.9142100.0000AID1490
RAR-related orphan receptor gammaMus musculus (house mouse)Potency7.69590.006038.004119,952.5996AID1159521; AID1159523
USP1 protein, partialHomo sapiens (human)Potency0.28180.031637.5844354.8130AID743255
TDP1 proteinHomo sapiens (human)Potency0.07280.000811.382244.6684AID686978; AID686979
AR proteinHomo sapiens (human)Potency11.92720.000221.22318,912.5098AID743035; AID743063
nuclear receptor subfamily 1, group I, member 3Homo sapiens (human)Potency0.09330.001022.650876.6163AID1224838; AID1224893
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency11.10440.01237.983543.2770AID1346984; AID1645841
EWS/FLI fusion proteinHomo sapiens (human)Potency0.46860.001310.157742.8575AID1259256
glucocorticoid receptor [Homo sapiens]Homo sapiens (human)Potency0.97700.000214.376460.0339AID720692
retinoic acid nuclear receptor alpha variant 1Homo sapiens (human)Potency14.56020.003041.611522,387.1992AID1159552; AID1159555
estrogen nuclear receptor alphaHomo sapiens (human)Potency9.57930.000229.305416,493.5996AID1259244; AID743069; AID743080
GVesicular stomatitis virusPotency23.91850.01238.964839.8107AID1645842
67.9K proteinVaccinia virusPotency0.44670.00018.4406100.0000AID720579
euchromatic histone-lysine N-methyltransferase 2Homo sapiens (human)Potency11.22020.035520.977089.1251AID504332
Histone H2A.xCricetulus griseus (Chinese hamster)Potency45.51300.039147.5451146.8240AID1224845; AID1224896
chromobox protein homolog 1Homo sapiens (human)Potency28.18380.006026.168889.1251AID488953
thyroid hormone receptor beta isoform aHomo sapiens (human)Potency2,238.72000.010039.53711,122.0200AID1479
nuclear factor erythroid 2-related factor 2 isoform 2Homo sapiens (human)Potency0.11580.00419.984825.9290AID504444
thyroid hormone receptor beta isoform 2Rattus norvegicus (Norway rat)Potency0.25780.000323.4451159.6830AID743065; AID743067
serine/threonine-protein kinase mTOR isoform 1Homo sapiens (human)Potency0.76990.00378.618923.2809AID2660; AID2667; AID2668
peptidyl-prolyl cis-trans isomerase NIMA-interacting 1Homo sapiens (human)Potency30.13130.425612.059128.1838AID504536
DNA polymerase iota isoform a (long)Homo sapiens (human)Potency89.12510.050127.073689.1251AID588590
gemininHomo sapiens (human)Potency9.81480.004611.374133.4983AID624296; AID624297
peripheral myelin protein 22Rattus norvegicus (Norway rat)Potency11.40510.005612.367736.1254AID624032; AID624044
lamin isoform A-delta10Homo sapiens (human)Potency0.03980.891312.067628.1838AID1487
Voltage-dependent calcium channel gamma-2 subunitMus musculus (house mouse)Potency21.68990.001557.789015,848.9004AID1259244
Interferon betaHomo sapiens (human)Potency23.91850.00339.158239.8107AID1645842
HLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)Potency23.91850.01238.964839.8107AID1645842
Cellular tumor antigen p53Homo sapiens (human)Potency0.42190.002319.595674.0614AID651631; AID720552
Glutamate receptor 2Rattus norvegicus (Norway rat)Potency21.68990.001551.739315,848.9004AID1259244
D(1A) dopamine receptorSus scrofa (pig)Potency0.92680.00378.108123.2809AID2667
Inositol hexakisphosphate kinase 1Homo sapiens (human)Potency23.91850.01238.964839.8107AID1645842
Ataxin-2Homo sapiens (human)Potency14.12540.011912.222168.7989AID588378
cytochrome P450 2C9, partialHomo sapiens (human)Potency23.91850.01238.964839.8107AID1645842
ATP-dependent phosphofructokinaseTrypanosoma brucei brucei TREU927Potency0.00760.060110.745337.9330AID485368
[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)
nuclear receptor coactivator 1 isoform 1 [Homo sapiens]Homo sapiens (human)IC50 (µMol)25.58701.15306.28039.9630AID602235
transactivating tegument protein VP16 [Human herpesvirus 1]Human alphaherpesvirus 1 (Herpes simplex virus type 1)IC50 (µMol)16.24700.94604.70169.4870AID602236
nuclear receptor coactivator 3 isoform aHomo sapiens (human)IC50 (µMol)7.53300.14764.33099.9200AID602234
ATP-binding cassette sub-family C member 3Homo sapiens (human)IC50 (µMol)133.00000.63154.45319.3000AID1473740
Multidrug resistance-associated protein 4Homo sapiens (human)IC50 (µMol)133.00000.20005.677410.0000AID1473741
Bile salt export pumpHomo sapiens (human)IC50 (µMol)133.00000.11007.190310.0000AID1473738
Thymidine kinase, cytosolicHomo sapiens (human)IC50 (µMol)1,000.00000.01601.21053.0000AID210895; AID306718
Thymidine kinaseHuman alphaherpesvirus 1 strain SC16IC50 (µMol)1,000.00000.10001.84917.9433AID210531
Cytochrome P450 3A4Homo sapiens (human)Ki190.00000.00011.41629.9000AID55393
Thymidine kinaseHuman herpesvirus 3 strain DumasIC50 (µMol)1,000.00003.20003.20003.2000AID210892; AID306722
Cytidine deaminaseHomo sapiens (human)Ki190.00000.00001.43332.3000AID55393
E3 ubiquitin-protein ligase Mdm2Homo sapiens (human)IC50 (µMol)0.02000.00060.358210.0000AID1422083
Enoyl-[acyl-carrier-protein] reductase [NADH] Francisella tularensis subsp. tularensis SCHU S4IC50 (µMol)59.00000.05001.91256.0000AID756998
Canalicular multispecific organic anion transporter 1Homo sapiens (human)IC50 (µMol)133.00002.41006.343310.0000AID1473739
Thymidine kinaseHuman alphaherpesvirus 1 (Herpes simplex virus type 1)IC50 (µMol)1,000.00000.15004.81679.0000AID306720
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Other Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
LMP1 [Human herpesvirus 4]human gammaherpesvirus 4 (Epstein-Barr virus)AbsAC35_uM7.24007.24007.24007.2400AID652043
LMP1 [Human herpesvirus 4]human gammaherpesvirus 4 (Epstein-Barr virus)AC500.91800.068039.9389277.4300AID504882; AID588398
Thymidylate synthaseMus musculus (house mouse)ED500.03000.03000.03000.0300AID212472
Deoxycytidine kinaseHomo sapiens (human)Km3.04000.40003.57908.5000AID1222351
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (304)

Processvia Protein(s)Taxonomy
nucleobase-containing compound metabolic processThymidine kinase 2, mitochondrialHomo sapiens (human)
deoxyribonucleoside monophosphate biosynthetic processThymidine kinase 2, mitochondrialHomo sapiens (human)
nucleotide biosynthetic processThymidine kinase 2, mitochondrialHomo sapiens (human)
pyrimidine nucleoside salvageThymidine kinase 2, mitochondrialHomo sapiens (human)
deoxycytidine metabolic processThymidine kinase 2, mitochondrialHomo sapiens (human)
thymidine metabolic processThymidine kinase 2, mitochondrialHomo sapiens (human)
DNA biosynthetic processThymidine kinase 2, mitochondrialHomo sapiens (human)
xenobiotic metabolic processATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
bile acid and bile salt transportATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transportATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
leukotriene transportATP-binding cassette sub-family C member 3Homo sapiens (human)
monoatomic anion transmembrane transportATP-binding cassette sub-family C member 3Homo sapiens (human)
transport across blood-brain barrierATP-binding cassette sub-family C member 3Homo sapiens (human)
prostaglandin secretionMultidrug resistance-associated protein 4Homo sapiens (human)
cilium assemblyMultidrug resistance-associated protein 4Homo sapiens (human)
platelet degranulationMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic metabolic processMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
bile acid and bile salt transportMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transportMultidrug resistance-associated protein 4Homo sapiens (human)
urate transportMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
cAMP transportMultidrug resistance-associated protein 4Homo sapiens (human)
leukotriene transportMultidrug resistance-associated protein 4Homo sapiens (human)
monoatomic anion transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
export across plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
transport across blood-brain barrierMultidrug resistance-associated protein 4Homo sapiens (human)
guanine nucleotide transmembrane transportMultidrug resistance-associated protein 4Homo sapiens (human)
fatty acid metabolic processBile salt export pumpHomo sapiens (human)
bile acid biosynthetic processBile salt export pumpHomo sapiens (human)
xenobiotic metabolic processBile salt export pumpHomo sapiens (human)
xenobiotic transmembrane transportBile salt export pumpHomo sapiens (human)
response to oxidative stressBile salt export pumpHomo sapiens (human)
bile acid metabolic processBile salt export pumpHomo sapiens (human)
response to organic cyclic compoundBile salt export pumpHomo sapiens (human)
bile acid and bile salt transportBile salt export pumpHomo sapiens (human)
canalicular bile acid transportBile salt export pumpHomo sapiens (human)
protein ubiquitinationBile salt export pumpHomo sapiens (human)
regulation of fatty acid beta-oxidationBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transportBile salt export pumpHomo sapiens (human)
bile acid signaling pathwayBile salt export pumpHomo sapiens (human)
cholesterol homeostasisBile salt export pumpHomo sapiens (human)
response to estrogenBile salt export pumpHomo sapiens (human)
response to ethanolBile salt export pumpHomo sapiens (human)
xenobiotic export from cellBile salt export pumpHomo sapiens (human)
lipid homeostasisBile salt export pumpHomo sapiens (human)
phospholipid homeostasisBile salt export pumpHomo sapiens (human)
positive regulation of bile acid secretionBile salt export pumpHomo sapiens (human)
regulation of bile acid metabolic processBile salt export pumpHomo sapiens (human)
transmembrane transportBile salt export pumpHomo 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)
nucleobase-containing compound metabolic processThymidine kinase, cytosolicHomo sapiens (human)
deoxyribonucleoside monophosphate biosynthetic processThymidine kinase, cytosolicHomo sapiens (human)
thymidine metabolic processThymidine kinase, cytosolicHomo sapiens (human)
thymidine biosynthetic processThymidine kinase, cytosolicHomo sapiens (human)
protein homotetramerizationThymidine kinase, cytosolicHomo sapiens (human)
DNA synthesis involved in mitotic DNA replicationThymidine kinase, cytosolicHomo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycle G2/M phase transitionCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
ER overload responseCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
mitophagyCellular tumor antigen p53Homo sapiens (human)
in utero embryonic developmentCellular tumor antigen p53Homo sapiens (human)
somitogenesisCellular tumor antigen p53Homo sapiens (human)
release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
hematopoietic progenitor cell differentiationCellular tumor antigen p53Homo sapiens (human)
T cell proliferation involved in immune responseCellular tumor antigen p53Homo sapiens (human)
B cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
T cell lineage commitmentCellular tumor antigen p53Homo sapiens (human)
response to ischemiaCellular tumor antigen p53Homo sapiens (human)
nucleotide-excision repairCellular tumor antigen p53Homo sapiens (human)
double-strand break repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
protein import into nucleusCellular tumor antigen p53Homo sapiens (human)
autophagyCellular tumor antigen p53Homo sapiens (human)
DNA damage responseCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrestCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in transcription of p21 class mediatorCellular tumor antigen p53Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
Ras protein signal transductionCellular tumor antigen p53Homo sapiens (human)
gastrulationCellular tumor antigen p53Homo sapiens (human)
neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of neuroblast proliferationCellular tumor antigen p53Homo sapiens (human)
protein localizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA replicationCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell population proliferationCellular tumor antigen p53Homo sapiens (human)
determination of adult lifespanCellular tumor antigen p53Homo sapiens (human)
mRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
rRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
response to salt stressCellular tumor antigen p53Homo sapiens (human)
response to inorganic substanceCellular tumor antigen p53Homo sapiens (human)
response to X-rayCellular tumor antigen p53Homo sapiens (human)
response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
positive regulation of gene expressionCellular tumor antigen p53Homo sapiens (human)
cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of cardiac muscle cell apoptotic processCellular tumor antigen p53Homo sapiens (human)
glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
viral processCellular tumor antigen p53Homo sapiens (human)
glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
cerebellum developmentCellular tumor antigen p53Homo sapiens (human)
negative regulation of cell growthCellular tumor antigen p53Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayCellular tumor antigen p53Homo sapiens (human)
mitotic G1 DNA damage checkpoint signalingCellular tumor antigen p53Homo sapiens (human)
negative regulation of telomere maintenance via telomeraseCellular tumor antigen p53Homo sapiens (human)
T cell differentiation in thymusCellular tumor antigen p53Homo sapiens (human)
tumor necrosis factor-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
regulation of tissue remodelingCellular tumor antigen p53Homo sapiens (human)
cellular response to UVCellular tumor antigen p53Homo sapiens (human)
multicellular organism growthCellular tumor antigen p53Homo sapiens (human)
positive regulation of mitochondrial membrane permeabilityCellular tumor antigen p53Homo sapiens (human)
cellular response to glucose starvationCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of apoptotic processCellular tumor antigen p53Homo sapiens (human)
entrainment of circadian clock by photoperiodCellular tumor antigen p53Homo sapiens (human)
mitochondrial DNA repairCellular tumor antigen p53Homo sapiens (human)
regulation of DNA damage response, signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
transcription initiation-coupled chromatin remodelingCellular tumor antigen p53Homo sapiens (human)
negative regulation of proteolysisCellular tumor antigen p53Homo sapiens (human)
negative regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of DNA-templated transcriptionCellular tumor antigen p53Homo sapiens (human)
positive regulation of RNA polymerase II transcription preinitiation complex assemblyCellular tumor antigen p53Homo sapiens (human)
positive regulation of transcription by RNA polymerase IICellular tumor antigen p53Homo sapiens (human)
response to antibioticCellular tumor antigen p53Homo sapiens (human)
fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
negative regulation of fibroblast proliferationCellular tumor antigen p53Homo sapiens (human)
circadian behaviorCellular tumor antigen p53Homo sapiens (human)
bone marrow developmentCellular tumor antigen p53Homo sapiens (human)
embryonic organ developmentCellular tumor antigen p53Homo sapiens (human)
positive regulation of peptidyl-tyrosine phosphorylationCellular tumor antigen p53Homo sapiens (human)
protein stabilizationCellular tumor antigen p53Homo sapiens (human)
negative regulation of helicase activityCellular tumor antigen p53Homo sapiens (human)
protein tetramerizationCellular tumor antigen p53Homo sapiens (human)
chromosome organizationCellular tumor antigen p53Homo sapiens (human)
neuron apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of cell cycleCellular tumor antigen p53Homo sapiens (human)
hematopoietic stem cell differentiationCellular tumor antigen p53Homo sapiens (human)
negative regulation of glial cell proliferationCellular tumor antigen p53Homo sapiens (human)
type II interferon-mediated signaling pathwayCellular tumor antigen p53Homo sapiens (human)
cardiac septum morphogenesisCellular tumor antigen p53Homo sapiens (human)
positive regulation of programmed necrotic cell deathCellular tumor antigen p53Homo sapiens (human)
protein-containing complex assemblyCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to endoplasmic reticulum stressCellular tumor antigen p53Homo sapiens (human)
thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of thymocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
necroptotic processCellular tumor antigen p53Homo sapiens (human)
cellular response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
cellular response to xenobiotic stimulusCellular tumor antigen p53Homo sapiens (human)
cellular response to ionizing radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to gamma radiationCellular tumor antigen p53Homo sapiens (human)
cellular response to UV-CCellular tumor antigen p53Homo sapiens (human)
stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
signal transduction by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
cellular response to actinomycin DCellular tumor antigen p53Homo sapiens (human)
positive regulation of release of cytochrome c from mitochondriaCellular tumor antigen p53Homo sapiens (human)
cellular senescenceCellular tumor antigen p53Homo sapiens (human)
replicative senescenceCellular tumor antigen p53Homo sapiens (human)
oxidative stress-induced premature senescenceCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
oligodendrocyte apoptotic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of execution phase of apoptosisCellular tumor antigen p53Homo sapiens (human)
negative regulation of mitophagyCellular tumor antigen p53Homo sapiens (human)
regulation of mitochondrial membrane permeability involved in apoptotic processCellular tumor antigen p53Homo sapiens (human)
regulation of intrinsic apoptotic signaling pathway by p53 class mediatorCellular tumor antigen p53Homo sapiens (human)
positive regulation of miRNA transcriptionCellular tumor antigen p53Homo sapiens (human)
negative regulation of G1 to G0 transitionCellular tumor antigen p53Homo sapiens (human)
negative regulation of miRNA processingCellular tumor antigen p53Homo sapiens (human)
negative regulation of glucose catabolic process to lactate via pyruvateCellular tumor antigen p53Homo sapiens (human)
negative regulation of pentose-phosphate shuntCellular tumor antigen p53Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to hypoxiaCellular tumor antigen p53Homo sapiens (human)
regulation of fibroblast apoptotic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
positive regulation of reactive oxygen species metabolic processCellular tumor antigen p53Homo sapiens (human)
negative regulation of stem cell proliferationCellular tumor antigen p53Homo sapiens (human)
positive regulation of cellular senescenceCellular tumor antigen p53Homo sapiens (human)
positive regulation of intrinsic apoptotic signaling pathwayCellular tumor antigen p53Homo sapiens (human)
lipid hydroxylationCytochrome P450 3A4Homo sapiens (human)
lipid metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid catabolic processCytochrome P450 3A4Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A4Homo sapiens (human)
steroid metabolic processCytochrome P450 3A4Homo sapiens (human)
cholesterol metabolic processCytochrome P450 3A4Homo sapiens (human)
androgen metabolic processCytochrome P450 3A4Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A4Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A4Homo sapiens (human)
monoterpenoid metabolic processCytochrome P450 3A4Homo sapiens (human)
calcitriol biosynthetic process from calciolCytochrome P450 3A4Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D metabolic processCytochrome P450 3A4Homo sapiens (human)
vitamin D catabolic processCytochrome P450 3A4Homo sapiens (human)
retinol metabolic processCytochrome P450 3A4Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A4Homo sapiens (human)
long-chain fatty acid biosynthetic processCytochrome P450 3A4Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A4Homo sapiens (human)
oxidative demethylationCytochrome P450 3A4Homo sapiens (human)
pyrimidine nucleotide metabolic processDeoxycytidine kinaseHomo sapiens (human)
CMP biosynthetic processDeoxycytidine kinaseHomo sapiens (human)
dAMP salvageDeoxycytidine kinaseHomo sapiens (human)
nucleoside phosphate biosynthetic processDeoxycytidine kinaseHomo sapiens (human)
cell surface receptor signaling pathwayCytidine deaminaseHomo sapiens (human)
pyrimidine-containing compound salvageCytidine deaminaseHomo sapiens (human)
cytidine deaminationCytidine deaminaseHomo sapiens (human)
cytosine metabolic processCytidine deaminaseHomo sapiens (human)
negative regulation of cell growthCytidine deaminaseHomo sapiens (human)
UMP salvageCytidine deaminaseHomo sapiens (human)
negative regulation of nucleotide metabolic processCytidine deaminaseHomo sapiens (human)
response to cycloheximideCytidine deaminaseHomo sapiens (human)
cellular response to external biotic stimulusCytidine deaminaseHomo sapiens (human)
regulation of cell cycleE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ubiquitin-dependent protein catabolic processE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein polyubiquitinationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
blood vessel developmentE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
blood vessel remodelingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
regulation of heart rateE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
atrioventricular valve morphogenesisE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
endocardial cushion morphogenesisE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ventricular septum developmentE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
atrial septum developmentE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ubiquitin-dependent protein catabolic processE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
apoptotic processE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrestE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
traversing start control point of mitotic cell cycleE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
positive regulation of cell population proliferationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to xenobiotic stimulusE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to toxic substanceE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to iron ionE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
positive regulation of gene expressionE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
negative regulation of protein processingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
negative regulation of neuron projection developmentE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein ubiquitinationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein sumoylationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein destabilizationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to magnesium ionE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein localization to nucleusE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
regulation of protein catabolic processE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to cocaineE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
negative regulation of DNA damage response, signal transduction by p53 class mediatorE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
establishment of protein localizationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to etherE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
negative regulation of DNA-templated transcriptionE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
positive regulation of mitotic cell cycleE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to antibioticE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
positive regulation of protein export from nucleusE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to steroid hormoneE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
positive regulation of muscle cell differentiationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
proteolysis involved in protein catabolic processE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein autoubiquitinationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cardiac septum morphogenesisE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein-containing complex assemblyE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to hydrogen peroxideE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to vitamin B1E3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to alkaloidE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to growth factor stimulusE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to peptide hormone stimulusE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to estrogen stimulusE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to hypoxiaE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to gamma radiationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to UV-CE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
fibroblast activationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cellular response to actinomycin DE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
negative regulation of signal transduction by p53 class mediatorE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathway by p53 class mediatorE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to formaldehydeE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
positive regulation of vascular associated smooth muscle cell proliferationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
positive regulation of vascular associated smooth muscle cell migrationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
amyloid fibril formationE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
response to water-immersion restraint stressE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
negative regulation of apoptotic processE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cell cycleE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
regulation of gene expressionE3 ubiquitin-protein ligase Mdm2Homo 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)
xenobiotic metabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
negative regulation of gene expressionCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bile acid and bile salt transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
heme catabolic processCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic export from cellCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transepithelial transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
leukotriene transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
monoatomic anion transmembrane transportCanalicular multispecific organic anion transporter 1Homo sapiens (human)
transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transport across blood-brain barrierCanalicular multispecific organic anion transporter 1Homo sapiens (human)
negative regulation of receptor internalizationAtaxin-2Homo sapiens (human)
regulation of translationAtaxin-2Homo sapiens (human)
RNA metabolic processAtaxin-2Homo sapiens (human)
P-body assemblyAtaxin-2Homo sapiens (human)
stress granule assemblyAtaxin-2Homo sapiens (human)
RNA transportAtaxin-2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (117)

Processvia Protein(s)Taxonomy
deoxycytidine kinase activityThymidine kinase 2, mitochondrialHomo sapiens (human)
thymidine kinase activityThymidine kinase 2, mitochondrialHomo sapiens (human)
ATP bindingThymidine kinase 2, mitochondrialHomo sapiens (human)
nucleoside kinase activityThymidine kinase 2, mitochondrialHomo sapiens (human)
deoxynucleoside kinase activityThymidine kinase 2, mitochondrialHomo sapiens (human)
ATP bindingATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type xenobiotic transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
glucuronoside transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type bile acid transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATP hydrolysis activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
xenobiotic transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
icosanoid transmembrane transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
ABC-type transporter activityATP-binding cassette sub-family C member 3Homo sapiens (human)
guanine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ATP bindingMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type xenobiotic transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
prostaglandin transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
urate transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
purine nucleotide transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type bile acid transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
efflux transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
15-hydroxyprostaglandin dehydrogenase (NAD+) activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATP hydrolysis activityMultidrug resistance-associated protein 4Homo sapiens (human)
glutathione transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
xenobiotic transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
ABC-type transporter activityMultidrug resistance-associated protein 4Homo sapiens (human)
protein bindingBile salt export pumpHomo sapiens (human)
ATP bindingBile salt export pumpHomo sapiens (human)
ABC-type xenobiotic transporter activityBile salt export pumpHomo sapiens (human)
bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
canalicular bile acid transmembrane transporter activityBile salt export pumpHomo sapiens (human)
carbohydrate transmembrane transporter activityBile salt export pumpHomo sapiens (human)
ABC-type bile acid transporter activityBile salt export pumpHomo sapiens (human)
ATP hydrolysis activityBile salt export pumpHomo 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)
thymidine kinase activityThymidine kinase, cytosolicHomo sapiens (human)
protein bindingThymidine kinase, cytosolicHomo sapiens (human)
ATP bindingThymidine kinase, cytosolicHomo sapiens (human)
zinc ion bindingThymidine kinase, cytosolicHomo sapiens (human)
identical protein bindingThymidine kinase, cytosolicHomo sapiens (human)
transcription cis-regulatory region bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
cis-regulatory region sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
core promoter sequence-specific DNA bindingCellular tumor antigen p53Homo sapiens (human)
TFIID-class transcription factor complex bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription repressor activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription activator activity, RNA polymerase II-specificCellular tumor antigen p53Homo sapiens (human)
protease bindingCellular tumor antigen p53Homo sapiens (human)
p53 bindingCellular tumor antigen p53Homo sapiens (human)
DNA bindingCellular tumor antigen p53Homo sapiens (human)
chromatin bindingCellular tumor antigen p53Homo sapiens (human)
DNA-binding transcription factor activityCellular tumor antigen p53Homo sapiens (human)
mRNA 3'-UTR bindingCellular tumor antigen p53Homo sapiens (human)
copper ion bindingCellular tumor antigen p53Homo sapiens (human)
protein bindingCellular tumor antigen p53Homo sapiens (human)
zinc ion bindingCellular tumor antigen p53Homo sapiens (human)
enzyme bindingCellular tumor antigen p53Homo sapiens (human)
receptor tyrosine kinase bindingCellular tumor antigen p53Homo sapiens (human)
ubiquitin protein ligase bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase regulator activityCellular tumor antigen p53Homo sapiens (human)
ATP-dependent DNA/DNA annealing activityCellular tumor antigen p53Homo sapiens (human)
identical protein bindingCellular tumor antigen p53Homo sapiens (human)
histone deacetylase bindingCellular tumor antigen p53Homo sapiens (human)
protein heterodimerization activityCellular tumor antigen p53Homo sapiens (human)
protein-folding chaperone bindingCellular tumor antigen p53Homo sapiens (human)
protein phosphatase 2A bindingCellular tumor antigen p53Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingCellular tumor antigen p53Homo sapiens (human)
14-3-3 protein bindingCellular tumor antigen p53Homo sapiens (human)
MDM2/MDM4 family protein bindingCellular tumor antigen p53Homo sapiens (human)
disordered domain specific bindingCellular tumor antigen p53Homo sapiens (human)
general transcription initiation factor bindingCellular tumor antigen p53Homo sapiens (human)
molecular function activator activityCellular tumor antigen p53Homo sapiens (human)
promoter-specific chromatin bindingCellular tumor antigen p53Homo sapiens (human)
monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
steroid bindingCytochrome P450 3A4Homo sapiens (human)
iron ion bindingCytochrome P450 3A4Homo sapiens (human)
protein bindingCytochrome P450 3A4Homo sapiens (human)
steroid hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A4Homo sapiens (human)
oxygen bindingCytochrome P450 3A4Homo sapiens (human)
enzyme bindingCytochrome P450 3A4Homo sapiens (human)
heme bindingCytochrome P450 3A4Homo sapiens (human)
vitamin D3 25-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
caffeine oxidase activityCytochrome P450 3A4Homo sapiens (human)
quinine 3-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1-alpha,25-dihydroxyvitamin D3 23-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 8,9 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 11,12 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
anandamide 14,15 epoxidase activityCytochrome P450 3A4Homo sapiens (human)
aromatase activityCytochrome P450 3A4Homo sapiens (human)
vitamin D 24-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
estrogen 2-hydroxylase activityCytochrome P450 3A4Homo sapiens (human)
1,8-cineole 2-exo-monooxygenase activityCytochrome P450 3A4Homo sapiens (human)
deoxyadenosine kinase activityDeoxycytidine kinaseHomo sapiens (human)
deoxycytidine kinase activityDeoxycytidine kinaseHomo sapiens (human)
deoxyguanosine kinase activityDeoxycytidine kinaseHomo sapiens (human)
ATP bindingDeoxycytidine kinaseHomo sapiens (human)
protein homodimerization activityDeoxycytidine kinaseHomo sapiens (human)
cytidine kinase activityDeoxycytidine kinaseHomo sapiens (human)
nucleoside bindingCytidine deaminaseHomo sapiens (human)
cytidine deaminase activityCytidine deaminaseHomo sapiens (human)
protein bindingCytidine deaminaseHomo sapiens (human)
zinc ion bindingCytidine deaminaseHomo sapiens (human)
identical protein bindingCytidine deaminaseHomo sapiens (human)
protein homodimerization activityCytidine deaminaseHomo sapiens (human)
p53 bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ubiquitin-protein transferase activityE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
5S rRNA bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
zinc ion bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ligase activityE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
SUMO transferase activityE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
enzyme bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein domain specific bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ubiquitin protein ligase bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
receptor serine/threonine kinase bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
identical protein bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
peroxisome proliferator activated receptor bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ribonucleoprotein complex bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ubiquitin bindingE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
ubiquitin protein ligase activityE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
NEDD8 ligase activityE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
disordered domain specific bindingE3 ubiquitin-protein ligase Mdm2Homo 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)
protein bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP bindingCanalicular multispecific organic anion transporter 1Homo sapiens (human)
organic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type xenobiotic transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
bilirubin transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type glutathione S-conjugate transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATP hydrolysis activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
xenobiotic transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ATPase-coupled inorganic anion transmembrane transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
ABC-type transporter activityCanalicular multispecific organic anion transporter 1Homo sapiens (human)
RNA bindingAtaxin-2Homo sapiens (human)
epidermal growth factor receptor bindingAtaxin-2Homo sapiens (human)
protein bindingAtaxin-2Homo sapiens (human)
mRNA bindingAtaxin-2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (55)

Processvia Protein(s)Taxonomy
mitochondrionThymidine kinase 2, mitochondrialHomo sapiens (human)
mitochondrial matrixThymidine kinase 2, mitochondrialHomo sapiens (human)
cytoplasmThymidine kinase 2, mitochondrialHomo sapiens (human)
plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basal plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
basolateral plasma membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
membraneATP-binding cassette sub-family C member 3Homo sapiens (human)
nucleolusMultidrug resistance-associated protein 4Homo sapiens (human)
Golgi apparatusMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
platelet dense granule membraneMultidrug resistance-associated protein 4Homo sapiens (human)
external side of apical plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
plasma membraneMultidrug resistance-associated protein 4Homo sapiens (human)
basolateral plasma membraneBile salt export pumpHomo sapiens (human)
Golgi membraneBile salt export pumpHomo sapiens (human)
endosomeBile salt export pumpHomo sapiens (human)
plasma membraneBile salt export pumpHomo sapiens (human)
cell surfaceBile salt export pumpHomo sapiens (human)
apical plasma membraneBile salt export pumpHomo sapiens (human)
intercellular canaliculusBile salt export pumpHomo sapiens (human)
intracellular canaliculusBile salt export pumpHomo sapiens (human)
recycling endosomeBile salt export pumpHomo sapiens (human)
recycling endosome membraneBile salt export pumpHomo sapiens (human)
extracellular exosomeBile salt export pumpHomo sapiens (human)
membraneBile salt export pumpHomo 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)
nucleusThymidine kinase, cytosolicHomo sapiens (human)
cytosolThymidine kinase, cytosolicHomo sapiens (human)
nuclear bodyCellular tumor antigen p53Homo sapiens (human)
nucleusCellular tumor antigen p53Homo sapiens (human)
nucleoplasmCellular tumor antigen p53Homo sapiens (human)
replication forkCellular tumor antigen p53Homo sapiens (human)
nucleolusCellular tumor antigen p53Homo sapiens (human)
cytoplasmCellular tumor antigen p53Homo sapiens (human)
mitochondrionCellular tumor antigen p53Homo sapiens (human)
mitochondrial matrixCellular tumor antigen p53Homo sapiens (human)
endoplasmic reticulumCellular tumor antigen p53Homo sapiens (human)
centrosomeCellular tumor antigen p53Homo sapiens (human)
cytosolCellular tumor antigen p53Homo sapiens (human)
nuclear matrixCellular tumor antigen p53Homo sapiens (human)
PML bodyCellular tumor antigen p53Homo sapiens (human)
transcription repressor complexCellular tumor antigen p53Homo sapiens (human)
site of double-strand breakCellular tumor antigen p53Homo sapiens (human)
germ cell nucleusCellular tumor antigen p53Homo sapiens (human)
chromatinCellular tumor antigen p53Homo sapiens (human)
transcription regulator complexCellular tumor antigen p53Homo sapiens (human)
protein-containing complexCellular tumor antigen p53Homo sapiens (human)
cytoplasmCytochrome P450 3A4Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A4Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A4Homo sapiens (human)
plasma membraneGlutamate receptor 2Rattus norvegicus (Norway rat)
nucleoplasmDeoxycytidine kinaseHomo sapiens (human)
cytosolDeoxycytidine kinaseHomo sapiens (human)
mitochondrionDeoxycytidine kinaseHomo sapiens (human)
cytoplasmDeoxycytidine kinaseHomo sapiens (human)
extracellular regionCytidine deaminaseHomo sapiens (human)
cytosolCytidine deaminaseHomo sapiens (human)
secretory granule lumenCytidine deaminaseHomo sapiens (human)
tertiary granule lumenCytidine deaminaseHomo sapiens (human)
ficolin-1-rich granule lumenCytidine deaminaseHomo sapiens (human)
cytosolCytidine deaminaseHomo sapiens (human)
nuclear bodyE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
nucleusE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
nucleoplasmE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
nucleolusE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cytoplasmE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
cytosolE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
plasma membraneE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
transcription repressor complexE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
endocytic vesicle membraneE3 ubiquitin-protein ligase Mdm2Homo sapiens (human)
protein-containing complexE3 ubiquitin-protein ligase Mdm2Homo 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)
plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cell surfaceCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
intercellular canaliculusCanalicular multispecific organic anion transporter 1Homo sapiens (human)
apical plasma membraneCanalicular multispecific organic anion transporter 1Homo sapiens (human)
cytoplasmAtaxin-2Homo sapiens (human)
Golgi apparatusAtaxin-2Homo sapiens (human)
trans-Golgi networkAtaxin-2Homo sapiens (human)
cytosolAtaxin-2Homo sapiens (human)
cytoplasmic stress granuleAtaxin-2Homo sapiens (human)
membraneAtaxin-2Homo sapiens (human)
perinuclear region of cytoplasmAtaxin-2Homo sapiens (human)
ribonucleoprotein complexAtaxin-2Homo sapiens (human)
cytoplasmic stress granuleAtaxin-2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (1002)

Assay IDTitleYearJournalArticle
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.
AID1159607Screen for inhibitors of RMI FANCM (MM2) intereaction2016Journal of biomolecular screening, Jul, Volume: 21, Issue:6
A High-Throughput Screening Strategy to Identify Protein-Protein Interaction Inhibitors That Block the Fanconi Anemia DNA Repair Pathway.
AID625282Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cirrhosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID100352Antitumor activity in mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 25 mg/kg/day x 5 was determined as weight change (g/mouse) on 8th day1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID124798Antitumor activity against L1210 tumor in mice, at a dose of 500 mg/kg peroral administration in the presence of 100 mg/kg of zebularine.1991Journal of medicinal chemistry, Nov, Volume: 34, Issue:11
Antitumor properties of 2(1H)-pyrimidinone riboside (zebularine) and its fluorinated analogues.
AID130778Antitumor activity against L1210/ara-C leukemia in mice (Percent increase in life span.) at the dose of 200 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID81283Anticancer activity of the compound against ara-C resistant promyelocytic leukemia cell line that is Cyd-kinase deficient (HL-60-HGPRT-/dCK-) is determined1986Journal of medicinal chemistry, Jul, Volume: 29, Issue:7
Synthesis and biological activity of 5-phenylselenenyl-substituted pyrimidine nucleosides.
AID338733Cytotoxicity against human KB cells after 3 days by trypan blue exclusion technique
AID200650In vitro cytotoxicity was evaluated againstCNS SNB-7 cells; 1 x 10E12000Journal of medicinal chemistry, Apr-20, Volume: 43, Issue:8
Synthesis and evaluation of several new (2-chloroethyl)nitrosocarbamates as potential anticancer agents.
AID1079946Presence of at least one case with successful reintroduction. [column 'REINT' in source]
AID338735Cytotoxicity against human HeLaS3 cells after 3 days by trypan blue exclusion technique
AID717966Antitumor activity against human BxPC3 cells xenografted in SCID mouse assessed as tumor growth inhibition at 10 mg/kg, ip administered 5 times a week for one cycle relative to control2012Journal of medicinal chemistry, Nov-26, Volume: 55, Issue:22
New adamantane phenylalkylamines with σ-receptor binding affinity and anticancer activity, associated with putative antagonism of neuropathic pain.
AID355584Antiproliferative activity against mouse P388 cells at 1 ug after 48 hrs by two-layer agar-diffusion method
AID1079938Chronic liver disease either proven histopathologically, or through a chonic elevation of serum amino-transferase activity after 6 months. Value is number of references indexed. [column 'CHRON' in source]
AID130780Antitumor activity against L1210/ara-C leukemia in mice (Percent increase in life span.) at the dose of 250 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1589124Selectivity index, ratio of IC50 for cytotoxicity in human NHDF cells to IC50 for cytotoxicity in human KB cells2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID451311Resistance ratio of IC50 for deoxynucleoside analogue-resistant mouse L1210 10K cells to IC50 for deoxynucleoside analogue-sensitive mouse L1210 cells2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID451294Cytotoxicity against deoxynucleoside analogue-sensitive human CCRF-CEM cells after 3 days by MTT assay2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID138050Survival time treated / survival time (T/C) for antitumor activity against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 200 mg/kg per day after treatment 1-5;16/71990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID475911Antitumor activity against human H460 cells after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Apr-01, Volume: 18, Issue:7
Synthesis, X-ray crystal structure study and antitumoral evaluations of 5,6-disubstituted pyrimidine derivatives.
AID130644Antitumor activity against L1210/ara-C leukemia in mice (Percent increase in life span.) at the dose of 1000 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID127729Average survival time includes only those mice that died prior to day 60 at 75 mg/kg1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID135726Toxicity in mice after 12-13 days at a dose of 100 mg/kg/day; 12/81988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1134736Toxicity in CDF1 mouse assessed as change in body weight at 20 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID389365Growth inhibition of human KB cells by MTT method2008Bioorganic & medicinal chemistry, Oct-15, Volume: 16, Issue:20
Antitumor studies. Part 5: Synthesis, antitumor activity, and molecular docking study of 5-(monosubstituted amino)-2-deoxo-2-phenyl-5-deazaflavins.
AID1135472Antiviral activity against Herpes simplex virus 1 Sheely assessed as inhibition of virus-induced cytopathic effect at 37 degC after 72 hrs by two-fold dilution method1977Journal of medicinal chemistry, Jun, Volume: 20, Issue:6
Antiviral activity of some beta-diketones. 1. Aryl alkyl diketones. In vitro activity against both RNA and DNA viruses.
AID496823Antimicrobial activity against Trichomonas vaginalis2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID138549Antitumor activity against L1210 leukemia in mice (Average 7-day weight change (%)) at the dose of 100 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1739160Antitumor activity against human MV4-11 cells xenografted in Balb/c nude mouse assessed as reduction in tumor volume at 40 mg/kg, iv administered once daily for 21 days (Rvb = 1.00 g)2020European journal of medicinal chemistry, Aug-15, Volume: 200Discovery of coumarin derivatives as potent and selective cyclin-dependent kinase 9 (CDK9) inhibitors with high antitumour activity.
AID1292259Cytotoxicity against human MCF7cells after 72 hrs by SRB assay2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID99173Cross resistance profile versus L1210/R71 cells.1990Journal of medicinal chemistry, Jul, Volume: 33, Issue:7
Structure-activity relationships of antineoplastic agents in multidrug resistance.
AID1135050Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as increase in life span at 50 mg/kg, ip qd administered for 5 days relative to control1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID1542690Induction of apoptosis in human U937 cells assessed as necrotic cells at 0.3 uM measured after 1 day by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 2.5 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID1222350Activity of human recombinant N-terminal His-tagged DCK Ala119Gly mutant expressed in Escherichia coli BL21(DE3) cells assessed as cytosine-beta-D-arabinofuranoside-5'-MP formation incubated for 3 mins by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID105668In vitro antiproliferative activity was measured in mouse myeloma MPC-11 cell line1982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID43716In vitro inhibitory activity against Human T-cell acute lymphoblastoid leukemia (CCRF CEM) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID138730Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change) at the dose of 1000 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID97223Antitumor activity against ip implanted L1210 leukemia in mice expressed as active dose range (1-5qd treatment schedule) required to produce increase in life span by >25% was determined; Range is 5-3501986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1135049Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as increase in life span at 25 mg/kg, ip qd administered for 5 days relative to control1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID98955Antitumor activity against the ic implanted L1210 lymphoid leukemia in mice expressed as optimal dose (1-9qd treatment schedule) required to produce greatest increase in life span was determined1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1135722Immunosuppressive activity against PHA-stimulated proliferation in rat T-lymphocytes assessed as cell viability at 0.01 to 0.1 uM after 6 days by [3H]thymidine incorporation method relative to control1978Journal of medicinal chemistry, Jul, Volume: 21, Issue:7
Synthesis of branched-chain apiosylpyrimidines and their inhibition of lymphocyte proliferation.
AID475909Antitumor activity against human SW620 cells after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Apr-01, Volume: 18, Issue:7
Synthesis, X-ray crystal structure study and antitumoral evaluations of 5,6-disubstituted pyrimidine derivatives.
AID44362Cytotoxicity against CCRF-CEM/ara-C human leukemic lymphoblastoid cell lines1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID116596Percent increase in life span calculated as (T/C-1)X100 at a dose of 20 mg/kg/day X51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID132045Increase of life span was determined against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 400 mg/kg per day after treatment 51990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID44009In vitro concentration required for 50% inhibition of growth of human leukemia cell line CCRF-CEM with hPAP (0.2 unit/mL)2001Bioorganic & medicinal chemistry letters, Nov-05, Volume: 11, Issue:21
Amino acid phosphoramidate nucleosides: potential ADEPT/GDEPT substrates.
AID229265Ratio of IC50 for Parental L1210 to that of multidrug resistant L1210.1991Journal of medicinal chemistry, Jul, Volume: 34, Issue:7
Additional nucleotide derivatives of mitosenes. Synthesis and activity against parental and multidrug resistant L1210 leukemia.
AID152827Antitumor activity was determined against the ip implanted P388 leukemia in mice by range in survival days at 500 mg/kg (treatment of 1qd schedule) dosage; Range is 14-151986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID154166Percent inhibition of [123I]UdR incorporation in the DNA synthesis assay in P388 cell lines at a concentration of 10 E-3 M1983Journal of medicinal chemistry, Nov, Volume: 26, Issue:11
Synthesis and biological evaluation of sparsomycin analogues.
AID133648Dose producing greatest increase in life span expressed as optimum dose (L1210/ara-C II) (qd 1-5)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1197746Stimulation of human OATP1B3-mediated [3H]CCK-8 at 100 uM after 5 mins relative to control2015European journal of medicinal chemistry, Mar-06, Volume: 92Interaction of human organic anion transporter polypeptides 1B1 and 1B3 with antineoplastic compounds.
AID1360495Growth inhibition of human SMMC7721 cells at 0.01 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID261688Activity against NCI60 cell line xenograft in nude mouse by HF assay2006Journal of medicinal chemistry, Mar-23, Volume: 49, Issue:6
Assessment of in vitro and in vivo activities in the National Cancer Institute's anticancer screen with respect to chemical structure, target specificity, and mechanism of action.
AID1222351Activity of human recombinant N-terminal His-tagged wild type DCK expressed in Escherichia coli BL21(DE3) cells assessed as cytosine-beta-D-arabinofuranoside-5'-MP formation incubated for 3 mins by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID353086Cytotoxicity against human SF268 cells by MTT assay2009Bioorganic & medicinal chemistry letters, May-01, Volume: 19, Issue:9
Synthesis of potent antitumor and antiviral benzofuran derivatives.
AID1123322Antiviral activity against Herpes simplex virus-1 KOS infected in human skin fibroblast cells assessed as inhibition of virus-induced cytopathogenicity treated immediately after virus adsorption measured 2 to 3 days after virus infection1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID427703Cytotoxicity against human SNU638 cells by sulforhodamine B method2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Discovery of a new template for anticancer agents: 2'-deoxy-2'-fluoro-4'-selenoarabinofuranosyl-cytosine (2'-F-4'-seleno-ara-C).
AID338736Cytotoxicity against human MB9812 cells after 3 days by trypan blue exclusion technique
AID654376Growth inhibition of HDF cells after 72 hrs by WST-1 assay2012Journal of natural products, Feb-24, Volume: 75, Issue:2
Pinophilins A and B, inhibitors of mammalian A-, B-, and Y-family DNA polymerases and human cancer cell proliferation.
AID118369Compound was tested for the ability to suppress humoral immune response in mice treated for days 0,+1,and +2 at dose of 25 mg/kg (control = 7.0)1981Journal of medicinal chemistry, Apr, Volume: 24, Issue:4
Synthesis and biological evaluation of certain 2'-deoxy-beta-D-ribo- and -beta-D-arabinofuranosyl nucleosides of purine-6-carboxamide and 4,8-diaminopyrimido[5,4-d]pyrimidine.
AID137885Ratio of survival time of treated to control mice at 50 mg/kg1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID1124452Antiviral activity against Herpes simplex virus type 1 infected in Swiss albino mouse assessed as increase in mouse survival at 0.14 mg/kg, ic single dose after 6 hrs of infection relative to control1979Journal of medicinal chemistry, May, Volume: 22, Issue:5
Synthesis and antitumor and antiviral activities of 1-beta-D-arabinofuranosyl-2-amino-1,4(2H)-iminopyrimidine and its derivatives.
AID1134740Toxicity in CDF1 mouse assessed as change in body weight at 5 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID152681Antitumor activity against the ip implanted P388 leukemia in mice expressed as optimal dose (1 qd treatment schedule) required to produce greatest increase in life span was determined1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1407817Antiproliferative activity against human Jurkat E6-1 cells2018European journal of medicinal chemistry, Sep-05, Volume: 157Pyrrolopyrimidines: An update on recent advancements in their medicinal attributes.
AID746834Therapeutic index, ratio of IC50 for human CD34-positive blood stem/progenitor cells to IC50 for human MA9.3 cells2013Bioorganic & medicinal chemistry letters, May-15, Volume: 23, Issue:10
Novel ROS-activated agents utilize a tethered amine to selectively target acute myeloid leukemia.
AID23702Partition coefficient (logP)1984Journal of medicinal chemistry, Mar, Volume: 27, Issue:3
Comparative structure-activity relationships of antifolate triazines inhibiting murine tumor cells sensitive and resistant to methotrexate.
AID42743In vitro cytotoxicity was evaluated against renal CAKI-1 cells2000Journal of medicinal chemistry, Apr-20, Volume: 43, Issue:8
Synthesis and evaluation of several new (2-chloroethyl)nitrosocarbamates as potential anticancer agents.
AID96698Antitumor activity was determined against the ic implanted L1210 lymphoid leukemia in mice by range in survival days at 100 mg/kg (treatment of 1-9qd schedule) dosage; Range is 11-181986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID124599Median T/C calculated based on survivors at 71 mg/kg (292 umol) per day against Ip-implanted L1210 lymphoid leukemia mice1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID140522Tested for in vivo antitumor activity against Lewis Lung Carcinoma cell line in mice and tumor weight change at 100 mg/Kg per day1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID137176Antitumor activity against L1210/ara-C leukemia in mice (median survival) at the dose of 250 mg/kg; Value represented as Test/Control = 9/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID138575Antitumor activity against L1210 leukemia in mice (Average 7-day weight change) at the dose of 100 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID116793Percent increase in life span of drug-treated mice relative to tumored, vehicle treated controls at a dose of 500 mg/kg orally in the presence of 500 mg/kg of zebularine.1991Journal of medicinal chemistry, Nov, Volume: 34, Issue:11
Antitumor properties of 2(1H)-pyrimidinone riboside (zebularine) and its fluorinated analogues.
AID630371Cytotoxicity against human KB cells assessed as cell survival after 72 hrs by sulforhodamine B colorimetric assay2011Bioorganic & medicinal chemistry, Nov-01, Volume: 19, Issue:21
Synthesis and anticancer activity of 5'-chloromethylphosphonates of 3'-azido-3'-deoxythymidine (AZT).
AID116601Percent increase in life span in mice bearing lymphoid leukemia, treatment schedule is 1-9 qd.1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID208532In vitro inhibitory effect on growth of human bladder transitional-carcinoma cell lines (T24).1988Journal of medicinal chemistry, Jun, Volume: 31, Issue:6
Design, synthesis, and antineoplastic activity of 2'-deoxy-2'-methylidenecytidine.
AID1360496Growth inhibition of human SMMC7721 cells at 0.1 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID1124448Antiviral activity against Vaccinia virus infected in human KB cells assessed as inhibition of virus-induced cytopathic effect after 72 hrs by microscopically1979Journal of medicinal chemistry, May, Volume: 22, Issue:5
Synthesis and antitumor and antiviral activities of 1-beta-D-arabinofuranosyl-2-amino-1,4(2H)-iminopyrimidine and its derivatives.
AID1145829Antiviral activity against Herpes Simplex virus HF infected in African green monkey BGM cells assessed as plaque reduction measured on day 5 by disk diffusion assay1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID138897Mean body weight change on 5th day at a dose of 100 mg/kg/day1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID208863In vitro inhibitory activity against Human esophagus adenocarcinoma (TE-2) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID678712Inhibition of human CYP1A2 assessed as ratio of IC50 in absence of NADPH to IC50 for presence of NADPH using ethoxyresorufin as substrate after 30 mins2012Chemical research in toxicology, Oct-15, Volume: 25, Issue:10
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
AID355586Antiproliferative activity against mouse P388 cells at 0.01 ug after 48 hrs by two-layer agar-diffusion method
AID416482Induction of erythroid differentiation in human K562 cells assessed as benzidine-positive cells at 500 mM after 6 days2009European journal of medicinal chemistry, Feb, Volume: 44, Issue:2
Synthesis of glycose carbamides and evaluation of the induction of erythroid differentiation of human erythroleukemic K562 cells.
AID1123146Cytotoxicity against mouse P815 cells assessed as growth inhibition after 96 hrs1979Journal of medicinal chemistry, Jan, Volume: 22, Issue:1
Nucleosides. 110. Synthesis and antiherpes virus activity of some 2'-fluoro-2'-deoxyarabinofuranosylpyrimidine nucleosides.
AID40606Tested in vitro inhibitory activity against murine B16 melanoma cells line1993Journal of medicinal chemistry, Dec-10, Volume: 36, Issue:25
Synthesis and biological evaluation of N4-substituted imidazo- and v-triazolo[4,5-d]pyridazine nucleosides.
AID116792Percent increase in life span of drug-treated mice relative to tumored, vehicle treated controls at a dose of 500 mg/kg oral treatment1991Journal of medicinal chemistry, Nov, Volume: 34, Issue:11
Antitumor properties of 2(1H)-pyrimidinone riboside (zebularine) and its fluorinated analogues.
AID289357Antitumor activity against human CCRF-HSB2 cells by modified MTT assay2007Bioorganic & medicinal chemistry, Oct-01, Volume: 15, Issue:19
Antitumor studies. part 3: design, synthesis, antitumor activity, and molecular docking study of novel 2-methylthio-, 2-amino-, and 2-(N-substituted amino)-10-alkyl-2-deoxo-5-deazaflavins.
AID496826Antimicrobial activity against Entamoeba histolytica2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID85383Growth inhibition against HSV-1 virus in BSCL cells at the dose of 1.4 ug/mL and 22h time of treatment.1983Journal of medicinal chemistry, Jan, Volume: 26, Issue:1
Antiviral compounds. 1. Structure-activity relationship of some antiviral enediones derived from aldehydo sugars.
AID97102Antitumor activity was determined against the ic implanted L1210 lymphoid leukemia in mice by percent increase in life span at 100 mg/kg (treatment of 1-9qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID284055Growth inhibition of human KB cells by MTT assay2007Bioorganic & medicinal chemistry, Jan-01, Volume: 15, Issue:1
Antitumor studies. Part 1: design, synthesis, antitumor activity, and AutoDock study of 2-deoxo-2-phenyl-5-deazaflavins and 2-deoxo-2-phenylflavin-5-oxides as a new class of antitumor agents.
AID1159292Cytotoxicity against human HCT116 cells by SRB assay2014European journal of medicinal chemistry, Aug-18, Volume: 83Structure-activity relationships of 2'-modified-4'-selenoarabinofuranosyl-pyrimidines as anticancer agents.
AID410582Inhibition of erythroid differentiation in human K562 cell assessed benzidine positive cells at 200 uM after 6 days by benzidine staining assay2009Journal of medicinal chemistry, Jan-08, Volume: 52, Issue:1
New uracil dimers showing erythroid differentiation inducing activities.
AID97615Antitumor activity was determined against the ic implanted L1210 lymphoid leukemia in mice by percent median survival rate (treated/control) at 100 mg/kg (treatment of 1-9qd schedule) dosage; 14/7.01986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID436680Antitumor activity against mouse S180 cells xenografted in ICR mouse assessed as inhibition of tumor growth at 89 umol/kg, ip administered 24 hrs after tumor implantation once daily for 7 consecutive days measured after 24 hrs last post dose2009European journal of medicinal chemistry, Oct, Volume: 44, Issue:10
Novel N-(3-carboxyl-9-benzyl-beta-carboline-1-yl)ethylamino acids: synthesis, anti-tumor evaluation, intercalating determination, 3D QSAR analysis and docking investigation.
AID1360489Cytotoxicity against human SMMC7721 cells assessed as inhibition of cell proliferation after 72 hrs by MTT assay2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID468160Cytotoxicity against human CEM cells after 2 days2010European journal of medicinal chemistry, Feb, Volume: 45, Issue:2
Synthesis and in vitro cytostatic activity of new beta-D-arabino furan[1',2':4,5]oxazolo- and arabino-pyrimidinone derivatives.
AID89631In vitro inhibitory effect measured against human stomach adenocarcinoma ST-KM tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID122629Change in weight of mouse on day 8 was measured in L1210 lymphoid leukemia inoculated mice after ip administration at dose of 20 mg/kg/day X51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID128055Antitumor activity against L1210 leukemia in mice (Average weight per mouse on day 1.) at the dose of 250 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID274776Distribution of araCTP in Swiss Webster mouse liver at 100 mg/kg, ip measured as AUC (0-4h)2006Journal of medicinal chemistry, Dec-28, Volume: 49, Issue:26
Synthesis and characterization of a novel liver-targeted prodrug of cytosine-1-beta-D-arabinofuranoside monophosphate for the treatment of hepatocellular carcinoma.
AID82976In vitro concentration required to induce apoptosis in HL60 cells2003Journal of medicinal chemistry, Jul-31, Volume: 46, Issue:16
Synthesis and biological evaluation of resveratrol and analogues as apoptosis-inducing agents.
AID139211Weight change on day 8 in L1210/ara-C (I) Lymphoid Leukemia in mice( ip implanted) at a dose of 60 mg/kg/day (qd=1-15)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID133642Dose producing greatest increase in life span expressed as optimum dose (L1210/ara-C I) (qd 1)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1123323Antiviral activity against Herpes simplex virus-2 LYONS infected in primary rabbit kidney cells assessed as inhibition of virus-induced cytopathogenicity treated immediately after virus adsorption measured 2 to 3 days after virus infection1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID44349Concentration required to inhibit replication of CCRF-CEM cell lines by 50% after 72 hr incubation1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID201681Concentration required for 50% inhibition of human lung squamous cell carcinoma SW 480 solid cell lines. 1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID1150988Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 200 umol/kg/day, po using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation1976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID284051Growth inhibition of human NCI-H460 cells by MTT assay2007Bioorganic & medicinal chemistry, Jan-01, Volume: 15, Issue:1
Antitumor studies. Part 1: design, synthesis, antitumor activity, and AutoDock study of 2-deoxo-2-phenyl-5-deazaflavins and 2-deoxo-2-phenylflavin-5-oxides as a new class of antitumor agents.
AID97116Antitumor activity was determined against the ip implanted L1210 leukemia in mice by percent increase in life span at 400 mg/kg (treatment of 1qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID116640Percent increase in life span of drug-treated mice relative to tumored, vehicle treated controls at a dose of 100 mg/kg oral treatment1991Journal of medicinal chemistry, Nov, Volume: 34, Issue:11
Antitumor properties of 2(1H)-pyrimidinone riboside (zebularine) and its fluorinated analogues.
AID98816Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 34 mg/kg dose (post inoculation life span, days)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID128064Antitumor activity against L1210 leukemia in mice (Average weight per mouse on day 1.) at the dose of 50 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID72420Tested in vitro against human ovarian cancer cells (G-401) for anticancer activity at dose 1 uM (Cell number 1*10 e-4)1994Journal of medicinal chemistry, Oct-14, Volume: 37, Issue:21
Synthesis of N-acetylglucosamine-modified ara-C and its effect on ovarian cancer cells.
AID97292Antitumor activity was determined against the ip implanted L1210 leukemia in mice by 45-day survivors in mice at 400 mg/kg (treatment of 1qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID93849The compound was tested in vitro for antitumor activity(50% inhibition of tumor cell growth) against KB human cell lines after 144 hr1987Journal of medicinal chemistry, Sep, Volume: 30, Issue:9
Improved synthesis and antitumor activity of 1-deazaadenosine.
AID101082Inhibitory concentration on multidrug-resistant L1210 leukemia cells.1991Journal of medicinal chemistry, Jul, Volume: 34, Issue:7
Additional nucleotide derivatives of mitosenes. Synthesis and activity against parental and multidrug resistant L1210 leukemia.
AID496832Antimicrobial activity against Trypanosoma brucei rhodesiense2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID210459Anticancer activity carried out in vitro against human T-lymphocyte CEM/02001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Synthesis and biological evaluation of purine-containing butenolides.
AID1079933Acute liver toxicity defined via clinical observations and clear clinical-chemistry results: serum ALT or AST activity > 6 N or serum alkaline phosphatases activity > 1.7 N. This category includes cytolytic, choleostatic and mixed liver toxicity. Value is
AID137174Antitumor activity against L1210/ara-C leukemia in mice (median survival) at the dose of 200 mg/kg; Value represented as Test/Control = 7/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID132913Percentage increase in life span after 12-13days1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID389364Growth inhibition of human CCRF-HSB-2 cells by MTT method2008Bioorganic & medicinal chemistry, Oct-15, Volume: 16, Issue:20
Antitumor studies. Part 5: Synthesis, antitumor activity, and molecular docking study of 5-(monosubstituted amino)-2-deoxo-2-phenyl-5-deazaflavins.
AID355579Antiproliferative activity against human HT-29 cells at 10 ug after 48 hrs by two-layer agar-diffusion method
AID625289Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver disease2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID96848Antitumor activity was determined against the ip implanted L1210 leukemia in mice by range in survival days at 400 mg/kg (treatment of 1qd schedule) dosage; Range is 9-111986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID98767Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 34 mg/kg dose(Mean increased life span, calculated as (T/C-1)X100)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID154855Antitumor activity was determined against the ip implanted P388 leukemia in mice by change in tumor weight at 200 mg/kg (treatment of 1-5qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID122631Change in weight of mouse on day 8 was measured in L1210 lymphoid leukemia inoculated mice after ip administration at dose of 40 mg/kg/day X51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID261687Antiproliferative activity against NCI60 cell line2006Journal of medicinal chemistry, Mar-23, Volume: 49, Issue:6
Assessment of in vitro and in vivo activities in the National Cancer Institute's anticancer screen with respect to chemical structure, target specificity, and mechanism of action.
AID1360490Cytotoxicity against human LO2 cells assessed as inhibition of cell proliferation after 72 hrs by MTT assay2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID1079944Benign tumor, proven histopathologically. Value is number of references indexed. [column 'T.BEN' in source]
AID427702Cytotoxicity against human A549 cells by sulforhodamine B method2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Discovery of a new template for anticancer agents: 2'-deoxy-2'-fluoro-4'-selenoarabinofuranosyl-cytosine (2'-F-4'-seleno-ara-C).
AID320428Antitumor activity against human CCRF-HSB2 cells by MTT assay2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Antitumor studies. Part 4: Design, synthesis, antitumor activity, and molecular docking study of novel 2-substituted 2-deoxoflavin-5-oxides, 2-deoxoalloxazine-5-oxides, and their 5-deaza analogs.
AID1360499Growth inhibition of human SMMC7721 cells at 5 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID89281In vitro inhibitory effect measured against human lung adenocarcinoma PC-9 tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID588219FDA HLAED, gamma-glutamyl transferase (GGT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID96540Antitumor activity was determined against the ic implanted L1210 lymphoid leukemia in mice by change in tumor weight at 100 mg/kg (treatment of 1-9qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID116595Percent increase in life span calculated as (T/C-1)X100 at a dose of 10 mg/kg/day *51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID338732Cytotoxicity against human TMOLT3 cells after 3 days by trypan blue exclusion technique
AID167741Dose required to reduce primary rabbit kidney cell growth1986Journal of medicinal chemistry, Feb, Volume: 29, Issue:2
Systematic synthesis and biological evaluation of alpha- and beta-D-xylofuranosyl nucleosides of the five naturally occurring bases in nucleic acids and related analogues.
AID411722Cytotoxicity against human 2209-23 cells by WST1 cell ciability assay2009Journal of medicinal chemistry, Jan-08, Volume: 52, Issue:1
The design, synthesis, and antiviral activity of 4'-azidocytidine analogues against hepatitis C virus replication: the discovery of 4'-azidoarabinocytidine.
AID306718Inhibition of human TK1 assessed as [methyl-3H]dThd phosphorylation2007Bioorganic & medicinal chemistry, Apr-15, Volume: 15, Issue:8
Novel selective human mitochondrial kinase inhibitors: design, synthesis and enzymatic activity.
AID502465Antitumor activity against mouse S180 cells transplanted in ICR mouse assessed as inhibition of tumor weight at 8.9 umol/kg, ip2010Bioorganic & medicinal chemistry, Sep-01, Volume: 18, Issue:17
A class of novel carboline intercalators: Their synthesis, in vitro anti-proliferation, in vivo anti-tumor action, and 3D QSAR analysis.
AID135728Toxicity in mice after 15-18 days at a dose of 60 mg/kg/day; 15.5/8.51988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID124261In vivo activity against transplanted Mel-B16 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID108522Number of animals survived after 30-days in mice bearing lymphoid leukemia, treatment schedule is 1-9 qd.1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID1292263Selectivity index, ratio of IC50 for HDF cells to IC50 for human KB cells2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID132923Percentage increase in life span after 7-8 days1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID341998Antimicrobial activity against Streptococcus sp. 07686-2 after 20 hrs2007Antimicrobial agents and chemotherapy, Aug, Volume: 51, Issue:8
Deoxyribonucleoside kinases activate nucleoside antibiotics in severely pathogenic bacteria.
AID1123325Antimetabolic activity against primary rabbit kidney cells assessed as inhibition of [methyl-3H]dThd incorporation into DNA after 16 hrs1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID75536Compound was tested for the inhibition of glioblastoma1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID137057Antitumor activity against L1210/ara-C leukemia in mice (median survival) at the dose of 1000 mg/kg; Value represented as Test/Control = 8/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID210892Inhibitory concentration against Varicella- zoster virus thymidine kinase(VZV TK)2001Bioorganic & medicinal chemistry letters, May-21, Volume: 11, Issue:10
Design, synthesis and enzymatic activity of highly selective human mitochondrial thymidine kinase inhibitors.
AID1222329Activity of human recombinant N-terminal His-SUMO fused wild type CDA expressed in Escherichia coli BL21(DE3) cells assessed as arabinofuranosyl uridine formation by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID625276FDA Liver Toxicity Knowledge Base Benchmark Dataset (LTKB-BD) drugs of most concern for DILI2011Drug discovery today, Aug, Volume: 16, Issue:15-16
FDA-approved drug labeling for the study of drug-induced liver injury.
AID411721Antiviral activity against Hepatitis C virus infected human 2209-23 cells by genotype 1b replicon assay2009Journal of medicinal chemistry, Jan-08, Volume: 52, Issue:1
The design, synthesis, and antiviral activity of 4'-azidocytidine analogues against hepatitis C virus replication: the discovery of 4'-azidoarabinocytidine.
AID416475Induction of erythroid differentiation in human K562 cells assessed as benzidine-positive cells after 6 days2009European journal of medicinal chemistry, Feb, Volume: 44, Issue:2
Synthesis of glycose carbamides and evaluation of the induction of erythroid differentiation of human erythroleukemic K562 cells.
AID96839Antitumor activity was determined against the ip implanted L1210 leukemia in mice by range in survival days at 200 mg/kg (treatment of 1-5qd schedule) dosage; Range is 15-171986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID155571Antiviral activity against Para influenza virus type 3 strain C243 was evaluated in african green monkey kidney1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID588214FDA HLAED, liver enzyme composite activity2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID588215FDA HLAED, alkaline phosphatase increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID588217FDA HLAED, serum glutamic pyruvic transaminase (SGPT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID88959In vitro inhibitory effect measured against human osteosarcoma KHOS-321H tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID361405Antiviral activity against HSV2 MS infected in african green monkey Vero cells assessed as inhibition of virus-induced cytopathic effect after 3 days by MTT assay2002Journal of natural products, Nov, Volume: 65, Issue:11
New saponins from the starfish Certonardoa semiregularis.
AID433254Cytotoxicity against human H441 cells after 72 hrs by trypan blue cell exclusion assay2009Bioorganic & medicinal chemistry letters, Aug-15, Volume: 19, Issue:16
Synthesis of radiolabeled cytarabine conjugates.
AID152349Effective dose to inhibit the replication of P388 cell line in vitro.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Synthesis and anticancer activity of various 3'-deoxy pyrimidine nucleoside analogues and crystal structure of 1-(3-deoxy-beta-D-threo-pentofuranosyl)cytosine.
AID1134746Half life of the compound1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID687160Ratio of IC50 for human HL60/MX2 cells to IC50 for human HL60 cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID57470Inhibitory activity against dihydrofolate reductase (DHFR) isolated from murine L5178Y tumor cells resistant and sensitive to methotrexate1984Journal of medicinal chemistry, Mar, Volume: 27, Issue:3
Comparative structure-activity relationships of antifolate triazines inhibiting murine tumor cells sensitive and resistant to methotrexate.
AID654377Growth inhibition of HUVEC cells after 72 hrs by WST-1 assay2012Journal of natural products, Feb-24, Volume: 75, Issue:2
Pinophilins A and B, inhibitors of mammalian A-, B-, and Y-family DNA polymerases and human cancer cell proliferation.
AID1135044Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as median survival time at 50 mg/kg, ip qd administered for 5 days (Rvb = 9 days)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID431398Cytotoxicity against human HL60 cells after 48 hrs by cell titer-blue assay2009Journal of medicinal chemistry, Oct-08, Volume: 52, Issue:19
Structure-activity relationship and molecular mechanisms of ethyl 2-amino-4-(2-ethoxy-2-oxoethyl)-6-phenyl-4h-chromene-3-carboxylate (sha 14-1) and its analogues.
AID89111In vitro inhibitory effect measured against human osteosarcoma MNNG-HOS tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID431520Antitumor activity against mouse S180 cells xenografted in ICR mouse assessed as tumor weight at 89.28 umol/kg/day, ip administered for 7 consecutive days after 24 hrs of tumor implantation measured after 24 hrs post dose2009Bioorganic & medicinal chemistry, Aug-15, Volume: 17, Issue:16
Synthesis, DNA intercalation and 3D QSAR analysis of cis-2,4,5-trisubstituted-1,3-dithiolanes as a novel class of antitumor agents.
AID338738Cytotoxicity against human EH118MG cells after 3 days by trypan blue exclusion technique
AID100203Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose of 25 mg/kg/day x 5 was determined as mean survival of treated group compared to untreated; 9.5/81980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID210538Phosphorylating activity of viral HSV -2 (333) induced thymidine kinase relative to dThdK was determined1987Journal of medicinal chemistry, Nov, Volume: 30, Issue:11
Acetylenic nucleosides. 4. 1-beta-D-arabinofuranosyl-5-ethynylcytosine. Improved synthesis and evaluation of biochemical and antiviral properties.
AID132925Percentage increase in life span after 7 days1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1145834Antitumor activity against mouse L1210 cells allografted in BDF1 mouse assessed as survival at 100 mg/kg, ip administered as single dose measured on day 30 relative to vehicle-treated control1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID98818Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 93 mg/kg dose (post inoculation life span, days)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID1135817Retention time of the compound by liquid chromatography1978Journal of medicinal chemistry, Nov, Volume: 21, Issue:11
Synthesis and biological activities of some uronic acids, uronates, uronamides, and urononitriles of pyrimidine nucleosides.
AID1549407Cytotoxicity against human L02 cells incubated for 72 hrs by MTT assay2019European journal of medicinal chemistry, May-01, Volume: 169Synthesis and biological activities of artemisinin-piperazine-dithiocarbamate derivatives.
AID1134725Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 6.25 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID1542692Induction of apoptosis in human U937 cells assessed as early apoptotic cells at 0.3 uM measured after 2 days by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 0.7 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID427705Cytotoxicity against human PC3 cells by sulforhodamine B method2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Discovery of a new template for anticancer agents: 2'-deoxy-2'-fluoro-4'-selenoarabinofuranosyl-cytosine (2'-F-4'-seleno-ara-C).
AID138713Antitumor activity against L1210 leukemia in mice (Average 7-day weight change) at the dose of 500 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1740798Antiproliferative activity against human PC-3 cells assessed as reduction in cell growth incubated for 72 hrs by SRB assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID1740806Selectivity index, ratio of IC50 for cytotoxicity against human HDF cells to IC50 for cytotoxicity against human A549 cells2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID81809Differentiating activity of compound in HL60 cells at conc 0.4 ug/mL1997Journal of medicinal chemistry, Nov-07, Volume: 40, Issue:23
Retinoic acid conjugates as potential antitumor agents: synthesis and biological activity of conjugates with Ara-A, Ara-C, 3(2H)-furanone, and aniline mustard moieties.
AID1632122Cytotoxicity against human HeLa cells assessed as reduction in cell survival after 72 hrs by sulforhodamine B assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID167039Cytotoxicity against RPMI-6410 human leukemic lymphoblastoid cell lines1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID654371Growth inhibition of human A549 cells after 24 hrs by WST-1 assay2012Journal of natural products, Feb-24, Volume: 75, Issue:2
Pinophilins A and B, inhibitors of mammalian A-, B-, and Y-family DNA polymerases and human cancer cell proliferation.
AID135620Long time survivors are the number mice that survived for >60 days relative to the number of treated mice 25 mg/kg; 0 / 51991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID678713Inhibition of human CYP2C9 assessed as ratio of IC50 in absence of NADPH to IC50 for presence of NADPH using 7-methoxy-4-trifluoromethylcoumarin-3-acetic acid as substrate after 30 mins2012Chemical research in toxicology, Oct-15, Volume: 25, Issue:10
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
AID94990Effect of compound on erythroid differentiation of K562 cells was determined after a period of 9 days at concentration 500 nM1999Bioorganic & medicinal chemistry letters, Nov-01, Volume: 9, Issue:21
Induction of erythroid differentiation of human K562 cells by 3-O-acyl-1,2-O-isopropylidene-D-glucofuranose derivatives.
AID1079943Malignant tumor, proven histopathologically. Value is number of references indexed. [column 'T.MAL' in source]
AID99057Tested in vitro for cytotoxicity against L1210 leukemia cell line in mouse1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID341999Antimicrobial activity against Staphylococcus aureus ATCC 29213 after 20 hrs2007Antimicrobial agents and chemotherapy, Aug, Volume: 51, Issue:8
Deoxyribonucleoside kinases activate nucleoside antibiotics in severely pathogenic bacteria.
AID127728Average survival time includes only those mice that died prior to day 60 at 50 mg/kg1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID208539Concentration required for 50% inhibition of human lung squamous cell carcinoma T24 solid cell lines. 1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID128050Antitumor activity against L1210 leukemia in mice (Average weight per mouse on day 1.) at the dose of 200 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID502466Antitumor activity against mouse S180 cells transplanted in ICR mouse assessed as inhibition of tumor weight at 89 umol/kg, ip2010Bioorganic & medicinal chemistry, Sep-01, Volume: 18, Issue:17
A class of novel carboline intercalators: Their synthesis, in vitro anti-proliferation, in vivo anti-tumor action, and 3D QSAR analysis.
AID451300Cytotoxicity against deoxynucleoside analogue-sensitive human MCF7 cells after 3 days by MTT assay2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID130645Antitumor activity against L1210/ara-C leukemia in mice (Percent increase in life span.) at the dose of 100 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1292264Selectivity index, ratio of IC50 for HDF cells to IC50 for human MCF7 cells2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID94986Effect of compound on erythroid differentiation of K562 cells was determined after a period of 7 days at concentration 500 nM1999Bioorganic & medicinal chemistry letters, Nov-01, Volume: 9, Issue:21
Induction of erythroid differentiation of human K562 cells by 3-O-acyl-1,2-O-isopropylidene-D-glucofuranose derivatives.
AID419502Volume of distribution at steady state in human administered 1 to 12 g doses with 2 to 3.5 hrs iv infusion2009Journal of medicinal chemistry, Jul-23, Volume: 52, Issue:14
In silico prediction of volume of distribution in human using linear and nonlinear models on a 669 compound data set.
AID133374Range of survival days for antitumor activity against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 400 mg/kg per day after treatment 51990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID124260In vivo activity against transplanted Mam-17/Adr tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID434133Cytotoxicity against human HeLa cells after 48 hrs by MTT assay2009European journal of medicinal chemistry, Sep, Volume: 44, Issue:9
Synthesis and evaluation of anti-tumor activities of N4 fatty acyl amino acid derivatives of 1-beta-arabinofuranosylcytosine.
AID100061Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 5 mg/kg/day x 5 was determined and number of animals survived on 30 th day were reported; 0/71980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID630372Cytotoxicity against human MCF7 cells assessed as cell survival after 72 hrs by sulforhodamine B colorimetric assay2011Bioorganic & medicinal chemistry, Nov-01, Volume: 19, Issue:21
Synthesis and anticancer activity of 5'-chloromethylphosphonates of 3'-azido-3'-deoxythymidine (AZT).
AID105312In vitro inhibitory activity against Human T-cell acute lymphoblastic leukemia (MOLT-4) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID1506323Growth inhibition of human bone marrow cells2018Journal of medicinal chemistry, Aug-09, Volume: 61, Issue:15
Exploring the Structure-Activity Relationship and Mechanism of a Chromene Scaffold (CXL Series) for Its Selective Antiproliferative Activity toward Multidrug-Resistant Cancer Cells.
AID1542697Induction of apoptosis in human U937 cells assessed as late apoptotic cells at 0.3 uM measured after 3 days by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 1.75 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID130784Antitumor activity against L1210/ara-C leukemia in mice (Percent increase in life span.) at the dose of 50 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID95873Tested for in vitro cytotoxicity against KB human oral epidermoid carcinoma cell line1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID1542691Induction of apoptosis in human U937 cells assessed as live cells at 0.3 uM measured after 2 days by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 95.05 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID588218FDA HLAED, lactate dehydrogenase (LDH) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID139205Weight change on day 8 in L1210/ara-C (I) Lymphoid Leukemia in mice( ip implanted) at a dose of 100 mg/kg/day (qd=1,5,9)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1134738Toxicity in CDF1 mouse assessed as change in body weight at 10 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID1422083Inhibition of MDM2 (unknown origin)2018European journal of medicinal chemistry, Nov-05, Volume: 159The development of piperidinones as potent MDM2-P53 protein-protein interaction inhibitors for cancer therapy.
AID496824Antimicrobial activity against Toxoplasma gondii2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID153311Antitumor activity was determined against the ip implanted P388 leukemia in mice by percent median survival rate (treated/control) at 500 mg/kg (treatment of 1qd schedule) dosage; 15.0/11.01986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1542693Induction of apoptosis in human U937 cells assessed as late apoptotic cells at 0.3 uM measured after 2 days by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 1.75 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID1740802Selectivity index, ratio of IC50 for cytotoxicity against human HDF cells to IC50 for cytotoxicity against human SK-BR-3 cells2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID284052Growth inhibition of human HCT116 cells by MTT assay2007Bioorganic & medicinal chemistry, Jan-01, Volume: 15, Issue:1
Antitumor studies. Part 1: design, synthesis, antitumor activity, and AutoDock study of 2-deoxo-2-phenyl-5-deazaflavins and 2-deoxo-2-phenylflavin-5-oxides as a new class of antitumor agents.
AID687162Ratio of IC50 for daunorubicin-resistant human HL60 cells to IC50 for human HL60 cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID361403Cytotoxicity against african green monkey Vero cells after 3 days by MTT assay2002Journal of natural products, Nov, Volume: 65, Issue:11
New saponins from the starfish Certonardoa semiregularis.
AID124800Antitumor activity against L1210 tumor in mice, at a dose of 500 mg/kg peroral administration in the presence of 500 mg/kg of zebularine.1991Journal of medicinal chemistry, Nov, Volume: 34, Issue:11
Antitumor properties of 2(1H)-pyrimidinone riboside (zebularine) and its fluorinated analogues.
AID678722Covalent binding affinity to human liver microsomes assessed per mg of protein at 10 uM after 60 mins presence of NADPH2012Chemical research in toxicology, Oct-15, Volume: 25, Issue:10
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
AID594972Antitumor activity against mouse S180 cells xenografted in ICR mouse assessed as tumor growth inhibition at 100 umol/kg/day, ip qd for 7 days measured 24 hrs after last dose relative to control2011European journal of medicinal chemistry, May, Volume: 46, Issue:5
A class of novel N-isoquinoline-3-carbonyl-L-amino acid benzylesters: synthesis, anti-tumor evaluation and 3D QSAR analysis.
AID83139In vitro concentration required to induce apoptosis in HL60R cells2003Journal of medicinal chemistry, Jul-31, Volume: 46, Issue:16
Synthesis and biological evaluation of resveratrol and analogues as apoptosis-inducing agents.
AID1855688Antiproliferative activity against human A549 cells measured after 48 hrs by MTT assay
AID89280In vitro inhibitory effect measured against human lung adenocarcinoma PC-8 tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID138718Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change (%)) at the dose of 100 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID496828Antimicrobial activity against Leishmania donovani2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID137884Ratio of survival time of treated to control mice at 25 mg/kg1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID687161Ratio of IC50 for human HL60/ADR cells to IC50 for human HL60 cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID100054Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 18.2 mg/kg/day x 5 was determined and number of animals survived on 30 th day were reported; 0/81980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID1079934Highest frequency of acute liver toxicity observed during clinical trials, expressed as a percentage. [column '% AIGUE' in source]
AID99793Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 18.2 mg/kg/day x 5 was determined as percentage increase in the life span as compared with control tumor bearers1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID137018Antitumor activity against L1210 leukemia in mice (median survival) at the dose of 100 mg/kg; Value represented as Test/Control = 16/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID88952In vitro inhibitory effect measured against human osteosarcoma HOS tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID135746Toxicity in mice after 13-15 days at a dose of 60 mg/kg/day; 14/8.51988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1292257Cytotoxicity against human HeLa cells after 72 hrs by SRB assay2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID88953In vitro inhibitory effect measured against human fibrosarcoma HT1080 tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID153288Antitumor activity was determined against the ip implanted P388 leukemia in mice by percent increase in life span at 500 mg/kg (treatment of 1qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1150997Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 200 umol/kg/day, po using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation in presence of THU 10 mg/kg/day on days 1 to 51976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID1079936Choleostatic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is < 2 (see ACUTE). Value is number of references indexed. [column 'CHOLE' in source]
AID1879227Cytotoxicity against human U-87 MG cells measured after 3 days by Celltiter-glo assay2022Bioorganic & medicinal chemistry letters, 04-01, Volume: 61Design, chemical synthesis and antiviral evaluation of 2'-deoxy-2'-fluoro-2'-C-methyl-4'-thionucleosides.
AID1222327Activity of human recombinant N-terminal His-SUMO fused CDA Ala70Thr mutant expressed in Escherichia coli BL21(DE3) cells assessed as arabinofuranosyl uridine formation by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID355642Cytotoxicity against mouse Panc03 cells at 1.5 ug/disk by corbett disk diffusion assay
AID139208Weight change on day 8 in L1210/ara-C (I) Lymphoid Leukemia in mice( ip implanted) at a dose of 200 mg/kg/day (qd=1)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1474167Liver toxicity in human assessed as induction of drug-induced liver injury by measuring verified drug-induced liver injury concern status2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID1222328Activity of human recombinant N-terminal His-SUMO fused CDA Lys27Gln mutant expressed in Escherichia coli BL21(DE3) cells assessed as arabinofuranosyl uridine formation by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID124263In vivo activity against transplanted Panc-03 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID1134726Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 5 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID1134734Toxicity in CDF1 mouse assessed as change in body weight at 40 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID1222346Drug metabolism assessed as intrinsic clearance for human recombinant N-terminal His-tagged DCK Ala119Gly mutant-mediated cytosine-beta-D-arabinofuranoside-5'-MP formation expressed in Escherichia coli BL21(DE3) incubated for 3 mins by liquid chromatograp2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID85387Inhibition of viral DNA synthesis in HSV-1 virus at the dose of 1.4 ug/mL.1983Journal of medicinal chemistry, Jan, Volume: 26, Issue:1
Antiviral compounds. 1. Structure-activity relationship of some antiviral enediones derived from aldehydo sugars.
AID120066Median survival time expressed as (T/C) was measured at dose of 20 mg/kg/day X51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID214064In vitro inhibitory activity against Human histiocytic lymphoma (U-937) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID105305Cytotoxicity against MOLT-4 human leukemic lymphoblastoid cell lines1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID72422Tested in vitro against human ovarian cancer cells (G-401) for anticancer activity at dose 10 uM (Cell number 1*10 e-4)1994Journal of medicinal chemistry, Oct-14, Volume: 37, Issue:21
Synthesis of N-acetylglucosamine-modified ara-C and its effect on ovarian cancer cells.
AID1159296Cytotoxicity against human PC3 cells by SRB assay2014European journal of medicinal chemistry, Aug-18, Volume: 83Structure-activity relationships of 2'-modified-4'-selenoarabinofuranosyl-pyrimidines as anticancer agents.
AID502463Cytotoxicity against human K562 cells after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Sep-01, Volume: 18, Issue:17
A class of novel carboline intercalators: Their synthesis, in vitro anti-proliferation, in vivo anti-tumor action, and 3D QSAR analysis.
AID98041Effective dose to inhibit the replication of L1210 cell line in vitro.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Synthesis and anticancer activity of various 3'-deoxy pyrimidine nucleoside analogues and crystal structure of 1-(3-deoxy-beta-D-threo-pentofuranosyl)cytosine.
AID1292267Cytotoxicity against human 143B cells after 72 hrs by SRB assay2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID1159297Cytotoxicity against human K562 cells by SRB assay2014European journal of medicinal chemistry, Aug-18, Volume: 83Structure-activity relationships of 2'-modified-4'-selenoarabinofuranosyl-pyrimidines as anticancer agents.
AID625283Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for elevated liver function tests2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID155995In vitro inhibitory activity against Human lung adenocarcinoma (PC-14) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID135621Long time survivors are the number mice that survived for >60 days relative to the number of treated mice at 50 mg/kg; 0 / 51991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID1145837Antitumor activity against mouse L1210 cells allografted in BDF1 mouse assessed as survival at 1000 mg/kg, ip administered as single dose measured on day 30 relative to vehicle-treated control1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID1222324Activity of human recombinant N-terminal His-SUMO fused CDA Ala70Thr mutant expressed in Escherichia coli BL21(DE3) cells assessed as intrinsic clearance for arabinofuranosyl uridine formation by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID135301No of survivors after 45 days at a dose of 200 mg/kg/day1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID116437Percent increase in life span at 100 mg/kg (411 umol) per day1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID341996Antimicrobial activity against Streptococcus sp. 07941-1 after 20 hrs2007Antimicrobial agents and chemotherapy, Aug, Volume: 51, Issue:8
Deoxyribonucleoside kinases activate nucleoside antibiotics in severely pathogenic bacteria.
AID142338Anticancer activity carried out in vitro against murine leukemia P3882001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Synthesis and biological evaluation of purine-containing butenolides.
AID341997Antimicrobial activity against Streptococcus sp. 07706-1 after 20 hrs2007Antimicrobial agents and chemotherapy, Aug, Volume: 51, Issue:8
Deoxyribonucleoside kinases activate nucleoside antibiotics in severely pathogenic bacteria.
AID127727Average survival time includes only those mice that died prior to day 60 at 25 mg/kg1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID81806Differentiating activity of compound in HL60 cells at conc 0.08 ug/mL1997Journal of medicinal chemistry, Nov-07, Volume: 40, Issue:23
Retinoic acid conjugates as potential antitumor agents: synthesis and biological activity of conjugates with Ara-A, Ara-C, 3(2H)-furanone, and aniline mustard moieties.
AID154291The compound was tested for anticancer activity against P815(arc-C resistant) cell lines of mouse leukemia1980Journal of medicinal chemistry, Oct, Volume: 23, Issue:10
Nucleosides. 116. 1-(beta-D-Xylofuranosyl)-5-fluorocytosines with a leaving group on the 3' position. Potential double-barreled masked precursors of anticancer nucleosides.
AID99782Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 5 mg/kg/day x 5 was determined as percentage increase in the life span as compared with control tumor bearers1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID135744Toxicity in mice after 9-12 days at a dose of 200 mg/kg/day; 10.5/9.51988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID138726Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change (%)) at the dose of 500 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1134718Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 80 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID746835Cytotoxicity against human MA9.3 cells after 48 hrs by MTT assay2013Bioorganic & medicinal chemistry letters, May-15, Volume: 23, Issue:10
Novel ROS-activated agents utilize a tethered amine to selectively target acute myeloid leukemia.
AID198969Concentration required to inhibit replication of S-180 cell lines by 50% after 72 hr incubation1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID342000Antimicrobial activity against Staphylococcus aureus CCM 885 after 20 hrs2007Antimicrobial agents and chemotherapy, Aug, Volume: 51, Issue:8
Deoxyribonucleoside kinases activate nucleoside antibiotics in severely pathogenic bacteria.
AID1150996Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 100 umol/kg/day, po using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation in presence of THU 10 mg/kg/day on days 1 to 51976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID1123153Antiviral activity against HSV1 2931 infected in African green monkey Vero cells assessed as inhibition of virus replication at 0.1 ug/ml after 24 hrs relative to control1979Journal of medicinal chemistry, Jan, Volume: 22, Issue:1
Nucleosides. 110. Synthesis and antiherpes virus activity of some 2'-fluoro-2'-deoxyarabinofuranosylpyrimidine nucleosides.
AID132914Percentage increase in life span after 13-15 days1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID99511The compound was tested for anticancer activity against L5178Y (sensitive) cell lines of mouse leukemia1980Journal of medicinal chemistry, Oct, Volume: 23, Issue:10
Nucleosides. 116. 1-(beta-D-Xylofuranosyl)-5-fluorocytosines with a leaving group on the 3' position. Potential double-barreled masked precursors of anticancer nucleosides.
AID117643In vivo activity against transplanted Mam-16/C/Adr tumors of mice; Not active2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID82836Compound was tested for the inhibition of human HL60 Leukemia1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1589117Cytotoxicity in human HeLa cells assessed as reduction in cell viability incubated for 72 hrs by SRB assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID55975Relative velocity was determined against deaminase purified from human acute myeloblastic leukemia cells1987Journal of medicinal chemistry, Nov, Volume: 30, Issue:11
Acetylenic nucleosides. 4. 1-beta-D-arabinofuranosyl-5-ethynylcytosine. Improved synthesis and evaluation of biochemical and antiviral properties.
AID1229558Cytotoxicity against human PC3 cells assessed as inhibition of cell proliferation after 72 hrs by sulforhodamine B assay2015Journal of medicinal chemistry, Jun-25, Volume: 58, Issue:12
Structure-Activity Relationships of Neplanocin A Analogues as S-Adenosylhomocysteine Hydrolase Inhibitors and Their Antiviral and Antitumor Activities.
AID135735Toxicity in mice after 7-8 days at a dose of 100 mg/kg/day; 7/91988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID97634Antitumor activity was determined against the ip implanted L1210 leukemia in mice by percent median survival rate (treated/control) at 400 mg/kg (treatment of 1qd schedule) dosage; 9.5/8.01986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1375116Antitumor activity against human HL60 cells xenografted in zebrafish embryo assessed as inhibition of cell migration at 200 ug/ml measured after 3 days of incubation by CM-Dil staining-based microscopic method relative to control2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Natural Product Micheliolide (MCL) Irreversibly Activates Pyruvate Kinase M2 and Suppresses Leukemia.
AID84938Antiviral activity against Herpes simplex virus -2 strain MS was evaluated in african green monkey kidney1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID1542694Induction of apoptosis in human U937 cells assessed as necrotic cells at 0.3 uM measured after 2 days by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 2.5 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID490287Antiproliferative activity against mouse r/m HM-SFME-1 cells after 24 hrs by MTT assay2010European journal of medicinal chemistry, Jul, Volume: 45, Issue:7
Novel effects of glycyrrhetinic acid on the central nervous system tumorigenic progenitor cells: induction of actin disruption and tumor cell-selective toxicity.
AID490286Antiproliferative activity against mouse SFME cells after 24 hrs by MTT assay2010European journal of medicinal chemistry, Jul, Volume: 45, Issue:7
Novel effects of glycyrrhetinic acid on the central nervous system tumorigenic progenitor cells: induction of actin disruption and tumor cell-selective toxicity.
AID746836Cytotoxicity against human CD34-positive blood stem/progenitor cells after 48 hrs by MTT assay2013Bioorganic & medicinal chemistry letters, May-15, Volume: 23, Issue:10
Novel ROS-activated agents utilize a tethered amine to selectively target acute myeloid leukemia.
AID1079947Comments (NB not yet translated). [column 'COMMENTAIRES' in source]
AID654372Growth inhibition of human BALL-1 cells after 24 hrs by WST-1 assay2012Journal of natural products, Feb-24, Volume: 75, Issue:2
Pinophilins A and B, inhibitors of mammalian A-, B-, and Y-family DNA polymerases and human cancer cell proliferation.
AID89279In vitro inhibitory effect measured against human lung small-cell carcinoma PC-6 tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID1134733Toxicity in CDF1 mouse assessed as change in body weight at 50 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID138731Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change) at the dose of 100 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1135093Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as increase in life span at 18.3 mg/kg, ip qd administered for 5 days co-administered with 27 mg/kg prednisone relative to control1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID122796Dose producing greatest increase in life span >/=25% in mice bearing lymphoid leukemia, treatment schedule is 1-5 qd; 5-80 [mg(umol)/kg] /day1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID138727Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change (%)) at the dose of 50 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID83137In vitro concentration required to induce apoptosis in HL60R cells2003Journal of medicinal chemistry, Jul-31, Volume: 46, Issue:16
Synthesis and biological evaluation of resveratrol and analogues as apoptosis-inducing agents.
AID433255Cytotoxicity in human H441 cells at 2 uM after 72 hrs by trypan blue cell exclusion assay2009Bioorganic & medicinal chemistry letters, Aug-15, Volume: 19, Issue:16
Synthesis of radiolabeled cytarabine conjugates.
AID150342Concentration required to inhibit replication of P388 cell lines by 50% after 72 hr incubation1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID99488Concentration required for 50% inhibition of cell growth of L1210 leukemic cell lines in mouse.1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID98957Antitumor activity against the ip implanted L1210 leukemia in mice expressed as optimal dose (1-5qd treatment schedule) required to produce greatest increase in life span was determined1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1549406Cytotoxicity against human SMMC7721 cells incubated for 72 hrs by MTT assay2019European journal of medicinal chemistry, May-01, Volume: 169Synthesis and biological activities of artemisinin-piperazine-dithiocarbamate derivatives.
AID98035Concentration required to inhibit replication of L1210 cell lines by 50% after 72 hr incubation1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID138895Mean body weight change on 17th day at a dose of 100 mg/kg/day1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID608163Antiproliferative activity against human K562 cells after 48 hrs by MTT assay2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID89105In vitro inhibitory effect measured against human malignant fibrous histiocytoma MFH-ST tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID135305No of survivors after 45 days at a dose of 60 mg/kg/day1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID98814Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 156 mg/kg dose (post inoculation life span, days)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID1542695Induction of apoptosis in human U937 cells assessed as live cells at 0.3 uM measured after 3 days by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 95.05 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID138898Mean body weight change on 5th day at a dose of 200 mg/kg/day1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID89130In vitro inhibitory effect measured against human osteosarcoma OST tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID155991Tested in vitro against PC-13 human lung large cell carcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID306719Inhibition of human TK2 assessed as [methyl-3H]dThd phosphorylation2007Bioorganic & medicinal chemistry, Apr-15, Volume: 15, Issue:8
Novel selective human mitochondrial kinase inhibitors: design, synthesis and enzymatic activity.
AID142336Inhibitory activity against murine leukemia L1210 cells1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Unsaturated and carbocyclic nucleoside analogues: synthesis, antitumor, and antiviral activity.
AID105661Concentration required for 50% inhibition of cell growth of MOLT-4 leukemic cell lines in human.1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID451297Cytotoxicity against deoxycytidine kinase deficient and nucleoside analogue-resistant mouse L1210 10K cells after 3 days by MTT assay2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID43616Anticancer activity carried out in vitro against breast carcinoma MCF-72001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Synthesis and biological evaluation of purine-containing butenolides.
AID96553Antitumor activity was determined against the ip implanted L1210 leukemia in mice by change in tumor weight at 400 mg/kg (treatment of 1-5qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID210863Rate of phosphorylation relative to human CytodCydK was determined1987Journal of medicinal chemistry, Nov, Volume: 30, Issue:11
Acetylenic nucleosides. 4. 1-beta-D-arabinofuranosyl-5-ethynylcytosine. Improved synthesis and evaluation of biochemical and antiviral properties.
AID1123319Antiviral activity against Vaccinia virus infected in primary rabbit kidney cells assessed as inhibition of virus-induced cytopathogenicity treated immediately after virus adsorption measured 2 to 3 days after virus infection1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID87474The compound was tested in vitro for antitumor activity(50% inhibition of tumor cell growth) against HeLa human cell lines after 144 hr1987Journal of medicinal chemistry, Sep, Volume: 30, Issue:9
Improved synthesis and antitumor activity of 1-deazaadenosine.
AID100491Inhibition of L1210 lymphoid leukemia cells 72 hr after administration (length of preincubation=0 hr)1992Journal of medicinal chemistry, Sep-04, Volume: 35, Issue:18
Synthesis and functional evaluation of a peptide derivative of 1-beta-D-arabinofuranosylcytosine.
AID89494In vitro inhibitory effect measured against human osteosarcoma SAOS-2 tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID1145836Antitumor activity against mouse L1210 cells allografted in BDF1 mouse assessed as survival at 500 mg/kg, ip administered as single dose measured on day 30 relative to vehicle-treated control1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID1124451Antiviral activity against Pseudorabies virus infected in human KB cells assessed as inhibition of virus-induced cytopathic effect after 72 hrs by microscopically1979Journal of medicinal chemistry, May, Volume: 22, Issue:5
Synthesis and antitumor and antiviral activities of 1-beta-D-arabinofuranosyl-2-amino-1,4(2H)-iminopyrimidine and its derivatives.
AID152363Antitumor activity against ip implanted P388 leukemia in mice expressed as active dose range (1-5qd treatment schedule) required to produce increase in life span by >25% was determined; Range is 20-2001986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID341995Antimicrobial activity against Escherichia coli ATCC 25922 after 20 hrs2007Antimicrobial agents and chemotherapy, Aug, Volume: 51, Issue:8
Deoxyribonucleoside kinases activate nucleoside antibiotics in severely pathogenic bacteria.
AID496829Antimicrobial activity against Leishmania infantum2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID1134723Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 12.5 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID155865In vitro inhibitory activity against Human lung large-cell carcinoma (PC-13) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID99918Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 5 mg/kg/day x 5 was determined as Survival time in days; Range is 9-22 days1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID53936In vitro cytotoxicity was evaluated against colon DLD-1 cells2000Journal of medicinal chemistry, Apr-20, Volume: 43, Issue:8
Synthesis and evaluation of several new (2-chloroethyl)nitrosocarbamates as potential anticancer agents.
AID687164Ratio of IC50 for doxorubicin-resistant human K562 cells to IC50 for human K562 cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID1135092Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as increase in life span at 18.2 mg/kg, ip qd administered for 5 days co-administered with 27 mg/kg prednisolone relative to control1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID977602Inhibition of sodium fluorescein uptake in OATP1B3-transfected CHO cells at an equimolar substrate-inhibitor concentration of 10 uM2013Molecular pharmacology, Jun, Volume: 83, Issue:6
Structure-based identification of OATP1B1/3 inhibitors.
AID1589119Cytotoxicity in human A549 cells assessed as reduction in cell viability incubated for 72 hrs by SRB assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID434132Cytotoxicity against human HL60 cells after 48 hrs by MTT assay2009European journal of medicinal chemistry, Sep, Volume: 44, Issue:9
Synthesis and evaluation of anti-tumor activities of N4 fatty acyl amino acid derivatives of 1-beta-arabinofuranosylcytosine.
AID496820Antimicrobial activity against Trypanosoma brucei2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID1879226Antiviral activity against HCV by replicon assay2022Bioorganic & medicinal chemistry letters, 04-01, Volume: 61Design, chemical synthesis and antiviral evaluation of 2'-deoxy-2'-fluoro-2'-C-methyl-4'-thionucleosides.
AID431395Cytotoxicity against human CCRF-CEM cells after 48 hrs by cell titer-blue assay2009Journal of medicinal chemistry, Oct-08, Volume: 52, Issue:19
Structure-activity relationship and molecular mechanisms of ethyl 2-amino-4-(2-ethoxy-2-oxoethyl)-6-phenyl-4h-chromene-3-carboxylate (sha 14-1) and its analogues.
AID1632125Cytotoxicity against human HepG2 cells assessed as reduction in cell survival after 72 hrs by sulforhodamine B assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID137030Antitumor activity against L1210 leukemia in mice (median survival) at the dose of 200 mg/kg; Value represented as Test/Control = 17/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID608261Toxicity in ICR mouse xenografted with mouse S180 cells assessed as effect on kidney weight at 2 umol/kg2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID1292268Cytotoxicity against human thymidine kinase deficient 143B cells after 72 hrs by SRB assay2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID1360503Growth inhibition of human LO2 cells at 2.5 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID306722Inhibition of VZV TK assessed as [methyl-3H]dThd phosphorylation2007Bioorganic & medicinal chemistry, Apr-15, Volume: 15, Issue:8
Novel selective human mitochondrial kinase inhibitors: design, synthesis and enzymatic activity.
AID137886Ratio of survival time of treated to control mice at 75 mg/kg1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID451313Resistance ratio of IC50 for deoxynucleoside analogue-resistant human MCF7 1K cells to IC50 for deoxynucleoside analogue-sensitive human MCF7 cells2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID124913Change in weight in mice bearing ip-implanted L1210 lymphoid leukemia on day 8 at 200 mg/kg or 822 umol/kg per day1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID687165Ratio of IC50 for human K562/HHT300 cells to IC50 for human K562 cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID89647In vitro inhibitory effect measured against human bladder transitional cell carcinoma T-24 tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID116799Percent increase in life span of leukemic mice administered, with compound at 100 mg/kgx5, intraperitoneally1989Journal of medicinal chemistry, Apr, Volume: 32, Issue:4
2'-Fluorinated isonucleosides. 1. Synthesis and biological activity of some methyl 2'-deoxy-2'-fluoro-2'-pyrimidinyl-D-arabinopyranosides.
AID155844In vitro inhibitory activity against Human lung squamous cell carcinoma (PC-10) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID678994TP_TRANSPORTER: uptake in Xenopus laevis oocytes2000Biochemical pharmacology, Jul-15, Volume: 60, Issue:2
Role of organic cation transporters in the renal secretion of nucleosides.
AID124262In vivo activity against transplanted Panc-02 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID130631Antitumor activity against L1210 leukemia in mice (Percent increase in life span.) at the dose of 250 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID44350Concentration required to inhibit replication of CCRF-CEM deoxycytidine kinase deficient variant CCRF-CEM(dCK-)1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID588216FDA HLAED, serum glutamic oxaloacetic transaminase (SGOT) increase2004Current drug discovery technologies, Dec, Volume: 1, Issue:4
Assessment of the health effects of chemicals in humans: II. Construction of an adverse effects database for QSAR modeling.
AID1134724Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 10 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID44363Antiproliferative effects on CCRF-HSB-2 (human leukemia) cells.1997Journal of medicinal chemistry, Jul-04, Volume: 40, Issue:14
A facile, alternative synthesis of 4'-thioarabinonucleosides and their biological activities.
AID1135720Immunosuppressive activity against PHA-stimulated proliferation in rat T-lymphocytes assessed as cell viability at 1 uM after 6 days by [3H]thymidine incorporation method relative to control1978Journal of medicinal chemistry, Jul, Volume: 21, Issue:7
Synthesis of branched-chain apiosylpyrimidines and their inhibition of lymphocyte proliferation.
AID97287Antitumor activity was determined against the ip implanted L1210 leukemia in mice by 45-day survivors in mice at 200 mg/kg (treatment of 1-5qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID451296Cytotoxicity against deoxynucleoside analogue-sensitive mouse L1210 cells after 3 days by MTT assay2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID338734Cytotoxicity against human SW480 cells after 3 days by trypan blue exclusion technique
AID1134727Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 3.12 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID496819Antimicrobial activity against Plasmodium falciparum2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID94681Tested in vitro against KKLS human stomach adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID1632126Cytotoxicity against HDF assessed as reduction in cell survival after 72 hrs by sulforhodamine B assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID1360500Growth inhibition of human LO2 cells at 0.01 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID1146019Antiviral activity against Herpes simplex virus 2 Curtis assessed as inhibition of virus-induced cytopathic effect at 37 degC after 72 hrs by two-fold dilution method1977Journal of medicinal chemistry, Jun, Volume: 20, Issue:6
Antiviral activity of some beta-diketones. 1. Aryl alkyl diketones. In vitro activity against both RNA and DNA viruses.
AID154858Antitumor activity was determined against the ip implanted P388 leukemia in mice by change in tumor weight at 500 mg/kg (treatment of 1qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID364005Inhibition of rat brain protein kinase C2008European journal of medicinal chemistry, Jul, Volume: 43, Issue:7
Antitumor studies -- part 2: structure-activity relationship study for flavin analogs including investigations on their in vitro antitumor assay and docking simulation into protein tyrosine kinase.
AID769615Cytotoxicity against human KB cells after 72 hrs by SRB assay2013European journal of medicinal chemistry, Sep, Volume: 67Synthesis of 3'-azido-2',3'-dideoxy-5-fluorouridine phosphoramidates and evaluation of their anticancer activity.
AID124632In vivo antitumor activity against intraperitoneally inoculated L1210 lymphoid leukemic mice (Weight change observed on day 8.Treatment schedule is 1-9 qd)1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID306720Inhibition of HSV1 TK assessed as [methyl-3H]dThd phosphorylation2007Bioorganic & medicinal chemistry, Apr-15, Volume: 15, Issue:8
Novel selective human mitochondrial kinase inhibitors: design, synthesis and enzymatic activity.
AID1855689Antiproliferative activity against human WI-38 cells measured after 48 hrs by MTT assay
AID98769Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 93 mg/kg dose(Mean increased life span, calculated as (T/C-1)X100)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID1123149Antiviral activity against HSV1 2931 infected in African green monkey Vero cells assessed as inhibition of virus replication at 10 ug/ml after 24 hrs relative to control1979Journal of medicinal chemistry, Jan, Volume: 22, Issue:1
Nucleosides. 110. Synthesis and antiherpes virus activity of some 2'-fluoro-2'-deoxyarabinofuranosylpyrimidine nucleosides.
AID1542698Induction of apoptosis in human U937 cells assessed as necrotic cells at 0.3 uM measured after 3 days by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 2.5 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID1360501Growth inhibition of human LO2 cells at 0.1 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID139213Weight change on day 8 in L1210/ara-C (I) Lymphoid Leukemia in mice( ip implanted) at a dose of 60 mg/kg/day (qd=1-9)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1135091Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as median survival time at 18.3 mg/kg, ip qd administered for 5 days co-administered with 27 mg/kg prednisone (Rvb = 9 days)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID124583Median T/C calculated based on survivors at 100 mg/kg (411 umol) per day against Ip-implanted L1210 lymphoid leukemia mice1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID137036Antitumor activity against L1210 leukemia in mice (median survival) at the dose of 250 mg/kg; Value represented as Test/Control = 13/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID133644Dose producing greatest increase in life span expressed as optimum dose (L1210/ara-C I) (qd 1-15)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1589126Selectivity index, ratio of IC50 for cytotoxicity in human NHDF cells to IC50 for cytotoxicity in human U87 cells2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID1123321Antiviral activity against Herpes simplex virus-1 KOS infected in primary rabbit kidney cells assessed as inhibition of virus-induced cytopathogenicity treated immediately after virus adsorption measured 2 to 3 days after virus infection1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID608166Antiproliferative activity against human HepG2 cells after 48 hrs by MTT assay2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID1135056Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as increase in life span at 18.2 mg/kg, ip qd administered for 5 days relative to control1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID137047Antitumor activity against L1210 leukemia in mice (median survival) at the dose of 50 mg/kg; Value represented as Test/Control = 14/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID100058Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 25 mg/kg/day x 5 was determined and number of animals survived on 30 th day were reported; 0/81980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID135618Long time survivors are the number mice that survived for >60 days relative to the number of treated mice at 75 mg/kg; 0 / 51991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID95323Tested in vitro against KATO-III human stomach adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID120070Median survival time expressed as (T/C) was measured at dose of 40 mg/kg/day X51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID1292266Selectivity index, ratio of IC50 for HDF cells to IC50 for human 143B cells2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID135299No of survivors after 45 days at a dose of 100 mg/kg/day1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1145826Cytotoxicity against human HeLa cells after 48 hrs by Coulter counter analysis1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID1473741Inhibition of human MRP4 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID608259Antitumor activity against mouse S180 cells xenografted in ICR mouse assessed as tumor growth inhibition at 0.89 umol/kg, ip administered 1 day after tumor inoculation qd for 7 days2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID1916657Cytotoxicity against human MCF7 cells assessed as cell growth inhibition incubated for 48 hrs by AlamarBlue assay2022European journal of medicinal chemistry, Aug-05, Volume: 238Melphalan: Recent insights on synthetic, analytical and medicinal aspects.
AID409956Inhibition of mouse brain MAOB2008Journal of medicinal chemistry, Nov-13, Volume: 51, Issue:21
Quantitative structure-activity relationship and complex network approach to monoamine oxidase A and B inhibitors.
AID1222349Activity of human recombinant N-terminal His-tagged DCK Ile24Val mutant expressed in Escherichia coli BL21(DE3) cells assessed as cytosine-beta-D-arabinofuranoside-5'-MP formation incubated for 3 mins by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID99780Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 25 mg/kg/day x 5 was determined as percentage increase in the life span as compared with control tumor bearers1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID687157Cytotoxicity against human HL60 cells assessed as cell viability after 48 hrs by celltiter-blue assay2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID1595846Antitumor activity against human Z138 cells xenografted in nu/nu mouse assessed as tumor weight at 40 mg/kg, iv administered 5 times a week for 3 weeks and measured after 21 days (Rvb = 1.74 g)2019European journal of medicinal chemistry, Jul-01, Volume: 173Discovery of (R)-5-((5-(1-methyl-1H-pyrazol-4-yl)-4-(methylamino)pyrimidin-2-yl)amino)-3-(piperidin-3-yloxy)picolinonitrile, a novel CHK1 inhibitor for hematologic malignancies.
AID594979Antitumor activity against mouse S180 cells xenografted in ICR mouse assessed as decrease of tumor weight at 100 umol/kg, ip qd for 7 days measured 24 hrs after last dose2011European journal of medicinal chemistry, May, Volume: 46, Issue:5
A class of novel N-isoquinoline-3-carbonyl-L-amino acid benzylesters: synthesis, anti-tumor evaluation and 3D QSAR analysis.
AID1222326Activity of human recombinant N-terminal His-SUMO fused wild type CDA expressed in Escherichia coli BL21(DE3) cells assessed as intrinsic clearance for arabinofuranosyl uridine formation by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID128068Antitumor activity against L1210/ara-C leukemia in mice (Average weight per mouse on day 1.) at the dose of 100 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID81672Differentiating activity of compound in HL60 cells at conc 0.016 ug/mL1997Journal of medicinal chemistry, Nov-07, Volume: 40, Issue:23
Retinoic acid conjugates as potential antitumor agents: synthesis and biological activity of conjugates with Ara-A, Ara-C, 3(2H)-furanone, and aniline mustard moieties.
AID124259In vivo activity against transplanted Mam-17 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID427706Cytotoxicity against human K562 cells by sulforhodamine B method2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Discovery of a new template for anticancer agents: 2'-deoxy-2'-fluoro-4'-selenoarabinofuranosyl-cytosine (2'-F-4'-seleno-ara-C).
AID1145831Antitumor activity against mouse L1210 cells allografted in BDF1 mouse assessed as increase in life span at 500 mg/kg, ip administered as single dose relative to vehicle-treated control1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID475910Antitumor activity against human MCF7 cells after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Apr-01, Volume: 18, Issue:7
Synthesis, X-ray crystal structure study and antitumoral evaluations of 5,6-disubstituted pyrimidine derivatives.
AID1589127Selectivity index, ratio of IC50 for cytotoxicity in human NHDF cells to IC50 for cytotoxicity in human HepG2 cells2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID1135905Antiviral activity against Vesicular stomatitis virus infected in rabbit primary kidney cells assessed as inhibition of viral cytopathogenicity after 2 days1979Journal of medicinal chemistry, May, Volume: 22, Issue:5
Antiviral activity of aliphatic nucleoside analogues: structure-function relationship.
AID1292265Selectivity index, ratio of IC50 for HDF cells to IC50 for human HepG2 cells2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID138721Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change (%)) at the dose of 200 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID105667In vitro antiproliferative activity was measured in mouse myeloma L1210 cell line1982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID475907Antitumor activity against human MOLT4 cells after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Apr-01, Volume: 18, Issue:7
Synthesis, X-ray crystal structure study and antitumoral evaluations of 5,6-disubstituted pyrimidine derivatives.
AID1123151Cytotoxicity against mouse L5178Y cells assessed as growth inhibition after 96 hrs1979Journal of medicinal chemistry, Jan, Volume: 22, Issue:1
Nucleosides. 110. Synthesis and antiherpes virus activity of some 2'-fluoro-2'-deoxyarabinofuranosylpyrimidine nucleosides.
AID1229560Cytotoxicity against human SNU638 cells assessed as inhibition of cell proliferation after 72 hrs by sulforhodamine B assay2015Journal of medicinal chemistry, Jun-25, Volume: 58, Issue:12
Structure-Activity Relationships of Neplanocin A Analogues as S-Adenosylhomocysteine Hydrolase Inhibitors and Their Antiviral and Antitumor Activities.
AID96383Tested in vitro inhibitory activity against human oral epidermoid carcinoma KB cells1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID1222338Ratio of Vmax for human recombinant N-terminal His-tagged wild type DCK expressed in Escherichia coli BL21(DE3) cells to Vmax for human recombinant N-terminal His-tagged DCK Pro122Ser mutant expressed in Escherichia coli BL21(DE3) cells2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID654373Growth inhibition of against human HCT116 cells after 24 hrs by WST-1 assay2012Journal of natural products, Feb-24, Volume: 75, Issue:2
Pinophilins A and B, inhibitors of mammalian A-, B-, and Y-family DNA polymerases and human cancer cell proliferation.
AID274780Ratio of AUC for araCTP level in liver to plasma of Swiss Webster mouse2006Journal of medicinal chemistry, Dec-28, Volume: 49, Issue:26
Synthesis and characterization of a novel liver-targeted prodrug of cytosine-1-beta-D-arabinofuranoside monophosphate for the treatment of hepatocellular carcinoma.
AID138588Antitumor activity against L1210 leukemia in mice (Average 7-day weight change) at the dose of 250 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID136892Antitumor activity against L1210 leukemia in mice (median survival) at the dose of 1000 mg/kg; Value represented as Test/Control = 15/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID106856Tested in vitro against MKN-45 human stomach adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID100073Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 9.2 mg/kg/day x 5 was determined and number of animals survived on 30 th day were reported; 1/71980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID138734Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change) at the dose of 200 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID218494In vitro cytotoxicity against Vero cells.1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID101083Inhibitory concentration on parentral (sensitive) L1210 leukemia cells.1991Journal of medicinal chemistry, Jul, Volume: 34, Issue:7
Additional nucleotide derivatives of mitosenes. Synthesis and activity against parental and multidrug resistant L1210 leukemia.
AID625291Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver function tests abnormal2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1473738Inhibition of human BSEP overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-taurocholate in presence of ATP measured after 15 to 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1123326Antimetabolic activity against primary rabbit kidney cells assessed as inhibition of [2-14C]dUrd incorporation into DNA after 16 hrs1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID138723Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change (%)) at the dose of 250 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID433260Cytotoxicity against human H441 cells by glucosaminidase assay2009Bioorganic & medicinal chemistry letters, Aug-15, Volume: 19, Issue:16
Synthesis of radiolabeled cytarabine conjugates.
AID1375115Antitumor activity against human HL60 cells xenografted in zebrafish embryo assessed as inhibition of cell proliferation at 200 ug/ml measured after 3 days of incubation by CM-Dil staining-based microscopic method relative to control2018Journal of medicinal chemistry, 05-10, Volume: 61, Issue:9
Natural Product Micheliolide (MCL) Irreversibly Activates Pyruvate Kinase M2 and Suppresses Leukemia.
AID137058Antitumor activity against L1210/ara-C leukemia in mice (median survival) at the dose of 100 mg/kg; Value represented as Test/Control = 7/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1134737Toxicity in CDF1 mouse assessed as change in body weight at 12.5 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID1134721Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 25 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID502467Antitumor activity against mouse S180 cells transplanted in ICR mouse assessed as inhibition of tumor weight at 0.89 umol/kg, ip2010Bioorganic & medicinal chemistry, Sep-01, Volume: 18, Issue:17
A class of novel carboline intercalators: Their synthesis, in vitro anti-proliferation, in vivo anti-tumor action, and 3D QSAR analysis.
AID89278In vitro inhibitory effect measured against human lung adenocarcinoma PC-3 tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID1360497Growth inhibition of human SMMC7721 cells at 0.5 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID409744Antitumor activity against human CCRF-HSB2 cells by MTT assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
Synthesis, biological active molecular design, and molecular docking study of novel deazaflavin-cholestane hybrid compounds.
AID342001Antimicrobial activity against Streptococcus pyogenes AP1 after 20 hrs2007Antimicrobial agents and chemotherapy, Aug, Volume: 51, Issue:8
Deoxyribonucleoside kinases activate nucleoside antibiotics in severely pathogenic bacteria.
AID1222325Activity of human recombinant N-terminal His-SUMO fused CDA Lys27Gln mutant expressed in Escherichia coli BL21(DE3) cells assessed as intrinsic clearance for arabinofuranosyl uridine formation by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID208867In vitro inhibitory effect on growth of human esophagus adenocarcinoma cell lines (TE2).1988Journal of medicinal chemistry, Jun, Volume: 31, Issue:6
Design, synthesis, and antineoplastic activity of 2'-deoxy-2'-methylidenecytidine.
AID85090In vitro antiviral activity against Herpes simplex virus -2 strain MS was evaluated in african green monkey kidney1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID152592The compound was tested in vitro for antitumor activity(50% inhibition of tumor cell growth) against the P388 murine cell lines after 72 hours1987Journal of medicinal chemistry, Sep, Volume: 30, Issue:9
Improved synthesis and antitumor activity of 1-deazaadenosine.
AID306721Inhibition of Drosophila melanogaster dNK assessed as [methyl-3H]dThd phosphorylation2007Bioorganic & medicinal chemistry, Apr-15, Volume: 15, Issue:8
Novel selective human mitochondrial kinase inhibitors: design, synthesis and enzymatic activity.
AID1266628Antitumor activity against human KG1 cells xenografted in mouse assessed as tumor regression at 100 mg/kg, po qd measured after 11 days post tumor implantation2015Journal of medicinal chemistry, Nov-12, Volume: 58, Issue:21
Identification of N-(4-((1R,3S,5S)-3-Amino-5-methylcyclohexyl)pyridin-3-yl)-6-(2,6-difluorophenyl)-5-fluoropicolinamide (PIM447), a Potent and Selective Proviral Insertion Site of Moloney Murine Leukemia (PIM) 1, 2, and 3 Kinase Inhibitor in Clinical Tria
AID625285Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic necrosis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID289358Antitumor activity against human KB cells by modified MTT assay2007Bioorganic & medicinal chemistry, Oct-01, Volume: 15, Issue:19
Antitumor studies. part 3: design, synthesis, antitumor activity, and molecular docking study of novel 2-methylthio-, 2-amino-, and 2-(N-substituted amino)-10-alkyl-2-deoxo-5-deazaflavins.
AID89110In vitro inhibitory effect measured against human stomach adenocarcinoma MKN-28 tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID1506322Growth inhibition of human umbilical cord cells2018Journal of medicinal chemistry, Aug-09, Volume: 61, Issue:15
Exploring the Structure-Activity Relationship and Mechanism of a Chromene Scaffold (CXL Series) for Its Selective Antiproliferative Activity toward Multidrug-Resistant Cancer Cells.
AID1632129Selectivity index, ratio of IC50 for HDF to IC50 for human KB cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID133410In vivo percent inhibition of Lewis Lung Carcinoma cell line in mice at 100 mg/Kg per day (p<0.001) compared with NCDAC1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID1632131Selectivity index, ratio of IC50 for HDF to IC50 for human HepG2 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID89467In vitro inhibitory effect measured against human lung suamous-cell carcinoma QG-56 tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID156483Tested in vitro against PC-8 human lung adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID1135804Antiviral activity against HSV1 CL101 infected in African green monkey Vero cells assessed as inhibition of plaque formation at 200 uM after 40 hrs relative to control1978Journal of medicinal chemistry, Nov, Volume: 21, Issue:11
Synthesis and biological activities of some uronic acids, uronates, uronamides, and urononitriles of pyrimidine nucleosides.
AID1135090Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as median survival time at 18.2 mg/kg, ip qd administered for 5 days co-administered with 27 mg/kg prednisolone (Rvb = 9 days)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID138064Survival time treated / survival time (T/C) for antitumor activity against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 400 mg/kg per day after treatment 1;8/71990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID1123320Antiviral activity against Vaccinia virus infected in human skin fibroblast cells assessed as inhibition of virus-induced cytopathogenicity treated immediately after virus adsorption measured 2 to 3 days after virus infection1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID1135721Immunosuppressive activity against PHA-stimulated proliferation in rat T-lymphocytes assessed as cell viability at 100 uM after 6 days by [3H]thymidine incorporation method relative to control1978Journal of medicinal chemistry, Jul, Volume: 21, Issue:7
Synthesis of branched-chain apiosylpyrimidines and their inhibition of lymphocyte proliferation.
AID44010In vitro concentration required for 50% inhibition of growth of human leukemia cell line CCRF-CEM without hPAP (0.2 unit/mL)2001Bioorganic & medicinal chemistry letters, Nov-05, Volume: 11, Issue:21
Amino acid phosphoramidate nucleosides: potential ADEPT/GDEPT substrates.
AID496827Antimicrobial activity against Leishmania amazonensis2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID1145832Antitumor activity against mouse L1210 cells allografted in BDF1 mouse assessed as increase in life span at 1000 mg/kg, ip administered as single dose relative to vehicle-treated control1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID84060In vitro antiviral activity against Herpes simplex virus -1 strain KOS was evaluated in african green monkey kidney1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID140005Death due to toxicity occurring at a maximum body weight decrease of 10 to 22% from the average post injection body weight at 25 mg/kg; 2 / 51991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID128074Antitumor activity against L1210/ara-C leukemia in mice (Average weight per mouse on day 1.) at the dose of 500 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID451298Cytotoxicity against deoxynucleoside analogue-sensitive human MESSA cells after 3 days by MTT assay2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID82982In vitro inhibitory concentration against proliferation of HL60 cells2003Journal of medicinal chemistry, Jul-31, Volume: 46, Issue:16
Synthesis and biological evaluation of resveratrol and analogues as apoptosis-inducing agents.
AID130617Antitumor activity against L1210 leukemia in mice (Percent increase in life span.) at the dose of 100 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID45400Effect on cross resistance of CHO cells resistant to colchicine (CHRC5)1990Journal of medicinal chemistry, Jul, Volume: 33, Issue:7
Structure-activity relationships of antineoplastic agents in multidrug resistance.
AID123366In vivo antitumor activity against intraperitoneally inoculated L1210 Lymphoid leukemic mice (Dose producing greatest increase in life span, treatment schedule is 1- 9 qd)1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID201690Tested in vitro against SW-48 human colon adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID1595843Antitumor activity against human Z138 cells xenografted in nu/nu mouse assessed as inhibition in tumor growth at 40 mg/kg, iv administered 5 times a week for 3 weeks and measured twice per week by caliper method relative to control2019European journal of medicinal chemistry, Jul-01, Volume: 173Discovery of (R)-5-((5-(1-methyl-1H-pyrazol-4-yl)-4-(methylamino)pyrimidin-2-yl)amino)-3-(piperidin-3-yloxy)picolinonitrile, a novel CHK1 inhibitor for hematologic malignancies.
AID128067Antitumor activity against L1210/ara-C leukemia in mice (Average weight per mouse on day 1.) at the dose of 1000 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1222348Activity of human recombinant N-terminal His-tagged DCK Pro122Ser mutant expressed in Escherichia coli BL21(DE3) cells assessed as cytosine-beta-D-arabinofuranoside-5'-MP formation incubated for 3 mins by liquid chromatography-tandem mass spectrometry2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID124799Antitumor activity against L1210 tumor in mice, at a dose of 500 mg/kg peroral administration in the presence of 10 mg/kg of zebularine.1991Journal of medicinal chemistry, Nov, Volume: 34, Issue:11
Antitumor properties of 2(1H)-pyrimidinone riboside (zebularine) and its fluorinated analogues.
AID1079940Granulomatous liver disease, proven histopathologically. Value is number of references indexed. [column 'GRAN' in source]
AID133645Dose producing greatest increase in life span expressed as optimum dose (L1210/ara-C I) (qd 1-5)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID124253In vivo activity against iv transplanted AML Leukemia 1498 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID1123152Antiviral activity against HSV1 2931 infected in African green monkey Vero cells assessed as inhibition of virus replication at 0.01 ug/ml after 24 hrs relative to control1979Journal of medicinal chemistry, Jan, Volume: 22, Issue:1
Nucleosides. 110. Synthesis and antiherpes virus activity of some 2'-fluoro-2'-deoxyarabinofuranosylpyrimidine nucleosides.
AID95313Concentration required for 50% inhibition of human lung squamous cell carcinoma KATO III solid cell lines. 1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID89119In vitro inhibitory effect measured against human renal cell carcinoma NCC-nu tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID1146018Antiviral activity against Herpes simplex virus 1 Sheely assessed as inhibition of virus-induced cytopathic effect at 37 degC after 72 hrs by two-fold dilution method1977Journal of medicinal chemistry, Jun, Volume: 20, Issue:6
Antiviral activity of some beta-diketones. 1. Aryl alkyl diketones. In vitro activity against both RNA and DNA viruses.
AID1151006Immunosuppression activity in rat assessed as reduction of adjuvant-induced acute inflammation administered po for days 1 to 51976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID116802Percent increase in life span of leukemic mice administered, with compound at 50 mg/kgx5, intraperitoneally1989Journal of medicinal chemistry, Apr, Volume: 32, Issue:4
2'-Fluorinated isonucleosides. 1. Synthesis and biological activity of some methyl 2'-deoxy-2'-fluoro-2'-pyrimidinyl-D-arabinopyranosides.
AID769616Cytotoxicity against human HeLa cells after 72 hrs by SRB assay2013European journal of medicinal chemistry, Sep, Volume: 67Synthesis of 3'-azido-2',3'-dideoxy-5-fluorouridine phosphoramidates and evaluation of their anticancer activity.
AID140648Tested for in vivo antitumor activity against Lewis Lung Carcinoma cell line in mice and tumor weight change at 200 mg/Kg per day1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID1740800Antiproliferative activity against human A549 cells assessed as reduction in cell growth incubated for 72 hrs by SRB assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID132036Increase of life span was determined against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 200 mg/kg per day after treatment 1-51990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID132915Percentage increase in life span after 15-18 days1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID42138Inhibition of cellular DNA synthesis in BSCL cells at the dose of 1.4 ug/mL.1983Journal of medicinal chemistry, Jan, Volume: 26, Issue:1
Antiviral compounds. 1. Structure-activity relationship of some antiviral enediones derived from aldehydo sugars.
AID1360504Growth inhibition of human LO2 cells at 5 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID98815Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 208 mg/kg dose (post inoculation life span, days)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID96703Antitumor activity was determined against the ic implanted L1210 lymphoid leukemia in mice by range in survival days at 300 mg/kg (treatment of 1-5qd schedule) dosage; Range is 15-181986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID138740Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change) at the dose of 50 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID100211Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose of 5 mg/kg/day x 5 was determined as mean survival of treated group compared to untreated; 13.5/91980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID210535Phosphorylating activity of viral HSV -1 (KOS) induced thymidine kinase relative to dThdK was determined1987Journal of medicinal chemistry, Nov, Volume: 30, Issue:11
Acetylenic nucleosides. 4. 1-beta-D-arabinofuranosyl-5-ethynylcytosine. Improved synthesis and evaluation of biochemical and antiviral properties.
AID625288Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for jaundice2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1229555Cytotoxicity against human A549 cells assessed as inhibition of cell proliferation after 72 hrs by sulforhodamine B assay2015Journal of medicinal chemistry, Jun-25, Volume: 58, Issue:12
Structure-Activity Relationships of Neplanocin A Analogues as S-Adenosylhomocysteine Hydrolase Inhibitors and Their Antiviral and Antitumor Activities.
AID72417Tested in vitro against human ovarian cancer cells (G-401) for anticancer activity at dose 0 uM (Cell number 1*10 e-4)1994Journal of medicinal chemistry, Oct-14, Volume: 37, Issue:21
Synthesis of N-acetylglucosamine-modified ara-C and its effect on ovarian cancer cells.
AID1474166Liver toxicity in human assessed as induction of drug-induced liver injury by measuring severity class index2016Drug discovery today, Apr, Volume: 21, Issue:4
DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans.
AID284053Growth inhibition of human A431 cells by MTT assay2007Bioorganic & medicinal chemistry, Jan-01, Volume: 15, Issue:1
Antitumor studies. Part 1: design, synthesis, antitumor activity, and AutoDock study of 2-deoxo-2-phenyl-5-deazaflavins and 2-deoxo-2-phenylflavin-5-oxides as a new class of antitumor agents.
AID274781Ratio of AUC for araCTP level in liver to bone marrow of Swiss Webster mouse2006Journal of medicinal chemistry, Dec-28, Volume: 49, Issue:26
Synthesis and characterization of a novel liver-targeted prodrug of cytosine-1-beta-D-arabinofuranoside monophosphate for the treatment of hepatocellular carcinoma.
AID654375Growth inhibition of human NUGC3 cells after 24 hrs by WST-1 assay2012Journal of natural products, Feb-24, Volume: 75, Issue:2
Pinophilins A and B, inhibitors of mammalian A-, B-, and Y-family DNA polymerases and human cancer cell proliferation.
AID608161Toxicity in ICR mouse xenografted with mouse S180 cells assessed as effect on liver weight at 2 umol/kg2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID133646Dose producing greatest increase in life span expressed as optimum dose (L1210/ara-C I) (qd 1-9)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID286459Cytotoxicity against SW1573 cells after 72 hrs by SRB assay2007Journal of medicinal chemistry, May-03, Volume: 50, Issue:9
In vitro optimization of non-small cell lung cancer activity with troxacitabine, L-1,3-dioxolane-cytidine, prodrugs.
AID201696In vitro inhibitory activity against Human colon adenocarcinoma (SW-480) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID451299Cytotoxicity against deoxycytidine kinase deficient and deoxynucleoside analogue-resistant human MESSA 10K cells after 3 days by MTT assay2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID98851Compound was tested for percent increase in life span against L1210 lymphoid leukemia1987Journal of medicinal chemistry, Feb, Volume: 30, Issue:2
Synthesis and antitumor activity of fluorine-substituted 4-amino-2(1H)-pyridinones and their nucleosides. 3-Deazacytosines.
AID116452Percent increase in life span at 71 mg/kg (292 umol) per day1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID1542687Induction of apoptosis in human U937 cells assessed as live cells at 0.3 uM measured after 1 day by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 95.05 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID87475The compound was tested in vitro for antitumor activity(50% inhibition of tumor cell growth) against HeLa human cell lines after 72 hr1987Journal of medicinal chemistry, Sep, Volume: 30, Issue:9
Improved synthesis and antitumor activity of 1-deazaadenosine.
AID130640Antitumor activity against L1210 leukemia in mice (Percent increase in life span.) at the dose of 50 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID202209In vitro inhibitory effect on growth of human colon adenocarcinoma cell lines (SW480).1988Journal of medicinal chemistry, Jun, Volume: 31, Issue:6
Design, synthesis, and antineoplastic activity of 2'-deoxy-2'-methylidenecytidine.
AID117642In vivo activity against transplanted Mam-16/C/ Taxol tumors of mice; Not active2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID124254In vivo activity against iv transplanted Leukemia L1210 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID95310Tested for in vitro cytotoxicity against KATO III human stomach adenocarcinoma cell line1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID94982Effect of compound on erythroid differentiation of K562 cells was determined after a period of 5 days at concentration 500 nM1999Bioorganic & medicinal chemistry letters, Nov-01, Volume: 9, Issue:21
Induction of erythroid differentiation of human K562 cells by 3-O-acyl-1,2-O-isopropylidene-D-glucofuranose derivatives.
AID39130In vitro inhibitory activity against Mouse melanoma (B16) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID427704Cytotoxicity against human T47D cells by sulforhodamine B method2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Discovery of a new template for anticancer agents: 2'-deoxy-2'-fluoro-4'-selenoarabinofuranosyl-cytosine (2'-F-4'-seleno-ara-C).
AID97625Antitumor activity was determined against the ip implanted L1210 leukemia in mice by percent median survival rate (treated/control) at 200 mg/kg (treatment of 1-5qd schedule) dosage; 17.0/7.01986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1222345Drug metabolism assessed as intrinsic clearance for human recombinant N-terminal His-tagged DCK Ile24Val mutant-mediated cytosine-beta-D-arabinofuranoside-5'-MP formation expressed in Escherichia coli BL21(DE3) incubated for 3 mins by liquid chromatograph2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID380586Cytotoxicity against human HeLa S3 cells1999Journal of natural products, Jul, Volume: 62, Issue:7
A cytotoxic triterpene saponin from the root bark of Aralia dasyphylla.
AID625279Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for bilirubinemia2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1589122Cytotoxicity in human NHDF cells assessed as reduction in cell viability incubated for 72 hrs by SRB assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID445922Cytotoxicity against human HeLa cells assessed as inhibition of cell proliferation after 48 hrs2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
Application of the dipeptidyl peptidase IV (DPPIV/CD26) based prodrug approach to different amine-containing drugs.
AID1079948Times to onset, minimal and maximal, observed in the indexed observations. [column 'DELAI' in source]
AID1123318Antiviral activity against Vesicular stomatitis virus infected in primary rabbit kidney cells assessed as inhibition of virus-induced cytopathogenicity treated immediately after virus adsorption measured 2 to 3 days after virus infection1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID89827In vitro inhibitory effect measured against human osteosarcoma U20-S tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID496821Antimicrobial activity against Leishmania2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID99510The compound was tested for anticancer activity against L5178Y (arc-C resistant) cell lines of mouse leukemia1980Journal of medicinal chemistry, Oct, Volume: 23, Issue:10
Nucleosides. 116. 1-(beta-D-Xylofuranosyl)-5-fluorocytosines with a leaving group on the 3' position. Potential double-barreled masked precursors of anticancer nucleosides.
AID608262Toxicity in ICR mouse xenografted with mouse S180 cells assessed as increase in body weight at 2 umol/kg2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID1360833Cytotoxicity against human SNU638 cells assessed as decrease in cell viability after 72 hrs by SRB assay
AID1589118Cytotoxicity in human KB cells assessed as reduction in cell viability incubated for 72 hrs by SRB assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID152249In vitro inhibitory activity against Human melanoma (P-36) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID1124453Antiviral activity against Herpes simplex virus type 1 infected in Swiss albino mouse assessed as increase in mouse survival at 0.033 mg/kg, ic single dose after 6 hrs of infection relative to control1979Journal of medicinal chemistry, May, Volume: 22, Issue:5
Synthesis and antitumor and antiviral activities of 1-beta-D-arabinofuranosyl-2-amino-1,4(2H)-iminopyrimidine and its derivatives.
AID95143Inhibition of cell growth against human myeloid leukemia K562(S) cells1999Bioorganic & medicinal chemistry letters, Nov-01, Volume: 9, Issue:21
Induction of erythroid differentiation of human K562 cells by 3-O-acyl-1,2-O-isopropylidene-D-glucofuranose derivatives.
AID100508The compound was tested in vitro for antitumor activity(50% inhibition of tumor cell growth) against the L1210 murine cell lines after 72 hours1987Journal of medicinal chemistry, Sep, Volume: 30, Issue:9
Improved synthesis and antitumor activity of 1-deazaadenosine.
AID122446Median survival day of mice bearing lymphoid leukemia was determined, treatment schedule is 1-5 qd.; 15.5/9.51985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID55385Inhibition of cytidine deaminase partially purified from mouse kidney.1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID124636Weight change observed on day 8 in mice bearing lymphoid leukemia, treatment schedule is 1-5 qd.1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID355581Antiproliferative activity against human HT-29 cells at 0.1 ug after 48 hrs by two-layer agar-diffusion method
AID147784Tested in vitro against OST human osteosarcoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID380585Cytotoxicity against human KB cells1999Journal of natural products, Jul, Volume: 62, Issue:7
A cytotoxic triterpene saponin from the root bark of Aralia dasyphylla.
AID280941Cytotoxicity against mouse L1210/0 cells after 48 hrs2007Journal of medicinal chemistry, Apr-05, Volume: 50, Issue:7
Probing the anticancer activity of nucleoside analogues: a QSAR model approach using an internally consistent training set.
AID608263Toxicity in ICR mouse xenografted with mouse S180 cells assessed as increase in spleen index at 2 umol/kg2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID135303No of survivors after 45 days at a dose of 300 mg/kg/day1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID769614Cytotoxicity against human MCF7 cells after 72 hrs by SRB assay2013European journal of medicinal chemistry, Sep, Volume: 67Synthesis of 3'-azido-2',3'-dideoxy-5-fluorouridine phosphoramidates and evaluation of their anticancer activity.
AID95309In vitro inhibitory activity against Human gastric carcinoma (KATO III) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID1135061Toxicity in C3D2F1/J mouse allografted with mouse L1210 cells assessed as body weight change at 18.2 mg/kg, ip qd administered for 5 days measured on day 8 (Rvb = 1.81 +/- 0.79 g)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID977599Inhibition of sodium fluorescein uptake in OATP1B1-transfected CHO cells at an equimolar substrate-inhibitor concentration of 10 uM2013Molecular pharmacology, Jun, Volume: 83, Issue:6
Structure-based identification of OATP1B1/3 inhibitors.
AID320429Antitumor activity against human KB cells by MTT assay2008Bioorganic & medicinal chemistry, Jan-15, Volume: 16, Issue:2
Antitumor studies. Part 4: Design, synthesis, antitumor activity, and molecular docking study of novel 2-substituted 2-deoxoflavin-5-oxides, 2-deoxoalloxazine-5-oxides, and their 5-deaza analogs.
AID608260Toxicity in ICR mouse xenografted with mouse S180 cells assessed as effect on brain weight at 2 umol/kg2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID210866Rate of phosphorylation relative to human MitoThdK was determined1987Journal of medicinal chemistry, Nov, Volume: 30, Issue:11
Acetylenic nucleosides. 4. 1-beta-D-arabinofuranosyl-5-ethynylcytosine. Improved synthesis and evaluation of biochemical and antiviral properties.
AID1124450Antiviral activity against Myxoma virus infected in human KB cells assessed as inhibition of virus-induced cytopathic effect after 72 hrs by microscopically1979Journal of medicinal chemistry, May, Volume: 22, Issue:5
Synthesis and antitumor and antiviral activities of 1-beta-D-arabinofuranosyl-2-amino-1,4(2H)-iminopyrimidine and its derivatives.
AID43713In vitro cytotoxicity of compounds were determined on Inhibition of human leukemia cell line(CCRF-CEM). 1996Journal of medicinal chemistry, Nov-08, Volume: 39, Issue:23
Synthesis and biological activity of aromatic amino acid phosphoramidates of 5-fluoro-2'-deoxyuridine and 1-beta-arabinofuranosylcytosine: evidence of phosphoramidase activity.
AID124266In vivo activity against transplanted colon-38 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID434136Lipophilicity, log P of the compound2009European journal of medicinal chemistry, Sep, Volume: 44, Issue:9
Synthesis and evaluation of anti-tumor activities of N4 fatty acyl amino acid derivatives of 1-beta-arabinofuranosylcytosine.
AID130616Antitumor activity against L1210 leukemia in mice (Percent increase in life span.) at the dose of 1000 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID274778Drug level in Swiss Webster mouse plasma at 100 mg/kg, ip measured as AUC (0-4h)2006Journal of medicinal chemistry, Dec-28, Volume: 49, Issue:26
Synthesis and characterization of a novel liver-targeted prodrug of cytosine-1-beta-D-arabinofuranoside monophosphate for the treatment of hepatocellular carcinoma.
AID274771Production of araCTP in rat hepatocytes at 100 uM measured as AUC (0-4h)2006Journal of medicinal chemistry, Dec-28, Volume: 49, Issue:26
Synthesis and characterization of a novel liver-targeted prodrug of cytosine-1-beta-D-arabinofuranoside monophosphate for the treatment of hepatocellular carcinoma.
AID364007Inhibition of pp60c-src2008European journal of medicinal chemistry, Jul, Volume: 43, Issue:7
Antitumor studies -- part 2: structure-activity relationship study for flavin analogs including investigations on their in vitro antitumor assay and docking simulation into protein tyrosine kinase.
AID108521Number of animals survived after 30-days in mice bearing lymphoid leukemia, treatment schedule is 1-5 qd.1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID1473739Inhibition of human MRP2 overexpressed in Sf9 cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 20 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID1123148Antiviral activity against HSV1 2931 infected in African green monkey Vero cells assessed as inhibition of virus replication at 1 ug/ml after 24 hrs relative to control1979Journal of medicinal chemistry, Jan, Volume: 22, Issue:1
Nucleosides. 110. Synthesis and antiherpes virus activity of some 2'-fluoro-2'-deoxyarabinofuranosylpyrimidine nucleosides.
AID1632124Cytotoxicity against human MCF7 cells assessed as reduction in cell survival after 72 hrs by sulforhodamine B assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID608165Antiproliferative activity against mouse H22 cells after 48 hrs by MTT assay2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID97620Antitumor activity was determined against the ic implanted L1210 lymphoid leukemia in mice by percent median survival rate (treated/control) at 300 mg/kg (treatment of 1-5qd schedule) dosage; 16.0/7.01986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID135302No of survivors after 45 days at a dose of 200 mg/kg/day1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID167740Concentration required to reduce protein synthesis in primary rabbit kidney cells usingi [3H2-4,5]leucine incorporation1986Journal of medicinal chemistry, Feb, Volume: 29, Issue:2
Systematic synthesis and biological evaluation of alpha- and beta-D-xylofuranosyl nucleosides of the five naturally occurring bases in nucleic acids and related analogues.
AID205131Compound was tested for the inhibition of small cell lung cancer1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID130613The compound was tested in vitro for antitumor activity(50% inhibition of tumor cell growth) against the P388/doxorubicin resistant murine cell lines after 72 hours1987Journal of medicinal chemistry, Sep, Volume: 30, Issue:9
Improved synthesis and antitumor activity of 1-deazaadenosine.
AID1292260Cytotoxicity against human HepG2 cells after 72 hrs by SRB assay2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID1772825Antitumor activity against human MV4-11 cells xenografted in female nude mouse assessed as tumor weight growth inhibition at 100 mg/kg, ip measured after 21 days2021European journal of medicinal chemistry, Nov-05, Volume: 223Discovery of a potent MLL1 and WDR5 protein-protein interaction inhibitor with in vivo antitumor activity.
AID625287Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatomegaly2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID123365Dose producing greatest increase in life span of mice bearing lymphoid leukemia, treatment schedule is 1-5 qd.1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID156495In vitro inhibitory effect on growth of human lung squamous cell carcinoma cell lines (PC10).1988Journal of medicinal chemistry, Jun, Volume: 31, Issue:6
Design, synthesis, and antineoplastic activity of 2'-deoxy-2'-methylidenecytidine.
AID1134720Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 40 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID687166Ratio of IC50 for human CCRF-CEM/C2 cells IC50 for human CCRF-CEM cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID654374Growth inhibition of human HeLa cells after 24 hrs by WST-1 assay2012Journal of natural products, Feb-24, Volume: 75, Issue:2
Pinophilins A and B, inhibitors of mammalian A-, B-, and Y-family DNA polymerases and human cancer cell proliferation.
AID138736Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change) at the dose of 250 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1407816Antiproliferative activity against human HL60 cells2018European journal of medicinal chemistry, Sep-05, Volume: 157Pyrrolopyrimidines: An update on recent advancements in their medicinal attributes.
AID138410Antitumor activity against L1210 leukemia in mice (Average 7-day weight change (%)) at the dose of 1000 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1135906Antiviral activity against Vaccinia virus infected in rabbit primary kidney cells assessed as inhibition of viral cytopathogenicity after 3 days1979Journal of medicinal chemistry, May, Volume: 22, Issue:5
Antiviral activity of aliphatic nucleoside analogues: structure-function relationship.
AID679291TP_TRANSPORTER: uptake in Xenopus laevis oocytes2000Biochemical pharmacology, Jul-15, Volume: 60, Issue:2
Role of organic cation transporters in the renal secretion of nucleosides.
AID124265In vivo activity against transplanted colon-26 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID138739Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change) at the dose of 500 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID431399Cytotoxicity against human HL60/MX2 cells after 48 hrs by cell titer-blue assay2009Journal of medicinal chemistry, Oct-08, Volume: 52, Issue:19
Structure-activity relationship and molecular mechanisms of ethyl 2-amino-4-(2-ethoxy-2-oxoethyl)-6-phenyl-4h-chromene-3-carboxylate (sha 14-1) and its analogues.
AID1360519Cytotoxicity against human HeLa cells2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID1150984Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 100 umol/kg/day, po using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation1976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID355632Cytotoxicity against mouse L1210 cells at 0.15 ug/disk by corbett disk diffusion assay
AID1145825Antitumor activity against mouse L1210 cells allografted in BDF1 mouse assessed as increase in life span at 100 mg/kg, ip administered as single dose relative to vehicle-treated control1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID209962In vitro inhibitory activity against Human bladder transitional-cell carcinoma (T-24) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID613827Ratio of IC50 for vinblastine100-resistant human CCRF-CEM cells to IC50 for human CCRF-CEM cells2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Structure-activity relationship and molecular mechanisms of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and its analogues.
AID120036Median survival day of mice bearing lymphoid leukemia was determined, treatment schedule is 1-9 qd.; 25/10.51985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID105082In vitro inhibitory effect on growth of human T-cell acute lymphoblastic leukemic MOLT 4 cell lines.1988Journal of medicinal chemistry, Jun, Volume: 31, Issue:6
Design, synthesis, and antineoplastic activity of 2'-deoxy-2'-methylidenecytidine.
AID451301Cytotoxicity against deoxynucleoside analogue-resistant human MCF7 1K cells overexpressing ribonucleotide reductase after 3 days by MTT assay2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID284054Growth inhibition of human CCRF-HSB-2 cells by MTT assay2007Bioorganic & medicinal chemistry, Jan-01, Volume: 15, Issue:1
Antitumor studies. Part 1: design, synthesis, antitumor activity, and AutoDock study of 2-deoxo-2-phenyl-5-deazaflavins and 2-deoxo-2-phenylflavin-5-oxides as a new class of antitumor agents.
AID678715Inhibition of human CYP2D6 assessed as ratio of IC50 in absence of NADPH to IC50 for presence of NADPH using 4-methylaminoethyl-7-methoxycoumarin as substrate after 30 mins2012Chemical research in toxicology, Oct-15, Volume: 25, Issue:10
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
AID1360832Cytotoxicity against human HCT116 cells assessed as decrease in cell viability after 72 hrs by SRB assay
AID477295Octanol-water partition coefficient, log P of the compound2010European journal of medicinal chemistry, Apr, Volume: 45, Issue:4
QSPR modeling of octanol/water partition coefficient of antineoplastic agents by balance of correlations.
AID55393Inhibitory constant was measured on cytidine/deoxycytidine deaminase1991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID214573Concentration required for 50% inhibition of cell growth of U937 leukemic cell lines in human.1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID431396Cytotoxicity against human CCRF-CEM/C2 cells after 48 hrs by cell titer-blue assay2009Journal of medicinal chemistry, Oct-08, Volume: 52, Issue:19
Structure-activity relationship and molecular mechanisms of ethyl 2-amino-4-(2-ethoxy-2-oxoethyl)-6-phenyl-4h-chromene-3-carboxylate (sha 14-1) and its analogues.
AID138717Antitumor activity against L1210/ara-C leukemia in mice (Average 7-day weight change (%)) at the dose of 1000 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID687168Ratio of IC50 for human CCRF-CEM/VLB100 cells IC50 for human CCRF-CEM cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID138896Mean body weight change on 17th day at a dose of 200 mg/kg/day1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID86027Antiviral activity against Herpes simplex virus -1 strain KOS was evaluated in african green monkey kidney1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID153001Antitumor activity was determined against the ip implanted P388 leukemia in mice by 45-day survivors in mice at 200 mg/kg (treatment of 1-5qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID225890Compound was tested for the inhibition of non- small cell lung cancer1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID97216Antitumor activity against ic implanted L1210 lymphoid leukemia in mice expressed as active dose range (1-5qd treatment schedule) required to produce increase in life span by >25% was determined; Range is 50-3001986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1589121Cytotoxicity in human HepG2 cells assessed as reduction in cell viability incubated for 72 hrs by SRB assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID153285Antitumor activity was determined against the ip implanted P388 leukemia in mice by percent increase in life span at 200 mg/kg (treatment of 1-5qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1134730Toxicity in CDF1 mouse assessed as change in body weight at 200 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID101605Tested in vitro against MCF-7 human breast adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID608162Antiproliferative activity against mouse S180 cells after 48 hrs by MTT assay2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID1135089Toxicity in C3D2F1/J mouse allografted with mouse L1210 cells assessed as body weight change at 18.3 mg/kg, ip qd administered for 5 days measured on day 8 co-administered with 27 mg/kg prednisone (Rvb = 1.81 +/- 0.79 g)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID99916Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 25 mg/kg/day x 5 was determined as Survival time in days; Range is 9-16 days1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID1740805Selectivity index, ratio of IC50 for cytotoxicity against human HDF cells to IC50 for cytotoxicity against human U-87 MG cells2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID97111Antitumor activity was determined against the ip implanted L1210 leukemia in mice by percent increase in life span at 200 mg/kg (treatment of 1-5qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID82992Concentration required for 50% inhibition of cell growth of HL60 leukemic cell lines in human.1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID1222335Ratio of Vmax for human recombinant N-terminal His-SUMO fused wild type CDA expressed in Escherichia coli BL21(DE3) cells to Vmax for human recombinant N-terminal His-SUMO fused CDA Ala70Thr mutant expressed in Escherichia coli BL21(DE3) cells2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID496817Antimicrobial activity against Trypanosoma cruzi2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID89624In vitro inhibitory effect measured against human osteosarcoma SK-ES-1 tumor cells1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID163147Tested in vitro against QG-95 human squamous cell carcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID392723Antiviral activity against HSV2 assessed as inhibition of virus-induced cytopathicity2009Bioorganic & medicinal chemistry letters, Feb-15, Volume: 19, Issue:4
Synthesis of 5-isoxazol-5-yl-2'-deoxyuridines exhibiting antiviral activity against HSV and several RNA viruses.
AID1135042Cytotoxicity against mouse L1210 cells assessed as growth inhibition measured at 72 hrs by trypan blue exclusion assay1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID100193Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose of 18.2 mg/kg/day x 5 was determined as mean survival of treated group compared to untreated; 14.5/101980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID98043In vitro cytotoxicity with cleavage by phosphodiesterase-I concentration for 50% loss of viability was determined at 72 hours.1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID625290Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for liver fatty2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1134741Toxicity in CDF1 mouse assessed as change in body weight at 3.12 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID202613Effective dose to inhibit the replication of S180 cell line in vitro.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Synthesis and anticancer activity of various 3'-deoxy pyrimidine nucleoside analogues and crystal structure of 1-(3-deoxy-beta-D-threo-pentofuranosyl)cytosine.
AID687167Ratio of IC50 for human CCRF-CEM/VM-1-5 cells IC50 for human CCRF-CEM cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID132917Percentage increase in life span after 17-21 days1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID138557Antitumor activity against L1210 leukemia in mice (Average 7-day weight change (%)) at the dose of 200 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID127909Antitumor activity against L1210 leukemia in mice (Average weight per mouse on day 1.) at the dose of 100 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID436683Antitumor activity against mouse S180 cells xenografted in ICR mouse assessed as decrease in tumor weight at 89 umol/kg, ip administered 24 hrs after tumor implantation once daily for 7 consecutive days measured after 24 hrs last post dose2009European journal of medicinal chemistry, Oct, Volume: 44, Issue:10
Novel N-(3-carboxyl-9-benzyl-beta-carboline-1-yl)ethylamino acids: synthesis, anti-tumor evaluation, intercalating determination, 3D QSAR analysis and docking investigation.
AID140007Death due to toxicity occurring at a maximum body weight decrease of 10 to 22% from the average post injection body weight at 75 mg/kg; 5 / 51991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID1159294Cytotoxicity against human SNU638 cells by SRB assay2014European journal of medicinal chemistry, Aug-18, Volume: 83Structure-activity relationships of 2'-modified-4'-selenoarabinofuranosyl-pyrimidines as anticancer agents.
AID1134735Toxicity in CDF1 mouse assessed as change in body weight at 25 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID89121In vitro inhibitory effect measured against human stomach adenocarcinoma NUGC-4 tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID1292258Cytotoxicity against human KB cells after 72 hrs by SRB assay2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID152367Antitumor activity against ip implanted P388 leukemia in mice expressed as active dose range (1 qd treatment schedule) required to produce increase in life span by >25% was determined; Range is 100-6001986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1124447Antiviral activity against Herpes simplex virus type 1 infected in human KB cells assessed as inhibition of virus-induced cytopathic effect after 72 hrs by microscopically1979Journal of medicinal chemistry, May, Volume: 22, Issue:5
Synthesis and antitumor and antiviral activities of 1-beta-D-arabinofuranosyl-2-amino-1,4(2H)-iminopyrimidine and its derivatives.
AID1134732Toxicity in CDF1 mouse assessed as change in body weight at 80 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID120462Number of mice survived after 30 days1982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID80938Inhibitory activity against HL-60 (human, promyelocytic leukemia) cell line2003Journal of medicinal chemistry, Sep-11, Volume: 46, Issue:19
Synthesis and cytotoxic activity of two novel 1-dodecylthio-2-decyloxypropyl-3-phosphatidic acid conjugates with gemcitabine and cytosine arabinoside.
AID97106Antitumor activity was determined against the ic implanted L1210 lymphoid leukemia in mice by percent increase in life span at 300 mg/kg (treatment of 1-5qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID104931Tested in vitro against MNNG/HOS cell line human osteosarcoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID678716Inhibition of human CYP3A4 assessed as ratio of IC50 in absence of NADPH to IC50 for presence of NADPH using diethoxyfluorescein as substrate after 30 mins2012Chemical research in toxicology, Oct-15, Volume: 25, Issue:10
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
AID133369Range of survival days for antitumor activity against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 400 mg/kg per day after treatment 1; range is 8-101990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID1542688Induction of apoptosis in human U937 cells assessed as early apoptotic cells at 0.3 uM measured after 1 day by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 0.7 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID1079942Steatosis, proven histopathologically. Value is number of references indexed. [column 'STEAT' in source]
AID445923Cytotoxicity against human CEM cells assessed as inhibition of cell proliferation after 48 hrs2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
Application of the dipeptidyl peptidase IV (DPPIV/CD26) based prodrug approach to different amine-containing drugs.
AID89107In vitro inhibitory effect measured against human osteosarcoma MG-63 tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID81636The cytotoxic activity in HL-60 cells1997Journal of medicinal chemistry, Nov-07, Volume: 40, Issue:23
Retinoic acid conjugates as potential antitumor agents: synthesis and biological activity of conjugates with Ara-A, Ara-C, 3(2H)-furanone, and aniline mustard moieties.
AID42636Percent inhibition of [123I]UdR incorporation in the DNA synthesis assay in bone marrow cells at a concentration of 10 e-3 M1983Journal of medicinal chemistry, Nov, Volume: 26, Issue:11
Synthesis and biological evaluation of sparsomycin analogues.
AID94978Effect of compound on erythroid differentiation of K562 cells was determined after a period of 3 days at concentration 500 nM1999Bioorganic & medicinal chemistry letters, Nov-01, Volume: 9, Issue:21
Induction of erythroid differentiation of human K562 cells by 3-O-acyl-1,2-O-isopropylidene-D-glucofuranose derivatives.
AID497005Antimicrobial activity against Pneumocystis carinii2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID128075Antitumor activity against L1210/ara-C leukemia in mice (Average weight per mouse on day 1.) at the dose of 50 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1292262Selectivity index, ratio of IC50 for HDF cells to IC50 for human HeLa cells2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID1151005Immunosuppression activity in rat assessed as reduction of adjuvant-induced polyarthritis administered po for days 1 to 5 measured on day 211976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID128072Antitumor activity against L1210/ara-C leukemia in mice (Average weight per mouse on day 1.) at the dose of 250 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID124255In vivo activity against transplanted Mam-16/C tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID1151001Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 400 umol/kg/day, ip using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation1976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID1632128Selectivity index, ratio of IC50 for HDF to IC50 for human HeLa cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID1150998Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 300 umol/kg/day, po using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation in presence of THU 10 mg/kg/day on days 1 to 51976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID150696In vitro antitumor activity against P388 cell line in mice.1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID105669In vitro lethal dose was measured in mouse myeloma MPC-11 cell line1982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID608164Antiproliferative activity against mouse B16 cells after 48 hrs by MTT assay2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID1222337Ratio of Km for human recombinant N-terminal His-tagged wild type DCK expressed in Escherichia coli BL21(DE3) cells to Km for human recombinant N-terminal His-tagged DCK Pro122Ser mutant expressed in Escherichia coli BL21(DE3) cells2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID274779Distribution of araCTP in Swiss Webster mouse bone marrow at 100 mg/kg, ip measured as AUC (0-4h)2006Journal of medicinal chemistry, Dec-28, Volume: 49, Issue:26
Synthesis and characterization of a novel liver-targeted prodrug of cytosine-1-beta-D-arabinofuranoside monophosphate for the treatment of hepatocellular carcinoma.
AID445924Cytotoxicity against mouse L1210 cells assessed as inhibition of cell proliferation after 48 hrs2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
Application of the dipeptidyl peptidase IV (DPPIV/CD26) based prodrug approach to different amine-containing drugs.
AID1360502Growth inhibition of human LO2 cells at 0.5 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID687158Cytotoxicity against human K562 cells assessed as cell viability after 48 hrs by celltiter-blue assay2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID124267In vivo activity against transplanted colon-51 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID43702Cytotoxicity against CCRF-CEM human leukemic lymphoblastoid cell lines1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID625284Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatic failure2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID411723Antiproliferative activity against human 2209-23 cells by [3H]thymidine incorporation scintillation proximity assay2009Journal of medicinal chemistry, Jan-08, Volume: 52, Issue:1
The design, synthesis, and antiviral activity of 4'-azidocytidine analogues against hepatitis C virus replication: the discovery of 4'-azidoarabinocytidine.
AID1589123Selectivity index, ratio of IC50 for cytotoxicity in human NHDF cells to IC50 for cytotoxicity in human HeLa cells2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID55974Rate of deamination was determined against deaminase purified from human acute myeloblastic leukemia cells1987Journal of medicinal chemistry, Nov, Volume: 30, Issue:11
Acetylenic nucleosides. 4. 1-beta-D-arabinofuranosyl-5-ethynylcytosine. Improved synthesis and evaluation of biochemical and antiviral properties.
AID201988In vitro cytotoxicity was evaluated against melanoma SK-MEL-28 cells2000Journal of medicinal chemistry, Apr-20, Volume: 43, Issue:8
Synthesis and evaluation of several new (2-chloroethyl)nitrosocarbamates as potential anticancer agents.
AID678714Inhibition of human CYP2C19 assessed as ratio of IC50 in absence of NADPH to IC50 for presence of NADPH using 3-butyryl-7-methoxycoumarin as substrate after 30 mins2012Chemical research in toxicology, Oct-15, Volume: 25, Issue:10
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
AID98958Antitumor activity against the ip implanted L1210 leukemia in mice expressed as optimal dose (1 qd treatment schedule) required to produce greatest increase in life span was determined1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID338731Cytotoxicity against mouse L1210 cells after 3 days by trypan blue exclusion technique
AID1150999Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 100 umol/kg/day, ip using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation1976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID81470In vitro inhibitory activity against Human promyelocytic leukemia (HL-60) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID97217Antitumor activity against ic implanted L1210 lymphoid leukemia in mice expressed as active dose range (1-9qd treatment schedule) required to produce increase in life span by >25% was determined; Range is 100-3001986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID361404Antiviral activity against HSV1 F infected in african green monkey Vero cells assessed as inhibition of virus-induced cytopathic effect after 3 days by MTT assay2002Journal of natural products, Nov, Volume: 65, Issue:11
New saponins from the starfish Certonardoa semiregularis.
AID116600Percent increase in life span in mice bearing lymphoid leukemia, treatment schedule is 1-5 qd.1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID81282Anticancer activity was evaluated for the compound against parent promyelocytic leukemia cell line (HL-60-GHPRT+/dCK+)1986Journal of medicinal chemistry, Jul, Volume: 29, Issue:7
Synthesis and biological activity of 5-phenylselenenyl-substituted pyrimidine nucleosides.
AID1145828Antiviral activity against Vaccinia virus IHD infected in African green monkey BGM cells assessed as plaque reduction measured on day 5 by disk diffusion assay1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID1123324Antiviral activity against Herpes simplex virus-2 196 infected in primary rabbit kidney cells assessed as inhibition of virus-induced cytopathogenicity treated immediately after virus adsorption measured 2 to 3 days after virus infection1979Journal of medicinal chemistry, Jun, Volume: 22, Issue:6
Synthesis and antiviral activities of arabinofuranosyl-5-ethylpyrimidine nucleosides. Selective antiherpes activity of 1-(beta-D-arabinofuranosyl)-5-ethyluracil.
AID100344Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose of 9.2 mg/kg/day x 5 was determined as mean survival of treated group compared to untreated; 17/91980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID96544Antitumor activity was determined against the ic implanted L1210 lymphoid leukemia in mice by change in tumor weight at 300 mg/kg (treatment of 1-5qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1740799Antiproliferative activity against human U-87 MG cells assessed as reduction in cell growth incubated for 72 hrs by SRB assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID217969In vitro antiviral activity against Vaccinia virus strain Elstree was evaluated in african green monkey kidney1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID128073Antitumor activity against L1210/ara-C leukemia in mice (Average weight per mouse on day 1.) at the dose of 468 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID23271Partition coefficient (logD7.4)1990Journal of medicinal chemistry, Jul, Volume: 33, Issue:7
Structure-activity relationships of antineoplastic agents in multidrug resistance.
AID1145780Half life in human1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. 6. Synthesis and biological evaluation of some 3'-acyl derivatives of 2,2'-anhydro-1-(beta-D-arabinofuranosyl)cytosine hydrochloride.
AID1229556Cytotoxicity against human HCT116 cells assessed as inhibition of cell proliferation after 72 hrs by sulforhodamine B assay2015Journal of medicinal chemistry, Jun-25, Volume: 58, Issue:12
Structure-Activity Relationships of Neplanocin A Analogues as S-Adenosylhomocysteine Hydrolase Inhibitors and Their Antiviral and Antitumor Activities.
AID678717Inhibition of human CYP3A4 assessed as ratio of IC50 in absence of NADPH to IC50 for presence of NADPH using 7-benzyloxyquinoline as substrate after 30 mins2012Chemical research in toxicology, Oct-15, Volume: 25, Issue:10
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
AID1150992Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 400 umol/kg/day, po using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation1976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID433259Cytotoxicity against human H441 cells by serum lactate dehydrogenase assay2009Bioorganic & medicinal chemistry letters, Aug-15, Volume: 19, Issue:16
Synthesis of radiolabeled cytarabine conjugates.
AID1222347Drug metabolism assessed as intrinsic clearance for human recombinant N-terminal His-tagged wild type DCK-mediated cytosine-beta-D-arabinofuranoside-5'-MP formation expressed in Escherichia coli BL21(DE3) incubated for 3 mins by liquid chromatography-tand2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID137179Antitumor activity against L1210/ara-C leukemia in mice (median survival) at the dose of 500 mg/kg; Value represented as Test/Control = 9/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1159295Cytotoxicity against human T47D cells by SRB assay2014European journal of medicinal chemistry, Aug-18, Volume: 83Structure-activity relationships of 2'-modified-4'-selenoarabinofuranosyl-pyrimidines as anticancer agents.
AID132043Increase of life span was determined against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 400 mg/kg per day after treatment 11990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID1589120Cytotoxicity in human U87 cells assessed as reduction in cell viability incubated for 72 hrs by SRB assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID208044Anticancer activity carried out in vitro against human T-lymphocyte Molt4/C82001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Synthesis and biological evaluation of purine-containing butenolides.
AID1134731Toxicity in CDF1 mouse assessed as change in body weight at 100 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID156496Concentration required for 50% inhibition of human lung squamous cell carcinoma PC10 solid cell lines. 1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID364006Growth inhibition of human KB cells by MTT assay2008European journal of medicinal chemistry, Jul, Volume: 43, Issue:7
Antitumor studies -- part 2: structure-activity relationship study for flavin analogs including investigations on their in vitro antitumor assay and docking simulation into protein tyrosine kinase.
AID678721Metabolic stability in human liver microsomes assessed as GSH adduct formation at 100 uM after 90 mins by HPLC-MS analysis2012Chemical research in toxicology, Oct-15, Volume: 25, Issue:10
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
AID1632123Cytotoxicity against human KB cells assessed as reduction in cell survival after 72 hrs by sulforhodamine B assay2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID1229559Cytotoxicity against human SKHEP1 cells assessed as inhibition of cell proliferation after 72 hrs by sulforhodamine B assay2015Journal of medicinal chemistry, Jun-25, Volume: 58, Issue:12
Structure-Activity Relationships of Neplanocin A Analogues as S-Adenosylhomocysteine Hydrolase Inhibitors and Their Antiviral and Antitumor Activities.
AID625292Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) combined score2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID1145835Antitumor activity against mouse L1210 cells allografted in BDF1 mouse assessed as survival at 200 mg/kg, ip administered as single dose measured on day 30 relative to vehicle-treated control1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID588212Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID1292261Cytotoxicity against HDF cells after 72 hrs by SRB assay2016Bioorganic & medicinal chemistry, 05-15, Volume: 24, Issue:10
Synthesis and anticancer activity of some 5-fluoro-2'-deoxyuridine phosphoramidates.
AID122628Change in weight of mouse on day 8 was measured in L1210 lymphoid leukemia inoculated mice after ip administration at dose of 10 mg/kg/day *51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID93850The compound was tested in vitro for antitumor activity(50% inhibition of tumor cell growth) against KB human cell lines after 72 hr1987Journal of medicinal chemistry, Sep, Volume: 30, Issue:9
Improved synthesis and antitumor activity of 1-deazaadenosine.
AID1135053Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as median survival time at 25 mg/kg, ip qd administered for 5 days (Rvb = 8 days)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID496825Antimicrobial activity against Leishmania mexicana2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID135300No of survivors after 45 days at a dose of 100 mg/kg/day1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID100490Inhibition of L1210 lymphoid leukemia cells 48 hr after administration (length of preincubation=0 hr)1992Journal of medicinal chemistry, Sep-04, Volume: 35, Issue:18
Synthesis and functional evaluation of a peptide derivative of 1-beta-D-arabinofuranosylcytosine.
AID100366Inhibition of L1210 lymphoid leukemia cells 24 hr after administration (length of preincubation=0 hr)1992Journal of medicinal chemistry, Sep-04, Volume: 35, Issue:18
Synthesis and functional evaluation of a peptide derivative of 1-beta-D-arabinofuranosylcytosine.
AID608168Antitumor activity against mouse S180 cells xenografted in ICR mouse assessed as tumor growth inhibition at 8.9 umol/kg, ip administered 1 day after tumor inoculation qd for 7 days2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID625280Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholecystitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID613826Ratio of IC50 for homoharringtonine300-resistant human K562 cells to IC50 for human K562 cells2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Structure-activity relationship and molecular mechanisms of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and its analogues.
AID1360835Cytotoxicity against human SKHEP1 cells assessed as decrease in cell viability after 72 hrs by SRB assay
AID98764Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 156 mg/kg dose(Mean increased life span, calculated as (T/C-1)X100)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID98765Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 208 mg/kg dose(Mean increased life span, calculated as (T/C-1)X100)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID588213Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in non-rodents2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID135736Toxicity in mice after 7-8 days at a dose of 200 mg/kg/day; 8/91988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1135058Toxicity in C3D2F1/J mouse allografted with mouse L1210 cells assessed as body weight change at 50 mg/kg, ip qd administered for 5 days measured on day 8 (Rvb = 1.81 +/- 0.79 g)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID1740801Cytotoxicity against human HDF cells assessed as reduction in cell growth incubated for 72 hrs by SRB assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID588211Literature-mined compound from Fourches et al multi-species drug-induced liver injury (DILI) dataset, effect in humans2010Chemical research in toxicology, Jan, Volume: 23, Issue:1
Cheminformatics analysis of assertions mined from literature that describe drug-induced liver injury in different species.
AID137180Antitumor activity against L1210/ara-C leukemia in mice (median survival) at the dose of 50 mg/kg; Value represented as Test/Control = 8/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1079937Severe hepatitis, defined as possibly life-threatening liver failure or through clinical observations. Value is number of references indexed. [column 'MASS' in source]
AID128063Antitumor activity against L1210 leukemia in mice (Average weight per mouse on day 1.) at the dose of 500 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID85582Tested in vitro against HT1080 cell line human fibrosarcoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID1740803Selectivity index, ratio of IC50 for cytotoxicity against human HDF cells to IC50 for cytotoxicity against human SK-OV-3 cells2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID1134722Antitumor activity in mouse L1210 cells allografted in CDF1 mouse assessed as increase in life span at 20 mg/kg, ip administered as qd for 9 days relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID89276In vitro inhibitory effect measured against human lung large-cell carcinoma PC-13 tumor cells at 100 ug/mL1991Journal of medicinal chemistry, Mar, Volume: 34, Issue:3
Nucleosides and nucleotides. 95. Improved synthesis of 1-(2-azido-2-deoxy-beta-D-arabinofuranosyl)cytosine (cytarazid) and -thymine. Inhibitory spectrum of cytarazid on the growth of various human tumor cells in vitro.
AID1360834Cytotoxicity against human MDA-MB-231 cells assessed as decrease in cell viability after 72 hrs by SRB assay
AID94833In vitro inhibitory effect on growth of human chronic myelogenous leukemic K562 cell lines.1988Journal of medicinal chemistry, Jun, Volume: 31, Issue:6
Design, synthesis, and antineoplastic activity of 2'-deoxy-2'-methylidenecytidine.
AID1079939Cirrhosis, proven histopathologically. Value is number of references indexed. [column 'CIRRH' in source]
AID100362Antitumor activity in mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 9.2 mg/kg/day x 5 was determined as weight change (g/mouse) on 8th day1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID427701Cytotoxicity against human HCT116 cells by sulforhodamine B method2009Journal of medicinal chemistry, Sep-10, Volume: 52, Issue:17
Discovery of a new template for anticancer agents: 2'-deoxy-2'-fluoro-4'-selenoarabinofuranosyl-cytosine (2'-F-4'-seleno-ara-C).
AID8811Tested in vitro against A-375 cell line human melanoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID152679Antitumor activity against the ip implanted P388 leukemia in mice expressed as optimal dose (1-5qd treatment schedule) required to produce greatest increase in life span was determined1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1145827Half life in human1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID94555Tested in vitro inhibitory activity against murine L 1210 leukemia cells line1993Journal of medicinal chemistry, Dec-10, Volume: 36, Issue:25
Synthesis and biological evaluation of N4-substituted imidazo- and v-triazolo[4,5-d]pyridazine nucleosides.
AID496830Antimicrobial activity against Leishmania major2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID1135057Toxicity in C3D2F1/J mouse allografted with mouse L1210 cells assessed as body weight change at 25 mg/kg, ip qd administered for 5 days measured on day 8 (Rvb = 1.81 +/- 0.79 g)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID1151000Antitumor activity against mouse L1210 cells allografted in mouse assessed as increase in life span at 200 umol/kg/day, ip using 0.9% (W/V) aqueous NaCl vehicle for 5 days 24 hrs after tumor inoculation1976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID1135088Toxicity in C3D2F1/J mouse allografted with mouse L1210 cells assessed as body weight change at 18.2 mg/kg, ip qd administered for 5 days measured on day 8 co-administered with 27 mg/kg prednisolone (Rvb = 1.81 +/- 0.79 g)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID94965Evaluated for erythroid induction of benzidine-positive K562 cells at concentration 3 mM1999Bioorganic & medicinal chemistry letters, Nov-01, Volume: 9, Issue:21
Induction of erythroid differentiation of human K562 cells by 3-O-acyl-1,2-O-isopropylidene-D-glucofuranose derivatives.
AID502462Cytotoxicity against human Bel7402 cells after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Sep-01, Volume: 18, Issue:17
A class of novel carboline intercalators: Their synthesis, in vitro anti-proliferation, in vivo anti-tumor action, and 3D QSAR analysis.
AID451312Resistance ratio of IC50 for deoxynucleoside analogue-resistant human MESSA 10K cells to IC50 for deoxynucleoside analogue-sensitive human MESSA cells2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID201189Tested in vitro against ST-KM human stomach adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID138583Antitumor activity against L1210 leukemia in mice (Average 7-day weight change) at the dose of 200 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID588220Literature-mined public compounds from Kruhlak et al phospholipidosis modelling dataset2008Toxicology mechanisms and methods, , Volume: 18, Issue:2-3
Development of a phospholipidosis database and predictive quantitative structure-activity relationship (QSAR) models.
AID1740797Antiproliferative activity against human SK-OV-3 cells assessed as reduction in cell growth incubated for 72 hrs by SRB assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID1740796Antiproliferative activity against human SK-BR-3 cells assessed as reduction in cell growth incubated for 72 hrs by SRB assay2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID1489833Antitumor activity against human MV4-11 cells xenografted in nu/nu mouse assessed as tumor growth inhibition at 50 mg/kg, iv qd measured on day 21 relative to vehicle-treated control2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Discovery of 4-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-N-(4-((4-methylpiperazin-1-yl)methyl)phenyl)-1H-pyrazole-3-carboxamide (FN-1501), an FLT3- and CDK-Kinase Inhibitor with Potentially High Efficiency against Acute Myelocytic Leukemia.
AID138066Survival time treated / survival time (T/C) for antitumor activity against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 400 mg/kg per day after treatment 5;10/71990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID130783Antitumor activity against L1210/ara-C leukemia in mice (Percent increase in life span.) at the dose of 500 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1151004Immunosuppression activity in rat assessed as reduction of adjuvant-induced polyarthritis administered po for days 1 to 5 measured on day 151976Journal of medicinal chemistry, Aug, Volume: 19, Issue:8
Nucleic acids. 16. Orally active derivatives of ara-cytidine.
AID1079935Cytolytic liver toxicity, either proven histopathologically or where the ratio of maximal ALT or AST activity above normal to that of Alkaline Phosphatase is > 5 (see ACUTE). Value is number of references indexed. [column 'CYTOL' in source]
AID1360831Cytotoxicity against human A549 cells assessed as decrease in cell viability after 72 hrs by SRB assay
AID124264In vivo activity against transplanted Squam lung-LC12 tumors of mice2001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Design, synthesis, and biological evaluation of analogues of the antitumor agent, 2-(4-[(7-chloro-2-quinoxalinyl)oxy]phenoxy)propionic acid (XK469).
AID217820Antiviral activity against Vaccinia virus strain Elstree was evaluated in african green monkey kidney1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID99799Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 18.2 mg/kg/day x 5 was determined as Survival time in days; Range is 10-19 days1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID409954Inhibition of mouse brain MAOA2008Journal of medicinal chemistry, Nov-13, Volume: 51, Issue:21
Quantitative structure-activity relationship and complex network approach to monoamine oxidase A and B inhibitors.
AID468158Cytotoxicity against mouse L1210 cells after 2 days2010European journal of medicinal chemistry, Feb, Volume: 45, Issue:2
Synthesis and in vitro cytostatic activity of new beta-D-arabino furan[1',2':4,5]oxazolo- and arabino-pyrimidinone derivatives.
AID135750Toxicity in mice after 7 days at a dose of 200 mg/kg/day; 7/91988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID137046Antitumor activity against L1210 leukemia in mice (median survival) at the dose of 500 mg/kg; Value represented as Test/Control = 13/81982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1473740Inhibition of human MRP3 overexpressed in Sf9 insect cell membrane vesicles assessed as uptake of [3H]-estradiol-17beta-D-glucuronide in presence of ATP and GSH measured after 10 mins by membrane vesicle transport assay2013Toxicological sciences : an official journal of the Society of Toxicology, Nov, Volume: 136, Issue:1
A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development.
AID127908Antitumor activity against L1210 leukemia in mice (Average weight per mouse on day 1.) at the dose of 1000 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID116442Percent increase in life span at 200 mg/kg (822 umol) per day1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID72424Tested in vitro against human ovarian cancer cells (G-401) for anticancer activity at dose 100 uM (Cell number 1*10 e-4)1994Journal of medicinal chemistry, Oct-14, Volume: 37, Issue:21
Synthesis of N-acetylglucosamine-modified ara-C and its effect on ovarian cancer cells.
AID98768Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 56 mg/kg dose(Mean increased life span, calculated as (T/C-1)X100)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID138570Antitumor activity against L1210 leukemia in mice (Average 7-day weight change (%)) at the dose of 500 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID625281Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for cholelithiasis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID124801Antitumor activity against L1210 tumor in mice, at a dose of 500 mg/kg peroral administration.1991Journal of medicinal chemistry, Nov, Volume: 34, Issue:11
Antitumor properties of 2(1H)-pyrimidinone riboside (zebularine) and its fluorinated analogues.
AID130304Percentage increase in life span after 9-12 days1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID96720Apparent inhibition of incorporation of labeled thymidine into DNA in murine leukemia L1210 cell;No inhibition1987Journal of medicinal chemistry, Feb, Volume: 30, Issue:2
Synthesis and biological properties of 9-(trans-4-hydroxy-2-buten-1-yl)adenine and guanine: open-chain analogues of neplanocin A.
AID355580Antiproliferative activity against human HT-29 cells at 1 ug after 48 hrs by two-layer agar-diffusion method
AID210896Inhibitory concentration against mitochondrial thymidine kinase (TK-2)2001Bioorganic & medicinal chemistry letters, May-21, Volume: 11, Issue:10
Design, synthesis and enzymatic activity of highly selective human mitochondrial thymidine kinase inhibitors.
AID1079932Highest frequency of moderate liver toxicity observed during clinical trials, expressed as a percentage. [column '% BIOL' in source]
AID122799Dose producing greatest increase in life span >/=25% in mice bearing lymphoid leukemia, treatment schedule is 1-9 qd; 5-20 [mg(umol)/kg] /day1985Journal of medicinal chemistry, Feb, Volume: 28, Issue:2
Nucleoside conjugates. 6. Synthesis and comparison of antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of corticosteroids and selected lipophilic alcohols.
AID463364Antiproliferative activity against human RS4:11 cells having FLT3-ITD mutation after 72 hrs by WST8 assay2010Bioorganic & medicinal chemistry letters, Mar-15, Volume: 20, Issue:6
Indirubin derivatives as potent FLT3 inhibitors with anti-proliferative activity of acute myeloid leukemic cells.
AID130639Antitumor activity against L1210 leukemia in mice (Percent increase in life span.) at the dose of 500 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1145830Antitumor activity against mouse L1210 cells allografted in BDF1 mouse assessed as increase in life span at 200 mg/kg, ip administered as single dose relative to vehicle-treated control1976Journal of medicinal chemistry, May, Volume: 19, Issue:5
Reactions of 2-acyloxyisobutyryl halides with nucleosides. Synthesis and biological evaluation of some 3"-acyl and 3',5'-diacyl derivatives of 1-beta-D-arabinofuranosylcytosine.
AID1079941Liver damage due to vascular disease: peliosis hepatitis, hepatic veno-occlusive disease, Budd-Chiari syndrome. Value is number of references indexed. [column 'VASC' in source]
AID451295Cytotoxicity against nucleoside transporter deficient and deoxynucleoside analogue-resistant human CEM/ARAC8C cells after 3 days by MTT assay2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID1632130Selectivity index, ratio of IC50 for HDF to IC50 for human MCF7 cells2016Journal of medicinal chemistry, 09-08, Volume: 59, Issue:17
Synthesis, Antibacterial, and Anticancer Evaluation of Novel Spiramycin-Like Conjugates Containing C(5) Triazole Arm.
AID133357Range of survival days for antitumor activity against Ip implanted L1210 / 0 lymphoid leukemia in mice at a dose of 200 mg/kg per day after treatment 1-5; range is 15-171990Journal of medicinal chemistry, May, Volume: 33, Issue:5
Nucleoside conjugates. 11. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine and cytidine conjugates of thioether lipids.
AID1222334Ratio of Km for human recombinant N-terminal His-SUMO fused wild type CDA expressed in Escherichia coli BL21(DE3) cells to Km for human recombinant N-terminal His-SUMO fused CDA Ala70Thr mutant expressed in Escherichia coli BL21(DE3) cells2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID608173Antitumor activity against mouse S180 cells xenografted in ICR mouse assessed as tumor growth inhibition at 0.089 umol/kg, ip qd administered 1 day after tumor inoculation for 7 days2011European journal of medicinal chemistry, Aug, Volume: 46, Issue:8
A class of Trp-Trp-AA-OBzl: Synthesis, in vitro anti-proliferation/in vivo anti-tumor evaluation, intercalation-mechanism investigation and 3D QSAR analysis.
AID1360836Cytotoxicity against human PC3 cells assessed as decrease in cell viability after 72 hrs by SRB assay
AID1079949Proposed mechanism(s) of liver damage. [column 'MEC' in source]
AID210864Rate of phosphorylation relative to human CytodThdK was determined1987Journal of medicinal chemistry, Nov, Volume: 30, Issue:11
Acetylenic nucleosides. 4. 1-beta-D-arabinofuranosyl-5-ethynylcytosine. Improved synthesis and evaluation of biochemical and antiviral properties.
AID120062Median survival time expressed as (T/C) was measured at dose of 10 mg/kg/day *51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID133413In vivo percent inhibition of Lewis Lung Carcinoma cell line in mice drug at 200 mg/Kg per day (p<0.001) compared with NCDAC1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID219715Tested in vitro against colo-320 human colon adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID475908Antitumor activity against human HCT116 cells after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Apr-01, Volume: 18, Issue:7
Synthesis, X-ray crystal structure study and antitumoral evaluations of 5,6-disubstituted pyrimidine derivatives.
AID142335Anticancer activity carried out in vitro against murine leukemia L12102001Journal of medicinal chemistry, May-24, Volume: 44, Issue:11
Synthesis and biological evaluation of purine-containing butenolides.
AID1135809Cytotoxicity against mouse S180 cells at 5 uM after 18 to 42 hrs relative to control1978Journal of medicinal chemistry, Nov, Volume: 21, Issue:11
Synthesis and biological activities of some uronic acids, uronates, uronamides, and urononitriles of pyrimidine nucleosides.
AID100224Tested for in vitro cytotoxicity against LLC Lewis lung carcinoma cell line1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 123. Synthesis of 1-(2-deoxy-2-isocyano-beta-D-arabinofuranosyl)cytosine and related nucleosides as potential antitumor agents.
AID116598Percent increase in life span calculated as (T/C-1)X100 at a dose of 40 mg/kg/day X51982Journal of medicinal chemistry, Nov, Volume: 25, Issue:11
Phospholipid-nucleoside conjugates. 3. Syntheses and preliminary biological evaluation of 1-beta-D-arabinofuranosylcytosine 5'-monophosphate-L-1,2-dipalmitin and selected 1-beta-D-arabinofuranosylcytosine 5-diphosphate-L-1,2-diacylglycerols.
AID138571Antitumor activity against L1210 leukemia in mice (Average 7-day weight change (%)) at the dose of 50 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID138714Antitumor activity against L1210 leukemia in mice (Average 7-day weight change) at the dose of 50 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID135304No of survivors after 45 days at a dose of 60 mg/kg/day1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID43234Concentration required for 50% inhibition of cell growth of CCRF CEM leukemic cell lines in human.1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID96789In vitro antitumor activity against L1210 cell line in mice.1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID551670Cytotoxicity against human KB cells assessed as inhibition of protein biosynthesis2011Bioorganic & medicinal chemistry letters, Jan-15, Volume: 21, Issue:2
Synthesis of 3'-azido-3'-deoxythymidine (AZT)--Cinchona alkaloid conjugates via click chemistry: Toward novel fluorescent markers and cytostatic agents.
AID355582Antiproliferative activity against human HT-29 cells at 0.01 ug after 48 hrs by two-layer agar-diffusion method
AID138562Antitumor activity against L1210 leukemia in mice (Average 7-day weight change (%)) at the dose of 250 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1123150Antiviral activity against HSV1 2931 infected in African green monkey Vero cells assessed as inhibition of virus replication at 100 ug/ml after 24 hrs relative to control1979Journal of medicinal chemistry, Jan, Volume: 22, Issue:1
Nucleosides. 110. Synthesis and antiherpes virus activity of some 2'-fluoro-2'-deoxyarabinofuranosylpyrimidine nucleosides.
AID133643Dose producing greatest increase in life span expressed as optimum dose (L1210/ara-C I) (qd 1,5,9)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID1360498Growth inhibition of human SMMC7721 cells at 2.5 ug/ml after 72 hrs by MTT assay relative to control2018European journal of medicinal chemistry, Jul-15, Volume: 155Synthesis and biological activities of dithiocarbamates containing 2(5H)-furanone-piperazine.
AID99791Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 9.2 mg/kg/day x 5 was determined as percentage increase in the life span as compared with control tumor bearers1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID154292The compound was tested for anticancer activity against P815 (sensitive) cell lines of mouse leukemia1980Journal of medicinal chemistry, Oct, Volume: 23, Issue:10
Nucleosides. 116. 1-(beta-D-Xylofuranosyl)-5-fluorocytosines with a leaving group on the 3' position. Potential double-barreled masked precursors of anticancer nucleosides.
AID212472Inhibition of thymidylate synthase in L1210 cells1986Journal of medicinal chemistry, Jul, Volume: 29, Issue:7
Synthesis and biological activity of 5-phenylselenenyl-substituted pyrimidine nucleosides.
AID451310Resistance ratio of IC50 for deoxynucleoside analogue-resistant human CEM/ARAC8C cells to IC50 for deoxynucleoside analogue-sensitive human CCRF-CEM cells2009Bioorganic & medicinal chemistry, Sep-01, Volume: 17, Issue:17
Special feature of mixed phosphotriester derivatives of cytarabine.
AID99935Antitumor activity in the mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 9.2 mg/kg/day x 5 was determined as Survival time in days; Range is 14-42 days1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID687163Ratio of IC50 for doxorubicin-resistant human HL60 cells to IC50 for human HL60 cells2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID1855690Selectivity index, ratio IC50 for antiproliferative activity against human WI-38 cells over IC50 for antiproliferative activity against human A549 cells
AID156634Concentration required for 50% inhibition of human lung squamous cell carcinoma PC14 solid cell lines. 1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID286458Cytotoxicity against A549 cells after 72 hrs by SRB assay2007Journal of medicinal chemistry, May-03, Volume: 50, Issue:9
In vitro optimization of non-small cell lung cancer activity with troxacitabine, L-1,3-dioxolane-cytidine, prodrugs.
AID156355In vitro inhibitory activity against Human lung small-cell carcinoma (PC-6) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID502460Cytotoxicity against human HepG2 cells after 24 hrs by MTT assay2010Bioorganic & medicinal chemistry, Sep-01, Volume: 18, Issue:17
A class of novel carboline intercalators: Their synthesis, in vitro anti-proliferation, in vivo anti-tumor action, and 3D QSAR analysis.
AID208868Concentration required for 50% inhibition of human lung squamous cell carcinoma TE2 solid cell lines. 1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID124295Number of survivors in mice bearing ip-implanted L1210 lymphoid leukemia after 45 day1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID1542689Induction of apoptosis in human U937 cells assessed as late apoptotic cells at 0.3 uM measured after 1 day by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 1.75 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID95139Concentration required for 50% inhibition of cell growth of K562 leukemic cell lines in human.1991Journal of medicinal chemistry, Jan, Volume: 34, Issue:1
Nucleosides and nucleotides. 94. Radical deoxygenation of tert-alcohols in 1-(2-C-alkylpentofuranosyl) pyrimidines: synthesis of (2'S)-2'-deoxy-2'-C-methylcytidine, an antileukemic nucleoside.
AID140006Death due to toxicity occurring at a maximum body weight decrease of 10 to 22% from the average post injection body weight at 50 mg/kg; 5 / 51991Journal of medicinal chemistry, Aug, Volume: 34, Issue:8
Synthesis and anticancer and antiviral activities of various 2'- and 3'-methylidene-substituted nucleoside analogues and crystal structure of 2'-deoxy-2'-methylidenecytidine hydrochloride.
AID687159Cytotoxicity against human CCRF-CEM cells assessed as cell viability after 48 hrs by celltiter-blue assay2012Journal of medicinal chemistry, Jun-14, Volume: 55, Issue:11
Structure-activity relationship (SAR) study of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and the potential of the lead against multidrug resistance in cancer treatment.
AID496831Antimicrobial activity against Cryptosporidium parvum2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID96554Antitumor activity was determined against the ip implanted L1210 leukemia in mice by change in tumor weight at 400 mg/kg (treatment of 1qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1542685Cytotoxicity against human U937 cells assessed as reduction in cell viability at 10 uM measured after 3 days by CCK8 assay2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID409745Antitumor activity against human KB cells by MTT assay2008Bioorganic & medicinal chemistry, Sep-15, Volume: 16, Issue:18
Synthesis, biological active molecular design, and molecular docking study of novel deazaflavin-cholestane hybrid compounds.
AID613825Ratio of IC50 for daunorubicin-resistant human HL60 cells to IC50 for human HL60 cells2011Journal of medicinal chemistry, Aug-25, Volume: 54, Issue:16
Structure-activity relationship and molecular mechanisms of ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017) and its analogues.
AID1542686Cytotoxicity against human U937 cells assessed as reduction in cell viability measured after 3 days by CCK8 assay2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID1079931Moderate liver toxicity, defined via clinical-chemistry results: ALT or AST serum activity 6 times the normal upper limit (N) or alkaline phosphatase serum activity of 1.7 N. Value is number of references indexed. [column 'BIOL' in source]
AID130625Antitumor activity against L1210 leukemia in mice (Percent increase in life span.) at the dose of 200 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID56589Inhibitory concentration against Drosophila melanogaster deoxyribonucleoside kinase (Dm-dNK)2001Bioorganic & medicinal chemistry letters, May-21, Volume: 11, Issue:10
Design, synthesis and enzymatic activity of highly selective human mitochondrial thymidine kinase inhibitors.
AID167739Concentration required to reduce RNA synthesis in primary rabbit kidney cells using [3H-5]-Urd incorporation1986Journal of medicinal chemistry, Feb, Volume: 29, Issue:2
Systematic synthesis and biological evaluation of alpha- and beta-D-xylofuranosyl nucleosides of the five naturally occurring bases in nucleic acids and related analogues.
AID496818Antimicrobial activity against Trypanosoma brucei brucei2010Bioorganic & medicinal chemistry, Mar-15, Volume: 18, Issue:6
Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species.
AID124916Change in weight in mice bearing ip-implanted L1210 lymphoid leukemia on day 8 at 71 mg/kg or 292 umol/kg per day1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID1134739Toxicity in CDF1 mouse assessed as change in body weight at 6.25 mg/kg, ip measured on day 5 relative to control1979Journal of medicinal chemistry, Oct, Volume: 22, Issue:10
Synthesis and antitumor activity of 5-azacytosine arabinoside.
AID145074In vitro cytotoxicity was evaluated against lung NCI-H23 cells; 1 x 10E12000Journal of medicinal chemistry, Apr-20, Volume: 43, Issue:8
Synthesis and evaluation of several new (2-chloroethyl)nitrosocarbamates as potential anticancer agents.
AID625286Drug Induced Liver Injury Prediction System (DILIps) training set; hepatic side effect (HepSE) score for hepatitis2011PLoS computational biology, Dec, Volume: 7, Issue:12
Translating clinical findings into knowledge in drug safety evaluation--drug induced liver injury prediction system (DILIps).
AID463363Antiproliferative activity against wild type FLT3 expressing human MV411 cells after 72 hrs by WST8 assay2010Bioorganic & medicinal chemistry letters, Mar-15, Volume: 20, Issue:6
Indirubin derivatives as potent FLT3 inhibitors with anti-proliferative activity of acute myeloid leukemic cells.
AID1740804Selectivity index, ratio of IC50 for cytotoxicity against human HDF cells to IC50 for cytotoxicity against human PC-3 cells2020European journal of medicinal chemistry, Sep-15, Volume: 202Synthesis, structure and anticancer activity of new geldanamycin amine analogs containing C(17)- or C(20)- flexible and rigid arms as well as closed or open ansa-bridges.
AID167738Concentration required to reduce DNA synthesis in primary rabbit kidney cells using [3H-Methyl]-dThd incorporation1986Journal of medicinal chemistry, Feb, Volume: 29, Issue:2
Systematic synthesis and biological evaluation of alpha- and beta-D-xylofuranosyl nucleosides of the five naturally occurring bases in nucleic acids and related analogues.
AID124912Change in weight in mice bearing ip-implanted L1210 lymphoid leukemia on day 8 at 100 mg/kg or 411 umol/kg per day1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID98817Antileukemic activity against L1210 leukemia cells in female B6D2F1/J mice at 56 mg/kg dose (post inoculation life span, days)1983Journal of medicinal chemistry, Feb, Volume: 26, Issue:2
2'-O-nitro-1-beta-D-arabinofuranosylcytosine. A new derivative of 1-beta-D-arabinofuranosylcytosine that resists enzymatic deamination and has antileukemic activity.
AID1135473Antiviral activity against Herpes simplex virus 2 Curtis assessed as inhibition of virus-induced cytopathic effect at 37 degC after 72 hrs by two-fold dilution method1977Journal of medicinal chemistry, Jun, Volume: 20, Issue:6
Antiviral activity of some beta-diketones. 1. Aryl alkyl diketones. In vitro activity against both RNA and DNA viruses.
AID355578Antiproliferative activity against human HT-29 cells after 96 hrs by MTT assay
AID98505Tested in vitro inhibitory activity against murine leukemia L12101993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID487904Cytotoxicity against human SF268 cells after 3 days by MTT assay2010European journal of medicinal chemistry, Jun, Volume: 45, Issue:6
Novel benzimidazole-pyrimidine conjugates as potent antitumor agents.
AID1229557Cytotoxicity against human MDA-MB-231 cells assessed as inhibition of cell proliferation after 72 hrs by sulforhodamine B assay2015Journal of medicinal chemistry, Jun-25, Volume: 58, Issue:12
Structure-Activity Relationships of Neplanocin A Analogues as S-Adenosylhomocysteine Hydrolase Inhibitors and Their Antiviral and Antitumor Activities.
AID1589125Selectivity index, ratio of IC50 for cytotoxicity in human NHDF cells to IC50 for cytotoxicity in human A549 cells2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Synthesis and in vitro anticancer activity of new gemcitabine-nucleoside analogue dimers containing methyltriazole or ester-methyltriazole linker.
AID44376In vitro inhibitory effect on growth of human T-cell acute lymphoblastoid leukemic CCRFCEM cell lines.1988Journal of medicinal chemistry, Jun, Volume: 31, Issue:6
Design, synthesis, and antineoplastic activity of 2'-deoxy-2'-methylidenecytidine.
AID124589Median T/C calculated based on survivors at 200 mg/kg (822 umol) per day against Ip-implanted L1210 lymphoid leukemia mice1993Journal of medicinal chemistry, Jun-11, Volume: 36, Issue:12
Nucleoside conjugates. 13. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of thioether lipids with improved water solubility.
AID155572In vitro antiviral activity against Para influenza virus type 3 strain C243 was evaluated in african green monkey kidney1985Journal of medicinal chemistry, Apr, Volume: 28, Issue:4
Synthesis and antitumor and antiviral activities of a series of 1-beta-D-ribofuranosyl-5-halocytosine (5-halocytidine) cyclic 3',5'-monophosphates.
AID138574Antitumor activity against L1210 leukemia in mice (Average 7-day weight change) at the dose of 1000 mg/kg1982Journal of medicinal chemistry, Feb, Volume: 25, Issue:2
Lipophilic 5'-(alkyl phosphate) esters of 1-beta-D-arabinofuranosylcytosine and its N4-acyl and 2,2'-anhydro-3'-O-acyl derivatives as potential prodrugs.
AID1222344Drug metabolism assessed as intrinsic clearance for cytosine-beta-D-arabinofuranoside-5'-MP formation incubated for 3 mins in presence of human recombinant N-terminal His-tagged DCK Pro122Ser mutant expressed in Escherichia coli BL21(DE3) cells by liquid 2013Drug metabolism and disposition: the biological fate of chemicals, Mar, Volume: 41, Issue:3
Pharmacogenomics of gemcitabine metabolism: functional analysis of genetic variants in cytidine deaminase and deoxycytidine kinase.
AID156490Tested in vitro against PC-9 human lung adenocarcinoma1993Journal of medicinal chemistry, Dec-24, Volume: 36, Issue:26
Nucleosides and nucleotides. 122. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
AID392722Antiviral activity against HSV1 assessed as inhibition of virus-induced cytopathicity2009Bioorganic & medicinal chemistry letters, Feb-15, Volume: 19, Issue:4
Synthesis of 5-isoxazol-5-yl-2'-deoxyuridines exhibiting antiviral activity against HSV and several RNA viruses.
AID355585Antiproliferative activity against mouse P388 cells at 0.1 ug after 48 hrs by two-layer agar-diffusion method
AID355583Antiproliferative activity against mouse P388 cells after 48 hrs by MTT assay
AID139212Weight change on day 8 in L1210/ara-C (I) Lymphoid Leukemia in mice( ip implanted) at a dose of 60 mg/kg/day (qd=1-5)1988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID100364Antitumor activity in mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 18.2 mg/kg/day x 5 was determined as weight change (g/mouse) on 8th day1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID210895Inhibitory concentration against cytosolic thymidine kinase (TK-1)2001Bioorganic & medicinal chemistry letters, May-21, Volume: 11, Issue:10
Design, synthesis and enzymatic activity of highly selective human mitochondrial thymidine kinase inhibitors.
AID1542696Induction of apoptosis in human U937 cells assessed as early apoptotic cells at 0.3 uM measured after 3 days by muse Annexin V/dead cell reagent staining-based flow cytometry (Rvb = 0.7 %)2019Bioorganic & medicinal chemistry letters, 06-01, Volume: 29, Issue:11
Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives.
AID45763Compound was tested for the inhibition of CML/BC(chronic myeloid leukemia, blast crisis)1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID153307Antitumor activity was determined against the ip implanted P388 leukemia in mice by percent median survival rate (treated/control) at 200 mg/kg (treatment of 1-5qd schedule) dosage; 23.0/11.01986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID95852In vitro inhibitory activity against Human oral epidermoid carcinoma (KB) cell line.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Nucleosides and nucleotides. 97. Synthesis of new broad spectrum antineoplastic nucleosides, 2'-deoxy-2'-methylidenecytidine (DMDC) and its derivatives.
AID153004Antitumor activity was determined against the ip implanted P388 leukemia in mice by 45-day survivors in mice at 500 mg/kg (treatment of 1qd schedule) dosage1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID416474Antiproliferative activity against human K562 cells2009European journal of medicinal chemistry, Feb, Volume: 44, Issue:2
Synthesis of glycose carbamides and evaluation of the induction of erythroid differentiation of human erythroleukemic K562 cells.
AID1159293Cytotoxicity against human A549 cells by SRB assay2014European journal of medicinal chemistry, Aug-18, Volume: 83Structure-activity relationships of 2'-modified-4'-selenoarabinofuranosyl-pyrimidines as anticancer agents.
AID31435Compound was tested for the inhibition of AMML (acute myelomonocytic leukemia)1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID135730Toxicity in mice after 17-21 days at a dose of 60 mg/kg/day; 21/8.51988Journal of medicinal chemistry, Sep, Volume: 31, Issue:9
Nucleoside conjugates. 10. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine 5'-diphosphate-1,2-dipalmitins.
AID96713In vitro inhibitory effect on growth of mouse leukemic L1210 cell lines.1988Journal of medicinal chemistry, Jun, Volume: 31, Issue:6
Design, synthesis, and antineoplastic activity of 2'-deoxy-2'-methylidenecytidine.
AID1079945Animal toxicity known. [column 'TOXIC' in source]
AID1135064Antitumor activity against mouse L1210 cells allografted in C3D2F1/J mouse assessed as median survival time at 18.2 mg/kg, ip qd administered for 5 days (Rvb = 10 days)1979Journal of medicinal chemistry, Nov, Volume: 22, Issue:11
Nucleoside conjugates as potential antitumor agents. 2. Synthesis and biological activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of prednisolone and prednisone.
AID152823Antitumor activity was determined against the ip implanted P388 leukemia in mice by range in survival days at 200 mg/kg (treatment of 1-5qd schedule) dosage; Range is 22-241986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID100354Antitumor activity in mice with intraperitoneal implanted L1210 lymphoid leukemia at the dose 5 mg/kg/day x 5 was determined as weight change (g/mouse) on 8th day1980Journal of medicinal chemistry, Dec, Volume: 23, Issue:12
Nucleoside conjugates as potential antitumor agents. 3. Synthesis and antitumor activity of 1-(beta-D-arabinofuranosyl)cytosine conjugates of corticosteroids.
AID210531Inhibitory concentration against Herpes simplex virus type 1 thymidine kinase(HSV-1 TK)2001Bioorganic & medicinal chemistry letters, May-21, Volume: 11, Issue:10
Design, synthesis and enzymatic activity of highly selective human mitochondrial thymidine kinase inhibitors.
AID43519Inhibit the replication of CCRF-CEM cell line in vitro.1991Journal of medicinal chemistry, Feb, Volume: 34, Issue:2
Synthesis and anticancer activity of various 3'-deoxy pyrimidine nucleoside analogues and crystal structure of 1-(3-deoxy-beta-D-threo-pentofuranosyl)cytosine.
AID1409746Clearance in human2018ACS medicinal chemistry letters, Mar-08, Volume: 9, Issue:3
Data Sets Representative of the Structures and Experimental Properties of FDA-Approved Drugs.
AID756998Inhibition of C-terminal His6-tagged Francisella tularensis SCHU S4 FabI expressed in Escherichia coli BL21 (DE3) using CrCoA as substrate assessed as oxidation of NADH to NAD+ by fluorescence assay2013Journal of medicinal chemistry, Jul-11, Volume: 56, Issue:13
3-substituted indole inhibitors against Francisella tularensis FabI identified by structure-based virtual screening.
AID233950Ratio of GI50 without hPAP/ GI50 with hPAP2001Bioorganic & medicinal chemistry letters, Nov-05, Volume: 11, Issue:21
Amino acid phosphoramidate nucleosides: potential ADEPT/GDEPT substrates.
AID98953Antitumor activity against the ic implanted L1210 lymphoid leukemia in mice expressed as optimal dose (1-5qd treatment schedule) required to produce greatest increase in life span was determined1986Journal of medicinal chemistry, Oct, Volume: 29, Issue:10
Nucleoside conjugates. 7. Synthesis and antitumor activity of 1-beta-D-arabinofuranosylcytosine conjugates of ether lipids.
AID1745850Viability Counterscreen for Confirmatory qHTS for Inhibitors of ATXN expression
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588497High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain F protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588501High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Lethal Factor Protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2010Current protocols in cytometry, Oct, Volume: Chapter 13Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2006Cytometry. Part A : the journal of the International Society for Analytical Cytology, May, Volume: 69, Issue:5
Microsphere-based protease assays and screening application for lethal factor and factor Xa.
AID588499High-throughput multiplex microsphere screening for inhibitors of toxin protease, specifically Botulinum neurotoxin light chain A protease, MLPCN compound set2010Assay and drug development technologies, Feb, Volume: 8, Issue:1
High-throughput multiplex flow cytometry screening for botulinum neurotoxin type a light chain protease inhibitors.
AID1745846Firefly Luciferase Counterscreen for Inhibitors of ATXN expression
AID1745845Primary qHTS for Inhibitors of ATXN expression
AID1745848Confirmatory qHTS for Inhibitors of ATXN expression
AID1745847CMV-Luciferase Counterscreen for Inhibitors of ATXN expression
AID651635Viability Counterscreen for Primary qHTS for Inhibitors of ATXN expression
AID1745849Viability Counterscreen for CMV-Luciferase Assay of Inhibitors of ATXN expression
AID504749qHTS profiling for inhibitors of Plasmodium falciparum proliferation2011Science (New York, N.Y.), Aug-05, Volume: 333, Issue:6043
Chemical genomic profiling for antimalarial therapies, response signatures, and molecular targets.
AID504812Inverse Agonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID504810Antagonists of the Thyroid Stimulating Hormone Receptor: HTS campaign2010Endocrinology, Jul, Volume: 151, Issue:7
A small molecule inverse agonist for the human thyroid-stimulating hormone receptor.
AID1347050Natriuretic polypeptide receptor (hNpr2) antagonism - Pilot subtype selectivity assay2019Science translational medicine, 07-10, Volume: 11, Issue:500
Inhibition of natriuretic peptide receptor 1 reduces itch in mice.
AID1508630Primary qHTS for small molecule stabilizers of the endoplasmic reticulum resident proteome: Secreted ER Calcium Modulated Protein (SERCaMP) assay2021Cell reports, 04-27, Volume: 35, Issue:4
A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome.
AID1347049Natriuretic polypeptide receptor (hNpr1) antagonism - Pilot screen2019Science translational medicine, 07-10, Volume: 11, Issue:500
Inhibition of natriuretic peptide receptor 1 reduces itch in mice.
AID1347045Natriuretic polypeptide receptor (hNpr1) antagonism - Pilot counterscreen GloSensor control cell line2019Science translational medicine, 07-10, Volume: 11, Issue:500
Inhibition of natriuretic peptide receptor 1 reduces itch in mice.
AID1347082qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: LASV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID1347086qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lymphocytic Choriomeningitis Arenaviruses (LCMV): LCMV Primary Screen - GLuc reporter signal2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID588349qHTS for Inhibitors of ATXN expression: Validation of Cytotoxic Assay
AID1347083qHTS for Inhibitors of the Functional Ribonucleoprotein Complex (vRNP) of Lassa (LASV) Arenavirus: Viability assay - alamar blue signal for LASV Primary Screen2020Antiviral research, 01, Volume: 173A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity.
AID504836Inducers of the Endoplasmic Reticulum Stress Response (ERSR) in human glioma: Validation2002The Journal of biological chemistry, Apr-19, Volume: 277, Issue:16
Sustained ER Ca2+ depletion suppresses protein synthesis and induces activation-enhanced cell death in mast cells.
AID588378qHTS for Inhibitors of ATXN expression: Validation
AID1347126qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347093qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID1347104qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347119qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347127qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347098qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
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.
AID1347118qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
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.
AID1347117qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347124qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for RD cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347112qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347107qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh30 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347092qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for A673 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347103qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347101qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-12 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347121qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347099qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347106qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for control Hh wild type fibroblast cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347122qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347097qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Saos-2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347100qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347102qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347095qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347115qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for NB-EBc1 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347114qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347123qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347110qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for A673 cells)2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347090qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for DAOY cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347111qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SK-N-MC cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347154Primary screen GU AMC qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1347089qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for TC32 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347094qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for BT-37 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347125qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for Rh18 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347108qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for Rh41 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347091qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347105qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for MG 63 (6-TG R) cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347109qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for NB1643 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347096qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Primary screen for U-2 OS cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347129qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SK-N-SH cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347128qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for OHS-50 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347113qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for LAN-5 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1347116qHTS of pediatric cancer cell lines to identify multiple opportunities for drug repurposing: Confirmatory screen for SJ-GBM2 cells2018Oncotarget, Jan-12, Volume: 9, Issue:4
Quantitative high-throughput phenotypic screening of pediatric cancer cell lines identifies multiple opportunities for drug repurposing.
AID1159550Human Phosphogluconate dehydrogenase (6PGD) Inhibitor Screening2015Nature cell biology, Nov, Volume: 17, Issue:11
6-Phosphogluconate dehydrogenase links oxidative PPP, lipogenesis and tumour growth by inhibiting LKB1-AMPK signalling.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (15,030)

TimeframeStudies, This Drug (%)All Drugs %
pre-19905726 (38.10)18.7374
1990's3071 (20.43)18.2507
2000's2676 (17.80)29.6817
2010's2683 (17.85)24.3611
2020's874 (5.82)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials0 (0.00%)5.53%
Trials2,424 (15.65%)5.53%
Reviews7 (3.13%)6.00%
Reviews1,051 (6.79%)6.00%
Case Studies2 (0.89%)4.05%
Case Studies2,172 (14.02%)4.05%
Observational0 (0.00%)0.25%
Observational26 (0.17%)0.25%
Other215 (95.98%)84.16%
Other9,814 (63.37%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (1164)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Randomised Controlled Phase-2 Trial to Determine the Efficacy of Adoptive Immunotherapy With Haploidentical Natural Killer Cells in High-risk Acute Myeloid Leukemia[NCT02229266]Phase 21 participants (Actual)Interventional2015-09-30Terminated(stopped due to low recruitment rate)
A Randomized Trial for Adults With Newly Diagnosed Acute Lymphoblastic Leukemia[NCT01085617]Phase 31,033 participants (Actual)Interventional2010-12-31Active, not recruiting
S0919, A Phase II Study of Idarubicin and Ara-C in Combination With Pravastatin for Poor-Risk Acute Myelogenous Leukemia[NCT00840177]Phase 2115 participants (Actual)Interventional2009-12-10Completed
Pilot Trial of Hyper-CVAD and Methotrexate/ARA C Plus Rituximab in Patients With Previously Untreated Mantle Cell Lymphoma[NCT00041132]Phase 256 participants (Actual)Interventional2002-09-30Completed
A Phase I/II Pilot Study of Ifosfamide, Carboplatin and Etoposide Therapy (ICE) and SGN-30 (NSC# 731636, IND#) in Children With CD30+ Recurrent Anaplastic Large Cell Lymphoma[NCT00354107]Phase 1/Phase 25 participants (Actual)Interventional2007-01-31Terminated
T-Cell or Natural Killer (NK) Cell Adback in Patients With Lymphoid Malignancies Receiving Allogeneic Stem Cell Transplantation With Campath-IH Containing Conditioning Regimens[NCT00536978]Phase 222 participants (Actual)Interventional2007-09-30Completed
ALinC 17, Classification ©), B-precursor Induction Treatment (I)[NCT01225874]3,762 participants (Actual)Interventional1999-12-31Completed
Phase I Study of Lenalidomide and Conventional Chemotherapy in Acute Myeloid Leukemia[NCT01132586]Phase 161 participants (Actual)Interventional2010-05-31Completed
Study of Low-Dose Cytarabine and Etoposide With or Without All-Trans Retinoic Acid in Older Patients Not Eligible for Intensive Chemotherapy With Acute Myeloid Leukemia and NPM1 Mutation[NCT01237808]Phase 3144 participants (Actual)Interventional2011-03-31Completed
Busulfan, Etoposide, Cytarabine and Melphalan (BuEAM) as a Conditioning for Autologous Stem Cell Transplantation in Patients With Diffuse Large B Cell Lymphoma (DLCBL) Previously Treated With Rituximab Based Regimen[NCT01063439]Phase 242 participants (Anticipated)Interventional2010-01-31Recruiting
Randomized Phase-II Trial Evaluating Induction Therapy With Idarubicin and Etoposide Plus Sequential or Concurrent Azacitidine and Maintenance Therapy With Azacitidine[NCT01180322]Phase 2277 participants (Actual)Interventional2010-11-30Completed
A Phase III Study for the Treatment of Children and Adolescents With Newly Diagnosed Low Risk Hodgkin Disease[NCT00302003]Phase 3287 participants (Actual)Interventional2006-02-28Completed
A Phase I Dose Finding Study Of Panobinostat In Children With Refractory Hematologic Malignancies[NCT01321346]Phase 130 participants (Actual)Interventional2011-03-31Completed
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 III Trial Comparing ARA-C/High-Dose Mitoxantrone (ALL-2') to A Standard Vincristine/Prednisone Based Regimen ('L-20') as Induction Therapy For Adult Patients With Acute Lymphoblastic Leukemia (ALL): The ALL-4 Protocol"[NCT00002766]Phase 3170 participants (Actual)Interventional1996-03-31Completed
A Phase I/Ib Study of JNJ-75276617 in Combination With Conventional Chemotherapy for Pediatric and Young Adult Participants With Relapsed/Refractory Acute Leukemias Harboring KMT2A, NPM1, or Nucleoporin Gene Alterations[NCT05521087]Phase 180 participants (Anticipated)Interventional2024-05-29Not yet recruiting
A Phase 1b Study of JNJ-75276617 in Combination With AML-Directed Therapies for Participants With Acute Myeloid Leukemia Harboring KMT2A or NPM1 Alterations[NCT05453903]Phase 1150 participants (Anticipated)Interventional2022-10-04Recruiting
A Phase 1b Study to Investigate the Safety, Tolerability, and Pharmacokinetics of Entospletinib (ENTO) as Monotherapy in Japanese Subjects With Relapsed or Refractory Hematologic Malignancies and in Combination With Chemotherapy in Japanese Subjects With [NCT03135028]Phase 19 participants (Actual)Interventional2017-05-19Terminated(stopped due to No signal of efficacy with Entospletinib)
Leukemia Stem Cell Detection in Acute Myeloid Leukemia[NCT02927938]Phase 318 participants (Actual)Interventional2016-09-30Terminated(stopped due to Study terminated 7/23/2019 due to limited participation and testing challenges.)
An Open Label, Single Arm, Single-Center Exploratory Study to Evaluate the Efficacy and Safety of Selinexor and HAAG +/- HMA in Relapsed/Refractory Acute Leukemia (AML) Patients[NCT05805072]20 participants (Anticipated)Interventional2023-05-01Not yet recruiting
A Single-arm, Open, Multicenter, Phase II Study to Investigator the Efficacy and Safety of HYML-122 and Cytarabine in Patients With FLT3 Positive Relapsed or Refractory Acute Myeloid Leukemia (AML)[NCT05241093]Phase 218 participants (Anticipated)Interventional2022-03-29Recruiting
A Clinical Study to Evaluate the Safety, Efficacy and Pharmacokinetics of Mitoxantrone Hydrochloride Liposome Injection Combined With Chemotherapy in Previously Untreated de Novo Acute Myeloid Leukemia[NCT05941585]90 participants (Anticipated)Interventional2023-07-31Not yet recruiting
CLAG-M or FLAG-Ida Chemotherapy Followed Immediately by Related/Unrelated Reduced-Intensity Conditioning (RIC) Allogeneic Hematopoietic Cell Transplantation for Adults With Myeloid Malignancies at High Risk of Relapse: A Phase 1 Study[NCT04375631]Phase 1120 participants (Anticipated)Interventional2020-12-03Recruiting
Busulfan, Etoposide, Cytarabine, and Melphalan (BuEAM) as a Conditioning for Autologous Stem Cell Transplantation in Patients With T Cell or NK Cell Lymphoma[NCT01178658]Phase 242 participants (Anticipated)Interventional2010-07-31Recruiting
Multicentric Phase II Trial, Prospective, Open, Single Group, to Discuss Induction Therapy With a Combination of Clofarabine and Low-dose Cytarabine Followed by Consolidation Therapy With Clofarabine and Low-dose Cytarabine for the Treatment of AML Patien[NCT01193400]Phase 275 participants (Anticipated)Interventional2010-09-30Terminated
A Phase I/II Study of Llme Treated Non-Myeloablative Allogeneic Hematopoietic Stem Cell Transplantation for Patients With Hematological Malignancies[NCT00429416]Phase 1/Phase 214 participants (Actual)Interventional2004-03-31Completed
Prexasertib in Combination With Mitoxantrone, Etoposide and Cytarabine (MEC) in Relapsed/Refractory Acute Myeloid Leukemia (AML) and High Risk Myelodysplastic Syndrome (MDS) - a Phase I Trial[NCT03735446]Phase 12 participants (Actual)Interventional2019-01-18Terminated(stopped due to Sponsor Decision)
A Multicenter, Randomized, Open-label, Parallel-group, Phase 3 Trial of Subcutaneous Azacitidine Plus Best Supportive Care Versus Conventional Care Regimens Plus Best Supportive Care for the Treatment of Myelodysplastic Syndromes (MDS)[NCT00071799]Phase 3358 participants (Actual)Interventional2003-11-01Completed
A Multicenter, Prospective, Non-randomized, Phase I-II Trial to Assess the Efficacy and Safety of the Combination of Oral Quizartinib and the FLAG-IDA Chemotherapy Regimen in First Relapsed/Refractory Acute Myeloid Leukemia (R/R AML) Patients[NCT04112589]Phase 1/Phase 263 participants (Actual)Interventional2019-12-26Active, not recruiting
Hematopoietic Stem Cell Transplantation Using Bone Marrow Or Peripheral Blood Stem Cells From Matched, Unrelated, Volunteer Donors[NCT00054327]Phase 234 participants (Actual)Interventional2000-11-30Completed
Phase II Study of the Combination of Low-Intensity Chemotherapy and Blinatumomab in Patients With Philadelphia Chromosome Negative Relapsed/Refractory Acute Lymphoblastic Leukemia (ALL)[NCT03518112]Phase 26 participants (Actual)Interventional2018-04-18Terminated(stopped due to Due to Competing Studies)
A Phase II Study of R-CHOP With Intensive CNS Prophylaxis and Scrotal Irradiation in Patients With Primary Testicular Diffuse Large B-cell Lymphoma[NCT00945724]Phase 254 participants (Actual)Interventional2009-04-30Active, not recruiting
A Phase 1/2 Study of CPX-351 (NSC# 775341) Alone Followed by Fludarabine, Cytarabine, and G-CSF (FLAG) for Children With Relapsed Acute Myeloid Leukemia (AML)[NCT02642965]Phase 1/Phase 238 participants (Actual)Interventional2016-05-02Completed
Phase I Study of 4'-Thio-araC in Patients With Advanced Hematologic Malignancies[NCT01139151]Phase 131 participants (Actual)Interventional2010-08-31Completed
Phase I Dose Escalation Study of the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamic Properties of Oral AT-406 in Combination With Daunorubicin and Cytarabine in Patients With Poor-risk, Acute Myelogenous Leukemia (AML)[NCT01265199]Phase 129 participants (Actual)Interventional2011-02-28Terminated(stopped due to Study was terminated before a MTD was determined for administrative reasons)
A Monocenter Prospective Single Arm Study of Cladribine Plus Homoharringtonine and Cytarabine Regimen (CHA) for Induced Therapy in Adults de Novo Acute Myeloid Leukemia (Non-M3)[NCT05906914]Phase 230 participants (Anticipated)Interventional2023-06-29Recruiting
Phase III Study Of Gemcitabine, Dexamethasone, And Cisplatin Compared To Dexamethasone, Cytarabine, And Cisplatin Plus/Minus Rituximab [(R)-GDP vs (R)-DHAP] As Salvage Chemotherapy For Patients With Relapsed Or Refractory Aggressive Histology Non-Hodgkin'[NCT00078949]Phase 3849 participants (Actual)Interventional2003-08-27Completed
A Phase I and Expansion Cohort Study of Venetoclax in Combination With Chemotherapy in Pediatric Patients With Refractory or Relapsed Acute Myeloid Leukemia[NCT03194932]Phase 162 participants (Actual)Interventional2017-07-11Completed
Adult Acute Lymphoblastic Leukemia Treated With Pediatric Regimen in Brazil - a Prospective Collaborative Study[NCT05959720]180 participants (Anticipated)Observational [Patient Registry]2023-09-05Recruiting
A Phase II Randomized Study to Assess the Efficacy on Outcome of Venetoclax Combined With Cytarabine Versus Idarubicin Combined With Cytarabine Administered as Post-remission Therapy to Elderly Patients With Acute Myeloid Leukemia in First Remission[NCT04968015]Phase 2134 participants (Anticipated)Interventional2022-05-25Recruiting
Open-Label Phase 2 Trial of Vyxeos in Patients With Intermediate and High-Risk Acute Myeloid Leukemia Who Have Failed an Initial Cycle of Standard Cytarabine and Daunorubicin Chemotherapy[NCT04049539]Phase 1/Phase 228 participants (Anticipated)Interventional2021-01-29Recruiting
Pilot Open-label Trial of Nivolumab Combined With Chemotherapy in Refractory Myelodysplastic Syndromes.[NCT03259516]Phase 1/Phase 22 participants (Actual)Interventional2017-05-25Terminated(stopped due to Slow recruitment rate)
A Phase II, Open-Label Clinical Efficacy Study Defining Genomic Signatures That Correlate With Midostaurin Response in Relapsed or Refractory Acute Myeloid Leukemia (AML)[NCT03207334]Phase 20 participants (Actual)Interventional2018-11-30Withdrawn(stopped due to Insufficient funding)
A Multicenter,Open-label,Radonmized Study on the Treatment of Older Adult Acute Myeloid Leukemia Patients Aged 55 to 65 Years Old[NCT02432872]300 participants (Anticipated)Interventional2015-04-30Recruiting
Prospective Study of Rituximab Combined With Chemotherapy for CD20+ Adult Acute Lymphoblastic Leukemia[NCT01358253]Phase 4100 participants (Actual)Interventional2010-12-31Completed
Phase 1b Study of Venetoclax in Combination With Intensive Induction and Consolidation Chemotherapy in Treatment Naïve Subjects With Acute Myelogenous Leukemia[NCT03709758]Phase 164 participants (Anticipated)Interventional2018-10-17Recruiting
A Randomized Phase 3 Trial of Fludarabine/Cytarabine/Gemtuzumab Ozogamicin With or Without Venetoclax in Children With Relapsed AML[NCT05183035]Phase 398 participants (Anticipated)Interventional2022-10-01Recruiting
A Phase II Study of Ibrutinib Plus Rituximab With Hyper-CVAD Consolidation in Newly Diagnosed Young Patients With Mantle Cell Lymphoma: A Window Period for Bioimmunotherapy Before Chemotherapy[NCT02427620]Phase 2131 participants (Anticipated)Interventional2015-06-03Active, not recruiting
A Phase I Study of AC220 for Children With Relapsed or Refractory ALL or AML[NCT01411267]Phase 124 participants (Actual)Interventional2011-09-01Completed
A Phase 2 Study Evaluating Efficacy and Safety of Chi-BEAC Combining With Auto-HSCT to Treat Aggressive Lymphoma Subjects[NCT03629873]Phase 269 participants (Anticipated)Interventional2018-02-01Active, not recruiting
High Dose Chemotherapy With Autologous Stem Cell Rescue for Aggressive B Cell Lymphoma and Hodgkin Lymphoma in HIV-infected Patients (BMT CTN #0803)[NCT01141712]Phase 243 participants (Actual)Interventional2010-04-30Completed
Phase 1 Study of FLAG-Ida With Pivekimab Sunirine (PVEK [IMGN632]) for Adults With Newly Diagnosed Adverse-Risk Acute Myeloid Leukemia and Other High-Grade Myeloid Neoplasms[NCT06034470]Phase 130 participants (Anticipated)Interventional2024-01-13Recruiting
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 Pilot Trial of Blinatumomab Consolidation Post Autologous Stem Cell Transplantation in Patients With DLBCL[NCT03072771]Phase 114 participants (Actual)Interventional2017-08-01Completed
A Phase II Trial of Oxaliplatin, Cytosine Arabinoside, Dexamethasone With Rituxan (ROAD) in Patients With Relapsed CD20+ B-Cell Non-Hodgkins Lymphoma[NCT00166439]Phase 250 participants (Actual)Interventional2005-03-31Completed
Total XV - Total Therapy Study XV for Newly Diagnosed Patients With Acute Lymphoblastic Leukemia[NCT00137111]Phase 3501 participants (Actual)Interventional2000-07-08Completed
Cladribine Combined With G-CSF and Cytarabine as a Salvage Treatment in Refractory/Relapsed Acute Lymphoblastic Leukemia[NCT05578378]Phase 2/Phase 332 participants (Anticipated)Interventional2022-01-01Recruiting
Phase II Study of Intensified CVP, Rituximab, and High Dose Cyclophosphamide for Adult Burkitt or Burkitt-Like Lymphoma[NCT00133991]Phase 223 participants (Actual)Interventional2005-07-31Completed
An Open-Label, One-Arm, Multi-Site Trial of Precision Diagnosis Directing Histone Deacetylase Inhibitor Chidamide Total Therapy for Adult T-lymphoblastic Lymphoma/Leukemia[NCT03564704]Phase 2/Phase 380 participants (Anticipated)Interventional2016-02-14Recruiting
Phase 1b, Open-label, Dose Escalation Study of Quizartinib in Combination With Induction and Consolidation Chemotherapy in Japanese Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)[NCT02834390]Phase 17 participants (Actual)Interventional2016-08-12Completed
A Phase 1 Study of MLN9708 in Combination With MEC (Mitoxantrone, Etoposide, and Intermediate-Dose Cytarabine) for Relapsed/ Refractory Acute Myelogenous Leukemia (AML)[NCT02070458]Phase 130 participants (Actual)Interventional2014-10-08Completed
A Phase II Study of Tosedostat in Combination With Either Cytarabine or Decitabine in Newly Diagnosed AML or High-Risk MDS[NCT01567059]Phase 234 participants (Actual)Interventional2012-05-31Completed
An Open-Label, One-Arm, Multi-Site Trial of Precision Diagnosis Directing Histone Deacetylase Inhibitor Chidamide Target Total Therapy for Adult Early T-cell Progenitor Acute Lymphoblastic Leukemia/Lymphoma[NCT03553238]Phase 2/Phase 370 participants (Anticipated)Interventional2016-02-14Recruiting
A Phase 2 Study of Blinatumomab (NSC# 765986) in Combination With Nivolumab (NSC # 748726), a Checkpoint Inhibitor of PD-1, in B-ALL Patients Aged >/= 1 to < 31 Years Old With First Relapse[NCT04546399]Phase 2550 participants (Anticipated)Interventional2020-12-04Suspended(stopped due to Other - FDA Partial Clinical Hold)
A Multicenter, Double-Blind, Randomized, Placebo-Controlled, Phase III Study of Idasanutlin, an MDM2 Antagonist, With Cytarabine Versus Cytarabine Plus Placebo in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML)[NCT02545283]Phase 3447 participants (Actual)Interventional2015-12-30Terminated(stopped due to The study was stopped for futility based on efficacy results at the interim analysis; no unexpected safety findings were observed.)
Treatment Protocol For All Fragile Patients Ph' Negative Over 55 Years[NCT01358201]Phase 4100 participants (Anticipated)Interventional2010-05-31Recruiting
Safety and Feasibility of Lenalidomide in Combination With HLA-mismatched Stem-cell Microtransplantation as Post-remission Therapy in Patients With Acute Myeloid Leukemia (AML)[NCT02255162]Phase 18 participants (Actual)Interventional2015-01-31Terminated(stopped due to Slow Accrual)
Modified TBF Regimen as Conditioning Regimen Prior to Allogeneic Hematopoietic Cell Transplantation for T Cell Acute Lymphoblastic Leukemia/Lymphoblastic Lymphoma:Phase II Study[NCT05598593]Phase 270 participants (Anticipated)Interventional2022-10-23Recruiting
CHOA-AML: A Pilot Study for Newly Diagnosed Pediatric Patients With Acute Myeloid Leukemia (AML)[NCT04326439]Phase 28 participants (Actual)Interventional2020-01-24Terminated(stopped due to Due to unforeseen circumstances, the study team was limited in enrolling patients on this trial; thus, it was terminated.)
A Phase I Study of Volasertib Combined With Standard Induction Chemotherapy for Previously Untreated Patients With Acute Myeloid Leukemia (VIAC)[NCT02527174]Phase 10 participants (Actual)Interventional2016-11-30Withdrawn(stopped due to Volasertib no longer available)
Phase 1 Study of Cladribine Based Induction Therapy (CLAG) With ATRA (All-Trans Retinoic Acid) and Midostaurin in Relapsed/Refractory AML[NCT01161550]Phase 111 participants (Actual)Interventional2010-11-30Completed
Multicenter Randomised Clinical Trial in Acute Myeloid Leukemia Treatment Based on Three Anthracyclines, Comparing Two Types of Consolidation With Different ARA-C Doses Followed by One Year Maintenance[NCT01587430]Phase 4245 participants (Anticipated)Interventional2010-01-31Active, not recruiting
Freiburg ZNS-NHL Study: Therapy for Patients With Primary Non-Hodgkin Lymphoma of the CNS - Sequential High Dosage Chemotherapy With Autologous Peripheral Blood Stem Cell Plantation[NCT00647049]Phase 278 participants (Anticipated)Interventional2007-01-31Recruiting
Phase I Study of the Combination of Midostaurin, Bortezomib, and Chemotherapy in Relapsed/Refractory Acute Myeloid Leukemia[NCT01174888]Phase 134 participants (Actual)Interventional2010-08-31Completed
A Phase 3 Open-Label, Multicenter, Randomized Study of ASP2215 Versus Salvage Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) With FLT3 Mutation[NCT02421939]Phase 3371 participants (Actual)Interventional2015-10-20Active, not recruiting
Clinical Study on the Safety and Efficacy of QN-030a in Acute Myeloid Leukemia[NCT05665114]Phase 118 participants (Anticipated)Interventional2022-12-24Recruiting
Randomized Phase II Trial On Primary Chemotherapy With High-Dose Methotrexate And High-Dose Cytarabine With Or Without Thiotepa, And With Or Without Rituximab, Followed By Brain Irradiation Vs. High-Dose Chemotherapy Supported By Autologous Stem Cells Tra[NCT01011920]Phase 2126 participants (Actual)Interventional2009-11-30Completed
A Phase II Study of Punctual, Cyclic, and Intensive Chemotherapy With Liposomal Cytarabine (Depocyt®) CNS Prophylaxis for Adults With Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma[NCT02043587]Phase 231 participants (Actual)Interventional2014-01-31Terminated(stopped due to Original investigator for the trial has left)
Intensive Chemotherapy And Immunotherapy In Patients With Newly Diagnosed Primary CNS Lymphoma[NCT00098774]Phase 247 participants (Actual)Interventional2004-10-31Completed
A Phase III Randomized Trial of Gemtuzumab Ozogamicin (Mylotarg) Combined With Conventional Chemotherapy for De Novo Acute Myeloid Leukemia (AML) in Children, Adolescents, and Young Adults[NCT00372593]Phase 31,070 participants (Actual)Interventional2006-08-31Completed
Treatment of Late Isolated Extramedullary Relapse From Acute Lymphoblastic Leukemia (ALL) (Initial CR1≥ 18 Months)[NCT00096135]168 participants (Actual)Interventional2004-11-30Completed
HEC73543 Versus Salvage Chemotherapy in Relapsed or Refractory FLT3-ITD Acute Myeloid Leukemia: a Multicenter, Open-label, Randomized Phase 3 Trial[NCT05586074]Phase 3324 participants (Anticipated)Interventional2023-03-03Recruiting
A Multicenter, Phase 1, Open-Label Study of the FGFR Inhibitor Pemigatinib (INCB054828) Administered After Chemotherapy in Newly Diagnosed Acute Myeloid Leukemia (AML) With Adverse or Intermediate Risk Cytogenetics[NCT04659616]Phase 132 participants (Anticipated)Interventional2021-01-14Recruiting
A Randomized Controlled Clinical Trial Comparing Chidamide,Carmustine,Etoposide,Cytarabine and Melphalan With BEAM Regimen Combined With Autologus Hematopoietic Stem Cell Transplantation for the Treatment of Newly Diagnosed PTCL[NCT05931263]Phase 3104 participants (Anticipated)Interventional2023-06-01Recruiting
Addition of Sorafenib to G-CSF, Cladribine, Cytarabine and Mitoxantrone (G-CLAM) in Adults With Newly-Diagnosed Acute Myeloid Leukemia (AML) Independent of FLT3-ITD Status: A Phase 1/2 Study[NCT02728050]Phase 1/Phase 284 participants (Actual)Interventional2016-12-01Completed
A Phase II Trial Involving Patients With Recurrent PCNSL Treated With Carboplatin/BBBD, by Adding Rituxan (Rituximab), An Anti CD-20 Antibody, To The Treatment Regimen[NCT00074165]Phase 217 participants (Actual)Interventional2003-01-31Terminated(stopped due to Lack of accrual)
A Randomized, Placebo-Controlled, Double Blind, Trial of the Administration of the MDR Modulator, Zosuquidar Trihydrochloride (LY335979), During Conventional Induction and Post-Remission Therapy in Patients Greater Than 60 Years of Age With Newly Diagnose[NCT00046930]Phase 3449 participants (Actual)Interventional2002-09-17Completed
LCH-IV, International Collaborative Treatment Protocol for Children and Adolescents With Langerhans Cell Histiocytosis[NCT02205762]Phase 2/Phase 31,400 participants (Anticipated)Interventional2016-11-02Recruiting
Early Systemic Central Nervous System Prophylaxis in Diffuse Large B-cell Lymphoma[NCT03719560]Phase 20 participants (Actual)Interventional2019-07-01Withdrawn(stopped due to lack of funding)
Clofarabine in Combination With a Standard Remission Induction Regimen (AraC and Idarubicin) in Patients 18-60 Years Old With Previously Untreated Intermediate and Bad Risk Acute Myelogenous Leukemia (AML) or High Risk Myelodysplasia (MDS) : a Phase I-II [NCT00838240]Phase 1/Phase 2114 participants (Anticipated)Interventional2008-11-30Recruiting
A Double- Blind, Placebo Controlled, Randomized, Multicenter, Phase II Study to Assess the Efficacy of BL-8040 Addition to Consolidation Therapy in AML Patients[NCT02502968]Phase 2194 participants (Anticipated)Interventional2015-09-30Recruiting
Phase Ib/II Study of Uproleselan Added to Cladribine Plus Low Dose Cytarabine (LDAC) Induction Followed by Consolidation With Uproleselan Plus Cladribine Plus LDAC in Patients With Treated Secondary AML (TS-AML)[NCT04848974]Phase 1/Phase 237 participants (Anticipated)Interventional2021-06-11Recruiting
A Phase I/II Study of CPI-613 in Combination With Induction/Consolidation in Older AML Patients[NCT02472626]Phase 1/Phase 20 participants (Actual)Interventional2015-12-31Withdrawn(stopped due to Drug Supply Issues)
A Randomized, Open-Label Study of the Efficacy and Safety of Galinpepimut-S (GPS) Maintenance Monotherapy Compared to Investigator's Choice of Best Available Therapy in Subjects With Acute Myeloid Leukemia Who Have Achieved Complete Remission After Second[NCT04229979]Phase 3140 participants (Anticipated)Interventional2021-02-08Recruiting
A Phase Ib Study of Oral Selinexor in Adult Patients With Relapsed/Refractory B-cell Lymphoma Receiving R-DHAOx or R-GDP[NCT02741388]Phase 139 participants (Actual)Interventional2016-10-31Completed
A Phase 3 Randomized Trial for Patients With De Novo AML Comparing Standard Therapy Including Gemtuzumab Ozogamicin (GO) to CPX-351 With GO, and the Addition of the FLT3 Inhibitor Gilteritinib for Patients With FLT3 Mutations[NCT04293562]Phase 31,400 participants (Anticipated)Interventional2020-07-21Recruiting
A Phase III Study for Patients With Newly Diagnosed Acute Promyelocytic Leukemia (APL) Using Arsenic Trioxide and All-Trans Retinoic Acid[NCT02339740]Phase 3158 participants (Actual)Interventional2015-07-21Active, not recruiting
Phase II Study of Combination of Hyper-CVAD and Dasatinib (NSC-732517) With or Without Allogeneic Stem Cell Transplant in Patients With Philadelphia (Ph) Chromosome Positive and/or BCR-ABL Positive Acute Lymphoblastic Leukemia (ALL) (A BMT Study)[NCT00792948]Phase 297 participants (Actual)Interventional2009-09-01Active, not recruiting
Multicentered Phase II Study Evaluating the Activity and Toxicity of Liposomal Cytarabine in the Treatment of Children and Adolescents With Acute Lymphoblastic Leukemia With Resistent or Relapsed Central Nervous System Involvement[NCT01593488]Phase 231 participants (Anticipated)Interventional2012-03-31Active, not recruiting
A Phase I/II, Open-label Single Institution Study Evaluating Rapamycin in Combination With High-dose Etoposide and Cytarabine in Relapsed or Refractory Aggressive Lymphoid Malignancies[NCT00776373]Phase 1/Phase 24 participants (Actual)Interventional2007-01-31Terminated
CBA Versus FBA Conditioning Followed by Allogeneic HSCT in Treatment of High Risk and Refractory AML[NCT03384212]Phase 3120 participants (Anticipated)Interventional2016-08-01Recruiting
A Phase 1 Investigator Sponsored Study of Selinexor in Combination With Daunorubicin and Cytarabine in Patients With Previously Untreated Poor-Risk Acute Myeloid Leukemia[NCT02403310]Phase 121 participants (Actual)Interventional2015-06-18Completed
A Phase IIB, Randomized, Double-Blinded, Placebo-Controlled Study of Low Dose Cytarabine and Lintuzumab Compared to Low Dose Cytarabine and Placebo in Patients 60 Years of Age and Older With Previously Untreated AML[NCT00528333]Phase 2211 participants (Actual)Interventional2007-09-30Completed
A Safety and Tolerability Trial of Crenolanib and Chemotherapy With Cytarabine and Anthracyclines in Patients With Newly Diagnosed Acute Myeloid Leukemia With FLT3 Activating Mutations[NCT02283177]Phase 244 participants (Actual)Interventional2015-01-31Completed
The Treatment of Down Syndrome Children With Acute Myeloid Leukemia (AML) and Myelodysplastic Syndromes (MDS) Under the Age of 4 Years[NCT00369317]Phase 3205 participants (Actual)Interventional2007-03-31Completed
The Efficacy and Safety of Azacytidine Combined With HAG Regimen Versus Azacytidine for Elderly Patients With Newly Diagnosed Myeloid Malignancy: a Prospective, Randomized Controlled Trial[NCT03873311]Phase 4114 participants (Anticipated)Interventional2019-09-01Recruiting
Phase II/III Randomized Study to Improve Overall Survival in 18 to 60 Year-old Patients, Comparing Daunorubicin Versus High Dose Idarubicin Induction Regimens, High Dose Versus Intermediate Dose Cytarabine Consolidation Regimens, and Standard Versus Mycop[NCT02416388]Phase 2/Phase 33,100 participants (Anticipated)Interventional2015-01-31Recruiting
AML08: A Phase II Randomized Trial of Clofarabine Plus Cytarabine Versus Conventional Induction Therapy And A Phase II Study Of Natural Killer Cell Transplantation In Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT00703820]Phase 3324 participants (Actual)Interventional2008-08-04Completed
Non Randomized, Multicenter, Prospective Pediatric Hodgkin Lymphoma Treatment Trial Stratified According to Initial Risk Factors and Response to Chemotherapy, Reduced Cumulative Doses of Antineoplastic Agents and Radiotherapy.[NCT03500133]Phase 4500 participants (Anticipated)Interventional2017-10-06Recruiting
A PHASE III, MULTICENTRE, RANDOMIZED, OPEN LABEL CLINICAL TRIAL OF AZACYTIDINE (VIDAZA®) VERSUS FLUDARABINE AND CYTARABINE (FLUGA SCHEME) IN ELDERLY PATIENTS WITH NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA.[NCT02319135]Phase 3289 participants (Actual)Interventional2014-10-31Completed
Dual Growth Factor (rhTPO + G-CSF) and Chemotherapy Combination Regimen for Elderly Patients With Acute Myeloid Leukemia: A Phase II Single-Arm Multicenter Study[NCT05258799]130 participants (Anticipated)Interventional2022-01-21Recruiting
A Phase 2 Study of Cytokine-induced Memory-like NK Cells in Combination With Chemotherapy in Pediatric Patients With Refractory or Relapsed AML[NCT04354025]Phase 20 participants (Actual)Interventional2023-06-30Withdrawn(stopped due to Principal investigator decided not to move forward with the study.)
A Phase II Study of Dasatinib (Sprycel®) (NSC #732517) as Primary Therapy Followed by Transplantation for Adults >/= 18 Years With Newly Diagnosed Ph+ Acute Lymphoblastic Leukemia by CALGB, ECOG and SWOG[NCT01256398]Phase 266 participants (Actual)Interventional2010-12-14Completed
Evaluation of the Safety and Efficacy of Reduced-intensity Immunoablation and Autologous Hematopoietic Stem Cell Transplantation (AHSCT) in Multiple Sclerosis[NCT03113162]Phase 115 participants (Anticipated)Interventional2015-05-29Recruiting
A Phase I-II Trial of DA-EPOCH-R Plus Ixazomib as Frontline Therapy for Patients With MYC-aberrant Lymphoid Malignancies: The DACIPHOR Regimen[NCT02481310]Phase 1/Phase 238 participants (Actual)Interventional2015-10-28Active, not recruiting
A 5 Day Course of Fludarabine and Cytarabine Followed by Full Intensity Allogeneic Stem Cell Transplantation (FA5-Bucy) in Treating Patients With High-risk, Recurrent or Refractory Acute Leukemia and Advanced Myelodysplastic Syndrome[NCT02328950]50 participants (Anticipated)Observational2014-12-31Recruiting
A Dose Escalation Phase I/II Study of Clofarabine Plus Cytarabine With Growth Factor Priming in Patients Who Are Not Felt to be Candidates for More Aggressive Treatment, With Int-2 and High-Risk MDS[NCT00503880]Phase 1/Phase 22 participants (Actual)Interventional2007-05-07Terminated(stopped due to Genzyme discontinued Funding)
Risk Stratified Treatment for Patients With Newly Diagnosed Juvenile Myelomonocytic Leukemia: A Phase I/II Non-randomized Study of Trametinib and Azacitidine With or Without Chemotherapy (IND #164058)[NCT05849662]Phase 1/Phase 258 participants (Anticipated)Interventional2023-11-30Not yet recruiting
An Open Label Study to Evaluate the Feasibility of CPI-613 Given With High Dose Cytarabine and Mitoxantrone in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML)[NCT02484391]Phase 250 participants (Anticipated)Interventional2015-09-30Completed
Phase II Study of Etoposide, Methylprednisolone, High-dose Cytarabine and Oxaliplatin (ESHAOX) for Patients With Refractory or Relapsed Non-Hodgkin's Lymphoma[NCT00336583]Phase 227 participants (Actual)Interventional2006-06-30Completed
A 2:1 Randomized Phase II Trial to Compare the Efficacy and Safety of Standard Chemotherapy Plus Quizartinib Versus Standard Chemotherapy Plus Placebo in Adult Patients With Newly Diagnosed FLT3 Wild-type AML[NCT04107727]Phase 2273 participants (Actual)Interventional2019-09-05Active, not recruiting
A Phase II, Open-label, Single Arm Study to Evaluate the Safety, Efficacy, and Pharmacokinetics of Twice Daily Midostaurin (PKC412) Combined With Standard Chemotherapy and as a Single Agent Post-consolidation Therapy in Children With Untreated FLT3-mutate[NCT03591510]Phase 223 participants (Anticipated)Interventional2019-03-13Recruiting
An Investigator Sponsored Phase I Trial of Selinexor (KPT-330) Plus FLAG-Ida for the Treatment of Relapsing/Refractory AML[NCT03661515]Phase 116 participants (Actual)Interventional2018-07-17Completed
A Prospective Randomized Comparison of Idarubicin and High-dose Daunorubicin in Combination With Cytarabine in the Induction Chemotherapy for Acute Myeloid Leukemia[NCT01145846]Phase 3316 participants (Anticipated)Interventional2010-05-31Recruiting
Prospective Evaluation of Sorafenib Combined With Standard Therapy in Newly Diagnosed Adult Core-binding Factor Acute Myeloid Leukemia: an Open-label , Randomised Controlled, Multicenter Phase II Trial[NCT05404516]Phase 288 participants (Anticipated)Interventional2020-01-01Recruiting
A Phase I/II Study of CP-4055 in Patients With Platinum Resistant Ovarian Cancer[NCT00831636]Phase 1/Phase 228 participants (Actual)Interventional2008-04-30Completed
"Comparing the Efficacy and Safety of Venetoclax Combined With Decitabine Versus Conventional 7+3 Induction Chemotherapy of Acute Myeloid Leukemia in Young Adults"[NCT05177731]Phase 3188 participants (Anticipated)Interventional2022-03-01Recruiting
A Prospective Randomized Comparison of High-dose Cytarabine an High-dose Daunorubicin in the Induction Chemothrapy for Acute Myeloid Leukemia[NCT03507842]Phase 3380 participants (Actual)Interventional2018-03-01Enrolling by invitation
A Phase III Multicentric Randomized Study of the Combination of Repeated Doses of Gemtuzumab Ozogamicin (GO) With Daunorubicin and Cytarabine Versus Daunorubicin and Cytarabine in Untreated Patients With Acute Myeloid Leukemia (AML) Aged of 50-70 Years Ol[NCT00927498]Phase 3280 participants (Actual)Interventional2007-12-31Completed
Phase II Investigator Sponsored Study of CPX-351 in Combination With Ivosidenib for Patients With IDH1 Mutated Acute Myeloid Leukemia or High-Risk MDS[NCT04493164]Phase 230 participants (Anticipated)Interventional2020-12-30Recruiting
A Phase III Randomized Trial of Blinatumomab for Newly Diagnosed BCR-ABL-Negative B Lineage Acute Lymphoblastic Leukemia in Adults[NCT02003222]Phase 3488 participants (Actual)Interventional2014-05-19Active, not recruiting
A Phase II Study of Rituximab/Bendamustine Followed by Rituximab/Cytarabine for Untreated Mantle Cell Lymphoma[NCT01661881]Phase 223 participants (Actual)Interventional2012-08-16Active, not recruiting
Immunotherapy With Ex Vivo-Expanded Cord Blood-Derived CAR-NK Cells Combined With High-Dose Chemotherapy and Autologous Stem Cell Transplantation for B-Cell Lymphoma[NCT03579927]Phase 1/Phase 20 participants (Actual)Interventional2019-10-03Withdrawn(stopped due to Lack of Funding)
A Phase 2 Study of the JAK1/JAK2 Inhibitor Ruxolitinib With Chemotherapy in Children With De Novo High-Risk CRLF2-Rearranged and/or JAK Pathway-Mutant Acute Lymphoblastic Leukemia[NCT02723994]Phase 2171 participants (Actual)Interventional2016-09-30Active, not recruiting
Phase II Study of Combination of Hyper-CVAD and Dasatinib in Patients With Philadelphia (Ph) Chromosome Positive and/or BCR-ABL Positive Acute Lymphoblastic Leukemia (ALL)[NCT00390793]Phase 2107 participants (Actual)Interventional2006-09-28Active, not recruiting
Phase I Study of MLN 9708 in Addition to Chemotherapy for the Treatment of Acute Lymphoblastic Leukemia in Older Adults[NCT02228772]Phase 119 participants (Actual)Interventional2014-12-31Completed
Outcomes of Patients After Allogenic Hematopoietic Cell Transplantation With Decitabine-containing Conditioning Regimen and Acetylcysteine Treatment[NCT04945096]Phase 3100 participants (Anticipated)Interventional2021-07-01Not yet recruiting
A Phase 1/2 Study of INCB053914 in Subjects With Advanced Malignancies[NCT02587598]Phase 1/Phase 297 participants (Actual)Interventional2015-12-29Terminated
A Phase I Study of MEK Inhibitor MEK162 Combined With Idarubicin and Cytarabine Induction in Patients With Relapsed/Refractory RAS-Mutated Acute Myeloid Leukemia[NCT02049801]Phase 11 participants (Actual)Interventional2014-12-31Terminated(stopped due to logistics)
CPX-351 Plus Enasidenib for Relapsed Acute Myelogenous Leukemia Characterized by the IDH2 Mutation[NCT03825796]Phase 22 participants (Actual)Interventional2019-04-12Active, not recruiting
Phase I/II Study of VELCADE®-BEAM and Autologous Hematopoietic Stem Cell Transplantation for Relapsed Indolent Non-Hodgkin's Lymphoma, Transformed or Mantle Cell Lymphoma[NCT00571493]Phase 1/Phase 242 participants (Actual)Interventional2006-04-14Completed
"Magrolimab Plus Intensive Chemotherapy in Newly Diagnosed ELN 2022 Intermediate or Adverse-risk AML or High Risk MDS Patients Intended to Undergo Allogeneic Stem Cell Transplantation, a Phase 2, Single-arm, Open-Label Study"[NCT05829434]Phase 2108 participants (Anticipated)Interventional2024-01-31Not yet recruiting
International Collaborative Treatment Protocol for Children and Adolescents With Acute Lymphoblastic Leukemia - AIEOP-BFM ALL 2017[NCT03643276]Phase 35,000 participants (Anticipated)Interventional2018-07-15Recruiting
Phase I Trial of Ruxolitinib in Combination With a Pediatric Based-regimen for Adolescents and Young Adults (AYAs) With Ph-like Acute Lymphoblastic Leukemia (ALL)[NCT03571321]Phase 115 participants (Anticipated)Interventional2019-05-28Recruiting
An Investigator-Sponsored Randomized Phase II Study of Selinexor in Combination With Induction/Consolidation Therapy in Acute Myeloid Leukemia Patients[NCT02835222]Phase 2100 participants (Anticipated)Interventional2018-02-02Recruiting
Randomized Phase III Study of Intensive Chemotherapy With or Without Dasatinib (Sprycel™) in Adult Patients With Newly Diagnosed Core-Binding Factor Acute Myeloid Leukemia (CBF-AML)[NCT02013648]Phase 3203 participants (Anticipated)Interventional2014-07-31Active, not recruiting
A Randomized Phase 2 Trial of Brentuximab Vedotin (SGN35, NSC# 749710), or Crizotinib (NSC#749005, Commercially Labeled) in Combination With Chemotherapy for Newly Diagnosed Patients With Anaplastic Large Cell Lymphoma (ALCL)[NCT01979536]Phase 2137 participants (Actual)Interventional2013-11-13Active, not recruiting
Phase II Study of Combination of Hyper-CVAD and Ponatinib in Patients With Philadelphia (PH) Chromosome Positive and/or BCR-ABL Positive Acute Lymphoblastic Leukemia (ALL)[NCT01424982]Phase 288 participants (Actual)Interventional2011-10-05Active, not recruiting
Evaluation of the Efficacy of Induction-consolidation Treatment Using a Double Induction in Patients With AML <60 Years Old, Depending on the Percentage of Blasts in the 14 Day, Residual Disease and Leukemic Hematopoietic Cells[NCT02072811]Phase 3400 participants (Anticipated)Interventional2014-02-28Recruiting
Phase Ib Study of Ficlatuzumab With High Dose Cytarabine (HiDAC) in Relapsed and Refractory AML[NCT02109627]Phase 117 participants (Actual)Interventional2015-05-01Terminated(stopped due to Low Accrual)
A Randomized, Open-Label Study of Oral CEP-701 Administered in Sequence With Standard Chemotherapy to Patients With Relapsed Acute Myeloid Leukemia (AML) Expressing FLT-3 Activating Mutations[NCT00079482]Phase 2224 participants (Actual)Interventional2003-10-31Completed
A Phase 1b/2 Study of Pinometostat in Combination With Standard Induction Chemotherapy in Newly Diagnosed Acute Myeloid Leukemia With MLL Rearrangement[NCT03724084]Phase 1/Phase 25 participants (Actual)Interventional2019-01-25Terminated(stopped due to Other - Study agent no longer available)
Randomized Phase 3 Trial of Decitabine Versus Patient's Choice With Physician's Advice of Either Supportive Care or Low-Dose Cytarabine for the Treatment of Older Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT00260832]Phase 3485 participants (Actual)Interventional2005-11-30Completed
Phase I Study of Decitabine, Vorinostat, and Cytarabine in Acute Myeloid Leukemia[NCT01130506]Phase 117 participants (Actual)Interventional2010-05-17Completed
Prospective Evaluation of Standard Chemotherapy Regimen of Idarubicin and Cytarabine in Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)[NCT00422591]Phase 2175 participants (Actual)Interventional2006-12-31Completed
Dose Finding Safety Run-in Phase Followed by a Randomized Phase II Trial of Intensive Chemotherapy With or Without Volasertib (BI 6727) Administered Prior or After Chemotherapy in Patients With Newly Diagnosed High-Risk Myelodysplastic Syndrome (MDS) and [NCT02198482]Phase 26 participants (Actual)Interventional2016-02-29Terminated(stopped due to development program of study drug volasertib was stopped by Boehringer Ingelheim due to manufacturing problems)
A Phase 3, Prospective, Randomized Multi-center Intervention Trial of Early Intensification in AML Patients Bearing FLT3 Mutations Based on Peripheral Blast Clearance: A MYNERVA-GIMEMA Study[NCT04174612]Phase 3172 participants (Anticipated)Interventional2020-04-24Recruiting
A Phase II Trial of Sequential Chemotherapy, Imatinib Mesylate (Gleevec, STI571) (NSC # 716051), and Transplantation for Adults With Newly Diagnosed Ph+ Acute Lymphoblastic Leukemia by the CALGB and SWOG[NCT00039377]Phase 258 participants (Actual)Interventional2002-04-30Completed
High-Dose Chemo-Radiotherapy for Patients With Primary Refractory and Relapsed Hodgkin's Disease[NCT00003631]Phase 2118 participants (Actual)Interventional1998-08-31Completed
Randomized Trial of Gilteritinib vs Midostaurin in FLT3 Mutated Acute Myeloid Leukemia (AML)[NCT03836209]Phase 2181 participants (Actual)Interventional2019-12-06Active, not recruiting
Phase 1b/2 Study of Carfilzomib in Combination With Induction Chemotherapy in Children With Relapsed or Refractory Acute Lymphoblastic Leukemia[NCT02303821]Phase 1130 participants (Anticipated)Interventional2015-02-16Recruiting
A Phase III Randomized Trial Investigating Bortezomib (NSC# 681239) on a Modified Augmented BFM (ABFM) Backbone in Newly Diagnosed T-Lymphoblastic Leukemia (T-ALL) and T-Lymphoblastic Lymphoma (T-LLy)[NCT02112916]Phase 3847 participants (Actual)Interventional2014-10-04Active, not recruiting
"A Novel Pediatric-Inspired Regimen With Reduced Myelosuppressive Drugs for Adults (Aged 18-60) With Newly Diagnosed Ph Negative Acute Lymphoblastic Leukemia"[NCT01920737]Phase 239 participants (Actual)Interventional2013-08-31Active, not recruiting
A Phase 1b/2, Open-Label Trial to Assess the Safety and Preliminary Efficacy of Epcoritamab (GEN3013; DuoBody®-CD3xCD20) in Combination With Other Agents in Subjects With B-cell Non-Hodgkin Lymphoma (B-NHL)[NCT04663347]Phase 1/Phase 2662 participants (Anticipated)Interventional2020-11-03Recruiting
Dose-Finding Run-in Phase I Followed by a Phase III, Multicenter, Randomized, Double-Blind, Placebo-controlled Study of Crenolanib in Combination With Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia and Activating FLT3 Mutation[NCT02298166]Phase 39 participants (Actual)Interventional2016-11-17Terminated(stopped due to The manufacturer Arog Pharmaceuticals Inc has terminated the Agreement Concerning the Support of an Investigator Initiated Trial this became valid on 2020/03/09)
Safety, Tolerability and Efficacy of MB07133 Plus Sintilimab in Patients With Hepatocellular Carcinoma,a Phase 1/2a Study[NCT06141109]Phase 1/Phase 239 participants (Anticipated)Interventional2022-01-18Recruiting
International Phase 3 Trial in Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia (Ph+ALL) Testing Imatinib in Combination With Two Different Cytotoxic Chemotherapy Backbones[NCT03007147]Phase 3475 participants (Anticipated)Interventional2017-08-08Recruiting
A Phase 2 Study of Inotuzumab Ozogamicin (NSC# 772518) in Children and Young Adults With Relapsed or Refractory CD22+ B-Acute Lymphoblastic Leukemia (B-ALL)[NCT02981628]Phase 280 participants (Anticipated)Interventional2017-06-19Active, not recruiting
Phase 1 Study of Ibrutinib and Immuno-Chemotherapy Using Temozolomide, Etoposide, Doxil, Dexamethasone, Ibrutinib, Rituximab (TEDDI-R) in Primary CNS Lymphoma[NCT02203526]Phase 193 participants (Anticipated)Interventional2014-08-14Recruiting
Risk-Stratified Randomized Phase III Testing of Blinatumomab (NSC#765986) in First Relapse of Childhood B-Lymphoblastic Leukemia (B-ALL)[NCT02101853]Phase 3669 participants (Actual)Interventional2014-12-17Active, not recruiting
Intensified Methotrexate, Nelarabine (Compound 506U78) and Augmented BFM Therapy for Children and Young Adults With Newly Diagnosed T-cell Acute Lymphoblastic Leukemia (ALL) or T-cell Lymphoblastic Lymphoma[NCT00408005]Phase 31,895 participants (Actual)Interventional2007-01-22Active, not recruiting
an Single Center,Single Arm, Phase 3 Study to Evaluate Efficacy and Safety of PD-1 Inhibitor Combined With Azacytidine and HAG Regimen for Patients With Relapsed or Refractory Acute Myeloid Leukemia.[NCT04722952]Phase 330 participants (Anticipated)Interventional2021-05-01Recruiting
High Risk B-Precursor Acute Lymphoblastic Leukemia (ALL)[NCT00075725]Phase 33,154 participants (Actual)Interventional2003-12-29Completed
Venetoclax, Cladribine Plus Low-dose Cytarabine for Relapsed/Refractory Philadelphia Chromosome-negative (Ph-) B-cell Acute Lymphoblastic Leukemia (B-ALL): a Single-arm, Multicenter Study[NCT05657652]36 participants (Anticipated)Interventional2022-10-01Recruiting
A Phase III Groupwide Study of Dose-Intensive Response-Based Chemotherapy and Radiation Therapy for Children and Adolescents With Newly Diagnosed Intermediate Risk Hodgkin Disease[NCT00025259]Phase 31,734 participants (Actual)Interventional2002-09-30Completed
Phase II Trial Of Yttrium-90-Ibritumomab Tiuxetan (Zevalin®) Radioimmunotherapy After Cytoreduction With ESHAP Chemotherapy In Patients With Relapsed Follicular Non-Hodgkin's Lymphoma[NCT00732498]Phase 228 participants (Actual)Interventional2006-05-15Completed
Phase II Study of the Sequential Combination of Low-Intensity Chemotherapy and Ponatinib Followed by Blinatumomab and Ponatinib in Patients With Philadelphia Chromosome (Ph)-Positive and/or BCR-ABL Positive Acute Lymphoblastic Leukemia (ALL)[NCT03147612]Phase 221 participants (Actual)Interventional2018-02-08Active, not recruiting
Clinical Study on the Safety and Efficacy of QN-023a Targeting CD33 in Acute Myeloid Leukemia[NCT05665075]Phase 119 participants (Anticipated)Interventional2022-12-24Recruiting
A Phase I Study of Bortezomib in Combination With MEC (Mitoxantrone, Etoposide, and Intermediate-Dose Cytarabine) for Relapsed/ Refractory Acute Myelogenous Leukemia (AML)[NCT01127009]Phase 13 participants (Actual)Interventional2010-07-31Completed
Intensive Treatment For T-CELL Acute Lymphoblastic Leukemia and Advanced Stage Lymphoblastic Non-Hodgkin's Lymphoma: A Pediatric Oncology Group Phase III Study[NCT01230983]Phase 3573 participants (Actual)Interventional1996-06-30Completed
iCare 2: Personalized Genomic Mutation Informed Treatment of Patients With Myelodysplastic Syndromes[NCT03446638]0 participants (Actual)Interventional2019-05-31Withdrawn(stopped due to administratively withdrawn)
Clinical Study of Mitoxantrone Hydrochloride Liposome, Carmostine, Etoposide and Cytarabine as Conditioning Regimen for Autologous Hematopoietic Stem Cell Transplantation in Patients With Lymphoma[NCT05681403]53 participants (Anticipated)Interventional2023-01-15Not yet recruiting
Autologous Hematopoietic Cell Transplantation for Core-binding Factor Positive Acute Myeloid Leukemia in the First Complete Remission[NCT01146977]Phase 243 participants (Anticipated)Interventional2010-01-31Recruiting
A Phase II Trial on Ofatumumab With IVAC Salvage Chemotherapy in Diffuse Large B Cell Lymphoma Patients Progressing or Relapsed After Prior R-CHOP Treatment Not Suitable for Autologous Stem Cell Transplant[NCT01481272]Phase 277 participants (Actual)Interventional2011-11-30Completed
Immune Reconstitution and Biomarker Identification in Patients With Newly Diagnosed Low and Intermediate Risk Hodgkins Lymphoma Receiving Chemotherapy With or Without Radiation Therapy: TXCH-HD-12A[NCT01858922]Phase 240 participants (Actual)Interventional2012-12-19Active, not recruiting
A Phase I/II Study of the Selective Inhibitor of Nuclear Export Selinexor (KPT-330) in Combination With Fludarabine and Cytarabine in Patients With Refractory or Relapsed Leukemia or Myelodysplastic Syndrome[NCT03071276]Phase 237 participants (Actual)Interventional2016-01-14Terminated(stopped due to Due to slow enrollment)
Young Adult Acute Lymphoid Leukemia (ALL): Intensification of Pediatric AIEOP LLA-2000 Treatment[NCT01156883]76 participants (Actual)Interventional2010-04-30Completed
A Randomized Study of Haploidentical Lymphocytes With Nivolumab and Intermediate Dose Cytarabine Versus Nivolumab and Intermediate Dose Cytarabine as Consolidation Treatment in Older Adults With Acute Myeloid Leukemia.[NCT03381118]Phase 216 participants (Actual)Interventional2017-06-30Terminated(stopped due to Unexpected toxicity)
Busulfan, Etoposide, Cytarabine, and Melphalan (BuEAM) as a Conditioning for Autologous Stem Cell Transplantation in Patients With B Cell Lymphoma Except for Diffuse Large B Cell Lymphoma[NCT01178645]Phase 242 participants (Anticipated)Interventional2010-07-31Recruiting
Clinical Efficacy and Safety of DLAAG Protocol in the Treatment of Refractory/Relapse of Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome With Blast Excess: a Multicenter, Single-arm, Prospective Clinical Study[NCT03356080]Phase 250 participants (Anticipated)Interventional2017-07-07Recruiting
A PHASE 1 DOSE ESCALATION STUDY TO EVALUATE THE SAFETY, PHARMACOKINETICS AND PHARMACODYNAMICS OF INTRAVENOUS PF-06747143, ADMINISTERED AS SINGLE AGENT OR IN COMBINATION WITH STANDARD CHEMOTHERAPY IN ADULT PATIENTS WITH ACUTE MYELOID LEUKEMIA[NCT02954653]Phase 18 participants (Actual)Interventional2016-11-28Terminated(stopped due to The study was terminated due to a change in sponsor prioritization.)
Efficacy and Safety of Venetoclax Combined With BEAM (Carmustine, Etoposide Cytarabine and Melphalan) Pretreatment in Autologous Stem Cell Transplantation for Diffuse Large B-cell Lymphoma: a Single-center, Randomized Clinical Study[NCT05863845]52 participants (Anticipated)Interventional2023-06-01Not yet recruiting
An International Phase II Trial Assessing Tolerability and Efficacy of Sequential Methotrexate-Aracytin-based Combination and R-ICE Combination, Followed by HD Chemotherapy Supported by ASCT, in Patients With Systemic B-cell Lymphoma With CNS Involvement [NCT02329080]Phase 276 participants (Actual)Interventional2014-12-31Active, not recruiting
A Phase II, Multicenter, Prospective, Non-randomised, Open-label, Clinical Trial to Evaluate Effectiveness and Safety of BeEAC Conditioning Regimen in Malignant Lymphoma Subjects With Indications to Autologous Hematopoietic Stem-cell Transplantation[NCT03315520]Phase 2100 participants (Anticipated)Interventional2016-01-22Recruiting
A Phase I/II Multi-center Study of HDM201 Added to Chemotherapy in Adult Subjects With Relapsed/Refractory (R/R) or Newly Diagnosed Acute Myeloid Leukemia (AML)[NCT03760445]Phase 1/Phase 20 participants (Actual)Interventional2019-11-15Withdrawn(stopped due to It was determined that the study design may not be optimal given the changing AML treatment landscape.)
Effectiveness of Microcurrents Therapy in Pressure Ulcers in Elderly People: Controlled and Randomized Triple Blind Clinical Trial[NCT03753581]30 participants (Actual)Interventional2018-10-31Completed
An Open-label Two Strata Study of Bisantrene in Combination With Cytarabine Arabinoside or Bisantrene in Combination With Oral Decitabine/Cedazuridine for the Treatment of Acute Myeloid Leukemia Patients With Extramedullary Disease[NCT05456269]Phase 10 participants (Actual)Interventional2022-07-29Withdrawn(stopped due to commercial reason)
An Open Phase I/IIa Trial to Investigate the Maximum Tolerated Dose, Safety, Pharmacokinetics, and Efficacy of Intravenous BI 6727 as Monotherapy or in Combination With Subcutaneous Cytarabine in Patients With Acute Myeloid Leukaemia[NCT00804856]Phase 2180 participants (Actual)Interventional2008-11-27Completed
A Phase 1 Trial of the Wee1 Kinase Inhibitor AZD1775 in Combination With Flag Chemotherapy in Children, Adolescents, and Young Adults With Relapsed or Refractory Acute Myeloid Leukemia[NCT02791919]Phase 10 participants (Actual)Interventional2017-05-25Withdrawn(stopped due to Other - Protocol moved to Disapproved)
A Pilot, Pharmacodynamic Correlate Trial of Sirolimus in Combination With Chemotherapy (Idarubicin, Cytarabine) for the Treatment of Newly Diagnosed Acute Myelogenous Leukemia[NCT01822015]Early Phase 155 participants (Actual)Interventional2013-03-15Completed
Phase 1 Study of ADI-PEG 20 Plus Low Dose Cytarabine in Older Patients With Acute Myeloid Leukemia[NCT02875093]Phase 123 participants (Actual)Interventional2017-01-20Terminated(stopped due to Study Termination)
Multicenter Automatic Defibrillator Implantation Trial With Cardiac Resynchronization Therapy Post Approval Registry[NCT01294449]394 participants (Actual)Observational2011-03-31Completed
A Phase II Study of Twice Daily Cytarabine and Fludarabine in Acute Myelogenous Leukemia and High-Risk Myelodysplastic Syndrome[NCT01019317]Phase 2151 participants (Actual)Interventional2009-11-30Completed
A Phase 3, Randomized, Controlled, Double-Blind, Multinational Clinical Study of the Efficacy and Safety of Vosaroxin and Cytarabine Versus Placebo and Cytarabine in Patients With First Relapsed or Refractory Acute Myeloid Leukemia (VALOR)[NCT01191801]Phase 3711 participants (Actual)Interventional2010-12-17Completed
A Pilot Study Incorporating Motexafin Gadolinium (MGd) Into High-dose Methotrexate (MTX)-Based Chemo-immunotherapy and Radiation for Patients With Newly Diagnosed Primary CNS Lymphoma[NCT00734773]Early Phase 10 participants (Actual)Interventional2008-11-30Withdrawn(stopped due to Lack of funding)
[NCT02662647]Phase 230 participants (Actual)Interventional2015-04-30Completed
An Open-Label, Multicenter, Phase 1b/2 Study of the Safety and Efficacy of KRT-232 When Administered Alone and in Combination With Low-Dose Cytarabine (LDAC) or Decitabine in Patients With Acute Myeloid Leukemia (AML)[NCT04113616]Phase 1/Phase 286 participants (Anticipated)Interventional2019-09-25Active, not recruiting
Phase II Study of Rituximab in Combination With Methotrexate, Doxorubicin, Cyclophosphamide, Leucovorin, Vincristine, Ifosfamide, Etoposide, Cytarabine and Mesna (R-MACLO/VAM) in Patients With Previously Untreated Mantle Cell Lymphoma[NCT00878254]Phase 225 participants (Actual)Interventional2009-03-25Active, not recruiting
EDOCH Alternating With DHAP Regimen Combined Rituximab or Not to Treat New Diagnosed Younger (Age≤65 Years) Mantle Cell Lymphoma in China: A Multicentre Phase III Trial[NCT02858804]Phase 455 participants (Anticipated)Interventional2016-01-31Recruiting
A Phase II Study of Sirolimus, Tacrolimus and Thymoglobulin®, as Graft-versus-Host- Disease Prophylaxis in Patients Undergoing Unrelated Donor Hematopoietic Cell Transplantation[NCT00691015]Phase 248 participants (Actual)Interventional2008-05-31Completed
A Dose Escalation Phase I/II Study of Lovastatin With High-Dose Cytarabine for Patients With Refractory or Relapsed Acute Myeloid Leukemia[NCT00583102]Phase 1/Phase 223 participants (Actual)Interventional2001-06-30Terminated(stopped due to Slow accrual, PI left institution)
Rituximab and Hyper-CVAD (Cyclophosphamide, Vincristine, Adriamycin, and Dexamethasone) for Burkitt's and Burkitt's -Like Leukemia/Lymphoma[NCT00669877]Phase 256 participants (Actual)Interventional2002-08-31Completed
Phase 1/2 Study of AMG 531 to Evaluate the Safety, Efficacy, and Pharmacokinetics in Patients With Aggressive Non-Hodgkin's Lymphoma Receiving R-HyperCVAD Alternating With R-Ara-C/MTX[NCT00299182]Phase 1/Phase 250 participants (Actual)Interventional2006-03-31Completed
Targeted Intensification by a Preparative Regimen for Patients With High-grade B-Cell Lymphoma Utilizing Standard-dose Yttrium-90 Ibritumomab Tiuxetan (Zevalin) Radioimmunotherapy (RIT) Combined With High-dose BEAM Followed by Autologous Stem Cell Transpl[NCT00689169]Phase 275 participants (Actual)Interventional2007-08-31Completed
Phase II Study of Idarubicin, Cytarabine, and Vorinostat in Patients With High-Risk MDS and AML[NCT00656617]Phase 2106 participants (Actual)Interventional2008-04-30Completed
Single Arm,Open Label,Phase I Clinical Study of Venetoclax Combined With CACAG Regimen in the Treatment of Newly Diagnosed Acute Myeloid Leukemia[NCT05659992]Phase 130 participants (Anticipated)Interventional2022-01-10Recruiting
A Phase I Study of Lenalidomide Therapy Prior to Re-induction Chemotherapy With Mitoxantrone, Etoposide, and Cytarabine (MEC) for the Treatment of Relapsed or Refractory Acute Myeloid Leukemia (AML)[NCT01904643]Phase 117 participants (Actual)Interventional2014-02-28Terminated(stopped due to Accrual factor)
Reduced Intensity Conditioning Regimen for Low- and Intermediate-risk Myelodysplastic Syndrome Patients Receiving Haploidentical Hematopoietic Stem Cell Transplantation[NCT03412266]Phase 250 participants (Anticipated)Interventional2018-02-01Recruiting
Randomized Phase 2 Study of Daunorubicin and Cytarabine Liposome + Pomalidomide Versus Daunorubicin and Cytarabine Liposome in Newly Diagnosed AML With MDS-Related Changes[NCT04802161]Phase 278 participants (Anticipated)Interventional2022-08-24Recruiting
Effect of Priming With Granulocyte-Macrophage Colony-Stimulating Factor During Chemotherapy and Comparison of Timed Sequential Chemotherapy vs 4 Courses of High Dose Cytarabine as Consolidation in Younger Adults With Newly Diagnosed AML[NCT00880243]Phase 3473 participants (Actual)Interventional1999-03-31Completed
A Phase I/II Study of Sorafenib (BAY 43-9006) in Combination With Low Dose ARA-C (CYTARABINE) in Elderly Patients With AML or High-Risk MDS[NCT00516828]Phase 1/Phase 221 participants (Actual)Interventional2007-11-27Completed
A Phase II Randomized Study of Rituximab-HCVAD Alternating With Rituximab-Methotrexate-Cytarabine Versus Standard Rituximab-CHOP Every 21 Days for Patients With Newly Diagnosed High Risk Aggressive B-Cell Non-Hodgkin's Lymphomas in Patients 60 Years Old o[NCT00290498]Phase 267 participants (Actual)Interventional2005-08-01Completed
A Dose-escalated Phase Ⅰ Trial to Assess the Tolerance and Pharmacokinetics of Combination of Donafenib and Cytarabine/Daunorubicin in Relapsed AML Patient[NCT04402723]Phase 18 participants (Actual)Interventional2018-11-06Terminated(stopped due to Corporate policy adjustments)
A Phase Ib Study to Investigate the Safety, Pharmacokinetics and Pharmacodynamics of APG-115 as a Single Agent or in Combination With Azacitidine or Cytarabine in Patients With Relapse/Refractory AML and Relapsed/Progressed High/Very High Risk MDS[NCT04275518]Phase 1102 participants (Anticipated)Interventional2020-07-06Recruiting
A Phase IV, Randomized Study to Evaluate the Safety and Efficacy of Idarubicin at Different Dosages Combined With Cytarabine as Induction Therapy for Newly Diagnosed Acute Myeloid Leukaemia[NCT02277847]Phase 4400 participants (Anticipated)Interventional2010-03-31Enrolling by invitation
Phase II Study of Multimodality Therapy in Mantle Cell Lymphoma[NCT00004231]Phase 20 participants Interventional1999-10-31Completed
A Phase I Study of Palbociclib in Combination With Chemotherapy in Pediatric Patients With Relapsed or Refractory Acute Lymphoblastic Leukemia[NCT03515200]Phase 112 participants (Actual)Interventional2018-04-20Terminated(stopped due to Due to departure of PI from St. Jude)
Combination of Rituximab and Methotrexate Followed by Rituximab and Cytarabine in Elderly Patients With Primary CNS Lymphoma[NCT03569995]Phase 235 participants (Anticipated)Interventional2018-11-30Recruiting
A Phase II Study of Crenolanib With Fludarabine and Cytarabine in Pediatric Patients With Relapsed/Refractory FLT3-Mutated Acute Myeloid Leukemia[NCT03324243]Phase 20 participants (Actual)Interventional2018-01-31Withdrawn(stopped due to Withdrawn: Study halted prior to enrollment of first participant)
A Phase I Study of Stem Cell Gene Therapy for HIV Mediated by Lentivector Transduced, Pre-Selected CD34+ Cells[NCT02797470]Phase 1/Phase 211 participants (Actual)Interventional2016-06-23Active, not recruiting
Fludarabine and Cytarabine Versus High-dose Cytarabine in Consolidation Treatment of Core-bing Factor Acute Myeloid Leukemia: A Prospective, Multicenter, Randomized Study[NCT02926586]Phase 4200 participants (Anticipated)Interventional2017-01-01Recruiting
Epigenetic Reprogramming in Relapse AML: A Phase 1 Study of Decitabine and Vorinostat Followed by Fludarabine, Cytarabine and G-CSF (FLAG) in Children and Young Adults With Relapsed/Refractory AML[NCT03263936]Phase 137 participants (Actual)Interventional2017-07-11Completed
"A Phase I Study of Intravenous (Emulsion) Fenretinide (4-HPR, NSC 374551) in Children With Recurrent or Resistant Acute Lymphoblastic Leukemia (ALL), Acute Myelogenous Leukemia (AML), and Non-Hodgkin's Lymphoma (NHL) IND #70,058"[NCT01187810]Phase 13 participants (Actual)Interventional2010-08-31Terminated(stopped due to drug supply)
AGORA-1 /ALFA 2100 Study : A Phase 2 Study of Gemtuzumab Ozogamicin (GO)-Gilteritinib Combination in Adults With FLT3-ITD Relapse/Refractory (R/R) AML[NCT05199051]Phase 250 participants (Anticipated)Interventional2023-06-03Recruiting
Inotuzumab Ozogamicin for Children With MRD Positive CD22+ Lymphoblastic Leukemia[NCT03913559]Phase 232 participants (Anticipated)Interventional2019-05-14Recruiting
"A Randomized Phase II/III Trial of Novel Therapeutics Versus Azacitidine in Newly Diagnosed Patients With Acute Myeloid Leukemia (AML) or High-Risk Myelodysplastic Syndrome (MDS), Age 60 or Older LEAP: Less-Intense AML Platform Trial"[NCT03092674]Phase 2/Phase 378 participants (Actual)Interventional2018-02-02Active, not recruiting
A Multi-center Randomized Phase II Study of the Impact of CD34+ Cell Dose on Absolute Lymphocyte Count Following High-Dose Therapy and Autologous Stem Cell Transplantation for Relapsed and Refractory Diffuse Large B-cell Lymphoma (DLBCL)[NCT02570542]Phase 259 participants (Actual)Interventional2015-10-31Active, not recruiting
Evaluation of the Incidence and Prognostic Impact of Molecular and Genetic Abnormalities in Patients With Acute Myeloid Leukemia, Myelodysplastic Syndrome and Myeloproliferative Neoplasms[NCT02084563]Phase 2455 participants (Actual)Interventional2012-10-31Completed
CBA Versus FBA Conditioning Followed by Haploidentical Allogeneic HSCT in Treatment of High Risk and Refractory AML[NCT03384225]Phase 3120 participants (Anticipated)Interventional2016-08-01Recruiting
A PALG Prospective Multicenter Clinical Trial to Compare the Efficacy of Two Standard Induction Therapies (DA-90 vs DAC) and Two Standard Salvage Regimens (FLAG-IDA vs CLAG-M) in AML Patients ≤ 60 Years Old[NCT03257241]Phase 3582 participants (Anticipated)Interventional2017-07-03Recruiting
A Phase II Study of Double Induction Chemotherapy for Newly Diagnosed Non-L3 Adult Acute Lymphoblastic Leukemia With Investigation of Minimal Residual Disease and Risk of Relapse Following Maintenance Chemotherapy[NCT00109837]Phase 279 participants (Actual)Interventional2005-04-30Completed
National Treatment Program of Philadelphia Chromosome-negative Adult Acute Lymphoblastic Leukemia With Pegylated Asparaginase Added to a Lineage-Targeted Risk- and Minimal Residual Disease-Oriented Strategy[NCT02067143]Phase 2204 participants (Actual)Interventional2014-05-20Completed
A Randomized, Double-blind, Multiple-Dose, Two-Cycle, Parallel-Group, Bioequivalence Pretrial of Daunorubicin Cytarabine Liposome for Injection in Older, Naive AML Patients[NCT04920500]16 participants (Anticipated)Interventional2020-09-04Recruiting
Safety and Efficacy of an Outpatient Schedule of Rituximab, Cytarabine, Carboplatin, and Dexamethasone in Relapsed/Refractory Non-Hodgkin Lymphoma. Phase I/II Trial.[NCT03892421]Phase 1/Phase 222 participants (Actual)Interventional2018-04-05Completed
Treatment of Newly Diagnosed Standard Risk Acute Lymphoblastic Leukemia in[NCT06099366]Phase 2116 participants (Anticipated)Interventional2024-01-15Not yet recruiting
Improving Risk Assessment of AML With a Precision Genomic Strategy to Assess Mutation Clearance[NCT02756962]Phase 2110 participants (Anticipated)Interventional2016-07-06Recruiting
Phase II Study of Cladribine Plus Idarubicin Plus Cytarabine (ARAC) in Patients With AML, HR MDS, or Myeloid Blast Phase of CML[NCT02115295]Phase 2458 participants (Anticipated)Interventional2014-05-19Recruiting
A Phase III Randomized Trial for Patients With De Novo AML Using Bortezomib and Sorafenib (NSC# 681239, NSC# 724772) for Patients With High Allelic Ratio FLT3/ITD[NCT01371981]Phase 31,645 participants (Actual)Interventional2011-06-20Active, not recruiting
Efficacy and Safety of Cladribine in Combination With G-CSF, Low-dose Cytarabine and Aclarubicin in Patients With Refractory/Relapsed Acute Myeloid Leukemia: a Phase 2 Clinical Trial[NCT03181815]Phase 248 participants (Anticipated)Interventional2016-01-01Recruiting
A Phase 1b Study of Menin Inhibitor SNDX-5613 in Combination With Daunorubicin and Cytarabine in Newly Diagnosed Patients With Acute Myeloid Leukemia and NPM1 Mutated/FLT3 Wildtype or MLL/KMT2A Rearranged Disease.[NCT05886049]Phase 128 participants (Anticipated)Interventional2023-12-19Not yet recruiting
Eradicating Measurable Residual Disease in Patients With Acute Myeloid Leukemia (AML) Prior to StEm Cell Transplantation (ERASE): A MyeloMATCH Treatment Trial[NCT05554419]Phase 2184 participants (Anticipated)Interventional2024-08-16Not yet recruiting
A Phase 3, Randomized, Open-Label Study Evaluating the Safety and Efficacy of Magrolimab in Combination With Azacitidine Versus Physician's Choice of Venetoclax in Combination With Azacitidine or Intensive Chemotherapy in Previously Untreated Patients Wit[NCT04778397]Phase 3346 participants (Anticipated)Interventional2021-07-01Active, not recruiting
A Pilot Study of Immunotherapy Including Haploidentical NK Cell Infusion Following CD133+ Positively-Selected Autologous Hematopoietic Stem Cells in Children With High Risk Solid Tumors or Lymphomas[NCT02130869]Phase 18 participants (Actual)Interventional2014-10-10Completed
Standard Risk B-precursor Acute Lymphoblastic Leukemia (ALL)[NCT00103285]Phase 35,377 participants (Actual)Interventional2005-04-11Completed
Phase II Study of Intensive Chemotherapy and Rituximab in Burkitt Lymphoma[NCT00126191]Phase 210 participants (Actual)Interventional2005-07-31Terminated(stopped due to closed due to slow accrual)
A Collaboration Phase 2 Study of Venetoclax in Combination With Conventional Chemotherapy in Pediatric Patients With Acute Myeloid Leukemia[NCT05955261]Phase 270 participants (Anticipated)Interventional2023-07-25Recruiting
A Phase 1 Study Combining Venetoclax With a Pediatric-Inspired Regimen for Newly Diagnosed Adults With B Cell Ph-Like Acute Lymphoblastic Leukemia[NCT05157971]Phase 16 participants (Anticipated)Interventional2022-03-17Recruiting
Phase I Study of Induction Therapy With Cytarabine, High-Dose Mitoxantrone and Dasatinib for Patients With Philadelphia-Chromosome Positive (Ph+) Acute Lymphoblastic Leukemia (ALL): ALL-6 Protocol[NCT00940524]Phase 17 participants (Actual)Interventional2009-07-31Completed
A Randomized, Phase II Study of CX-01 Combined With Standard Induction Therapy for Newly Diagnosed Acute Myeloid Leukemia[NCT02873338]Phase 275 participants (Actual)Interventional2016-08-31Completed
Phase I Investigator Sponsored Study to Assess the Tolerability and Efficacy of Selinexor in Combination With High Dose Cytarabine (HiDAC) and Mitoxantrone Chemotherapy for Remission Induction in Acute Myelogenous Leukemia (AML)[NCT02573363]Phase 125 participants (Actual)Interventional2015-10-07Completed
Phase III Open-Label Randomized Study of Amonafide L-Malate in Combination With Cytarabine Compared to Daunorubicin in Combination With Cytarabine in Patients With Secondary Acute Myeloid Leukemia (AML)- The ACCEDE Study[NCT00715637]Phase 3420 participants (Anticipated)Interventional2007-06-30Active, not recruiting
A Phase II Study of Reduced Intensity Allogeneic Transplantation for Refractory Hodgkin Lymphoma[NCT00908180]Phase 247 participants (Anticipated)Interventional2009-07-31Not yet recruiting
Venetoclax-Navitoclax With Cladribine-based Salvage Therapy in Patients With Relapsed/Refractory Acute Myeloid Leukemia[NCT06007911]Phase 136 participants (Anticipated)Interventional2024-02-29Not yet recruiting
Phase II Prospective Randomized Control Trial of Cladribine and Low-Dose Cytarabine (LoDAC) Alternating With Decitabine vs. Hypomethylating Agents (HMA) Plus Venetoclax as Frontline Therapy for AML or High-Grade MDS in Patients Unfit for Intensive Inducti[NCT05766514]Phase 298 participants (Anticipated)Interventional2024-03-31Not yet recruiting
A Phase I Study of Tagraxofusp With or Without Chemotherapy in Pediatric Patients With Relapsed or Refractory CD123 Expressing Hematologic Malignancies[NCT05476770]Phase 154 participants (Anticipated)Interventional2022-11-11Recruiting
Phase 1/2 Study of Liposomal Annamycin in Combination With Cytarabine for the Treatment of Subjects With Acute Myeloid Leukemia (AML)[NCT05319587]Phase 1/Phase 263 participants (Anticipated)Interventional2022-09-29Recruiting
Randomized Phase III Study of Standard Intensive Chemotherapy Versus Intensive Chemotherapy With CPX-351 in Adult Patients With Newly Diagnosed AML and Intermediate- or Adverse Genetics[NCT03897127]Phase 3882 participants (Anticipated)Interventional2019-09-04Recruiting
Phase II Study of the Combination of Blinatumomab and Ponatinib in Patients With Philadelphia Chromosome (Ph)-Positive and/or BCR-ABL Positive Acute Lymphoblastic Leukemia (ALL)[NCT03263572]Phase 290 participants (Anticipated)Interventional2017-11-29Recruiting
Phase II Study of the Hyper-CVAD Regimen in Sequential Combination With Blinatumomab With or Without Inotuzumab Ozogamicin as Frontline Therapy for Adults With B-Cell Lineage Acute Lymphocytic Leukemia[NCT02877303]Phase 280 participants (Anticipated)Interventional2016-11-01Recruiting
A Phase 1/2, Multicenter, Open-label Study of FT-2102 as a Single Agent and in Combination With Azacitidine or Cytarabine in Patients With Acute Myeloid Leukemia or Myelodysplastic Syndrome With an IDH1 Mutation[NCT02719574]Phase 1/Phase 2336 participants (Actual)Interventional2016-04-30Active, not recruiting
Clofarabine in Combination With Cytarabine (Ara-C) and G-CSF Priming Followed by Infusion of Ex Vivo Expanded Cord Blood Progenitors for Patients With AML[NCT01031368]Phase 129 participants (Actual)Interventional2009-12-31Completed
Total Therapy Study XVI for Newly Diagnosed Patients With Acute Lymphoblastic Leukemia[NCT00549848]Phase 3600 participants (Actual)Interventional2007-10-29Completed
An Open-label Randomized Controlled Phase II Study of AS1411 Combined With Cytarabine in the Treatment of Patients With Primary Refractory or Relapsed Acute Myeloid Leukemia[NCT01034410]Phase 290 participants (Anticipated)Interventional2010-01-31Terminated
Aezea (Cenersen) in Combination With Chemotherapy for Treatment of Acute Myelogenous Leukemia Subjects ≥55 Years of Age With No Response to Single Frontline Induction Course in a Randomized Double-Blind Placebo-Controlled Multi-Center Study[NCT00967512]Phase 20 participants (Actual)Interventional2012-01-31Withdrawn(stopped due to Study Was Terminated due to lack of Funding.)
Phase II Study of the Combination of CPX-351 and Glasdegib in Previously Untreated Patients With Acute Myelogenous Leukemia With MDS Related Changes or Therapy-related Acute Myeloid Leukemia[NCT04231851]Phase 230 participants (Anticipated)Interventional2020-02-19Recruiting
A Feasibility Pilot and Phase II Study Of Chemoimmunotherapy With Epratuzumab (IND #12034) for Children With Relapsed CD22-Positive Acute Lymphoblastic Leukemia (ALL)[NCT00098839]Phase 1/Phase 2134 participants (Actual)Interventional2005-02-28Completed
Phase 1b/2, Open-Label, Multicenter, Dose-Escalating, Clinical Study of the Safety, Tolerability, and PK and PD Profiles of Voreloxin Injection in Combination With Cytarabine in Patients With Relapsed or Refractory AML[NCT00541866]Phase 1/Phase 2110 participants (Actual)Interventional2007-10-06Completed
A Phase II Single Arm Study of the Use of CODOX-M/IVAC With Rituximab (R-CODOX-M/IVAC) in the Treatment of Patients With Diffuse Large B-Cell Lymphoma (International Prognostic Index High or High-Intermediate Risk)[NCT00974792]Phase 2150 participants (Anticipated)Interventional2006-01-31Recruiting
Safety and Efficacy of Liposomal Cytarabine in Combination With Radiotherapy (RT) and Lomustine for the Treatment of Leptomeningeal Metastasis From Malignant Melanoma[NCT01563614]Phase 11 participants (Actual)Interventional2012-03-31Terminated(stopped due to No patients can be recruited for this trial anymore due to other therapeutical approaches that became available.)
A PHASE I-II MULTICENTER STUDY OF THE CLORETAZINE-DAUNORUBICIN-ARACYTINE COMBINATION FOR THE TREATMENT OF ACUTE MYELOID LEUKEMIA (AML) WITH UNFAVORABLE CYTOGENETICS[NCT00840684]Phase 1/Phase 2135 participants (Anticipated)Interventional2009-01-31Completed
A Pilot Study To Determine The Toxicity Of The Addition Of Rituximab To The Induction And Consolidation Phases And The Addition Of Rasburicase To The Reduction Phase In Children With Newly Diagnosed Advanced B-Cell Leukemia/Lymphoma Treated With LMB/FAB T[NCT00057811]Phase 297 participants (Actual)Interventional2004-06-30Completed
A Phase I/II Dose Escalation Study of Subcutaneous Campath-1H (NSC #715969, IND #10864) During Intensification Therapy in Adults With Untreated Acute Lymphoblastic Leukemia (ALL)[NCT00061945]Phase 1/Phase 2302 participants (Actual)Interventional2003-06-30Completed
Phase II Study of Rituximab in Combination With Methotrexate, Doxorubicin, Cyclophosphamide, Leucovorin, Vincristine, Ifosfamide, Etoposide, Cytarabine and Mesna (MACLO/IVAM) in Patients With Previously Untreated Mantle Cell Lymphoma[NCT00450801]Phase 222 participants (Actual)Interventional2004-04-30Completed
A Phase I Study of Intravenous Plerixafor in Combination With Mitoxantrone Etoposide and Cytarabine for Relapsed or Refractory Acute Myeloid Leukemia[NCT01027923]Phase 16 participants (Actual)Interventional2010-05-31Terminated(stopped due to Withdrawal of support from sponsor)
Randomized Phase III Study for the Treatment of Newly Diagnosed Disseminated Lymphoblastic Lymphoma or Localized Lymphoblastic Lymphoma[NCT00004228]Phase 3393 participants (Actual)Interventional2000-06-30Completed
S0910, A Phase II Study of Epratuzumab (NSC-716711) in Combination With Cytarabine and Clofarabine for Patients With Relapsed or Refractory Ph- Negative Precursor B-Cell Acute Lymphoblastic Leukemia[NCT00945815]Phase 235 participants (Actual)Interventional2010-09-30Completed
A Pilot Pharmacokinetic, Pharmacodynamic and Feasibility Study of Sorafenib in Combination With Cytarabine and Clofarabine in Patients With Refractory or Relapsed Hematologic Malignancies[NCT00908167]Phase 144 participants (Actual)Interventional2009-09-30Completed
Randomized Phase III Trial Evaluating the Role of Autologous Stem Cell Transplantation in Previously Untreated Patients With Stage B and C Chronic Lymphocytic Leukemia[NCT00931645]Phase 3241 participants (Actual)Interventional2001-04-30Completed
A Phase I, Open-label, Multi-centre, Multiple Ascending Dose Study to Assess the Safety and Tolerability of AZD1152 in Combination With Low Dose Cytosine Arabinoside (LDAC) in Patients With Acute Myeloid Leukaemia (AML)[NCT00926731]Phase 14 participants (Actual)Interventional2009-06-30Completed
A Phase I/II Study of the Oral MDM2 Inhibitor DS-3032b (Milademetan) in Combination With Low Dose Cytarabine (LDAC) in Patients With Newly Diagnosed or Relapsed/Refractory Acute Myeloid Leukemia (AML)[NCT03634228]Phase 1/Phase 216 participants (Actual)Interventional2018-12-17Terminated(stopped due to This study was terminated early due to lack of adequate response, and did not move to the Phase II portion of the study)
Phase III, Randomized, Multicenter Study to Assess the Efficacy and Safety of HuM195 (Recombinant Humanized Anti-CD33 Monoclonal Antibody) in Combination With Standardized Chemotherapy Compared to Standardized Chemotherapy Alone in the Treatment of Patien[NCT00006045]Phase 30 participants Interventional2000-03-31Active, not recruiting
A Phase III Trial in Adult Acute Myeloid Leukemia: Daunorubicin Dose-Intensification Prior to Risk-Allocated Autologous Stem Cell Transplantation[NCT00049517]Phase 3657 participants (Actual)Interventional2002-12-19Completed
Phase I Study of Inotuzumab Ozogamicin With 3 and 4 Drug Augmented Berlin-Frankfurt-Münster (BFM) Re-Induction for Patients With Relapsed or Refractory B-cell Acute Lymphoblastic Leukemia (B-ALL)[NCT03962465]Phase 136 participants (Anticipated)Interventional2022-07-22Active, not recruiting
A Phase 1 Dose-Escalation Study of SCH 900776 in Combination With Cytarabine in Subjects With Acute Leukemias (Protocol No. P05247)[NCT00907517]Phase 124 participants (Actual)Interventional2009-07-29Terminated
A Single-Arm, Open-Label Phase 2 Pilot Study of Vyxeos (CPX-351) in Adults With Relapsed or Refractory Acute Lymphoblastic Leukemia[NCT03575325]Phase 211 participants (Actual)Interventional2018-10-11Completed
Genomics-Based Target Therapy for Children With Relapsed or Refractory Malignancy[NCT02638428]Phase 290 participants (Anticipated)Interventional2015-12-31Recruiting
Phase I Study Evaluating the Chemosensitizing Effect of Everolimus Administered With Cytarabine and Daunorubicin in Patients With Acute Myeloid Leukemia in Relapse[NCT00544999]Phase 121 participants (Anticipated)Interventional2007-09-30Recruiting
A Phase I Dose Escalation Study of Ibrutinib With Idarubicin/Cytarabine for Relapsed/Refractory Acute Myeloid Leukemia[NCT02635074]Phase 12 participants (Actual)Interventional2016-11-30Terminated(stopped due to safety)
A Phase II Pilot Trial of Bortezomib (PS-341, Velcade) in Combination With Intensive Re-Induction Therapy for Children With Relapsed Acute Lymphoblastic Leukemia (ALL) and Lymphoblastic Lymphoma (LL)[NCT00873093]Phase 2148 participants (Actual)Interventional2009-03-31Completed
A Phase Ib Feasibility Study of Personalized Kinase Inhibitor Therapy Combined With Induction in Acute Leukemias Who Exhibit In Vitro Kinase Inhibitor Sensitivity[NCT02779283]Phase 17 participants (Actual)Interventional2016-01-13Completed
A Prospective Phase II Trial of Modified MRC UKALL Ⅻ/ECOG E2993 Regimen in the Treatment of Low Risk Philadelphia Chromosome Negative Acute Lymphoblastic Leukemia for Young Adults[NCT02660762]Phase 2100 participants (Anticipated)Interventional2016-01-31Recruiting
A Phase II/III Study of Immunomodulation After High Dose Myeloablative Therapy With Autologous Stem Cell Rescue for Refractory/Relapsed Hodgkin Disease[NCT00070187]Phase 2/Phase 324 participants (Actual)Interventional2003-11-30Completed
Phase 2 Study of Cytarabine in Association With Bendamustine and Rituximab in the Treatment of Relapsed/Refractory Diffuse Large B Cell Lymphoma[NCT02758925]Phase 278 participants (Anticipated)Interventional2016-06-30Not yet recruiting
An Open-Label Phase 2 Trial of LY2181308 Sodium Administered in Combination With Idarubicin and Cytarabine to Patients With Refractory or Relapsed Acute Myeloid Leukemia[NCT00620321]Phase 224 participants (Actual)Interventional2008-03-31Completed
A Pilot Study of Dose Intensification of Methotrexate in Patients With Advanced-Stage (III/IV) Small Non-Cleaved Cell Non-Hodgkins Lymphoma and B-Cell All[NCT00005977]Phase 383 participants (Actual)Interventional2000-09-30Completed
DFCI ALL Adult Consortium Protocol: Adult ALL Trial[NCT01005758]Phase 2180 participants (Anticipated)Interventional2009-01-31Not yet recruiting
Eradicating Measurable Residual Disease in Patients With Acute Myeloid Leukemia (AML) Prior to StEm Cell Transplantation (ERASE): A MyeloMATCH Treatment Trial[NCT05628623]Phase 2184 participants (Anticipated)Interventional2023-10-23Not yet recruiting
Randomization of Cytarabine Monotherapy Versus Standard-of-Care Vinblastine/Prednisone for Frontline Treatment of Langerhans Cell Histiocytosis (TXCH LCH0115)[NCT02670707]Phase 3124 participants (Anticipated)Interventional2016-03-07Recruiting
European Proof-of-Concept Therapeutic Stratification Trial of Molecular Anomalies in Relapsed or Refractory Tumors[NCT02813135]Phase 1/Phase 2460 participants (Anticipated)Interventional2016-08-03Recruiting
A Phase II Study of Combination Daunorubicin and Cytarabine (Ara-c) and Nilotinib (Tasigna) (DATA) in Patients Newly Diagnosed With Acute Myeloid Leukemia and KIT Overexpression[NCT01806571]Phase 234 participants (Actual)Interventional2015-03-12Completed
Phase II Trial of Targeted Immune-Depleting Chemotherapy and Reduced-Intensity Allogeneic Hematopoietic Stem Cell Transplantation Using 8/8 and 7/8 HLA-matched Unrelated Donors and Utilizing Two Graft-versus-Host Disease Prophylaxis Regimens for the Treat[NCT00520130]Phase 1/Phase 292 participants (Actual)Interventional2007-10-30Completed
A Multi-center Prospective Single Arm Clinical Study of Reduced Intensive 3 + 5 Idarubicin and Cytarabine Chemotherapy Plus Venetoclax as First-line Treatment for Adults With Acute Myeloid Leukaemia and High-risk Myelodysplastic Syndrome[NCT06050941]Phase 260 participants (Anticipated)Interventional2023-10-20Not yet recruiting
A Dose Escalation And Phase II Study Of Gemtuzumab Ozogamicin (CMA-676; Mylotarg) With High-Dose Cytarabine For Patients With Refractory Or Relapsed Acute Myeloid Leukemia (AML)[NCT00006265]Phase 260 participants (Actual)Interventional2001-03-31Completed
Clinical Evaluation of ASTX727 in Combination With Venetoclax All-Oral Therapy vs Standard of Care Cytarabine and Anthracycline Induction Chemotherapy for Younger FLT3WT Patients With ELN High- Risk Acute Myeloid Leukemia[NCT04817241]Phase 1/Phase 255 participants (Anticipated)Interventional2022-02-10Active, not recruiting
A Phase 3 Trial Investigating Blinatumomab (NSC# 765986) in Combination With Chemotherapy in Patients With Newly Diagnosed Standard Risk or Down Syndrome B-Lymphoblastic Leukemia (B-ALL) and the Treatment of Patients With Localized B-Lymphoblastic Lymphom[NCT03914625]Phase 36,720 participants (Anticipated)Interventional2019-07-03Recruiting
A Phase II Study of Modified VR-CAP and Acalabrutinib as First Line Therapy for Transplant-Eligible Patients With Mantle Cell Lymphoma[NCT04626791]Phase 245 participants (Anticipated)Interventional2021-08-03Recruiting
A Phase I/II Study of AMD3100 With Mitoxantrone, Etoposide and Cytarabine (AMD3100+MEC) in Relapsed or Refractory AML[NCT00512252]Phase 1/Phase 252 participants (Actual)Interventional2007-07-31Completed
A Phase II Trial of Cytarabine and Clofarabine in Patients With Relapsed or Refractory Acute Lymphoblastic Leukemia (ALL)[NCT00337168]Phase 236 participants (Actual)Interventional2006-10-31Completed
Dose Densified Chemoimmunotherapy With Early CNS Prophylaxis in Patients Less Than 65 Years With High Risk (aaIPI≥ 2) Diffuse Large B-Cell Lymphoma[NCT01325194]Phase 2143 participants (Actual)Interventional2011-03-31Completed
CD-34 Selection for Ex-vivo T-Cell Depletion of Mobilized Peripheral Blood Stem Cells for Recipients of HLA Haploidentical Related Donor Stem Cell Grafts Receiving Intensive Conditioning[NCT00368355]Phase 246 participants (Actual)Interventional2000-04-30Completed
Phase II Study of Maintenance Therapy With Decitabine (NSC #127716) Following Standard Induction and Cytogenetic Risk-Adapted Intensification in Previously Untreated Patients With AML < 60 Years[NCT00416598]Phase 2546 participants (Actual)Interventional2006-11-15Completed
A Randomized Double-Blind Phase III Study of Ibrutinib During and Following Autologous Stem Cell Transplantation Versus Placebo in Patients With Relapsed or Refractory Diffuse Large B-Cell Lymphoma of the Activated B-Cell Subtype[NCT02443077]Phase 3302 participants (Anticipated)Interventional2016-10-12Active, not recruiting
A Phase I Study of Liposomal Cytarabine and Daunorubicin (CPX-351) in Combination With Gemtuzumab Ozogamicin (GO) in Relapsed Refractory Pediatric Patients With Acute Myeloid Leukemia (AML)[NCT04915612]Phase 118 participants (Anticipated)Interventional2021-05-21Recruiting
Phase 2 Clinical Trial for Comprehensive Treatment Program for Patients With Blastic Plasmacytoid Dendritic Cell Neoplasm (BPDCN): Tagraxofusp (SL-401) in Combination With HCVAD/Mini-CVD and VENETOCLAX[NCT04216524]Phase 240 participants (Anticipated)Interventional2020-05-29Recruiting
Phase 1 Dose Escalation Study of CPX-351 for Patients With Int-2 or High Risk IPSS Myelodysplastic Syndromes and Chronic Myelomonocytic Leukemia After Failure to Hypomethylating Agents[NCT03896269]Phase 138 participants (Anticipated)Interventional2019-05-14Recruiting
A Pilot Study of Liposomal Cytarabine and Daunorubicin (CPX-351) in Combination With Gemtuzumab Ozogamicin (GO) in Relapsed Refractory Patients With Acute Myeloid Leukemia (AML) and Post-Hypomethylating Agent (Post-HMA) Failure High-Risk Myelodysplastic S[NCT03672539]Phase 250 participants (Anticipated)Interventional2018-11-07Recruiting
Phase II Study of CPX-351 in Combination With Venetoclax in Patients With Acute Myeloid Leukemia (AML)[NCT03629171]Phase 252 participants (Anticipated)Interventional2018-10-29Recruiting
Risk Adapted Treatment of Newly Diagnosed Childhood Acute Promyelocytic Leukemia (APL) Using Arsenic Trioxide (Trisenox® ) During Consolidation[NCT00866918]Phase 3106 participants (Actual)Interventional2009-03-09Completed
A Phase I and Dose Expansion Cohort Study of Panobinostat in Combination With Fludarabine and Cytarabine in Pediatric Patients With Refractory or Relapsed Acute Myeloid Leukemia or Myelodysplastic Syndrome[NCT02676323]Phase 119 participants (Actual)Interventional2016-05-03Terminated(stopped due to Slow accrual)
Phase 1B Study of Venetoclax in Combination With Standard Intensive Chemotherapy With Daunorubicin Plus Cytarabine Followed by High-Dose Cytarabine in Adult Patients With Newly Diagnosed Acute Myeloid Leukemia and Advanced Myelodysplastic Syndrome[NCT05342584]Phase 199 participants (Anticipated)Interventional2022-05-25Recruiting
Zevalin/BEAM/Rituximab vs BEAM/Rituximab With or Without Rituximab Maintenance in Autologous Stem Cell Transplantation for Diffuse Large B-Cell Lymphomas[NCT00591630]Phase 230 participants (Actual)Interventional2007-11-14Completed
A Pilot Study of Combined Immunochemotherapy Followed by Reduced Dose RT for Patients With Newly Diagnosed Primary Central Nervous System Lymphoma[NCT00594815]52 participants (Actual)Interventional2002-08-28Completed
A Randomized Trial for Patients With High-Grade Myeloid Neoplasms With Measurable Residual Disease (MRD): CPX-351 vs. Immediate Allogeneic Hematopoietic Cell Transplantation[NCT04526288]Phase 21 participants (Actual)Interventional2021-08-09Terminated(stopped due to Terminated due to low accrual)
A Phase 2, Randomized, Open-Label, Multicenter Study of Ficlatuzumab in Combination With High-Dose Cytarabine (HiDAC) and HiDAC Alone in Adults With Relapsed or Refractory Acute Myeloid Leukemia (AML)[NCT04100330]Phase 20 participants (Actual)Interventional2020-01-31Withdrawn(stopped due to Urgent shift among clinical sites toward efforts to combat COVID-19 pandemic;impacted feasibility of completing study within shelf-life of current IP supply)
Ph 1b Dose Escalation Study of OXi4503 as a Single Agent and in Combination With Cytarabine With Subsequent Phase 2 Cohorts for Subjects With Relapsed/Refractory Acute AML and MDS[NCT02576301]Phase 1/Phase 2105 participants (Anticipated)Interventional2015-10-31Recruiting
Phase II Study of Low Intensity Allogeneic Transplantation in Mantle Cell Lymphoma[NCT00720447]Phase 225 participants (Anticipated)Interventional2008-11-30Not yet recruiting
Dose-Intensive Chemotherapy Combined With Monoclonal Antibody Therapy and Targeted Radioimmunotherapy for Untreated Patients With High-Risk B-Cell Non-Hodgkin's Lymphoma[NCT00577629]Phase 239 participants (Actual)Interventional2005-06-18Completed
A Randomized, Multi-center Phase II Trial to Assess the Efficacy of 5-azacytidine Added to Standard Primary Therapy in Elderly Patients With Newly Diagnosed AML[NCT00915252]Phase 2214 participants (Actual)Interventional2009-07-31Completed
Phase II Trial of Gleevec and Low-Dose Ara-C for Elderly Patients With C-Kit Positive Acute Myeloid Leukemia and High-Risk Myelodysplastic Syndromes[NCT00451997]Phase 210 participants (Actual)Interventional2004-03-31Completed
High Dose Cytarabine And Mitoxantrone Therapy For Relapsed And Refractory Acute Myeloid And Lymphocytic Leukemia: Effects Of GM-CSF Versus G-CSF On Dendritic Cells And Leukemia Associated Antigen-Specific T-Lymphocytes[NCT00053131]Phase 20 participants Interventional1999-01-31Completed
Randomized Comparison of Consolidation Treatment in Elderly Patients With Acute Myeloid Leukemia: Idarubicin (IDA) Combined With Intermediate-dose Cytarabine Versus Intermediate-dose Cytarabine Alone[NCT04216771]Phase 2/Phase 3320 participants (Anticipated)Interventional2020-01-31Recruiting
A Randomized Trial of the I-BFM-SG for the Management of Childhood Non-B Acute Lymphoblastic Leukemia[NCT00764907]Phase 34,000 participants (Anticipated)Interventional2002-11-30Recruiting
Randomized Multicenter Treatment Optimization Study In Chronic Myeloid Leukemia (CML) Interferon-a Vs. Allogeneic Stem Cell Transplantation Vs. High-Dose Chemotherapy Followed By Autografting And Interferon-a Maintainance In Early Chronic Phase[NCT00025402]Phase 31,000 participants (Anticipated)Interventional1997-07-31Active, not recruiting
Dasatinib in Combination With Chemotherapy for Relapsed or Refractory Core Binding Factor Acute Myeloid Leukemia: A Phase I Study (AflacLL1401)[NCT02680951]Phase 10 participants (Actual)Interventional2015-12-31Withdrawn(stopped due to Withdrawn due to lack of participants.)
A Phase II Study of CPX-351 as a Novel Therapeutic Approach for Patients With Myelodysplastic Syndromes (MDS) After Hypomethylating Agent Failure[NCT03957876]Phase 225 participants (Anticipated)Interventional2019-07-25Recruiting
The Efficiency and Safety of N-acetylcysteine for Prevention of Thrombotic Events After Allogenic Hematopoietic Stem Cell Transplantation[NCT05907486]Phase 3260 participants (Anticipated)Interventional2023-08-01Not yet recruiting
Phase III Randomized Trial of Autologous and Allogeneic Stem Cell Transplantation Versus Intensive Conventional Chemotherapy in Acute Lymphoblastic Leukemia in First Remission[NCT00002514]Phase 31,929 participants (Actual)Interventional1993-05-07Completed
Phase II Study Of Bryostatin 1 (NSC 339555) And High-Dose 1-B-D-Arabinofuranosylcytosine (HiDAC) In Patients With Refractory Leukemia[NCT00017342]Phase 20 participants Interventional2001-07-31Completed
Phase I/II Study of Hyper-CVAD Plus RAD001 (Everolimus) for Patients With Relapsed or Refractory Acute Lymphocytic Leukemia (ALL)[NCT00968253]Phase 1/Phase 224 participants (Actual)Interventional2009-11-30Completed
A Single Arm Pilot Study of Intrathecally Administered DepoCyt® With Systemic Sorafenib in the Treatment of Neoplastic Meningitis From Solid Tumors[NCT00964743]2 participants (Actual)Interventional2009-08-31Terminated(stopped due to Low Accrual)
A Phase 1b, Open-label Study of LY2510924, Idarubicin and Cytarabine in Patients With Relapsed or Refractory Acute Myeloid Leukemia[NCT02652871]Phase 136 participants (Anticipated)Interventional2016-05-09Completed
Dose-intense Idarubicin Induction in Young Patients With Acute Myeloid Leukemia[NCT04069208]Phase 242 participants (Anticipated)Interventional2019-09-03Recruiting
PETHEMA LAL-RI/96: Treatment for Patients With Standard Risk Acute Lymphoblastic Leukemia[NCT00494897]Phase 4374 participants (Actual)Interventional1996-06-30Completed
A Phase II Study of the Impact of Clinicogenetic Risk-Stratified Management on Outcomes of Acute Myeloid Leukemia in Older Patients[NCT03226418]Phase 275 participants (Actual)Interventional2017-07-07Active, not recruiting
The Clinical Application of Tumor Reversion: A Phase I Study of Sertraline (Zoloft) in Combination With Timed-sequential Cytosine Arabinoside (Ara-C) in Adults With Relapsed and Refractory Acute Myeloid Leukemia (AML)[NCT02891278]Phase 16 participants (Actual)Interventional2016-08-11Completed
Phase II Trial of the Addition of PEG-Asparaginase to the Hyper-CVAD Regimen in Adult Newly-Diagnosed Acute Lymphoblastic Leukemia[NCT01005914]Phase 211 participants (Actual)Interventional2009-06-30Terminated(stopped due to Increased rate of bacterial infections)
Risk-Stratified Therapy for Acute Myeloid Leukemia in Down Syndrome[NCT02521493]Phase 3312 participants (Anticipated)Interventional2015-12-23Active, not recruiting
A Randomized Phase III Study of Intensive Consolidation With High Dose Cytosine Arabinoside in Acute Myelogenous Leukemia (AML-8B)[NCT01324063]Phase 3160 participants (Anticipated)Interventional1986-11-30Completed
A Phase III Randomized, Double-Blind Study of Induction (Daunorubicin/Cytarabine) and Consolidation (High-Dose Cytarabine) Chemotherapy + Midostaurin (PKC412) (IND #101261) or Placebo in Newly Diagnosed Patients < 60 Years of Age With FLT3 Mutated Acute M[NCT00651261]Phase 3717 participants (Actual)Interventional2008-04-30Active, not recruiting
Phase II Clinical Evaluation of Vorinostat Combined With Salvage Reinduction Chemotherapy Including Gemtuzumab Ozogamicin, Idarubicin and Cytarabine and Vorinostat Maintenance in Relapse or Refractory Acute Myeloid Leukemia Patients With 50 Years or Older[NCT01039363]Phase 227 participants (Anticipated)InterventionalNot yet recruiting
A Multicenter and Randomized Prospective Study for Improving the Outcome of Childhood Acute Promyeloid Leukemia[NCT02200978]Phase 4176 participants (Actual)Interventional2011-09-30Completed
Phase I/II Study of Sensitization of Non-M3 Acute Myeloid Leukemia (AML) Blasts to All-trans Retinoic Acid (ATRA) by Epigenetic Treatment With Tranylcypromine (TCP), an Inhibitor of the Histone Lysine Demethylase 1 (LSD1)[NCT02717884]Phase 1/Phase 260 participants (Anticipated)Interventional2015-05-31Recruiting
Treatment of AML in Adults 50 to 70 Years, Study of Two Anthracyclines and the Interest of Maintenance Treatment With Interleukin 26- ALFA 9801[NCT00931138]Phase 3420 participants Interventional1999-12-31Completed
A Phase 1 Study Of Palbociclib (IND#141416), A CDK 4/6 Inhibitor, In Combination With Chemotherapy In Children With Relapsed Acute Lymphoblastic Leukemia (ALL) Or Lymphoblastic Lymphoma (LL)[NCT03792256]Phase 115 participants (Anticipated)Interventional2019-04-11Active, not recruiting
A Phase I/II Study of CLOLAR® (Clofarabine, IND# 73, 789) in Combination With Cytarabine in Pediatric Patients With Refractory/Relapsed Leukemia[NCT00372619]Phase 1/Phase 274 participants (Actual)Interventional2007-03-31Completed
Clofarabine, Idarubicin, and Cytarabine Combination as Induction Therapy for Younger Patients With Acute Myeloid Leukemia (AML)[NCT01025154]Phase 263 participants (Actual)Interventional2010-01-31Completed
A RANDOMIZED (1:1), DOUBLE-BLIND, MULTI-CENTER, PLACEBO CONTROLLED STUDY EVALUATING INTENSIVE CHEMOTHERAPY WITH OR WITHOUT GLASDEGIB (PF-04449913) OR AZACITIDINE (AZA) WITH OR WITHOUT GLASDEGIB IN PATIENTS WITH PREVIOUSLY UNTREATED ACUTE MYELOID LEUKEMIA[NCT03416179]Phase 3730 participants (Actual)Interventional2018-04-20Completed
A Phase II Study of Metronomic and Targeted Anti-angiogenesis Therapy for Children With Recurrent/Progressive Medulloblastoma, Ependymoma and ATRT[NCT01356290]Phase 2100 participants (Anticipated)Interventional2014-04-30Recruiting
"A Phase I Trial of Volasertib (BI 6727), an Intravenous Polo-Like Kinase Inhibitor, in Combination With 7+3 Induction Chemotherapy for Patients With Acute Myeloid Leukemia"[NCT02905994]Phase 10 participants (Actual)Interventional2016-09-30Withdrawn(stopped due to Funding Withdrawn)
Prospective Multicentre Phase II Study to Evaluate the Combination of Rituximab and DepoCyte® by Intrathecal Injection in the C5R Chemotherapy Protocol in Patients Between the Ages of 18 and 60 Years With Primary Cerebral Non-Hodgkin Lymphoma and Systemic[NCT00553943]Phase 260 participants (Actual)Interventional2007-07-31Completed
The Prospective Study of FTD Program and HD-MTX-Ara-C Program Contrast in the Treatment of PCNSL Lymphoma.[NCT05274139]Phase 220 participants (Actual)Interventional2017-03-02Completed
Impact of Treatment Intensity on Survival, Quality of Life, and Resource Utilization in Medically Less Fit Adults With Acute Myeloid Leukemia and Analogous Myeloid Neoplasms: A Randomized Pilot Study[NCT03012672]Phase 250 participants (Actual)Interventional2016-12-30Completed
Phase I Study of Escalating Doses of Carfilzomib With Hyper-CVAD in Patients With Newly Diagnosed Acute Lymphoblastic Leukemia/Lymphoma[NCT02293109]Phase 110 participants (Actual)Interventional2015-12-17Completed
A Phase I/II Study of Lintuzumab-Ac225 in Older Patients With Untreated Acute Myeloid Leukemia[NCT02575963]Phase 1/Phase 240 participants (Actual)Interventional2012-10-31Completed
Phase II Randomized Study of Rituximab, Methotrexate, Procarbazine, Vincristine, and Cytarabine With and Without Low-Dose Whole-Brain Radiotherapy for Primary Central Nervous System Lymphoma[NCT01399372]Phase 291 participants (Actual)Interventional2011-09-30Completed
Open-Label, Multicenter Phase Ib/IIa Study For the Evaluation of Dasatinib (Sprycel™) Following Induction and Consolida-tion Therapy as Well as in Maintenance Therapy in Patients With Newly Diagnosed Core Binding Factor (CBF) Acute Myeloid Leukemia (AML)[NCT00850382]Phase 1/Phase 289 participants (Actual)Interventional2009-06-30Completed
A Phase 3, Open Label, Single Arm, Multi-Center Study to Evaluate the Efficacy and Safety of Decitabine Combined With HAAG Regimen in Elderly Newly Diagnosed Acute Myeloid Leukemia Patients.[NCT04083911]Phase 350 participants (Anticipated)Interventional2019-04-01Recruiting
HLA-Mismatched Allogeneic Cellular Therapy (Microtransplantation) After Chemotherapy in Patients With Intermediate-Risk Acute Myeloid Leukemia < 60 Years[NCT02587871]Phase 20 participants (Actual)Interventional2018-12-12Withdrawn(stopped due to PI decided not to pursue study)
Study Characterizing the Impact of Different Therapeutic Strategies on Event Occurrence at 2 Years, 5 Years, 10 Years, and 15 Years, According to Prognostic Groups in Patients With Hodgkin Lymphoma[NCT00920153]Phase 3442 participants (Actual)Interventional2008-05-31Terminated(stopped due to Other new drugs)
Acalabrutinib Plus RICE for Patients With Relapsed/Refractory DLBCL Followed by Autologous Hematopoietic Cell Transplantation and Acalabrutinib Maintenance Therapy[NCT03736616]Phase 247 participants (Anticipated)Interventional2019-08-16Recruiting
Loncastuximab Tesirine in Combination With BEAM (Carmustine, Etoposide, Ara-C, Melphalan) Conditioning Regimen Prior to Autologous Stem Cell Transplant (ASCT) and for Maintenance Therapy in Diffuse Large B-Cell Lymphoma (DLBCL)[NCT05228249]Phase 10 participants (Actual)Interventional2023-04-30Withdrawn(stopped due to PI left institution and funding sponsor closed study. Study did not open to accrual, and no participants were enrolled.)
Phase II Study of The Modified Hyper-CVAD Program for Acute Lymphoblastic Leukemia[NCT00671658]Phase 2220 participants (Actual)Interventional2002-11-30Completed
A Pilot Study of Intravenous EZN-2285 (SC-PEG E. Coli L-asparaginase) or Intravenous Oncaspar® in the Treatment of Patients With High-Risk Acute Lymphoblastic Leukemia (ALL)[NCT00671034]Phase 3166 participants (Actual)Interventional2008-07-21Completed
A Phase II Pilot Study of Bortezomib (PS-341, Velcade) Combined With Reinduction Chemotherapy in Children and Young Adults With Recurrent, Refractory or Secondary Acute Myeloid Leukemia[NCT00666588]Phase 252 participants (Actual)Interventional2008-04-30Completed
A Phase 1 Trial of Indoximod in Combination With Idarubicin and Cytarabine in Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)[NCT02835729]Phase 154 participants (Actual)Interventional2016-07-31Completed
A Programme of Treatment Development for Older Patients With Acute Myeloid Leukemia and High Risk Myelodysplastic Syndrome[NCT00454480]Phase 2/Phase 32,000 participants (Anticipated)Interventional2006-08-31Completed
A Randomized Study on CNS Prophylaxis With Liposome-Encapsulated Cytarabine in Association With a Lineage-Targeted and MRD-Oriented Postremission Strategy in Adult ALL[NCT00795756]Phase 2/Phase 3145 participants (Actual)Interventional2008-01-31Completed
Clofarabine Plus Low-Dose Cytarabine Induction Followed by Consolidation of Clofarabine Plus Low-Dose Cytarabine Alternating With Decitabine in Frontline Acute Myeloid Leukemia (AML) and High-Risk Myelodysplastic Syndrome (MDS)[NCT00778375]Phase 2122 participants (Actual)Interventional2008-10-31Completed
Venetoclax in Combination With Homoharringtonine and Cytarabine in Newly Diagnosed Subjects With Acute Myeloid Leukemia: a Phase 2/3, the Single-arm, Open-label, Monocentric Study[NCT05805098]Phase 2/Phase 360 participants (Anticipated)Interventional2023-03-01Recruiting
A Phase II Study of Cladribine and Low Dose Cytarabine in Combination With Venetoclax, Alternating With Azacitidine and Venetoclax, in Patients With Higher-risk Myeloproliferative Chronic Myelomonocytic Leukemia or Higher-risk Myelodysplastic Syndromes Wi[NCT05365035]Phase 260 participants (Anticipated)Interventional2022-09-23Recruiting
Phase III Randomised Study on Liposomal Cytarabine (DepoCyte®) vs. Intrathecal Triple for CNS-Treatment During Maintenance Therapy in High-Risk Acute Lymphoblastic Leukemia Patients in NOPHO ALL 2008 Treatment Protocol[NCT00991744]Phase 3100 participants (Anticipated)Interventional2009-01-31Suspended(stopped due to Sterility problems in DepoCyte production)
A Phase III Study, Randomized, to Evaluate the Reduction of Chemotherapy Intensity in Association With Nilotinib (Tasigna®) in Philadelphia Chromosome-positive (Ph+) ALL of Young Adults (18-59 Years Old) (GRAAPH-2014)[NCT02611492]Phase 3265 participants (Anticipated)Interventional2016-04-30Recruiting
Safety and Efficacy Study of Busulfan/FLAG Conditioning Regimen in Patients With Relapsed/Refractory Acute Leukemia Undergoing Allogeneic Peripheral Blood Stem Cell Transplantation[NCT02784561]Phase 480 participants (Anticipated)Interventional2016-07-31Not yet recruiting
Treatment of Elderly Patients (>60 Years) With Acute Myeloblastic Leukemia or Advanced MDS (RAEB-T): An Open Randomized Study to Test the Efficacy of G-CSF-Priming and a Feasibility Trial of Dose-Reduced Allogeneic Transplantation and of Autologous Stem C[NCT00199147]Phase 4250 participants Interventional2000-01-31Recruiting
High-Dose Ara-C Followed by Continuous Infusion Interleukin-2 for Acute Myelogenous Leukemia in First Remission[NCT00136448]Phase 230 participants Interventional1993-02-28Completed
A Phase I Study of M3814 in Combination With MEC in Patients With Relapsed or Refractory Acute Myeloid Leukemia[NCT03983824]Phase 148 participants (Anticipated)Interventional2020-05-05Recruiting
An Open-label, Multicenter, Phase 2 Study Evaluating the Efficacy and Safety of Daratumumab in Pediatric and Young Adult Subjects >=1 and <=30 Years of Age With Relapsed/Refractory Precursor B-cell or T-cell Acute Lymphoblastic Leukemia or Lymphoblastic L[NCT03384654]Phase 247 participants (Actual)Interventional2018-05-14Completed
Prospectively Randomized Phase III Study of an Individualized Sensitivity-Directed Combination Chemotherapy Versus DTIC as First-Line Treatment in Stage IV Metastatic Melanoma[NCT00779714]Phase 3360 participants (Anticipated)Interventional2008-10-31Recruiting
Immunochemotherapy With Rituximab-Bendamustine-Cytarabine for Patients With Mantle Cell Lymphoma Not Eligible for Intensive Regimens or Autologous Transplantation.[NCT00992134]Phase 241 participants (Actual)Interventional2009-06-30Completed
A Prospective Single Institution Pilot Study Evaluating the Pharmacokinetics of Sirolimus in Combination With MEC (Mitoxantrone + Etoposide + Cytarabine) in Patients With High Risk Leukemias[NCT00780104]Phase 116 participants (Actual)Interventional2007-07-31Completed
Treatment of Acute Myelogenous Leukemia With the Histone Deacetylase Inhibitor Valproic Cid in Combination With All-trans Retinoic Acid (ATRA) and Low Dose Cytarabine[NCT00995332]Phase 1/Phase 236 participants (Actual)Interventional2009-09-30Completed
Phase I/II Study of Plitidepsin (Aplidin®) in Combination With Cytarabine in Patients With Relapsed/Refractory Leukemia[NCT00780143]Phase 1/Phase 23 participants (Actual)Interventional2007-11-30Terminated(stopped due to Poor recruitment)
Multicenter Trial for Treatment of Acute Lymphoblastic Leukemia in Adults (05/93)[NCT00199069]Phase 4720 participants Interventional1993-04-30Completed
Intensified Tyrosine Kinase Inhibitor Therapy (Dasatinib NSC# 732517) in Philadelphia Chromosome Positive Acute Lymphoblastic Leukemia (ALL)[NCT00720109]Phase 2/Phase 363 participants (Actual)Interventional2008-07-14Completed
Phase I Study of Temozolomide and Intrathecal DepoCyt in Patients With Neoplastic Meningitis[NCT00515788]Phase 111 participants (Actual)Interventional2006-02-28Terminated(stopped due to Study terminated due to slow accrual with no expansion to additional phase.)
A Phase II Study of GM-CSF (Sargramostim) and Rituximab Following Autologous Transplantation For Relapsed Follicular Lymphoma[NCT00521014]Phase 214 participants (Actual)Interventional2007-10-31Completed
UARK 2006-15: A Phase III Randomized Study of Tandem Transplants With or Without Bortezomib (Velcade) and Thalidomide (Thalomid) to Evaluate Its Effect on Response Rate and Durability of Response in Multiple Myeloma Patients[NCT00574080]Phase 320 participants (Actual)Interventional2006-07-31Terminated(stopped due to low accrual)
A Randomized Trial for Patients With Acute Myeloid Leukemia or High Risk Myelodysplatic Syndrome Aged 60 or Over[NCT00005823]Phase 32,000 participants (Anticipated)Interventional1998-12-31Completed
Treatment of High Risk Adult Acute Lymphoblastic Leukemia[NCT00853008]Phase 4100 participants (Anticipated)Interventional2003-01-31Completed
Primed Peripheral Blood Stem Cell Autologous Transplantation for Lymphoma and Hodgkin's Disease[NCT00005985]Phase 2213 participants (Actual)Interventional2000-08-31Completed
Autologous Transplantation for Non-Hodgkin's Lymphoma and Hodgkin's Disease Using Retrovirally Marked Peripheral Blood Progenitor Cells Obtained After In Vivo Mobilization Using Hematopoietic Cytokines Plus Chemotherapy[NCT00005998]Phase 20 participants (Actual)Interventional2000-01-31Withdrawn(stopped due to Withdrawn because study never enrolled patients)
Allogeneic Stem Cell Transplantation for Mantle Cell Lymphoma[NCT00006747]Phase 24 participants (Actual)Interventional2000-11-30Completed
A Phase III Study of STI571 Versus Interferon-a (IFN-a) Combined With Cytarabine (Ara-C) in Patients With Newly Diagnosed Previously Untreated Philadelphia Chromosome Positive (Ph+) Chronic Myelogenous Leukemia in Chronic Phase (CML-CP)[NCT00006343]Phase 30 participants Interventional2000-06-30Completed
Autologous Peripheral Blood Stem Cell Mobilization and Transplantation for Myelofibrosis[NCT00006367]Phase 20 participants Interventional2000-05-31Completed
The Clinical Observationg on HAM for Acute Myeloid Leukemia[NCT04024241]250 participants (Anticipated)Observational2017-09-01Recruiting
Phase II Study of Cladribine Plus Low Dose Cytarabine (LDAC) Induction Followed By Consolidation With Cladribine Plus LDAC Alternating With Decitabine in Patients With Untreated Acute Myeloid Leukemia (AML) or High-Risk Myelodysplastic Syndrome (MDS)[NCT01515527]Phase 2160 participants (Anticipated)Interventional2012-02-07Recruiting
Phase I/II Study of Decitabine (DAC) Followed by Clofarabine, Idarubicin, and Cytarabine (CIA) in Acute Leukemia[NCT01794702]Phase 1/Phase 265 participants (Actual)Interventional2013-02-20Completed
Prospective , Multicentric, Randomized Phase II Study, Evaluating the Role of Cranial Radiotherapy or Intensive Chemotherapy With Hematopoietic Stem Cell Rescue After Conventional Chemotherapy for Primary Central Nervous System in Young Patients (< 60 y)[NCT00863460]Phase 2140 participants (Actual)Interventional2008-10-03Active, not recruiting
LSA5 PROTOCOL FOR THE TREATMENT OF ADVANCED PEDIATRIC AND ADOLESCENT NON-HODGKIN'S LYMPHOMA (NHL)[NCT00002691]Phase 20 participants Interventional1995-08-31Completed
Escalating Dose Intravenous Methotrexate Without Leucovorin Rescue Versus Oral Methotrexate and Single Versus Double Delayed Intensification for Children With Standard Risk Acute Lymphoblastic Leukemia[NCT00005945]Phase 33,054 participants (Actual)Interventional2000-06-30Completed
Dose Escalation and Phase II Study of Bortezomib (IND #58443) Added to Standard Daunorubicin and Cytarabine Therapy for Patients With Previously Untreated Acute Myeloid Leukemia Age 60-75 Years[NCT00742625]Phase 1/Phase 295 participants (Actual)Interventional2008-09-30Completed
Intensification Therapy of Mature B-ALL, Burkitt and Burkitt Like and Other High Grade Non-Hodgkin's Lymphoma in Adults[NCT00797810]Phase 425 participants (Anticipated)Interventional2006-12-31Recruiting
AML2003 - Randomized Comparison Between Standard-Therapy and Intensified Therapy for Adult Acute Myeloid Leukemia Patients <= 60 Years. A Prospective, Randomized, Multi-center Therapy-Optimizing-Study.[NCT00180102]Phase 4600 participants (Anticipated)Interventional2003-12-31Completed
A Phase III Study of Daunorubicin and Cytarabine +/- G3139 (Genasense, Oblimersen Sodium, NSC #683428, IND #58842), a BCL2 Antisense Oligodeoxynucleotide, in Previously Untreated Patients With Acute Myeloid Leukemia (AML) > / = 60 Years[NCT00085124]Phase 3500 participants (Actual)Interventional2003-12-31Completed
Treatment of Older Adults With Acute Lymphoblastic Leukemia[NCT00973752]Phase 230 participants (Actual)Interventional2009-08-31Completed
A Phase 3, Randomized, Controlled, Multi-Center Study to Evaluate the Efficacy and Safety of Decitabine Combined With HAAG Regimen in Newly Diagnosed Acute Myeloid Leukemia Patients Younger Than 60 Years[NCT04087967]Phase 3162 participants (Anticipated)Interventional2019-04-01Recruiting
Open Label, Multicenter, Dose Escalation Phase 1a/b Study of RO5429083, Administered as Intravenous Infusion Alone or in Combination With Cytarabine in Patients With Acute Myelogenous Leukemia (AML).[NCT01641250]Phase 144 participants (Actual)Interventional2012-08-31Completed
A Study of Bortezomib With Chemotherapy for Relapsed/Refractory Acute Lymphoblastic Leukemia[NCT00440726]Phase 1/Phase 231 participants (Actual)Interventional2006-08-04Completed
Efficacy and Safety of Cladribine in Combination With G-CSF,Low-dose Cytarabine and Aclarubicin in Newly Diagnosed Unfit Patients With Acute Myeloid Leukemia[NCT04254640]Phase 234 participants (Anticipated)Interventional2021-03-01Not yet recruiting
Bortezomib (Velcade®) and Reduced-Intensity Allogeneic Stem Cell Transplantation for Patients With Lymphoid Malignancies[NCT00439556]Phase 240 participants (Actual)Interventional2007-02-13Completed
A Phase II Study of the Efficacy of the HCVIDDOXIL Regimen in Patients With Newly Diagnosed Peripheral T Cell Lymphoma[NCT00290433]Phase 255 participants (Actual)Interventional2003-09-30Completed
A Multicenter Phase II Study Incorporating DOXIL® and Rituximab Into the Magrath Regimen for HIV-Negative and HIV-Positive Patients With Newly Diagnosed Burkitt's and Burkitt-like Lymphoma[NCT00392990]Phase 225 participants (Actual)Interventional2007-02-06Completed
Phase II Study of Fludarabine, Cytarabine (ARA-C) and Erwinase IV in Patients With Relapsed or Refractory Hematologic Malignancies[NCT02718755]Phase 20 participants (Actual)Interventional2018-05-31Withdrawn(stopped due to Problem with drug supply)
A Collaborative Trial for the Treatment of Patients With Newly Diagnosed Acute Myeloid Leukemia or Myelodysplasia[NCT00136084]Phase 3238 participants (Actual)Interventional2002-08-31Completed
Phase Ib Study of Fludarabine, Cytarabine (Ara-C) and Pegylated Erwinase (Pegcrisantaspase) in Patients With Relapsed or Refractory Leukemia[NCT04526795]Phase 162 participants (Anticipated)Interventional2021-04-09Recruiting
A Phase 1/2, Multicenter, Open-Label, Single Arm, Dose Escalation and Expansion Study of Gilteritinib (ASP2215) Combined With Chemotherapy in Children, Adolescents and Young Adults With FMS-like Tyrosine Kinase 3 (FLT3)/Internal Tandem Duplication (ITD) P[NCT04240002]Phase 1/Phase 297 participants (Anticipated)Interventional2020-09-04Recruiting
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
An Open-Label, Randomized Study of Low-Dose Cytarabine in Combination With Arsenic Trioxide Compared With Low-Dose Cytarabine Alone for the Treatment of Elderly Patients With Acute Myeloid Leukemia[NCT00513305]Phase 367 participants (Actual)Interventional2007-10-31Terminated(stopped due to Study has been stopped by sponsor decision)
Intensive Treatment for Intermediate-Risk Relapse of Childhood B-precursor Acute Lymphoblastic Leukemia (ALL): A Randomized Trial of Vincristine Strategies[NCT00381680]Phase 3275 participants (Actual)Interventional2007-03-31Completed
A Phase III Study of STI571 Versus Interferon-α (IFN-α) Combined With Cytarabine (Ara-C) in Patients With Newly Diagnosed Previously Untreated Philadelphia Chromosome Positive (Ph+) Chronic Myelogenous Leukemia in Chronic Phase (CML-CP)[NCT00333840]Phase 31,106 participants (Actual)Interventional2000-06-30Completed
A Phase IIa Pharmacokinetic and Efficacy Study of Amonafide L-malate (AS1413) in Combination With Cytarabine in Adult Patients With Acute Myeloid Leukemia (AML)[NCT01066494]Phase 220 participants (Anticipated)Interventional2010-01-31Active, not recruiting
A Randomized Multicenter Phase III Study to Evaluate the Role of All-trans Retinoic Acid (ATRA) in Combination With Induction Chemotherapy, or Azacitidine and Idarubicin as Salvage Therapy and Idarubicin With Cytarabine or Azacitidine as Maintenance Thera[NCT01067274]Phase 30 participants (Actual)Interventional2010-04-30Withdrawn(stopped due to Study abandoned)
A Phase III Randomized, Double-blind, Controlled Study Comparing Clofarabine and Cytarabine Versus Cytarabine Alone in Adult Patients 55 Years and Older With Acute Myelogenous Leukemia (AML) Who Have Relapsed or Are Refractory After Receiving up to Two Pr[NCT00317642]Phase 3326 participants (Actual)Interventional2006-08-31Completed
A Phase Ib Study of Belinostat With RDHAP Chemotherapy (Dexamethasone, Cytarabine, Cisplatinum) in Adults With Relapsed or Refractory Diffuse Large B-cell Lymphoma[NCT02532192]Phase 10 participants (Actual)Interventional2015-12-31Withdrawn(stopped due to Withdrawal of study support.)
A Pilot Study of Lestaurtinib (CEP-701) in Combination With Chemotherapy in Young Patients With Relapsed or Refractory FLT3-mutant Acute Myeloid Leukemia[NCT00469859]Phase 1/Phase 214 participants (Actual)Interventional2007-06-30Completed
A Phase II Study of High-Dose Immunosuppressive Therapy Using Carmustine, Etoposide, Cytarabine, Melphalan, Thymoglobulin and Autologous CD34+ Hematopoietic Stem Cell Transplant for the Treatment of Poor Prognosis Multiple Sclerosis[NCT00288626]Phase 225 participants (Actual)Interventional2006-07-31Completed
An Open, Multi-center, Randomized Trial Comparing Haploidentical HSCTs From Young Non-first-degree and Older First-degree Donors in Hematological Malignancies[NCT04547049]Phase 3160 participants (Anticipated)Interventional2020-09-01Recruiting
Pilot Study Evaluating the Use of Ex Vivo Expanded Cord Blood Progenitors as Supportive Care Following Chemotherapy (FLAG) in Patients With AML or Acute Leukemia of Ambiguous Lineage[NCT01701323]Phase 17 participants (Actual)Interventional2012-12-10Terminated
Phase II Trial of Intermediate-Dose Cytarabine to Modulate EWS/FLI for Children and Young Adults With Recurrent or Refractory Ewing Sarcoma[NCT00470275]Phase 210 participants (Actual)Interventional2007-05-31Completed
A Phase II Trial Of BEAM/Rituximab/Autologous Hematopoietic Stem Cell Transplantation (AHSCT) For Patients With CD20 Positive Non-Hodgkin's Lymphoma[NCT00080886]Phase 268 participants (Actual)Interventional2002-05-08Completed
German Multicenter Trial for Treatment Optimisation in Acute Lymphoblastic Leukemia in Adults and Adolescents Above 15 Years With Rituximab for Improvement of Prognosis in CD20 Positive Standard Risk ALL (Amend 2)[NCT00199004]Phase 460 participants (Anticipated)Interventional2004-04-30Completed
Phase I/II Study of Bortezomib (VELCADE) Plus Rituximab-HyperCVAD Alternating With Bortezomib Plus Rituximab-High Dose Methotrexate/Cytarabine in Patients With Untreated Aggressive Mantle Cell Lymphoma[NCT00477412]Phase 1/Phase 2107 participants (Actual)Interventional2007-04-03Completed
Efficacy of 6x R-CHOP Followed by Myeloablative Radiochemotherapy and Autologous Stem Cell Transplantation vs. 3 x (R-CHOP/R-DHAP) Followed by a High Dose ARA-C Containing Myeloablative Regimen and Autologous Stem Cell Transplantation[NCT00209222]Phase 3360 participants (Anticipated)Interventional2004-07-31Recruiting
Autologous Blood or Marrow Transplantation for Aggressive Non-Hodgkin's Lymphoma Based on Early [18F] FDG-PET Scanning[NCT00238368]Phase 259 participants (Actual)Interventional2004-02-29Completed
SPINOZA / שפינוזה. Study With Preparatory INduction Of Zevalin in Aggressive Lymphoma. A Randomized Phase 3 Study of BEAM Versus 90Yttrium Ibritumomab Tiuxetan (Zevalin) / BEAM in Patients Requiring ASCT for Relapsed DLBCL[NCT02366663]Phase 33 participants (Actual)Interventional2015-01-31Terminated(stopped due to Withdrawal of sponsor support)
A Phase I Trial of High Dose Therapy and Autologous Stem Cell Transplantation Followed by Infusion of Chimeric Antigen Receptor (CAR) Modified T-Cells Directed Against CD19+ B-Cells for Relapsed and Refractory Aggressive B Cell Non-Hodgkin Lymphoma[NCT01840566]Phase 117 participants (Actual)Interventional2013-04-30Active, not recruiting
Moscow-Berlin 2008 Multicenter Randomised Study for Treatment of Acute Lymphoblastic Leukemia in Children and Adolescents[NCT01953770]3,000 participants (Anticipated)Interventional2008-02-29Active, not recruiting
A Phase III Randomized Trial of Steroids + Tyrosine Kinase Inhibitor (TKI) Induction With Chemotherapy or Blinatumomab for Newly Diagnosed BCR-ABL-Positive Acute Lymphoblastic Leukemia (ALL) in Adults[NCT04530565]Phase 3348 participants (Anticipated)Interventional2021-01-25Recruiting
Randomized Controlled Trial to Test Efficacy of High-Dose Methotrexate Consolidation Therapy for BCR-ABL-Negative Acute Lymphoblastic Leukemia in Adults[NCT00131027]Phase 3240 participants (Anticipated)Interventional2002-09-30Recruiting
Pilot Study of Crenolanib Combined With Standard Salvage Chmetherapy in Subjects With Relapsed/Refractory Acute Myeloid Leukemia[NCT02626338]Phase 1/Phase 216 participants (Actual)Interventional2016-02-29Completed
Combined Retinoic Acid,Arsenic Trioxide and Chemotherapy for Newly-diagnosed Acute Promyelocytic Leukemia: Chinese National Multi-center Randomized Study[NCT01987297]Phase 4738 participants (Anticipated)Interventional2012-06-30Active, not recruiting
Pilot Study of the Efficacy and Tolerance of the Adjunction of a Fish Oil Emulsion to Daunorubicin and Cytarabine Chemotherapy for the Treatment of Acute MYeloblastic Leukemia of Younger Patients With High-risk Cytogenetics[NCT01999413]Phase 230 participants (Actual)Interventional2013-11-30Completed
A Randomized, Double-Blind, Placebo Controlled Phase 3 Study of Venetoclax Co-Administered With Low Dose Cytarabine Versus Low Dose Cytarabine in Treatment Naïve Patients With Acute Myeloid Leukemia Who Are Ineligible for Intensive Chemotherapy[NCT03069352]Phase 3211 participants (Actual)Interventional2017-05-23Active, not recruiting
Accelerated Dose Schedule of Cytarabine Consolidation Therapy for Patients With Acute Myeloid Leukemia (AML) in Complete Remission[NCT04914676]Phase 258 participants (Anticipated)Interventional2022-03-08Recruiting
Phase III Randomized Trial of Clofarabine as Induction and Post-Remission Therapy vs. Standard Daunorubicin &Amp; Cytarabine Induction and Intermediate Dose Cytarabine Post-Remission Therapy, Followed by Decitabine Maintenance vs. Observation in Newly-Dia[NCT02085408]Phase 3727 participants (Actual)Interventional2011-02-04Active, not recruiting
A Phase 2 Multicenter Study of High Dose Chemotherapy With Autologous Stem Cell Transplant Followed by Maintenance Therapy With Romidepsin for the Treatment of T Cell Non-Hodgkin Lymphoma[NCT01908777]Phase 247 participants (Actual)Interventional2013-07-16Active, not recruiting
Phase II Study of Rituximab Given in Conjunction With Standard Chemotherapy in Primary Central Nervous System (CNS) Lymphoma[NCT00335140]Phase 226 participants (Actual)Interventional2007-08-23Terminated(stopped due to slow accrual)
A Phase I/II, Open-Label, Multi-Center Study Evaluating the Safety and Efficacy of Ruxolitinib and CPX-351 in Combination for the Treatment of Advanced Phase Myeloproliferative Neoplasms[NCT03878199]Phase 1/Phase 247 participants (Anticipated)Interventional2019-02-20Recruiting
A Multicenter Phase II Study Evaluating BeEAM (Bendamustine, Etoposide, Cytarabine, Melphalan) Prior to Autologous Stem Cell Transplant for First and Second Chemosensitive Relapses in Patients With Follicular Lymphoma[NCT02008006]Phase 221 participants (Actual)Interventional2014-07-09Terminated(stopped due to Insufficient recruitment and unavailability of the treatment)
A Phase II Study of Cladribine, Cytarabine, and Granulocyte-Colony Stimulating Factor With Fractionated Gemtuzumab Ozogamicin (CLAG-GO) for the Treatment of Patients With Persistent, Relapsed or Refractory Acute Myeloid Leukemia[NCT04050280]Phase 239 participants (Anticipated)Interventional2019-11-01Recruiting
"Phase I Study of Yttrium-90 Labeled Anti-CD25 (a-Tac) Monoclonal Antibody Plus BEAM for Autologous Hematopoietic Cell Transplantation (AHCT) in Patients With Primary Refractory or Relapsed Hodgkin Lymphoma, the a-Tac BEAM Regimen"[NCT01476839]Phase 125 participants (Actual)Interventional2012-11-09Active, not recruiting
A Phase I Study of Clofarabine & Cytosine Arabinoside Therapy for Older Adults With Acute Myeloid Leukemia[NCT00081822]Phase 123 participants (Actual)Interventional2004-01-31Completed
A Randomized Comparison of Fludarabine in Combination With Cytarabine Versus High -Dose Cytarabine in Post-remission Therapy for AML1-ETO Acute Myeloid Leukemia[NCT02024308]Phase 462 participants (Anticipated)Interventional2010-11-30Recruiting
A Phase I Study of Pomalidomide Given at the Time of Lymphocyte Recovery Following Induction Timed Sequential Chemotherapy With Cytarabine, Daunorubicin and Etoposide (AcDVP16) in Patients With Newly Diagnosed Acute Myeloid Leukemia (AML) and High-Risk MD[NCT02029950]Phase 150 participants (Actual)Interventional2013-12-16Completed
Protocol for Patients With High Risk (Resistant, Refractory, Relapsed or Adverse Cytogenetic) AML[NCT00005863]Phase 30 participants Interventional1998-08-31Completed
An ALFA 2101 Multicenter Randomized Phase II Study: CPX-351 Versus Intensive Chemotherapy in Patients With de Novo Intermediate or Adverse Risk AML Stratified by Genomics[NCT05260528]Phase 2210 participants (Anticipated)Interventional2023-05-03Recruiting
Phase III Randomized Study of Induction Chemotherapy With or Without MDR-Modulation With PSC-833 (NSC # 648265, IND # 41121) Followed by Cytogenetic Risk-Adapted Intensification Therapy Followed by Immunotherapy With rIL-2 (NSC # 373364, IND # 1969) vs. O[NCT00006363]Phase 3720 participants (Actual)Interventional2000-11-30Completed
A Phase II Trial of Rituxan and BEAM High-Dose Chemotherapy and Autologous Peripheral Blood Progenitor Transplant for Lymphoma[NCT00007852]Phase 244 participants (Actual)Interventional2000-09-01Completed
A Phase II Multicenter Study Comparing Antiretroviral Therapy Alone to Antiretroviral Therapy Plus Cytosine Arabinoside (Cytarabine; Ara-C) for the Treatment of Progressive Multifocal Leukoencephalopathy (PML) in Human Immunodeficiency Virus (HIV)-Infecte[NCT00001048]Phase 290 participants InterventionalCompleted
Phase 1 Study of CPX-351(Cytarabine:Daunorubicin) Liposome Injection in Patients With Advanced Hematologic Malignancies.[NCT00389428]Phase 148 participants (Actual)Interventional2006-09-30Completed
CINJALL: Treatment for Children With Acute Lymphocytic Leukemia[NCT00176462]Phase 260 participants (Actual)Interventional2001-02-28Completed
Programma di Terapia Per Pazienti Affetti da Linfoma Diffuso a Grandi Cellule B CD20 Positive[NCT00556127]Phase 294 participants (Actual)Interventional2002-06-30Completed
A Phase II, Single Arm Study of Tislelizumab Combined With DNA Demethylation Agent +/- CAG Regimen in the Treatment of Patients With High-risk AML or AML Patients Older Than 60 Years of Age Who Are Unfit for Intensive Chemotherapy[NCT04541277]Phase 255 participants (Anticipated)Interventional2020-09-01Recruiting
Safety and Efficacy of Stem Cell Mobilization Using G-CSF (Filgrastim) Alone Compared to Intermediate-dose Cytosine Arabinoside Plus G-CSF in Multiple Myeloma Patients.[NCT01908621]Phase 390 participants (Actual)Interventional2013-03-20Completed
A Randomized, Open-label, Two-period, Two-way Crossover Bioequivalence Study of Two (Cytarabine: Daunorubicin) Liposome for Injection in Elderly AML Subjects[NCT05801835]36 participants (Anticipated)Interventional2023-08-25Recruiting
A Phase 1, Multicenter, Open-Label, Dose Escalation Study to Assess the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Clinical Activity of Orally Administered FHD-286, as Monotherapy or Combination Therapy, in Subjects With Advanced Hemato[NCT04891757]Phase 1144 participants (Anticipated)Interventional2021-06-14Recruiting
A Phase II Study of CPX-351 in Younger Patients < 60 Years Old With Secondary Acute Myeloid Leukemia[NCT04269213]Phase 246 participants (Anticipated)Interventional2021-07-29Recruiting
A Phase I Clinical Trial Testing the Safety of IL-21-Expanded, Off-the-shelf, Third-party Natural Killer Cells (KDS-1001) for the Induction of Relapsed/Refractory Acute Myeloid Leukemia[NCT04220684]Phase 121 participants (Anticipated)Interventional2020-06-01Recruiting
Dexamethasone Plus Salvage Chemotherapy Versus Salvage Chemotherapy Alone in Patients With First Relapsed or Refractory Acute Myeloid Leukemia: a Randomized, Controlled, Open-label, Multicenter, Phase III Study[NCT03765541]Phase 3142 participants (Anticipated)Interventional2020-01-13Recruiting
A Phase II Study of Ibrutinib Plus Rituximab Followed by Venetoclax and Hyper-CVAD Consolidation in Newly Diagnosed Young Patients With Mantle Cell Lymphoma: Window II Protocol[NCT03710772]Phase 250 participants (Anticipated)Interventional2019-05-01Active, not recruiting
A Phase II Trial of Epigenetic Priming in Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT03164057]Phase 2206 participants (Actual)Interventional2017-06-15Active, not recruiting
Treatment of Patients With Newly Diagnosed Standard Risk B-Lymphoblastic Leukemia (B-ALL) or Localized B-Lineage Lymphoblastic Lymphoma (B-LLy)[NCT01190930]Phase 39,350 participants (Actual)Interventional2010-08-09Active, not recruiting
A Phase II Study of CLAG Regimen in Combination With Imatinib Mesylate (Gleevec) in Refractory or Relapsed Acute Myeloid Leukemia[NCT00594555]Phase 220 participants (Anticipated)Interventional2007-11-30Withdrawn(stopped due to Principal Investigator resigned position with the University of Cincinnati, closing study at this site will reopen study in new position)
Open Randomized Prospective Clinical Study of Rituximab Combined With Fotemustine, Pemetrexed, Dexamethasone Versus Rituximab Plus Methotrexate, Cytarabine, and Dexamethasone in the Treatment of Primary Central Nervous System Lymphoma[NCT04083066]Phase 420 participants (Anticipated)Interventional2019-09-05Recruiting
A Phase I/II Study of Ribavirin and Low-dose Cytarabine Arabinoside (Ara-C) in Acute Myeloid Leukemia (AML) M4 and M5 Subtypes, and AML With High eIF4E Expression[NCT01056523]Phase 1/Phase 229 participants (Actual)Interventional2010-01-31Completed
Cardioprotection With Dexrazoxane in Acute Myeloid Leukemia (AML), High-Risk Myelodysplastic Syndrome (MDS), Myeloid Blast Phase of Chronic Myeloid Leukemia (CML), Ph+ AML, and Myeloid Blast Phase of Myeloproliferative Neoplasms[NCT03589729]Phase 2100 participants (Anticipated)Interventional2018-09-19Recruiting
A Trial of Atovaquone (Mepron®) Combined With Conventional Chemotherapy for de Novo Acute Myeloid Leukemia (AML) in Children, Adolescents, and Young Adults (ATACC AML)[NCT03568994]Early Phase 126 participants (Actual)Interventional2018-07-10Active, not recruiting
A Prospective, Single Arm, Multicenter Clinical Study to Evaluate the Efficacy of Venetoclax Combined With Azacytidine or DA Regimen in Prevention the Relapse of Consecutive MRD Positive AML Patients[NCT05361057]Phase 240 participants (Anticipated)Interventional2022-05-01Recruiting
S0629, Observational Study of Asymptomatic Waldenstrom's Macroglobulinemia and Phase II Study of Tandem Autologous Transplant and Maintenance Treatment for Patients With Symptomatic Disease[NCT00723658]Phase 20 participants (Actual)Interventional2008-09-30Withdrawn(stopped due to lack of accrual)
Safety and Efficacy of Stem Cell Mobilization Using G-CSF (Filgrastim) Alone Compared to Intermediate-dose Cytosine Arabinoside Plus G-CSF in Hodgkin's Lymphoma and Non-Hodgkin's Lymphoma Patients.[NCT02722733]Phase 390 participants (Anticipated)Interventional2016-03-31Recruiting
Intensified PEG-Asparaginase in High Risk Acute Lymphoblastic Leukemia (ALL): A Pilot Study[NCT00866307]Phase 1104 participants (Actual)Interventional2009-02-23Completed
A Phase 2 Multicenter Study of First-line Dasatinib Plus Conventional Chemotherapy in Adults With Newly Diagnosed Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia[NCT01004497]Phase 251 participants (Actual)Interventional2010-03-31Completed
AIEOP LLA 2000 Multicenter Study for the Diagnosis and Treatment of Childhood Acute Lymphoblastic Leukemia[NCT00613457]Phase 32,039 participants (Actual)Interventional2000-09-30Completed
A Phase 1/2, Multicenter, Dose-Escalating Study To Evaluate the Safety, Pharmacokinetics, Pharmacodynamics, and Efficacy Of Quizartinib Administered in Combination With Re-Induction Chemotherapy, and as a Single-Agent Continuation Therapy, in Pediatric Re[NCT03793478]Phase 1/Phase 265 participants (Anticipated)Interventional2018-08-15Recruiting
An Open Label Study of the Effect of Rituxan, High Dose Methotrexate and High Dose Cytarabine on Response Rate in Patients With Primary Central Nervous System Lymphoma.[NCT00517699]Phase 25 participants (Actual)Interventional2007-09-30Terminated(stopped due to Study was terminated early due to lack of enrollment.)
Phase 3 Open-label, Multicenter, Randomized Study of ASP2215 Versus Salvage Chemotherapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) With FLT3 Mutation[NCT03182244]Phase 3276 participants (Actual)Interventional2018-01-15Active, not recruiting
A Pilot Study of a Novel Sequential Treatment Utilizing CPX-351 as Salvage Chemotherapy Followed by Allogeneic Stem-Cell Transplantation (SCT) Utilizing a Haplo-cord Graft for Patients With Relapsed or Refractory Leukemia or Myelodysplastic Syndrome[NCT03393611]Phase 114 participants (Actual)Interventional2012-11-30Completed
Autologous and Allogeneic Transplantation for T-Cell Lymphoma: Impact of Campath -1H and Soluble CD52[NCT00505921]Phase 227 participants (Actual)Interventional2003-03-31Terminated(stopped due to Slow Accrual.)
A Phase II Evaluation of High Dose Chemotherapy and Autologous Stem Cell Transplantation for Intestinal T-cell Lymphomas[NCT00669812]Phase 260 participants (Anticipated)Interventional2008-02-29Recruiting
A Phase 2 Study of SNDX-5613 in Combination With Chemotherapy for Patients With Relapsed or Refractory KMT2A-Rearranged Infant Leukemia[NCT05761171]Phase 278 participants (Anticipated)Interventional2023-11-20Recruiting
Treatment of Transient Myeloproliferative Disorder (TMD) in Children With Down Syndrome (DS)[NCT00411281]Phase 30 participants (Actual)Interventional2006-03-31Withdrawn(stopped due to Per Group Chair: This study will not move forward.)
A Phase I Study of Venetoclax in Combination With Cytotoxic Chemotherapy, Including Calaspargase Pegol, for Children, Adolescents and Young Adults With High-Risk Hematologic Malignancies[NCT05292664]Phase 192 participants (Anticipated)Interventional2023-03-29Recruiting
International Randomised Phase III Clinical Trial in Children With Acute Myeloid Leukaemia - Incorporating an Embedded Dose Finding Study for Gemtuzumab Ozogamicin in Combination With Induction Chemotherapy[NCT02724163]Phase 3700 participants (Anticipated)Interventional2016-04-30Recruiting
Modified BFM-95 Regimen for the Treatment of Newly Diagnosed T-lymphoblastic Lymphoma in Adults:a Prospective Phase II Study[NCT02396043]Phase 250 participants (Anticipated)Interventional2015-03-31Recruiting
International Proof of Concept Therapeutic Stratification Trial of Molecular Anomalies in Relapsed or Refractory HEMatological Malignancies in Children, Subprotocol D: Trametinib + Dexamethasone + Cyclophosphamide and Cytarabine in Pediatric Patients With[NCT05658640]Phase 1/Phase 226 participants (Anticipated)Interventional2023-04-01Not yet recruiting
A Clinico-Pathologic Study of Primary Mediastinal B-Cell Lymphoma (IELSG 26)[NCT00689845]120 participants (Anticipated)Interventional2007-06-30Recruiting
A Phase I Study Evaluating the Safety, Tolerability and Biological Activity of EZN-3042, a Survivin mRNA Antagonist, Administered With Re-induction Chemotherapy in Children With Relapsed Acute Lymphoblastic Leukemia (ALL)[NCT01186328]Phase 16 participants (Actual)Interventional2010-08-24Terminated(stopped due to Enzon Pharmaceuticals decided to end its development of EZN-3042.)
A Phase 1 Clinical Study of DSP-2033 (Alvocidib) in Combination With Cytarabine/Mitoxantrone or Cytarabine/Daunorubicin (7+3) in Patients With Acute Myeloid Leukemia[NCT03563560]Phase 110 participants (Actual)Interventional2018-05-15Completed
D-CTAG in the Treatment of Newly Diagnosed Acute Myeloid Leukemia in Elderly Patients[NCT04168138]20 participants (Anticipated)Interventional2019-11-01Recruiting
Phase I/II Study of the Combination of Quizartinib (AC220) With 5-Azacytidine or Low-Dose Cytarabine for the Treatment of Patients With Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS)[NCT01892371]Phase 1/Phase 2200 participants (Anticipated)Interventional2013-11-12Completed
Optimal Treatment Strategy Based on Prognostic Groups for Pediatric de Novo Acute Myeloid Leukemia[NCT02848183]Phase 2350 participants Interventional2016-01-31Recruiting
Bortezomib* and Vorinostat as Maintenance Therapy After Autologous Transplant for Non-Hodgkin Lymphoma Using R-BEAM or BEAM Conditioning Transplant Regimen[NCT00992446]Phase 227 participants (Actual)Interventional2010-09-02Completed
Chemosensitization With Plerixafor Plus G-CSF in Relapsed or Refractory Acute Myeloid Leukemia[NCT00906945]Phase 1/Phase 239 participants (Actual)Interventional2011-02-28Completed
Pediatric-inspired Regimen Combined With Venetoclax for Adolescent and Adult Patients With de Novo Philadelphia Chromosome-Negative Acute Lymphoblastic Leukemia[NCT05660473]Phase 2100 participants (Anticipated)Interventional2022-10-31Recruiting
ACUTE MYELOID LEUKAEMIA TRIAL 12[NCT00002658]Phase 32,000 participants (Anticipated)Interventional1994-01-31Active, not recruiting
A Phase 1b/2 Study of PCM-075 (Onvansertib) in Combination With Either Low-Dose Cytarabine or Decitabine in Subjects With Acute Myeloid Leukemia (AML)[NCT03303339]Phase 1/Phase 272 participants (Actual)Interventional2017-11-17Completed
Descriptive Study Evaluating the Presence and Function of Natural Killer Cells in Elderly Patients With Acute Myeloid Leukemia in First Remission.[NCT00540956]40 participants (Anticipated)Interventional2006-11-30Completed
A Phase 1 Study of AC220 (ASP2689) in Combination With Induction and Consolidation Chemotherapy in Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT01390337]Phase 119 participants (Actual)Interventional2011-10-31Completed
A Phase 1 Study of the Deglycosylated Ricin A Chain-containing Combined Anti-CD19 and Anti-CD22 Immunotoxin Combotox in Combination With High-Dose Cytarabine in Adult Relapsed or Refractory B-lineage Acute Lymphoblastic Leukemia[NCT01408160]Phase 118 participants (Actual)Interventional2013-04-30Terminated
Randomised Comparison of OSHO Induction vs. the German AML Intergroup Standard, Randomised Comparison of AraC/Mtx vs. Flu/AraC/Mtx in Pts Without CR After One Induction Cycle and Randomized Comparison of One vs. Two Consolidation Therapies.[NCT01414231]Phase 3850 participants (Anticipated)Interventional2002-04-30Recruiting
Phase II Trial of Venetoclax in Combination With Azacitidine and CAG as Induction Therapy in Patients With Refractory/Relapse Acute Myeloid Leukemia[NCT05807347]Phase 242 participants (Anticipated)Interventional2023-02-01Recruiting
Phase 2 Study Evaluating the Efficacy of Rituximab Plus Modified VPDL for Newly Diagnosed CD20-Positive Adult Acute Lymphoblastic Leukemia[NCT01429610]Phase 278 participants (Actual)Interventional2011-11-30Active, not recruiting
Pivotal Study for High Dose Therapy and Autologous Stem Cell Transplantation in Early Stages of CLL[NCT00275015]Phase 2169 participants (Actual)Interventional1998-01-31Completed
A Pilot Study of Arsenic Trioxide-Based Consolidation Therapy for the Primary Treatment of Acute Promyelocytic Leukemia[NCT00276601]Phase 20 participants Interventional2004-10-31Completed
Ofatumumab (O) in Combination With Chemotherapy: Hyper-Fractionated Cyclophosphamide, Doxorubicin, Vincristine and Dexamethasone (O-HyperCVAD) Alternating With Ofatumumab High-Dose Cytarabine and Methotrexate (O-MA) for Patients With Newly Diagnosed Mantl[NCT01527149]Phase 237 participants (Actual)Interventional2011-12-06Active, not recruiting
The Prospective Non-randomized Phase II Clinical Trial of Vemurafenib in Combination With Cytarabine and 2-chlorodeoxyadenosine in Children With Langerhans-cell Hisitocytosis With BRAF V600E Mutation[NCT03585686]Phase 212 participants (Anticipated)Interventional2018-06-26Recruiting
Phase II Evaluation of Gemtuzumab Ozogamicin in Combination With Cytarabine in Untreated Patients Above the Age of 60 Years With Acute Myeloid Leukemia and High-Risk Myelodysplastic Syndrome[NCT00195000]Phase 243 participants (Actual)Interventional2003-05-31Completed
Gemtuzumab Ozogamicin (CMA-676) Followed or Not by Intensive Chemotherapy as Initial Treatment for Elderly Patients With Acute Myeloid Leukemia: An EORTC-LG Pilot Phase II Study[NCT00006122]Phase 2106 participants (Actual)Interventional2000-06-30Completed
A Pilot Study of Double Cord Blood Stem Cell Transplantation in Patients With Hematologic Malignancies[NCT00423826]0 participants Expanded Access2007-01-31No longer available
Randomized Phase II Trial of Idarubicin + Ara-C +/- Bevacizumab in Patients Age < 60 With Untreated Acute Myeloid Leukemia[NCT00096148]Phase 2120 participants (Actual)Interventional2004-10-31Terminated(stopped due to Administratively complete.)
Multicenter Trial for Treatment Optimization in T-lymphoblastic Lymphoma in Adults and Adolescents Older Than 15 Years (GMALL T-LBL 1/2004) (Amend 1)[NCT00199017]Phase 475 participants (Anticipated)Interventional2004-04-30Completed
Dual Growth Factor (rhTPO + G-CSF) and Chemotherapy Combination Regimen in Acute Myeloid Leukemia: Study Protocol for a Randomized Controlled Trial[NCT05382390]Phase 3130 participants (Anticipated)Interventional2022-01-21Recruiting
Idarubicin, Cytarabine and Pravastatin (IAP) for Induction of Newly Diagnosed Acute Myeloid Leukemia (AML)[NCT01831232]24 participants (Actual)Interventional2013-05-31Completed
High-dose Cytarabine/Mitoxantrone Followed by Autotransplantation for Therapy-Related Myelodysplastic Syndrome/Therapy -Related Acute Myeloid Leukemia[NCT00774046]Phase 232 participants (Actual)Interventional2002-12-31Completed
Clinical Observation on the Efficacy and Safety of CLAE Regimen (Cladribine + Cytarabine + Etoposide) in the Treatment of Relapsed/Refractory T- Acute Lymphoblastic Leukemia/Lymphoblastic Lymphoma[NCT04679506]50 participants (Anticipated)Observational [Patient Registry]2020-12-31Not yet recruiting
A Phase I Study of 5-Azacytidine in Combination With Chemotherapy for Children With Relapsed or Refractory ALL or AML[NCT01861002]Phase 115 participants (Actual)Interventional2013-05-22Completed
Gleevec (Imatinib) Plus Multi-Agent Chemotherapy For Newly-Diagnosed Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia[NCT00618501]Phase 250 participants (Anticipated)Interventional2005-10-31Completed
A Randomized, Multi-center, Open-label Phase III Bridging Study to Compare Efficacy of Liposomal Cytarabine-Daunorubicin for Injection With Cytarabine and Daunorubicin in Treating Older Patients With High-Risk (Secondary) Acute Myeloid Leukemia[NCT06182592]Phase 3120 participants (Anticipated)Interventional2024-01-31Not yet recruiting
A Phase I, Single-arm, Open Label, Dose Escalation, Multicenter Study of Off-the-shelf Natural Killer (NK) Cells (SAR445419) in Participants With Relapsed or Refractory Acute Myeloid Leukemia (R/R AML)[NCT05712278]Phase 112 participants (Anticipated)Interventional2023-06-16Recruiting
A Randomized Phase III Study of the Treatment of Children and Adolescents With Refractory or Relapsed Acute Myeloid Leukemia[NCT00186966]Phase 3394 participants (Actual)Interventional2002-03-31Completed
Dose-Finding (Phase 1) Study of Continuous Infusion Cladribine, Cytarabine and Mitoxantrone (CI-CLAM) for Adults With Relapsed/Refractory Acute Myeloid Leukemia or Other High-Grade Myeloid Neoplasms Treated at UW/SCCA[NCT04196010]Phase 113 participants (Actual)Interventional2020-05-08Terminated(stopped due to Terminated due to unfavorable risk-benefit ratio of investigational regimen.)
AIDA2000 - Risk-Adapted Therapy for Patients With Acute Promyelocytic Leukemia(APL)[NCT00180128]Phase 480 participants (Anticipated)Interventional2000-01-31Recruiting
A Randomized Phase II/III Study of Conventional Chemotherapy +/- Uproleselan (GMI-1271) in Older Adults With Acute Myeloid Leukemia Receiving Intensive Induction Chemotherapy[NCT03701308]Phase 2/Phase 3670 participants (Anticipated)Interventional2019-03-28Suspended(stopped due to End of Initial Phase of Multi-phase protocol)
Treatment for Newly Diagnosed Patients With Stage III/IV Non-Hodgkin Lymphoma-Study XIII (A Therapeutic Pilot Study)[NCT00187122]42 participants (Actual)Interventional1993-03-31Completed
Small Noncleaved Cell (SNCC), Non-Hodgkin Lymphoma (NHL), Large Cell NHL (B-Cell) and B-Cell Acute Lymphoblastic Leukemia (B-ALL) Study II[NCT00187161]Phase 268 participants (Actual)Interventional1994-11-30Completed
Arsenic Trioxide, High-Dose Cytarabine and Idarubicin Induction Therapy in Previously Untreated de Novo and Secondary Adult Acute Myeloid Leukemia Patients < 60 Years Old - A Phase I Study[NCT00093483]Phase 161 participants (Actual)Interventional2002-04-30Completed
A Randomised Multicentric Phase III Study for the Treatment of Young Patients With High Risk (IPI 2-3) Diffuse Large B-Cell Lymphoma. Dose Dense Chemotherapy + Rituximab +/- Intensified High Dose Chemoimmunotherapy With Support of Peripheral Autologous St[NCT00499018]Phase 3399 participants (Anticipated)Interventional2006-01-31Active, not recruiting
Phase I Study of a Pharmacologically Derived Hybrid Bolus-Infusion Schedule of Flavopiridol (NSC 649890, IND 46,211) Given in Timed Sequential Combination With Cytosine Arabinoside (Ara-C) and Mitoxantrone for Adults With Relapsed and Refractory Acute Leu[NCT00470197]Phase 135 participants (Actual)Interventional2007-04-30Completed
Randomized Phase II/III-Study on All-Trans Retinoic Acid in Combination With Induction and Consolidation Therapy as Well as Pegfilgrastim After Consolidation Therapy in Younger Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT00151242]Phase 2/Phase 3920 participants (Actual)Interventional2004-07-31Completed
Large Cell Lymphoma, Pilot Study III[NCT00187070]8 participants (Actual)Interventional1997-12-31Completed
Phase I/II Trial to Study the Dose, Tolerability and the Effectiveness of Imatinib in Combination With Daunorubicine and Cytarabine for Patients With Chronic Myelogenous Leukemia in Myeloid Acute Phase[NCT00219765]Phase 1/Phase 230 participants Interventional2001-05-31Terminated
Pilot Study I for Treatment of Cancer in Children With Ataxia-Telangiectasia[NCT00187057]6 participants (Actual)Interventional2002-09-30Completed
Phase I/II Study of the Combination of Inotuzumab Ozogamycin (CMC-544) With Low-Intensity Chemotherapy in Patients With Acute Lymphoblastic Leukemia (ALL)[NCT01371630]Phase 1/Phase 2276 participants (Anticipated)Interventional2011-08-26Recruiting
Treatment of Acute Lymphoblastic Leukemia in Children[NCT00400946]Phase 3800 participants (Actual)Interventional2005-04-30Completed
LAL-AR-N-2005: Study Treatment for Children High Risk Acute Lymphoblastic Leukemia[NCT00526409]Phase 440 participants (Anticipated)Interventional2005-06-30Completed
[NCT02937662]Phase 260 participants (Anticipated)Interventional2016-10-31Recruiting
International Collaborative Treatment Protocol for Infants Under One Year With Acute Lymphoblastic or Biphenotypic Leukemia[NCT00550992]445 participants (Anticipated)Interventional2006-01-31Recruiting
DLCL002 Protocol for Young Patients With Newly Diagnosed High Risk Aggressive B-cell Lymphoma, a Multicenter Phase II Study[NCT03837873]Phase 2118 participants (Anticipated)Interventional2019-01-21Recruiting
Phase II Study of the Hyper-CVAD Regimen in Sequential Combination With Inotuzumab Ozogamicin as Frontline Therapy for Adults With B-Cell Lineage Acute Lymphocytic Leukemia[NCT03488225]Phase 24 participants (Actual)Interventional2018-03-28Terminated(stopped due to the study was closed early due to competing trials)
PHASE II TRIAL OF CHEMOTHERAPY PLUS RADIOTHERAPY FOR MANAGEMENT OF PRIMARY CENTRAL NERVOUS SYSTEM NON-HODGKIN'S LYMPHOMA (PCNSL)[NCT00002676]Phase 236 participants (Actual)Interventional1995-07-31Completed
A Phase II Trial of ICE Chemotherapy Followed by High Dose BEAM Chemotherapy With Autologous Peripheral Blood Progenitor Cell Transplantation in Patients >= 60 Years Old With Refractory or Relapsed Intermediate Grade Non-Hodgkin's Lymphoma[NCT00002982]Phase 20 participants Interventional1997-01-31Completed
A Phase I Trial of a Combined Regimen of Chemotherapy and 90Y-Labeled, Humanized LL2 (Anti-CD22) Antibody With Peripheral Stem Cell Rescue for the Treatment of Relapsed or Refractory Non-Hodgkin's Lymphoma[NCT00004086]Phase 10 participants Interventional1997-06-30Active, not recruiting
Phase I Investigation of the Feasibility of Combining 5-azacytidine With Highdose Cytarabine (HiDAC) and Mitoxantrone Chemotherapy in a Sequential Manner for Remission Induction in High-risk Acute Myelogenous Leukemia (AML)[NCT01839240]Phase 150 participants (Actual)Interventional2012-06-06Completed
A Single-center Open-label Phase I Study of ALT-801 for ex Vivo Maturation and in Vivo Retargeting of Haploidentical Natural Killer Cells Delivered Following Fludarabine, Cytarabine, and G-CSF in Patients With Relapsed/Refractory Acute Myeloid Leukemia[NCT01478074]Phase 10 participants (Actual)Interventional2011-11-30Withdrawn(stopped due to The treatment planned was determined to be of low feasibility as no subject was found eligible and able to enroll after screening over 30 subjects)
A Phase 3 Trial of Systematic Versus Response-adapted Timed-Sequential Induction in Patients With Core Binding Factor (CBF) Acute Myeloid Leukemia (AML)[NCT00428558]Phase 3200 participants (Actual)Interventional2007-07-31Completed
ALL-BFM 2000 Multi-Center Study for the Treatment of Children and Adolescents With Acute Lymphoblastic Leukemia[NCT00430118]Phase 34,559 participants (Actual)Interventional2000-07-31Completed
Randomised Induction and Post Induction Therapy in Older Patients (≥61 Years of Age) With Acute Myeloid Leukemia (AML) and Refractory Anemia With Excess Blasts (RAEB, RAEB-t)[NCT00121303]Phase 3600 participants (Anticipated)Interventional2005-01-31Completed
A Phase II Study of Ara-C (Cytarabine) in Men With Androgen Independent Prostate Cancer[NCT00480090]Phase 210 participants (Actual)Interventional2007-04-30Completed
FLAT: Fludarabine, Cytarabine and Topotecan in Treating Patients With Refractory or Relapsed Acute Myeloid Leukemia[NCT00488709]Phase 447 participants (Actual)Interventional2003-05-31Completed
Randomised Trial on Allogeneic Haematopoietic Stem Cell Transplantation in Patients Under the Age of 60 Years With Acute Myeloid Leukemia of Intermediate Risk in First Complete Remission and a Matched Sibling or Unrelated Donor (ETAL-1)[NCT01246752]Phase 3143 participants (Actual)Interventional2011-02-10Terminated(stopped due to decreased recrucial rate as the likelihood of achieving the envisioned patient number in a realistic time-frame was very low.)
Multicenter, Phase 2 Clinical Trial Evaluating the Efficacy and Tolerability of Induction and Consolidation Chemotherapy Comprising Fludarabine, Cytarabine, and Attenuated-dose Idarubicin in Elderly Patients With AML(Acute Myeloid Leukemia)[NCT01247493]Phase 2108 participants (Actual)Interventional2007-06-30Completed
FLAG-IDA Chemotherapy Induction Follow by Intensive Chemotherapy Postremission +/- Autologous Hemopoietic Stem Cell Transplantation or Bone Marrow Transplantation in Patients With High Risk Myelodysplastic Syndromes or Secondary Acute Myeloblastic Leukemi[NCT00487448]Phase 4200 participants (Anticipated)Interventional1998-07-31Completed
Phase I Trial of Cytarabine and Lenalidomide in Relapsed or Refractory Acute Myeloid Leukemia Patients[NCT01246622]Phase 132 participants (Actual)Interventional2011-02-07Completed
A Phase 1 and Pharmacokinetic Study of Uproleselan (GMI-1271, NSC #801708) in Combination With Fludarabine and Cytarabine for Patients With Acute Myeloid Leukemia Myelodysplastic Syndrome or Mixed Phenotype Acute Leukemia That Expresses E-Selectin Ligand [NCT05146739]Phase 118 participants (Anticipated)Interventional2023-12-23Recruiting
A Phase 1 Study of Vyxeos Plus Gilteritinib in Relapsed or Refractory, FLT3-Mutated Acute Myeloid Leukemia[NCT05024552]Phase 122 participants (Anticipated)Interventional2021-08-23Recruiting
A Study of Temozolomide, Etoposide, Doxil, Dexamethasone, Ibrutinib, and Rituximab (TEDDI-R) in Aggressive B-cell Lymphomas With Secondary Involvement of the Central Nervous System (CNS)[NCT03964090]Phase 265 participants (Anticipated)Interventional2019-06-27Recruiting
A Groupwide Pilot Study to Test the Tolerability and Biologic Activity of the Addition of Azacitidine (NSC# 102816) to Chemotherapy in Infants With Acute Lymphoblastic Leukemia (ALL) and KMT2A (MLL) Gene Rearrangement[NCT02828358]Phase 278 participants (Actual)Interventional2017-04-01Active, not recruiting
A Phase 1/2 Study of ASP2215 in Combination With Induction and Consolidation Chemotherapy in Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT02310321]Phase 1/Phase 284 participants (Actual)Interventional2015-02-26Active, not recruiting
Phase II Study of CPX-351 (Cytarabine:Daunorubicin) Liposome Injection in Patients With Newly Diagnosed AML at High Risk for Induction Mortality[NCT02286726]Phase 256 participants (Actual)Interventional2015-05-04Completed
Multicenter Randomized Phase II Study of Methotrexate (MTX) and Temozolomide Versus MTX, Procarbazine, Vincristine and Cytarabine for Primary CNS Lymphoma (PCNSL) in the Elderly[NCT00503594]Phase 292 participants (Anticipated)Interventional2007-07-31Recruiting
Phase I Dose Escalating Trial of VELCADE (PS-341) in Combination With Idarubicin and Cytosine Arabinoside in Patients With Acute Myelogenous Leukemia[NCT00505700]Phase 136 participants (Anticipated)Interventional2003-07-31Completed
LAL-Ph-2000: Treatment of Acute Lymphoblastic Leukemia Chromosome Philadelphia Positive[NCT00526305]Phase 4100 participants (Anticipated)Interventional2000-01-31Completed
A Pilot Study Of Cytarabine And High-Dose Mitoxantrone For Relapsed Or Refractory Hematologic Malignancies[NCT00047021]Phase 23 participants (Actual)Interventional2001-11-30Completed
Phase III - Study on All-Trans Retinoic Acid in Combination With Standard Induction and Consolidation Therapy in Older Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT00151255]Phase 3500 participants (Anticipated)Interventional2004-06-30Completed
A Phase I-II Study of Triciribine Phosphate Monohydrate (PTX-200) Plus Cytarabine in Refractory or Relapsed Acute Leukemia[NCT02930109]Phase 1/Phase 240 participants (Anticipated)Interventional2016-09-30Recruiting
LAL-BR/2001: Study Treatment to Low Risk ALL[NCT00526175]Phase 4150 participants (Anticipated)Interventional2001-06-30Completed
[NCT00529880]Phase 219 participants (Anticipated)Interventional2004-12-31Recruiting
A Phase 1 Study of NKX101, an Activating Chimeric Receptor Natural Killer Cell Therapy, in Subjects With Hematological Malignancies or Dysplasias[NCT04623944]Phase 190 participants (Anticipated)Interventional2020-09-21Recruiting
Modified BFM (Berlin-Frankfurt-Munster)Backbone Therapy for Chinese Children or Adolescents With Newly Diagnosed Lymphoblastic Lymphoma[NCT02845882]Phase 3150 participants (Anticipated)Interventional2016-01-31Active, not recruiting
A Phase III Study of Risk Directed Therapy for Infants With Acute Lymphoblastic Leukemia (ALL): Randomization of Highest Risk Infants to Intensive Chemotherapy +/- FLT3 Inhibition (CEP-701, Lestaurtinib; NSC#617807)[NCT00557193]Phase 3218 participants (Actual)Interventional2008-01-15Active, not recruiting
A Phase 1B/2A, Open-label Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Fosciclopirox Alone and In Combination With Cytarabine in Patients With Relapsed/Refractory Acute Myeloid Leukemia (AML)[NCT04956042]Phase 1/Phase 228 participants (Anticipated)Interventional2021-08-27Recruiting
A Phase 1 Study of ASP2215 in Combination With Induction and Consolidation Chemotherapy in Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT02236013]Phase 180 participants (Actual)Interventional2015-01-07Completed
LBL 2018 - International Cooperative Treatment Protocol for Children and Adolescents With Lymphoblastic Lymphoma[NCT04043494]Phase 3683 participants (Anticipated)Interventional2019-08-23Recruiting
Ma-Spore ALL 2010 Study[NCT02894645]Phase 4500 participants (Anticipated)Interventional2008-10-31Recruiting
A Phase Ib/II Study Evaluating the Safety and Efficacy of Idasanutlin in Combination With Cytarabine and Daunorubicin in Patients Newly Diagnosed With Acute Myeloid Leukemia (AML) and the Safety and Efficacy of Idasanutlin in the Maintenance of First AML [NCT03850535]Phase 1/Phase 224 participants (Actual)Interventional2019-03-25Terminated(stopped due to Sponsor's decision to terminate the study after Phase 1; will not proceed with Phase 2.)
A Trial Comparing Single Agent Melphalan to Carmustine, Etoposide, Cytarabine and Melphalan (BEAM) as a Preparative Regimen for Patients With Multiple Myeloma Undergoing High Dose Therapy Followed by Autologous Stem Cell Reinfusion[NCT03570983]Phase 2100 participants (Anticipated)Interventional2018-09-05Recruiting
A Phase I/II, Open-label Multicenter Trial to Determine Safety, Pharmacokinetics and Efficacy of GMI-1271 in Combination With Chemotherapy in Patients With Acute Myeloid Leukemia[NCT02306291]Phase 1/Phase 291 participants (Actual)Interventional2015-03-31Completed
A Phase II Study of Cloretazine® (VNP40101M) for Elderly Patients With De Novo Poor Risk Acute Myelogenous Leukemia[NCT00354276]Phase 285 participants (Anticipated)Interventional2006-05-31Active, not recruiting
A PedAL/EuPAL Phase 1/2 Trial of IMGN632 in Pediatric Patients With Relapsed or Refractory Leukemia[NCT05320380]Phase 1/Phase 20 participants (Actual)Interventional2023-08-01Withdrawn(stopped due to Withdrawn per CS0150757)
A Phase 1/1b Open-Label Study to Determine the Safety and Tolerability of ALRN-6924 Alone and in Combination With Cytarabine (Ara-C) in Patients With Relapsed/Refractory Acute Myeloid Leukemia or Advanced Myelodysplastic Syndrome With Wild-Type TP53[NCT02909972]Phase 155 participants (Actual)Interventional2016-09-30Completed
Primary Comparison of Liposomal Anthracycline Based Treatment Versus Conventional Care Strategies Before Allogeneic Stem Cell Transplantation in Patients With Higher Risk MDS and Oligoblastic AML[NCT04061239]Phase 2150 participants (Anticipated)Interventional2019-08-19Recruiting
Phase I Trial of Brentuximab Vedotin With Re-induction Chemotherapy in Patients With Relapsed, CD30 Expressing, Acute Myeloid Leukemia (AML)[NCT01830777]Phase 122 participants (Actual)Interventional2013-05-31Completed
A Phase I Dose Escalation Study With Expansion to Evaluate the Safety of SEPHB4-HSA in Combination With Cytarabine or Liposomal Vincristine in Patients With Relapsed or Refractory Acute Leukemia[NCT03519984]Phase 13 participants (Actual)Interventional2018-05-09Terminated(stopped due to Study drug supply issue)
Treatment of Peripheral T-cell Lymphoma With Aggressive Induction Chemotherapy Followed by Autologous Stem Cell Transplant Using Denileukin Diftitox (Ontak) for In-vivo Purging and Post-Transplant Therapy: A Multicenter Phase II Clinical Trial[NCT00632827]Phase 221 participants (Actual)Interventional2008-07-01Terminated(stopped due to Manufacturing shortage of both Diftitox and Doxil)
A Phase II Study of Intensive Consolidation and Stem Cell Mobilization Therapy With Ofatumumab, Etoposide, and High-dose Ara-C (OVA), Followed by Autologous Stem Cell Transplantation in High-risk Patients With Relapsed/Refractory Diffuse Large B-Cell Lymp[NCT01555541]Phase 219 participants (Actual)Interventional2012-05-25Completed
A Phase 2 Randomized Study Comparing Venetoclax and Azacitidine to Induction Chemotherapy for Newly Diagnosed Fit Adults With Acute Myeloid Leukemia[NCT04801797]Phase 2172 participants (Anticipated)Interventional2021-05-20Recruiting
A Prospective Randomized Phase I/II Study of Clofarabine (Clo) and Ara-C vs Clo and Ida vs Clo Plus Ida and Ara-C in Patients With First Relapse or First Salvage of Primary Refractory AML; and High-Grade MDS(>/= 10% Blasts); or CML in Myeloid Blasts Phase[NCT00067028]Phase 2116 participants (Actual)Interventional2003-12-31Completed
Liposomal Cytarabine and Daunorubicin (CPX-351) for Adults With Untreated High-Risk MDS and Non-APL AML at High Risk of Treatment-Related Mortality[NCT01804101]48 participants (Actual)Interventional2013-05-07Completed
BUSULFAN AND CYCLOPHOSPHAMIDE FOR CYTOREDUCTION OF PATIENTS WITH ACUTE AND CHRONIC LEUKEMIAS AND MYELODYSPLASTIC SYNDROMES UNDERGOING ALLOGENEIC BONE MARROW TRANSPLANTATION WHO CANNOT BE TREATED WITH TOTAL BODY IRRADIATION[NCT00002502]Phase 20 participants Interventional1992-07-31Completed
TREATMENT OF ADULT PATIENTS WITH RELAPSING ACUTE LYMPHOCYTIC LEUKEMIA, A MULTICENTER TRIAL[NCT00002532]Phase 20 participants Interventional1993-01-31Active, not recruiting
A PHASE II STUDY OF MITOXANTRONE AND HIGH-DOSE ARA-C FOLLOWED BY INTENSIVE CONSOLIDATION WITH CYCLOPHOSPHAMIDE AND ETOPOSIDE FOR MYELOID BLAST CRISIS OF CHRONIC MYELOGENOUS LEUKEMIA (CML)[NCT00002598]Phase 230 participants (Anticipated)Interventional1994-06-30Completed
A RANDOMIZED PHASE III TRIAL COMPARING DEXAMETHASONE WITH PREDNISONE IN INDUCTION TREATMENT AND BONE MARROW TRANSPLANTATION WITH INTENSIVE MAINTENANCE TREATMENT IN ADOLESCENT AND ADULT ACUTE LYMPHOBLASTIC LEUKEMIA (ALL-4)[NCT00002700]Phase 3392 participants (Anticipated)Interventional1995-08-31Completed
A Phase II Trial of T-Cell Depleted Marrow Grafts Combined With Infusions of G-CSF Stimulated, CD34 Ceprate Stem Cell Column Selected, E-Rosette Depleted Peripheral Blood Progenitor Cells Derived From HLA Haplotype Matched Related Donors for Patients With[NCT00002718]Phase 231 participants (Actual)Interventional1995-11-30Completed
A Phase II Study of Compound 506U78 in Patients With Refractory T-Cell Malignancies-POG/CCG Intergroup Study[NCT00002970]Phase 2148 participants (Actual)Interventional1997-06-30Completed
Multicenter Trial for Treatment of Acute Lymphocytic Leukemia in Adults (Pilot Study 06/99)[NCT00199056]Phase 4225 participants Interventional1999-10-31Completed
Treatment of Newly Diagnosed Childhood AML Using a Timed-Sequential Remission Induction and Consolidation Followed by Single Dose Melphalan With Peripheral Stem Cell Rescue: A POG Pilot Study[NCT00004056]Phase 135 participants (Actual)Interventional1999-10-31Completed
The Value of High Dose Versus Standard Dose ARA-C During Induction and of IL-2 After Intensive Consolidation/Autologous Stem Cell Transplantation in Patients (Age 15-60 Years) With Acute Myelogenous Leukemia. A Randomized Phase II Trial of the EORTC and t[NCT00004128]Phase 32,000 participants (Anticipated)Interventional1999-09-30Active, not recruiting
A Study to Determine the Safety and Efficacy of Using CD8-High Density Microparticles (CD8-HDM) to Deplete CD8+ Cells From Donor Lymphocyte Infusion in Order to Reduce Graft-versus-Host Disease (GvHD) Without Compromising an Anti-Leukemia Effect in Patien[NCT00004878]Phase 20 participants (Actual)InterventionalWithdrawn(stopped due to study never opened)
Chemotherapy (CT) Followed by Donor Lymphocyte Infusion (DLI) Plus Interleukin 2 (IL-2) for Patients With Relapse Acute Myeloid or Lymphoid Leukemia After Allogeneic Hematopoietic Transplant[NCT00005802]Phase 1/Phase 20 participants Interventional1999-06-30Completed
A Data Collection Study to Compare the Outcome for Children With Advanced Unilateral Retinoblastoma Treated With or Without Post-Enucleation Chemotherapy ± Radiotherapy on RB 2005 11 With Historical Controls Receiving no Additional Therapy[NCT00360750]0 participants Interventional2005-09-30Active, not recruiting
A Multi-Center, Open-Label, Phase 1/1b Study of Escalating Doses of RO5503781 Administered Orally as 1) a Single Agent, 2) In Combination With Cytarabine, or 3) With Cytarabine and Anthracycline and 4) Assessing PK and Safety of New Optimized Formulation [NCT01773408]Phase 1122 participants (Actual)Interventional2013-02-28Completed
A Single-Arm Phase I/II Study Evaluating the Safety and Clinical Efficacy Of the 2-nd Generation CD19 Autologous CAR T Cells on the CliniMACS Prodigy Automated Manufacturing Platform in Treatment of Paediatric And Young Adult Patients With Relapsed/Refrac[NCT03467256]Phase 1/Phase 218 participants (Anticipated)Interventional2018-05-14Active, not recruiting
Phase I-II Clinical Trial for the Evaluation of Brentuximab Vedotin Plus Etoposide, Solumoderin (Methylprednisolone), High Dose ARA-C (Cytarabine) and Cisplatin in the Transplant and Post-transplant Management for Relapsed or Refractory Classical Hodgkin [NCT02243436]Phase 1/Phase 267 participants (Actual)Interventional2014-11-11Completed
A Phase 1, Multicenter, Open-Label, Safety Study of AG-120 or AG-221 in Combination With Induction Therapy and Consolidation Therapy in Patients With Newly Diagnosed Acute Myeloid Leukemia With an IDH1 and/or IDH2 Mutation[NCT02632708]Phase 1153 participants (Actual)Interventional2015-12-31Active, not recruiting
Phase I/II Study of Lenalidomide Maintenance Following BEAM (+/- Rituximab) for Chemo-Resistant or High Risk Non-Hodgkin?s Lymphoma[NCT01035463]Phase 1/Phase 274 participants (Actual)Interventional2009-11-12Completed
Phase-II Study Evaluating Midostaurin in Induction, Consolidation and Maintenance Therapy Also After Allogeneic Blood Stem Cell Transplantation in Patients With Newly Diagnosed Acute Myeloid Leukemia Exhibiting a FLT3 Internal Tandem Duplication[NCT01477606]Phase 2451 participants (Actual)Interventional2012-05-31Completed
A Phase II Study Of Induction With Daunorubicin, Cytarabine, And Cyclosporine All By Continuous IV Infusion For Previously Untreated Non-M3 Acute Myeloid Leukemia (AML) In Patients Of Age 56 Or Older[NCT00066794]Phase 269 participants (Actual)Interventional2004-07-31Completed
A Phase I Study of Triapine in Combination With High Dose Ara-C (Hi-DAC) in Patients With Advanced Hematologic Malignancies[NCT00077181]Phase 148 participants (Actual)Interventional2004-01-31Completed
A Phase I Study Of VNP40101M And Cytarabine For Patients With Hematologic Malignancies[NCT00070538]Phase 10 participants Interventional2003-06-30Completed
A Phase I Study of CLAG Regimen in Combination With Imatinib Mesylate (Gleevec) in Refractory or Relapsed Leukemias[NCT00258271]Phase 118 participants (Anticipated)Interventional2005-03-31Completed
A Phase II Study of the Efficacy and Pharmacogenomics of Cladribine-based Salvage Chemotherapy in Patients With Relapse/Refractory and Secondary Acute Myeloid Leukemia (AML) and High Risk Myelodysplastic Syndrome (MDS)[NCT03150004]Phase 290 participants (Anticipated)Interventional2017-06-14Recruiting
A Phase 1/2 Open-Label Study to Assess the Safety, Tolerability, and Pharmacokinetics of Intravenous Infusion of MB07133 in Subjects With Unresectable Hepatocellular Carcinoma and Child-Pugh Class A Liver Function[NCT00073736]Phase 1/Phase 228 participants (Actual)Interventional2003-09-30Completed
A Phase I Trial Evaluating the Effect of the Addition of HMGCoA-Reductase Inhibition With Pravastatin to Salvage Chemotherapy Idarubicin-HDAC in Patients With Relapsed or Refractory Acute Myelogenous Leukemia[NCT00107523]Phase 10 participants Interventional2005-01-31Completed
Intensive Induction for Newly Diagnosed Acute Myelogenous Leukemia[NCT00274807]Phase 240 participants (Actual)Interventional2001-06-30Completed
A Phase III Randomized of Cloretazine™ (VNP40101M) and Cytosine Arabinoside (AraC) in Patients With Acute Myeloid Leukemia in First Relapse[NCT00112554]Phase 3420 participants (Anticipated)Interventional2005-03-31Completed
Phase 2 Study of Imatinib-Combined Chemotherapy for Newly Diagnosed BCR-ABL-Positive Acute Lymphoblastic Leukemia[NCT00130195]Phase 2100 participants (Actual)Interventional2002-09-30Completed
A Phase 2 Multi-Arm Study of Magrolimab Combinations in Patients With Myeloid Malignancies[NCT04778410]Phase 259 participants (Anticipated)Interventional2021-06-28Active, not recruiting
Phase 1 Study to Determine the Safety and Tolerability of Ziftomenib Combinations for the Treatment of KMT2A-rearranged or NPM1-mutant Relapsed/Refractory Acute Myeloid Leukemia[NCT06001788]Phase 1171 participants (Anticipated)Interventional2023-12-31Recruiting
Phase II Study Of Rituximab And Short Duration, High Intensity Chemotherapy With G-CSF Support In Previously Untreated Patients With Burkitt Lymphoma/Leukemia[NCT00039130]Phase 2105 participants (Actual)Interventional2002-05-31Completed
A Phase IB, Open-Label Study to Determine the Safety and Pharmacokinetics of Twice Daily Oral Dosing of PKC412 Administered in Combinations Sequentially and Concomitantly With Daunorubicin and Cytarabine for Standard Induction Therapy, and High Dose Cytar[NCT00093600]Phase 169 participants (Actual)Interventional2004-02-29Completed
A Phase I Study of R115777 (Zarnestra) in Combination With Induction Chemotherapy in Patients With Newly Diagnosed, High Risk Acute Myeloid Leukemia[NCT00124644]Phase 130 participants Interventional2006-03-31Terminated(stopped due to "Withdrawn due to toxicity problems")
A Single-arm Open-label Multicenter Clinical Study of Selinexor in Combination With HAD or CAG Rregimens in Relapsed or Refractory Acute Myeloid Leukemia[NCT05726110]Phase 350 participants (Anticipated)Interventional2023-01-29Recruiting
Multicentric Study for the Treatment of Children With Acute Lymphoblastic Leukemia[NCT00343369]550 participants (Anticipated)Interventional2003-01-31Recruiting
A Phase I And Pharmacological Trial Of 17-Allylamino -17-demethoxygeldanamycin (17-AAG) And Cytarabine In Refractory Leukemia And Myelodysplastic Syndrome[NCT00098423]Phase 142 participants (Actual)Interventional2004-11-30Completed
A Phase I Study Of Therapy With The Farnesyl Transferase Inhibitor R115777 (Zarnestra) Combined With Conventional Induction And Consolidation Chemotherapy For Previously Untreated Patients Over Age 55 With Acute Myeloid Leukemia (AML)[NCT00101153]Phase 124 participants (Actual)Interventional2007-04-30Completed
Phase I Study of Adoptive Immunotherapy With CD8 Minor Histocompatibility (H) Antigen-Specific CTL Clones for Patients With Relapsed of AML or ALL After Allogeneic Hematopoietic Stem Cell Transplant[NCT00107354]Phase 10 participants Interventional1998-12-31Completed
Randomized Prospective Study of Adding Lomustine to Idarubicin and Cytarabine for Induction Chemotherapy and Adding Intermediate Dose Cytarabine to Consolidation in Older Patients With Acute Myeloid Leukaemia[NCT00480064]Phase 3360 participants (Actual)Interventional1995-07-31Completed
Phase II Study: 2-Chlorodeoxyadenosine (2-CdA) and Cytarabine (Ara-C) in Idiopathic Hypereosinophilic Syndrome (HES)[NCT00483067]Phase 213 participants (Actual)Interventional1998-03-31Completed
A Phase I Study Investigating the Combination of RAD001 With Standard Induction and Consolidation Therapy in Older Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)[NCT01154439]Phase 111 participants (Actual)Interventional2010-10-31Completed
Phase I Study to Evaluate the Safety and Efficacy of NK Cell Therapy in Acute Myeloid Leukemia (AML).[NCT05987696]Phase 1102 participants (Anticipated)Interventional2023-08-10Not yet recruiting
Pilot Study in AIDS-Related Lymphomas[NCT00002524]Phase 246 participants (Actual)Interventional1993-06-30Completed
Phase 2 Study of 5 Days Azacytidine Priming Prior to Fludarabine, Cytarabine and Granulocyte-Colony Stimulating Factor (G-CSF) Combination for Patients With Relapsed or Refractory AML[NCT02275663]Phase 237 participants (Anticipated)Interventional2014-12-31Recruiting
Phase II Study on Gemtuzumab Ozogamicin in Combination With All-trans-Retinoic Acid, High-dose Cytarabine and Mitoxantrone in Patients With Primary Refractory Acute Myeloid Leukemia[NCT00143975]Phase 295 participants (Actual)Interventional2004-06-30Completed
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
HLA-mismatched Allogeneic Cellular Therapy (HMMACT) After Chemotherapy in High Risk Acute Myeloid Leukemia[NCT01801046]Phase 110 participants (Actual)Interventional2013-03-06Terminated(stopped due to Insufficient Accrual)
A Phase 2 Study of Fludarabine, Cytarabine, Filgrastim-sndz,Gemtuzumab Ozogamicin and Idarubicin in Newly Diagnosed Core Binding Factor Associated Acute Myelogenous Leukemia[NCT00801489]Phase 2270 participants (Anticipated)Interventional2007-04-04Recruiting
A Randomized, Multicenter, Open-Label Study of Single Dose Filgrastim-SD/01 Versus Daily Filgrastim Following ESHAP Chemotherapy for Non-Hodgkin's Lymphoma[NCT00004192]Phase 260 participants (Anticipated)Interventional2000-05-01Completed
A Randomized Trial to Evaluate Early High Dose Therapy and Autologous Bone Marrow Transplantation as Part of Planned Initial Therapy for Poor Risk Intermediate/High Grade NHL[NCT00003578]Phase 3500 participants (Anticipated)Interventional1993-01-31Active, not recruiting
A Randomized Phase II Study of the Combination of 5-Azacytidine With Valproic Acid (VPA) Versus Low-Dose Ara-C in Patients With AML/MDS Not Eligible for Other Studies[NCT00382590]Phase 211 participants (Actual)Interventional2005-08-31Completed
Randomized Trial Using Standard Dose Versus High Dose Rituximab in Addition to Autologous Transplantation With BEAM for Patients With Diffuse Large B Cell Lymphomas[NCT00472056]Phase 293 participants (Actual)Interventional2005-03-31Completed
A Phase I/II Trial of ABT-751 Combined With Dexamethasone, PEG-asparaginase, and Doxorubicin in Relapsed Acute Lymphoblastic Leukemia (ALL)[NCT00439296]Phase 1/Phase 29 participants (Actual)Interventional2006-05-22Terminated(stopped due to The study was stopped due to poor accrual and lack of funding.)
"Randomized Phase I/II Study of 5-Azacytidine in Combination With Cytosine Arabinoside in Patients With Relapsed/Refractory Acute Myelogenous Leukemia or High Risk Myelodysplastic Syndrome - SPORE"[NCT00569010]Phase 1/Phase 236 participants (Actual)Interventional2005-12-31Completed
Phase IIB, Multicenter, Randomized, Open Label Trial of CPX-351 (Cytarabine:Daunorubicin) Liposome Injection Versus Cytarabine and Daunorubicin in Patients With Untreated AML 60-75 Years of Age.[NCT00788892]Phase 2126 participants (Actual)Interventional2008-10-31Completed
Study of Oral Clofarabine Plus Low-dose Cytarabine in Previously Treated AML and High-Risk MDS Patients at Least 60 Years of Age[NCT00839982]Phase 1/Phase 235 participants (Actual)Interventional2008-11-30Completed
Intravenous Erwinase for Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia and Allergy to E. Coli Asparaginase (IND 104224)[NCT00928200]Phase 11 participants (Actual)Interventional2009-04-13Terminated(stopped due to Study was terminated due to lack of accrual.)
Sequential Autologous HCT / Nonmyeloablative Allogeneic HCT Using Related, HLA-Haploidentical Donors for Patients With High-Risk Lymphoma, Multiple Myeloma, or Chronic Lymphocytic Leukemia[NCT01008462]Phase 216 participants (Actual)Interventional2010-03-18Completed
A Phase 2 Study of Clofarabine, Idarubicin, Cytarabine, Vincristine, and Corticosteroids for Patients With Newly Diagnosed or Relapsed Mixed Phenotype Acute Leukemia[NCT02135874]Phase 218 participants (Actual)Interventional2014-10-27Completed
Phase II Study of Hyper-CVAD Plus Nelarabine in Previously Untreated T-ALL and Lymphoblastic Lymphoma[NCT00501826]Phase 2160 participants (Anticipated)Interventional2007-07-11Recruiting
Multicenter,Open Label,Phase 2 Clinical Study of Venetoclax Combined With CACAG Regimen in the Treatment of Newly Diagnosed Acute Myeloid Leukemia[NCT06068621]Phase 2200 participants (Anticipated)Interventional2023-08-01Recruiting
A Phase 1b Study With Expansion Cohort of Escalating Doses of KRT-232 (AMG 232) Administered in Combination With Standard Induction Chemotherapy (Cytarabine and Idarubicin) in Newly Diagnosed Acute Myelogenous Leukemia (AML)[NCT04190550]Phase 124 participants (Anticipated)Interventional2021-02-04Active, not recruiting
A Pilot, Pharmacodynamic Correlate, Multi-Institutional Trial of Sirolimus in Combination With Chemotherapy (Mitoxantrone, Etoposide, Cytarabine) for the Treatment of High Risk, Acute Myelogenous Leukemia[NCT01184898]36 participants (Actual)Interventional2010-07-31Completed
Phase II Study of the Hyper - CVAD Regimen in Combination With Ofatumumab as Frontline Therapy for Patients With CD-20 Positive Acute Lymphoblastic Leukemia[NCT01363128]Phase 272 participants (Actual)Interventional2011-07-12Completed
Autologous Stem Cell Transplant In Patients With Hodgkin Lymphoma (HL) and Non-Hodgkin Lymphomas (NHL)[NCT03125642]Phase 2150 participants (Anticipated)Interventional2017-04-20Recruiting
BLockade of PD-1 Added to Standard Therapy to Target Measurable Residual Disease in Acute Myeloid Leukemia 1 (BLAST MRD AML-1): A Randomized Phase 2 Study of the Anti-PD-1 Antibody Pembrolizumab in Combination With Conventional Intensive Chemotherapy as F[NCT04214249]Phase 2124 participants (Anticipated)Interventional2021-02-17Recruiting
Treatment Of Newly Diagnosed Adult Acute Lymphoblastic Leukemia With Intensified Post Remission Therapy Containing PEG-Asparaginase.[NCT00184041]Phase 247 participants (Actual)Interventional2004-07-31Completed
Early Assessment of Treatment Response in AML Using FLT PET/CT Imaging[NCT02392429]Phase 257 participants (Anticipated)Interventional2016-04-20Active, not recruiting
Phase II Study to Evaluate the Efficacy of Upfront Obinutuzumab in Mantle Cell Lymphoma Patients Treated by DHAP Followed by Autologous Transplantation Plus Obinutuzumab Maintenance Then MRD Driven Maintenance[NCT02896582]Phase 286 participants (Actual)Interventional2016-10-31Active, not recruiting
Total Therapy Study XIV for Newly Diagnosed Patients With Acute Lymphoblastic Leukemia[NCT00187005]Phase 353 participants (Actual)Interventional1998-07-31Terminated(stopped due to Toxicity)
A Phase I-II Study of the Combination of Ruxolitinib or Dasatinib With Chemotherapy in Patients With Philadelphia Chromosome (Ph)-Like Acute Lymphoblastic Leukemia (ALL)[NCT02420717]Phase 211 participants (Actual)Interventional2015-07-15Terminated(stopped due to Study was closed early due to low accrual and lack of response.)
Phase I/II Study of Arsenic Trioxide in Combination With Cytosine Arabinoside in Patients With High-risk Myelodysplastic Syndrome and Poor-prognosis Acute Myelogenous Leukemia[NCT00195104]Phase 1/Phase 287 participants (Actual)Interventional2003-09-17Completed
Randomized Phase II Trial on Primary Chemotherapy With High-dose Methotrexate, Alone or Associated With High-dose Cytarabine, Followed by Response- and Age-tailored Radiotherapy for Immunocompetent Patients With Newly Diagnosed Primary Central Nervous Sys[NCT00210314]Phase 279 participants (Actual)Interventional2003-07-31Completed
A Phase 1 Study of Revumenib, Azacitidine, and Venetoclax in Pediatric and Young Adult Patients With Refractory or Relapsed Acute Myeloid Leukemia[NCT06177067]Phase 124 participants (Anticipated)Interventional2024-01-31Not yet recruiting
Prospective Pilot Trial of Arsenic Trioxide (Trisenox®) in Combination With Cytosine Arabinoside in Patients With Advanced or Transformed Myelofibrosis[NCT00572065]Early Phase 121 participants (Actual)Interventional2008-02-29Completed
Study of the Efficacy and Safety of Venetoclax Plus Azacytidine Versus Daunorubicin Plus Cytarabine in Adult Acute Myeloid Leukemia (AML) Patients With Adverse Risk Features[NCT05939180]Phase 2/Phase 3108 participants (Anticipated)Interventional2023-07-01Recruiting
A Randomized Trial Assessing the Roles of AraC in Newly Diagnosed Acute Promyelocytic Leukemia (APL)[NCT00591526]Phase 3250 participants (Actual)Interventional2000-06-30Completed
Mitoxantrone, Etoposide, and Cytarabine (MEC) Following Epigenetic Priming With Decitabine in Adults With Relapsed/Refractory Acute Myeloid Leukemia (AML) or High-Risk Myelodysplastic Syndromes (MDS): A Phase 1/2 Study[NCT01729845]Phase 1/Phase 252 participants (Actual)Interventional2012-12-20Completed
Phase III Trial in AML Comparing Standard-dose vs High-dose Remission Induction and, Within a Risk-oriented Postremission Strategy, Autologous Blood Stem Cell Transplantation vs Blood Stem Cell-supported Multicycle High-dose Program[NCT00495287]Phase 3573 participants (Actual)Interventional2006-11-30Completed
A Phase 1 Study of Azacitidine in Combination With MEC (Mitoxantrone, Etoposide, Cytarabine) in Relapsed and Refractory Acute Myeloid Leukemia[NCT01249430]Phase 124 participants (Actual)Interventional2011-01-20Completed
A Pilot Study of the Efficacy of the Chop-Montak Regimen in Patients With Newly Diagnosed Peripheral T Cell Lymphoma[NCT00513188]0 participants (Actual)Interventional2007-02-28Withdrawn
Phase II Trial of CLAG-M in Relapsed ALL[NCT01513603]Phase 250 participants (Anticipated)Interventional2012-01-31Recruiting
Intergroup Trial for Children or Adolescents With B-Cell Non Hodgkin Lymphoma or B-Acute Leukemia: Evaluation of Rituximab Efficacy and Safety in High Risk Patients - Phase III Trial[NCT01516580]Phase 3482 participants (Actual)Interventional2011-12-31Active, not recruiting
A Phase II Study of Microtransplantation in Patients With Refractory or Relapsed Hematologic Malignancies[NCT02433483]Phase 24 participants (Actual)Interventional2015-05-22Terminated(stopped due to The study was closed due to poor accrual and because of competing protocols.)
Phase I/II Study on Cytarabine and Idarubicin Combined With Escalating Doses of Clofarabine as Induction Therapy in Patients With Acute Myeloid Leukemia and High Risk for Induction Failure (AMLSG 17-10)[NCT01534702]Phase 1/Phase 260 participants (Anticipated)Interventional2012-01-31Recruiting
[NCT01540812]418 participants (Actual)Observational2012-02-29Completed
Hyper-CVAD With Liposomal Vincristine (Hyper-CMAD) in Acute Lymphoblastic Leukemia[NCT01319981]Phase 231 participants (Actual)Interventional2013-03-05Completed
A RANDOMIZED TRIAL OF UNMODIFIED VERSUS T-CELL DEPLETED ALLOGENEIC HLA-IDENTICAL BONE MARROW TRANSPLANTATION FOR THE TREATMENT OF ACUTE LEUKEMIAS[NCT00002534]Phase 30 participants Interventional1993-05-31Completed
A Phase III Study of Large Cell Lymphomas in Children and Adolescents: Comparison of APO vs APO + IDMTX/HDARA-C and Continuous vs Bolus Infusion of Doxorubicin[NCT00002618]Phase 3242 participants (Anticipated)Interventional1994-12-31Completed
Randomized Comparisons of Oral Mercaptopurine vs Oral Thioguanine and IT Methotrexate vs ITT for Standard Risk Acute Lymphoblastic Leukemia[NCT00002744]Phase 31,970 participants (Actual)Interventional1996-05-31Completed
Induction Intensification in Infant ALL: A Children's Oncology Group Study[NCT00002756]Phase 2221 participants (Actual)Interventional1996-06-30Completed
Phase II Study of Intrathecal Therapy With DepoCyt for Active Lymphomatous or Leukemic Meningitis[NCT00523939]Phase 24 participants (Actual)Interventional2006-06-30Terminated(stopped due to Low accrual.)
Phase III Study of Combination Chemotherapy in Children With T Cell and Pre-B Cell Non-Hodgkin's Lymphoma[NCT00003650]Phase 3179 participants (Actual)Interventional1997-02-28Completed
Evaluating the MBVP Chemotherapy Schedule Followed by Consolidating Radiotherapy in Non-AIDS Related Primary Central Nervous System Lymphoma (NAPCL)[NCT00003061]Phase 250 participants (Anticipated)Interventional1997-07-31Completed
A Phase I Dose Escalation Study of Intrathecal DepoFoam Encapsulated Cytarabine (DTC 101) in Pediatric Patients With Advanced Meningeal Malignancies[NCT00003073]Phase 115 participants (Anticipated)Interventional1997-02-28Active, not recruiting
BEAM Plus Iodine-131 Anti-B1 Antibody and Autologous Hematopoietic Stem Cell Transplantation for Treatment of Recurrent Diffuse Large B-Cell Non-Hodgkin's Lymphoma[NCT00006695]Phase 250 participants (Actual)Interventional2000-04-01Completed
A Pilot Study of Dose Intensification of Methotrexate and Cyclophosphamide in Patients With Advanced-Stage (III/IV) Small Non-Cleaved Non-Hodgkins Lymphoma and B-Cell All- A Limited Institution Phase III Pilot Study[NCT00003217]Phase 120 participants (Actual)Interventional1998-03-31Completed
Transplantation of Unrelated Donor Hematopoietic Stem Cells for the Treatment of Hematological Malignancies[NCT00281879]Phase 2200 participants (Actual)Interventional2006-02-28Terminated
MULTICENTRE TRIAL OF INTENSIFIED THERAPY FOR ADULT ALL (O5/93)[NCT00002531]Phase 20 participants Interventional1993-01-31Active, not recruiting
Phase II Trial of Sequential Modification of Immunosuppression, Interferon Alpha, and Promace-Cytabom For Treatment of Post-Cardiac Transplant Lymphoproliferation.[NCT00002657]Phase 220 participants (Actual)Interventional1995-05-31Completed
A Study of Modified Augmented BFM Therapy for Infants With Acute Lymphoblastic Leukemia[NCT00022126]Phase 26 participants (Actual)Interventional2002-11-30Completed
A Phase II Study of Bevacizumab (rhuMab VEGF, NSC 704865), Idarubicin and Cytarabine in Patients With Chronic Myeloid Leukemia in Blast Phase[NCT00023920]Phase 260 participants (Actual)Interventional2001-07-31Terminated(stopped due to Administratively complete.)
A Randomized Trial Of BEAM Plus PBSCT Versus Single Agent High-Dose Therapy Followed By BEAM Plus PBSCT In Patients With Relapsed Hodgkin's Disease[NCT00025636]Phase 3220 participants (Anticipated)Interventional2001-07-31Active, not recruiting
Nonmyeloablative Allogeneic Hematopoietic Cell Transplantation From HLA Matched Sibling Donors For Treatment Of Patients With High Risk Acute Lymphocytic Leukemia In Complete Remission[NCT00027547]Phase 1/Phase 20 participants Interventional2001-07-31Completed
A Phase II Study of CPX-351 for Treatment of AML or Higher Risk MDS Relapsed or Refractory to Prior Therapy With Hypomethylating (HMA) Agent[NCT02019069]Phase 211 participants (Actual)Interventional2014-02-03Completed
A Randomised Phase III Study On The Effect Of The Chimeric Anti-CD20 Monoclonal Antibody (Mabthera) During Sequential Chemotherapy Followed By Autologous Stem Cell Transplantation In Patients With Relapse B-Cell Non-Hodgkin Lymphoma(HOVON 44 STUDY)[NCT00012051]Phase 3340 participants (Anticipated)Interventional2000-09-30Completed
Phase I Trial of mBACOD and Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) in AIDS-Associated Large Cell, Immunoblastic, and Small Non-cleaved Lymphoma[NCT00000689]Phase 118 participants InterventionalCompleted
A Phase I/II Trial of STI571 and High-Dose Cytarabine in Myeloid Blast Crisis of Chronic Myeloid Leukemia[NCT00015834]Phase 1/Phase 246 participants (Actual)Interventional2001-05-31Completed
International Collaborative Treatment Protocol for Infants Under One Year With Acute Lymphoblastic Leukemia[NCT00015873]Phase 3350 participants (Anticipated)Interventional1999-05-31Completed
A Phase II Study of the Recombinant Human Monoclonal Anti-Vascular Endothelial Growth Factor Antibody (rhuMAB VEGF) Bevacizumab (NSC #704865, IND # 7,921) Administered in Times Sequential Combination With Cytosine Arabinoside (Ara-C) and Mitoxantrone for [NCT00015951]Phase 20 participants Interventional2001-04-30Completed
The Use of Modified BFM +/- Compound 506U78 (Nelarabine) (NSC# 686673, IND #52611) in an Intensive Chemotherapy Regimen for the Treatment of T-Cell Leukemia[NCT00016302]100 participants (Actual)Interventional2001-04-30Completed
Treatment of Mantle Cell Lymphomas at Advanced Stages: Prospective Randomized Comparison of Myeloablative Radiochemotherapy Followed by Blood Stem Cell Transplantation Versus Maintenance With Interferon Alpha in First Remission After Initial Cytoreductive[NCT00016887]Phase 30 participants Interventional2000-12-31Active, not recruiting
A Clinicopathological Study In Burkitts's And Burkitt-Like Non-Hodgkin's Lymphoma[NCT00040690]Phase 2120 participants (Anticipated)Interventional2008-11-30Completed
Autologous Peripheral Blood Stem Cell Transplant for Patients With Lymphoma[NCT00345865]Phase 2473 participants (Actual)Interventional2005-08-24Completed
A Phase I Study of Triapine and Cytarabine in Patients With Hematologic Malignancies[NCT00064090]Phase 10 participants Interventional2003-03-31Completed
Treatment of Newly Diagnosed Childhood Acute Myeloid Leukemia (AML) Using Intensive MRC-Based Therapy and Gemtuzumab Ozogamicin (GMTZ): A COG Pilot Study[NCT00070174]Phase 2350 participants (Actual)Interventional2003-12-31Completed
Combination Chemotherapy (Methotrexate, Procarbazine And CCNU), Intraventricular Cytarabine And Methotrexate, +/- Intra-Ocular Chemotherapy For Patients With Primary Central Nervous System Lymphoma[NCT00074191]Phase 21 participants (Actual)Interventional2000-01-31Completed
Open Label, Phase II Dosing Study of Ara-C Chemotherapy in Combination With EL625 and Idarubicin in Refractory and Relapsed Acute Myelogenous Leukemia (AML)[NCT00074737]Phase 253 participants (Actual)Interventional2004-04-30Completed
Idarubicin and Ara-C in Combination With Gemtuzumab-Ozogamicin (IAGO) for Young Untreated Patients, Without an HLA Identical Sibling, With High Risk MDS or AML Developing After a Preceding Period With MDS During 6 Months Duration: A Phase II Study[NCT00077116]Phase 231 participants (Actual)Interventional2003-11-30Completed
Randomized Study of ICE Plus RITUXIMAB Versus DHAP Plus Rituximab in Previously Treated Patients With Diffuse Large B-cell Lymphoma, Followed by Randomized Maintenance With Rituximab[NCT00137995]Phase 3481 participants (Actual)Interventional2003-06-30Completed
Immunotherapy With Ex Vivo-Expanded Cord Blood-Derived NK Cells Combined With Rituximab High-Dose Chemotherapy and Autologous Stem Cell Transplant for B-Cell Non-Hodgkin's Lymphoma[NCT03019640]Phase 222 participants (Actual)Interventional2017-10-10Completed
Phase I-II Study of Idarubicin, Cytarabine, and Sorafenib (BAY43-9006), an Oral Vascular Endothelial Growth Factor (VEGF), Rapidly Accelerated Fibrosarcoma (RAF) and FMS-like Tyrosine Kinase 3 (FLT3), in Patients With High-risk MDS and AML[NCT00542971]Phase 1/Phase 278 participants (Actual)Interventional2007-10-31Completed
Treatment Protocol for T-Cell and B-Precursor Cell Lymphoblastic Lymphoma of the European Inter-group Co-operation on Childhood Non-Hodgkin-Lymphoma (EICNHL)[NCT00275106]Phase 3600 participants (Anticipated)Interventional2004-09-30Terminated(stopped due to Withdrawn due to an excess of toxic deaths)
A Phase I/II Study of Flavopiridol (NSC 649890, IND 46,211) in Timed Sequential Combination With Cytosine Arabinoside (Ara-C) and Mitoxantrone for Adults With Poor-Risk Acute Leukemias[NCT00016016]Phase 1/Phase 253 participants (Anticipated)Interventional2001-02-28Completed
An Open-Label, Phase I/II, Multicenter Dose Escalation Study of Zosuquidar, Daunorubicin, and Cytarabine in Patients Ages 55-75 With Newly Diagnosed Acute Myeloid Leukemia[NCT00129168]Phase 1/Phase 2100 participants (Anticipated)Interventional2005-08-31Completed
PILOT MULTINATIONAL PROTOCOLS IN ACUTE LYMPHOBLASTIC LEUKEMIA AND DIFFUSE NON-HODGKIN'S LYMPHOMA[NCT00018954]Phase 20 participants Interventional1992-10-31Completed
A Phase II Study Of Intensive Induction Chemotherapy Followed By Autologous Stem Cell Transplantation Plus Immunotherapy For Mantle Cell Lymphoma[NCT00020943]Phase 279 participants (Actual)Interventional2001-06-30Completed
A Children's Oncology Group Pilot Study for the Treatment of Very High Risk Acute Lymphoblastic Leukemia in Children and Adolescents (Imatinib (STI571, GLEEVEC) NSC#716051)[NCT00022737]Phase 3220 participants (Actual)Interventional2002-10-31Completed
Phase II Window Evaluation of the Farnesyl Transferase Inhibitor (R115777) Followed by 13-CIS Retinoic Acid, Cytosine Arabinoside and Fludarabine Plus Hematopoietic Stem Cell Transplantation in Children With Juvenile Myelomonocytic Leukemia[NCT00025038]Phase 2100 participants (Actual)Interventional2001-06-30Completed
A Dose Finding Study of the Safety of Gemtuzumab Ozogamicin Combined With Conventional Chemotherapy for Patients With Relapsed or Refractory Acute Myeloid Leukemia[NCT00028899]Phase 147 participants (Actual)Interventional2002-07-31Completed
A Randomized Clinical Study to Determine the Patient Benefit and Safety of Depocyt (Cytarabine Liposome Injection) for the Treatment of Neoplastic Meningitis[NCT00029523]Phase 4100 participants Interventional2001-04-30Completed
A Dose Ranging Study of the Safety and Efficacy of Gemtuzumab Ozogamicin (GO) Given in Combination With Cytarabine in Relapsed or Refractory Patients and Alder De Novo Patients With Acute Myeloid Leukemia.[NCT00037596]Phase 20 participants Interventional2000-08-31Completed
Campath 1H (Alemtuzumab) Combined With High-Dose Therapy and Autologous Stem Cell Transplantation in Chronic Lymphocytic Leukemia[NCT00276809]Phase 230 participants (Anticipated)Interventional2001-06-30Completed
A Phase I Study of G3139 ( NSC # 683428) in Combination With Cytarabine and Daunorubicin in Previously Untreated Patients With Acute Myeloid Leukemia (AML)>= 60 Years of Age[NCT00039117]Phase 132 participants (Actual)Interventional2002-04-30Completed
A Randomized Study of Two Methods of CNS Prophylaxis in Patients With Acute Lymphoblastic Leukemia[NCT00019409]Phase 30 participants (Actual)Interventional1999-10-31Withdrawn
A Phase II Study of Gemtuzumab Ozogamicin (Mylotarg) and Standard Dose ARA-C for Patients With Relapsed Acute Myeloid Leukemia (AML)[NCT00049179]Phase 233 participants (Actual)Interventional2003-04-30Completed
Intensive Induction Therapy for Children With Acute Lymphoblastic Leukemia (ALL) Who Experience a Bone Marrow Relapse[NCT00049569]126 participants (Anticipated)Interventional2003-01-31Completed
A Phase II Study Of Daunomycin And ARA-C, Both Given By Continous IV Infusion For Previously Untreated Non-M3 Acute Myeloid Leukemia (AML) In Patients Of Age 56 Or Older[NCT00023777]Phase 271 participants (Actual)Interventional2001-08-31Completed
Gemtuzumab Ozogamicin (GO) Combined With Standard Intensive Chemotherapy Versus Standard Intensive Chemotherapy Alone For Induction/Consolidation In Patients 61-75 Years Old With Previously Untreated AML: A Randomized Phase III Trial (AML-17) Of The EORTC[NCT00052299]Phase 3472 participants (Actual)Interventional2002-09-30Completed
A Dose-ranging Phase I/II Study of STI571 in Combination With Cytarabin in Patients With First Chronic Phase Chronic Myeloid Leukemia[NCT00028847]Phase 1/Phase 260 participants (Anticipated)Interventional2001-04-30Active, not recruiting
A Phase II Study of Rituximab (IND #7028) and Ifosfamide, Carboplatin and Etoposide (ICE) Chemotherapy in Children With Recurrent/Refractory B-cell (CD20+) Non-Hodgkin Lymphoma and B-cell Acute Lymphoblastic Leukemia[NCT00058461]Phase 282 participants (Anticipated)Interventional2003-11-30Terminated
Cytarabine Plus Continuous Infusion Daunorubicin Induction Therapy for Adults With Acute Myeloid Leukemia: A Feasibility Study With Cardiac MRI Monitoring[NCT02971397]Phase 240 participants (Actual)Interventional2016-11-30Active, not recruiting
A Phase II Trial of BBR 2778 in Combination With Cytarabine, Methylprednisolone and Cisplatin (BSHAP) as Salvage in Patients With Relapsed Aggressive Non-Hodgkin's Lymphoma[NCT00069966]Phase 20 participants Interventional2003-04-30Active, not recruiting
Randomised Controlled Study Comparing Fast Track and Standard Care Protocol on the Functional Outcomes and Hospital Stay of Total Knee Arthroplasty[NCT03869996]64 participants (Anticipated)Interventional2019-02-25Recruiting
A Phase I Study of GTI2040 (NSC 722929; IND 67368) in Combination With High-dose Cytarabine in Refractory or Relapsed Acute Myeloid Leukemia (AML)[NCT00070551]Phase 151 participants (Actual)Interventional2003-09-30Completed
Protocol for the Treatment of Adults Aged NCT00209833]Phase 2/Phase 3200 participants Interventional1999-01-31Active, not recruiting
A Phase III Prospective Randomized Comparison of Imatinib at a Dose of 400mg in Combination With Peg-Interferon-alpha2a (Peg-IFNa2a) or Cytarabine (Ara-C)Versus Imatinib at a Dose of 600mg Versus Imatinib a Dose of 400mg for Previously Untreated Chronic M[NCT00219739]Phase 3789 participants (Actual)Interventional2003-09-30Completed
Combination Chemotherapy (Methotrexate, Cyclophosphamide, And Etoposide Phosphate) Delivered In Conjunction With Osmotic Blood-Brain Barrier Disruption (BBBD), With Intraventricular Cytarabine +/- Intra-Ocular Chemotherapy, In Patients With Primary Centra[NCT00074178]Phase 222 participants (Actual)Interventional2000-01-31Completed
Phase 2 Study of Applying Pediatric Regimens to Younger Adult Patients With BCR-ABL-Negative Acute Lymphoblastic Leukemia[NCT00131053]Phase 2120 participants (Anticipated)Interventional2002-09-30Recruiting
T-Cell Depleted, Reduced-Intensity Allogeneic Stem Cell Transplantation From Haploidentical Related Donors For Hematologic Malignancies[NCT00080925]Phase 120 participants (Anticipated)Interventional2004-02-29Completed
A Phase I Trial of BBR 2778 in Combination With Cytarabine, Methylprednisolone and Cisplatin in the Treatment of Patients With Relapsed or Refractory Aggressive Non-Hodgkin's Lymphoma[NCT00053105]Phase 10 participants Interventional2002-02-28Active, not recruiting
Irinotecan And Cytarabine In Refractory or Relapsed Acute Myeloid Leukemia And In Chronic Myelogenous Leukemia In Myeloid Blast Transformation: Efficacy And In Vitro Correlates[NCT00053144]Phase 10 participants Interventional1999-11-30Completed
Treatment of Acute Lymphoblastic Leukemia in Children[NCT00165178]Phase 3498 participants (Actual)Interventional2000-09-30Completed
Treatment Optimization Trial in Chronic Myeloid Leukemia (CML) - Randomized Controlled Comparison of Imatinib vs. Imatinib/Interferon-alpha vs. Imatinib/Low-Dose AraC vs. Interferon-alpha Standard Therapy and Determination of the Role of Allografting in N[NCT00055874]Phase 31,551 participants (Actual)Interventional2002-06-30Completed
AML96 - Risk-Adapted and Randomized Postremission-Therapy for Adult Acute Myeloid Leukemia Patients. A Cooperative AML-Study of the German SHG-Study Group.[NCT00180115]Phase 4400 participants Interventional1996-02-29Completed
A Phase I Trial Combining IDEC-Y2B8 And High-Dose Beam Chemotherapy With Hematopoietic Progenitor Cell Transplant In Patients With Relapsed Or Refractory B-Cell Non-Hodgkin's Lymphoma[NCT00058292]Phase 144 participants (Actual)Interventional2000-04-30Completed
Prospective Study of the Diagnosis and Treatment of Myelodysplastic Syndromes (MDS) in Childhood[NCT00047268]Phase 30 participants Interventional1998-07-31Active, not recruiting
A Multicenter, Phase 2 Study, to Evaluate Safety and Efficacy of an Acute Lymphoblastic Leukemia (ALL) Intensive Chemotherapy for Adult Lymphoblastic Lymphoma (LL).[NCT00195871]Phase 2155 participants (Actual)Interventional2004-02-29Active, not recruiting
A Randomized, Multicenter Open Label Phase II Study to Determine the Safety and Efficacy of Induction Therapy With Imatinib in Comparison With Standard Induction Chemotherapy in Elderly (> 55 Years) Patients With Ph Positive Acute Lymphoblastic Leukemia ([NCT00199186]Phase 20 participants Interventional2002-03-31Recruiting
ALL Adult Consortium Trial: Adult ALL Trial[NCT00476190]Phase 2112 participants (Anticipated)Interventional2007-04-30Active, not recruiting
Azacitidine Compared to Conventional Chemotherapy in Consolidation of Elderly Patients (65 Years or Older) With AML in First Complete Remission[NCT01794169]Phase 2130 participants (Anticipated)Interventional2013-03-31Terminated(stopped due to Poor recruitment)
An Intergroup Phase II Clinical Trial for Adolescents and Young Adults With Untreated Acute Lymphoblastic Leukemia (ALL)[NCT00558519]Phase 2318 participants (Actual)Interventional2008-03-12Active, not recruiting
Epigenetic Reprogramming in Relapse AML: A Phase 1 Study of Decitabine and Vorinostat Followed by Fludarabine, Cytarabine and G-CSF (FLAG) in Children and Young Adults With Relapsed/Refractory AML[NCT02412475]Phase 13 participants (Actual)Interventional2015-02-21Terminated(stopped due to We opened a competing study with the TACL consortium)
Medical Research Council Working Party on Leukaemia in Children UK National Lymphoblastic Leukaemia (ALL) Trial[NCT00003437]Phase 31,800 participants (Anticipated)Interventional1997-01-31Active, not recruiting
Treatment of Children With Down Syndrome (DS) and Acute Myeloid Leukemia (AML), Myelodysplastic Syndrome (MDS), and Transient Myeloproliferative Disorder (TMD): A Phase III Group-Wide Study[NCT00003593]Phase 3254 participants (Actual)Interventional1999-06-30Completed
A Phase I-II Study of Oxaliplatin, Fludarabine, Cytarabine, and Rituximab in Patients With Richter's Transformation, Prolymphocytic Leukemia, Aggressive, Relapsed or Refractory B-Cell Chronic Lymphocytic Leukemia[NCT00472849]Phase 1/Phase 292 participants (Actual)Interventional2007-05-31Completed
Phase I/II Trial of VELCADE® (Bortezomib) in Combination With Mitoxantrone and Etoposide for Relapsed or Refractory Acute Leukemias[NCT00410423]Phase 1/Phase 255 participants (Actual)Interventional2006-01-31Completed
A Randomized Multi-Center Treatment Study (COALL 08-09) to Improve the Survival of Children With Acute Lymphoblastic Leukemia on Behalf of the German Society of Pediatric Hematology and Oncology[NCT01228331]Phase 2/Phase 3745 participants (Actual)Interventional2010-10-31Active, not recruiting
A Prospective, Multicenter, Single Arm Clinical Study to Evaluate Efficacy of HAD Induction With Intensified Cytarabine in Newly-diagnosed CEBPA Double Mutated Acute Myeloid Leukemia[NCT04415008]Phase 240 participants (Anticipated)Interventional2020-06-01Recruiting
Efficacy of Intermediate-Dose Cytarabine Induction Regimen in Adult AML[NCT03021330]Phase 31,100 participants (Anticipated)Interventional2017-02-08Recruiting
Cytosine Arabinoside and Mitoxantrone for Patients With Juvenile Myelomonocytic Leukemia Receiving Repeat Stem Cell Transplantation[NCT00609739]Phase 1/Phase 21 participants (Actual)Interventional1999-06-30Terminated(stopped due to Low accrual)
T-Regulatory Cell Kinetics Post Transplant For Patients Undergoing Matched Sibling Stem Cell Transplantation[NCT00578461]26 participants (Actual)Interventional2007-10-31Terminated
T-Regulatory Cell Kinetics for Patients Receiving Alemtuzamb and Undergoing Stem Cell Transplantation From HLA Mismatched-Related, or HLA Matched, or One Antigen Mismatched-Unrelated Donors[NCT00578539]24 participants (Actual)Interventional2007-10-31Terminated
A Phase II Study of Combination Nilotinib and Hyper-CVAD in Patients Newly Diagnosed With Philadelphia-Chromosome Positive Acute Lymphoblastic Leukemia or Chronic Myeloid Leukemia Blast-Phase Lymphoid Lineage[NCT01670084]Phase 20 participants (Actual)Interventional2012-12-31Withdrawn(stopped due to No Accrual)
A Phase II Study of Doxil (Liposomal Doxorubicin), Cyclophosphamide, Vincristine and Prednisone for AIDS-Related Systemic Lymphoma[NCT00003388]Phase 238 participants (Anticipated)Interventional1999-07-26Completed
Treatment Regimen or Children or Adolescent With Mature B-cell NHL or B-AL in China[NCT02405676]Phase 2/Phase 3200 participants (Anticipated)Interventional2015-01-31Active, not recruiting
An Open-label, Multi-center Phase Ib/II Study to Evaluate the Tolerance and Efficacy of Mitoxantrone Hydrochloride Liposome Injection Combined With Cytarabine in Patients With Acute Myeloid Leukemia (AML)[NCT05100303]Phase 1/Phase 258 participants (Anticipated)Interventional2021-12-31Not yet recruiting
Asia-wide, Multicenter Open-label, Phase II Non-randomised Study Involving Children With Down Syndrome Under 21 Year-old With Newly Diagnosed, Treatment naïve Acute Lymphoblastic Leukemia[NCT03286634]Phase 260 participants (Anticipated)Interventional2017-04-18Recruiting
A Non-Myeloablative Conditioning Regimen for Allogeneic Transplantation With Clofarabine, Cytarabine, and Thymoglobulin for Myelodysplastic Syndrome and Acute Myeloid Leukemia[NCT00593645]Phase 27 participants (Actual)Interventional2007-11-30Terminated(stopped due to toxicities were worse than expected)
The Efficiency of CAMS-2016 Trial for the Newly Diagnosed Pediatric Acute Myeloid Leukemia: A Prospective Single Centre Trial From China[NCT03173612]132 participants (Anticipated)Interventional2016-08-31Recruiting
A Randomized Phase II Study of Nuclear Factor-kappa B (NF-κB) Inhibition During Induction Chemotherapy for Patients With Acute Myelogenous Leukemia[NCT02144675]Phase 227 participants (Actual)Interventional2009-01-31Completed
A Randomized Phase II Study Comparing Inotuzumab Plus Chemotherapy Versus Standard Chemotherapy in Older Adults With Philadelphia-Chromosome-Negative B-Cell Acute Lymphoblastic Leukemia[NCT05303792]Phase 266 participants (Anticipated)Interventional2023-02-27Recruiting
Parent-Administered Low-Dose Cytarabine to Children and Adolescents With Cancer at Home - a Feasibility Study[NCT05372536]15 participants (Anticipated)Interventional2022-03-11Recruiting
Augmented Berlin-Frankfurt-Munster Therapy for Adolescents/Young Adults With Acute Lymphoblastic Leukemia or Lymphoblastic Lymphoma[NCT00866749]Phase 2120 participants (Actual)Interventional2006-09-12Completed
A PHASE IIA, MULTICENTER, OPEN-LABEL STUDY DESIGNED TO EVALUATE THE SAFETY AND EFFICACY OF ESCALATING DOSES OF BL-8040 IN ADULT SUBJECTS WITH RELAPSED/REFRACTORY ACUTE MYELOID LEUKEMIA[NCT01838395]Phase 242 participants (Actual)Interventional2013-04-30Completed
"A Feasibility Study of Early Allogeneic Hematopoietic Cell Transplantation for Relapsed or Refractory High-Grade Myeloid Neoplasms"[NCT02756572]Phase 230 participants (Actual)Interventional2016-09-22Completed
Phase I Dose Finding/Phase II Placebo-Controlled Trial of Eltrombopag During Consolidation Therapy in Adults With Acute Myeloid Leukemia (AML) in Complete Remission[NCT01656252]Phase 1/Phase 215 participants (Actual)Interventional2012-07-31Terminated(stopped due to Study stopped per Novartis request due to futility from another study.)
A Two-Part Study Evaluating the Combination of Tazemetostat and CPX-351 (Part 1) and Palbociclib With CPX-351 (Part 2) for the Treatment of Relapsed or Refractory Acute Myeloid Leukemia[NCT05627232]Phase 124 participants (Anticipated)Interventional2023-08-28Recruiting
A Phase II Study Using a BFM Regimen Plus Tyrosine Kinase Inhibitor in Adult Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia[NCT04845035]Phase 223 participants (Anticipated)Interventional2024-01-31Recruiting
A Phase III Trial to Evaluate the Efficacy of the Addition of Inotuzumab Ozogamicin (a Conjugated Anti-CD22 Monoclonal Antibody) to Frontline Therapy in Young Adults (Ages 18-39 Years) With Newly Diagnosed Precursor B-Cell ALL[NCT03150693]Phase 3310 participants (Anticipated)Interventional2017-06-01Suspended(stopped due to Unacceptable Toxicity)
Randomized Phase II Trial of Timed Sequential Cytosine Arabinoside (Ara-C) With and Without the Checkpoint Kinase 1 (CHK1) Inhibitor MK-8776 in Adults With Relapsed AML[NCT01870596]Phase 232 participants (Actual)Interventional2013-05-31Completed
An Multicenter, Randomized, Controlled, Prospective Clinical Study of Mitoxantrone Liposome Combined With PTCy as Conditioning Regimen in Allo-HSCT in Acute Leukemia[NCT05739630]Phase 2/Phase 360 participants (Anticipated)Interventional2023-01-01Recruiting
A Phase I/II Trial of Venetoclax and BEAM Conditioning Followed by Autologous Stem Cell Transplantation for Patients With Primary Refractory Non-Hodgkin Lymphoma[NCT03583424]Phase 1/Phase 219 participants (Actual)Interventional2018-09-10Active, not recruiting
A Phase II Study Incorporating Panobinostat, Bortezomib and Liposomal Vincristine Into Re-Induction Therapy for Relapsed Pediatric T-Cell Acute Lymphoblastic Leukemia or Lymphoma[NCT02518750]Phase 23 participants (Actual)Interventional2016-11-23Terminated(stopped due to Due to slow accrual)
Phase I and Clinical Pharmacokinetic De-Escalation Study of 2'-Deoxycitidine Administered as a Continuous Infusion in Conjunction With a Continuous Infusion of High-Dose ARA-C in Patients With Refractory Acute Myelogenous Leukemia[NCT00002818]Phase 115 participants (Actual)Interventional1995-02-28Completed
A Prospective,Randomized,and Comparative Study on the Efficacy of Venetoclax Combined With CACAG Regimen and BAT Regimen in the Treatment of Relapsed/Refractory Acute Myeloid Leukemia[NCT06084819]Phase 2200 participants (Anticipated)Interventional2023-08-01Recruiting
Phase Ib/II Study of the Combination of Low-Intensity Chemotherapy and Tagraxofusp in Patients With Acute Lymphoblastic Leukemia (ALL)[NCT05032183]Phase 1/Phase 240 participants (Anticipated)Interventional2022-02-17Recruiting
Risk-stratified Therapy for Primary and Secondary AML and MDS. A Randomized Study by AMLCG in Relation to Cytogenetically Defined Prognostic Factors (1) on the Role of High-dose AraC as Part of Double Induction, (2) on G-CSF Priming, and (3) on High-dose [NCT00266136]Phase 33,500 participants (Actual)Interventional1999-06-30Completed
A Phase II Open-Label Study of High-Dose Cytarabine and Clofarabine in Adult Patients With Refractory or Relapsed Acute Myelogenous Leukemia or Refractory or Relapsed Acute Lymphoblastic Leukemia[NCT01656031]Phase 239 participants (Actual)Interventional2005-02-28Completed
An Open-Label Phase 1/2 Multi-Arm Study of DS-1594b as a Single-Agent and in Combination With Azacitidine and Venetoclax or Mini-HCVD for the Treatment of Patients With Acute Myeloid Leukemia (AML) and Acute Lymphoblastic Leukemia (ALL)[NCT04752163]Phase 1/Phase 217 participants (Actual)Interventional2021-03-25Completed
A Phase I/II Study of the Selective Inhibitor of Nuclear Export Selinexor (KPT-330) in Combination With Fludarabine and Cytarabine in Patients With Refractory or Relapsed Leukemia or Myelodysplastic Syndrome[NCT02212561]Phase 119 participants (Actual)Interventional2014-08-31Completed
Immune Consolidation With Activated T Cells Armed With OKT3 x Rituxan (Anti-CD3 x Anti-CD20) Bispecific Antibody (CD20Bi) After Peripheral Blood Stem Cell Transplant for High Risk CD20+ Non-Hodgkin's Lymphomas[NCT00244946]Phase 115 participants (Actual)Interventional2004-03-31Completed
A Phase I Study of Lenalidomide Plus Chemotherapy With Mitoxantrone, Etoposide, and Cytarabine for the Reinduction of Patients With Acute Myelogenous Leukemia[NCT01681537]Phase 136 participants (Actual)Interventional2012-09-30Completed
Multicenter, Prospective, Open-label, Single-arm, Phase I-II Clinical Trial to Analyze Induction Therapy With a Combination of Fludarabine, Idarubicin, Cytarabine, G-CSF and Plerixafor for the Treatment of Young Patients With Relapsed or Refractory AML[NCT01435343]Phase 1/Phase 255 participants (Actual)Interventional2012-07-31Completed
Dose Escalation Study of Clofarabine in Combination With Cytarabine (Ara-C) and G-CSF Priming for Relapsed or Refractory Acute Myeloid Leukemia (AML) Patients[NCT00602225]Phase 1/Phase 250 participants (Actual)Interventional2007-12-31Completed
A Phase Ib/II Multicenter Open-label Study of BGB324 as a Single Agent and in Combination With Cytarabine or Decitabine in Patients With Acute Myeloid Leukemia or as a Single Agent in Patients With Myelodysplastic Syndrome[NCT02488408]Phase 1/Phase 2121 participants (Actual)Interventional2014-09-30Active, not recruiting
A Phase I Pharmacokinetic, Pharmacodynamic and Feasibility Study of Crenolanib in Combination With Sorafenib in Patients With Refractory or Relapsed Hematologic Malignancies (ARO-008)[NCT02270788]Phase 110 participants (Actual)Interventional2015-04-02Completed
A Phase I Trial of Imatinib Mesylate (Gleevec, Formerly Known as STI571) in Combination With Daunorubicin and Cytarabine for C-kit Positive Relapsed AML[NCT00268229]Phase 121 participants (Actual)Interventional2003-07-31Completed
Phase 1/2 Study of Cord Blood Transplantation From Unrelated Donor for Adult Patients With Hematologic Malignancies Using Myeloablative Conditioning Regimen[NCT00270881]Phase 1/Phase 233 participants (Actual)Interventional2006-01-31Completed
Phase 2 Open-Label Study of Amonafide L-Malate in Combination With Cytarabine in Subjects With Secondary Acute Myeloid Leukemia (AML)[NCT00273884]Phase 280 participants Interventional2005-08-31Completed
Phase II Study of Induction Therapy Comprising Etoposide, Methylprednisolone, Cytarabine, and Cisplatin (ESHAP) Followed by Consolidation Therapy Comprising Rituximab and Yttrium Y 90 Ibritumomab Tiuxetan in Patients With Relapsed or Refractory AIDS-Relat[NCT00310128]Phase 20 participants (Actual)Interventional2006-02-28Withdrawn(stopped due to Drug supply unavailable)
Intensive Chemotherapy and Immunotherapy in Patients With Newly Diagnosed Primary CNS Lymphoma: A Pilot Study[NCT00416819]10 participants (Actual)Interventional2003-09-30Completed
AML Therapy With Irradiated Allogeneic Cells[NCT02105116]6 participants (Actual)Interventional2014-02-28Terminated(stopped due to No patients were eligible to receive the experimental component of the protocol therapy.)
Phase 1 Study of Venetoclax/Azacitidine or Venetoclax in Combination With Ziftomenib or Standard Induction Cytarabine/Daunorubicin (7+3) Chemotherapy in Combination With Ziftomenib for the Treatment of Patients With Acute Myeloid Leukemia[NCT05735184]Phase 1212 participants (Anticipated)Interventional2023-07-18Recruiting
A Phase Ib Trial With Dose Expansion Evaluating CPX-351 Plus Gemtuzumab Ozogamicin for Relapsed Acute Myelogenous Leukemia[NCT03904251]Phase 113 participants (Actual)Interventional2019-07-18Terminated(stopped due to slow accrual)
A Phase I Study of Lintuzumab-Ac225 in Combination With CLAG-M Chemotherapy in Patients With Relapsed/Refractory Acute Myeloid Leukemia[NCT03441048]Phase 126 participants (Actual)Interventional2018-05-22Active, not recruiting
A Phase 3, Double-Blind, Placebo-controlled Study of Quizartinib Administered in Combination With Induction and Consolidation Chemotherapy, and Administered as Continuation Therapy in Subjects 18 to 75 Years Old With Newly Diagnosed FLT3-ITD (+) Acute Mye[NCT02668653]Phase 3539 participants (Actual)Interventional2016-09-01Completed
A Phase I Open-Label Study of High-Dose Cytarabine and Clofarabine in Adult Patients With Refractory or Relapsed Acute Myelogenous Leukemia or Refractory or Relapsed Acute Lymphoblastic Leukemia[NCT00295841]Phase 153 participants (Actual)Interventional2005-02-28Completed
Phase I/II Study of the Combination of Dasatinib With Chemotherapy for High Risk Acute Myeloid Leukemia (AML) Patients[NCT01876953]Phase 1/Phase 220 participants (Actual)Interventional2013-09-13Terminated(stopped due to Due to the budget issues, the study discontinued at Phase II.)
Therapy of Early Chronic Phase Chronic Myelogenous Leukemia (CML) With SCH54031 (PEG Interferon Alpha 2B/PEG Intron) and Low-Dose Cytosine Arabinoside (Ara-C)[NCT00303290]Phase 176 participants (Actual)Interventional2000-01-31Completed
Randomized Phase III Study Comparing the OSHO Arm to the Standard Intergroup Arm.[NCT01497002]Phase 31,222 participants (Actual)Interventional2005-04-30Completed
A Phase II Study Of Pooled Unrelated Donor Umbilical Cord Blood (UCB) Transplant For Patients With Hematologic Malignancies Needing Allogeneic Stem Cell Transplant But Do Not Have A Related HLA-Matched Donor[NCT01500161]Phase 21 participants (Actual)Interventional2011-11-30Terminated(stopped due to Insufficient accruals)
A Randomized Phase II Trial of Myeloablative Versus Non-Myeloablative Consolidation Chemotherapy for Newly Diagnosed Primary CNS B-cell Lymphoma[NCT01511562]Phase 2113 participants (Actual)Interventional2012-09-30Active, not recruiting
Randomized Phase II Study of Epigenetic Priming Using Decitabine With Induction Chemotherapy in Patients With Acute Myelogenous Leukemia (AML)[NCT01627041]Phase 2178 participants (Actual)Interventional2011-09-16Active, not recruiting
A Multicenter, Open Label, Phase 1B Study of Escalating Doses of RO5045337 Administered Orally, With Cytarabine Administered A) Subcutaneously, or B) Intravenously, in Patients With Acute Myelogenous Leukemia (AML)[NCT01635296]Phase 143 participants (Actual)Interventional2012-07-31Completed
Phase I/II Study of Cytarabine or 5-Azacitidine Combined With Tosedostat to Evaluate the Safety and Tolerability in Older Patients With Acute Myeloid Leukemia (AML) or High Risk MDS[NCT01636609]Phase 118 participants (Actual)Interventional2012-11-20Terminated(stopped due to In 2013 the FDA put a temporary hold on the trial and the Phase II portion of this study was cancelled.)
Phase II Trial Examining Epigenetic Priming With Decitabine Followed by Idarubicin and Cytarabine for Patients With Relapsed or Refractory AML.[NCT01607645]Phase 27 participants (Actual)Interventional2012-07-31Terminated(stopped due to The study was terminated early because there were other competing protocols.)
Efficacy and Safety of Chidamide in CBF Leukemia[NCT03031262]Phase 1/Phase 2250 participants (Anticipated)Interventional2017-02-08Recruiting
Role of Intrathecal Chemotherapy With Liposomal Cytarabine (DepoCyte®) in Patients Wih Leptomeningeal Metastasis of Breast Cancer. A Randomized Phase III Study.[NCT01645839]Phase 374 participants (Actual)Interventional2011-08-30Completed
A Randomised Study of High Dose Chemotherapy/Radiotherapy and Autologous Bone Marrow Transplantation in Patients With High Grade Malignant Non-Hodgkin's Lymphoma (Kiel Classification) According to Prognostic Groups[NCT00003815]Phase 30 participants Interventional1994-06-30Active, not recruiting
A Phase II Trial to Evaluate the Safety, Feasibility and Efficacy of a Salvage Therapy Consisting of Temsirolimus Added to the Standard Therapy R-DHAP for the Treatment of Patients With Relapsed or Refractory DLBCL - the STORM Trial[NCT01653067]Phase 288 participants (Anticipated)Interventional2012-09-30Active, not recruiting
Phase II Trial Utilizing Idarubicin in Combination With High Dose Ara-C for Induction Therapy for Adult Acute Myelogenous Leukemia (AML)[NCT00528398]Phase 2111 participants (Actual)Interventional1994-09-30Completed
A Phase I Study of Venetoclax to Augment Epigenetic Modification and Chemotherapy in Pediatric and Young Adult Patients With Relapsed and Refractory Acute Myeloid Leukemia[NCT05317403]Phase 140 participants (Anticipated)Interventional2023-03-31Recruiting
Phase 1/2 Study of Decitabine Combined With Modified CAG Followed by HLA Haploidentical T Cell Infusion in Treating Elderly Patients With Intermediate-high Risk Myelodysplastic Syndrome(MDS) or Acute Myeloid Leukemia(AML)[NCT01690507]Phase 1/Phase 229 participants (Actual)Interventional2012-11-30Completed
A Pilot Study of CPX-351 (Vyxeos™) for Transplant Eligible, Higher Risk Patients With Myelodysplastic Syndrome[NCT03572764]Phase 120 participants (Actual)Interventional2018-12-14Active, not recruiting
A Phase II Study of Therapy for Pediatric Relapsed or Refractory Precursor B-Cell Acute Lymphoblastic Leukemia and Lymphoma[NCT01700946]Phase 280 participants (Actual)Interventional2013-04-15Completed
A Randomized, Blinded, Placebo-Controlled, Dose Finding Study to Assess the Safety and Efficacy of the Oral Thrombopoietin Receptor Agonist, Eltrombopag, Administered to Subjects With Acute Myelogenous Leukaemia (AML) Receiving Induction Chemotherapy[NCT01890746]Phase 2148 participants (Actual)Interventional2013-09-05Completed
Treatment Protocol for Newky Diagnosed Adult Ph-Chromosome Positive (BCR::ABL1) Acute Lymphoblastic Leukemia (LALPh2022)[NCT06175702]150 participants (Anticipated)Observational2023-12-25Not yet recruiting
Consolidation With ADCT-402 (Loncastuximab Tesirine) After a Short Course of Immunochemotherapy: a Phase II Study in BTKi-treated (or BTKi Intolerant) Relapsed/Refractory (R/R) Mantle Cell Lymphoma (MCL) Patients[NCT05249959]Phase 256 participants (Anticipated)Interventional2022-04-21Recruiting
CD19-specific T Cell Infusion in Patients With B-Lineage Lymphoid Malignancies[NCT00968760]Phase 134 participants (Actual)Interventional2011-06-20Completed
A Study of Umbilical Cord Blood-Derived Natural Killer Cells in Conjunction With Lymphodepleting Chemotherapy and Lenalidomide as Immunotherapy in Patients With Hematologic Malignancies[NCT02280525]Phase 18 participants (Actual)Interventional2015-03-05Completed
A Phase II Study of Yttrium-90-Labeled Anti-CD20 Monoclonal Antibody in Combination With High-Dose Beam Followed by Autologous Stem Cell Transplantation for Poor Risk/Relapsed B-Cell Lymphoma[NCT00695409]Phase 2122 participants (Actual)Interventional2008-03-18Completed
An Open Label, Dose-Escalation Study to Evaluate Safety, Tolerability, Maximum Tolerated Dose (MTD), Efficacy, and Pharmacokinetics (PKs) of CPI-613 Given With High Dose Cytarabine and Mitoxantrone in Patients With Relapsed or Refractory Acute Myeloid Leu[NCT01768897]Phase 167 participants (Actual)Interventional2013-01-31Completed
AUTOLOGOUS TRANSPLANTATION AND STEM CELL BASED-GENE THERAPY FOR THE TREATMENT OF HIV-ASSOCIATED LYMPHOMA[NCT01769911]0 participants (Actual)Interventional2015-02-28Withdrawn
Safety and Efficacy of Implementing Low-Dose Coronary Computed Tomographic Angiography for Early Triage of Acute Chest Pain in Emergency Department[NCT01770444]681 participants (Actual)Interventional2012-12-31Completed
A Phase I Study of the Aurora A Kinase Inhibitor Alisertib in Combination With 7+3 Induction Chemotherapy in Patients With Acute Myeloid Leukemia[NCT01779843]Phase 122 participants (Actual)Interventional2013-04-30Completed
An Open-Label, Single-Dose Study Designed to Assess the Mass Balance Recovery of an Intravenous Microdose of [14C]-Elacytarabine in Healthy Male Subjects[NCT01783964]Phase 16 participants (Actual)Interventional2013-02-28Completed
A Phase I/II Clinical Trial Testing the Safety and Feasibility of IL-21-Expanded Natural Killer Cells for the Induction of Relapsed/Refractory Acute Myeloid Leukemia[NCT01787474]Phase 130 participants (Actual)Interventional2014-05-19Completed
Phase III Randomized Study of Crenolanib Versus Midostaurin Administered Following Induction Chemotherapy and Consolidation Therapy in Newly Diagnosed Subjects With FLT3 Mutated Acute Myeloid Leukemia[NCT03258931]Phase 3510 participants (Anticipated)Interventional2018-08-15Recruiting
A Phase II Study of Intrathecal and Systemic Chemotherapy With Radiation Therapy for Children With Central Nervous System Atypical Teratoid/Rhabdoid Tumor (AT/RT) Tumor[NCT00084838]Phase 225 participants (Actual)Interventional2003-02-28Completed
A Phase II Study of Sulindac, a COX Inhibitor, in Older Patients With Acute Myeloid Leukemia in First Complete Remission[NCT01843179]Phase 20 participants (Actual)Interventional2014-01-31Withdrawn(stopped due to Lack of Funding)
Phase 1 Trial of Ivosidenib and FLAG Chemotherapy in Relapsed/Refractory IDH1+ Acute Myeloid Leukemia (AML)[NCT04250051]Phase 125 participants (Anticipated)Interventional2020-12-21Recruiting
Treatment of Poor Prognosis Acute Myeloid Leukemia and Blastic Crisis Chronic Myelogenous Leukemia With Mylotarg, High-Dose Cytarabine, Mitozantrone and Ethyol AML/CML 2000-06.[NCT00003407]Phase 20 participants (Actual)Interventional2001-02-13Withdrawn(stopped due to The P.I deceased.)
"Randomized Phase II Trial of Timed Sequential Therapy (TST) With Alvocidib (Flavopiridol), Ara-C and Mitoxantrone (FLAM) vs. 7+3 for Adults Age 70 and Under With Newly Diagnosed Acute Myelogenous Leukemia (AML)"[NCT01349972]Phase 2172 participants (Actual)Interventional2011-04-30Completed
Phase 2 Study of Rituximab and ESHAP (Etoposide, Methylprednisolone, Cytarabine, and Cisplatin) in Relapsed and Refractory Aggressive Non-Hodgkin's Lymphoma[NCT00367497]Phase 25 participants (Actual)Interventional2005-08-31Terminated(stopped due to The stopping rule was applied because of low response rates.)
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
Phase 1/2 Study of Concurrent Decitabine in Combination With G-CSF, Cladribine, Cytarabine, and Mitoxantrone (G-CLAM) in Adults With Newly Diagnosed Acute Myeloid Leukemia (AML) or High-Risk Myelodysplastic Syndromes (MDS)[NCT02921061]Phase 1/Phase 228 participants (Actual)Interventional2016-11-17Completed
Safety and Efficiency of Anti-CD19/CD22 Tandem Fully Human Chimeric Antigen Receptor (CAR)-Transduced T-cell Therapy for Pediatric and Young Adult Patients With Relapsed/Refractory B-cell Acute Lymphoblastic Leukemia: a Single Centre, Non-randomised, Open[NCT04499573]Phase 1/Phase 250 participants (Anticipated)Interventional2020-07-27Active, not recruiting
Phase I Study of Vorinostat (Suberoylanilide Hydroxamic Acid, or SAHA) in Combination With Cytosine Arabinoside (Ara-C) and Etoposide for Patients With Relapsed and/or Refractory Acute Leukemias, Myelodysplasias and Myeloproliferative Disorders[NCT00357305]Phase 125 participants (Actual)Interventional2006-05-31Completed
A PHASE 1B/2 STUDY TO EVALUATE THE SAFETY AND EFFICACY OF PF-04449913, AN ORAL HEDGEHOG INHIBITOR, IN COMBINATION WITH INTENSIVE CHEMOTHERAPY, LOW DOSE ARA-C OR DECITABINE IN PATIENTS WITH ACUTE MYELOID LEUKEMIA OR HIGH-RISK MYELODYSPLASTIC SYNDROME[NCT01546038]Phase 2255 participants (Actual)Interventional2012-06-27Completed
Phase II Trial of Sequential Chemotherapy and Radiotherapy for AIDS-Related Primary Central Nervous System Lymphoma[NCT00000801]Phase 233 participants InterventionalCompleted
Phase II Study of Venetoclax Added to Cladribine Plus Low Dose Cytarabine (LDAC) Induction Followed by Consolidation With Cladribine Plus LDAC Alternating With 5-Azacitidine With Venetoclax in Patients With Untreated AML[NCT03586609]Phase 2145 participants (Anticipated)Interventional2018-10-25Recruiting
Phase II Study of High-Dose Cytarabine, Cisplatin, and Dexamethasone Followed By Cyclophosphamide, Etoposide, Total Body Irradiation, and Autologous Bone Marrow Rescue in Patients With Relapsed or Refractory Non-Hodgkin's Lymphoma[NCT00002481]Phase 20 participants Interventional1990-03-31Active, not recruiting
TREATMENT OF ALL IN FIRST BONE MARROW RELAPSE AFTER BFM PROTOCOLS[NCT00002499]Phase 2/Phase 30 participants Interventional1990-01-31Active, not recruiting
IDA VS MTZ IN INDUCTION AND INTENSIFICATION TREATMENT OF AML OR MDS IN CHILDREN, A PHASE III RANDOMIZED STUDY[NCT00002517]Phase 30 participants Interventional1993-03-31Completed
ALLOGENEIC AND SYNGENEIC MARROW TRANSPLANTATION IN PATIENTS WITH ACUTE NON-LYMPHOCYTIC LEUKEMIA[NCT00002547]Phase 2280 participants (Actual)Interventional1987-08-31Completed
A Phase I and Expansion Cohort Study of Selinexor and Venetoclax in Combination With Chemotherapy in Pediatric and Young Adult Patients With Refractory or Relapsed Acute Myeloid Leukemia[NCT04898894]Phase 142 participants (Anticipated)Interventional2021-11-15Recruiting
"RANDOMIZED COMPARISON OF ALTERNATING TRIPLE THERAPY (ATT) VERUS CHOP IN PATIENTS WITH INTERMEDIATE GRADE LYMPHOMAS AND IMMUNOBLASTIC LYMPHOMAS WITH INTERNATIONAL INDEX 2-5"[NCT00002565]Phase 361 participants (Actual)Interventional1994-05-25Completed
Study of Promace-Cytabom With Trimethoprim Sulfamethoxazole, Zidovudine (AZT), and Granulocyte Colony Stimulating Factor (G-CSF) in Patients With AIDS-Related Lymphoma, Phase II[NCT00002571]Phase 252 participants (Actual)Interventional1994-06-30Completed
International Proof of Concept Therapeutic Stratification Trial of Molecular Anomalies in Relapsed or Refractory HEMatological Malignancies in Children, Sub-protocol B Dasatinib + Venetoclax + Dexamethasone + Cyclophosphamide and Cytarabine in Pediatric P[NCT05751044]Phase 1/Phase 226 participants (Anticipated)Interventional2023-10-01Not yet recruiting
FAB LMB 96 -- Treatment of Mature B-CELL Lymphoma/Leukemia: A SFOP LMB 96/CCG 5961/UKCCSG NHL 9600 Cooperative Study[NCT00002757]Phase 31,148 participants (Actual)Interventional2001-06-30Completed
T-Cell Replete Haploidentical Donor Hematopoietic Stem Cell Plus Natural Killer (NK) Cell Transplantation in Patients With Hematologic Malignancies Relapsed or Refractory Despite Previous Allogeneic Transplant[NCT01621477]Phase 234 participants (Actual)Interventional2012-08-31Terminated(stopped due to Study was terminated in August 2016 due to replacement by a new study.)
Phase II Study of the Combination of High-Dose Methotrexate and Intrathecal Liposomal Cytarabine in Patients With Leptomeningeal Metastases With or Without Parenchymal Brain Involvement[NCT00992602]Phase 23 participants (Actual)Interventional2011-04-30Completed
A Phase 2 Trial of Yttrium-90 Labeled Anti-CD25 Monoclonal Antibody Combined With BEAM Chemotherapy (aTac-BEAM) Conditioning for Autologous Hematopoietic Cell Transplantation (AHCT) in Patients With Primary Refractory or Relapsed Hodgkin Lymphoma[NCT04871607]Phase 233 participants (Anticipated)Interventional2021-11-02Recruiting
Efficacy and Safety of Rituximab, High-dose Ara-C and Dexamethasone (R-HAD) Alone or in Combination With Bortezomib in Patients With Relapsed or Refractory Mantle Cell Lymphoma[NCT01449344]Phase 3128 participants (Actual)Interventional2009-05-09Active, not recruiting
Sequential Chemotherapy and Lenalidomide Followed by Rituximab and Lenalidomide Maintenance for Untreated Mantle Cell Lymphoma: A Phase II Study[NCT02633137]Phase 249 participants (Actual)Interventional2015-12-14Completed
RAVEN: A Phase I/II Trial Treating Relapsed Acute Lymphoblastic Leukemia With Venetoclax and Navitoclax[NCT05192889]Phase 1/Phase 290 participants (Anticipated)Interventional2022-08-25Recruiting
High-Dose Immunosuppressive Therapy Using Carmustine, Etoposide, Cytarabine, and Melphalan (BEAM) + Thymoglobulin Followed by Syngeneic or Autologous Hematopoietic Cell Transplantation for Patients With Autoimmune Neurologic Diseases[NCT00716066]Phase 280 participants (Anticipated)Interventional2008-06-30Recruiting
A Phase I Trial of Temsirolimus (CCI-779, Pfizer, Inc.) in Combination With Etoposide and Cyclophosphamide in Children With Relapsed Acute Lymphoblastic Leukemia and Non-Hodgkins Lymphoma[NCT01614197]Phase 116 participants (Actual)Interventional2015-07-03Completed
A Phase I Dose Finding and Proof-of-concept Study of the Histone Deacetylase Inhibitor Panobinostat (LBH589) in Combination With Standard Dose Cytarabine and Daunorubicin for Older Patients With Untreated Acute Myeloid Leukemia or Advanced Myelodysplastic[NCT01463046]Phase 129 participants (Actual)Interventional2012-01-31Completed
A Prospective, Multicenter, Single-Arm Pilot Study of CPX-351 (VYXEOS) in Individuals < 22 Years With Secondary Myeloid Neoplasms[NCT05656248]Phase 225 participants (Anticipated)Interventional2023-01-17Recruiting
A Phase 1/1b Dose Escalation and Expansion of CPX-351 in Combination With Gemtuzumab Ozogamicin in Newly Diagnosed Acute Myeloid Leukemia[NCT05558124]Phase 118 participants (Anticipated)Interventional2022-09-22Recruiting
Phase I/II Trial Of Sequential Therapy With Cytarabine And Bismuth-213-Labeled HuM195 (Humanized Anti-CD33) In Patients With Advanced Myeloid Malignancies[NCT00014495]Phase 1/Phase 232 participants (Actual)Interventional2000-11-30Completed
A Pilot Study of Acalabrutinib With Bendamustine / Rituximab Followed by Cytarabine / Rituximab for Untreated Mantle Cell Lymphoma[NCT03623373]Phase 213 participants (Actual)Interventional2018-11-29Active, not recruiting
A Phase II Randomized Trial of Carboplatin and Topotecan; Flavopiridol, Mitoxantrone and Cytosine Arabinoside; and Sirolimus, Mitoxantrone, Etoposide and Cytosine Arabinoside for the Treatment of Adults With Primary Refractory or Initial Relapse of Acute [NCT00634244]Phase 292 participants (Actual)Interventional2008-10-31Completed
T2007-002 A Phase II Study of Clofarabine With Etoposide and Cyclophosphamide in Relapsed/Refractory AML (IND 104,650)[NCT00939653]Phase 26 participants (Actual)Interventional2009-07-10Terminated(stopped due to Due to insufficient research institution participation and patient enrollment)
Treateament of Newly Diagnosed Acute Monocytic Leukemia in Children: A Prospective Multicenter Study in South China[NCT05313958]Phase 2/Phase 343 participants (Anticipated)Interventional2021-12-01Recruiting
Clinical Study on the Safety and Efficacy of QN-030a in Acute Myeloid Leukemia With Minimal Residual Disease[NCT05601830]Phase 118 participants (Anticipated)Interventional2022-10-28Recruiting
A Prospective Clinical Study of Hanlikang and BTK Inhibitors in the Treatment of Newly Diagnosed Mantle Cell Lymphoma[NCT05506410]100 participants (Anticipated)Observational [Patient Registry]2022-08-12Recruiting
Clinical Study of Bcl-2 Inhibitors Combined With Azacytidine and Chemotherapy in Elderly Patients With Previously Untreated Acute Myeloid Leukemia[NCT05053425]30 participants (Anticipated)Interventional2021-10-20Recruiting
An Investigator Sponsored Phase I/II Study of CLAG + Selinexor in Relapsed or Refractory Acute Myeloid Leukemia[NCT02416908]Phase 1/Phase 240 participants (Actual)Interventional2015-06-16Completed
Reduced Intensity Allogeneic Hematopoietic Stem Cell Transplantation for Sickle Cell Anemia From HLA Matched or Partially-Matched Related Donors[NCT01350232]2 participants (Actual)Interventional2009-09-30Terminated(stopped due to Poor accrual)
A Randomized Phase III Study to Compare Arsenic Trioxide (ATO) Combined to ATRA and Idarubicin Versus Standard ATRA and Anthracyclines-based Chemotherapy (AIDA Regimen) for Patients With Newly Diagnosed, High-risk Acute Promyelocytic Leukemia[NCT02688140]Phase 3280 participants (Anticipated)Interventional2016-06-30Active, not recruiting
A Phase III Randomized Trial of Low-Dose Versus Standard-Dose mBACOD Chemotherapy With rGM-CSF for Treatment of AIDS-Associated Non-Hodgkin's Lymphoma[NCT00000658]Phase 3250 participants InterventionalCompleted
HIGH INTENSITY, BRIEF DURATION CHEMOTHERAPY FOR DIFFUSE SMALL NONCLEAVED CELL LYMPHOMA AND THE L-3 SUBTYPE OF ALL: A PILOT STUDY OF A MULTIDRUG REGIMEN[NCT00002494]Phase 2134 participants (Actual)Interventional1992-05-31Completed
Multicenter Study to Optimise Therapy of B-ALL, Burkitt's NHL and High-Grade Non-Hodgkin's Lymphoma in Adults (Amend 7)[NCT00199082]Phase 4650 participants (Anticipated)Interventional2002-07-31Completed
HIGH-DOSE CHEMOTHERAPY FOLLOWED BY AUTOLOGOUS PERIPHERAL BLOOD STEM CELL TRANSPLANTATION FOR CHILDREN WITH RELAPSED ACUTE LYMPHOCYTIC LEUKEMIA[NCT00002638]Phase 230 participants (Anticipated)Interventional1995-03-31Completed
CYTOREDUCTIVE CHEMOTHERAPY WITH MITOXANTRONE, CYTOSINE ARABINOSIDE AND ETOPOSIDE FOLLOWED BY RECOMBINANT HUMAN G-CSF FOR MOBILIZATION OF PERIPHERAL BLOOD STEM CELLS IN PATIENTS WITH CHRONIC MYELOID LEUKEMIA[NCT00002674]Phase 230 participants (Anticipated)Interventional1994-10-31Completed
TREATMENT OF ISOLATED CNS RELAPSE OF ACUTE LYMPHOBLASTIC LEUKEMIA -- A PEDIATRIC ONCOLOGY GROUP-WIDE PHASE II STUDY[NCT00002704]Phase 2156 participants (Actual)Interventional1996-01-31Completed
A Pilot Study For The Treatment of Newly-Diagnosed Disseminated Anaplastic Large Cell Ki-1 Lymphoma and T-Large Cell Lymphoma[NCT00002590]Phase 2221 participants (Actual)Interventional1994-07-31Completed
Phase II Study of Age-Adjusted R-BAC (Rituximab, Bendamustine, Cytarabine) as Induction Therapy in Older Patients With Mantle Cell Lymphoma (MCL)[NCT01662050]Phase 257 participants (Actual)Interventional2012-03-20Completed
Autologous Peripheral Blood Stem Cell Transplantation (PSCT) Versus a Second Intensive Consolidation Course After a Common Induction and Consolidation Course in Patients With Bad Prognosis Myelodysplastic Syndromes (MDS) and Acute Myelogenous Leukemia Sec[NCT00002926]Phase 380 participants (Anticipated)Interventional1996-12-31Active, not recruiting
A Phase lb Study of the Safety, Feasibility, and Pharmacokinetics of AMG 386 Alone and in Combination With Low Dose Cytarabine in Acute Myeloid Leukemia (AML) Patients[NCT01555268]Phase 124 participants (Actual)Interventional2011-10-31Completed
Medical Research Council Working Party on Leukaemia in Childhood Acute Myeloid Leukaemia Trial 12[NCT00003436]Phase 32,000 participants (Anticipated)Interventional1998-07-31Completed
PHASE II STUDY OF HIGH DOSE CYTARABINE COMBINED WITH A SINGLE HIGH DOSE OF IDARUBICIN FOR NEWLY DIAGNOSED PATIENTS WITH AML: THE AML-3 PROTOCOL[NCT00002800]Phase 260 participants (Anticipated)Interventional1996-07-31Completed
Treatment of Patients With Acute Lymphoblastic Leukemia With Unfavorable Features: A Phase III Group-wide Study[NCT00002812]Phase 32,078 participants (Actual)Interventional1996-09-30Completed
Phase III Study of MDR Modulation With PSC-833 (NSC #648265) Followed by Immunotherapy With rIL-2 (NSC #373364) vs. No Further Therapy in Previously Untreated Patients With AML >60 Years[NCT00003190]Phase 3640 participants (Actual)Interventional1998-01-31Completed
Phase III Randomized Study of Concurrent Tretinoin and Chemotherapy With or Without Arsenic Trioxide (AS2O3) (NSC # 706363) as Initial Consolidation Therapy Followed by Maintenance Therapy With Intermittent Tretinoin Versus Intermittent Tretinoin Plus Mer[NCT00003934]Phase 3420 participants (Actual)Interventional1999-06-30Completed
A Phase II Study of Newly Diagnosed Patients With BCR/ABL (+) Chronic Myelogenous Leukemia Treated With Combined Homoharringtonine (NSC #141633) and Low-Dose Cytarabine[NCT00003694]Phase 260 participants (Actual)Interventional1999-03-31Completed
Phase II Study in Adults With Untreated Acute Lymphoblastic Leukemia Testing Increased Doses of Daunorubicin During Induction, and Cytarabine During Consolidation, Followed by High-Dose Methotrexate and Intrathecal Methotrexate in Place of Cranial Irradia[NCT00003700]Phase 2163 participants (Actual)Interventional1999-01-31Completed
A Phase I Study of Cytosine Arabinoside, Idarubicin, and Amifostine as Induction Therapy for Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT00003268]Phase 10 participants Interventional1998-01-31Completed
PHASE I-II STUDY OF IDARUBICIN, CYTARABINE AND R115777 (TIPIFARNIB, ZARNESTRA; 702818; IND 58359), A FARNESYLTRANSFERASE INHIBITOR, IN PATIENTS WITH HIGH-RISK MYELODYSPLASTIC SYNDROMES AND ACUTE MYELOID LEUKEMIAS[NCT00096122]Phase 1/Phase 295 participants (Actual)Interventional2004-09-30Completed
Non-Myeloablative Chemotherapy Followed by Unrelated Allogeneic Stem Cell Transplantation in Patients With Advanced Hematologic Malignancies: A Pilot Study[NCT00004114]Phase 10 participants (Actual)InterventionalWithdrawn(stopped due to Study never opened)
Phase I/II Study of the Combination of Low-Intensity Chemotherapy and Venetoclax (ABT-199) in Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia (ALL)[NCT03808610]Phase 1/Phase 250 participants (Anticipated)Interventional2019-04-03Recruiting
A Phase 2 Trial to Evaluate the Efficacy of Bortezomib, Cytarabine, and Dexamethasone in Patients With Relapsed or Refractory Mantle Cell Lymphoma[NCT02840539]Phase 219 participants (Actual)Interventional2016-10-11Completed
Clinical Study on QN-023a Targeting CD33 in Relapsed/Refractory Acute Myeloid Leukemia[NCT05601466]Phase 118 participants (Anticipated)Interventional2022-10-28Recruiting
Phase I Study of Yttrium-90 Labeled Anti-CD25 Monoclonal Antibody Plus Standard BEAM Conditioning for Autologous Hematopoietic Cell Transplantation in Patients With Mature T-Cell Non-Hodgkin Lymphoma: the aTAC BEAM Regimen[NCT02342782]Phase 120 participants (Actual)Interventional2020-06-08Active, not recruiting
Pilot Study for Treatment of Elderly Patients (>65 Years) With Acute Lymphoblastic Leukemia[NCT00199095]Phase 440 participants Interventional1997-02-28Completed
Multicenter Study To Optimize Treatment in Elderly Patients (> 55 Years, No Upper Age Limit) With Acute Lymphoblastic Leukemia (GMALL Elderly 1/2003)(Amend 2)[NCT00198978]Phase 4377 participants (Actual)Interventional2003-01-31Completed
A Biomarker Validation Study to Establish Whether Serial Flow Cytometric Measurements Predict Clinical Response to Sirolimus and MEC (Mitoxantrone Etoposide Cytarabine) Treatment in Patients With High-Risk Acute Myelogenous Leukemia[NCT02583893]Phase 239 participants (Actual)Interventional2015-10-07Completed
A Phase II Study To Determine The Anti-Leukemic Effects Of STI571 In Combination With Ara-C In Patients With Chronic Myelogenous Leukemia In Chronic Phase[NCT00022490]Phase 224 participants (Actual)Interventional2001-06-30Terminated
ALinC 17: Protocol for Patients With Newly Diagnosed Standard Risk Acute Lymphoblastic Leukemia (ALL): A Phase III Study[NCT00005596]Phase 31,076 participants (Actual)Interventional2000-04-30Completed
Reduced Burden of Oncologic Therapy in Advanced B-cell Lymphoma (REBOOT ABLY) in Children, Adolescents and Young Adults With CD20+ Mature B-Cell Lymphoma[NCT01859819]Phase 245 participants (Actual)Interventional2013-01-31Completed
An International Randomised Clinical Trial of Therapeutic Interventions to Assess the Effects on Outcome in Adults With Acute Myeloid Leukaemia and High Risk Myelodysplasia Undergoing Allogeneic Stem Cell Transplantation.[NCT04217278]Phase 2/Phase 3333 participants (Actual)Interventional2020-01-27Active, not recruiting
A Phase III Study of the Addition of Gemtuzumab Ozogamicin (Mylotarg®) During Induction Therapy Versus Standard Induction With Daunomycin and Cytosine Arabinoside Followed by Consolidation and Subsequent Randomization to Post-Consolidation Therapy With Ge[NCT00085709]Phase 3637 participants (Actual)Interventional2004-07-31Completed
Clofarabine With Cytarabine for MRD Positive Leukemia[NCT01158885]Phase 22 participants (Actual)Interventional2010-08-31Terminated(stopped due to Study terminated for lack of accrual.)
An Open Phase I/IIa Trial to Investigate the Maximum Tolerated Dose, Safety, Efficacy and Pharmacokinetics of BI 811283 in Combination With Cytarabine in Patients With Previously Untreated Acute Myeloid Leukaemia Ineligible for Intensive Treatment[NCT00632749]Phase 268 participants (Actual)Interventional2008-05-31Completed
Phase I/II Randomized Study of Clofarabine, Idarubicin, and Cytarabine (CIA) Versus Fludarabine, Idarubicin, and Cytarabine (FLAI) in Acute Myelogenous Leukemia and High-Risk Myelodysplastic Syndrome[NCT01289457]Phase 1/Phase 2282 participants (Actual)Interventional2011-02-02Completed
Reducing the Burden of Oncologic Chemoradiotherapy And Radiation Exposure From Diagnostic Imaging by Utilizing Targeted Immunotherapy in Children, Adolescents and Young Adults With Lymphoma[NCT05253495]Phase 280 participants (Anticipated)Interventional2022-02-01Recruiting
Phase I Dose Escalation Study of Millennium 9708 in Combination With Induction and Consolidation Chemotherapy in Adults >= 60 Years With Acute Myeloid Leukemia[NCT02582359]Phase 139 participants (Actual)Interventional2016-01-31Completed
Evaluation of Functional Rehabilitation Fast Track Total Hip Arthroplasty vs Standard Care: a Randomized Controlled Trial[NCT04211987]93 participants (Actual)Interventional2018-03-13Completed
Phase 1 Study of Selinexor in Combination With Topoisomerase-II Inhibition in Acute Myeloid Leukemia[NCT02299518]Phase 123 participants (Actual)Interventional2015-05-18Completed
Cytarabine Monotherapy for Adult Patients With Newly Diagnosed Langerhans Cell Histiocytosis: A Single Arm, Single Center, Prospective Phase 2 Study[NCT04121819]Phase 240 participants (Anticipated)Interventional2019-10-01Recruiting
German Multicenter Study for Treatment Optimisation in Acute Lymphoblastic Leukemia in Adults and Adolescents Above 15 Years (Amend 3) (GMALL 07/2003)[NCT00198991]Phase 41,883 participants (Actual)Interventional2003-04-30Completed
Open-label, Multicenter Phase I/II Study: Salvage Therapy of Progressive and Relapsed Aggressive Non-Hodgkin-Lymphoma by Combination of Lenalidomide (Revlimid®) With Rituximab, Dexamethason, High-dose ARA-C and Cisplatinum (R²-DHAP)[NCT02983097]Phase 1/Phase 234 participants (Actual)Interventional2010-11-30Terminated(stopped due to Phase II: no scientific interests are given anymore)
Zevalin And Rituxan For The Treatment Of Relapsed Or Refractory Diffuse Large B-Cell Non-Hodgkin's Lymphoma[NCT00073957]Phase 225 participants (Actual)Interventional2003-12-31Completed
UARK 98-026, Total Therapy II - A Phase III Study for Newly Diagnosed Multiple Myeloma Evaluating Anti-Angiogenesis With Thalidomide and Post-Transplant Consolidation Chemotherapy[NCT00083551]Phase 3668 participants (Actual)Interventional1998-08-31Completed
ChiCGB Versus BEAM With Autologous Stem-Cell Transplantation in High-risk Hodgkin and Non-Hodgkin Lymphoma - A Prospective, Multi-centered, Randomized Clinical Trial[NCT05466318]Phase 3306 participants (Anticipated)Interventional2022-07-01Recruiting
A Prospective Institutional Study for the Treatment of Children With Newly Diagnosed Langerhans Cell Histiocytosis Using a Cytarabine Contained Protocol[NCT04773366]Phase 3200 participants (Anticipated)Interventional2018-07-01Recruiting
A Study of Therapy for Pediatric Relapsed or Refractory Acute Lymphoblastic Leukemia[NCT00186875]Phase 247 participants (Actual)Interventional2003-11-30Completed
Clofarabine Plus Low-Dose Cytarabine for the Treatment of Patients With Higher-Risk Myelodysplastic Syndrome (MDS) Who Have Been Relapsing After, or Are Refractory to, Hypomethylator Therapy[NCT01444742]Phase 281 participants (Actual)Interventional2011-11-16Completed
A Phase 2, Open-label, Randomized, Two-stage Clinical Study of Alvocidib in Patients With Relapsed/Refractory Acute Myeloid Leukemia Following Treatment With Venetoclax Combination Therapy[NCT03969420]Phase 211 participants (Actual)Interventional2020-01-15Terminated(stopped due to Business decision to terminate the development of alvocidib program on 17 November 2020. Patients permitted on treatment until April 22, 2021. The last end-of-treatment follow-up completed on May 14, 2021.)
A Phase 2 Study of E7070, Idarubicin and Cytarabine in Patients With Relapsed or Refractory Acute Myeloid Leukemia and High-risk Myelodysplastic Syndromes[NCT01692197]Phase 243 participants (Actual)Interventional2013-02-01Completed
Pilot Study of Vosaroxin and Cytarabine for the Treatment of Adults 60 Years of Age or Older With Previously Untreated AML[NCT02485353]2 participants (Actual)Interventional2015-10-20Terminated(stopped due to Safety concerns)
A Phase 1b/2 Study of Entospletinib (GS-9973) Monotherapy and in Combination With Chemotherapy in Patients With Acute Myeloid Leukemia (AML)[NCT02343939]Phase 1/Phase 2148 participants (Actual)Interventional2015-07-01Terminated
A Single-arm, Single-center Trial of Prophylactic Tocilizumab for Acute GVHD Prevention After Haploidentical HSCT.[NCT04688021]Phase 246 participants (Anticipated)Interventional2020-12-03Recruiting
Induction Therapy With Pacritinib Combined With Decitabine or Cytarabine in Older Patients With Acute Myeloid Leukemia (AML)[NCT02532010]Phase 213 participants (Actual)Interventional2015-06-15Terminated(stopped due to The study was put on clinical hold by the sponsor since Feb 9th 2016, and was later decided not to re-open due to financial constraints.)
Efficacy and Safety of ATO Plus ATRA in Nucleophosmin-1 Mutated Acute Myeloid Leukemia[NCT03031249]Phase 1/Phase 280 participants (Anticipated)Interventional2017-02-08Recruiting
A Therapeutic Trial of Decitabine and Vorinostat in Combination With Chemotherapy (Vincristine, Prednisone, Doxorubicin and PEG-Asparaginase) for Relapsed/Refractory Acute Lymphoblastic Leukemia (ALL) or Lymphoblastic Lymphoma (LL)[NCT00882206]Phase 215 participants (Actual)Interventional2009-04-30Terminated(stopped due to Slow accrual)
A Phase III Randomised, Double-blind, Controlled, Parallel Group Study of Intravenous Volasertib in Combination With Subcutaneous Low-dose Cytarabine vs. Placebo + Low-dose Cytarabine in Patients >=65 Years With Previously Untreated Acute Myeloid Leukaemi[NCT01721876]Phase 3666 participants (Actual)Interventional2013-01-29Completed
Phase II Evaluation of Gallium Nitrate (NSC 15200) in Non-Hodgkin's Lymphoma in Patients With Acquired Immunodeficiency Syndrome[NCT00002578]Phase 235 participants (Anticipated)Interventional1994-08-31Completed
Comparison of Ara-c 12 gm/m2 vs 18 gm/m2 Per Cycle for 3 Cycles Each as Consolidation in AML ; An Open Label Randomized Non-inferiority Study[NCT01615757]Phase 3180 participants (Anticipated)Interventional2012-08-31Recruiting
A Phase II Study Incorporating Sorafenib (NSC 724772) Into the Therapy of Patients >/= 60 Years of Age With FLT3 Mutated Acute Myeloid Leukemia[NCT01253070]Phase 254 participants (Actual)Interventional2011-04-01Completed
Phase II Study of Hyper-CVAD Plus Imatinib Mesylate (Gleevec, STI571) for Philadelphia-Positive Acute Lymphocytic Leukemia[NCT00038610]Phase 254 participants (Actual)Interventional2001-03-31Completed
Randomized Phase II Study of Clofarabine Alone Versus Clofarabine in Combination With Low-Dose Cytarabine in Previously Untreated Patients >= 60 Years With AML and High-Risk MDS[NCT00088218]Phase 295 participants (Actual)Interventional2004-07-31Completed
Phase II Trial of Clofarabine and Cytarabine in Relapsed Standard-Risk AML and Untreated High-Risk MDS in Adult Patients, and Untreated AML in Selected Elderly Patients at High Risk of Anthracycline Toxicity[NCT00334074]Phase 230 participants (Actual)Interventional2005-08-31Completed
Phase 2 Study Of Clofarabine With High Dose Cytarabine And G-CSF Priming In Adult Patients Less Than Age 65 With Newly Diagnosed Acute Myeloid Leukemia Or Advanced Myelodysplastic Syndrome and/or Advanced Myeloproliferative Neoplasm[NCT01101880]Phase 250 participants (Actual)Interventional2010-08-31Completed
Phase 2 Study of Mitoxantrone, Etoposide, and Cytarabine (MEC) Plus Lenalidomide for the Treatment of Adult Patients With Relapsed or Refractory Acute Myeloid Leukemia[NCT03118466]Phase 241 participants (Actual)Interventional2017-09-25Active, not recruiting
A Phase 1 Trial of the CD123 X CD3 DART Molecule Flotetuzumab (NSC#808294) in Children, Adolescents, and Young Adults With Relapsed or Refractory Acute Myeloid Leukemia[NCT04158739]Phase 116 participants (Actual)Interventional2020-01-22Active, not recruiting
A Phase I/II, Multicenter, Open-Label, Multi-Arm Study Evaluating the Safety, Tolerability, Pharmacokinetics, and Preliminary Activity of Idasanutlin in Combination With Either Chemotherapy or Venetoclax in the Treatment of Pediatric and Young Adult Patie[NCT04029688]Phase 1/Phase 2183 participants (Anticipated)Interventional2020-01-27Recruiting
B-NHL 2013 - Treatment Protocol of the NHL-BFM and the NOPHO Study Groups for Mature Aggressive B-cell Lymphoma and Leukemia in Children and Adolescents[NCT03206671]Phase 3650 participants (Anticipated)Interventional2017-08-03Recruiting
Treatment Protocol for Relapsed Anaplastic Large Cell Lymphoma of Childhood and Adolescence[NCT00317408]96 participants (Anticipated)Interventional2004-04-30Active, not recruiting
A Randomized, Open Label, Phase 2 Study of the Selective Inhibitor of Nuclear Export (SINE) Selinexor (KPT-330) Versus Specified Physician's Choice in Patients ≥ 60 Years Old With Relapsed or Refractory Acute Myeloid Leukemia (AML) Who Are Ineligible for [NCT02088541]Phase 2317 participants (Actual)Interventional2014-03-31Completed
AUTOLOGOUS, ALLOGENEIC, OR SYNGENEIC BONE MARROW TRANSPLANTATION IN HODGKIN'S DISEASE, NON-HODGKIN'S LYMPHOMA, AND MULTIPLE MYELOMA[NCT00002552]Phase 240 participants (Anticipated)Interventional1993-10-31Completed
AUTOLOGOUS STEM CELL TRANSPLANTATION FOR ACUTE MYELOID LEUKEMIA IN SECOND REMISSION: A PHASE II STUDY[NCT00002768]Phase 251 participants (Actual)Interventional1996-06-30Completed
Chemotherapy and Azidothymidine, With or Without Radiotherapy, for High Grade Lymphoma in AIDS-Risk Group Members[NCT00000703]45 participants InterventionalCompleted
A PHASE III STUDY IN CHILDREN WITH UNTREATED ACUTE MYELOGENOUS LEUKEMIA (AML) OR MYELODYSPLASTIC SYNDROME (MDS)[NCT00002798]Phase 3880 participants (Actual)Interventional1996-08-31Completed
EXTRAMEDULLARY RELAPSE AND OCCULT BONE MARROW INVOLVEMENT IN CHILDHOOD ACUTE LYMPHOBLASTIC LEUKEMIA: A PHASE III GROUP-WIDE STUDY[NCT00002816]Phase 3120 participants (Anticipated)Interventional1996-12-31Completed
TREATMENT OF ADULT ACUTE LYMPHOBLASTIC LEUKEMIA: PHASE II TRIALS OF AN INDUCTION REGIMEN INCLUDING PEG-L-ASPARAGINASE, WITH OR WITHOUT PIXY, IN PREVIOUSLY UNTREATED PATIENTS, FOLLOWED BY ALLOGENEIC BONE MARROW TRANSPLANTATION OR FURTHER CHEMOTHERAPY IN FI[NCT00002665]Phase 250 participants (Anticipated)Interventional1995-07-31Completed
INDUCTION WITH ALL-TRANS RETINOIC ACID IN COMBINATION WITH IDARUBICIN AND INTENSIVE CONSOLIDATION FOLLOWED BY BONE MARROW TRANSPLANTATION OR A RANDOMIZED MAINTENANCE TREATMENT DEPENDING UPON THE AMOUNT OF MINIMAL RESIDUAL DISEASE IN ACUTE PROMYELOCYTIC LE[NCT00002701]Phase 3750 participants (Anticipated)Interventional1995-10-31Active, not recruiting
A Randomised Study Comparing an Oral Regimen (Idarubicin and Etoposide) With an Intravenous Regimen (MAE) for Consolidation in Patients Over 55 Years With Acute Myeloid Leukaemia in First Complete Remission[NCT00003602]Phase 3400 participants (Anticipated)Interventional1998-03-31Active, not recruiting
A Phase II Study of Intensive Methotrexate and Cytarabine Followed by High Dose Beam Chemotherapy With Autologous Peripheral Blood Progenitor Cell Transplantation in Patients With Newly Diagnosed Primary Central Nervous System Lymphoma[NCT00003632]Phase 230 participants (Anticipated)Interventional1998-09-30Completed
CAMP-010: PHASE I/II STUDY OF IN VIVO PURGING FOLLOWED BY HIGH DOSE CHEMOTHERAPY, AUTOLOGOUS HEMATOPOIETIC STEM CELL INFUSION AND IMMUNOTHERAPY IN PATIENTS WITH CHRONIC MYELOID LEUKEMIA[NCT00002761]Phase 1/Phase 20 participants (Actual)Interventional1996-02-29Withdrawn(stopped due to PI left institution)
Treatment of Newly Diagnosed Acute Lymphoblastic Leukemia in Infants Less Than 1 Year of Age.[NCT00002785]Phase 20 participants Interventional1996-07-31Completed
A Phase I Study of G3139 (NSC 683428) in Combination With Salvage Chemotherapy for Treatment of Refractory and Relapsed Acute Myeloid Leukemia (AML) and Acute Lymphoblastic Leukemia (ALL)[NCT00004862]Phase 124 participants (Actual)Interventional1999-10-31Completed
The Value of Dexamethasone Versus Prednisolone During Induction and Maintenance Therapy of Prolonged Versus Conventional Duration of L-Asparaginase Therapy During Consolidation and Late Intensification, and of Corticosteroid + VCR Pulses During Maintenanc[NCT00003728]Phase 31,500 participants (Anticipated)Interventional1998-12-31Active, not recruiting
Treatment of Relapsed Acute Myelogenous Leukemia Consisting of Intermediate Dose Cytosine Arabinoside (ARA-C) Plus Interspaced Continuous Infusion Idarubicin, Followed by Continuous Infusion of Low-Dose ARA-C, A Phase II Study by the EORTC-LCG[NCT00003758]Phase 260 participants (Anticipated)Interventional1998-12-31Active, not recruiting
An Investigator-Initiated Study To Evaluate Ara-C and Idarubicin in Combination With the Selective Inhibitor Of Nuclear Export (SINE) Selinexor (KPT-330) in Patients With Relapsed Or Refractory AML[NCT02249091]Phase 242 participants (Actual)Interventional2014-09-30Completed
Use of G-CSF Stimulated HLA-Identical Allogeneic Peripheral Blood Stem Cells for Patients With High Risk Acute Myelogenous Leukemia or CML in Blast Crisis[NCT00002833]Phase 253 participants (Actual)Interventional1994-10-31Completed
PROSPECTIVE RANDOMISED STUDY TO COMPARE LOW-DOSE INTERFERON-ALPHA-n1 (WELLFERON) +/- HYDROXYUREA VS HIGH-DOSE INTERFERON-ALPHA-n1 (WELLFERON) +/- HYDROXYUREA IN PATIENTS WITH NEWLY DIAGONISED CHRONIC PHASE CHRONIC MYELOID LEUKAEMIA[NCT00002869]Phase 3800 participants (Anticipated)Interventional1995-04-30Active, not recruiting
A Phase I/II Combination Study of Topotecan, Fludarabine, Cytosine Arabinoside and G-CSF (T-FLAG) Induction Therapy in Patients With Poor Prognosis AML, MDS and Relapsed/Refractory ALL Followed by Maintenance of Either PBSC Transplant or 13 Cis-Retinoic A[NCT00003619]Phase 1/Phase 20 participants Interventional1998-02-28Completed
The Unrelated Donor Marrow Transplantation Trial[NCT00003187]Phase 2/Phase 319 participants (Actual)Interventional1995-05-31Completed
A Phase II Study of High Dose Late Intensification Therapy in Patients With Chemotherapy Sensitive Multiple Myeloma[NCT00004903]Phase 20 participants Interventional1999-10-31Active, not recruiting
Phase 2, Randomized, Biomarker-driven Clinical Study in Patients With Relapsed or Refractory Acute Myeloid Leukemia (AML) With MCL-1 Dependence ≥30%[NCT02520011]Phase 2104 participants (Actual)Interventional2016-03-14Terminated(stopped due to Due to slow enrollment, the extensive time projected to conclude the study hypothesis rendered the study no longer reasonably feasible to complete.)
A Phase I Study of Metformin and Cytarabine for the Treatment of Relapsed/Refractory Acute Myeloid Leukemia[NCT01849276]Phase 12 participants (Actual)Interventional2015-03-11Terminated(stopped due to Slow accrual)
Augmented Berlin-Frankfurt-Munster Therapy Plus Ofatumumab for Young Adults With Acute Lymphoblastic Leukemia or Lymphoblastic Lymphoma[NCT02419469]Phase 21 participants (Actual)Interventional2015-11-13Terminated(stopped due to Slow Accrual)
Phase II Trial of Intensive, Short-Course Combination Chemotherapy in the Treatment of Newly Diagnosed Patients With Poor-Risk Nonlymphoblastic Lymphoma and Acute B-Lymphoblastic Leukemia and in Patients With Recurrent Non-Hodgkin's Lymphoma[NCT00002471]Phase 20 participants Interventional1990-02-28Completed
RANDOMIZED PHASE III STUDY OF INDUCTION (ICE VS MICE VS DCE) AND INTENSIVE CONSOLIDATION (IDIA VS NOVIA VS DIA) FOLLOWED BY BONE MARROW TRANSPLANTATION IN ACUTE MYELOGENOUS LEUKEMIA: AML 10 PROTOCOL[NCT00002549]Phase 31,520 participants (Anticipated)Interventional1993-11-30Active, not recruiting
PHASE II TRIAL OF POST-REMISSION THERAPY WITH HuM195 AND CYTOTOXIC CHEMOTHERAPY FOR ACUTE PROMYELOCYTIC LEUKEMIA[NCT00002609]Phase 240 participants (Actual)Interventional1994-08-31Completed
PROSPECTIVE CONTROLLED STUDY FOR THE OPTIMIZATION OF THERAPY IN CHRONIC MYELOID LEUKEMIA (CML): MULTICENTRIC STUDY FOR THE EVALUATION OF INTERFERON ALPHA VS ALLOGENIC BM TRANSPLANTATION WITH CHEMOTHERAPY IN CML[NCT00002771]Phase 3750 participants (Anticipated)Interventional1995-01-31Active, not recruiting
High-Dose Cytarabine and Idarubicin Induction, High Dose Etoposide and Cyclophosphamide Intensification, Autologous Stem Cell Transplantation and Interleukin-2 Immune Modulation in Previously Untreated De Novo and Secondary Adult Myeloid Leukemia[NCT00002945]Phase 361 participants (Actual)Interventional1996-12-31Completed
A Phase II Study of Subcutaneous Bortezomib in Combination With Chemotherapy (VXLD) for Relapsed/Refractory Adult Acute Lymphoblastic Leukemia[NCT01769209]Phase 218 participants (Actual)Interventional2013-03-31Completed
Phase II Study of Decitabine and Cytarabine for Older Patients With Newly Diagnosed AML[NCT01829503]Phase 244 participants (Actual)Interventional2013-02-28Completed
A Phase II Study of Clofarabine and Cytarabine for Patients With Newly Diagnosed Acute Myeloid Leukemia Who Have Persistent Disease After Treatment With an Anthracycline and Cytarabine[NCT01960387]Phase 22 participants (Actual)Interventional2013-10-31Terminated(stopped due to lack of accrual)
A Phase II Study of Melphalan HCl for Injection (Propylene Glycol-free), Combined With Carmustine, Etoposide, and Cytarabine (BEAM Regimen) for Myeloablative Conditioning in Lymphoma Patients Undergoing Autologous Stem Cell Transplantation[NCT01969435]Phase 250 participants (Actual)Interventional2014-03-19Completed
A Phase 1/2 Trial of G-CSF, Cladribine, Cytarabine, and Dose-Escalated Mitoxantrone (G-CLAM) in Adults With Newly Diagnosed or Relapsed/Refractory Acute Myeloid Leukemia (AML) or High-Risk Myelodysplastic Syndromes (MDS)[NCT02044796]Phase 1/Phase 2199 participants (Actual)Interventional2014-01-23Completed
Phase 1 Study of the Dual MDM2/MDMX Inhibitor ALRN-6924 in Pediatric Cancer[NCT03654716]Phase 121 participants (Actual)Interventional2018-11-01Completed
A Phase II Study of Induction (Daunorubicin/Cytarabine) and Consolidation (High-Dose Cytarabine) Chemotherapy Plus Dasatinib (NSC #732517) and Continuation Therapy With Dasatinib Alone in Newly Diagnosed Patients With Core Binding Factor Acute Myeloid Leu[NCT01238211]Phase 261 participants (Actual)Interventional2010-12-14Completed
Treatment Protocol for Patients With Standard Risk Acute Myelogenous Leukemia and Its Variants: Induction Using High-Dose Cytarabine, Mitoxantrone and Ethyol; Consolidation With Cytarabine and Idarubicin and Maintenance With 13 Cis Retinoic Acid and Alpha[NCT00003405]Phase 20 participants (Actual)Interventional1998-04-30Withdrawn(stopped due to No enrollment)
Phase 3 Multicenter Randomized Trial to Evaluate Efficacy and Safety of CPI-613 in Combination With HD Cyt. and Mito. vs HD Cyt. and Mito. Therapy and Control Sub-groups in Older Patients With R/R AML[NCT03504410]Phase 3200 participants (Actual)Interventional2018-11-12Terminated(stopped due to Futile)
Non-Hodgkin's Lymphoma T Cell Protocol[NCT00003423]Phase 3100 participants (Anticipated)Interventional1995-05-31Active, not recruiting
Acute Myeloid Leukemia Salvage Therapy for Patients in First Relapse or Who Fail to Achieve an Initial Remission or Who Develop AML as a Second Malignant Neoplasm[NCT00002805]Phase 2115 participants (Actual)Interventional1997-08-31Completed
A Randomized Prospective Study of Early Intensification Versus Alternating Triple Therapy for Patients With Poor Prognosis Lymphoma[NCT00002835]Phase 3116 participants (Actual)Interventional1995-10-30Completed
HIGH INTENSITY, BRIEF DURATION CHEMOTHERAPY FOR RELAPSED OR REFRACTORY ALL: A PHASE II STUDY OF A MULTIDRUG REGIMEN[NCT00002865]Phase 225 participants (Actual)Interventional1995-04-30Completed
A Phase II Trial of Multiple Cycles of Sequential High Dose Chemotherapy for Patients With Chemotherapy Sensitive Relapsed Non-Hodgkin's Lymphoma[NCT00003957]Phase 23 participants (Actual)Interventional1998-12-31Completed
Pilot Study of Intensive Chemotherapy Followed by Peripheral Blood Stem Cell Harvesting for Autotransplantation of Adults With Chronic Myelogenous Leukemia and High Risk Acute Leukemia[NCT00004905]Phase 20 participants Interventional1999-10-31Completed
"Clinical Protocol for a Phase II Study of Leridistim (SC-70935) in Adult Patients (Age>55) With Acute Myeloid Leukemia (AML) Receiving Chemotherapy With the Cytarabine and Daunorubicin 7+3 Regimen"[NCT00004215]Phase 20 participants Interventional1999-08-31Completed
Phase II Study of Fludarabine + Idarubicin + Aracytine in Refractory or Relapsed ALL in Children[NCT00003729]Phase 213 participants (Actual)Interventional1998-12-31Terminated(stopped due to low accrual)
Phase I Study of UCN-01 and Cytarabine (ARA-C) in Patients With Acute Myelogenous Leukemia, and Myelodysplastic Syndromes[NCT00004263]Phase 116 participants (Actual)Interventional1999-12-31Completed
ALinC 17: Continuous Intensification for Very High Risk Acute Lymphocytic Leukemia (A.L.L.): A Pediatric Oncology Group Pilot Study[NCT00003783]Phase 236 participants (Actual)Interventional1999-03-31Completed
Randomized Study of Rituximab (Mabthera) in Patients With Relapsed Follicular Lymphoma Prior to High-Dose Therapy as In Vivo Purging and to Maintain Remission Following High-Dose Therapy[NCT00005589]Phase 3460 participants (Anticipated)Interventional1999-10-31Completed
A Phase II Trial of Cisplatinum, Cytosine Arabinoside, Dexamethasone (DHAP) With Rituxan in Patients With Relapsed CD20+ B-Cell Non-Hodgkin's Lymphoma[NCT00005601]Phase 258 participants (Actual)Interventional2000-10-31Completed
A Phase I/II Study of Induction Chemotherapy With Daunorubicin, Cytarabine, Topotecan and Etoposide (DATE) for De Novo AML: In the Treatment of Young Patients Ages 16-59[NCT00005793]Phase 1/Phase 241 participants (Actual)Interventional1999-07-31Completed
Evaluation of Pretargeted Anti-CD20 Radioimmunotherapy Combined With BEAM Chemotherapy and Autologous Stem Cell Transplantation for High-Risk B-Cell Malignancies[NCT02483000]Phase 13 participants (Actual)Interventional2017-02-01Terminated(stopped due to Closed early due to lack of funding)
A Pilot Study of Mitoxantrone-Based Four Drug Reinduction in Combination With Bortezomib for Relapsed or Refractory Acute Lymphoblastic Leukemia or Lymphoblastic Lymphoma in Children and Young Adults[NCT02535806]Phase 22 participants (Actual)Interventional2015-07-31Terminated(stopped due to Funding source discontinued)
Phase I/II Study of Idarubicin, Cytarabine, and Nivolumab in Patients With High-Risk Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukemia (AML)[NCT02464657]Phase 1/Phase 244 participants (Actual)Interventional2015-07-31Completed
Phase 3 Randomized Trial of DFP-10917 vs Non-Intensive Reinduction (LoDAC, Azacitidine, Decitabine, Venetoclax Combination Regimens) or Intensive Reinduction (High & Intermediate Dose Cytarabine Regimens) for Acute Myelogenous Leukemia Patients in Second,[NCT03926624]Phase 3450 participants (Anticipated)Interventional2019-11-22Recruiting
Autologous Bone Marrow Transplantation for Non-M3 Acute Myeloid Leukemia (AML) in First Remission in Patients NCT00534469]Phase 260 participants (Actual)Interventional2000-02-08Active, not recruiting
Phase Ib Trial of Gilteritinib in Combination With Mitoxantrone, Cladribine, Cytarabine and Filgrastim (GM-CLAG) for Relapsed/Refractory FLT3-mutated Acute Myeloid Leukemia[NCT05330377]Phase 10 participants (Actual)Interventional2023-03-31Withdrawn(stopped due to Sponsor withdrew funding)
A Phase II Study of Bortezomib and Vorinostat in Patients With Refractory or Relapsed MLL Rearranged Hematologic Malignancies[NCT02419755]Phase 212 participants (Actual)Interventional2015-04-14Terminated(stopped due to Accrual goals were no longer feasible based on restrictions imposed by the DSMB.)
Treatment Optimization in Adult Patients With Newly Diagnosed Acute Lymphoblastic Leukemia (ALL) or Lymphoblastic Lymphoma by Individualised, Targeted and Intensified Treatment - a Phase IV-trial With a Phase III-part to Evaluate Safety and Efficacy of Ne[NCT02881086]Phase 31,000 participants (Actual)Interventional2016-08-31Active, not recruiting
A Phase 1b Dose-escalation Study of SGN-CD33A in Combination With Standard-of-care for Patients With Newly Diagnosed Acute Myeloid Leukemia[NCT02326584]Phase 1116 participants (Actual)Interventional2014-12-31Completed
AN OPEN-LABEL, RANDOMIZED PHASE 3 STUDY OF INOTUZUMAB OZOGAMICIN COMPARED TO A DEFINED INVESTIGATOR'S CHOICE IN ADULT PATIENTS WITH RELAPSED OR REFRACTORY CD22-POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA (ALL)[NCT01564784]Phase 3326 participants (Actual)Interventional2012-08-02Completed
A Phase I-II Study of Oxaliplatin, Fludarabine, and Cytarabine in Patients With Acute Myeloid Leukemia or High-Risk Myelodysplastic Syndromes at First Relapse With Complete Remission Duration < 1 Year[NCT00480987]Phase 1/Phase 227 participants (Actual)Interventional2007-07-31Terminated(stopped due to Lack of support.)
Efficacy and Safety of Cladribine in Combination With G-CSF, Low-dose Cytarabine and Pegaspargase in Patients With Refractory/Relapsed Acute Lymphoblastic Leukemia: a Phase 2 Clinical Trial[NCT03318419]Phase 250 participants (Anticipated)Interventional2016-01-01Recruiting
The Feasibility of Safely Managing Patients Receiving Induction With Liposomal Daunorubicin and Cytarabine (CPX-351) for Acute Myeloid Leukemia (AML) in an Outpatient Environment[NCT03988205]Phase 43 participants (Actual)Interventional2019-08-28Terminated(stopped due to Low accrual)
NHL16: Study For Newly Diagnosed Patients With Acute Lymphoblastic Lymphoma[NCT01451515]Phase 223 participants (Actual)Interventional2012-05-25Completed
A Multi-center Randomized Open-label Clinical Trial of Ara-C, Aclarubicin Combined With PEG-G-CSF for Initial Treatment of Acute Myeloid Leukemia Patients[NCT03045627]Phase 2120 participants (Anticipated)Interventional2017-01-31Recruiting
A Phase 1/2, Open-label Randomized Study of Ulocuplumab (BMS-936564) in Combination With Low Dose Cytarabine in Subjects With Newly Diagnosed Acute Myeloid Leukemia[NCT02305563]Phase 1/Phase 270 participants (Actual)Interventional2015-01-27Terminated(stopped due to Business objectives have changed, slow accrual, the standard of care for the patient population changed and we were unable to accrue any longer.)
B-cell Mature Non-Hodgkin's Lymphoma Treatment Protocol in Children and Adolescents 2021 (B-NHL-M-2021)[NCT05518383]Phase 4300 participants (Anticipated)Interventional2022-05-25Recruiting
RANDOMIZED PHASE III STUDY TO EVALUATE THE VALUE OF rHuG-CSF IN INDUCTION AND OF AN ORAL SCHEDULE AS CONSOLIDATION TREATMENT IN ELDERLY PATIENTS WITH ACUTE MYELOGENOUS LEUMEKIA (AML-13 PROTOCOL)[NCT00002719]Phase 3500 participants (Anticipated)Interventional1995-12-31Completed
Phase I-II Study of Crenolanib Combined With Standard Salvage Chemotherapy, and Crenolanib Combined With 5-Azacitidine in Acute Myeloid Leukemia Patients With FLT3 Activating Mutations[NCT02400281]Phase 1/Phase 228 participants (Actual)Interventional2015-09-30Completed
Allogeneic Hematopoietic Stem Cell Transplantation as Initial Salvage Therapy for Patients With Primary Induction Failure Acute Myeloid Leukemia Refractory to High-Dose Cytarabine-Based Induction Chemotherapy[NCT02441803]Phase 211 participants (Actual)Interventional2015-09-14Active, not recruiting
A Multicenter,Open-label,Randomized Study on the Treatment of Elderly Chinese Acute Myeloid Leukemia Patients Aged 65 to 75 Years Old[NCT02432911]300 participants (Anticipated)Interventional2015-04-30Recruiting
A Phase I - II Study to Assess Safety and Efficacy of the Combination of Ponatinib With High or Intermediate-Dose Cytarabine as Consolidation Therapy for Patients With Intermediate-Risk Cytogenetic FLT3-ITD AML iIn First Complete Remission[NCT02428543]Phase 1/Phase 249 participants (Actual)Interventional2013-07-31Active, not recruiting
Phase I Study of Bryostatin 1 (NSC 339555) and High-Dose 1-Beta-D-Arabinofuranosylcytosine (HiDAC) in Patients With Refractory Leukemia[NCT00003079]Phase 130 participants (Actual)Interventional1997-09-30Completed
Phase 1 Study of Deferasirox in Acute Leukemia Patients Not Treated by Standard Chemotherapy Regimens[NCT02413021]Phase 140 participants (Anticipated)Interventional2016-05-31Not yet recruiting
A Phase 3, Randomized, Double-blind, Placebo-controlled Study to Assess the Efficacy and Safety of Entospletinib in Combination With Intensive Induction and Consolidation Chemotherapy in Adults With Newly Diagnosed Nucleophosmin 1-mutated Acute Myeloid Le[NCT05020665]Phase 315 participants (Actual)Interventional2021-11-30Terminated(stopped due to Termination was due to significant challenges associated with study enrollment in a genetic subset of fit participants in the front-line acute myeloid leukemia (AML) setting and other challenges associated with post-COVID impacts.)
Treatment of Mature B-ALL and Burkitt Lymphoma (BL) in Adult Patients. BURKIMAB-14.[NCT05049473]Phase 2100 participants (Anticipated)Interventional2014-01-31Recruiting
A Randomized Controlled Study of Dasatinib Combined With Reduced Intensive Consolidation Chemotherapy in Newly Diagosed Philadelphia Chromesome Positive Adult Lymphoblastic Leukemia[NCT05026229]60 participants (Anticipated)Interventional2021-09-06Recruiting
Phase II CCOP Trial of High Dose Methotrexate/ARA-C and HCVAD for Newly Diagnosed Nodular and Diffuse Mantle Cell Lymphoma and Their Blastic Variants[NCT00003311]Phase 219 participants (Actual)Interventional1998-05-20Completed
A Randomized 3-Arm Phase II Study Comparing 1.) Bendamustine, Rituximab and High Dose Cytarabine (BR/CR) 2.) Bendamustine, Rituximab, High Dose Cytarabine and Acalabrutinib (BR/CR-A), and 3.) Bendamustine, Rituximab and Acalabrutinib (BR-A) in Patients &l[NCT04115631]Phase 2360 participants (Actual)Interventional2019-12-13Active, not recruiting
A Phase II Randomized Trial of Immunologic and Chemotherapeutic Agents for Treatment of Patients With Relapsed or Refractory Acute Myelogenous Leukemia[NCT00005962]Phase 20 participants Interventional2000-10-04Completed
PROSPECTIVE RANDOMISED STUDY TO COMPARE INTERFERON-ALPHA-n1 (WELLFERON) VS 'IDAC' CHEMOTHERAPY AND AUTOGRAFTING FOLLOWED BY INTERFERON ALPHA-n1 (WELLFERON) IN PATIENTS WITH NEWLY DIAGNOSED CHRONIC PHASE CHRONIC MYELOID LEUKEMIA[NCT00002868]Phase 3744 participants (Anticipated)Interventional1997-11-20Completed
A Randomized Phase II Trial of R-HCVAD/MTX/ARA-C Induction Followed by Consolidation With an Autologous Stem Cell Transplant Vs. R-Bendamustine Induction Followed by Consolidation With an Autologous Stem Cell Transplant for Patients ≤ 65 Years of Age With[NCT01412879]Phase 253 participants (Actual)Interventional2011-11-30Completed
A Phase 1/2 Study of Venetoclax in Combination With Low-Dose Cytarabine in Treatment-Naïve Subjects With Acute Myelogenous Leukemia Who Are ≥ 60 Years of Age and Who Are Not Eligible for Standard Anthracycline-Based Induction Therapy[NCT02287233]Phase 1/Phase 294 participants (Actual)Interventional2014-12-31Completed
Bortezomib Combined With Fludarabine and Cytarabine for Mantle Cell Lymphoma Patients:a Single Arm, Open-labelled, Phase 2 Study[NCT03016988]Phase 250 participants (Anticipated)Interventional2017-01-31Not yet recruiting
A Phase II Study of CLAG Regimen in Combination With Imatinib Mesylate (Gleevec) in Relapsed or Refractory Acute Myeloid Leukemia[NCT00955916]Phase 238 participants (Actual)Interventional2009-08-31Completed
A Phase II Study of the Aurora A Kinase Inhibitor Alisertib in Combination With 7+3 Induction Chemotherapy in Patients With High-risk Acute Myeloid Leukemia[NCT02560025]Phase 242 participants (Actual)Interventional2015-12-31Completed
Total Therapy for Infants With Acute Lymphoblastic Leukemia (ALL) I[NCT02553460]Phase 1/Phase 250 participants (Actual)Interventional2016-01-29Active, not recruiting
Phase 1-2 Safety and Efficacy Study of DACOGEN in Sequential Administration With Cytarabine in Children With Relapsed or Refractory Acute Myeloid Leukemia[NCT01853228]Phase 1/Phase 217 participants (Actual)Interventional2013-10-22Terminated(stopped due to EMA/PDCO acknowledged that available results from this study do not support further clinical studies in relapsed/refractory AML paediatric patients.)
A Phase I/II Study Evaluating Escalating Doses of 90Y-BC8-DOTA (Anti-CD45) Antibody Followed by BEAM Chemotherapy and Autologous Stem Cell Transplantation for High-Risk Lymphoid Malignancies[NCT01921387]Phase 1/Phase 220 participants (Actual)Interventional2013-10-09Completed
Dasatinib Plus Multi-agent Chemotherapy for New Diagnosed Philadelphia Chromosome-positive Acute Lymphoblastic Leukemia[NCT02523976]Phase 230 participants (Actual)Interventional2015-08-01Completed
A Randomized Controlled Study on the Efficacy and Safety of MA-BUCY2 Protocol in the Conditioning of Haploidentical Stem Cell Transplantation in Patients With High-risk Acute Myeloid Leukemia[NCT05814731]264 participants (Anticipated)Interventional2023-04-15Not yet recruiting
Multicenter Randomized Controlled Clinical Study of Mitoxantrone Hydrochloride Liposome Injection Combined With Carmustine, Etoposide and Cytarabine (Modified BEAM Protocol) for T Cell Lymphoma Underwent Autologous Stem Cell Transplantation[NCT05814718]122 participants (Anticipated)Interventional2023-04-15Not yet recruiting
A Phase 1 Single-Center Trial Combining Venetoclax With G-CSF, Cladribine, Cytarabine, and Mitoxantrone (CLAG-M) for Patients With AML and High-Grade Myeloid Neoplasms[NCT04797767]Phase 120 participants (Anticipated)Interventional2022-02-04Recruiting
Open-label, Single-arm Trial to Evaluate Antitumor Activity, Safety, and Pharmacokinetics of Isatuximab Used in Combination With Chemotherapy in Pediatric Patients From 28 Days to Less Than 18 Years of Age With Relapsed/Refractory B or T Acute Lymphoblast[NCT03860844]Phase 267 participants (Actual)Interventional2019-08-06Terminated(stopped due to Study was prematurely stopped due to sponsor decision (stage 2 efficacy criteria not met); not due to safety concerns.)
A Phase 1, Open-label, Dose-escalation, Safety and Biomarker Prediction of Alvocidib and Cytarabine/Daunorubicin (7+3) in Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)[NCT03298984]Phase 132 participants (Actual)Interventional2017-09-25Completed
A Phase 3, Multicenter, Open-label, Randomized Study Comparing the Efficacy and Safety of AG-221 (CC-90007) Versus Conventional Care Regimens in Older Subjects With Late Stage Acute Myeloid Leukemia Harboring an Isocitrate Dehydrogenase 2 Mutation[NCT02577406]Phase 3319 participants (Actual)Interventional2015-12-30Active, not recruiting
A Single-Center Phase 1/2 Study of Single- or Fractioned-Dose Gemtuzumab Ozogamicin in Combination With G-CSF, Cladribine, Cytarabine, and Mitoxantrone for Previously Untreated Adult Acute Myeloid Leukemia or High-Grade Myeloid Neoplasm[NCT03531918]Phase 1/Phase 266 participants (Actual)Interventional2018-09-14Completed
A Multicenter Randomized Control Clinical Trail of Evaluating Effect of Demethylation Drug Combined With Chemotherapy in Patients With Intermediate-risk AML After Hematological Complete Remission[NCT03417427]Phase 2100 participants (Anticipated)Interventional2018-02-01Recruiting
A Phase I-II Study of Oxaliplatin, Fludarabine, Cytarabine and Rituximab in Patients With Richter's Transformation, Prolymphocytic Leukemia or Refractory/Relapsed B-Cell Chronic Lymphocytic Leukemia[NCT00452374]Phase 1/Phase 248 participants (Actual)Interventional2004-11-30Completed
Decitabine,Cytarabine(Ara-C) and Arsenic Trioxide(ATO) in the Treatment of Acute Myeloid Leukemia With p53 Mutations[NCT03381781]Phase 2100 participants (Anticipated)Interventional2018-03-31Not yet recruiting
Decitabine Plus Cytarabine for Induction of Remission in Newly Diagnosed Elderly Acute Myeloid Leukemia (AML) and Advanced Myelodysplastic Syndrome (MDS)[NCT02121418]12 participants (Actual)Interventional2014-06-30Completed
A PHASE IB/II STUDY TO EVALUATE THE SAFETY AND EFFICACY OF VISMODEGIB IN RELAPSED/REFRACTORY ACUTE MYELOGENOUS LEUKEMIA (AML) AND RELAPSED/REFRACTORY HIGH-RISK MYELODYSPLASTIC SYNDROME (MDS)[NCT01880437]Phase 238 participants (Actual)Interventional2013-09-30Terminated
Clinical Study on the Efficacy and Safety of Auto-HSCT in Adult Patients With Burkitt Lymphoma, Lymphoblastic Lymphoma, and Acute Lymphoblastic Leukemia Who Received TCCA Conditioning Regimen[NCT06060782]Phase 128 participants (Anticipated)Interventional2023-10-01Recruiting
BeEAM (Bendamustine, Etoposide, Cytarabine, Melphalan) Versus CEM (Carboplatin, Etoposide, Melphalan) in Lymphoma Patients as a Conditioning Regimen Before Autologous Hematopoietic Cell Transplantation[NCT05813132]60 participants (Actual)Interventional2022-12-01Completed
Randomized Phase 2 Trial of CPX-351 (Vyxeos) vs. CLAG-M (Cladribine, Cytarabine, G-CSF, and Mitoxantrone) in Medically Less-Fit Adults With Acute Myeloid Leukemia (AML) or Other High-Grade Myeloid Neoplasm[NCT04195945]Phase 260 participants (Anticipated)Interventional2020-03-11Recruiting
LS1781: Phase 2 Trial of High Dose Intravenous Ascorbic Acid as an Adjunct to Salvage Chemotherapy in Relapsed / Refractory Lymphoma and Patients With Clonal Cytopenia of Undetermined Significance[NCT03418038]Phase 255 participants (Anticipated)Interventional2018-03-23Recruiting
Total Therapy XVII for Newly Diagnosed Patients With Acute Lymphoblastic Leukemia and Lymphoma[NCT03117751]Phase 2/Phase 3790 participants (Actual)Interventional2017-03-29Active, not recruiting
A Phase III Randomized Trial for Newly Diagnosed High Risk B-Lymphoblastic Leukemia (B-ALL) Including a Stratum Evaluating Dasatinib (NSC#732517) in Patients With Ph-like Tyrosine Kinase Inhibitor (TKI) Sensitive Mutations[NCT02883049]Phase 35,937 participants (Actual)Interventional2012-02-29Active, not recruiting
Phase II Trial of Vosaroxin in Combination With Infusional Cytarabine in Patients With Untreated AML[NCT02658487]Phase 242 participants (Actual)Interventional2016-03-31Active, not recruiting
A Study of Combination of Venetoclax, Hypomethylation Agent and Low-dose Cytarabine as a Salvage Therapy in Patients With Acute Myeloid Leukemia Who Had Relapsed/Refractory Disease or Positive Minimal Residual Disease[NCT05362942]Phase 252 participants (Anticipated)Interventional2022-05-01Not yet recruiting
An Expanded Access Study for Venetoclax in Combination With Low-Dose Cytarabine in Treatment-Naïve Subjects With Acute Myeloid Leukemia Who Are Ineligible for Intensive Induction Therapy in Japan[NCT04509622]Phase 314 participants (Actual)Interventional2020-10-05Completed
Phase III, Multicenter, Randomized, Trial of CPX-351 (Cytarabine:Daunorubicin) Liposome Injection Versus Cytarabine and Daunorubicin in Patients 60-75 Years of Age With Untreated High Risk (Secondary) AML[NCT01696084]Phase 3309 participants (Actual)Interventional2012-12-13Completed
A Phase 1/2 Study of Sequential Idarubicin + Cytarabine, Followed by Lenalidomide, in Patients With Myelodysplastic Syndrome (RAEB-2) or With Previously Untreated Acute Myeloid Leukemia[NCT00831766]Phase 1/Phase 251 participants (Actual)Interventional2009-06-25Completed
A Phase II Trial of Clofarabine and Cytarabine to Treat Minimal Residual Disease (MRD) in Acute Myeloid Leukemia[NCT00863434]Phase 22 participants (Actual)Interventional2009-02-28Terminated
Consolidation Therapy for Acute Myeloid Leukemia Guided by Leukemia Stem Cell Behavior[NCT01588951]Phase 2/Phase 310 participants (Actual)Interventional2013-12-31Terminated(stopped due to Low accrual)
A Single-arm, Prospective, Single-center, Phase II Clinical Study of Penpulimab Combined With RMA in the Treatment of Newly Diagnosed Primary CNS Lymphoma[NCT05347641]Phase 223 participants (Anticipated)Interventional2022-06-01Not yet recruiting
A Phase I Study of Venetoclax Combined With Vyxeos (CPX-351) for Children, Adolescents and Young Adults With Relapsed or Refractory Acute Leukemia[NCT03826992]Phase 121 participants (Anticipated)Interventional2018-12-27Recruiting
A TACL Phase 1/2 Study of PO Ixazomib in Combination With Chemotherapy for Childhood Relapsed or Refractory Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma IND# 140730[NCT03817320]Phase 1/Phase 231 participants (Anticipated)Interventional2019-02-12Recruiting
A Randomized International Phase 3 Trial of Imatinib and Chemotherapy With or Without Blinatumomab in Patients With Newly-Diagnosed Philadelphia Chromosome-Positive or Philadelphia Chromosome-Like ABL-Class B-Cell Acute Lymphoblastic Leukemia[NCT06124157]Phase 3680 participants (Anticipated)Interventional2024-01-22Not yet recruiting
International Collaborative Treatment Protocol For Children And Adolescents With Acute Lymphoblastic Leukemia[NCT01117441]Phase 36,136 participants (Actual)Interventional2010-06-30Completed
A Multicenter Phase II Study in Adults With Untreated Acute Lymphoblastic Leukemia: Testing Pharmacokinetically Individualized Doses of L-Asparaginase Following the DFCI Pediatric Consortium Protocol[NCT00136435]Phase 2100 participants (Anticipated)Interventional2002-06-30Active, not recruiting
A Dose-finding Phase I Study of the Tumor-targeting Human F16IL2 Monoclonal Antibody-cytokine Fusion Protein in Combination With Very Low-dose Cytarabine in Patients With AML Relapse After Allogeneic Hematopoietic Stem Cell Transplantation[NCT02957032]Phase 130 participants (Anticipated)Interventional2016-04-13Terminated(stopped due to New agents for the same indication impacted on patients' recruitment)
Venetoclax Combining Chidamide and Azacitidine (VCA) Regimen Followed by Dicitabine Combined With Liposome Mitoxantrone, Cytarabine, and G-CSF (D-MAG) Regimen on the Treatment of Elderly Patients With Newly Diagnosed Acute Myeloid Leukemia (AML) : A Multi[NCT05603884]Phase 266 participants (Anticipated)Interventional2022-12-01Recruiting
Prospective, Single-arm, Multicenter Exploratory Clinical Study of the Combination of Etoposide, Cytarabine and PEG-rhG-CSF (EAP Regimen) as First Line Mobilization Regimen of Hematopoietic Stem Cells in Patients With Hematological Malignancies[NCT05536154]68 participants (Anticipated)Interventional2022-11-01Recruiting
Standard Chemotherapy (CHOP Regimen) Versus Sequential High-Dose Chemotherapy With Autologous Stem Cell Transplantation in Patients With Newly Diagnosed Aggressive Non-Hodgkin's Lymphomas and Poor Prognostic Factors: A Randomized Phase III Study (MISTRAL)[NCT00003215]Phase 3400 participants (Anticipated)Interventional1997-04-30Completed
Therapy of Early Chronic Phase Chronic Myelogenous Leukemia (CML) With Alpha Interferon (IFN-A), Low-Dose Cytosine Arabinoside (ARA-C), and Homoharringtonine (HHT)[NCT00003239]Phase 290 participants (Actual)Interventional1998-03-31Completed
A Phase II Study of Daunomycin and ARA-C Given by Continuous IV Infusion With PSC-833 for Previously Untreated Non-M3 Acute Myeloid Leukemia (AML) in Patients of Age 56 or Older[NCT00004217]Phase 255 participants (Anticipated)Interventional2000-02-29Completed
[NCT00004643]Phase 210 participants Interventional1995-02-28Completed
ALINC #17 Treatment for Patients With Low Risk Acute Lymphoblastic Leukemia: A Pediatric Oncology Group Phase III Study[NCT00005585]Phase 3838 participants (Actual)Interventional2000-04-30Completed
ALinC 17: Protocol for Patients With Newly Diagnosed High Risk Acute Lymphoblastic Leukemia (ALL) - Evaluation of the Augmented BFM Regimen: A Phase III Study[NCT00005603]Phase 3276 participants (Actual)Interventional2000-03-31Completed
Protocol for Patients With Newly Diagnosed Better Risk Acute Lymphoblastic Leukemia (ALL): A POG Pilot Study[NCT00003671]Phase 259 participants (Actual)Interventional1998-12-31Completed
Treatment of Poor Risk Myelodysplasia With the Combination of Amifostine, Topotecan and ARA-C: A Phase II Study[NCT00003827]Phase 225 participants (Anticipated)Interventional1999-01-31Active, not recruiting
Prospective Phase II Study of a High Dose, Short Course Regimen (R-CODOX-M/IVAC) Including CNS Penetration and Intensive IT Prophylaxis in HIV-Associated Burkitt's and Atypical Burkitt's Lymphoma[NCT00392834]Phase 234 participants (Actual)Interventional2006-09-30Completed
Phase II Study of Alvocidib (NSC 649890, Flavopiridol) in Timed Sequential Combination With Cytosine Arabinoside (Ara-C) and Mitoxantrone for Adults With Newly Diagnosed, Previously Untreated, Poor-Risk Acute Myelogenous Leukemias[NCT00407966]Phase 245 participants (Actual)Interventional2006-10-31Completed
Randomized Phase II Study Comparing Two Administration Schedules of Flavopiridol (Alvocidib, NSC 649890, IND 46, 211) Given in Timed Sequential Combination With Cytosine Arabinoside (Ara-C) and Mitoxantrone Hydrochloride for Adults With Newly Diagnosed, P[NCT00795002]Phase 278 participants (Actual)Interventional2008-11-30Completed
A Phase II Study of Omacetaxine (OM) and Low Dose Cytarabine (LDAC) in Older Patients With Acute Myelogenous Leukemia (AML) and High-Risk Myelodysplastic Syndrome (MDS)[NCT01272245]Phase 236 participants (Actual)Interventional2011-07-31Completed
Tasigna® (Nilotinib) Plus Multi-Agent Chemotherapy for Newly-Diagnosed Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia[NCT00844298]Phase 291 participants (Actual)Interventional2009-01-31Completed
A Phase II Trial of Ifosfamide, Etoposide, Cytarabine, and Methotrexate (IVAM) Chemotherapy for Refractory or Relapsed Diffuse Large B Cell Lymphoma[NCT03383406]Phase 230 participants (Anticipated)Interventional2016-12-01Enrolling by invitation
A Randomized Phase III Study of Standard Cytarabine Plus Daunorubicin (7+3) Therapy or Idarubicin With High Dose Cytarabine (IA) Versus IA With Vorinostat (NSC-701852) (IA + V) in Younger Patients With Previously Untreated Acute Myeloid Leukemia (AML)[NCT01802333]Phase 3754 participants (Actual)Interventional2013-02-12Completed
A Phase II Study of V-BEAM (Bortezomib, Carmustine, Etoposide, Cytarabine, and Melphalan) as Conditioning Regimen Prior to Second Autologous Stem Cell Transplantation for Multiple Myeloma[NCT01653418]Phase 210 participants (Actual)Interventional2012-09-30Terminated(stopped due to Due to the high rate of morbidity and mortality)
A Phase 3 Randomized Trial of Inotuzumab Ozogamicin (NSC#: 772518) for Newly Diagnosed High-Risk B-ALL; Risk-Adapted Post-Induction Therapy for High-Risk B-ALL, Mixed Phenotype Acute Leukemia, and Disseminated B-LLy[NCT03959085]Phase 34,772 participants (Anticipated)Interventional2019-10-31Recruiting
Mature B-Cell Lymphoma And Leukemia Study III[NCT01046825]Phase 2/Phase 3128 participants (Actual)Interventional2010-09-09Active, not recruiting
Phase II Study on Safety and Activity of a Short Term Intensified Chemo-immunotherapy Combination in HIV-positive Patients Affected by Burkitt Lymphoma[NCT01516593]Phase 219 participants (Actual)Interventional2011-11-30Completed
Risk-stratified Sequential Treatment of Post-transplant Lymphoproliferative Disease (PTLD) With 4 Courses of Rituximab SC Followed by 4 Courses of Rituximab SC, 4 Courses of Rituximab SC Combined With CHOP-21 or 6 Courses of Rituximab SC Combined With Alt[NCT02042391]Phase 260 participants (Actual)Interventional2015-02-03Completed
A Pilot Study of Bendamustine and Rituximab Alternating With Cytarabine and Rituximab for Untreated Mantle Cell Lymphoma[NCT02728531]Phase 118 participants (Actual)Interventional2016-04-18Completed
Phase III Randomized, Double-blind, Placebo-controlled Study Investigating the Efficacy of the Addition of Crenolanib to Salvage Chemotherapy Versus Salvage Chemotherapy Alone in Subjects ≤ 75 Years of Age With Relapsed/Refractory FLT3 Mutated Acute Myelo[NCT03250338]Phase 3322 participants (Anticipated)Interventional2018-06-05Recruiting
Y Zevalin, BEAM and Rituximab In Autologous Stem Cell Transplantation (ASCT) For Lymphoma[NCT01538472]Phase 1/Phase 240 participants (Actual)Interventional2003-09-30Completed
Efficacy and Safety of Decitabine in Combination With Low-dose Cytarabine as Inductive Treatment in Newly Diagnosed Elderly Patients With Acute Myeloid Leukemia[NCT02985372]Phase 330 participants (Anticipated)Interventional2016-12-31Enrolling by invitation
A Phase 3, Randomized, Open-label, Multicenter Study Comparing Ponatinib Versus Imatinib, Administered in Combination With Reduced-Intensity Chemotherapy, in Patients With Newly Diagnosed Philadelphia Chromosome Positive Acute Lymphoblastic Leukemia (Ph+ [NCT03589326]Phase 3245 participants (Actual)Interventional2018-10-04Active, not recruiting
MidOStaurin + Gemtuzumab OzogAmIcin Combination in First-line Standard Therapy for Acute Myeloid Leukemia (MOSAIC)[NCT04385290]Phase 1/Phase 2214 participants (Anticipated)Interventional2020-09-04Recruiting
A Phase I/II Study of CP-4055 in Patients With Refractory/Relapsed Hematologic Malignancies[NCT00405743]Phase 1/Phase 2153 participants (Actual)Interventional2006-05-31Completed
AML-02: Study of the Activity and Safety of the Addition of Omacetaxine to the Standard-of-Care Induction Therapy Regimen of Cytarabine and Idarubicin in Newly-Diagnosed AML Patients[NCT02440568]Phase 1/Phase 222 participants (Actual)Interventional2015-06-05Terminated
Phase I Study of Pacritinib and Chemotherapy in Patients With Acute Myeloid Leukemia and FLT3 Mutations[NCT02323607]Phase 113 participants (Actual)Interventional2016-01-12Completed
Epacadostat With Cladribine and Cytarabine (ECC) in Relapsed / Refractory AML Patients Fit for Intensive Chemotherapy; a Phase I Study.[NCT03491579]Phase 10 participants (Actual)Interventional2018-12-31Withdrawn(stopped due to IMP will not be further developed)
Epacadostat With Idarubicin and Cytarabine (EIC) for First-line Treatment of AML Patients Fit for Intensive Chemotherapy; a Phase I Study[NCT03444649]Phase 10 participants (Actual)Interventional2018-09-30Withdrawn(stopped due to IMP will not be further developed)
International Protocol for the Treatment of Childhood Anaplastic Large Cell Lymphoma[NCT00006455]Phase 3885 participants (Actual)Interventional1999-11-26Completed
Reduced Intensity Conditioning Regimen for Elderly or High Comorbidity Burden Patients Receiving Haploidentical Hematopoietic Stem Cell Transplantation[NCT03412409]Phase 250 participants (Anticipated)Interventional2018-02-01Recruiting
Master Screening and Reassessment Protocol (MSRP) for the NCI MyeloMATCH Clinical Trials[NCT05564390]Phase 2750 participants (Anticipated)Interventional2024-02-01Not yet recruiting
A Randomized Phase II Study Comparing Cytarabine + Daunorubicin (7 + 3) to (Daunorubicin and Cytarabine) Liposome, Cytarabine + Daunorubicin + Venetoclax, and Azacitidine + Venetoclax in Patients Aged 59 or Younger With High-Risk (Adverse) Acute Myeloid L[NCT05554406]Phase 2268 participants (Anticipated)Interventional2024-02-01Not yet recruiting
A Measurable Residual Disease (MRD) Driven, Phase II Study of Venetoclax Plus Chemotherapy for Newly Diagnosed Younger Patients With Intermediate Risk Acute Myeloid Leukemia: A Tier 1 MYELOMATCH Clinical Trial[NCT05554393]Phase 2153 participants (Anticipated)Interventional2023-12-31Not yet recruiting
Treatment of Newly Diagnosed Acute Lymphoblastic Leukemia in Children and Adolescents[NCT03020030]Phase 3560 participants (Actual)Interventional2017-03-03Active, not recruiting
A Phase 3, Open-Label, Multicenter, Randomized Study of SKLB1028 Versus Salvage Chemotherapy in Patients With FLT3-mutated Acute Myeloid Leukemia Refractory to or Relapsed After First-line Treatment(ALIVE)[NCT04716114]Phase 3315 participants (Anticipated)Interventional2021-03-24Recruiting
A Phase I/II Study of Autologous Stem Cell Transplantation Followed by Nonmyeloablative Allogeneic Stem Cell Transplantation for Patients With Relapsed or Refractory Lymphoma - A Multi-center Trial[NCT00005803]Phase 1/Phase 276 participants (Actual)Interventional1999-09-30Completed
A Phase 2 Study of WEE1 Inhibition With AZD1775 Alone or Combined With Cytarabine in Patients With Advanced Acute Myeloid Leukemia and Myelodysplastic Syndrome[NCT02666950]Phase 23 participants (Actual)Interventional2017-05-05Completed
A Prospective, Randomized, Controlled Trial of Decitabine Versus Conventional Chemotherapy for Maintenance Therapy of Patients With Acute Myeloid Leukemia With t(8;21)[NCT03026842]Phase 4180 participants (Anticipated)Interventional2017-01-31Active, not recruiting
Safety and Efficacy of Chemotherapy Combined With Adoptive Transfer of Human Leukocyte Antigen (HLA)-Haploidentical Donor Lymphocyte Infusion (DLI) in Older Patients With Righ-Risk Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS)[NCT02046122]Phase 1/Phase 219 participants (Actual)Interventional2014-07-31Completed
OAG and Decitabine for Newly Diagnosed Acute Myeloid Leukemia Patients Greater Than or Equal to 65 Years of Age[NCT02029417]Phase 22 participants (Actual)Interventional2014-07-31Terminated(stopped due to Major revisions needed in study)
A PHASE 1 STUDY TO EVALUATE THE SAFETY, TOLERABILITY, EFFICACY, PHARMACOKINETICS, AND PHARMACODYNAMICS OF PF-04449913 (GLASDEGIB), AN ORAL HEDGEHOG INHIBITOR, ADMINISTERED AS A SINGLE AGENT IN JAPANESE PATIENTS WITH SELECT HEMATOLOGIC MALIGNANCIES AND IN [NCT02038777]Phase 148 participants (Actual)Interventional2014-03-25Active, not recruiting
A Phase I/II Study of Liposomal Cytarabine and Daunorubicin (CPX-351) in Combination With Quizartinib in Patients With Acute Myeloid Leukemia (AML) and High Risk Myelodysplastic Syndrome (MDS)[NCT04128748]Phase 1/Phase 252 participants (Anticipated)Interventional2020-05-27Recruiting
A Combination of Cladribine, Idarubicin, Cytarabine (CLIA) and Quizartinib for the Treatment of Patients With Newly Diagnosed or Relapsed/Refractory Acute Myeloid Leukemia (AML) and High-Risk Myelodysplastic Syndrome (MDS))[NCT04047641]Phase 1/Phase 280 participants (Anticipated)Interventional2019-10-22Recruiting
A Phase 1b/2 Study of the BCL-2 Inhibitor Venetoclax in Combination With Standard Intensive AML Induction/Consolidation Therapy With FLAG-IDA in Patients With Newly Diagnosed or Relapsed/Refractory AML[NCT03214562]Phase 1/Phase 2116 participants (Anticipated)Interventional2017-09-26Recruiting
A Feasibility Study of Myeloablative BEAM Allogeneic Transplantation Followed by Oral Ixazomib Maintenance Therapy in Patients With Relapsed High-Risk Multiple Myeloma[NCT02504359]Phase 111 participants (Actual)Interventional2015-07-20Completed
A Phase I Study to Evaluate the Safety and Tolerability of Gemtuzumab Ozogamicin and Midostaurin When Used in Combination With Standard Cytarabine and Daunorubicin Induction for Newly Diagnosed FLT3-mutated Acute Myeloid Leukemia[NCT03900949]Phase 118 participants (Anticipated)Interventional2019-03-13Recruiting
Phase II Study of Pedi-cRIB: Mini-Hyper-CVD With Condensed Rituximab, Inotuzumab Ozogamicin and Blinatumomab (cRIB) for Relapsed Therapy for Pediatric With B-Cell Lineage Acute Lymphocytic Leukemia[NCT05645718]Phase 227 participants (Anticipated)Interventional2023-07-14Recruiting
Phase 1 A/B Study of LY2606368 in Combination With Cytarabine and Fludarabine in Patients With Relapsed/Refractory Acute Myelogenous Leukemia (AML) and High-Risk Myelodysplastic Syndrome (HRMDS)[NCT02649764]Phase 115 participants (Actual)Interventional2016-05-04Completed
A Randomized Trial Using a Modified COG ABFM Regimen Backbone to Investigate Capizzi Escalating Methotrexate Versus High Dose Methotrexate in Children With Newly Diagnosed T-cell Lymphoblastic Lymphoma (T-LBL)[NCT05681260]Phase 3200 participants (Anticipated)Interventional2023-02-06Recruiting
Prospective, Single-arm, Multicenter Exploratory Clinical Study of the Combination of Etoposide, Cytarabine and PEG-rhG-CSF (EAP Regimen) on Hematopoietic Stem Cell Mobilization in Poor Mobilization Patients With Hematological Malignancies[NCT05510089]62 participants (Anticipated)Interventional2022-09-01Recruiting
BEAM + 131Iodine-Anti-B1 Radioimmunotherapy and Autologous Hematopoietic Stem Cell Transplantation for the Treatment of Recurrent Non-Hodgkin's Lymphoma[NCT00574509]Phase 134 participants (Actual)Interventional1996-03-04Completed
A Current Practice Study of Rituxan in Patient Receiving BEAM Chemotherapy and Autologous Blood Stem Cell Transplantation for High Risk Lymphoma or Hodgkin's Disease[NCT01702961]75 participants (Actual)Interventional2002-06-30Completed
International Proof of Concept Therapeutic Stratification Trial of Molecular Anomalies in Relapsed or Refractory HEMatological Malignancies in Children, Subprotocol C Ruxolitinib + Venetoclax + Dexamethasone + Cyclophosphamide and Cytarabine in Pediatric [NCT05745714]Phase 1/Phase 226 participants (Anticipated)Interventional2023-10-01Not yet recruiting
Feasibility of Outpatient Daily High Dose Cytarabine as Consolidation Therapy for Older Patients With Acute Myeloid Leukemia[NCT02101983]11 participants (Actual)Interventional2011-05-31Completed
A Pilot Study of a Sequential Regimen of Intensive Chemotherapy Followed by Autologous or Allogeneic Transplantation for Refractory Lymphoma (Non-Hodgkin's and Hodgkin's) and Phase 2 Expansion Cohort[NCT02059239]Phase 1/Phase 234 participants (Actual)Interventional2014-06-04Completed
A Phase I-II, Multicentre, Open Label Clinical Trial to Assess the Safety and Tolerability of the Combination of Low-dose Cytarabine or Azacitidine, Plus Venetoclax and Quizartinib in Newly Diagnosed Acute Myeloid Leukemia Patients Aged Equal or More Than[NCT04687761]Phase 1/Phase 284 participants (Anticipated)Interventional2020-11-04Recruiting
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

Complete Remission (CR) Rate (Including CR With Incomplete Recovery)

"Participants who achieved morphological complete remission with or without incomplete blood count recovery.~Per the Revised Recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia, morphologic complete remission requires that the patient achieve the morphologic leukemia-free state and have an absolute neutrophil count of more than 1,000/μL and platelets of ≥ 100,000/μL." (NCT00840177)
Timeframe: Up to 5 years after registration

InterventionParticipants (Count of Participants)
Initial Cohort: Relapsed AML With Previous Remission ≥ 3 Month27
Poor-risk Cohort: MDS Transformed to AML14
Poor-risk Cohort: Refractory/Relapsed AML, < 6 Mths Remission14

Overall Survival (OS)

OS is calculated for all participants from the date of initial registration on study until death from any cause. Observations for participants last known to be alive were censored. (NCT00840177)
Timeframe: OS assessed for up to 5 years, median OS reported

Interventionmonths (Median)
Initial Cohort: Relapsed AML With Previous Remission ≥ 3 Month10
Poor-risk Cohort: MDS Transformed to AML10
Poor-risk Cohort: Refractory/Relapsed AML, < 6 Mths Remission4

Relapse-free Survival (RFS)

"RFS is calculated for participants who have achieved a complete response (CR) or CR with incomplete blood count recovery (CRi). RFS will be measured from the date of CR or CRi until relapse from CR or CRi for death from any cause. Observation is censored at the date of last follow-up for patients last known to be alive without report of relapse.~Per the Revised Recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia, morphologic complete remission requires that the patient achieve the morphologic leukemia-free state and have an absolute neutrophil count of more than 1,000/μL and platelets of ≥ 100,000/μL." (NCT00840177)
Timeframe: RFS assessed for up to 5 years, median RFS reported

Interventionmonths (Median)
Initial Cohort: Relapsed AML With Previous Remission ≥ 3 Month9
Poor-risk Cohort: MDS Transformed to AML19.4
Poor-risk Cohort: Refractory/Relapsed AML, < 6 Mths Remission2.7

Frequency and Severity of Toxicity as Assessed by NCI CTCAE Version 3.0

Number of patients with Grade 3-5 adverse events that were possibly, probably or definitely related to study drug are reported by given type of adverse event (NCT00840177)
Timeframe: Up to 5 years post registration

,,
InterventionParticipants (Number)
ALT, SGPT (serum glutamic pyruvic transaminase)AST, SGOTAcidosis (metabolic or respiratory)Albumin, serum-low (hypoalbuminemia)Alkaline phosphataseAlkalosis (metabolic or respiratory)AnorexiaBilirubin (hyperbilirubinemia)Blood/Bone Marrow-Other (Specify)Bone marrow cellularityCalcium, serum-low (hypocalcemia)Carbon monoxide diffusion capacity (DL(co))Cardiac General-Other (Specify)Cardiac-ischemia/infarctionColitis, infectious (e.g., Clostridium difficile)Conduction abnormality - AsystoleConfusionConstitutional Symptoms-Other (Specify)CreatinineDiarrheaDistention/bloating, abdominalDry mouth/salivary gland (xerostomia)Dyspnea (shortness of breath)Edema: limbEsophagitisFatigue (asthenia, lethargy, malaise)Febrile neutropeniaGlucose, serum-high (hyperglycemia)HemoglobinHemorrhage, GI - DuodenumHemorrhage, GU - VaginaHemorrhage, pulmo/upper resp- Bronchopulmonary NOSHemorrhage, pulmonary/upper respiratory - NoseHemorrhage/Bleeding-Other (Specify)HypertensionHypotensionHypoxiaIleus, GI (functional obstruction of bowel)Inf (clin/microbio) w/Gr 3-4 neuts - BloodInf (clin/microbio) w/Gr 3-4 neuts - BronchusInf (clin/microbio) w/Gr 3-4 neuts - KidneyInf (clin/microbio) w/Gr 3-4 neuts - LungInf (clin/microbio) w/Gr 3-4 neuts - MeningesInf (clin/microbio) w/Gr 3-4 neuts - SinusInf (clin/microbio) w/Gr 3-4 neuts - SkinInf (clin/microbio) w/Gr 3-4 neuts - Small bowelInf (clin/microbio) w/Gr 3-4 neuts - StomachInfection with unknown ANC - BloodInfection with unknown ANC - Lung (pneumonia)Infection with unknown ANC - MucosaInfection with unknown ANC - SinusInfection with unknown ANC - Small bowel NOSInfection-Other (Specify)Left ventricular diastolic dysfunctionLeukocytes (total WBC)LipaseLymphopeniaMagnesium, serum-high (hypermagnesemia)Mental statusMucositis/stomatitis (clinical exam) - Oral cavityMucositis/stomatitis (clinical exam) - PharynxMucositis/stomatitis (functional/symp) - Oral cavMusculoskeletal/Soft Tissue-Other (Specify)NauseaNecrosis, GI - Small bowel NOSNeutrophils/granulocytes (ANC/AGC)Obstruction, GI - Small bowel NOSOcular/Visual-Other (Specify)Opportunistic inf associated w/gt=Gr 2 lymphopeniaPain - Abdomen NOSPain - BonePain - EsophagusPain - Extremity-limbPain - Oral cavityPain - PeritoneumPhosphate, serum-low (hypophosphatemia)PlateletsPleural effusion (non-malignant)Potassium, serum-high (hyperkalemia)Potassium, serum-low (hypokalemia)Pulmonary hypertensionPulmonary/Upper Respiratory-Other (Specify)Rash/desquamationRenal failureRenal/Genitourinary-Other (Specify)SVT and nodal arrhythmia - Atrial fibrillationSVT and nodal arrhythmia - Atrial flutterSodium, serum-high (hypernatremia)Sodium, serum-low (hyponatremia)Syncope (fainting)Typhlitis (cecal inflammation)Ulcer, GI - DuodenumUlcerationVomitingWeight loss
Initial Cohort: Relapsed AML With Previous Remission ≥ 3 Month0001001221211001100300201320111010011111410501010100101216160100102014010100000217006100001111100020
Poor-risk Cohort: MDS Transformed to AML2313011210540110023421120218214100000222400200100210011011051000011115100300001416123010011000001020
Poor-risk Cohort: Refractory/Relapsed AML, < 6 Mths Remission2106100000310010002800200232021001100130711610001401000017090011103016002011130219003001200001010102

Overall Survival

Overall Survival rate at 1 year. Time to death is from date of registration to date of death due to any cause. (NCT00041132)
Timeframe: assessed after cycle 4, after completion of treatment, then every 3 months for 2 years, then every 6 months thereafter until 5 years

Interventionpercentage of participants (Number)
Hyper-CVAD + MTX/Ara-C + Rituximab92

Progression-free Survival

Progression-Free Survival (PFS) rate at 1 year. PFS measured from date of registration to date of first observation of progressive disease or death due to any cause. Progressive disease is a 50% increase in the sum of products of greatest diameters (SPD) of target measurable lesions over the smallest sum observed if a complete response (confirmed, or unconfirmed) was not previously achieved; appearance of a new lesion/site; unequivocal progression of non-measurable disease; or death due to disease without prior documentation of progression. (NCT00041132)
Timeframe: assessed after cycle 4, after completion of treatment, then every 3 months until 1 year after registration

Interventionpercentage of participants (Number)
Hyper-CVAD + MTX/Ara-C + Rituximab90

Response

Complete (CR), complete unconfirmed (CRU) and partial responses (PR). CR is complete disappearance of all measurable and non-measurable disease with the exception of nodes; no new lesions; previously enlarged organs must have regressed in size; and if bone marrow positive at baseline, it must be negative. CRU is complete disappearance of all measurable and non-measurable disease; regressed, non-palpable organs; and one or more exceptions not qualifying for CR (see protocol section 10). PR applies to patients with at least one measurable lesion who do not qualify for CR or CRU. PR is a 50% decrease in sum of products of greatest diameters (SPD) for up to six identified dominant lesions identified at baseline; no new lesions; no increase in the size of liver, spleen or other nodes; and splenic and hepatic nodules must have regressed in size by at least 50% in SPD. (NCT00041132)
Timeframe: assessed after cycle 4 and after completion of treatment (168 days)

Interventionparticipants (Number)
Hyper-CVAD + MTX/Ara-C + Rituximab42

Response

Anti tumor activity as assessed by computed tomography of neck/chest/abdomen/pelvis, positron emission tomography scan and/or gallium scan. Assessed by physical examination appropriate imaging studies. Bone marrow aspirate/biopsy must be normal and any macroscopic nodules in any organs detectable on imaging techniques should no longer be present. Gallium scans must be negative if initially positive. (NCT00354107)
Timeframe: Week 4

Interventionpercent (Number)
Treatment (Monoclonal Antibody Therapy, Chemotherapy)50

Event Free Survival (EFS)

Survival is defined as the minimum time from study entry to a relapse of any kind, death from any cause, or occurrence of a second malignant neoplasm. Patients without report of such events where censored at last contact. This will be used to compute event free survival (EFS). (NCT00302003)
Timeframe: At 60 months

InterventionProbability of survival (Number)
Group 10.79

Event Free Survival Without Receiving Radiation Therapy (EFSnoRT).

Survival is defined as the minimum time from study entry to requirement for additional chemotherapy and IFRT for retrieval, occurrence of a second malignant neoplasm, or death from any cause. Patients without report of such events where censored at last contact. Patients who achieve less than CR after 3 cycles of AV-PC will require IFRT and hence will satisfy this definition at the time of response evaluation. Patients who achieve a CR but who relapse will receive addition chemotherapy and IFRT or intense retrieval and hence will satisfy this definition at the time of the first relapse of Hodgkin disease. This endpoint will be used to compute event free survival without receiving radiation therapy (EFSnoRT). (NCT00302003)
Timeframe: At 60 months

InterventionProbability of survival (Number)
Group 10.49

Intensive Therapy Free Survival (ITFS).

Survival is defined as the minimum time from study entry to a relapse of higher risk at any time, any relapse following treatment with protocol mandated IFRT, death from any cause, or the occurrence of a second malignant neoplasm. This will be used to compute intensive therapy free survival (ITFS). Patients without report of such events where censored at last contact. This differs from traditional EFS in that relapse after AVPC* x3 therapy alone that does not place the patient in a higher risk category is not considered a treatment failure. In this definition, higher-risk relapse refers to relapse involving sites and extent of disease that place the patient in the current COG definition of intermediate or high-risk disease. If a patient with CR who experiences a LR relapse is not retreated with protocol-mandated chemotherapy and IFRT, subsequent disease relapses will nevertheless be counted in the analysis of the treatment strategy. (NCT00302003)
Timeframe: At 60 months

InterventionProbability of survival (Number)
Group 10.89

Overall Survival

Survival is defined as time from study entry to death due to any cause. Patients alive at last contact where censored at last contact. (NCT00302003)
Timeframe: At 60 months

InterventionProbability of survival (Number)
Group 10.99

Complete Remission (CR)

complete remission (CR) Disappearance of all clinical evidence of leukemia for a minimum of four weeks. The patient should have a neutrophil count > 1,000 x 10^6/1, a platelet count > 100,000 x 10^9/1, no circulating blasts, and < than or = to blasts on bone marrow differential in a qualitatively normal or hypercellular marrow. Progressive disease or failure: Increasing bone marrow infiltrate or development of organ failure or extramedullary infiltrates due to leukemia. (NCT00002766)
Timeframe: 2 years

,
Interventionparticipants (Number)
Complete RemissionComplete Response (CR)FailureFailure-ProgressionRelapse
All-25014581
L-20501114100

Percentage of Participants With Adverse Events (AEs) and Laboratory Abnormalities Defined as Dose Limiting Toxicities (DLT)

"Occurrence of any of the following toxicities, attributable to ENTO, during Cycle 1 was considered DLT:~Grade 4 non-hematologic toxicity except alopecia, nausea, and vomiting controllable with anti-emetic therapy, line associated venous thrombosis, infection-related toxicities such as fever/sepsis, and fatigue.~In participants without bone marrow evidence of hematologic malignancy, hematologic toxicity defined as failure to recover absolute neutrophil count (ANC > 500/μL) or platelet count (>25000/μL) within 28 days~Grade 4, clinically significant, electrolyte abnormalities that were not correctable within 24 hours~Liver function test abnormalities that did not resolve to Grade 2 within 10 days~Infection that resulted from unexpectedly complicated prolonged myelosuppression~Toxicities that required temporary interruption of treatment and did not resolve to Grade 2 within 10 days or ENTO was suspended or dose reduced for a period of more than 10 days." (NCT03135028)
Timeframe: Cycle 1 (28-day cycle)

Interventionpercentage of participants (Number)
ENTO 400 mg0.0

Percentage of Participants With AEs and Laboratory Abnormalities Not Defined as DLT

(NCT03135028)
Timeframe: Day 1 Up to 30 days after permanent discontinuation of study treatment (maximum approximately 80 weeks)

Interventionpercentage of participants (Number)
ENTO 400 mg100.0

Plasma Concentration of ENTO

Plasma concentration of drug (ENTO) over different time points is reported. (NCT03135028)
Timeframe: Cycle 1 (28-days cycle) Day 8 at 0 (predose), 1, 2, 3, 4, 6, 8, and 12 hours postdose

Interventionng/mL (Mean)
Cycle 1 Day 8, predoseCycle 1 Day 8, 1 hour postdoseCycle 1 Day 8, 2 hours postdoseCycle 1 Day 8, 3 hours postdoseCycle 1 Day 8, 4 hours postdoseCycle 1 Day 8, 6 hours postdoseCycle 1 Day 8, 8 hours postdoseCycle 1 Day 8, 12 hours postdose
ENTO 400 mg921.91395.71892.71529.11350.01139.31075.9855.1

2 Year Relapse Free Survival (RFS)

Comparison of 2 year RFS in patient with detectable LSCs in the marrow at the end of consolidation to the 2 year RFS of patients without detectable LSCs. IWG Criteria (Cheson 2003) was utilized to classify relapse, with relapse defined as ≥ 5% blasts in the marrow or peripheral blood, extramedullary disease, or disease presence determined by a physician upon clinical assessment. (NCT02927938)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
eLSC- (Evaluable Cohort)NA

Number of Patients Who Achieve a CD4 Count > 200/Micro-liters

Determine the number of patients who achieve a CD4 count > 200/micro-liters by 60 days after transplant. (NCT00429416)
Timeframe: Through 60 Days Post Transplant

Interventionparticipants (Number)
LLME to Decrease GVHD Following HSC T13

Rate of Engraftment of Non-Myeloablative Transplants

Determine the engraftment rate of non-myeloablative transplants using CD34+ stem cells and LLME treated CD34- products. (NCT00429416)
Timeframe: Through 30 days post-transplant

Interventionparticipants (Number)
LLME to Decrease GVHD Following HSC T13

Rate of Serious Infectious Complications

"Determine the rate of serious infectious complications. A serious infection will be defined as any requiring hospitalization or parenteral therapy.~CD4 counts will be measured monthly for the first 3 months after transplant." (NCT00429416)
Timeframe: Through 3 months post-transplant

Interventionparticipants (Number)
LLME to Decrease GVHD Following HSC T2

Safety of CD34+ Stem Cell Infusions Followed by LLME as Measured by 100-Day Mortality

"Determine the safety of CD34+ stem cell infusions followed by the LLME treated CD34- fraction. This includes monitoring the patients for any side effects associated with the LLME treated cell infusion or any other unexpected adverse events.~This regimen will be gauged as to its safety using 100 day mortality as the measured endpoint. Deaths from all causes will be included." (NCT00429416)
Timeframe: Through 100 days post-transplant or death

Interventionparticipants (Number)
LLME to Decrease GVHD Following HSC T1

Incidence of Grade II-IV Acute Graft-Versus-Host-Disease (GVHD)

Determine the incidence of grade II-IV acute GVHD after administration of grafts when combined with Cyclosporine/Mycophenolate Mofetil for GVHD prophylaxis. GVHD assessments occur daily as an in patient and at each out patient visit. (NCT00429416)
Timeframe: Through 24 months post-treatment

Interventionparticipants (Number)
Developed grade II-IV GVHDDeveloped cGVHD (Chronic GVHD)
LLME to Decrease GVHD Following HSC T31

Duration of Any Hematologic Improvement

The duration of improvement was defined as the time from the date of hematologic improvement until the date of first documented progression or relapse after hematologic improvement or death from any cause. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

Interventionmonths (Median)
Azacitidine13.57
Conventional Care5.18

Kaplan-Meier Estimate for Median Time to Transformation to Acute Myeloid Leukemia (AML) or Death From Any Cause, Whichever Occurred First

The time to transformation to AML or death from any cause (whichever occurred first) was defined as the number of days from the date of randomization until the date of documented AML transformation or death from any cause. Patients who did not transform to AML or die were censored at the date of last follow-up. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

Interventionmonths (Median)
Azacitidine13.02
Conventional Care7.61

Kaplan-Meier Estimates for Median Time to Death From Any Cause

Kaplan-Meier estimates for the median months until death from any cause within the intent-to-treat population. Patients surviving at the end of the follow-up period were censored at the date of last contact. If a patient withdrew consent to follow-up or was lost to follow-up, the patient was censored as of the last date of contact. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

Interventionmonths (Number)
Azacitidine24.46
Conventional Care15.02

Kaplan-Meier Estimates for Median Time to Transformation to Acute Myeloid Leukemia (AML) Based on the Last Bone Marrow Assessment

A sensitivity analysis of time to transformation to AML during the entire study was performed based on the last bone marrow assessment. Patients were censored based on the last bone marrow assessment. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

Interventionmonths (Median)
Azacitidine17.80
Conventional Care11.48

Kaplan-Meier Estimates for Median Time to Transformation to Acute Myeloid Leukemia (AML)

The time to transformation to AML was defined as the number of days from the date of randomization until the date of documented AML transformation, defined as a bone marrow blast count ≥ 30% independent of baseline bone marrow count. Patients who did not transform to AML were censored at the date of last follow-up or date of death. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

Interventionmonths (Median)
Azacitidine20.66
Conventional Care15.44

Number of Infections Per Treatment Year Requiring Intravenous Antibiotics, Antifungals or Antivirals

The on-treatment adverse event rate of infection requiring IV antibiotics, antifungals, or antivirals per patient-years. The on-treatment period was considered the period from the date of randomization to the last treatment study visit. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

Interventioninfections per treatment year (Number)
Azacitidine0.16
Conventional Care0.24

Number of Participants Who Died

Count of participants who died during the study (NCT00071799)
Timeframe: 42 months

Interventionparticipants (Number)
Azacitidine82
Conventional Care113

Time to Disease Progression, Relapse After Complete or Partial Remission, or Death From Any Cause

The time to disease progression, relapse after complete or partial remission (CR, PR), or death from any cause was defined as the time from the date of randomization until the first date of documented disease progression, relapse after CR or PR, or death from any cause. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

Interventionmonths (Median)
Azacitidine14.13
Conventional Care8.82

Number of Participants Considered Hematologic Responders by Investigator Determinations Using International Working Group (IWG 2000) Criteria for Myelodysplastic Syndrome (MDS)

"Investigator determined responses followed IWG criteria for~complete remission(CR): repeat bone marrow show <5% myeloblasts, and peripheral blood evaluations lasting >=2 months of hemoglobin(>110 g/L), neutrophils(>=1.5x10^9/L), platelets(>=100x10^9/L), blasts (0%) and no dysplasia~partial remission(PR) is the same as CR for peripheral blood: bone marrow shows blasts decrease by >=50% or a less advanced FAB classification from pretreatment~stable disease(SD) is a failure to achieve at least a partial remission, but with no evidence of progression for at least 2 months." (NCT00071799)
Timeframe: Day 1 to 42 months

,
Interventionparticipants (Number)
Overall (Complete + Partial Remission)Complete RemissionPartial RemissionStable Disease
Azacitidine51302175
Conventional Care2114765

Number of Participants in Different Categories of Adverse Experiences During Core Study Period

Patient counts for a variety of subsets of adverse experiences for the core study period (day 1 to 42 months). The individual options for Conventional Care Regimens (Best Supportive Care Only, Low-Dose Cytarabine, and Standard Chemotherapy) are presented as separate treatments. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

,,,
Interventionparticipants (Number)
Patients with >=1 treatment emergent AE (TEAE)Patients with >=1 treatment related TEAEPatients with >=1 serious TEAEPatients with >=1 serious treatment related TEAEPatients w TEAE leading to discontinued treatmentPatients w TEAE leading to dose reductionPatients w TEAE leading to dose interruption
Azacitidine17516911443222082
Best Supportive Care Only971710400
Low-dose Cytarabine443427136212
Standard Chemotherapy19191413200

Number of Participants Showing Hematologic Improvement Using International Working Group (IWG 2000) Criteria for Myelodysplastic Syndrome (MDS) Assessed by Independent Review Committee

"IWG 2000 Criteria: Pretreatment=hemoglobin <100g/L or RBC transfusion-dependent, platelet count <100x10^9/L or platelet transfusion dependent, absolute neutrophil count <1.5x10^9/L.~Erythroid response: Major->20g/L increase or transfusion independent. Minor- 10-20g/L increase or >=50% decrease in transfusion requirements.~Platelet response: Major-absolute increase of >=30x10^9/L or platelet transfusion independence. Minor->=50% increase.~Neutrophil response: Major->=100% increase or an absolute increase of >0.5x10^9/L. Minor->=100% increase and absolute increase of <0.5x10^9/L." (NCT00071799)
Timeframe: Day 1 to 42 months

,
Interventionparticipants (Number)
Any Improvement n=177, 178Erythroid Response - Major n=157, 160Erythroid Response - Minor n=157, 160Platelet Response - Major n=141, 129Platelet Response - Minor n=138, 127Neutrophil Response - Major n=131, 111Neutrophil Response - Minor n=131, 111
Azacitidine87622466255
Conventional Care51171184209

Summary of Participants' Platelet Transfusion Status for Participants Who Were Transfusion Dependent at Baseline

Summary of dependence and independence from platelet transfusion at baseline and during treatment for patients who were dependent at baseline. A patient was considered transfusion independent at baseline if the patient had no transfusions during the 56 days prior to randomization. During study, a patient was considered transfusion independent during the on-treatment period if the patient had no transfusions during any 56 consecutive days or more. Otherwise, the patient was considered transfusion dependent. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

,
Interventionparticipants (Number)
Baseline Dependent; On-Treatment IndependentBaseline Dependent; On-Treatment Dependent
Azacitidine1622
Conventional Care1116

Summary of Participants' Platelet Transfusion Status for Participants Who Were Transfusion Independent at Baseline

Summary of dependence and independence from platelet transfusion at baseline and during treatment for patients who were independent at baseline. A patient was considered transfusion independent at baseline if the patient had no transfusions during the 56 days prior to randomization. During study, a patient was considered transfusion independent during the on-treatment period if the patient had no transfusions during any 56 consecutive days or more. Otherwise, the patient was considered transfusion dependent. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

,
Interventionparticipants (Number)
Baseline Independent; On-Treatment IndependentBaseline Independent; On-Treatment Dependent
Azacitidine12615
Conventional Care10250

Summary of Participants' Red Blood Cell (RBC) Transfusion Status for Participants Who Were Transfusion Dependent at Baseline

Summary of dependence and independence from red blood cell (RBC) transfusion at baseline and during treatment, for patients who were dependent at baseline. A patient was considered transfusion independent at baseline if the patient had no transfusions during the 56 days prior to randomization. During study, a patient was considered transfusion independent during the on-treatment period if the patient had no transfusions during any 56 consecutive days or more. Otherwise, the patient was considered transfusion dependent. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

,
Interventionparticipants (Number)
Baseline Dependent; On-Treatment IndependentBaseline Dependent; On-Treatment Dependent
Azacitidine5061
Conventional Care13101

Summary of Participants' Red Blood Cell (RBC) Transfusion Status for Participants Who Were Transfusion Independent at Baseline

Summary of dependence and independence from red blood cell (RBC) transfusion at baseline and during treatment, for patients who were independent at baseline. A patient was considered transfusion independent at baseline if the patient had no transfusions during the 56 days prior to randomization. During study, a patient was considered transfusion independent during the on-treatment period if the patient had no transfusions during any 56 consecutive days or more. Otherwise, the patient was considered transfusion dependent. (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

,
Interventionparticipants (Number)
Baseline Independent; On-Treatment IndependentBaseline Independent; On-Treatment Dependent
Azacitidine5810
Conventional Care3728

Summary of Subgroup Analyses for Kaplan-Meier Estimates for Time to Death From Any Cause

"Kaplan-Meier estimates for the median months until death from any cause within the intent-to-treat population. Patients surviving at the end of the follow-up period were censored at the date of last contact. If a patient withdrew consent to follow-up or was lost to follow-up, the patient was censored as of the last date of contact.~Subgroups that were analyzed are age, gender, French-American-British (FAB) classification, World Health Organization (WHO) classification and International Prognostic Scoring System (IPSS) classification." (NCT00071799)
Timeframe: Day 1 (randomization) to 42 months

,
Interventionmonths (Number)
Age <65 yearsAge >= 65 yearsAge >= 75 yearsGender: MaleGender: FemaleFAB: Refractory anemia with excess blasts (RAEB)FAB: RAEB in transformationWHO: RAEB 1WHO: RAEB 2WHO: Other (AML, CMMoL-1 and 2, indeterminate)IPSS: Intermediate 2IPSS: High
Azacitidine11.3124.468.9224.4625.1134.6617.2511.5421.1120.4634.6619.21
Conventional Care7.8713.876.2015.0214.8515.2115.256.7215.0215.2516.8914.52

Incidence of Recurrent Disease

Number of patients that have disease recurrence. (NCT00054327)
Timeframe: at day 100 post transplant

Interventionparticipants (Number)
Regimen A4
Regimen B-12
Regimen B-20
Regimen B-31
Regimen C2
Regimen D0

Number of Patients With Overall Survival at 2 Years.

(NCT00054327)
Timeframe: at 2 years from transplant

Interventionparticipants (Number)
Regimen A5
Regimen B-12
Regimen B-22
Regimen B-31
Regimen C5
Regimen D1

Rates of Durable Engraftment

Number of days that patients take to reach engraftment defined as time to hematologic engraftment will be defined as ANC >500/µl and platelets >20K/µl without transfusion support. (NCT00054327)
Timeframe: at day 42

Interventiondays (Mean)
Regimen A17.9
Regimen B-115.75
Regimen B-213
Regimen B-318.25
Regimen C13.9
Regimen D10.5

Toxicity as Measured by CTC v2.0

Number of patients that experience grade 3 or above toxicity. See serious adverse event list for toxicities. (NCT00054327)
Timeframe: at 100 days post transplant

Interventionparticipants (Number)
Regimen A0
Regimen B-10
Regimen B-21
Regimen B-32
Regimen C2
Regimen D0

Graft-versus-host Disease (GVHD)

Number of patients that develop acute graft-versus-host disease by grades 0-4. Grade O is no development of GVHD. Grade 1-4 is increase severity of skin, liver and gut involvement with 1 being least severe and 4 being most severe. (NCT00054327)
Timeframe: at 100 days post transplant

,,,,,
Interventionparticipants (Number)
Grade 0Grade 1Grade 2Grade 3Grade 4
Regimen A02620
Regimen B-111200
Regimen B-210101
Regimen B-310030
Regimen C11530
Regimen D00002

Duration of Response

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

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

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

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

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

Number of Participants Negative for Minimal Residual Disease (MRD)

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

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

Overall Survival

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

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

Participants With a Response

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

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

Liposome-encapsulated Cytarabine Area Under the Curve

Geometric mean liposome-encapsulated cytarabine area under the plasma concentration-time curve from time zero to the time of the last quantifiable concentration will be determined for patients in the dose-finding phase. (NCT02642965)
Timeframe: Prior to infusion on day 5 and, 45 and 90 minutes post day 5 infusion of cycle 1

Intervention(NANOGRAM x HOUR) / MILLILITER (Geometric Mean)
Treatment (CPX-351 and FLAG)4418582.5

Liposome-encapsulated Cytarabine Clearance

Geometric mean liposome-encapsulated cytarabine clearance following IV infusion will be determined for patients in the dose-finding phase. (NCT02642965)
Timeframe: Prior to infusion on day 5 and, 45 and 90 minutes post day 5 infusion of cycle 1

InterventionMILLILITER / HOUR (Geometric Mean)
Treatment (CPX-351 and FLAG)71.76

Liposome-encapsulated Cytarabine Time of Maximum Concentration

Median liposome-encapsulated cytarabine time of maximum observed plasma concentration will be determined for patients in the dose-finding phase. (NCT02642965)
Timeframe: Prior to infusion on day 5 and, 45 and 90 minutes post day 5 infusion of cycle 1

InterventionHOUR (Median)
Treatment (CPX-351 and FLAG)5

Liposome-encapsulated Cytarabine Volume of Distribution

Geometric mean liposome-encapsulated cytarabine volume of distribution following IV infusion will be determined for patients in the dose-finding phase. (NCT02642965)
Timeframe: Prior to infusion on day 5 and, 45 and 90 minutes post day 5 infusion of cycle 1

InterventionMILLILITER (Geometric Mean)
Treatment (CPX-351 and FLAG)4158.0

Liposome-encapsulated Daunorubicin Area Under the Curve

Geometric mean liposome-encapsulated daunorubicin area under the plasma concentration-time curve from time zero to the time of the last quantifiable concentration will be determined for patients in the dose-finding phase. (NCT02642965)
Timeframe: Prior to infusion on day 5 and, 45 and 90 minutes post day 5 infusion of cycle 1

Intervention(NANOGRAM x HOUR) / MILLILITER (Geometric Mean)
Treatment (CPX-351 and FLAG)1288010.3

Liposome-encapsulated Daunorubicin Clearance

Geometric mean liposome-encapsulated daunorubicin clearance following IV infusion will be determined for patients in the dose-finding phase. (NCT02642965)
Timeframe: Prior to infusion on day 5 and, 45 and 90 minutes post day 5 infusion of cycle 1

InterventionMILLILITER / HOUR (Geometric Mean)
Treatment (CPX-351 and FLAG)94.7

Liposome-encapsulated Daunorubicin Time of Maximum Concentration

Median liposome-encapsulated daunorubicin time of maximum observed plasma concentration will be determined for patients in the dose-finding phase. (NCT02642965)
Timeframe: Prior to infusion on day 5 and, 45 and 90 minutes post day 5 infusion of cycle 1

InterventionHOUR (Median)
Treatment (CPX-351 and FLAG)2

Liposome-encapsulated Daunorubicin Volume of Distribution

Geometric mean liposome-encapsulated daunorubicin volume of distribution following IV infusion will be determined for patients in the dose-finding phase. (NCT02642965)
Timeframe: Prior to infusion on day 5 and, 45 and 90 minutes post day 5 infusion of cycle 1

InterventionMILLILITER (Geometric Mean)
Treatment (CPX-351 and FLAG)3827.7

Number of Participants With a Dose-limiting Toxicity

Number of patients in the dose-finding phase with a dose-limiting toxicity, graded using the National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. (NCT02642965)
Timeframe: 28 days

InterventionParticipants (Count of Participants)
Treatment (CPX-351 and FLAG)1

Percentage of Responders (Complete Response or Complete Remission With Partial or Incomplete Platelet Recovery) After First Cycle of Therapy

Response (complete response or complete remission with partial or incomplete platelet recovery) after first cycle of therapy, where response is assessed using the revised Acute Myeloid Leukemia International Working Group Criteria. Percentage of responders is calculated by the total of number of patients with complete response or complete remission with partial or incomplete platelet recovery divided by the number of patients evaluable for response after the first cycle. 95% confidence interval is determined using a binomial exact method. (NCT02642965)
Timeframe: Up to 4 weeks

InterventionPercentage of responders (Number)
Treatment (CPX-351 and FLAG)75.68

Percentage of Responders (Complete Response or Complete Remission With Partial Platelet Recovery) After up to 2 Cycles

Best response (complete response or complete remission with partial platelet recovery) after up to 2 cycles of therapy, where response is assessed using the revised Acute Myeloid Leukemia International Working Group Criteria. Percentage of responders is calculated using the methods of Jung and Kim. 90% confidence interval is determined using the methods of Koyama and Chen. (NCT02642965)
Timeframe: Up to 8 weeks

InterventionPercantage of best responders (Number)
Treatment (CPX-351 and FLAG)68.30

Event-free Survival of Patients on Maintenance Randomization (Period 2)

Number of patients who develop EFS event during maintenance randomization (period 2) (NCT00078949)
Timeframe: during the period 2 (up to10 years)

Interventionparticipants (Number)
Maintenance53
Observation65

Response Rate of Patients After 2 Courses of Chemotherapy

The overall response rate by arm is calculated as total number of responders (CR + CRu + PR) / (all patients in the ITT analysis population). (NCT00078949)
Timeframe: After 2 cycle of treatment

Interventionpercentage of response (Number)
Salvage GDP45.2
Salvage DHAP44.0

Toxic Effect

Number of patients affected by adverse events graded according to CTC Version 2.0. See adverse event (others) for details. (NCT00078949)
Timeframe: 48 months

InterventionParticipants (Count of Participants)
Salvage GDP36
Salvage DHAP26
Maintenance16
Observation8

Transplantation Rate of Patients After 2 Courses of Chemotherapy

Transplantation rate is defined as the number of patients who respond sufficiently to protocol salvage chemotherapy to be planned for transplantation minus those who do not meet the endpoint of successful transplantation, divided by the number of all randomized patients (NCT00078949)
Timeframe: During period 1 (salvage chemotherapy)

Interventionpercentage of transplantation (Number)
Salvage GDP51.0
Salvage DHAP48.9

Cumulative Incidence of Neutrophil Recovery

Neutrophil recovery is defined as two consecutive days of absolute neutrophil count (ANC) > 500 neutrophils/μL following the expected nadir. (NCT01141712)
Timeframe: Day 28

Interventionpercentage of participants (Number)
Autologous Transplant97.5

Cumulative Incidence of Platelet Recovery

Platelet recovery is defined as a platelet count greater than 20,000/μL for the first of two consecutive labs with no platelet transfusions 7 days prior. (NCT01141712)
Timeframe: Day 100

Interventionpercentage of participants (Number)
Autologous Transplant92.5

Number of Participants Experiencing Infections

Microbiologically documented infections will be reported by site of disease, date of onset, severity, and resolution, if any. (NCT01141712)
Timeframe: Year 1

Interventionparticipants (Number)
Autologous Transplant22

Number of Participants Experiencing Toxicity

Toxicities will be defined by using the version 3.0 NCI Common Terminology Criteria for Adverse Events (CTCAE) criteria. Only grade 3 and higher toxicities will be collected. (NCT01141712)
Timeframe: Year 2

Interventionparticipants (Number)
Autologous Transplant9

Progression-Free Survival (PFS)

The time to this event is the time from enrollment until death, relapse/progression, receipt of anti-lymphoma therapy, or last follow up, whichever comes first. Progression-free survival (PFS) will be estimated using the Kaplan Meier product limit estimator. The PFS probability and confidence interval will be calculated at two years post-transplant. (NCT01141712)
Timeframe: Year 2

Interventionpercentage of participants (Number)
Autologous Transplant79.8

Relapse/Progression

Relapse is defined as the appearance of any new lesion more than 1.5 cm in any axis during or at the end of therapy, even if other lesions are decreasing in size. Progression is defined as a lymph node with a diameter of the short axis of less than 1.0 cm must increase by >= 50% and to a size of 1.5 x 1.5 cm or more than 1.5 cm in the long axis. (NCT01141712)
Timeframe: Year 2

Interventionpercentage of participants (Number)
Autologous Transplant12.5

Treatment-Related Mortality (TRM)

TRM is defined as death occurring in a patient from causes other than relapse or progression. A cumulative incidence curve will be computed along with a 95% confidence interval at 100 days post-transplant. (NCT01141712)
Timeframe: Day 100

Interventionpercentage of participants (Number)
Autologous Transplant5.2

Complete Remission (CR) and/or Partial Response (PR)

The frequencies and proportions of patients who have a CR or PR. CR is defined as the disappearance of all evidence of disease, and PR is defined as the regression of measurable disease and no new sites. (NCT01141712)
Timeframe: Day 100

Interventionparticipants (Number)
CRPRRelapse/Progression
Autologous Transplant3612

Hematologic Function

Hematologic function will be defined as ANC > 1500 neutrophils/μL, Hemoglobin> 10g/dL without transfusion support, and platelets > 100,000/μL (NCT01141712)
Timeframe: Days 100 and 365

Interventionparticipants (Number)
Day 100Day 365
Autologous Transplant1123

HIV Single-Copy Polymerase Chain Reaction (PCR)

HIV RNA assay will be performed at the specified time points and summarize the assessments of the viral copy number using descriptive statistics. (NCT01141712)
Timeframe: Baseline, Days 100, 180, 365, and 730

Interventioncopies/mL (Median)
BaselineDay 100Day 180Day 365Day 730
Autologous Transplant802988497130

Immunologic Reconstitution

Immunologic Reconstitution will be assessed by quantitative immunoglobulin measurement (IgM, IgG and IgA) (NCT01141712)
Timeframe: Year 1

Interventionmg/dL (Median)
IgGIgAIgM
Autologous Transplant109012348.5

Overall Survival (OS)

Assess the OS after autologous hematopoietic stem cell transplantation (HCT) for chemotherapy-sensitive aggressive B cell lymphoma or Hodgkin's lymphoma in patients with HIV using BEAM for pre-transplant conditioning. The primary analysis will consist of estimating the 1 year OS probability from the time of transplantation based on the Kaplan-Meier product limit estimator. The 1 year and 2 year OS and confidence interval will be calculated. (NCT01141712)
Timeframe: Year 1 and 2

Interventionpercentage of participants (Number)
1 year2 years
Autologous Transplant87.382

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

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

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

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

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

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

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

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

Overall Response Rate After Two Cycles of ROAD

The overall response rate is defined as the percentage of patients who achieve a response after two cycles of oxaliplatin with rituximab, cytarabine, and dexamethasone (ROAD). A response was considered a Complete Response (CR) or Partial Response (PR) as defined by the NCI Sponsored International Working Group (IWG). CR: Complete disappearance of all detectable clinical and radiographic evidence of disease and disappearance of all disease-related symptoms if present before therapy, and normalization of those biochemical abnormalities PR: ≥ 50% decrease in SPD of the six largest dominant nodes or nodal masses. (NCT00166439)
Timeframe: Up to 42 days

Interventionpercentage of patients (Number)
Treatment (ROAD)58

Overall Survival

Survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. (NCT00166439)
Timeframe: Up to 10 years

Interventionmonths (Median)
Treatment (ROAD)26

Progression-free Survival

The progression-free survival time is defined as the time from registration to progression or death due to any cause. The distribution of progression-free survival will be estimated using the method of Kaplan-Meier. Progression is defined by the NCI Sponsored IWG as a ≥ 50% increase from nadir in the sum of the products of the greatest diameters (SPD) of any previously identified abnormal node for PRs or nonresponders and/or Appearance of any new lesion during or at the end of therapy. (NCT00166439)
Timeframe: Up to 10 years

Interventionmonths (Median)
Treatment (ROAD)11

Circulating Leukemia Cells in Peripheral Blood Change From Prior to the Methotrexate Infusion to Three Days After Between Two Arms (4 Hours vs. 24 Hours)

"White blood cell (leukocytes) counts in peripheral blood by Complete Blood Count~Measurement: Percentage change of leukemia cells from baseline" (NCT00137111)
Timeframe: Immediately before the methotrexate infusion and three days after subsequent infusion

InterventionPercent change (Mean)
4 hr-44
24 hr-50

Continuous Complete Remission Since Week 56 Therapy.

CCR was measured from end of week 56 therapy to the date of first treatment failure of any kind (relapse, death, lineage switch, or second malignancy) or to the last date of follow-up. Measurement was determined by Kaplan-Meyer estimate. (NCT00137111)
Timeframe: Median follow up time (range) 4.5 (1 to 7.8) years

InterventionPercentage of participants (Number)
Patients With High Risk of CNS Relapse92.2

Mean Difference of Active Methotrexate Polyglutamates (MTXPG) in Leukemia Cells Between Two Arms (4 Hours vs. 24 Hours).

Children were randomly assigned to receive initial single-agent treatment with HDMTX (1g/m^2) as either a 24-hour infusion or a 4-hour infusion and the outcome measure was the accumulation of MTXPG in leukemia cells. (NCT00137111)
Timeframe: 42 hours after start of high dose methotrexate infusion (HDMTX)

Interventionpmol/1,000,000,000 cells (Mean)
4 hr1688
24 hr2521

Overall Event-free Survival (EFS)

EFS was measured from the start of on-study to the date of first treatment failure of any kind (relapse, death, lineage switch, or second malignancy) or to the last date of follow-up. Failure to enter remission was considered an event at time zero. Measurement was determined by Kaplan-Meyer estimate. (NCT00137111)
Timeframe: Median follow-up time (range) 5.6 (1.3 to 8.9) years

InterventionPercentage of Participants (Number)
Total Therapy87.3

Median Difference in CASP1 Gene Expression in Primary Leukemia Cells of Patients in Glucocorticoid-resistant Cells vs Glucocorticoid-sensitive Cells

Prednisolone sensitivity was measured in primary leukemia cells from bone marrow collected at diagnosis. Expression of CASP1 was determined by HG-U133A microarray. Values given are gene expression values, and the unit is arbitrary units (AU) defined as scaled fluorescence measured on microarray. (NCT00137111)
Timeframe: Pre-treatment

Interventionarbitrary units (Median)
Prednisolone-sensitive cellsPrednisolone-resistant cells
Total Therapy341.3447.9

Median Difference in NLRP3 Gene Expression in Primary Leukemia Cells of Patients in Glucocorticoid-resistant Cells vs. Glucocorticoid-sensitive Cells

Prednisolone sensitivity was measured in primary leukemia cells from bone marrow collected at diagnosis. Expression of NLRP3 was determined by HG-U133A microarray. Values given are gene expression values, and the unit is arbitrary units (AU) defined as scaled fluorescence measured on microarray. (NCT00137111)
Timeframe: Pre-treatment

Interventionarbitrary units (Median)
Prednisolone-sensitive cellsPrednisolone-resistant cells
Total Therapy41.2110.7

Minimal Residual Disease (MRD)

Detection of MRD at end of induction where positive MRD was defined as one or more leukemic cell per 10,000 mononuclear bone-marrow cells (>=0.01%). (NCT00137111)
Timeframe: End of Induction (Day 46 MRD measurement)

Interventionparticipants (Number)
Negative <0.01%Positive >= 0.01%
Total Therapy390102

Percentage of Participants Experiencing Grade 3-5 Toxicity

Percentage of participants experiencing at least one grade 3-5 adverse event (by CTCAE 3.0 criteria). (NCT00133991)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
R-CVP + HiCy21

Event-free Survival

Percentage of participants alive without relapse at 1 year and 3 years. (NCT00133991)
Timeframe: 1 year and 3 years

Interventionpercentage of participants (Number)
1 year3 years
R-CVP + HiCy5252

Overall Response Rate

Number of participants who have a complete or partial remission (2007 International Working Group criteria). (NCT00133991)
Timeframe: Up to 3 months

InterventionParticipants (Count of Participants)
Complete remissionPartial remission
R-CVP + HiCy112

Overall Survival

Percentage of participants alive at 1 year and at 3 years. (NCT00133991)
Timeframe: 1 year and 3 years

Interventionpercentage of participants (Number)
1 year3 years
R-CVP + HiCy5757

Relapse Pattern

Percentage of participants experiencing central nervous system (CNS) and systemic relapse. (NCT00133991)
Timeframe: Up to 6 months

InterventionParticipants (Count of Participants)
Systemic relapse onlySystemic and CNS relapse
R-CVP + HiCy32

Number of Participants Treatment-Emergent Adverse Events Reported During Consolidation Phase After a Daily Dose of 20 mg or 40 mg in Combination With Standard Chemotherapy to Japanese Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)

A treatment-emergent adverse event (TEAE) was defined as an adverse event (AE) that emerges (or worsens) from the first dose of study drug and coadministered drugs to the follow-up visit, 35 days after the last dose of study drug and coadministered drugs, or the start date of new AML post-treatment, whichever was earlier. (NCT02834390)
Timeframe: Consolidation phase Cycle 1, Day 1 up to 35 days after last dose (each cycle 28 days), up to approximately 1 year

,
InterventionParticipants (Count of Participants)
Participants with TEAEsInfections and InfestationsLung infectionSkin infectionUpper respiratory tract infectionBlood and Lymphatic System DisordersFebrile neutropeniaNervous System DisordersDysgeusiaGastrointestinal DisordersDiarrhoeaNauseaStomatitisSkin and Subcutaneous Tissue DisordersRashRash maculo-papularGeneral Disorders & Administration Site ConditionsOedema peripheralInvestigationsPlatelet count decreasedWhite blood cell count decreasedAlanine aminotransferase increasedElectrocardiogram QT prolongedInjury, Poisoning, and Procedural ComplicationsContusionFall
Quizartinib 20 mg/Day21011221122111101121110000
Quizartinib 40 mg/Day11100110000001010011101111

Number of Participants Treatment-Emergent Adverse Events Reported During Induction Phase After a Daily Dose of 20 mg or 40 mg in Combination With Standard Chemotherapy to Japanese Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)

A treatment-emergent adverse event (TEAE) was defined as an adverse event (AE) that emerges (or worsens) from the first dose of study drug and coadministered drugs to the follow-up visit, 35 days after the last dose of study drug and coadministered drugs, or the start date of new AML post-treatment, whichever was earlier. (NCT02834390)
Timeframe: Induction phase Cycle 1, Day 1 up to 35 days after last dose in Cycle 2 (each cycle 28 days), up to approximately 1 year

,
InterventionParticipants (Count of Participants)
Participants with TEAEsInfections and InfestationsPneumoniaSkin infectionStaphylococcal bacteraemiaBlood and Lymphatic System DisordersFebrile neutropeniaAnaemiaLeukopeniaNeutropeniaThrombocytopeniaImmune DisordersHypersensitivityMetabolism and Nutrition DisordersDecreased appetiteHypoalbuminaemiaHypokalaemiaHypomagnesaemiaHyponatraemiaHypophosphataemiaHypouricaemiaTumour lysis syndromeNervous System DisordersDysgeusiaDizzinessEye DisordersPhotophobiaCardiac DisordersPalpitationsVascular DisordersHypotensionRespiratory, Thoracic, and Mediastinal DisordersEpistaxisOropharyngeal painRespiratory disorderGastrointestinal DisordersAbdominal pain upperNauseaConstipationDiarrhoeaStomatitisCheilitisVomitingMelaenaHepatobiliary DisordersHepatic function abnormalSkin and Subcutaneous Tissue DisordersAlopeciaRashRash maculo-papularDermatitis bullousDry skinMusculoskeletal and Connective Tissue DisordersArthralgiaMusculoskeletal painMyalgiaPain in extremityRenal and Urinary DisordersHaematuriaProteinuriaUrinary tract painReproductive System and Breast DisordersMenorrhagiaGeneral Disorders & Administration Site ConditionsOedema peripheralPyrexiaInvestigationsGamma-glutamyltransferase increasedAlanine aminotransferase increasedBlood alkaline phosphatase increasedElectrocardiogram QT prolongedPlatelet count decreasedWeight decreasedWhite blood cell count decreasedAspartate aminotransferaseAspartate aminotransferase increasedBlood bilirubin increasedBlood lactate dehydrogenase increasedBlood pressure decreasedLipase increased
Quizartinib 20 mg/Day41100442111003312111012211100000000332112110004311001001011110000043210111101000
Quizartinib 40 mg/Day33111331000112110000101100011111111312221111113221113210100001111131112111010111

Pharmacokinetic Parameter Accumulation Ratio (AR) of Quizartinib and Active Metabolite (AC886) After a Daily Dose of 20 mg or 40 mg Combined With Chemotherapy to Japanese Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)

The accumulation ratio of geometric means for Cmax and AUCtau is reported at Induction phase Cycle 1, Day 21 for serum quizartinib, active metabolite AC886, and quizartinib and AC886. (NCT02834390)
Timeframe: Induction phase Cycle 1, Day 21 (each cycle 28 days)

,
Interventionaccumulation ratio (Geometric Mean)
Cycle 1, Day 21: Quizartinib AR (Cmax)Cycle 1, Day 21: AC886 AR (Cmax)Cycle 1, Day 21: Quizartinib + AC886 AR (Cmax)Cycle 1, Day 21: Quizartinib AR (AUCtau)Cycle 1, Day 21: AC886 AR (AUCtau)Cycle 1, Day 21: Quizartinib + AC886 AR (AUCtau)
Quizartinib 20 mg/Day1.522.652.092.373.502.99
Quizartinib 40 mg/Day1.982.552.532.843.313.24

Pharmacokinetic Parameter Area Under the Concentration-Time Curve During Dosing Interval of Quizartinib and Metabolite (AC886) After Daily Dose of 20 mg or 40 mg With Chemotherapy to Japanese Participants With Newly Diagnosed Acute Myeloid Leukemia

The area under the plasma concentration-time curve during dosing interval (AUCtau) of geometric means is reported at Induction phase Cycle 1, Day 9 and the area under the plasma concentration-time curve during dosing interval at steady state (AUCtauss) of geometric means are reported at Induction phase Cycle 1, Day 21 for quizartinib, active metabolite AC886, and quizartinib and AC886. (NCT02834390)
Timeframe: Induction phase Cycle 1, Day 8 to Cycle 1, Day 21 (each cycle 28 days)

,
Interventionng*h/mL (Geometric Mean)
Cycle 1, Day 8: Quizartinib, AUCtauCycle 1, Day 8: AC886, AUCtauCycle 1, Day 8: Quizartinib + AC886, AUCtauCycle 1, Day 21: Quizartinib, AUCtau,ssCycle 1, Day 21: AC886, AUCtau, ssCycle 1, Day 21: Quizartinib + AC886, AUCtau
Quizartinib 20 mg/Day418555101099119403020
Quizartinib 40 mg/Day92116402640294053108850

Pharmacokinetic Parameter Maximum Serum Concentration (Cmax) of Geometric Means of Quizartinib and Active Metabolite (AC886) After Daily Dose of 20 mg or 40 mg Combined With Chemotherapy to Japanese Participants With Newly Diagnosed Acute Myeloid Leukemia

The maximum serum plasma concentration (Cmax) is reported at Induction phase Cycle 1, Day 9 and the maximum serum plasma concentration at steady state (Cmax,ss) is reported at Induction phase Cycle 1, Day 21 for quizartinib, active metabolite AC886, and quizartinib and AC886. (NCT02834390)
Timeframe: Induction phase Cycle 1, Day 8 to Cycle 1, Day 21 (each cycle 28 days)

,
Interventionng/mL (Geometric Mean)
Cycle 1, Day 8: Quizartinib, CmaxCycle 1, Day 8: AC886, CmaxCycle 1, Day 8: Quizartinib + AC886, CmaxCycle 1, Day 21: Quizartinib, Cmax, ssCycle 1, Day 21: AC886, Cmax, ssCycle 1, Day 21: Quizartinib + AC886, Cmax
Quizartinib 20 mg/Day42.436.577.464.296.7162
Quizartinib 40 mg/Day91.3103180212256480

Pharmacokinetic Parameter Metabolite to Parent Ratio (MR) of Active Metabolite (AC886) After a Daily Dose of 20 mg or 40 mg Combined With Chemotherapy to Japanese Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)

The metabolite to parent ratio of geometric means for Cmax and AUCtau is reported at Induction phase Cycle 1, Day 9 for active metabolite AC886 is reported. (NCT02834390)
Timeframe: Induction phase Cycle 1, Day 8 (each cycle 28 days)

,
Interventionmetabolite to parent ratio (Geometric Mean)
Cycle 1, Day 8: MR (Cmax)Cycle 1, Day 8: MR (AUCtau)
Quizartinib 20 mg/Day0.8611.33
Quizartinib 40 mg/Day1.131.78

Pharmacokinetic Parameter Time of Maximum Plasma Concentration (Tmax) of Quizartinib and the Active Metabolite (AC886) After a Daily Dose of 20 mg or 40 mg Combined With Chemotherapy to Japanese Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)

The time of maximum plasma concentration (Tmax) of geometric means is reported at Induction phase Cycle 1, Day 9 and the time of maximum plasma concentration at steady state (Tmax,ss) of geometric means is reported at Induction phase Cycle 1, Day 21 for quizartinib, active metabolite AC886, and quizartinib and AC886. (NCT02834390)
Timeframe: Induction phase Cycle 1, Day 8 to Cycle 1, Day 21 (each cycle 28 days)

,
Interventionhour (Median)
Cycle 1, Day 8: Quizartinib, TmaxCycle 1, Day 8: AC886, TmaxCycle 1, Day 8: Quizartinib + AC886, TmaxCycle 1, Day 21: Quizartinib, Tmax,ssCycle 1, Day 21: AC886, Tmax,ssCycle 1, Day 21: Quizartinib + AC886, Tmax
Quizartinib 20 mg/Day3.035.014.014.035.024.03
Quizartinib 40 mg/Day2.176.084.174.086.094.08

Pharmacokinetic Parameter Trough Plasma Concentration (Ctrough) of Quizartinib and Active Metabolite (AC886) After a Daily Dose of 20 mg or 40 mg Combined With Chemotherapy to Japanese Patients With Newly Diagnosed Acute Myeloid Leukemia (AML)

The trough plasma concentration (Ctrough) of geometric means is reported at Induction phase Cycle 1, Day 21 for serum quizartinib, active metabolite AC886, and quizartinib and AC886. (NCT02834390)
Timeframe: Induction phase Cycle 1, Day 21 (each cycle 28 days)

,
Interventionng/mL (Geometric Mean)
Cycle 1, Day 21: QuizartinibCycle 1, Day 21: AC886Cycle 1, Day 21: Quizartinib + AC886
Quizartinib 20 mg/Day22.768.694.6
Quizartinib 40 mg/Day77.1195299

Summary of Best Response Rates After the First Oral Dose of Quizartinib in Participants With Relapsed or Refractory Acute Myeloid Leukemia

Best response was defined as the best measured complete remission (CR), CR with incomplete hematological recovery (CRi), CR with incomplete platelet recovery (CRp), Partial remission (PR), or No response (NR) in the overall response assessments at all time points after the first dose. (NCT02834390)
Timeframe: Day 0 to Day 21 of last completed cycle (each cycle 28 days), up to approximately 1 year

,
Interventionpercentage of participants (Number)
Composite CR rate (CRc rate: CR+CRp+CRi)Response rate (CRc+PR)
Quizartinib 20 mg/Day75.0100
Quizartinib 40 mg/Day66.766.7

Summary of Best Responses After a Daily Dose of 20 mg or 40 mg in Combination With Standard Chemotherapy to Japanese Participants With Newly Diagnosed Acute Myeloid Leukemia (AML)

Best response was defined as the best measured complete remission (CR), CR with incomplete hematological recovery (CRi), CR with incomplete platelet recovery (CRp), Partial remission (PR), or No response (NR) in the overall response assessments at all time points after the first dose. (NCT02834390)
Timeframe: Day 0 to Day 21 of last completed cycle (each cycle 28 days), up to approximately 1 year

,
InterventionParticipants (Count of Participants)
Complete remission (CR)CR with incomplete platelet recovery (CRp)CR with incomplete hematological recovery (CRi)Partial remission (PR)No response (NR)
Quizartinib 20 mg/Day00310
Quizartinib 40 mg/Day00201

Duration of Remission Following CR (DOR) in TP53 WT Population

"DOR is defined for patients achieving complete remission and is the time from clinical remission until relapse or death from any cause, whichever occurs first. For patients with none of these events before time of analysis, DOR is censored at the date of the patient's last response assessment.~The Median upper limits were set to 999 because they were not reached and therefore it was Not Evaluable (NE) and the field is numeric only. The study was terminated because of futility, therefore did not reach the planned end of the study." (NCT02545283)
Timeframe: From achieving CR until relapse or death from any cause (up to approximately 4.5 years)

InterventionMonths (Median)
Placebo Plus Cytarabine18.73
Idasanutlin Plus Cytarabine16.76

Event-Free Survival (EFS) According to HMRA in TP53 WT Population

"Event Free Survival (EFS) is defined as the time from the date of randomization to whichever occurs first: treatment failure (failure to achieve CR, set as day of final response assessment), relapse from CR, or death from any cause. The study was terminated because of futility, therefore did not reach the planned end of the study.~The design followed a hierarchical statistical testing framework." (NCT02545283)
Timeframe: From randomization up to treatment failure, relapse, or death from any cause (up to approximately 4.5 years)

InterventionWeeks (Median)
Placebo Plus Cytarabine6.29
Idasanutlin Plus Cytarabine4.36

Number of Participants Who Experienced at Least One Adverse Event by Severity, According to National Cancer Institute Common Terminology Criteria for Adverse Events, Version 4.03 (NCI-CTCAE v4.03)

Participants who experienced at least one Adverse Event by severity, According to National Cancer Institute common terminology criteria for Adverse Events, version 4.03 (NCI-CTCAE v4.03) have been reported. The study was terminated because of futility, therefore did not reach the planned end of the study. (NCT02545283)
Timeframe: Baseline up to approximately 4.5 years

InterventionParticipants (Number)
Placebo Plus Cytarabine149
Idasanutlin Plus Cytarabine232

Number of Participants With Adverse Events Leading to Death up to Day 30

The number of participants with AE resulted by death within 30 days from dosing is reported (NCT02545283)
Timeframe: Up to Day 30

InterventionParticipants (Number)
Placebo Plus Cytarabine9
Idasanutlin Plus Cytarabine23

Number of Participants With Adverse Events Leading to Death up to Day 60

The number of participants with AE resulted by death within 60 days from dosing is reported (NCT02545283)
Timeframe: Up to Day 60

InterventionParticipants (Number)
Placebo Plus Cytarabine24
Idasanutlin Plus Cytarabine60

Number of Participants With Adverse Events Leading to Discontinuation

Participants with Adverse Events leading to discontinuation of the study have been reported. The study was terminated because of futility, therefore did not reach the planned end of the study. (NCT02545283)
Timeframe: Baseline up to approximately 4.5 years

InterventionParticipants (Number)
Placebo Plus Cytarabine0
Idasanutlin Plus Cytarabine0

Number of Treatment Cycles Started

Participants who started the study treatment cycles are reported. (NCT02545283)
Timeframe: Up to 3 cycles (1 cycle is 28 days)

InterventionTreatment Cycles (Mean)
Placebo Plus Cytarabine1.3
Idasanutlin Plus Cytarabine1.2

Overall Survival in TP53 WT Population

"P53 tumor protein Wild Type (TP53 WT) population's Overall Survival was compared in participants with relapsed or refractory (R/R) acute myeloid leukemia (AML) who had been randomized to idasanutlin in combination with cytarabine versus those who had been randomized to cytarabine and placebo.~The study was terminated because of futility, therefore did not reach the planned end of the study." (NCT02545283)
Timeframe: From randomization to death from any cause (up to approximately 4.5 years)

InterventionMonths (Median)
Placebo Plus Cytarabine9.13
Idasanutlin Plus Cytarabine8.28

Percentage of Participants in Complete Response (CR) at the End of Induction According to Hematologic Malignancy Response Assessment (HMRA) in TP53 WT Population

"Rate of complete response at the end of induction in the TP53 WT population was compared in participants who had been randomized to idasanutlin in combination with cytarabine versus those who had been randomized to cytarabine and placebo.~The design followed a hierarchical statistical testing framework." (NCT02545283)
Timeframe: At the end of induction (up to Day 56)

InterventionPercentage of Participants (Number)
Placebo Plus Cytarabine20.3
Idasanutlin Plus Cytarabine17.1

Percentage of Participants Undergoing HSCT Following Complete Response (CR), in TP53 WT Population

Rate of complete response at the end of induction in mutation-defined subgroups (positive for mutation of FLT3, IDH1 and IDH2, respectively) of the TP53 WT population was compared in participants who had been randomized to idasanutlin in combination with cytarabine versus those who had been randomized to cytarabine and placebo. The study was terminated because of futility, therefore did not reach the planned end of the study. (NCT02545283)
Timeframe: Baseline up to approximately 4.5 years

InterventionPercentage of Participants (Number)
Placebo Plus Cytarabine10.6
Idasanutlin Plus Cytarabine11.6

Percentage of Participants With Overall Remission (CR, CRp, and CRi) at the End of Induction According to HMRA in TP53 WT Population

"Rate of overall remission (complete remission, complete remission with incomplete hematologic recovery, complete remission with incomplete platelet count recovery) at the end of induction in the TP53 WT population was compared in participants who had been randomized to idasanutlin in combination with cytarabine versus those who had been randomized to cytarabine and placebo.~The design followed a hierarchical statistical testing framework." (NCT02545283)
Timeframe: At the end of induction (up to Day 56)

InterventionPercentage of Participants (Number)
Placebo Plus Cytarabine38.8
Idasanutlin Plus Cytarabine22.0

Total Duration of Study Treatment

Participants were planned to be treated up to 3 Cycles. (NCT02545283)
Timeframe: Up to 3 cycles (1 cycle is 28 days)

InterventionDays (Mean)
Idasanutlin Plus Cytarabine16.5
Placebo Plus Cytarabine17.6

Change From Baseline in Body Temperature Over Time

"Vital signs were measured prior to the infusion while the participant was in a seated position. The baseline value at visit and the change from baseline value at each timepoint are reported. The change from baseline value was calculated by subtracting the post-baseline value from the baseline value.~The study was terminated because of futility, therefore did not reach the planned end of study." (NCT02545283)
Timeframe: Baseline; Cycles 1-3, Days 8, 15, 22, 28 (1 cycle is 28 days); and, if incomplete blood count recovery, Days 29-42 and Days 43-56, Cycles 2 -3 Days 1, 8, 15, 22, 28 and 29-56 (max delay between cycles is 56 days)

,
InterventionC, Celsius Degree (Mean)
BaselineCycle 1 Day 8Cycle 1 Day 15Cycle 1 Day 22Cycle 1 Day 28Cycle 1 Day 29-42Cycle 1 Day 43-56Cycle 2 Day 1Cycle 2 Day 8Cycle 2 Day 15Cycle 2 Day 22Cycle 2 Day 28Cycle 2 Day 29-56Cycle 3 Day 1Cycle 3 Day 8Cycle 3 Day 15Cycle 3 Day 22Cycle 3 Day 28Cycle 3 Day 29-56
Idasanutlin Plus Cytarabine36.520.320.460.360.230.000.07-0.100.070.340.100.04-0.09-0.19-0.040.290.04-0.15-0.25
Placebo Plus Cytarabine36.490.070.300.220.060.08-0.020.11-0.080.170.04-0.22-0.06-0.250.10-0.09-0.07-0.260.27

Change From Baseline in Diastolic Blood Pressure Over Time

"Vital signs were measured prior to the infusion while the participant was in a seated position. The baseline value at visit and the change from baseline value at each timepoint are reported. The change from baseline value was calculated by subtracting the post-baseline value from the baseline value.~The study was terminated because of futility, therefore did not reach the planned end of study." (NCT02545283)
Timeframe: Baseline; Cycles 1-3, Days 8, 15, 22, 28 (1 cycle is 28 days); and, if incomplete blood count recovery, Days 29-42 and Days 43-56, Cycles 2 -3 Days 1, 8, 15, 22, 28 and 29-56 (max delay between cycles is 56 days)

,
InterventionMillimeters of mercury (mmHg) (Mean)
BaselineCycle 1 Day 8Cycle 1 Day 15Cycle 1 Day 22Cycle 1 Day 28Cycle 1 Day 29-42Cycle 1 Day 43-56Cycle 2 Day 1Cycle 2 Day 8Cycle 2 Day 15Cycle 2 Day 22Cycle 2 Day 28Cycle 2 Day 29-56Cycle 3 Day 1Cycle 3 Day 8Cycle 3 Day 15Cycle 3 Day 22Cycle 3 Day 28Cycle 3 Day 29-56
Idasanutlin Plus Cytarabine71.5-3.1-1.3-1.4-0.70.60.90.9-2.2-1.51.73.93.20.3-3.3-1.8-2.11.32.6
Placebo Plus Cytarabine70.2-1.5-1.3-0.51.03.81.71.10.4-0.63.14.6-2.95.02.00.710.12.35.0

Change From Baseline in Electrocardiogram Parameters: PQ, PR, RR, QRS, QT and QTcF Intervals

"Single 12-lead ECGs was obtained using an ECG machine that automatically calculates the heart rate and measures PR, QRS, QT, and QTc intervals. The baseline value at visit and the change from baseline value at each timepoint are reported. The change from baseline value was calculated by subtracting the post-baseline value from the baseline value.~The study was terminated because of futility, therefore did not reach the planned end of study." (NCT02545283)
Timeframe: Baseline, Days 1, 2, and 5 of Cycle 1, Days 1, 2 of Cycles 2 and 3 (1 cycle is 28 days), Treatment Discontinuation Visit (28 days after last dose of study drug)

,
InterventionMillisecond (msec) (Mean)
PR Duration BaselinePR Duration Cycle 1 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboPR Duration Cycle 1 Day 1 - Post-CytarabinePR Duration Cycle 1 Day 1 6 Hours Post-Idasanutlin/PlaceboPR Duration Cycle 1 Day 2PR Duration Cycle 1 Day 5 Within 2 Hours Pre-Idasanutlin/PlaceboPR Duration Cycle 1 Day 5 Post-CytarabinePR Duration Cycle 1 Day 5 6 Hours Post-Idasanutlin/PlaceboPR Duration Cycle 2 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboPR Duration Cycle 2 Day 1 Post-CytarabinePR Duration Cycle 2 Day 1 6 Hours Post-Idasanutlin/PlaceboPR Duration Cycle 2 Day 2PR Duration Cycle 3 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboPR Duration Cycle 3 Day 1 Post-CytarabinePR Duration Cycle 3 Day 1 6 Hours Post-Idasanutlin/PlaceboPR Duration Cycle 3 Day 2PR Duration Study Drug Completion/DiscontinuationQRS Duration BaselineQRS Duration Cycle 1 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQRS Duration Cycle 1 Day 1 Post-CytarabineQRS Duration Cycle 1 Day 1 6 Hours Post-Idasanutlin/PlaceboQRS Duration Cycle 1 Day 2QRS Duration Cycle 1 Day 5 Within 2 Hours Pre-Idasanutlin/PlaceboQRS Duration Cycle 1 Day 5 Post-CytarabineQRS Duration Cycle 1 Day 5 6 Hours Post-Idasanutlin/PlaceboQRS Duration Cycle 2 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQRS Duration Cycle 2 Day 1 Post-CytarabineQRS Duration Cycle 2 Day 1 6 Hours Post-Idasanutlin/PlaceboQRS Duration Cycle 2 Day 2QRS Duration Cycle 3 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQRS Duration Cycle 3 Day 1 Post-CytarabineQRS Duration Cycle 3 Day 1 6 Hours Post-Idasanutlin/PlaceboQRS Duration Cycle 3 Day 2QRS Duration Study Drug Completion/DiscontinuationQT Duration BaselineQT Duration Cycle 1 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQT Duration Cycle 1 Day 1 Post-CytarabineQT Duration Cycle 1 Day 1 6 Hours Post-Idasanutlin/PlaceboQT Duration Cycle 1 Day 2QT Duration Cycle 1 Day 5 Within 2 Hours Pre-Idasanutlin/PlaceboQT Duration Cycle 1 Day 5 Post-CytarabineQT Duration Cycle 1 Day 5 6 Hours Post-Idasanutlin/PlaceboQT Duration Cycle 2 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQT Duration Cycle 2 Day 1 Post-CytarabineQT Duration Cycle 2 Day 1 6 Hours Post-Idasanutlin/PlaceboQT Duration Cycle 2 Day 2QT Duration Cycle 3 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQT Duration Cycle 3 Day 1 Post-CytarabineQT Duration Cycle 3 Day 1 - 6 Hours Post-Idasanutlin/PlaceboQT Duration Cycle 3 Day 2QT Duration Study Drug Completion/DiscontinuationQTcB BaselineQTcB Cycle 1 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQTcB Cycle 1 Day 1 Post-CytarabinQTcB Cycle 1 Day 1 6 Hours Post-Idasanutlin/PlaceboQTcB Cycle 1 Day 2QTcB Cycle 1 Day 5 Within 2 Hours Pre-Idasanutlin/PlacebQTcB Cycle 1 Day 5 Post-CytarabineQTcB Cycle 1 Day 5 6 Hours Post-Idasanutlin/PlaceboQTcB Cycle 2 Day 1 - Within 2 Hours Pre-Idasanutlin/PlaceboQTcB Cycle 2 Day 1 Post-CytarabineQTcB Cycle 2 Day 1 6 Hours Post-Idasanutlin/PlaceboQTcB Cycle 2 Day 2QTcB Cycle 3 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQTcB Cycle 3 Day 1 Post-CytarabineQTcB Cycle 3 Day 1 6 Hours Post-Idasanutlin/PlaceboQTcB Cycle 3 Day 2QTcB Study Drug Completion/DiscontinuationQTcF BaselineQTcF Cycle 1 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQTcF Cycle 1 Day 1 Post-CytarabineQTcF Cycle 1 Day 1 6 Hours Post-Idasanutlin/PlaceboQTcF Cycle 1 Day 2QTcF Cycle 1 Day 5 Within 2 Hours Pre-Idasanutlin/PlaceboQTcF Cycle 1 Day 5 Post-CytarabineQTcF Cycle 1 Day 5 6 Hours Post-Idasanutlin/PlaceboQTcF Cycle 2 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQTcF Cycle 2 Day 1 Post-CytarabineQTcF Cycle 2 Day 1 6 Hours Post-Idasanutlin/PlaceboQTcF Cycle 2 Day 2QTcF Cycle 3 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboQTcF Cycle 3 Day 1 - Post-CytarabineQTcF Cycle 3 Day 1 6 Hours Post-Idasanutlin/PlaceboQTcF Cycle 3 Day 2QTcF Study Drug Completion/DiscontinuationRR Duration BaselineRR Duration Cycle 1 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboRR Duration Cycle 1 Day 1 Post-CytarabineRR Duration Cycle 1 Day 1 6 Hours Post-Idasanutlin/PlaceboRR Duration Cycle 1 Day 2RR Duration Cycle 1 Day 5 Within 2 Hours Pre-Idasanutlin/PlaceboRR Duration Cycle 1 Day 5 Post-CytarabineRR Duration Cycle 1 Day 5 6 Hours Post-Idasanutlin/PlaceboRR Duration Cycle 2 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboRR Duration Cycle 2 Day 1 Post-CytarabineRR Duration Cycle 2 Day 1 6 Hours Post-Idasanutlin/PlaceboRR Duration Cycle 2 Day 2RR Duration Cycle 3 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboRR Duration Cycle 3 Day 1 Post-CytarabineRR Duration Cycle 3 Day 1 6 Hours Post-Idasanutlin/PlaceboRR Duration Cycle 3 Day 2RR Duration Study Drug Completion/Discontinuation
Idasanutlin Plus Cytarabine155.5-2.71.61.61.4-2.1-1.6-3.52.24.96.05.96.73.96.914.1-7.789.3-1.9-1.0-1.11.11.60.2-0.30.4-2.10.10.2-2.4-2.0-2.7-1.6-0.4385.64.24.40.03.11.64.73.48.510.64.19.915.922.019.028.0-4.8437.54.83.86.42.4-3.2-3.7-5.51.64.24.6-1.0-1.113.621.914.35.8419.05.14.14.22.6-1.1-0.4-2.14.06.44.01.114.016.820.719.22.9787.94.83.6-19.14.119.536.837.825.723.3-15.214.165.538.93.362.0-14.8
Placebo Plus Cytarabine160.312.44.80.62.1-3.01.2-5.730.0-4.2-3.0-3.09.3-7.7-4.5-1.0-5.392.8-0.4-0.9-0.6-0.51.10.60.30.86.52.00.3-0.12.70.41.5-1.3388.76.00.71.15.424.825.027.8-9.1-11.5-7.512.21.921.2-3.4-9.4-5.6435.38.71.31.4-1.8-8.3-4.4-5.20.53.06.50.813.116.66.91.41.9418.07.61.00.91.33.66.16.00.00.51.56.114.018.53.5-2.60.2797.9-2.5-4.7-3.037.6159.3133.7144.2-5.4-38.8-66.957.915.111.1-49.4-42.9-14.8

Change From Baseline in Heart Rate, as Measured by Electrocardiogram

"Single 12-lead ECGs was obtained using an ECG machine that automatically calculates the heart rate and measures PR, QRS, QT, and QTc intervals. The baseline value at visit and the change from baseline value at each timepoint are reported. The change from baseline value was calculated by subtracting the post-baseline value from the baseline value.~The study was terminated because of futility, therefore did not reach the planned end of study." (NCT02545283)
Timeframe: Baseline; Cycles 1-3 Day 1 (2 hrs pre-dose, post-Cytarabine and 6 hrs post-Idasanutlin/Placebo ); Cycle 1-3 Day 2; Cycle 1 Day 5 (within 2 hrs Idanasanutlin/Placebo, post-Cytarabine and 6 hrs post-Idasanutlin/Placebo), Study Drug Completion

,
InterventionBeats per Minute (Mean)
BaselineCycle 1 Day 1 2 Hours Pre-doseCycle 1 Day 1 Post-CytarabineCycle 1 Day 1 6 Hours Post-Idasanutlin/PlaceboCycle 1 Day 2Cycle 1 Day 5 Within 2 Hours Pre-Idasanutlin/PlaceboCycle 1 Day 5 - Post-CytarabineCycle 1 Day 5 6 Hours Post-Idasanutlin/PlaceboCycle 2 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboCycle 2 Day 1 Post-CytarabineCycle 2 Day 1 6 Hours Post-Idasanutlin/PlaceboCycle 2 Day 2Cycle 3 Day 1 Within 2 Hours Pre-Idasanutlin/PlaceboCycle 3 Day 1 Post-CytarabineCycle 3 Day 1 6 Hours Post-Idasanutlin/PlaceboCycle 3 Day 2Study Drug Completion/Discontinuation
Idasanutlin Plus Cytarabine78.6-0.8-0.41.4-0.4-1.2-3.0-3.1-4.8-4.8-0.7-4.8-7.1-4.5-0.1-9.34.5
Placebo Plus Cytarabine77.31.1-0.10.1-2.2-10.3-9.9-10.7-0.50.8-0.7-5.2-3.0-8.9-2.00.13.4

Change From Baseline in Pulse Rate Over Time

"Vital signs were measured prior to the infusion while the participant was in a seated position. The baseline value at visit and the change from baseline value at each timepoint are reported. The change from baseline value was calculated by subtracting the post-baseline value from the baseline value.~The study was terminated because of futility, therefore did not reach the planned end of study." (NCT02545283)
Timeframe: Baseline; Cycles 1-3, Days 8, 15, 22, 28 (1 cycle is 28 days); and, if incomplete blood count recovery, Days 29-42 and Days 43-56, Cycles 2 -3 Days 1, 8, 15, 22, 28 and 29-56 (max delay between cycles is 56 days)

,
InterventionBeats per Minute (Mean)
BaselineCycle 1 Day 8Cycle 1 Day 15Cycle 1 Day 22Cycle 1 Day 28Cycle 1 Day 29-42Cycle 1 Day 43-56Cycle 2 Day 1Cycle 2 Day 8Cycle 2 Day 15Cycle 2 Day 22Cycle 2 Day 28Cycle 2 Day 29-56Cycle 3 Day 1Cycle 3 Day 8Cycle 3 Day 15Cycle 3 Day 22Cycle 3 Day 28Cycle 3 Day 29-56
Idasanutlin Plus Cytarabine79.23.41.14.14.05.16.8-0.92.41.70.31.24.01.02.41.93.40.95.2
Placebo Plus Cytarabine78.4-4.31.50.24.82.05.52.0-3.3-0.42.3-2.31.80.41.32.54.80.4-5.0

Change From Baseline in Respiratory Rate Over Time

"Vital signs were measured prior to the infusion while the participant was in a seated position. The baseline value at visit and the change from baseline value at each timepoint are reported. The change from baseline value was calculated by subtracting the post-baseline value from the baseline value.~The study was terminated because of futility, therefore did not reach the planned end of study." (NCT02545283)
Timeframe: Up to Approximately 4.5 Years

,
InterventionBreaths per Minute (Mean)
BaselineCycle 1 Day 8Cycle 1 Day 15Cycle 1 Day 22Cycle 1 Day 28Cycle 1 Day 29-42Cycle 1 Day 43-56Cycle 2 Day 1Cycle 2 Day 8Cycle 2 Day 15Cycle 2 Day 22Cycle 2 Day 28Cycle 2 Day 29-56Cycle 3 Day 1Cycle 3 Day 8Cycle 3 Day 15Cycle 3 Day 22Cycle 3 Day 28Cycle 3 Day 29-56
Idasanutlin Plus Cytarabine16.60.00.40.70.60.50.0-0.20.10.00.40.50.30.2-1.00.90.80.80.1
Placebo Plus Cytarabine16.3-0.10.70.60.40.70.3-0.3-0.20.50.8-0.30.21.10.21.51.10.30.7

Change From Baseline in Systolic Blood Pressure Over Time

"Vital signs were measured prior to the infusion while the participant was in a seated position. The baseline value at visit and the change from baseline value at each timepoint are reported. The change from baseline value was calculated by subtracting the post-baseline value from the baseline value.~The study was terminated because of futility, therefore did not reach the planned end of study." (NCT02545283)
Timeframe: Baseline; Cycles 1-3, Days 8, 15, 22, 28 (1 cycle is 28 days); and, if incomplete blood count recovery, Days 29-42 and Days 43-56, Cycles 2 -3 Days 1, 8, 15, 22, 28 and 29-56 (max delay between cycles is 56 days)

,
InterventionMillimeters of mercury (mmHg) (Mean)
BaselineCycle 1 Day 8Cycle 1 Day 15Cycle 1 Day 22Cycle 1 Day 28Cycle 1 Day 29-42Cycle 1 Day 43-56Cycle 2 Day 1Cycle 2 Day 8Cycle 2 Day 15Cycle 2 Day 22Cycle 2 Day 28Cycle 2 Day 29-56Cycle 3 Day 1Cycle 3 Day 8Cycle 3 Day 15Cycle 3 Day 22Cycle 3 Day 28Cycle 3 Day 29-56
Idasanutlin Plus Cytarabine122.1-6.9-0.1-1.5-0.53.43.80.6-5.1-0.22.55.06.62.3-1.70.2-1.17.15.1
Placebo Plus Cytarabine120.6-3.7-2.80.71.24.49.86.95.3-1.75.55.8-1.011.26.03.313.95.327.7

Cumulative Dose of Idasanutlin and Cytarabine

The cumulative doses of idasanutlin and cytaradine are reported. (NCT02545283)
Timeframe: Up to 3 cycles (1 cycle is 28 days)

InterventionMilligram (mg) (Mean)
Cytarabine cumulative dose (mg)
Placebo Plus Cytarabine11500

Cumulative Dose of Idasanutlin and Cytarabine

The cumulative doses of idasanutlin and cytaradine are reported. (NCT02545283)
Timeframe: Up to 3 cycles (1 cycle is 28 days)

InterventionMilligram (mg) (Mean)
Idasanutlin/Placebo cumulative dose (mg)Cytarabine cumulative dose (mg)
Idasanutlin Plus Cytarabine3340.111200

Number of Participants With Clinical Laboratory Abnormalities in Biochemistry Tests at the Greatest Severity, According to NCI-CTCAE v4.03

"Laboratory parameters for blood biochemistry will be measured and compared with a standard reference range. Values outside the standard reference range are considered abnormalities. A laboratory test result will be reported as an adverse event if it meets any of the following criteria: is accompanied by clinical symptoms; results in a change in study treatment or a medical intervention; or is clinically significant in the investigator's judgment.~For each patient, baseline is the last observation prior to initiation of study drug. For each laboratory test, patients with at least 1 post-baseline assessment are included in the analysis. For each cell, the denominator is the number of patients with baseline NCI-CTCAE Grade 0-2 in the specified direction of abnormality, or Grade 1-4 in the opposite direction of abnormality." (NCT02545283)
Timeframe: Up to Approximately 4.5 Years

,
InterventionParticipants (Number)
Albumin LowAlkaline Phosphatase HighSGPT/ALT HighSGOT/AST HighCalcium LowCreatinine HighGlucose LowGlucose HighMagnesium LowMagnesium HighPhosphorus LowPotassium HighSodium LowSodium HighBilirubin HighUric Acid HighPotassium Low
Idasanutlin Plus Cytarabine22916133611025810728177425376
Placebo Plus Cytarabine821398419051856192921

Number of Participants With Clinical Laboratory Abnormalities in Hematology Tests at the Greatest Severity, According to NCI-CTCAE v4.03

"Laboratory parameters for hematology will be measured and compared with a standard reference range. Values outside the standard reference range are considered abnormalities. A laboratory test result will be reported as an adverse event if it meets any of the following criteria: is accompanied by clinical symptoms; results in a change in study treatment or a medical intervention; or is clinically significant in the investigator's judgment.~For each patient, baseline is the last observation prior to initiation of study drug. For each laboratory test, patients with at least 1 post-baseline assessment are included in the analysis. For each cell, the denominator is the number of patients with baseline NCI-CTCAE Grade 0-2 in the specified direction of abnormality, or Grade 1-4 in the opposite direction of abnormality." (NCT02545283)
Timeframe: Up to Approximately 4.5 Years

,
InterventionParticipants (Number)
Hemoglobin LowHemoglobin HighLymphocytes Abs LowLymphocytes Abs HighNeutrophils, Total, Abs LowPlatelet LowTotal Leukocyte Count LowTotal Leukocyte Count High
Idasanutlin Plus Cytarabine19112291971281841
Placebo Plus Cytarabine83110914059871

Overall Survival in Clinically Actionable Mutation-Defined Subpopulation (FLT3, IDH1 and IDH2) in TP53 WT Population

Overall survival in mutation-defined subgroups (positive for mutation of FLT3, IDH1 and IDH2 respectively) of the TP53 WT population was compared in participants who had been randomized to idasanutlin in combination with cytarabine versus those who had been randomized to cytarabine and placebo. The Median upper limits were set to 999 because they were not reached and therefore it was Not Evaluable (NE) and the field is numeric only. The study was terminated because of futility, therefore did not reach the planned end of the study. (NCT02545283)
Timeframe: From randomization to death from any cause (up to approximately 4.5 years)

,
InterventionMonths (Median)
IDH2IDH1FLT3
Idasanutlin Plus Cytarabine11.018.255.55
Placebo Plus Cytarabine11.379.134.76

Percentage of Participants With Complete Response (CR) in Clinically Actionable Mutation-Defined Subpopulation (FLT3, IDH1 and IDH2) in TP53 WT Population

Rate of complete response at the end of induction in mutation-defined subgroups (positive for mutation of FLT3, IDH1 and IDH2, respectively) of the TP53 WT population was compared in participants who had been randomized to idasanutlin in combination with cytarabine versus those who had been randomized to cytarabine and placebo. (NCT02545283)
Timeframe: At the end of induction (up to Day 56)

,
InterventionPercentage of Participants (Number)
FLT3IDH1IDH2
Idasanutlin Plus Cytarabine15.334.829.5
Placebo Plus Cytarabine12.511.123.1

Disease-free Survival (DFS) for Patients Who Are MRD Negative

Disease-free survival for patients who are MRD negative but lack high or low risk molecular and cytogenetic features, defined as time from end of first course of therapy to death or relapse (NCT04326439)
Timeframe: Up to 2 years post-intervention

Interventionyears (Median)
Patients Who Are MRD Negative1.08

Event-free Survival (EFS) in Low Risk Patients and High Risk Patients

Event-free survival defined as the time from on study to death, failure to achieve remission or relapse in newly diagnosed patients with pediatric acute myeloid leukemia in the Low risk stratification group and High risk stratification group (NCT04326439)
Timeframe: Up to 2 years post-intervention

Interventionyears (Median)
Aflac-AML Low Risk Patients1.20
Aflac-AML High Risk Patients0.76

Minimal Residual Disease (MRD) Negative Status

Number of patients that are in remission (MRD negative) after course 1 in participants receiving GO and those that did not receive GO. (NCT04326439)
Timeframe: Post-induction I, an average of 28 days

InterventionParticipants (Count of Participants)
Induction I With GO5
Induction I Without GO3

Overall Survival (OS)

Time from study entry and from end of first course of therapy for newly diagnosed patients with pediatric acute myeloid leukemia (NCT04326439)
Timeframe: Up to 2 years post-intervention

Interventionyears (Median)
Aflac-AML Low Risk Patients1.97
Aflac-AML High Risk Patients1.98

Cardiotoxicity in Patients With de Novo AML That Receive the Four-cycle Aflac-AML Regimen With the Inclusion of Dexrazoxane

Number of patients that develop cardiac ejection fraction <50% (CTCAE V5.0 grade 2 or greater dysfunction) either during therapy (early cardiotoxicity) or after completion of therapy (late cardiotoxicity) (NCT04326439)
Timeframe: At completion of Cycle 4 (each cycle average is 28 days)

,
InterventionParticipants (Count of Participants)
Early cardiotoxicityLate cardiotoxicity
Aflac-AML High Risk Patients00
Aflac-AML Low Risk Patients00

Duration of Hospital Admission in Patients That Developed Infection and Febrile Neutropenia at the End of Each Treatment Cycle

Duration of hospital admission in patients that developed infection and febrile neutropenia at the end of each treatment cycle (NCT04326439)
Timeframe: At the end of each cycle (each cycle average is 28 days)

Interventiondays (Mean)
Cycle 1 - Induction 1Cycle 2 - Induction 2
Aflac-AML High Risk Patients3030.0

Duration of Hospital Admission in Patients That Developed Infection and Febrile Neutropenia at the End of Each Treatment Cycle

Duration of hospital admission in patients that developed infection and febrile neutropenia at the end of each treatment cycle (NCT04326439)
Timeframe: At the end of each cycle (each cycle average is 28 days)

Interventiondays (Mean)
Cycle 1 - Induction 1Cycle 2 - Induction 2Cycle 3 - Induction 3Cycle 4 - Induction 4
Aflac-AML Low Risk Patients3329.32927.5

Proportion of Patients Who Develop Infection and/or Febrile Neutropenia During Each Treatment Course

Number of participants that developed infection and/or febrile neutropenia at the end of each treatment cycle. (NCT04326439)
Timeframe: At the end of each cycle (each cycle average is 28 days)

,
InterventionParticipants (Count of Participants)
Cycle 1 - Induction 1Cycle 2 - Induction 2Cycle 3 - Induction 3Cycle 4 - Induction 4
Aflac-AML High Risk Patients2200
Aflac-AML Low Risk Patients5322

Duration of Event-Free Survival (EFS)

"EFS was defined as the time from the date of randomization until the date of documented relapse (excluding relapse after PR), treatment failure or death from any cause within 30 days after the last dose of study drug, whichever occurred first (earliest of [relapse date, treatment failure date, death date] - randomization date + 1). If a participant experienced relapse or death within 30 days after the last dose of study drug, the participant was defined as having an EFS event related to either relapse or death, and the event date was the date of relapse or death. For a participant who was not known to have had a relapse or treatment failure or death event, EFS was censored at the date of last relapse-free disease assessment (last relapse-free disease assessment date - randomization date + 1). Data was estimated based on Kaplan-Meier estimates." (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, median time of follow-up for OS was 17.8 months

InterventionMonths (Median)
Gilteritinib2.8
Salvage Chemotherapy0.7

Duration of Leukemia-Free Survival (LFS)

The LFS was defined as the time from the date of first CRc until the date of documented relapse (excluding relapse from PR) or death for participants who achieved CRc (relapse date or death date - first CRc disease assessment date + 1). For a participant who was not known to have relapsed or died, LFS was censored on the date of last relapse-free disease assessment date (last relapse-free disease assessment date - first CRc disease assessment date + 1). For a participant who was not known to have relapsed or died, LFS was censored on the date of last relapse-free disease assessment date (last relapse-free disease assessment date - first CRc disease assessment date + 1). (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, median time of follow-up for OS was 17.8 months

InterventionMonths (Median)
Gilteritinib4.4
Salvage Chemotherapy6.7

Duration of Overall Survival (OS)

Overall survival was defined as the time from the date of randomization until the date of death from any cause (death date - randomization date + 1). For a participant who was not known to have died by the end of study follow-up, OS was censored at the date of last contact (date of last contact - randomized date + 1). The date of last contact was the latest date that the participant was known to be alive by the cutoff date. The last contact date was derived for participants alive at the analysis cutoff date. Survival rate and 95% CI were estimated using the Kaplan-Meier method and the Greenwood formula. (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, median time of follow-up for OS was 17.8 months

InterventionMonths (Median)
Gilteritinib9.3
Salvage Chemotherapy5.6

Duration of Remission

Duration of remission included duration of composite complete remission (CRc), duration of complete remission (CR)/ complete remission with partial hematologic recovery (CRh), duration of CRh, duration of CR and duration of response (CRc + partial remission (PR). The duration of response was defined as the time from the date of either first CRc or PR until the date of documented relapse (i.e., the date of first NR after CRc or PR) for participants who achieved CRc or PR (relapse date - first CRc or PR disease assessment date + 1). Participants who died without report of relapse were considered nonevents and censored at their last relapse-free disease assessment date (last relapse-free disease assessment date - first CRc or PR disease assessment date + 1). Other participants who did not relapse during the study were considered nonevents and censored at the last relapse-free assessment date. Duration of CR was only applicable to participants with best overall response of CR. (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, median time of follow-up for OS was 17.8 months

InterventionMonths (Median)
Gilteritinib14.8
Salvage Chemotherapy1.8

Percentage of Participants Who Underwent Hematopoietic Stem Cell Transplant

Transplantation rate is defined as the percentage of participants undergoing Hematopoietic stem cell transplant (HSCT) during the study period. (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, median time of follow-up for OS was 17.8 months

InterventionPercentage of participants (Number)
Gilteritinib25.5
Salvage Chemotherapy15.3

Percentage of Participants With Complete Remission (CR) Rate

The CR rate was defined as the number of participants who achieved the best response of CR divided by the number of participants in the analysis population. (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, all participants included in the primary analysis of CR rate were followed up at least 6 months

InterventionPercentage of participants (Number)
Gilteritinib21.1
Salvage Chemotherapy10.5

Percentage of Participants With Complete Remission (CR) With Partial Hematological Recovery (CRh)

CRh rate was defined as the number of participants who achieved CRh at any of the postbaseline visits and did not have a best response of CR divided by the number of participants in the analysis population. (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, median time of follow-up for OS was 17.8 months

InterventionPercentage of participants (Number)
Gilteritinib34.0
Salvage Chemotherapy15.3

Percentage of Participants With Composite Complete Remission (CRc Rate)

CRc rate was defined as the number of participants who achieved the best response of CRc (CR,complete remission with incomplete platelet recovery (CRp) or complete remission with incomplete hematologic recovery (CRi) divided by the number of participants in the analysis population. (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, median time of follow-up for OS was 17.8 months

InterventionPercentage of participants (Number)
Gilteritinib54.3
Salvage Chemotherapy21.8

Change From Baseline in Brief Fatigue Inventory (BFI)

The Brief Fatigue Inventory (BFI) is a screening tool designed to assess the severity and impact of fatigue on daily functioning of participants with cancer during the 24 hours. There are 9 items on the scale. The first three questions ask participants to rate their fatigues on a scale from 0 (no fatigue) - 10 (as bad as you can imagine), with higher scores indicating worse outcome. The remaining six questions ask participants to rate how much fatigue has interfered with their daily activities on a scale from 0 (Does not interfere) to 10 (Completely interferes). A global fatigue score can be obtained by averaging all the items on the BFI. The global BFI score will be calculated only if at least 5 of the 9 items are answered. A higher BFI fatigue score indicates worse outcome. (NCT02421939)
Timeframe: Baseline and cycle 1, day 8 and cycle 2 day 1 (up to data cut off date of 17 Sep 2018)

,
InterventionUnits on a scale (Mean)
Cycle 1 day 8 (C1D8)Cycle 2 day 1 (C2D1)
Gilteritinib-0.40.0
Salvage Chemotherapy1.00.4

Number of Participants With Adverse Events

"A treatment-emergent adverse event (TEAE) was defined as an AE observed after starting administration of the study drug (gilteritinib or salvage chemotherapy). If the AE occurred on day 1 and the onset check box was marked Onset after first dose of study drug or the onset check box was left blank, then the AE was considered treatment emergent. If the AE occurred on day 1 and the onset check box was marked Onset before first dose of study drug, then the AE was not considered treatment emergent. Majority of salvage chemotherapy participants finished the study by cycle 2 of treatment, the duration of exposure was longer in the gilteritinib arm compared with the salvage chemotherapy arm (126.00 [4.0, 885.0] days versus 28.0 [5.0, 217.0] days). The NCI-CTCAE is defined as National Cancer Institute-Common Terminology Criteria for Adverse Events." (NCT02421939)
Timeframe: From first dose of study drug up to 30 days after the last dose of study drug (median treatment duration for gilteritinib was (126.00 [4.0, 885.0]) days versus salvage chemotherapy 28.0 [5.0, 217.0] days)

,
InterventionParticipants (Count of Participants)
Drug-related TEAESerious TEAEDrug-related serious TEAETEAE leading to deathDrug-related TEAE leading to deathTEAE leading to withdrawal of treatmentDrug-related TEAE lead withdrawal of treatmentNCI-CTCAE Grade 3 or higher TEAEDrug-related Grade 3 or higher TEAEDeath
Gilteritinib2062058871105827236153170
Salvage Chemotherapy713416165135945781

Percentage of Participants Who Achieved Transfusion Conversion and Maintenance

Transfusion conversion & maintenance rate was defined for gilteritinib arm. Participants were classified as transfusion independent if there were no RBC or platelet transfusions within 28 days prior to the first dose to 28 days after the first dose; otherwise they were classified as transfusion dependent at baseline. Participants were considered independent postbaseline if they had 1 consecutive 8 week period without any RBC or platelet transfusion from 29 days after the first dose until the last dose date. For participants who were on treatment ≤ 4 weeks or > 4 weeks but < 12 weeks and there was no RBC or platelet transfusion within postbaseline period, they were considered not evaluable; otherwise, they were considered postbaseline transfusion dependent. Transfusion conversion rate was defined for participants who had evaluable postbaseline transfusion status. Transfusion status (independent vs. dependent) at baseline and postbaseline was reported in a 2 by 2 contingency table. (NCT02421939)
Timeframe: From randomization until the data cut-off date of 17 Sep 2018, median treatment duration for gilteritinib was (126.00 [4.0, 885.0]) days versus salvage chemotherapy 28.0 [5.0, 217.0] days)

InterventionPercentage of participants (Number)
Baseline Independent/ Post baseline IndependentBaseline Independent/Post baseline DependentBaseline Independent/Post baseline Not EvaluableBaseline Dependent/Post baseline IndependentBaseline Dependent/Post baseline DependentBaseline Dependent/Post baseline Not Evaluable
Gilteritinib59.224.516.334.555.89.6

Percentage of Participants With Complete Remission and Complete Remission With Partial Hematological Recovery (CR/CRh) in the Gilteritinib Arm

The CR/CRh rate was defined as the number of participants who achieved either CR or CRh at any of the postbaseline visits divided by the number of participants in the analysis population. (NCT02421939)
Timeframe: From randomization until the data cut-off date 04 Aug 2017, the 142 patients included in the primary analysis of CR/CRh rate were followed up at least 112 days

InterventionPercentage of participants (Number)
CR/CRh rateCR rateCRh rate
Gilteritinib28.219.09.2

4 Year Overall Survival Rate

Percentage of patients who were alive at 4 years. The 4-year survival rate was estimated using the Kaplan Meier method. (NCT00098774)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Intensive Combination Chemo & Immunotherapy65

4 Year Progression Free Rate

"Percentage of patients who were progression free at 4 years. The 4-year progression free rate was estimated using the Kaplan Meier method.~Relapse was assessed by investigator according to Revised Response Criteria for Malignant Lymphoma. Progression required a 25% increase of previous area of gadolinium enhancement, appearance of new areas of T1 gadolinium enhancement or new appearance of malignant cells in the spinal fluid or new tumor appearance in other sites of the body" (NCT00098774)
Timeframe: 4 years

Interventionpercentage of participants (Number)
Intensive Combination Chemo & Immunotherapy48

Complete Response Rate After Remission Induction

Response is assessed by investigator according to Revised Response Criteria for Malignant Lymphoma. Complete response requires disappearance of all evidence of disease. (NCT00098774)
Timeframe: 4 months

Interventionpercentage of participants (Number)
Intensive Combination Chemo & Immunotherapy66

Change From Baseline in Mini-Mental Status Evaluation at 4 Months

Neurologic functioning will be assessed using the Mini-Mental Status Evaluation (MMSE), a standardized, bedside tool for evaluation of higher mental function. This assessment is based on a 30-point scale (0-30) with higher scores associated with better performance. (NCT00098774)
Timeframe: Baseline & month 4

Interventionunits on a scale (Median)
Baseline Score4 month ScoreChange from Baseline
Intensive Combination Chemo & Immunotherapy27281

Disease-free Survival (DFS)

Time from end of Intensification I to relapse, death or last contact (NCT00372593)
Timeframe: At 3 years from end of Intensification I

InterventionPercentage participants DFS at 3 years (Number)
Arm A: Standard Arm - No GMTZ, AML Pts w/Out Down Syndrome56.6
Arm B: Experimental - With GMTZ, AML Pts w/Out Down Syndrome63.0
Arm A: Standard Arm - No GMTZ, AML Patients With Down Syndrome75.0

Event-free Survival at 3 Years

The Kaplan-Meier method will be used to calculate estimates of Event Free Survival (EFS). The log-rank test will be used to compare survival between treatment groups. Analysis of EFS of Down syndrome patients will be performed separately. Monitoring for efficacy of GMTZ with respect to Overall Survival (OS) and EFS will utilize monitoring based on the Lan-DeMets criterion with α-spending function αt^2 (truncated at 3 standard deviations) and 2.5% type I error. (NCT00372593)
Timeframe: Time from study entry to time of induction failure, relapse, or death, assessed at 3 years

Interventionpercentage of participants (Number)
Arm A: Standard Arm - No GMTZ, AML Pts w/Out Down Syndrome46.9
Arm B: Experimental - With GMTZ, AML Pts w/Out Down Syndrome53.1
Arm A: Standard Arm - No GMTZ, AML Patients With Down Syndrome50.0

Mortality

Number of participants who died during the first three courses of therapy. (NCT00372593)
Timeframe: During the first three courses of therapy

InterventionNumber of participants (Number)
Arm A: Standard Arm - No GMTZ, AML Pts w/Out Down Syndrome11
Arm B: Experimental - With GMTZ, AML Pts w/Out Down Syndrome13
Arm A: Standard Arm - No GMTZ, AML Patients With Down Syndrome1

Overall Survival at 3 Years

The Kaplan-Meier method will be used to calculate estimates of OS. Analysis of OS of Down syndrome patients will be performed separately. Monitoring for efficacy of GMTZ with respect to OS and EFS will utilize monitoring based on the Lan-DeMets criterion with α-spending function αt^2 (truncated at 3 standard deviations) and 2.5% type I error. (NCT00372593)
Timeframe: Time from study entry, assessed at 3 years

Interventionpercentage of participants (Number)
Arm A: Standard Arm - No GMTZ, AML Pts w/Out Down Syndrome65.4
Arm B: Experimental - With GMTZ, AML Pts w/Out Down Syndrome69.4
Arm A: Standard Arm - No GMTZ, AML Patients With Down Syndrome50.0

Remission Induction Rate After 2 Courses of Induction Therapy

Patients without an evaluable bone marrow at the end of Induction I will be excluded from the calculation of remission rate after 2 courses of therapy because their responses are not evaluable. The following patients will be considered to not be in complete remission (CR) after 2 courses of therapy: (1) patients who die during Induction I and II; (2) patients with ≥ 5% blasts or extramedullary disease at the end of Induction II. (NCT00372593)
Timeframe: After 2 courses of induction (I and II) therapy, assessed for up to 10 years

InterventionProportion of participants (Number)
Arm A: Standard Arm - No GMTZ, AML Pts w/Out Down Syndrome0.851324
Arm B: Experimental - With GMTZ, AML Pts w/Out Down Syndrome0.882716
Arm A: Standard Arm - No GMTZ, AML Patients With Down Syndrome0.666667

Toxicities, Including Infectious Complications

Number of participants with at least one grade 3 or higher adverse event during therapy. (NCT00372593)
Timeframe: From the time therapy is initiated, assessed up to 10 years

InterventionNumber of participants (Number)
Arm A: Standard Arm - No GMTZ, AML Pts w/Out Down Syndrome482
Arm B: Experimental - With GMTZ, AML Pts w/Out Down Syndrome477
Arm A: Standard Arm - No GMTZ, AML Patients With Down Syndrome5

Time to Marrow Recovery

Mean time to ANC recovery - defined as ANC greater than 500/MicroLiter for 3 consecutive days. (NCT00372593)
Timeframe: At 25 days after treatment with Induction I, Induction II, and Intensification I

,,
InterventionMean days (Mean)
During Induction I (days 0 - 28 of therapy)During Induction II (days 29 - 56 of therapy)During Intensification I (days 57 - 84 of therapy)
Arm A: Standard Arm - No GMTZ, AML Patients With Down Syndrome29.1726.624.5
Arm A: Standard Arm - No GMTZ, AML Pts w/Out Down Syndrome30.5628.5427.51
Arm B: Experimental - With GMTZ, AML Pts w/Out Down Syndrome30.5628.5228.10

Event-free Survival

Monitoring of efficacy results will be performed in comparison with historical results. (NCT00096135)
Timeframe: 3 years

Interventionpercentage of participants (Number)
CNS Patients - Treatment (Combination Chemotherapy)64.9
Testicular Relapse Patients (Combination Chemotherapy)70

Event-free Survival (EFS)

12-month event free survival (NCT02728050)
Timeframe: 12 months

Interventionpercentage of participants (Number)
CLAGM+Sorafenib Phase 281

Number of Participants With Adverse Events

Will be assessed using National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. (NCT02728050)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Phase 1, Dose Level 16
Phase 1, Dose Level 26
Phase 1, Dose Level 311
Phase 1, Dose Level 48
Phase 1, Dose Level 59
Phase 1, Dose Level 67
Phase 2, Dose Level 632

Overall Response Rate (ORR)

ORR, defined as CR+CRi, rates of patients treated with CLAG-M with sorafenib. (NCT02728050)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Phase 1, Dose Level 16
Phase 1, Dose Level 26
Phase 1, Dose Level 38
Phase 1, Dose Level 45
Phase 1, Dose Level 59
Phase 1, Dose Level 67
Phase 2, Dose Level 634

Overall Survival (OS)

12-month overall survival (NCT02728050)
Timeframe: 12 months

Interventionpercentage of participants (Number)
CLAGM+Sorafenib Phase 286

Phase 1: Maximum Tolerated Dose (MTD)/Recommended Phase 2 Dose (RP2D) of Mitoxantrone

MTD/RP2D will be defined as the highest dose studied in which the incidence of dose-limiting toxicity (DLT) is < 33% assuming at least 6 patients have been treated at this dose. DLTs were defined as: 1) grade ≥3 non-hematologic toxicity lasting >48 hours leading to >7-day delay of the next cycle; 2) grade ≥4 non-hematologic toxicity if no recovery to grade ≤2 in 14 days (both excluding febrile neutropenia/ infection); 3) Absolute neutrophil count <500/ µL or platelet count <50,000/µL for >49 days after CLAGM+S without marrow evidence of AML. Doses were escalated up to dose level six if <2/6 patients out of each cohort of 6 had a DLT (some cohorts were expanded to 12 patients while awaiting completion of DLT monitoring period). The dose level at which dose escalation was stopped was the recommended phase 2 dose (RP2D). (NCT02728050)
Timeframe: First 28 days of treatment

Interventionmg/m^2 (Number)
CLAGM+Sorafenib Phase 118

Phase 1: Maximum Tolerated Dose (MTD)/Recommended Phase 2 Dose (RP2D) of Sorafenib

MTD/RP2D will be defined as the highest dose studied in which the incidence of dose-limiting toxicity (DLT) is < 33% assuming at least 6 patients have been treated at this dose. DLTs were defined as: 1) grade ≥3 non-hematologic toxicity lasting >48 hours leading to >7-day delay of the next cycle; 2) grade ≥4 non-hematologic toxicity if no recovery to grade ≤2 in 14 days (both excluding febrile neutropenia/ infection); 3) Absolute neutrophil count <500/ µL or platelet count <50,000/µL for >49 days after CLAGM+S without marrow evidence of AML. Doses were escalated up to dose level six if <2/6 patients out of each cohort of 6 had a DLT (some cohorts were expanded to 12 patients while awaiting completion of DLT monitoring period). The dose level at which dose escalation was stopped was the recommended phase 2 dose (RP2D). (NCT02728050)
Timeframe: First 28 days of treatment

Interventionmg BID (Number)
CLAGM+Sorafenib Phase 1400

Phase I and II: Rate of Minimal Residual Disease Negative (MRDneg) Complete Response (CR)

We will determine if the addition of sorafenib to CLAG-M improves the rate of MRDneg CR compared to our institution's historical control of CLAG-M alone in adults with newly-diagnosed AML/high-risk MDS. (NCT02728050)
Timeframe: 56 days (2 cycles of induction chemotherapy)

InterventionParticipants (Count of Participants)
Phase 1, Dose Level 13
Phase 1, Dose Level 26
Phase 1, Dose Level 38
Phase 1, Dose Level 44
Phase 1, Dose Level 59
Phase 1, Dose Level 67
Phase 2, Dose Level 631

Relapse-free Survival (RFS)

12-month relapse free survival (RFS) (NCT02728050)
Timeframe: 12 months

Interventionpercentage of participants (Number)
CLAGM+Sorafenib Phase 282

Number of Participants With a Complete Response Rate to Chemotherapy Regimen Assessed by Radiographic Response at 2 Years.

Per RECIST criteria (v1.1) and assessed by magnetic resonance imaging (MRI): Complete response (CR), Disappearance of all target lesions. (NCT00074165)
Timeframe: 2 years

InterventionParticipants (Number)
Rituximab and Carboplatin1

Overall Survival (OS)

Time from randomization to death. Patients alive at last follow-up were censored. (NCT00046930)
Timeframe: Assessed every 3 months for 2 years, then every 6 months for 3 years, then annually thereafter

InterventionMonths (Median)
Zosuquidar7.23
Placebo9.43

Progression-free Survival (PFS)

Time from randomization to the earlier of disease progression or death. Patients alive and progression-free at last follow-up were censored. (NCT00046930)
Timeframe: Assessed every 3 months for 2 years, then every 6 months for 3 years, then annually thereafter

InterventionMonths (Median)
Zosuquidar3.02
Placebo2.04

Response

Number of eligible participants in each response category. Categories, based on peripheral blood counts and bone marrow aspirate and biopsy, include complete remission (CR), partial remission (PR), morphologic complete remission (MCR), and relapse. (NCT00046930)
Timeframe: Assessed at the end of induction

,
InterventionParticipants (Number)
Complete RemissionMorphologic Complete RemissionPartial RemissionRelapseUnevaluable
Placebo961209023
Zosuquidar981226733

EFS in High Risk APL Patients

EFS is defined as the time from on study to failure to achieve hematological CR prior to start of consolidation, persistence of molecular positive disease after MRD positive consolidation course, relapse (molecular, morphologic or extramedullary), or death. The Kaplan-Meier method will be used to estimate 2-year EFS along with 90% log-minus-log transformed confidence limits. (NCT02339740)
Timeframe: Up to 24 months

Interventionpercentage of participants (Number)
High Risk96.1

Event-free Survival (EFS) in Standard Risk Acute Promyelocytic Leukemia (APL) Patients

EFS is defined as the time from on study to failure to achieve hematological complete response (CR) prior to start of consolidation, persistence of molecular positive disease after minimal residual disease (MRD) positive consolidation course, relapse (molecular, morphologic or extramedullary), or death. The Kaplan-Meier method will be used to estimate 2-year EFS along with 90% log-minus-log transformed confidence limits. (NCT02339740)
Timeframe: Up to 24 months

Interventionpercentage of participants (Number)
Standard Risk97.9

Continuous Complete Remission (CCR) Rate

Will be testing using an exact binomial test (NCT00792948)
Timeframe: 18 months

Interventionpercentage of participants (Number)
Treatment57

Overall Survival (OS)

OS will be estimated using the method of Kaplan-Meier. (NCT00792948)
Timeframe: From the date of initial registration on the study until death from any cause, assessed up to 5 years

InterventionProbability of surviving 12 months (Number)
Treatment0.88

Relapse-free Survival (RFS) After Allogeneic Stem Cell Transplantation

Will be estimated using the method of Kaplan-Meier. (NCT00792948)
Timeframe: 12 months

InterventionProbability of 12-month RFS (Number)
Treatment0.83

Cytarabine Drug Sensitivity by R-Strip (MicroMath) Curve Fitting Program

Mean and standard deviation of area under the concentration time curve. Specimen draws were performed only with dose 1, day 1 of induction II of AraC. First sample drawn pre-infusion, then drawn 30 mins prior to the end of the infusion, and then drawn for 6 time periods post infusion up to 8 hours post infusion (and before the 2nd dose of AraC). Results are based from these multiple time points on Day 1 of Induction II only. (NCT00369317)
Timeframe: Days 1, 2, 8, and 9 of induction II

InterventionMean micromolar x minutes (Mean)
Treatment (Combination Chemotherapy)1337.279

Cytarabine Drug Sensitivity by R-Strip (MicroMath) Curve Fitting Program

Mean and standard deviation of half-life of elimination. Specimen draws were performed only with dose 1, day 1 of induction II of AraC. First sample drawn pre-infusion, then drawn 30 mins prior to the end of the infusion, and then drawn for 6 time periods post infusion up to 8 hours post infusion (and before the 2nd dose of AraC). Results are based from these multiple time points on Day 1 of Induction II only. (NCT00369317)
Timeframe: Days 1, 2, 8, and 9 of induction II

InterventionMean minutes (Mean)
Treatment (Combination Chemotherapy)306.156034

Cytarabine Drug Sensitivity by R-Strip (MicroMath) Curve Fitting Program

Mean and standard deviation of peak plasma concentration. Specimen draws were performed only with dose 1, day 1 of induction II of AraC. First sample drawn pre-infusion, then drawn 30 mins prior to the end of the infusion, and then drawn for 6 time periods post infusion up to 8 hours post infusion (and before the 2nd dose of AraC). Results are based from these multiple time points on Day 1 of Induction II only. (NCT00369317)
Timeframe: Days 1, 2, 8, and 9 of induction II

InterventionMean micromolar (Mean)
Treatment (Combination Chemotherapy)32.69931

Event-free Survival (EFS) at 3 Years

(NCT00369317)
Timeframe: Time from study entry to induction failure, relapse, or death assessed at 3 years.

Interventionpercentage (Number)
Treatment (Combination Chemotherapy)90.1

Induction Remission Rate

Proportion of participants with a remission after four courses of Induction therapy. (NCT00369317)
Timeframe: End of induction therapy (day 112)

InterventionProportion of participants (Number)
Treatment (Combination Chemotherapy)0.984615

Overall Survival (OS) at 3 Years

(NCT00369317)
Timeframe: Time from study entry to death, assessed at 3 years.

Interventionpercentage (Number)
Treatment (Combination Chemotherapy)92.7

Percentage of Patients Experiencing Grade 3 or 4 Toxicity Assessed by the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0

Proportion of participants with at least one grade 3 or higher adverse event during therapy. (NCT00369317)
Timeframe: From the beginning of induction therapy to the end of intensification therapy

InterventionProportion of participants (Number)
Treatment (Combination Chemotherapy)0.911765

Prevalence of Leukemia Phenotype of DS Patients < 4 Years of Age at Diagnosis by Flow Cytometry

Proportion of participants having megakaryoblastic subtype (AMkL) phenotype among patients with phenotype data available. (NCT00369317)
Timeframe: At the start of therapy

InterventionProportion of participants (Number)
Treatment (Combination Chemotherapy)0.45122

Prevalence of of GATA1 Mutations of DS Patients < 4 Years of Age at Diagnosis

Proportion of participants having GATA1 mutation among patients with phenotype data available. (NCT00369317)
Timeframe: At baseline and at the end of therapy (intensification) or disease relapse

InterventionProportion of participants (Number)
Treatment (Combination Chemotherapy)0.891304

Proportions of Patients in Morphologic Remission With Positive MRD by Flow Cytometry

Proportion of participants in complete remission by morphology and with positive MRD by flow cytometry among patients having evaluable remission and MRD assessment. (NCT00369317)
Timeframe: After Induction I therapy (day 28 from start of therapy)

InterventionProportion of participants (Number)
Treatment (Combination Chemotherapy)0.0935254

Event-free Survival of Standard Risk Patients Who Receive Chemotherapy Alone.

Kaplan-Meier estimate of the probability of being alive and free of relapse or second malignancy three years after protocol enrollment (NCT00703820)
Timeframe: 3 years after completion of therapy

InterventionPercentage of participants (Number)
Cytarabine+Daunorubicin+Etoposide55.6
Clofarabine+Cytarabine54.3

Event-free Survival of Standard Risk Patients Who Receive Chemotherapy Followed by Natural Killer Cell Transplantation.

Kaplan-Meier estimate of the probability of being alive and free of relapse or second malignancy three years after protocol enrollment (NCT00703820)
Timeframe: 3 years after completion of therapy

InterventionPercentage of participants (Number)
Cytarabine+Daunorubicin+Etoposide55.6
Clofarabine+Cytarabine77.8

Disease Free Survival (DFS)

Estimated using the Kaplan-Meier estimator. Proportions will be estimated using point as well as interval estimators. All interval estimators will be constructed using the finite sample size sampling distribution at the unadjusted two-sided level of 0.05. (NCT01256398)
Timeframe: 10 years

InterventionMonths (Median)
Treatment (Chemotherapy, Transplant)29.7

Disease Free Survival Defined From the Date of First Induction Complete Response (CR) to Relapse or Death Due to Any Cause

Estimated using the Kaplan-Meier estimator. Proportions will be estimated using point as well as interval estimators. All interval estimators will be constructed using the finite sample size sampling distribution at the unadjusted two-sided level of 0.05. (NCT01256398)
Timeframe: At 3 years after CR

Interventionpercentage of patients (Number)
Treatment (Chemotherapy, Transplant)52.6

Feasibility of Maintenance Therapy in This Patient Population (Restricted to Those Patients Achieving a CR). Feasibility Will be Defined as the Number of Deaths Ocuring.

Proportions will be estimated based on the combined and individual cohorts. (NCT01256398)
Timeframe: 10 years

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, Transplant)5

Overall Survival (OS)

Estimated using the Kaplan-Meier estimator. Proportions will be estimated using point as well as interval estimators. All interval estimators will be constructed using the finite sample size sampling distribution at the unadjusted two-sided level of 0.05. (NCT01256398)
Timeframe: 10 years

InterventionMonths (Median)
Treatment (Chemotherapy, Transplant)55.9

Probability of Being BCR-ABL Negative in the Bone Marrow and Peripheral Blood at the Completion of the CNS Prophylaxis Course (Restricted to Those Patients Achieving a CR)

Proportions will be estimated based on the combined and individual cohorts. (NCT01256398)
Timeframe: 10 years

Interventionproportion of participants (Number)
Treatment (Chemotherapy, Transplant)0.667

Response

Proportion of patients reaching a CR. A CR requires the following: an absolute neutrophil count (segs and bands) >1000/μL, no circulating blasts, platelets >100,000/μL; adequate bone marrow cellularity with trilineage hematopoiesis, and <5% marrow leukemia blast cells. All previous extramedullary manifestations of disease must be absent (e.g., lymphadenopathy, splenomegaly, skin or gum infiltration, testicular masses, or CNS involvement). (NCT01256398)
Timeframe: 10 years

Interventionproportion of participants (Number)
Treatment (Chemotherapy, Transplant).9846

12-month PFS (Progression Free Survival) of Treatment With Ixazomib in Combination With DA-EPOCH-R (Phase II)

12-month PFS will be defined as the percentage of patients alive and progression free 12 months from start of therapy. Patients lost to follow up will be censored from last point of contact. PFS was calculated using a Kplan-meier estimation with the probability of patients being progression free at 12 months, presented as the percentage based on this probability. (NCT02481310)
Timeframe: Up to 12 months from initiation of treatment

Interventionprobability (%) of patients alive (Number)
Treatment (Combination Chemotherapy, Rituximab, Ixazomib)75.84

To Determine the Recommended Phase II Dose (RP2D) of Ixazomib in Combination With DA-EPOCH-R.

"The RP2D will be determined by the evaluation of dose-limiting toxicities (DLT) to find the Maximum Tolerated Dose (MTD) which will constitute the RP2D. DLTs will be defined as the occurrence of ≥ Grade 3 toxicity experienced during the first cycle (3 weeks or 21 days) of study treatment, will be determined, except for cytopenias.~The dose of ixazomib will start at 2.3 mg weekly (for phase I patients) when given concurrently with DA-EPOCH-R. Single-subject cohorts (escalating in ixazomib dose) will be enrolled until the MTD is encountered, after which expansion to 3-subject cohorts will be undertaken. The MTD from phase I will equate the RP2D for ixazomib given in conjunction with DA-EPOCH-R." (NCT02481310)
Timeframe: The first 21 days of treatment

Interventionmg (Number)
Treatment (Combination Chemotherapy, Rituximab, Ixazomib)3

Overall Response Rate

The Overall Response Rate was measured by the number of patients per the total treatment population who partially or completely responded to treatment. Response was evaluated according to the International Workshop to Standardize Response Criteria for Non-Hodgkin's Lymphomas. (NCT00336583)
Timeframe: up to 24 weeks

Interventionpariticipants (Number)
ESHAOx17

Worst Toxicity Grade by Patient

graded by National Cancer Institute Common Toxicity Criteria of Adverse Event version 3.0 (NCT00336583)
Timeframe: up to 24 weeks

Interventionparticipants (Number)
Grade 1 neutropeniaGrade 2 neutropeniaGrade 3 neutropeniaGrade 4 neutropeniaGrade 1 thrombocytopeniaGrade 2 thrombocytopeniaGrade 3 thrombocytopeniaGrade 4 thrombocytopenia
ESHAOx332134336

1 Year Progression-Free Survival

1-year progression-free survival is the probability of patients remaining alive and progression-free at 1 year from study entry estimated using Kaplan-Meier methods. Disease progression was based on the International Working Group (IWG) Criteria (Cheson et al, 1999). (NCT01661881)
Timeframe: Disease was assessed after three- and six-cycles of therapy and in long-term follow-up per standard practice every 6 months until the earliest of relapse, death or 5 years. Median follow-up in this study cohort was 13 months.

Interventionprobability (Number)
RB/RC.96

Autologous Stem Cell Transplant (ASCT) Rate

ASCT rate is the proportion of patients who completed therapy and proceeded to autologous stem cell transplant (ASCT) (NCT01661881)
Timeframe: All patients were followed for continuation to ASCT upon completion of induction therapy. Patients usually proceed to ASCT within 3 months of completing induction.

Interventionproportion of participants (Number)
RB/RC.91

Complete Remission (CR) Rate After 6 Cycles

The CR rate is defined as the proportion of patients who after 6 cycles of therapy achieve complete remission based on the International Working Group (IWG) Criteria (Cheson et al, 1999), using CT scans. CR or CRu (CR unconfirmed) by CT scans was defined by standard IWG criteria, ie resolution of all abnormal adenopathy and organomegaly, and clearance of marrow disease when present at baseline. (NCT01661881)
Timeframe: Disease was assessed after three- and six-cycles of therapy, up to approximately 25 weeks. All patients completed 6 cycles of therapy with a cycle duration of 28 days.

Interventionproportion of participants (Number)
RB/RC.96

Determination of the Safety and Tolerability of INCB053914 as Measured by the Number of Participants With Adverse Events

(NCT02587598)
Timeframe: Approximately 7 months

InterventionParticipants (Number)
Parts 1 and 2: INCB053914 100 mg QD4
Parts 1 and 2: INCB053914 50 mg BID11
Parts 1 and 2: INB053914 65 mg BID4
Parts 1 and 2: INB053914 80 mg BID21
Parts 1 and 2: INB053914 100 mg BID12
Parts 1 and 2: INB053914 115 mg BID6
Parts 3 and 4: INCB053914 50 mg BID + Cytarabine6
Parts 3 and 4: INCB053914 50 mg BID + Azacitidine7
Parts 3 and 4: INCB053914 80 mg BID + Azacitine9
Parts 3 & 4: INCB 053914 50 mg BID + Ruxolitinib3
Parts 3 & 4: INCB 053914 80 mg + Ruxolitinib14

Evaluation of Phosphorylated BCL--2 Associated Death Promoter Protein (pBAD)

Percent Inhibition of pBAD at the C1D15 trough from the pBAD at pre-dose by ex vivo cellular assay (NCT02587598)
Timeframe: 1 month

InterventionPercentage of Inhibition (Mean)
Parts 1 and 2: INCB053914 100 mg QD41
Parts 1 and 2: INCB053914 50 mg BID37
Parts 1 and 2: INB053914 65 mg BID68
Parts 1 and 2: INB053914 80 mg BID78
Parts 1 and 2: INB053914 100 mg BID55
Parts 1 and 2: INB053914 115 mg BID58

Pharmacokinetics: AUCtau of Combination Group B INCB053914 80 mg + Azatcitidine

(NCT02587598)
Timeframe: Cycle 1 Day 8

InterventionnM*h (Mean)
Parts 3 & 4: INCB053914 80 mg BID + Azacitidine11000

Pharmacokinetics: AUCtau of Combination Treatment Group A INCB053914 50 mg + Cytarabine

(NCT02587598)
Timeframe: Cycle 1 Day 5

InterventionnM*h (Mean)
Parts 3 & 4: INCB053914 50 mg BID + Cytarabine2860

Pharmacokinetics: AUCtau of Combination Treatment Group C INCB053914 80 mg + Ruxolitinib

(NCT02587598)
Timeframe: Regimen 2 Week 4

InterventionnM*h (Mean)
Parts 3 & 4: INB053914 80 mg BID + Ruxolitinib3060

Pharmacokinetics: AUCtau of INCB053914 Monotherapy

(NCT02587598)
Timeframe: Cycle 1 Day 8

InterventionnM*h (Mean)
Parts 1 and 2: INCB053914 100 mg QD4140
Parts 1 and 2: INCB053914 50 mg BID1290
Parts 1 and 2: INB053914 65 mg BID2480
Parts 1 and 2: INB053914 80 mg BID3940
Parts 1 and 2: INB053914 100 mg BID5410
Parts 1 and 2: INB053914 115 mg BID5630

Pharmacokinetics: Cl/F of Combination Group B INCB053914 80 mg + Azatcitidine

(NCT02587598)
Timeframe: Cycle 1 Day 8

InterventionL/h (Mean)
Parts 3 & 4: INCB053914 80 mg BID + Azacitidine30.8

Pharmacokinetics: Cl/F of Combination Treatment Group A INCB053914 50 mg + Cytarabine

(NCT02587598)
Timeframe: Cycle 1 Day 5

InterventionL/hr (Mean)
Parts 3 & 4: INCB053914 50 mg BID + Cytarabine122

Pharmacokinetics: Cl/F of Combination Treatment Group C INCB053914 80 mg + Ruxolitinib

(NCT02587598)
Timeframe: Regimen 2 Week 4

InterventionL/h (Mean)
Parts 3 & 4: INB053914 80 mg BID + Ruxolitinib69.7

Pharmacokinetics: CL/F of INCB053914 Monotherapy

(NCT02587598)
Timeframe: Cycle 1 Day 8

InterventionL/h (Mean)
Parts 1 and 2: INCB053914 100 mg QD47.2
Parts 1 and 2: INCB053914 50 mg BID132
Parts 1 and 2: INB053914 65 mg BID95.1
Parts 1 and 2: INB053914 80 mg BID71.2
Parts 1 and 2: INB053914 100 mg BID85.2
Parts 1 and 2: INB053914 115 mg BID39.8

Pharmacokinetics: Cmax of Combination Group B INCB053914 80 mg + Azatcitidine

(NCT02587598)
Timeframe: Cycle 1 Day 8

InterventionnM (Mean)
Parts 3 & 4: INCB053914 80 mg BID + Azacitidine1320

Pharmacokinetics: Cmax of Combination Treatment Group A INCB053914 50 mg + Cytarabine

(NCT02587598)
Timeframe: Cycle 1 Day 5

InterventionnM (Mean)
Parts 3 & 4: INCB053914 50 mg BID + Cytarabine423

Pharmacokinetics: Cmax of Combination Treatment Group C INCB053914 80 mg + Ruxolitinib

(NCT02587598)
Timeframe: Regimen 2 Week 4

InterventionnM (Mean)
Parts 3 & 4: INB053914 80 mg BID + Ruxolitinib541

Pharmacokinetics: Cmax of INCB053914 Monotherapy

(NCT02587598)
Timeframe: Cycle 1 Day 8

InterventionnM (Mean)
Parts 1 and 2: INCB053914 100 mg QD352
Parts 1 and 2: INCB053914 50 mg BID227
Parts 1 and 2: INB053914 65 mg BID333
Parts 1 and 2: INB053914 80 mg BID591
Parts 1 and 2: INB053914 100 mg BID796
Parts 1 and 2: INB053914 115 mg BID578

Pharmacokinetics: Cmin of Combination Group B INCB053914 80 mg + Azatcitidine

(NCT02587598)
Timeframe: Cycle 1 Day 8

InterventionnM (Mean)
Parts 3 & 4: INCB053914 80 mg BID + Azacitidine513

Pharmacokinetics: Cmin of Combination Treatment Group A INCB053914 50 mg + Cytarabine

(NCT02587598)
Timeframe: Cycle 1 Day 5

InterventionnM (Mean)
Parts 3 & 4: INCB053914 50 mg BID + Cytarabine139

Pharmacokinetics: Cmin of Combination Treatment Group C INCB053914 80 mg + Ruxolitinib

(NCT02587598)
Timeframe: Regimen 2 Week 4

InterventionnM (Mean)
Parts 3 & 4: INB053914 80 mg BID + Ruxolitinib104

Pharmacokinetics: Ctau of INCB053914 Monotherapy

(NCT02587598)
Timeframe: Cycle 1 Day 8

InterventionL/h (Mean)
Parts 1 and 2: INCB053914 100 mg QD98.2
Parts 1 and 2: INCB053914 50 mg BID65.7
Parts 1 and 2: INB053914 65 mg BID132
Parts 1 and 2: INB053914 80 mg BID213
Parts 1 and 2: INB053914 100 mg BID293
Parts 1 and 2: INB053914 115 mg BID410

Pharmacokinetics: Tmax of Combination Group B INCB053914 80 mg + Azatcitidine

(NCT02587598)
Timeframe: Cycle 1 Day 8

Interventionh (Median)
Parts 3 & 4: INCB053914 80 mg BID + Azacitidine2

Pharmacokinetics: Tmax of Combination Treatment Group A INCB053914 50 mg + Cytarabine

(NCT02587598)
Timeframe: Cycle 1 Day 5

Interventionh (Median)
Parts 3 & 4: INCB053914 50 mg BID + Cytarabine1.52

Pharmacokinetics: Tmax of Combination Treatment Group C INCB053914 80 mg + Ruxolitinib

(NCT02587598)
Timeframe: Regimen 2 Week 4

Interventionh (Median)
Parts 3 & 4: INB053914 80 mg BID + Ruxolitinib2.02

Pharmacokinetics: Tmax of INCB053914 Monotherapy

(NCT02587598)
Timeframe: Cycle 1 Day 8

Interventionhour (Median)
Parts 1 and 2: INCB053914 100 mg QD2.0
Parts 1 and 2: INCB053914 50 mg BID1.0
Parts 1 and 2: INB053914 65 mg BID2.0
Parts 1 and 2: INB053914 80 mg BID1.5
Parts 1 and 2: INB053914 100 mg BID1.0
Parts 1 and 2: INB053914 115 mg BIDNA

Maximum Tolerated Dose (MTD) of Bortezomib

The maximum tolerated dose (MTD) is defined to be the dose cohort below which 3 of 6 patients experience dose limiting toxicity (DLT), or the highest dose cohort of 1.5 mg/m², if 2 DLT were not observed at any dose cohort. (NCT00571493)
Timeframe: 14 months

Interventionmg/m² (Number)
Phase I1.5
Phase II1.0

Preliminary Estimate of Overall Response Rate (ORR)

To obtain a preliminary estimate of overall response rate (ORR). The overall response rate is calculated as the number of patients who achieved complete response (CR) and partial response (PR) divided by the total number of evaluable patients. (NCT00571493)
Timeframe: 100 day post autologous hematopoietic stem cell transplantation (ASCT), one year post ASCT

Interventionparticipants (Number)
Overall Response Rate (100 days after transplant)Overall Response Rate (1 year after transplant)
Phase II: Overall Response Rate3833

Progression-free Survival (PFS), and Overall Survival (OS)

To obtain a preliminary estimate of PFS and OS. Overall survival (OS) is defined as time from the first chemotherapy administered on the transplant trial until death from any cause. Progression free survival (PFS)is defined as time from therapy until relapse, progression, or death from any cause. (NCT00571493)
Timeframe: one year post autologous hematopoietic stem cell transplantation (ASCT) , 5 years post ASCT

Interventionpercentage of participants (Number)
Progression Free Survival 1 year after transplantOverall Survival 1 year after transplantProgression Free Survival 5 years after transplantOverall Survival 5 years after transplant
Phase II: Progression Free Survival and Overall Survival83913267

Event Free Survival (EFS)

The Kaplan-Meier method will be used to estimate the 2-year EFS for each of the treatment regimens. (NCT01979536)
Timeframe: Time from study entry until progressive disease, relapse, or death, assessed up to 2 years

InterventionPercentage of patients (Number)
Arm BV (Brentuximab Vedotin, Combination Chemotherapy)78.8
Arm CZ (Crizotinib, Combination Chemotherapy)76.8

Occurrence of Grade 3+ Non-hematologic Adverse Events

Will be assessed using the National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. Toxicities will be summarized by individual toxicity counts separated by arm. (NCT01979536)
Timeframe: Up to 60 months

InterventionPercentage of patients (Number)
Arm BV (Brentuximab Vedotin, Combination Chemotherapy)80.6
Arm CZ (Crizotinib, Combination Chemotherapy)87.9

Prognostic Significance of Minimal Residual Disease

Analyzed by estimating the 2-year EFS of negative MRD and positive MRD by arm. Minimal disease was performed using serial assessments of the t(2;5)(p23;q35) NPM-ALK fusion transcript using quantitative RT-PCR. Quantitative RT-PCR was performed by extracting total RNA from peripheral blood specimens. Peripheral blood samples were obtained at baseline, on day 1 of cycle 1, and on day 1 of cycle 2. The normalized copy numbers (NCN) were expressed as copy numbers of NPM-ALK per 104 copies of ABL. Minimal disease (MDD) was defined as >10 NCN at baseline. (NCT01979536)
Timeframe: Baseline up to progressive disease, relapse, or death, assessed up to 2 years

InterventionPercentage of patients (Number)
MRD Negative Arm BV (Brentuximab Vedotin, Combination Chemotherapy)89
MRD Positive Arm BV (Brentuximab Vedotin, Combination Chemotherapy)52.6
MRD Negative Arm CZ (Crizotinib, Combination Chemotherapy)85.6
MRD Positive Arm CZ (Crizotinib, Combination Chemotherapy)58.1

Overall Survival in Patients 65 Years or Older Who Have Newly Diagnosed de Novo or Secondary AML.

The interval from date of randomization to the date of death from any cause or the last date the subject was known to be alive or 5 years whichever occurs first. (NCT00260832)
Timeframe: The interval from date of randomization to the date of death from any cause or the last date the subject was known to be alive or 5 years whichever occurs first.

Interventionmonths (Median)
Cytarabine or Supportive Care5.0
Dacogen (Decitabine) Only7.7

Percentage of Participants With Complete Remission (CR) Plus Complete Remission With Incomplete Platelet Recovery (CRp)

Morphologic CR plus CRp rate where Morphologic leukemia-free state defined as less that (<) 5 percent (%) blasts in an aspirate sample with marrow spicules and a count of greater than or equal to (>=) 200 nucleated cells (there should have been no blasts with Auer rods or persistence of extramedullary disease) plus absolute neutrophil count (ANC) greater than (>)1,000 per microliter (/mcL), platelet count of >=100,000/mcL, and the participant must have been independent of transfusions for at least 1 week before each assessment. There was no duration requirement for confirmation of this designation and Morphologic CR without the requirement of platelet count >=100,000/mcL. (NCT00260832)
Timeframe: Post randomization when at least one post-baseline bone marrow assessment or peripheral blood count data available (up to 29.5 months)

Interventionpercentage of participants (Number)
Cytarabine or Supportive Care7.8
Dacogen (Decitabine) Only17.8

Event-Free Survival (EFS)

Event -Free Survival (EFS) EFS was calculated with Kaplan-Meier estimates. Event-free survival (EFS), defined as the time to no response to intensive induction therapy, relapse, or death of any cause, whichever comes first. (NCT00422591)
Timeframe: from treatment initiation until treatment failure, relapse, or death

InterventionMonths (Median)
Idarubicin + Cytarabine4.7

Overall Survival (OS)

Overall Survival (OS) was calculated with Kaplan-Meier estimates. OS was calculated from the time of treatment initiation until death. (NCT00422591)
Timeframe: Until death or loss of follow-up

InterventionMonths (Median)
Idarubicin + Cytarabine11.3

5 Year Disease-free Survival for Autologous & Allogeneic Transplant Groups

Percentage of patients who achieved a complete remission (CR) and were alive and relapse free at 5 years. The 5-year progression free survival was estimated using the Kaplan Meier method. (NCT00039377)
Timeframe: 5 years from CR

Interventionpercentage of patients (Number)
Patients With HLA-matched Sibling Donor46
Patients Without HLA-matched Sibling Donors47

5 Year Overall Survival for Autologous & Allogeneic Transplant Groups

Percentage of patients who were alive at 5 years. The 5-year progression free survival was estimated using the Kaplan Meier method. (NCT00039377)
Timeframe: 5 years from registration

Interventionpercentage of patients (Number)
Patients With HLA-matched Sibling Donor53
Patients Without HLA-matched Sibling Donors51

Disease Free Survival

"Disease-free survival (DFS) was measured as the interval from achievement of complete remission (CR) until relapse or death, regardless of cause; patients alive and in CR were censored at last follow-up. DFS was estimated using the Kaplan Meier method.~A complete remission (CR) was defined as recovery of morphologically normal bone marrow and blood counts (i.e., neutrophils >= 1.5 x 10^9/L and platelets > 100 x 10^9/L) and no circulating leukemic blasts or evidence of extramedullary leukemia and persisting for at least one month." (NCT00039377)
Timeframe: Duration of treatment (up to 10 years)

Interventionyears (Median)
Entire Cohort1.7

Overall Survival

Overall survival (OS) as the interval from the on-study date until death. OS was estimated using the Kaplan Meier method. (NCT00039377)
Timeframe: Duration of study (up to 10 years)

Interventionyears (Median)
Entire Cohort3.6

Number of Participants Who Achieved a BCR-ABL Response at 12 Months

"BCR-ABL response is defined in two ways: complete molecular response (CMR) and major molecular response (MMR).~Complete Molecular Response is defined as a Bcr-Abl (a fusion of gene of Bcr and ABl genes) ratio ≤0.0032% on the International Scale Bcr = breakpoint cluster gene Abl = abelson proto-oncogene~MMR is defined as Bcr-Abl (A fusion gene of the breakpoint cluster region [Bcr] gene and Abelson proto-oncogene [Abl] genes) transcript ratio ≤0.1% (≥ 3 log reduction of BCR-ABL transcripts from a standardized baseline), as detected by reverse transcriptase polymerase chain reaction [RT-PCR] (performed centrally)." (NCT00039377)
Timeframe: 12 months

Interventionparticipants (Number)
Complete Molecular ResponseMajor Molecular Response
Entire Cohort94

Objective Response

Determine the overall objective response. CR rate [as measured from the start of ICE, (or high dose CTX) to the end of transplant for those who receive it, or the end of ICE for those who do not].Complete response (CR): No evidence of Hodgkin's disease determined clinically, radiologically or pathologically when indicated (NCT00003631)
Timeframe: 2 years

,,
Interventionparticipants (Number)
FailureMinor Response (MR)Partial Response (PR)Progression Free (P-Free)Complete Response (CR)Progression of Disease (POD)
Group A - Favorable Prognostic Group3214100
Group B - Intermediate Prognostic Group6123121
Group C - Unfavorable Prognostic Group304710

EFS for Standard (SR) and Intermediate Risk (IR) T-ALL Patients on the Non-bortezomib Containing Arm on This Study (no Cranial Radiation Therapy [CRT]) and Similar Patients on AALL0434 (Received CRT)

EFS is calculated as time from randomization at study entry to first event (induction failure, induction death, relapse, second malignancy, remission death) or date of last contact. (NCT02112916)
Timeframe: 3 years

InterventionPercentage of participants (Number)
AALL1231 T-ALL Patients88.3
AALL0434 T-ALL Patients88.8

EFS for Very High Risk (VHR) T-ALL Patients Treated With High Risk (HR) Berlin-Frankfurt-Munster (BFM) Intensification Blocks Who Become Minimal Residual Disease (MRD) Negative and Those Who Remain MRD Positive at the End of HR Block 3

EFS will be calculated as time from the end of the three high-risk blocks of therapy to first event (relapse, second malignancy, remission death) or date of last contact. (NCT02112916)
Timeframe: 3 years

InterventionPercentage of participants (Number)
VHR T-ALL MRD Undetectable25.0
VHR T-ALL MRD Detectable88.9

EFS for Very High Risk (VHR) T-LLy Patients Treated With HR Berlin-Frankfurt-Munster (BFM) Intensification Blocks Who Have Complete or Partial Remission and Those Who do Not Respond

EFS for very high risk (VHR) T-LLy patients treated with HR Berlin-Frankfurt-Munster (BFM) intensification blocks who have complete or partial remission and those who do not respond (NCT02112916)
Timeframe: 3 years

InterventionPercentage of participants (Number)
VHR T-LLy (CR/PR)0

Event-free Survival (EFS) for Modified Augmented Berlin-Frankfurt-Munster Backbone With or Without Bortezomib in All Randomized Patients

EFS is calculated as time from randomization at study entry to first event (induction failure, induction death, relapse, second malignancy, remission death) or date of last contact. Three-year EFS rates will be calculated for both groups. (NCT02112916)
Timeframe: 3 years

InterventionPercentage of participants (Number)
Arm A (Combination Chemotherapy)81.7
Arm B (Combination Chemotherapy, Bortezomib)85.1

Toxicity Rates Associated With Modified Standard Therapy, Including Dexamethasone and Additional Pegaspargase

Percentage of patients who experienced Grade 3 or higher Toxicity Assessed by the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0 (NCT02112916)
Timeframe: 3 years from start of therapy by patient

InterventionPercentage of participants (Number)
Total Patients78.0

Cumulative Incidence Rates of Isolated Central Nervous System (CNS) Relapse for SR and IR T-ALL Patients on the Non-bortezomib Containing Arm on This Study (no CRT) and Similar Patients on AALL0434 (Receive CRT)

Cumulative incidence of isolated CNS relapse adjusting for competing risks using the method of: Gray R, A class of K-sample tests for comparing the cumulative incidence of a competing risk. Ann Stat 1141:1154, 1988 (NCT02112916)
Timeframe: 3 years

,
InterventionPercentage of participants (Number)
Isolated CNS RelapseIsolated Bone Marrow RelapseCombined Bone Marrow Relapse
AALL0434 T-ALL Patients2.23.01.8
AALL1231 T-ALL Patients3.61.41.3

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

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

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

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

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

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

Overall Survival (OS) of HR and IR Relapse Patients

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

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

Overall Survival (OS) of LR Relapse Patients

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

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

Disease-free Survival (DFS) for Randomized Methotrexate T-ALL Cohort (Arm I + Arm II vs. Arm III + Arm IV)

Disease Free Probability where DFS time is defined as time from randomization end of induction to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00408005)
Timeframe: 4 years from randomization at the end of induction

Interventionpercent probability (Number)
ARM I and ARM II (Combination Chemotherapy)91.45
ARM III and ARM IV (Combination Chemotherapy)85.78

Disease-free Survival (DFS) for Randomized Methotrexate T-ALL Cohort (Arm I vs. Arm II vs. Arm III vs. Arm IV)

Disease-free survival defined as time from randomization end of induction to first event (relapse, second malignant neoplasm, remission death) or date of last contact for those who are event-free. (NCT00408005)
Timeframe: 4 years from randomization at the end of induction

Interventionpercent probability (Number)
ARM I (Combination Chemotherapy)91.76
ARM II (Combination Chemotherapy)90.53
ARM III (Combination Chemotherapy)86.06
ARM IV (Combination Chemotherapy)84.89

Disease-free Survival (DFS) for Randomized Nelarabine T-ALL Cohort (Arm I + Arm III vs. Arm II + Arm IV)

Disease Free Probability where DFS time is defined as time from randomization end of induction to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event (NCT00408005)
Timeframe: 4 years from randomization at the end of induction

Interventionpercent probability (Number)
ARM I and ARM III (Combination Chemotherapy)82.96
ARM II and ARM IV (Combination Chemotherapy)88.30

Disease-free Survival (DFS) for Randomized Nelarabine T-ALL Cohort (Arm I vs. Arm II vs. Arm III vs. Arm IV)

Disease Free Probability where DFS time is defined as time from randomization end of induction to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00408005)
Timeframe: 4 years from randomization at the end of induction

Interventionpercent probability (Number)
ARM I (Combination Chemotherapy)89.01
ARM II (Combination Chemotherapy)90.53
ARM III (Combination Chemotherapy)78.07
ARM IV (Combination Chemotherapy)86.46

Cumulative Incidence of CNS Relapse for T-ALL by Risk Group

Cumulative incidence of CNS relapse adjusting for DFS events, was calculated using the method Gray et. al. High risk patients receive cranial radiation and low risk patients receive no cranial radiation. (NCT00408005)
Timeframe: 4 years from randomization at the end of induction

,
Interventionpercent probability (Number)
Intermediate RiskHigh Risk
ARM II (Combination Chemotherapy)1.080
ARM IV (Combination Chemotherapy)0.853.45

Cumulative Incidence of CNS Relapse for T-ALL by Risk Group

Cumulative incidence of CNS relapse adjusting for DFS events, was calculated using the method Gray et. al. High risk patients receive cranial radiation and low risk patients receive no cranial radiation. (NCT00408005)
Timeframe: 4 years from randomization at the end of induction

,
Interventionpercent probability (Number)
Low RiskIntermediate RiskHigh Risk
ARM I (Combination Chemotherapy)1.851.163.64
ARM III (Combination Chemotherapy)1.929.16.52

Disease-free Survival (DFS) for T-cell Lymphoblastic Lymphoma (T-LLy) Cohort

Disease Free Probability where DFS time is defined as time from randomization end of induction to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00408005)
Timeframe: 4 years from end of induction

Interventionpercent probability (Number)
High RiskInduction Failure
ARM II (Combination Chemotherapy)85.0100

Disease-free Survival (DFS) for T-cell Lymphoblastic Lymphoma (T-LLy) Cohort

Disease Free Probability where DFS time is defined as time from randomization end of induction to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00408005)
Timeframe: 4 years from end of induction

Interventionpercent probability (Number)
Standard RiskHigh Risk
ARM I (Combination Chemotherapy)87.485.1

Comparison of the Increase in Cure Rate of High Risk ALL Without Causing More Serious Side Effects Between Interventions

Event Free Probability. (NCT00075725)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Dexamethasone and Capizzi Methotrexate Patients < 10 Years83.2
Dexamethasone, High Dose Methotrexate (Non Randomly Assigned)81.6
Dexamethasone & Capizzi Methotrexate Patients => 10 Years Old69.1
Dexamethasone, High Dose Methotrexate (IM) < 10 Years91.2
Prednisone, Capizzi Methotrexate <10 Years82.1
Prednisone, Capezzi Methotrexate >= 10 Years73.5
Predisone and High Dose Methotrexate < 10 Yrs Old80.8
Prenisone and High Dose Methotrexate >=10 Years75.8
Dexamethasone, High Dose Methotrexate (IM) >= 10 Years77.0
Prenisone, Capezzi Methotrexate (Down's Syndrome)61.8
Dexamethasone, Capizzi Methotrexate Down Syndrome (Non Random)44.4

Correlation of Early Marrow Response Status With MRD Negative.

Bone marrow status is defined as: M1: < 5% lymphoblasts; M2: 5-25% lymphoblasts; M3: > 25% lymphoblasts. Bone marrow MRD status is defined as positive with >= 0.1 detectable leukemia cells, and negative with < 0.1 detectable leukemia cells. (NCT00075725)
Timeframe: Day 29

Interventionparticipants (Number)
Dexamethasone and Capizzi Methotrexate Patients < 10 Years182
Dexamethasone, High Dose Methotrexate (Non Randomly Assigned)72
Dexamethasone & Capizzi Methotrexate Patients => 10 Years Old198
Dexamethasone, High Dose Methotrexate (IM) < 10 Years188
Prednisone, Capizzi Methotrexate <10 Years195
Prednisone, Capezzi Methotrexate >= 10 Years471
Prednisone and High Dose Methotrexate < 10 Yrs Old190
Prednisone and High Dose Methotrexate >=10 Years479
Dexamethasone, High Dose Methotrexate (IM) >= 10 Years208
Prednisone, Capezzi Methotrexate (Down's Syndrome)25
Dexamethasone, Capizzi Methotrexate Down Syndrome (Non Random)3
Prednisone and High Dose Methotrexate (Non Randomly Assigned)18

Correlation of Early Marrow Response Status With MRD Positive.

Bone marrow status is defined as: M1: < 5% lymphoblasts; M2: 5-25% lymphoblasts; M3: > 25% lymphoblasts. Bone marrow MRD status is defined as positive with >= 0.1 detectable leukemia cells, and negative with < 0.1 detectable leukemia cells. (NCT00075725)
Timeframe: Day 29

Interventionparticipants (Number)
Dexamethasone and Capizzi Methotrexate Patients < 10 Years26
Dexamethasone, High Dose Methotrexate (Non Randomly Assigned)12
Dexamethasone & Capizzi Methotrexate Patients => 10 Years Old43
Dexamethasone, High Dose Methotrexate (IM) < 10 Years14
Prednisone, Capizzi Methotrexate <10 Years16
Prednisone, Capezzi Methotrexate >= 10 Years95
Prednisone and High Dose Methotrexate < 10 Yrs Old17
Prednisone and High Dose Methotrexate >=10 Years98
Dexamethasone, High Dose Methotrexate (IM) >= 10 Years39
Prednisone, Capezzi Methotrexate (Down's Syndrome)3
Dexamethasone, Capizzi Methotrexate Down Syndrome (Non Random)3
Prednisone and High Dose Methotrexate (Non Randomly Assigned)3

Correlation of Minimal Residual Disease (MRD) Negative With Event Free Survival (EFS).

Bone marrow MRD status is defined as negative with < 0.1 detectable leukemia cells. (NCT00075725)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Dexamethasone and Capizzi Methotrexate Patients < 10 Years86.4
Dexamethasone, High Dose Methotrexate (Non Randomly Assigned)93.6
Dexamethasone & Capizzi Methotrexate Patients => 10 Years Old80.5
Dexamethasone, High Dose Methotrexate (IM) < 10 Years93.1
Prednisone, Capizzi Methotrexate <10 Years86.5
Prednisone, Capezzi Methotrexate >= 10 Years83.4
Prednisone and High Dose Methotrexate < 10 Yrs Old84.2
Prednisone and High Dose Methotrexate >=10 Years83.9
Dexamethasone, High Dose Methotrexate (IM) >= 10 Years85.3
Prednisone, Capezzi Methotrexate (Down's Syndrome)74.4
Dexamethasone, Capizzi Methotrexate Down Syndrome (Non Random)25

Correlation of Minimal Residual Disease (MRD) Negative With Overall Survival (OS).

Bone marrow MRD status is defined as negative with < .01 detectable leukemia cells. (NCT00075725)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Dexamethasone and Capizzi Methotrexate Patients < 10 Years95.4
Dexamethasone, High Dose Methotrexate (Non Randomly Assigned)92.9
Dexamethasone & Capizzi Methotrexate Patients => 10 Years Old87.4
Dexamethasone, High Dose Methotrexate (IM) < 10 Years98.1
Prednisone, Capizzi Methotrexate <10 Years93.3
Prednisone, Capizzi Methotrexate >= 10 Years90.2
Prednisone and High Dose Methotrexate < 10 Yrs Old94.5
Prednisone and High Dose Methotrexate >=10 Years90.5
Dexamethasone, High Dose Methotrexate (IM) >= 10 Years91.6
Prednisone, Capizzi Methotrexate (Down's Syndrome)78.3
Dexamethasone, Capizzi Methotrexate Down Syndrome (Non Random)25.0

Correlation of Minimal Residual Disease (MRD) Positive With Event Free Survival (EFS)

Bone marrow MRD status is defined as positive with >= 0.1 detectable leukemia cells. (NCT00075725)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Dexamethasone and Capizzi Methotrexate Patients < 10 Years66.5
Dexamethasone, High Dose Methotrexate (Non Randomly Assigned)43.3
Dexamethasone & Capizzi Methotrexate Patients => 10 Years Old35.4
Dexamethasone, High Dose Methotrexate (IM) < 10 Years80
Prednisone, Capizzi Methotrexate <10 Years34.7
Prednisone, Capizzi Methotrexate >= 10 Years39
Prednisone and High Dose Methotrexate < 10 Yrs Old55
Prednisone and High Dose Methotrexate >=10 Years47.8
Dexamethasone, High Dose Methotrexate (IM) >= 10 Years49.4

Correlation of Minimal Residual Disease (MRD) Positive With Overall Survival (OS)

Bone marrow MRD status is defined as positive with >= 0.1 detectable leukemia cells, and negative with < 0.1 detectable leukemia cells. (NCT00075725)
Timeframe: 5 Years

Interventionpercentage of participants (Number)
Dexamethasone and Capizzi Methotrexate Patients < 10 Years79.2
Dexamethasone, High Dose Methotrexate (Non Randomly Assigned)69.9
Dexamethasone & Capizzi Methotrexate Patients => 10 Years Old65.6
Dexamethasone, High Dose Methotrexate (IM) < 10 Years86.2
Prednisone, Capizzi Methotrexate <10 Years93.8
Prednisone, Capizzi Methotrexate >= 10 Years63.1
Predisone and High Dose Methotrexate < 10 Yrs Old84.2
Prednisone and High Dose Methotrexate >=10 Years73.6
Dexamethasone, High Dose Methotrexate (IM) >= 10 Years74.6

Disease Response Assessed by Modified RECIST Criteria

Number of participants with complete response and very good partial response at the end of protocol therapy. (NCT00025259)
Timeframe: Protocol therapy: the overall duration of which is: (n=1527) an average of 137.1 days, median 133.0 days, interquartile range: 101.0, 164.0 days.

InterventionNumber of participants (Number)
Arm I (Patients Off-therapy Before Callback-Induction Only)5
Arm II (RER With CR [ABVE-PC, IFRT])370
Arm III (RER With CR [ABVE-PC])380
Arm IV (RER With Less Than CR [ABVE-PC, IFRT])538
Arm V (RER With PD)29
Arm VI (SER [DECA, ABVE-PC, IFRT])105
Arm VII (SER [ABVE-PC, IFRT])100

Event-free Survival

Probability of event-Free survival which is defined as the time from study entry to treatment failure (disease progression, disease recurrence, biopsy positive residual after completion of all protocol therapy), occurrence of a second malignant neoplasm, or death from any cause. Patients without report of such events where censored at last contact. (NCT00025259)
Timeframe: 5 years

InterventionProbability of survival (Number)
Arm I (Patients Off-therapy Before Callback-Induction Only)0.89
Arm II (RER With CR [ABVE-PC, IFRT])0.87
Arm III (RER With CR [ABVE-PC])0.84
Arm IV (RER With Less Than CR [ABVE-PC, IFRT])0.87
Arm V (RER With PD)0.70
Arm VI (SER [DECA, ABVE-PC, IFRT])0.79
Arm VII (SER [ABVE-PC, IFRT])0.74

Grade 3 or 4 Non-hematologic Toxicity

Occurrence of any grade 4 non-hematologic toxicity or grade 3 non-hematologic toxicity which doesn't respond to treatment within 7 days despite recommended therapy modification, or toxic death, which is any death primarily attributable to treatment. Grade 3 is defined to be severe or medically significant but not immediate life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self-care ADL. Grade 4 refers to toxicities with life-threatening consequences; urgent intervention indicated. (NCT00025259)
Timeframe: Protocol therapy: the overall duration of which is: (n=1684) an average of 137.3 days, median 133.0 days, interquartile range: 101.0, 164.0 days.

InterventionNumber of participants (Number)
Arm I (Patients Off-therapy Before Callback-Induction Only)10
Arm II (RER With CR [ABVE-PC, IFRT])153
Arm III (RER With CR [ABVE-PC])130
Arm IV (RER With Less Than CR [ABVE-PC, IFRT])216
Arm V (RER With PD)11
Arm VI (SER [DECA, ABVE-PC, IFRT])62
Arm VII (SER [ABVE-PC, IFRT])45

Overall Survival

Probability of overall survival which is defined as the time from study entry to death from any cause. Patients alive where censored at last contact. (NCT00025259)
Timeframe: 5 years

InterventionProbability of survival (Number)
Arm I (Patients Off-therapy Before Callback-Induction Only)0.93
Arm II (RER With CR [ABVE-PC, IFRT])0.98
Arm III (RER With CR [ABVE-PC])0.98
Arm IV (RER With Less Than CR [ABVE-PC, IFRT])0.98
Arm VI (SER [DECA, ABVE-PC, IFRT])0.96
Arm VII (SER [ABVE-PC, IFRT])0.93

Complete Response Rate

To evaluate the complete (CR) response rate with relapsed follicular NHL treated with ESHAP chemotherapy for cytoreduction (2 cycles) followed by Ibritumomab tiuxetan (Zevalin) radioimmunotherapy (NCT00732498)
Timeframe: 5 years

InterventionParticipants (Count of Participants)
ESHAP Followed by Zevalin and Rituximab10

Median Time to Progression

To evaluate the median TTP of patients with relapsed follicular NHL treated with ESHAP chemotherapy for cytoreduction (2 cycles) followed by Ibritumomab tiuxetan (Zevalin) radioimmunotherapy. (NCT00732498)
Timeframe: 5 years

Interventionmonths (Median)
ESHAP Followed by Zevalin and Rituximab10

Overall Response Rate

To evaluate the overall (ORR) response rate with relapsed follicular NHL treated with ESHAP chemotherapy for cytoreduction (2 cycles) followed by Ibritumomab tiuxetan (Zevalin) radioimmunotherapy. Descriptive and summary statistics for demographic and clinical variables obtained. The incidences of reported adverse events (AEs) tabulated. Kaplan-Meier survival analysis for PFS and OS performed on TPP. All the analyses were performed using Stata [12]. (NCT00732498)
Timeframe: 5 years

Interventionpercentage of participants (Number)
ESHAP Followed by Zevalin and Rituximab77.3

Progression-free Survival at 1 Year

To evaluate the 1-year progression-free survival (PFS) of patients with relapsed follicular non-Hodgkin's lymphoma (NHL) treated with ESHAP chemotherapy for cytoreduction (2 cycles) followed by Ibritumomab tiuxetan (Zevalin) radioimmunotherapy. (NCT00732498)
Timeframe: 1 year

Interventionpercentage of participants (Number)
ESHAP Followed by Zevalin and Rituximab38

Complete Response or Complete Response With Incomplete Count Recovery

Complete response or complete response with incomplete count recovery, of selinexor in combination with fludarabine and cytarabine for patients with relapsed or refractory AML in the phase II portion of the study (NCT03071276)
Timeframe: Between days 28 and 35 of cycle 1 (cycle length is 42-56 days)

Interventionpercentage of participants (Number)
Interventions: Selinexor, Fludarabine, and Cytarabine50.0

Complete Response

Complete response, of selinexor in combination with fludarabine and cytarabine for patients with relapsed or refractory AML in the phase II portion of the study (NCT03071276)
Timeframe: Between days 28 and 35 of cycle 1 (cycle length is 42-56 days)

Interventionpercentage of participants (Number)
Interventions: Selinexor, Fludarabine, and Cytarabine46.4

The Overall Response (OR) Rate (Complete Response + Incomplete Count Recovery + Partial Response)

Complete response or complete response with incomplete count recovery or partial response, of selinexor in combination with fludarabine and cytarabine for patients with relapsed or refractory AML in the phase II portion of the study (NCT03071276)
Timeframe: Between days 28 and 35 of cycle 1 (cycle length is 42-56 days)

Interventionpercentage of participants (Number)
Interventions: Selinexor, Fludarabine, and Cytarabine53.6

Duration of Objective Response Rate (ORR) [Part 1]

"Duration of ORR is the time from first documentation of MLFS, CR, CRc, CRm, PR, CRi or PRi to date of first documentation of disease progression or death due to any cause.~MLFS: BM blasts <5%; absence of blasts with Auer rods and EMD. CR: MLFS criteria; ANC>1000/ul and platelet >100,000/ul; independence from red cell transfusions.~CRc: Reversion to a normal karyotype at the time of CR in cases with an abnormal karyotype at the time of diagnosis; based on the evaluation of 20 metaphase cells from BM.~CRm: Reversion to a molecular-negative phenotype at the time of CR. CRi: All CR criteria except for ANC <1000/ul or platelet <100,000/ul. PR: ANC >1000/ul and platelet >100,000/ul; BM blasts decreased to 5-25% and ≥50% decrease from pre-treatment levels.~PRi: ANC <1000/ul or platelet <100,000/ul; BM blasts decreased to 5-25% and ≥50% decrease from pre-treatment levels.~Disease progression/relapse: BM blast ≥5%; or reappearance of blast in the blood; or development of EMD." (NCT02954653)
Timeframe: 16 weeks

Interventionmonths (Median)
Part 1: PF-06747143 0.3 mg/kgNA
Part 1: PF-06747143 1 mg/kgNA

Number of Participants With Dose-Limiting Toxicities (DLTs) [Part 1]

DLTs were classified according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) version 4.03 and were defined as any of a predefined set of unacceptable hematologic and non-hematologic adverse events (AEs) occurring in the first treatment cycle unless clearly determined unrelated to PF-06747143. In addition, clinically important or persistent toxicities that were not included in the pre-specified criteria could be considered a DLT following review by the investigators and sponsor. (NCT02954653)
Timeframe: Day 1 to Day 28 of Cycle 1

InterventionParticipants (Count of Participants)
Part 1: PF-06747143 0.3 mg/kg0
Part 1: PF-06747143 1 mg/kg1

Objective Response Rate (ORR) - Percentage of Participants With Objective Response [Part 1]

"Objective Response was defined as morphologic leukemia-free state (MLFS), complete remission (CR), cytogenetic CR (CRc), molecular CR (CRm), partial remission (PR), or CR or PR with incomplete blood count recovery (CRi or PRi).~MLFS: Bone marrow (BM) blasts <5%; absence of blasts with Auer rods and extramedullary disease (EMD).~CR: MLFS criteria; absolute neutrophil count (ANC)>1000/ul and platelet >100,000/ul; independence from red cell transfusions.~CRc: Reversion to a normal karyotype at the time of CR in cases with an abnormal karyotype at the time of diagnosis; based on the evaluation of 20 metaphase cells from BM.~CRm: Reversion to a molecular-negative phenotype at the time of CR. CRi: All CR criteria except for ANC <1000/ul or platelet <100,000/ul. PR: ANC >1000/ul and platelet >100,000/ul; BM blasts decreased to 5-25% and ≥50% decrease from pre-treatment levels.~PRi: ANC <1000/ul or platelet <100,000/ul; BM blasts decreased to 5-25% and ≥50% decrease from pre-treatment levels (NCT02954653)
Timeframe: 16 weeks

Interventionpercentage of participants (Number)
Part 1: PF-06747143 0.3 mg/kgNA
Part 1: PF-06747143 1 mg/kgNA

Progression Free Survival [Part 1]

Progression/relapse free survival is the time from the start of study treatment to first documentation of disease progression or to death due to any cause, whichever occurrs first. Disease progression/relapse: Bone marrow blast ≥ 5%; or reappearance of blast in the blood; or development of extramedullary disease (EMD). (NCT02954653)
Timeframe: 16 weeks

Interventionmonths (Median)
Part 1: PF-06747143 0.3 mg/kgNA
Part 1: PF-06747143 1 mg/kgNA

Incidence of Anti-Drug Antibodies (ADA) Against PF-06747143 [Part 1]

Samples were tested for ADA using a validated assay. Number of participants with positive ADA samples was determined. (NCT02954653)
Timeframe: Cycle 1 Day 1, Cycle 1 Day 15, Cycle 2 Day 1, end of treatment

,
InterventionParticipants (Count of Participants)
Cycle 1 Day 1Cycle 1 Day 15Cycle 2 Day 1End of treatment
Part 1: PF-06747143 0.3 mg/kg0000
Part 1: PF-06747143 1 mg/kg1000

Number of Participants With Treatment-Emergent Adverse Events (AEs) by Maximum National Cancer Institute Common Terminology Criteria for Adverse Event (NCI CTCAE) Grade [Part 1]

An AE was any untoward medical occurrence in a participant administered a product or medical device without regard to possibility of causal relationship. Treatment-emergent AEs were those with initial onset or increasing in severity after the first dose of study treatment. AEs were graded by the investigator according to NCI CTCAE version 4.03 (Grade 1: mild AE; Grade 2: moderate AE; Grade 3: severe AE; Grade 4: life-threatening consequences, urgent intervention indicated; Grade 5: death related to AE). AEs included non-serious AEs and SAEs. (NCT02954653)
Timeframe: 1 year

,
InterventionParticipants (Count of Participants)
Grade 1Grade 2Grade 3Grade 4Grade 5
Part 1: PF-06747143 0.3 mg/kg01011
Part 1: PF-06747143 1 mg/kg00031

Number of Participants With Treatment-Emergent Adverse Events (AEs) or Serious Adverse Events (SAEs) [Part 1]

An AE was any untoward medical occurrence in a participant administered a product or medical device without regard to possibility of causal relationship. An SAE was an AE resulting in any of the following outcomes or deemed significant for any other reason: death; life-threatening experience (immediate risk of dying); initial or prolonged inpatient hospitalization; persistent or significant disability/incapacity; congenital anomaly/birth defect. Treatment-emergent AEs were those with initial onset or increasing in severity after the first dose of study treatment. AEs included non-serious AEs and SAEs. Causality to study treatment was determined by the investigator. (NCT02954653)
Timeframe: 1 year

,
InterventionParticipants (Count of Participants)
AE (all causality)AE (treatment related)SAE (all causality)SAE (treatment related)
Part 1: PF-06747143 0.3 mg/kg3310
Part 1: PF-06747143 1 mg/kg4331

Number of Participants With Chemistry Laboratory Abnormalities by Type and Maximum National Cancer Institute Common Terminology Criteria for Adverse Event (NCI CTCAE) Grade [Part 1]

Chemistry laboratory abnormalities included alanine aminotransferase (ALT), alkaline phosphatase, aspartate aminotransferase (AST), bilirubin (total), creatinine, hypercalcemia, hyperglycemia, hyperkalemia, hypermagnesemia, hypernatremia, hypoalbuminemia, hypocalcemia, hypoglycemia, hypokalemia, hypomagnesemia, hyponatremia, and hypophosphatemia. Each laboratory parameter was graded per NCI CTCAE version 4.03. (NCT02954653)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
ALT72525522ALT72525523Alkaline phosphatase72525522Alkaline phosphatase72525523AST72525522AST72525523Bilirubin (total)72525522Bilirubin (total)72525523Creatinine72525522Creatinine72525523Hypercalcemia72525522Hypercalcemia72525523Hyperglycemia72525522Hyperglycemia72525523Hyperkalemia72525522Hyperkalemia72525523Hypermagnesemia72525522Hypermagnesemia72525523Hypernatremia72525522Hypernatremia72525523Hypoalbuminemia72525522Hypoalbuminemia72525523Hypocalcemia72525522Hypocalcemia72525523Hypoglycemia72525522Hypoglycemia72525523Hypokalemia72525522Hypokalemia72525523Hypomagnesemia72525522Hypomagnesemia72525523Hyponatremia72525522Hyponatremia72525523Hypophosphatemia72525522Hypophosphatemia72525523
Grade 0Grade 1Grade 2Grade 3Grade 4
Part 1: PF-06747143 1 mg/kg4
Part 1: PF-06747143 0.3 mg/kg0
Part 1: PF-06747143 0.3 mg/kg3
Part 1: PF-06747143 1 mg/kg1
Part 1: PF-06747143 1 mg/kg3
Part 1: PF-06747143 0.3 mg/kg2
Part 1: PF-06747143 0.3 mg/kg1
Part 1: PF-06747143 1 mg/kg2
Part 1: PF-06747143 1 mg/kg0

Number of Participants With Hematology Laboratory Abnormalities by Type and Maximum National Cancer Institute Common Terminology Criteria for Adverse Event (NCI CTCAE) Grade [Part 1]

Hematology laboratory abnormalities included anemia, hemoglobin increased, lymphocyte count increased, lymphopenia, neutrophil count decreased, platelet count decreased, and white blood cell (WBC) decreased. Each laboratory parameter was graded per NCI CTCAE version 4.03. (NCT02954653)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Anemia72525522Anemia72525523Hemoglobin increased72525523Hemoglobin increased72525522Lymphocyte count increased72525523Lymphocyte count increased72525522Lymphopenia72525523Lymphopenia72525522Neutrophil count decreased72525523Neutrophil count decreased72525522Platelet count decreased72525523Platelet count decreased72525522WBC decreased72525523WBC decreased72525522
Grade 2Grade 4Grade 0Grade 1Grade 3
Part 1: PF-06747143 1 mg/kg3
Part 1: PF-06747143 1 mg/kg0
Part 1: PF-06747143 0.3 mg/kg3
Part 1: PF-06747143 0.3 mg/kg0
Part 1: PF-06747143 1 mg/kg1
Part 1: PF-06747143 1 mg/kg2
Part 1: PF-06747143 1 mg/kg4
Part 1: PF-06747143 0.3 mg/kg1

Apparent Volume of Distribution of Volasertib at Steady State (VSS)

Apparent Volume of Distribution of volasertib at steady state (VSS) following Intravenous (i.v.) administration. (NCT00804856)
Timeframe: Cycle 1: -0:05 hour (h), 0:30h, 1:00h, 1:30h, 2h, 3h, 4h, 24h, 96h, 216h, 335:55h, 336:30h, 337h, 337:30h, 338h, 339h, 340h, 648h after first drug administration.

InterventionLiter (L) (Geometric Mean)
Phase I Schedule A. Volasertib 150 mg+LDAC10600
Phase I Schedule A. Volasertib 200 mg+LDAC8640
Phase I Schedule A. Volasertib 250 mg+LDAC7000
Phase I Schedule A. Volasertib 300 mg+LDAC6320
Phase I Schedule A. Volasertib 350 mg+LDAC5270
Phase I Schedule A. Volasertib 400 mg+LDAC4830
Phase I Schedule B. Volasertib 150 mg10300
Phase I Schedule B. Volasertib 200 mgNA
Phase I Schedule B. Volasertib 350 mg5800
Phase I Schedule B. Volasertib 400 mg7150
Phase I Schedule B. Volasertib 450 mg5740
Phase I Schedule B. Volasertib 500 mg6360
Phase I Schedule B. Volasertib 550 mg5680
Phase II Schedule A. Volasertib 350 mg+LDAC.6130

Dose Normalized Area Under the Concentration-Time Curve of Cytarabine in Plasma Over the Time Interval From 0 Extrapolated up to 4 Hours

"AUC0-4,norm: Area under the concentration-time curve of Cytarabine in plasma over the time interval from zero extrapolated to 4 hours. The dose normalisation was done by dividing by dose applied. Unit: nanogram*hour/milliliter/milligram: ((ng*h/mL)/mg).~Dose groups of Phase I were combined by dose normalizing as pre specified in the study protocol." (NCT00804856)
Timeframe: Cycle 1: -0:05 hour (h), 0:30h, 1:00h, 1:30h, 2h, 3h, 4h after first drug administration.

Intervention(ng*h/mL)/mg (Geometric Mean)
Dose Normalized Volasertib+LDAC(Phase I,Schedule A). 150 to 400 mg Volasertib.3.84
Dose Normalized Volasertib+LDAC (Schedule A). Phase II. 350 mg Volasertib.4.00
Dose Normalized LDAC (Schedule C). Phase II. LDAC Monotherapy.3.94

Dose Normalized Maximum Measured Concentration of Cytarabine in Plasma (Cmax, Norm)

Cmax, norm: Maximum measured concentration of Cytarabine in plasma. The dose normalisation was done by dividing by the dose applied. Unit: nanogram/milliliter/milligram: ((ng/mL)/mg). Dose groups of Phase I were combined by dose normalizing as pre specified in the study protocol. (NCT00804856)
Timeframe: Cycle 1: -0:05 hour (h), 0:30h, 1:00h, 1:30h, 2h, 3h, 4h, 24h, 96h, 216h, 335:55h, 336:30h, 337h, 337:30h, 338h, 339h, 340h, 648h after first drug administration.

Intervention(ng/mL)/mg (Geometric Mean)
Dose Normalized Volasertib+LDAC(Phase I,Schedule A). 150 to 400 mg Volasertib.2.92
Dose Normalized Volasertib+LDAC (Schedule A). Phase II. 350 mg Volasertib.2.83
Dose Normalized LDAC (Schedule C). Phase II. LDAC Monotherapy.2.36

Phase I: Maximum Tolerated Dose (MTD) of Volasertib in Combination With LDAC (Schedule A) and Volasertib Monotherapy (Schedule B)

"To determine the Maximum tolerated dose (MTD), dose escalation was conducted following the 3+3 design with de-escalation. The MTD was defined as the highest dose at which 6 patients had been treated and less than 2 patients experienced a Dose limiting toxicity (DLT) within the first cycle of treatment. The MTD was defined based on safety data from the first cycle only.~DLT is defined as drug related Common terminology criteria for adverse events (CTCAE) grade ≥ 3 nonhaematological toxicity (excluding: untreated nausea, untreated vomiting, CTCAE grade 3 untreated diarrhoea, CTCAE grade 3 febrile neutropenia and CTCAE grade 3 infection with grade 3 or 4 neutrophils).~In patients with complete remission with incomplete blood count recovery (CRi) or partial remission (PR), persistent CTCAE grade 4 neutropenia or thrombocytopenia until three weeks after the end of the treatment cycle will be regarded a DLT." (NCT00804856)
Timeframe: First Treatment cycle, up to 28 days.

Interventionmilligram (mg) (Number)
Phase I Schedule A. Volasertib+LDAC350
Phase I Schedule B. Volasertib450

Phase I: Number of Participants With Dose Limiting Toxicities (DLTs) in the First Cycle for the Determination of the Maximum Tolerated Dose (MTD)

A DLT was defined as a drug related CTCAE (Common Toxicity Criteria for Adverse Events) Grade ≥3 non-haematological toxicity (excluding untreated nausea, untreated vomiting, Grade 3 untreated diarrhea, Grade 3 febrile neutropenia, and Grade 3 infection with Grade 3 or 4 neutrophils). In patients with CRi (Complete Remission with Incomplete Blood Count Recovery) or PR (Partial Remission), persistent Grade 4 neutropenia or thrombocytopenia for 3 weeks after the end of the treatment cycle was regarded as a DLT unless the respective Grade 4 cytopenia was preexistent. In patients who required platelet substitution to maintain a Grade <4 before treatment, a Grade 4 thrombocytopenia after treatment did not constitute a DLT. (NCT00804856)
Timeframe: First Treatment cycle, up to 28 days.

InterventionParticipants (Count of Participants)
Phase I Schedule A. Volasertib 150 mg+LDAC0
Phase I Schedule A. Volasertib 200 mg+LDAC0
Phase I Schedule A. Volasertib 250 mg+LDAC0
Phase I Schedule A. Volasertib 300 mg+LDAC1
Phase I Schedule A. Volasertib 350 mg+LDAC1
Phase I Schedule A. Volasertib 400 mg+LDAC2
Total Phase I Combined. Schedule A.4
Phase I Schedule B. Volasertib 150 mg1
Phase I Schedule B. Volasertib 200 mg0
Phase I Schedule B. Volasertib 350 mg0
Phase I Schedule B. Volasertib 400 mg1
Phase I Schedule B. Volasertib 450 mg1
Phase I Schedule B. Volasertib 500 mg2
Phase I Schedule B. Volasertib 550 mg2
Total Phase I Combined. Schedule B.7

Phase I: Number of Patients With Objective Response: Complete Remission or Complete Remission With Incomplete Blood Count Recovery (CR+CRi)

"Phase I: Number of patients with Objective Response (Complete remission (CR) + Complete remission with incomplete blood count recovery (CRi)).~Complete remission (CR): morphologically leukaemia-free state (i.e. bone marrow with <5% blasts by morphologic criteria and no Auer rods, no evidence of extramedullary leukaemia) and absolute neutrophil count ≥1,000/microliter (μL) and platelets >100,000/μL.~Complete remission with incomplete blood count recovery (incomplete CR, CRi): all of the above criteria for CR were met except that neutrophils <1,000/μL or platelets <100,000/μL in the blood were not achieved." (NCT00804856)
Timeframe: The best overall response recorded during the time period from the start of the treatment until end of the treatment period, progression or death (whichever was earlier), up to 594 days.

InterventionParticipants (Count of Participants)
Phase I Schedule A. Volasertib 150 mg+LDAC0
Phase I Schedule A. Volasertib 200 mg+LDAC2
Phase I Schedule A. Volasertib 250 mg+LDAC2
Phase I Schedule A. Volasertib 300 mg+LDAC1
Phase I Schedule A. Volasertib 350 mg+LDAC1
Phase I Schedule A. Volasertib 400 mg+LDAC0
Total Phase I Combined. Schedule A.6
Phase I Schedule B. Volasertib 150 mg0
Phase I Schedule B. Volasertib 200 mg0
Phase I Schedule B. Volasertib 350 mg1
Phase I Schedule B. Volasertib 400 mg2
Phase I Schedule B. Volasertib 450 mg2
Phase I Schedule B. Volasertib 500 mg0
Phase I Schedule B. Volasertib 550 mg0
Total Phase I Combined. Schedule B.5

Phase II: Event Free Survival

"Event-free survival (EFS) [days] was the shortest duration of the following: (a) Date of assessment indicating PD on the response page of the eCRF (Electronic Case Report Form) - date of randomisation + 1 day (b) Date of assessment indicating clinical progressive disease (PD) on the disease or responses pages of the of eCRF- date of randomisation + 1 day (c) Date of assessment indicating PD on the patient status page of the eCRF - date of randomisation +1 day (for patients who had not been censored before this time point) (d) Death date - date of randomisation +1 day Patients not being assessed PD, clinical PD, or death during the trial were censored.~EFS was analysed with the Kaplan-Meier method for each of the treatment arms." (NCT00804856)
Timeframe: The patients that entered the trial, and measured from the date of randomization to the date of disease progression (treatment failure), relapse or death from any cause, whichever occurred first, up to 1000 days..

InterventionDays (Median)
Phase II Schedule C. LDAC69.0
Phase II Schedule A. Volasertib 350 mg+LDAC169.0

Phase II: Number of Patients With Objective Response (Complete Remission (CR) + Complete Remission With Incomplete Blood Count Recovery (CRi))

"Phase II: Number of patients with Objective Response (Complete remission (CR) + Complete remission with incomplete blood count recovery (CRi)).~Complete remission (CR): morphologically leukaemia-free state (i.e. bone marrow with <5% blasts by morphologic criteria and no Auer rods, no evidence of extramedullary leukaemia) and absolute neutrophil count ≥1,000/microliter (μL) and platelets >100,000/μL.~Complete remission with incomplete blood count recovery (incomplete CR, CRi): all of the above criteria for CR were met except that neutrophils <1,000/μL or platelets <100,000/μL in the blood were not achieved." (NCT00804856)
Timeframe: The best overall response recorded during the time period from the start of the treatment until end of the treatment period, progression or death (whichever was earlier), up to 869 days.

InterventionParticipants (Count of Participants)
Phase II Schedule C. LDAC6
Phase II Schedule A. Volasertib 350 mg+LDAC13

Phase II: Overall Survival

"Overall survival [days] = (date of death - date of randomisation + 1 day), for patients with known date of death. Overall survival (censored) [days] = (date of last trial visit or follow-up - date of randomisation + 1 day), for patients who were still alive at time of database lock.~Overall survival (OS) was analysed with the Kaplan-Meier method for each of the treatment arms." (NCT00804856)
Timeframe: The patients that entered the trial, and measured from the date of randomisation until death from any cause, up to 1100 days..

InterventionDays (Median)
Phase II Schedule C. LDAC158.0
Phase II Schedule A. Volasertib 350 mg+LDAC245.0

Phase II: Relapse - Free Survival

"Relapse-free survival [days] = (date of first recurrence of disease or death after entering the trial - date of first occurrence of CR or CRi after entering the trial+ 1 day) for patients with a recurrence (this value should be positive).~Relapse-free survival (censored) [days] = (date of last trial visit or follow-up - date of first occurrence of CR or CRi after entering the trial + 1 day) for patients who did not experience recurrence of disease or death at the time of analysis." (NCT00804856)
Timeframe: The patients who achieved CR or CRi, and was measured from the date of attaining CR or CRi until the date of disease recurrence or death from any cause, whichever occurred first, up to 900 days.

InterventionDays (Median)
Phase II Schedule C. LDAC304.0
Phase II Schedule A. Volasertib 350 mg+LDAC563.0

Phase II: Remission Duration

Remission duration analysis was defined only for patients who achieved Complete remission (CR) or Complete remission with incomplete blood count recovery (CRi), and was measured from the date of attaining CR or CRi until the date of disease recurrence (relapse). For patients who died without report of relapse, remission duration was censored on the date of death, regardless of cause. (NCT00804856)
Timeframe: The patients who achieved CR or CRi, and was measured from the date of attaining CR or CRi until the date of disease recurrence (relapse), up to 900 days.

InterventionDays (Median)
Phase II Schedule C. LDAC367.0
Phase II Schedule A. Volasertib 350 mg+LDAC687.0

Phase II: Time to Remission

Time to remission [days] = (date of first occurrence of CR or CRi after entering the trial-date of randomisation + 1 day) for patients with an objective response. (NCT00804856)
Timeframe: The patients who achieved CR or CRi, and was measured from the date of attaining CR or CRi until the date of disease recurrence (relapse), up to 158 days.

InterventionDays (Median)
Phase II Schedule C. LDAC63.5
Phase II Schedule A. Volasertib 350 mg+LDAC71.0

Total Clearance (CL) of Volasertib in Plasma After i.v (Intravenous) Administration of Volasertib

Total Clearance (CL) of Volasertib in Plasma after Intravenous (i.v.) Administration of Volasertib. (NCT00804856)
Timeframe: Cycle 1: -0:05 hour (h), 0:30h, 1:00h, 1:30h, 2h, 3h, 4h, 24h, 96h, 216h, 335:55h, 336:30h, 337h, 337:30h, 338h, 339h, 340h, 648h after first drug administration.

Interventionmillilitre/minute (mL/min) (Geometric Mean)
Phase I Schedule A. Volasertib 150 mg+LDAC1280
Phase I Schedule A. Volasertib 200 mg+LDAC972
Phase I Schedule A. Volasertib 250 mg+LDAC864
Phase I Schedule A. Volasertib 300 mg+LDAC1150
Phase I Schedule A. Volasertib 350 mg+LDAC1000
Phase I Schedule A. Volasertib 400 mg+LDAC852
Phase I Schedule B. Volasertib 150 mg1330
Phase I Schedule B. Volasertib 200 mgNA
Phase I Schedule B. Volasertib 350 mg810
Phase I Schedule B. Volasertib 400 mg1120
Phase I Schedule B. Volasertib 450 mg920
Phase I Schedule B. Volasertib 500 mg1140
Phase I Schedule B. Volasertib 550 mg939
Phase II Schedule A. Volasertib 350 mg+LDAC.897

Absolute QTcF (QT Interval Corrected for Heart Rate Using Fridericia's Formula) Intervals

ECG (Electro Cardio Gram) Measurements: Absolute QTcF (QT Interval (interval from the beginning of the QRS complex to the end of the T wave) Corrected for Heart Rate Using Fridericia's Formula) Intervals. The 350 mg + LDAC arm of phase I and II were combined into one arm to provide maximum information on QT changes and presented together with the Phase I schedule B 450 mg arm, as pre specified in the study protocol. (NCT00804856)
Timeframe: Baseline (Days -14 to -1) and day 1 (1 hour and 24 hours) after start of infusion.

,
Interventionmilliseconds (ms) (Mean)
Individual baseline1 hour after start of infusion24 hour after start of infusion
Phase I Schedule B. Volasertib 450 mg412.4441.1411.9
Phase I+II Volasertib 350 mg+LDAC411.6430.0414.0

Best Eastern Co-operative Oncology Group (ECOG) Performance Score From Baseline Until End of Treatment

ECOG performance score change from baseline to last visit of last cycle = ECOG performance score at last visit of the last cycle - ECOG performance score at baseline. ECOG performance score changes from baseline were also categorised on a 3-point categorical scale: deteriorated (-1), unchanged (0), and improved (1). The number of participants for category of ECOG score change is reported. (NCT00804856)
Timeframe: Baseline and End of Treatment (up to 869 days).

,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
UnchangedImprovedDeteriorated
Phase I Schedule A. Volasertib 150 mg+LDAC301
Phase I Schedule A. Volasertib 200 mg+LDAC120
Phase I Schedule A. Volasertib 250 mg+LDAC500
Phase I Schedule A. Volasertib 300 mg+LDAC212
Phase I Schedule A. Volasertib 350 mg+LDAC503
Phase I Schedule A. Volasertib 400 mg+LDAC101
Phase I Schedule B. Volasertib 150 mg901
Phase I Schedule B. Volasertib 200 mg101
Phase I Schedule B. Volasertib 350 mg014
Phase I Schedule B. Volasertib 400 mg411
Phase I Schedule B. Volasertib 450 mg1435
Phase I Schedule B. Volasertib 500 mg212
Phase I Schedule B. Volasertib 550 mg300
Phase II Schedule A. Volasertib 350 mg+LDAC151311
Phase II Schedule C. LDAC22812
Total Phase I Combined. Schedule A.1737
Total Phase I Comined. Schedule B.33614
Total Phase II. Schedule A and C.372123

Best Overall Response

"CR~CR+CRi~Partial remission: CR except bone marrow (BM) contained ≥5% but <25% blasts (or ≤50% initial blasts), or <5% blasts in presence of Auer rods or abnormal morphology.~No change: survived ≥7 days (d) after 1st cycle with persistent leukemia in last peripheral blood smear or BM, or with persistent extramedullary disease, without further deterioration due to leukemia or increase of blasts in BM or peripheral blood.~Aplasia: survived ≥7d after 1st cycle, died whilst cytopenic, with last post-treatment BM result aplastic or hypoplastic and without leukemia blasts.~Indeterminate: survived <7 d after 1st cycle or survived ≥7d after 1st cycle, died with no persistent leukemia in peripheral smear but no post-treatment BM examination or did not complete 1st cycle.~Progressive disease: survived ≥7d after 1st cycle with increase of blast population in BM or peripheral blood or aggravation or new extramedullary disease or further deterioration or death due to leukemia." (NCT00804856)
Timeframe: The best overall response recorded during the time period from the start of the treatment until end of the treatment period, progression or death (whichever was earlier), up to 869 days.

,,,,,,,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Complete remissionCRiPartial remissionNo changeAplasiaIndeterminateProgressive diseaseNot evaluableMissing
Phase I Schedule A. Volasertib 150 mg+LDAC000200200
Phase I Schedule A. Volasertib 200 mg+LDAC020100000
Phase I Schedule A. Volasertib 250 mg+LDAC200200100
Phase I Schedule A. Volasertib 300 mg+LDAC010102311
Phase I Schedule A. Volasertib 350 mg+LDAC010103300
Phase I Schedule A. Volasertib 400 mg+LDAC000101100
Phase I Schedule B. Volasertib 150 mg000401600
Phase I Schedule B. Volasertib 200 mg000000200
Phase I Schedule B. Volasertib 350 mg011200100
Phase I Schedule B. Volasertib 400 mg020210100
Phase I Schedule B. Volasertib 450 mg022730720
Phase I Schedule B. Volasertib 500 mg000310100
Phase I Schedule B. Volasertib 550 mg001010110
Phase II Schedule A. Volasertib 350 mg+LDAC6721505700
Phase II Schedule C. LDAC33218031420
Total Phase I Combined. Schedule A.2408061011
Total Phase I Combined. Schedule B.05418611930
Total Phase II Combined. Schedule A and C.910433082120

Phase I Schedule A: Number of Participants With Adverse Events (AEs) Graded According to Common Terminology Criteria for Adverse Events (CTCAE), Based on the Number of Patients With AEs With CTCAE Grade ≥3 During All Cycles

Number of patients with AEs following in the categories 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related AE) of CTCAE is reported. (NCT00804856)
Timeframe: From first drug administration until 21 days after the last trial drug administration in the last treatment cycle, up to 615 days.

,,,,,,
InterventionParticipants (Count of Participants)
Grade 3Grade 4Grade 5
Phase I Schedule A. Volasertib 150 mg+LDAC111
Phase I Schedule A. Volasertib 200 mg+LDAC030
Phase I Schedule A. Volasertib 250 mg+LDAC050
Phase I Schedule A. Volasertib 300 mg+LDAC153
Phase I Schedule A. Volasertib 350 mg+LDAC232
Phase I Schedule A. Volasertib 400 mg+LDAC111
Total Phase I Combined. Schedule A.5187

Phase I Schedule A: Number of Participants With Adverse Events (AEs) Graded According to Common Terminology Criteria for Adverse Events (CTCAE), Based on the Number of Patients With AEs With CTCAE Grade ≥3 During Cycle 1

Number of patients with AEs following in the categories 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related AE) of CTCAE is reported. (NCT00804856)
Timeframe: First treatment cycle, up to 28 days.

,,,,,,
InterventionParticipants (Count of Participants)
Grade 3Grade 4Grade 5
Phase I Schedule A. Volasertib 150 mg+LDAC020
Phase I Schedule A. Volasertib 200 mg+LDAC030
Phase I Schedule A. Volasertib 250 mg+LDAC130
Phase I Schedule A. Volasertib 300 mg+LDAC153
Phase I Schedule A. Volasertib 350 mg+LDAC232
Phase I Schedule A. Volasertib 400 mg+LDAC111
Total Phase I Combined. Schedule A.5176

Phase I Schedule B: Number of Participants With Adverse Events (AEs) Graded According to Common Terminology Criteria for Adverse Events (CTCAE), Based on the Number of Patients With AEs With CTCAE Grade ≥3 During All Cycles

Number of patients with AEs following in the categories 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related AE) of CTCAE is reported. (NCT00804856)
Timeframe: From first drug administration until 21 days after the last trial drug administration in the last treatment cycle, up to 597 days.

,,,,,,,
InterventionParticipants (Count of Participants)
Grade 3Grade 4Grade 5
Phase I Schedule B. Volasertib 150 mg542
Phase I Schedule B. Volasertib 200 mg011
Phase I Schedule B. Volasertib 350 mg130
Phase I Schedule B. Volasertib 400 mg050
Phase I Schedule B. Volasertib 450 mg3153
Phase I Schedule B. Volasertib 500 mg012
Phase I Schedule B. Volasertib 550 mg031
Total Phase I Schedule B.9329

Phase I Schedule B: Number of Participants With Adverse Events (AEs) Graded According to Common Terminology Criteria for Adverse Events (CTCAE), Based on the Number of Patients With AEs With CTCAE Grade ≥3 During Cycle 1

Number of patients with AEs following in the categories 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related AE) of CTCAE is reported. (NCT00804856)
Timeframe: First treatment cycle, up to 28 days.

,,,,,,,
InterventionParticipants (Count of Participants)
Grade 3Grade 4Grade 5
Phase I Schedule B. Volasertib 150 mg541
Phase I Schedule B. Volasertib 200 mg011
Phase I Schedule B. Volasertib 350 mg130
Phase I Schedule B. Volasertib 400 mg040
Phase I Schedule B. Volasertib 450 mg5142
Phase I Schedule B. Volasertib 500 mg011
Phase I Schedule B. Volasertib 550 mg031
Total Phase I Schedule B.11306

Phase II: Number of Participants With Adverse Events (AEs) Graded According to Common Terminology Criteria for Adverse Events (CTCAE), Based on the Number of Patients With AEs With CTCAE Grade ≥3 During All Cycles

Number of patients with AEs following in the in the categories 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related AE) of CTCAE is reported. (NCT00804856)
Timeframe: From first drug administration until 21 days after the last trial drug administration in the last treatment cycle, up to 890 days.

,
InterventionParticipants (Count of Participants)
Grade 3Grade 4Grade 5
Phase II Schedule A. Volasertib 350 mg+LDAC12208
Phase II Schedule C. LDAC13136

QTcF (QT Interval Corrected for Heart Rate Using Fridericia's Formula) Change From Baseline at Cycle 1

ECG Measurements: QTcF (QT Interval (interval from the beginning of the QRS complex to the end of the T wave) changes from baseline at each time point: The QTcF post baseline measurement obtained at time t minus baseline QTcF measurement. The 350 mg + LDAC arm of phase I and II were combined into one arm to provide maximum information on QT changes and presented together with the Phase I schedule B 450 mg arm, as pre specified in the study protocol. (NCT00804856)
Timeframe: Baseline (Days -14 to -1) and day 1 (1 hour and 24 hours) after start of infusion.

,
Interventionmillisecond (ms) (Mean)
Change from baseline after 1 hourChange from baseline after 24 hour
Phase I Schedule B. Volasertib 450 mg29.6-0.5
Phase I+II, Volasertib 350 mg+LDAC18.51.9

All-Cause Mortality

Outcome measured for total population. Not broken down by indication received. (NCT01294449)
Timeframe: 5 years

Interventionparticipants (Number)
MADIT-CRT ICD11
MADIT-CRT CRT-D12

Participants With a Complete Response

Complete Response (CR) was defined as: Neutrophil count ≥ 1.0 ×109/L, Platelet count ≥ 100 ×109/L, Bone marrow aspirate ≤5% blasts and No extramedullary leukemia. Response evaluation following Induction Therapy (Cycle 1) and every 2-3 cycles during Consolidation Therapy (Cycles 2 - 7) where Cycle is 4-6 weeks. (NCT01019317)
Timeframe: Minimally 6 weeks (Cycle 1) up to 1 year (7 cycles)

InterventionParticipants (Number)
Cytarabine + Fludarabine34

All Cause Mortality

Vosaroxin + cytarabine mortality versus placebo + cytarabine mortality (NCT01191801)
Timeframe: 30 Days

Interventionpercentage of Participants in the Group (Number)
Group A (Vosaroxin/Cytarabine)7.9
Group B (Placebo/Cytarabine)6.6

All Cause Mortality

Vosaroxin + cytarabine mortality versus placebo + cytarabine mortality (NCT01191801)
Timeframe: 60 Days

Interventionpercentage of Participants (Number)
Group A (Vosaroxin/Cytarabine)19.7
Group B (Placebo/Cytarabine)19.4

Complete Remission (CR) Rate Based on Modified International Working Group (IWG) Criteria.

Group A (Vosaroxin + cytarabine) patient CR as compared to Group B (placebo + cytarabine) patient CR. Complete remission (CR) is typically defined using IWG criteria as bone marrow blast count of less than 5% with adequate recovery of peripheral blood counts. (NCT01191801)
Timeframe: Up to 5 years or duration of study

Interventionpercentage of particpants (Number)
Group A (Vosaroxin/Cytarabine)30.1
Group B (Placebo/Cytarabine)16.3

Event Free Survival (EFS)

(NCT01191801)
Timeframe: Up to 5 years or duration of study

Interventionmonths (Median)
Group A (Vosaroxin/Cytarabine)1.9
Group B (Placebo/Cytarabine)1.3

Leukemia-Free Survival (LFS)

Durability of remission (CR) assessed by LFS (NCT01191801)
Timeframe: Up to 5 years or the duration of the study

InterventionMonths (Median)
Group A (Vosaroxin/Cytarabine)11
Group B (Placebo/Cytarabine)8.7

Overall Remission (OR) Rate Based on the IWG Response Criteria

"Group A patient OR compared to Group B patient OR~Overall Remission includes Complete Remission (CR), Complete Remission with incomplete platelet recovery (CRp), Complete Remission with incomplete blood count recovery (CRi), and Partial Remission (PR). Complete remission means bone marrow blast count of less than 5% with adequate recovery of peripheral blood counts as typically defined by the IWG. Both CRi and CRp refer complete remission but with incomplete blood count and platelet recovery, respectively. PR, or partial remission, refers to remission in which bone marrow contains blast counts between 5 and 25 percent." (NCT01191801)
Timeframe: Up to 5 years or the duration of the study

Interventionpercentage of participants (Number)
Group A (Vosaroxin/Cytarabine)37.9
Group B (Placebo/Cytarabine)18.9

Overall Survival

Vosaroxin + cytarabine patient survival versus placebo + cytarabine patient survival (NCT01191801)
Timeframe: Up to 5 years or duration of study

InterventionMonths (Median)
Group A (Vosaroxin/Cytarabine)7.5
Group B (Placebo/Cytarabine)6.1

Incidence of Acute Graft-versus-host Disease (GVHD)

(NCT00691015)
Timeframe: Within 100 days after donor peripheral blood stem cell transplantation (PBSCT) as assessed by Glucksberg criteria

Interventionpercentage of participants (Number)
All Participants44.7

Incidence of Chronic GVHD.

(NCT00691015)
Timeframe: Within 2 years after PBSCT

Interventionpercentage of participants (Number)
All Participants44.68

Incidence of Infections, Including Bacterial, Fungal, and Viral Infections (i.e., CMV and EBV Reactivation, Including Post-transplant Lymphoproliferative Disorders)

(NCT00691015)
Timeframe: Within 6 months after PBSCT

Interventionpercentage of participants (Number)
All Participants80.85

Karnofsky Performance Status Performance Status

"100 - Normal; no complaints; no evidence of disease. 90 - Able to carry on normal activity; minor signs or symptoms of disease. 80 - Normal activity with effort; some signs or symptoms of disease. 70 - Cares for self; unable to carry on normal activity or to do active work. 60 - Requires occasional assistance, but is able to care for most of their personal needs.~50 - Requires considerable assistance and frequent medical care. 40 - Disabled; requires special care and assistance. 30 - Severely disabled; hospital admission is indicated although death not imminent.~20 - Very sick; hospital admission necessary; active supportive treatment necessary.~10 - Moribund; fatal processes progressing rapidly. 0 - Dead" (NCT00691015)
Timeframe: At 90 days after PBSCT

Interventionunits on a scale (Median)
All Participants80

Overall Survival.

(NCT00691015)
Timeframe: At 2 years after PBSCT

Interventionpercentage of participants (Number)
All Participants57.4

Safety, as Defined by Serious Adverse Events and Adverse Events Related to Study Treatment.

(NCT00691015)
Timeframe: Within 6 months after PBSCT

Intervention% of participants with a reported SAE (Number)
All Participants93.62

Severity of Acute Graft-versus-host Disease (GVHD)

(NCT00691015)
Timeframe: Within 100 days after donor peripheral blood stem cell transplantation (PBSCT) as assessed by Glucksberg criteria

Intervention% of participants with severe aGVHD (Number)
All Participants33.3

Time to Engraftment (i.e., Absolute Neutrophil Recovery [ANC > 500/mm³] )

(NCT00691015)
Timeframe: post transplant, up to 4 weeks

InterventionDays (Median)
All Participants11

Complete Remission Rate: Percentage of Participants With Complete Remission (CR)

Complete Remission (CR) was defined as the presence of 5% or less blasts in the bone marrow, with a granulocyte count ≥1.0 × 10^9/L, a platelet count ≥100 × 10^9/L, and no extramedullary disease. Complete recovery except platelets (CRp) was defined as for CR, except for recovery of platelet count to <100 × 10^9/L. Partial remission (PR) was defined as a bone marrow with >5% and <25% blasts with a granulocyte count of ≥1.0 × 109/L and a platelet count of ≥100 × 10^9/L. Relapse was defined by recurrence of more than 5% lymphoblasts in the bone marrow aspirate or by the presence of extramedullary disease after achieving CR. (NCT00669877)
Timeframe: After two 21-day courses, response to treatment checked for Complete Remission (CR)

Interventionpercentage of participants (Number)
Hyper-CVAD85

Overall Response Rate: Percentage of Participants With Complete Remission (CR) or Partial Remission (PR)

Complete Remission (CR) was defined as the presence of 5% or less blasts in the bone marrow, with a granulocyte count ≥1.0 × 10^9/L, a platelet count ≥100 × 10^9/L, and no extramedullary disease. Complete recovery except platelets (CRp) was defined as for CR, except for recovery of platelet count to <100 × 10^9/L. Partial remission (PR) was defined as a bone marrow with >5% and <25% blasts with a granulocyte count of ≥1.0 × 109/L and a platelet count of ≥100 × 10^9/L. Relapse was defined by recurrence of more than 5% lymphoblasts in the bone marrow aspirate or by the presence of extramedullary disease after achieving CR. (NCT00669877)
Timeframe: After two 21-day courses, response to treatment checked for Complete Remission (CR)

Interventionpercentage of participants (Number)
Hyper-CVAD89

Days Platelets Count of < 100K/μL

The optimal biologic dose was defined as the dose of AMG 531 at which the greatest proportion of patients avoided grade 4 thrombocytopenia (Platelet nadir < 25K/μL) in the absence of platelet transfusion in the blinded study cycle. (NCT00299182)
Timeframe: Prior to start of treatment and then at least 2 times a week during treatment until end of cycle 2 (1 cycle = 21 days)

Interventiondays (Mean)
Arm A 1mcg/kg5.5
Arm A 3mcg/kg5.3
Arm A 10mcg/kg8.3
Arm B 1mcg/kg6.8
Arm B 3mcg/kg8
Arm B 10mcg/kg9.3
Placebo9.8

Platelet (PLT) Nadir

Blood counts were performed at least two times a week and when clinically indicated during the study and daily when PLT count is < 50K/μL until recovery (two consecutive counts showing upward trend). The optimal biologic dose was defined as the dose of AMG 531 at which the greatest proportion of patients avoided grade 4 thrombocytopenia (Platelet nadir < 25K/μL) in the absence of platelet transfusion in the blinded study cycle. (NCT00299182)
Timeframe: Prior to start of treatment and then at least 2 times a week during treatment until end of cycle 2 (1 cycle = 21 days)

InterventionK/μL (Mean)
Arm A 1mcg/kg20.3
ARM A 3mcg/kg26.3
Arm A 10mcg/kg24.8
Arm B 1mcg/kg22.5
Arm B 3mcg/kg18.3
Arm B 10mcg/kg8
Placebo11.3

Progression Free Survival (PFS) at 7 Months

Progression-free survival defined as time from date of randomization to first occurrence of having documented disease progression or death due to any cause, whichever comes first. Progression based on tumor assessments according to Response Evaluation Criteria in Solid Tumors (RECIST). Participants were followed from baseline to disease progression with PFS evaluation at 7 months. (NCT00656617)
Timeframe: PFS Evaluation at 7 months

Interventionpercentage of participants (Number)
Idarubicin + Ara-C + Vorinostat65

Participant Response

Number of participants with response assessed according RECIST: Complete Response (CR) defined as normalization of marrow (< 5% blasts) and of peripheral blood counts (neutrophil count > 1.109/L, platelet count > 100 x 109/L). Partial response (PR) defined as for CR in terms of peripheral counts but with reduction of marrow blasts by >50% compared to pretreatment values but above <5%. Complete Response without platelet recovery (CRp) = CR, but platelets <100 x 109/L. Progressive disease (PD) defined as increase of blasts to > 10% after an initial response. (NCT00656617)
Timeframe: Monitoring with each 4 week cycle, up to 18 cycles of treatment

Interventionparticipants (Number)
Complete Response (CR)Partial Response (PR)Complete Response without platelet recovery (CRp)Progressive Disease (PD)No response
Idarubicin + Ara-C + Vorinostat67011024

Progression Free Survival (Rate)

Progression free survival (PFS) for three years following therapy with Rituxan-HCVAD alternating with Rituximab with high -dose methotrexate/ara-C and standard R-CHOP in patients with newly diagnosed B-cell aggressive non-Hodgkin's lymphomas younger than 60 years old and with adjusted IPI 2 or higher adverse prognostic features. (NCT00290498)
Timeframe: 3 years post-therapy

InterventionParticipants (Count of Participants)
R-HCVAD/MA35
R-CHOP7

Response Rate R-HCVAD vs. R-CHOP

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: 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. (NCT00290498)
Timeframe: 3 years

,
InterventionParticipants (Count of Participants)
Complete RemissionInevaluableProgressive DiseasePartial RemissionComplete Remission Unconfirmed
R-CHOP71101
R-HCVAD402142

Percentage of Participants Who Achieve a Timely Engraftment

Timely engraftment is defined as a persistent an absolute neutrophil count (ANC) of at least 500 cells/mm3 and a platelet count of at least 20,000 cells/mm3 for at least 3 days (NCT02797470)
Timeframe: 1 month post-transplant

Interventionpercentage of participants who achieve a (Mean)
Treatment (Anti-HIV Gene Transduced CD34+ Cells): 1:0 Ratio60
Treatment (Anti-HIV Gene Transduced CD34+ Cells): 1:1 Ratio75
Treatment (Anti-HIV Gene Transduced CD34+ Cells): 5:1 Ratio100

Continuous Complete Remission at 1 Year

A patient has a continuous complete remission at 1 year if they achieve a CR and are alive 365 days after registering to the study. (NCT00109837)
Timeframe: After induction, after consolidation, every 3 months during maintenance, and every three months after off treatment for up to a year

Interventionparticipants (Number)
Treatment21

Toxicity

Number of patients with Grade 3-5 adverse events that are related to study drug by given type of adverse event (NCT00109837)
Timeframe: Patients were assessed for adverse events after the induction cycle

InterventionParticipants with a given type of AE (Number)
ALT, SGPT (serum glutamic pyruvic transaminase)AST, SGOT (serum glut oxaloacetic transaminase)Albumin, serum-low (hypoalbuminemia)Alkaline phosphataseAnorexiaAscites (non-malignant)Bilirubin (hyperbilirubinemia)Calcium, serum-low (hypocalcemia)CholecystitisCholesterol, serum-high (hypercholesterolemia)Coagulation-Other (Specify)Colitis, infectious (e.g., Clostridium difficile)ConstipationDIC (disseminated intravascular coagulation)Death not assoc with CTCAE term-Multi-organ failEdema: limbFatigue (asthenia, lethargy, malaise)Febrile neutropeniaFever (in the abs of neutropenia)FibrinogenGlucose, serum-high (hyperglycemia)Glucose, serum-low (hypoglycemia)HemoglobinHypertensionHypotensionHypoxiaIleus, GI (functional obstruction of bowel)Infec(doc clin or mibio) w/ Gr 3/4 neut-AnalInfec(doc clin or mibio) w/ Gr 3/4 neut-BladderInfec(doc clin or mibio) w/ Gr 3/4 neut-BloodInfec(doc clin or mibio) w/ Gr 3/4 neut-BronchusIInfec(doc clin or mibio) w/ Gr 3/4 neut-CatheterInfec(doc clin or mibio) w/ Gr 3/4 neut-Eye NOSInfec(doc clin or mibio) w/ Gr 3/4 neut-LungInfec(doc clin or mibio) w/ Gr 3/4 neut-NoseInfec(doc clin or mibio) w/ Gr 3/4 neut-PharynxInfec(doc clin or mibio) w/ Gr 3/4 neut-Ur tractInfec with nor ANC or Gr 1/2 neut-Lung (pneumonia)Infection-Other (Specify)Leukocytes (total WBC)LipaseLiver dysfunction/failure (clinical)LymphopeniaMagnesium, serum-high (hypermagnesemia)Mucositis/stomatitis (clinical exam) - Oral cavityMucositis/stomatitis (funct/symp) - Oral cavityMucositis/stomatitis (func/symp) - PharynxMuscle weak,gen spec area-Whole bodyNauseaNeuropathy: motorNeutrophils/granulocytes (ANC/AGC)Pain - Abdomen NOSPain - BonePain - NeckPancreatic endocrine: glucose intolerancePancreatitisPhosphate, serum-low (hypophosphatemia)PlateletsPotassium, serum-high (hyperkalemia)Potassium, serum-low (hypokalemia)Rash/desquamationRenal failureSodium, serum-low (hyponatremia)Thrombosis/thrombus/embolismThrombotic microangiopathyTriglyceride, serum-high (hypertriglyceridemia)Tumor lysis syndromeTyphlitis (cecal inflammation)Uric acid, serum-high (hyperuricemia)Vomiting
Induction171332216712111211318111161332321111111111122143121912112314711111144171261215111

Bortezomib Clearance

Median and range of bortezomib clearance during Induction II. (NCT01371981)
Timeframe: Day 8 of Induction II

InterventionLiters/hour/m^2 (Median)
Arm B8.42

Change in Ejection Fraction

The mean percentage change in ejection fraction from baseline to the end of Induction I will be determined for eligible patients enrolled on Arms A, B and C. (NCT01371981)
Timeframe: Up to 4 weeks

InterventionPercentage change (Mean)
Arm A-2.0272
Arm B-2.3453
Arm C (Cohort 1)-7.5000
Arm C (Cohort 2)-5.1997
Arm C (Cohort 3)-3.4624

Change in Shortening Fraction

Mean percentage change in shortening fraction from baseline to the end of Induction I will be determined for eligible patients enrolled on Arms A, B and C. (NCT01371981)
Timeframe: Up to 4 weeks

InterventionPercentage change (Mean)
Arm A-1.8445
Arm B-2.6298
Arm C (Cohort 1)-2.2333
Arm C (Cohort 2)-3.6700
Arm C (Cohort 3)-3.4246

EFS for Patients on Arm C, Cohort 1

The Kaplan-Meier method will be used to estimate 3-year EFS, defined as the time from study entry until induction failure, relapse, secondary malignancy, or death. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm C (Cohort 1)25.00

EFS for Patients on Arm C, Cohort 2

The Kaplan-Meier method will be used to estimate 3-year EFS, defined as the time from study entry until induction failure, relapse, secondary malignancy, or death. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm C (Cohort 2)56.12

EFS for Patients on Arm C, Cohort 3

The Kaplan-Meier method will be used to estimate 3-year EFS, defined as the time from study entry until induction failure, relapse, secondary malignancy, or death. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm C (Cohort 3)58.18

Event-free Survival (EFS) for Patients Without High Allelic Ratio FLT3/ITD+ Mutations

The Kaplan-Meier method will be used to estimate 3-year EFS, defined as the time from study entry until induction failure, relapse, secondary malignancy, or death. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm A45.64
Arm B46.95

OS for Patients on Arm C, Cohort 1

The Kaplan-Meier method will be used to estimate 3-year OS, defined as the time from study entry until death. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm C (Cohort 1)41.67

OS for Patients on Arm C, Cohort 2

The Kaplan-Meier method will be used to estimate 3-year OS, defined as the time from study entry until death. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm C (Cohort 2)64.77

OS for Patients on Arm C, Cohort 3

The Kaplan-Meier method will be used to estimate 3-year OS, defined as the time from study entry until death. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm C (Cohort 3)61.84

Overall Survival (OS) for Patients Without High Allelic Ratio FLT3/ITD+ Mutations

The Kaplan-Meier method will be used to estimate 3-year OS, defined as the time from study entry until death. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm A65.04
Arm B68.45

Proportion of High Risk Children Without HR FLT3/ITD+ Converting From Positive MRD at End of Induction I to Negative MRD at the End of Induction II

The proportion of high risk children without HR FLT3/ITD+ converting from positive MRD at end of Induction I to negative MRD at the end of Induction II will be estimated as well as the corresponding 95% confidence interval determined using a binomial exact method. (NCT01371981)
Timeframe: Up to 8 weeks

InterventionProportion of patients (Number)
Arm A0.5000
Arm B0.5238

Proportion of Patients Experiencing Grade 3 or Higher Non-hematologic Toxicities and Infections While on Protocol Therapy

The proportion of patients experiencing at least one grade 3 or higher non-hematologic toxicity and infection while on protocol therapy will be estimated along with the corresponding 95% confidence interval determined using a binomial exact method. Toxicity will be assessed by Common Terminology Criteria for Adverse Events version 4.0 (CTCAE v4.0). (NCT01371981)
Timeframe: Up to 2 years

InterventionProportion of patients (Number)
Arm A0.8819
Arm B0.9217
Arm C (Cohort 1)0.9167
Arm C (Cohort 2)0.9394
Arm C (Cohort 3)0.9149
Arm D0.0239

Relapse Rate for Patients Without High Allelic Ratio FLT3/ITD+ Mutations

Cumulative incidence estimates 3 year relapse rate defined as time from study entry to induction failure or relapse where deaths or secondary malignancies are competing events. (NCT01371981)
Timeframe: Up to 3 years

Interventionpercentage of patients (Number)
Arm A46.67
Arm B46.65

Sorafenib Steady State Concentration

Median and range of sorafenib steady state concentration for Induction I. (NCT01371981)
Timeframe: Up to 30 days

InterventionNanogram/Milliliter (Median)
Arm C1090.0

Total Scale Score From Parent-reported Cancer Module

"Results represent the total scale scores from the parent report of the PedsQL™ 3.0 Cancer Module for timepoint 1 (up to 14 days from start of therapy). Items are reverse-scored and linearly transformed to a 0-100 scale as follows: 0=100, 1=75, 2=50, 3=25, 4=0. Therefore, a higher number is a better outcome. The total score is the sum of all the items divided by the number of items answered on all the scales. Scores on a scale is used for a unit of measure." (NCT01371981)
Timeframe: Up to 14 days

InterventionScores on a scale (Mean)
Arm A66.2
Arm B65.8
Arm C (Cohort 1)74.9
Arm C (Cohort 2)63.7

Total Scale Score From Parent-reported Multidimensional Fatigue Scale Module

"Results represent the total scale scores from the parent report of the PedsQL™ Multidimensional Fatigue Scale for timepoint 1 (up to 14 days from start of therapy). Items are reverse-scored and linearly transformed to a 0-100 scale as follows: 0=100, 1=75, 2=50, 3=25, 4=0. Therefore, a higher number is a better outcome. The total score is the sum of all the items divided by the number of items answered on all the scales. Scores on a scale is used for a unit of measure." (NCT01371981)
Timeframe: Up to 14 days

InterventionScores on a scale (Mean)
Arm A60.5
Arm B58.1
Arm C (Cohort 1)71.2
Arm C (Cohort 2)48.2

Total Scale Score From Parent-reported Pediatric Quality of Life Inventory Module

"Results represent the total scale scores from the parent report of the PedsQL™ 4.0 Generic Core Scales for timepoint 1 (up to 14 days from start of therapy). Items are reverse-scored and linearly transformed to a 0-100 scale as follows: 0=100, 1=75, 2=50, 3=25, 4=0. Therefore, a higher number is a better outcome. The total score is the sum of all the items divided by the number of items answered on all the scales. Scores on a scale is used for a unit of measure." (NCT01371981)
Timeframe: Up to 14 days

InterventionScores on a scale (Mean)
Arm A68.3
Arm B67.8
Arm C (Cohort 1)71.3
Arm C (Cohort 2)61.6

Early Marrow Status (EMS) by MRD Status End Induction (Day 29)

Early Marrow Status defined as M1 versus M2/M3 marrow is correlated with MRD (Positive vs. Negative) (NCT00103285)
Timeframe: Early Marrow Status at Day 15, MRD Status at Day 29 of therapy.

InterventionParticipants (Count of Participants)
All Patients for Induction, MRD Negative4378
All Patients for Induction, MRD Positive258

Event-Free Survival (EFS) for Low MRD (Negative) Subjects by Genetic Subset (TEL/Trisomy Positive vs Negative)

Event-free probability where EFS is defined as time from randomization to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00103285)
Timeframe: 6 years

InterventionPercent probability (Number)
Group 1-SR-low ALL, Arm I-combination Chemotherapy95.22
Group 2-SR-avg ALL, Arm I-combination Chemotherapy88.52

Event-free Survival (EFS) for SR-High Patients.

Event Free Probability where EFS time is defined as time from randomization to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00103285)
Timeframe: 6 years

Interventionpercent probability (Number)
Group 3-SR-high ALL, Combination Chemotherapy85.58

Event-free Survival (EFS) for SR-Low Patients

Event Free Probability where EFS time is defined as time from randomization to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00103285)
Timeframe: 6 years

InterventionPercent probability (Number)
SR-low ALL, Arm I-combination Chemotherapy95.22
SR-low ALL, Arm II-combination Chemotherapy93.96

Event-Free Survival Probability According to MRD Status End Induction (Day 29)

Event-Free survival by Day 29 MRD status (negative vs positive), Event Free Probability (time from study entry to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00103285)
Timeframe: MRD at Day 29 of therapy

InterventionPercent Probability (Number)
Induction Therapy, MRD Negative91.39
Induction Therapy, MRD Positive79.86

Overall Survival Probability (OS) According to Induction Day 29 MRD Status

Overall survival by Day 29 MRD status (negative vs positive), Overall survival defined as time from study entry to death or date of last contact for patients who are alive. (NCT00103285)
Timeframe: Overall Survival Probability of 6 years

Interventionpercent probability (Number)
Induction Therapy, MRD Negative97.07
Induction Therapy, MRD Positive90.47

Event-free Survival (EFS) for SR-Average ALL Patients

EFS for SR-Average with standard and Intensified Consolidation. Event Free Probability where EFS time is defined as time from randomization to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00103285)
Timeframe: 6 years

,
Interventionpercent probability (Number)
Standard and Intensified therapy
Group 2-SR-avg ALL, Arm III-combination Chemotherapy88.34
Group 2-SR-avg ALL, Arm IV-combination Chemotherapy90.51

Event-free Survival (EFS) for SR-Average ALL Patients

EFS for SR-Average with standard and Intensified Consolidation. Event Free Probability where EFS time is defined as time from randomization to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. (NCT00103285)
Timeframe: 6 years

,
Interventionpercent probability (Number)
Standard and Intensified therapyStandard therapy
Group 2-SR-avg ALL, Arm I-combination Chemotherapy83.8287.41
Group 2-SR-avg ALL, Arm II-combination Chemotherapy88.8988.29

Health-related Quality of Life Relative to Physical, Social and Emotional Impairment

To identify potentially modifiable factors associated with impaired health related quality of life (HRQOL) at different periods of therapy in the patients who are SR-average enrolled on the standard risk ALL study.Standardized scores will be computed for child function using the gender and age-adjusted scores available from normative data from a healthy population of about 10,000 children. The various domains of family functioning will be assessed using well-validated instruments and analyzed as a dichotomous variable (impaired vs. non-impaired family functioning). Multiple regression analysis will be used to test the effect of family functioning (adjusted for therapy given, age at diagnosis, gender, socioeconomic status and other factors) on child function. (NCT00103285)
Timeframe: At 1, 6 and 12 months after diagnosis and, 3 months post-therapy

InterventionPercentage of participants (Number)
Physical Impairment 1 mth after diagnosisPhysical Impairment 6 mths after diagnosisPhysical Impairment 12 mths after diagnosisPhysical Impairment 3 mths post therapySocial Impairment 1 mth after diagnosisSocial Impairment 6 mths after diagnosisSocial Impairment 12 mths after diagnosisSocial Impairment 3 mths post-therapyEmotional Impairment 1 mth after diagnosisEmotional Impairment 6 mths after diagnosisEmotional Impairment 12 mths after diagnosisEmotional Impairment 3 mths post-therapy
Induction Therapy76.3942.7529.4127.8443.3623.6623.5325.7738.8917.5613.978.25

Optimal Time Point for Advance Health Related Quality of Life Intervention

Percentage of patients with elevated Anxiety. (NCT00103285)
Timeframe: At 1 month after diagnosis and 3 months post-therapy.

InterventionPercentage of participants (Number)
At 1 month after diagnosisAt 3 months post-therapy
Induction Therapy25.224.0

Disease Free Survival

Participants are followed after completion of protocol therapy until disease progression to determine disease free survival. (NCT00126191)
Timeframe: Until disease progression up to 120 months

InterventionMonths (Mean)
Low Risk84
High Risk52

Response Rates (CR and PR) in Adults With Burkitt/Atypical Burkitt

"Complete Response (CR): Disappearance of all measurable or evaluable disease confirmed.~Partial Response (PR): Reduction of 50% or greater in the sum of the products of the perpendicular diameters of all measurable.~Of 8 High Risk participants, 7 met the primary response outcome. 1 High Risk participant did not meet protocol defined primary outcome response and died two months following enrollment." (NCT00126191)
Timeframe: 3 years

Interventionparticipants (Number)
Low Risk2
High Risk7

Duration of Event-free Survival

Event-free survival was measured as date of randomization until treatment failure. Treatment failure was defined as failure to achieve composite completed morphological remission during the induction and re-induction phase of the study lasting up to 60 days, relapse from complete response, or death from any cause, whichever occurred first. (NCT02873338)
Timeframe: Randomization up to 30 months

Interventiondays (Median)
Control (Idarubicin+Cytarabine)243.5
Dociparstat 0.125 mg/kg1
Dociparstat 0.25 mg/kg171.5

Duration of Morphologic Complete Remission

"The duration of morphologic complete remission was assessed only in subjects who had achieved morphologic complete remission and was defined as the time from achievement of complete response to the detection or relapse. Relapse was defined as the reappearance of leukemia blasts in the peripheral blood, >5% blasts in the bone marrow not attributable to another cause, or appearance/reappearance of extramedullary disease and with a bone marrow blast percentage of >5% but ≤20%. If the latter, a repeat bone marrow examination was performed at least 7 days after the first marrow examination; documentation of the bone marrow blast percentage of >5% was necessary to establish relapse. Assessments were performed every 3 months and continued until death or 18 months after the last subject was randomized, whichever occurred first.~Duration of morphologic complete remission (time from the achievement of complete response to the detection of relapse)" (NCT02873338)
Timeframe: Randomization to end of study (18 months)

Interventiondays (Median)
Control (Idarubicin+Cytarabine)233
Dociparstat 0.125 mg/kg494
Dociparstat 0.25 mg/kg294

Number of Subjects Who Achieved Composite Complete Remission

The composite complete remission (CR) rate included CR, CR without recovery of platelets (CRp), and CR without recovery of neutrophils and/or platelets (CRi), as defined by the International Working Group criteria during the induction and re-induction phases of treatment. CR was defined as an Absolute Neutrophil Count (ANC) >1000/μL, platelet count >100,000/μL, <5% blasts in bone marrow aspirate, no blasts with Auer rods, and no evidence of extramedullary disease. CRi was defined as an ANC <1000/μL and/or platelet count <100,000/, <5% blasts in bone marrow aspirate, no blasts with Auer rods, and no evidence of extramedullary disease. (NCT02873338)
Timeframe: Up to 60 days after the start of each treatment cycle

InterventionParticipants (Count of Participants)
Control (Idarubicin+Cytarabine)16
Dociparstat 0.125 mg/kg9
Dociparstat 0.25 mg/kg15

Number of Subjects Who Achieved Morphologic Complete Remission

Morphologic complete remission (CR) was evaluated by International Working Group (IWG) criteria and defined as absolute neutrophil count (ANC) >1000/microliter; platelet count >100,000, <5% blasts in bone marrow aspirate, no blasts with Auer rods, and no evidence of extramedullary disease. (NCT02873338)
Timeframe: During induction and re-induction phases of treatment (up to 60 days after the start of each treatment cycle)

InterventionParticipants (Count of Participants)
Control (Idarubicin+Cytarabine)14
Dociparstat 0.125 mg/kg8
Dociparstat 0.25 mg/kg11

Number of Subjects Who Died by Day 30

Mortality (rate of death) of subjects by Day 30, measured from the first day of induction treatment to 30 days post-induction. (NCT02873338)
Timeframe: 30 days (from first day of induction treatment to 30 days after)

InterventionParticipants (Count of Participants)
Control (Idarubicin+Cytarabine)2
Dociparstat 0.125 mg/kg1
Dociparstat 0.25 mg/kg3

Number of Subjects Who Died by Day 60.

Mortality (rate of death) of subjects by Day 60, measured from the first day of induction treatment to 60 days post-induction. (NCT02873338)
Timeframe: 60 days (from the first day of induction treatment to 60 days after)

InterventionParticipants (Count of Participants)
Control (Idarubicin+Cytarabine)2
Dociparstat 0.125 mg/kg2
Dociparstat 0.25 mg/kg3

Number of Subjects Who Died by Day 90

Mortality (rate of death) of subjects at Day 90, measured from the first day of induction treatment to 90 days post-induction. (NCT02873338)
Timeframe: 90 days (from the first day of induction treatment to 90 days after)

InterventionParticipants (Count of Participants)
Control (Idarubicin+Cytarabine)2
Dociparstat 0.125 mg/kg3
Dociparstat 0.25 mg/kg3

Time to Leukemia-free Survival

Leukemia-free survival was assessed as a secondary endpoint only in subjects who achieved composite complete remission and was measured from randomization until disease relapse or death from any cause, whichever occurred first. Assessments were performed every 3 months until death or 18 months after the last subject was randomized, whichever occurred first. (NCT02873338)
Timeframe: Randomization until disease relapse or patient death from any cause, whichever occurs first, assessed up to 30 months

Interventiondays (Median)
Control (Idarubicin+Cytarabine)292
Dociparstat 0.125 mg/kg448
Dociparstat 0.25 mg/kg166

Time to Platelet Recovery

Platelet recovery was measured from randomization to platelet recovery (platelet count >20,000/µL and >100,000/µL) (NCT02873338)
Timeframe: Randomization to platelet recovery, for up to 60 days after the start of each treatment cycle

,,
Interventiondays (Median)
Recovery to >20,000µLRecovery to >100,000µL
Control (Idarubicin+Cytarabine)3538
Dociparstat 0.125 mg/kg3650
Dociparstat 0.25 mg/kg2932

Time to Recovery of Neutrophils

Neutrophil recovery was assessed from randomization to Absolute Neutrophil Count (ANC) recovery (ANC >500/µL and >1000/µL) for up to 60 days after the start of each treatment cycle. (NCT02873338)
Timeframe: Randomization to ANC recovery, for up to 60 days after the start of each treatment cycle

,,
Interventiondays (Median)
Recovery to >500/µLRecovery to >1000/µL
Control (Idarubicin+Cytarabine)3237
Dociparstat 0.125 mg/kg4342
Dociparstat 0.25 mg/kg2935

Percentage of Participants With Continuous Complete Remission of Patients Receiving High-dose and Conventional Dose PEG-asparaginase.

"The primary objective of this study is to compare the distributions of continuous complete remission of patients randomized on the first day of the continuation phase to receive a higher dose of PEG-asparaginase or to receive the conventional dose (2,500 units/m2).~The randomization will occur on the starting day of the continuation phase, at which time all information necessary for performing the randomization should be available. In the rare case that immunophenotype and/or Day-15 MRD is not available, we will make the following assumptions: If immunophenotype is unknown at the time the randomization is to be executed, then it will be assumed B-lineage. If Day-15 MRD is unknown at the time of randomization, then it will be assumed negative (<0.01%). Past experience indicate that few patients will fall into these unknown categories." (NCT00549848)
Timeframe: 3.5 years after the last enrollment up to 12.5 years

InterventionPercentage of participants (Number)
PEG 3500 Units/m^291.6
PEG 2500 Units/m^290.7

Probability of CNS Relapse

To assess whether intensification of CNS-directed intrathecal and systemic chemotherapy will improve outcome in patients at high-risk of CNS relapse. (NCT00549848)
Timeframe: For Total XVI: 3.5 years after the last enrollment, up to approximately 13.5 years For Total XV: patients are followed continuously, up to 20.5 years

InterventionPercentage of participants (Number)
TOTXVI PEG 35000.8
TOTXVI PEG 25001.8
TOTXVI Not Randomized2.7
All Eligible Patients in TOTXV5.7

Probability of Event-free Survival

"To estimate the event-free survival of children with ALL who are treated with risk-directed therapy and to compare the EFS results of TOTXVI with that of TOTXV (NCT00137111).~EFS will be measured from the date of complete response to the date of initial failure for patients who fail. Failure includes the traditional endpoints of failure to achieve a complete remission, relapse in any site, secondary malignancy, and death during induction or remission. EFS time will be measured to the date of last contact for patients who are failure free at the time of analysis. The EFS time is defined to be zero (0) for patients who die during induction therapy or fail to achieve a complete remission." (NCT00549848)
Timeframe: For Total XVI: 3.5 years after the last enrollment, up to approximately 13.5 years For Total XV: patients are followed continuously, up to 20.5 years

InterventionPercentage of participants (Number)
TOTXVI PEG 350092.4
TOTXVI PEG 250091.1
TOTXVI Not Randomized86.3
All Eligible Patients in TOTXV87.1

Probability of Overall Survival

To estimate the overall survival of children with ALL who are treated with risk-directed therapy and to compare the EFS results of TOTXVI with that of TOTXV. (NCT00549848)
Timeframe: For Total XVI: 3.5 years after the last enrollment, up to approximately 13.5 years For Total XV: patients are followed continuously, up to 20.5 years

InterventionPercentage of participants (Number)
TOTXVI PEG 350097.5
TOTXVI PEG 250095.6
TOTVI Not Randomized90.8
All Eligible Patients in TOTXV93.5

Proportion of Participants With Minimal Residual Disease (MRD) at End of Remission Induction ≥ 0.01%

To study whether intensifying induction, including fractionated cyclophosphamide and thioguanine, in patients with day 15 MRD ≥ 5% will result in improved leukemia cytoreduction in this subgroup compared to therapy followed in the TOTXV protocol. (NCT00549848)
Timeframe: End of remission induction; day 42 in Total XVI and day 46 in Total XV

InterventionParticipants (Count of Participants)
TOTXVI PEG 35007
TOTXVI PEG 250012
TOTXVI Not Randomized20
All Eligible Patients in TOTXV44

Proportion of Participants With Minimal Residual Disease (MRD) on the 15th Day of Remission Induction ≥ 5%

To study whether intensifying induction, including fractionated cyclophosphamide and thioguanine, in patients with day 15 MRD ≥ 5% will result in improved leukemia cytoreduction in this subgroup compared to therapy followed in the TOTXV protocol. (NCT00549848)
Timeframe: Middle of remission induction, Day 15 in Total XVI and Day 19 in Total XV

InterventionParticipants (Count of Participants)
TOTXVI PEG 350022
TOTXVI PEG 250026
TOTXVI Not Randomized31
All Eligible Patients in TOTXV55

Event-free Survival Rate

Proportion of patients who were event free at 4 months (NCT00098839)
Timeframe: At 4 months after enrollment

InterventionProportion of participants (Number)
Reinduction Chemoimmunotherapy With Epratuzumab Once Weekly.604
Reinduction Chemoimmunotherapy With Epratuzumab Twice Weekly.640

Pharmacokinetics

Mean trough serum concentration measured before final dose of epratuzumab. (NCT00098839)
Timeframe: Up to day 36

Interventionug/mL (Mean)
Twice Weekly Dosing Schedule501

Rate of Minimal Residual Disease (MRD) < 0.01%

Proportion of patients (evaluable and had MRD measured at the end of Block 1) who had MRD < 0.01%. (NCT00098839)
Timeframe: At the end of Block 1 of re-induction therapy (day 36)

InterventionProportion of participants (Number)
Reinduction Chemoimmunotherapy With Epratuzumab Once Weekly.195
Reinduction Chemoimmunotherapy With Epratuzumab Twice Weekly.295

Remission Re-induction (CR2) Rate

The proportion of patients who achieved complete response at the end Block 1 of re-induction therapy. Complete Remission (CR) - Attainment of M1 bone marrow (<5% blasts) with no evidence of circulating blasts or extramedullary disease and with recovery of peripheral counts (ANC >1000/uL and platelet count >100,000/uL). Partial Remission (PR) - Complete disappearance of circulating blasts and achievement of M2 marrow status (5% or < 25% blast cells and adequate cellularity). Partial Remission Cytolytic (PRCL) - Complete disappearance of circulating blasts and achievement of at least 50% reduction from baseline in bone marrow blast count. Minimal Response Cytolytic (MRCL) - 50% reduction in the peripheral blast count with no increase in peripheral white blood cell count. (NCT00098839)
Timeframe: At the end of Block 1 of re-induction therapy (day 36)

Interventionproportion of participants (Number)
Reinduction Chemoimmunotherapy With Epratuzumab Once Weekly.646
Reinduction Chemoimmunotherapy With Epratuzumab Twice Weekly.660

Incidence of Dose-Limiting Toxicity (DLT) to Determine Maximum Tolerated Dose in Schedule A and Schedule B of Dose Escalation Phase (Group 1 and Group 2)

Patients were treated in cohorts with escalating doses of vosaroxin administered in combination with cytarabine in Schedule A, and with vosaroxin in escalating doses in Schedule B. For both Schedules, the highest dose at which fewer than 2 of 6 (<0.33) patients experienced a dose-limiting toxicity (DLT) during induction became the MTD and the recommended future dose. (NCT00541866)
Timeframe: From start of treatment (Day 1) through Induction Day 29 or the start of reinduction, whichever occurred first.

InterventionParticipants (Count of Participants)
Sch A, Cohort 10mg/m20
Sch A, Cohort 20mg/m20
SchA, Cohort 34mg/m20
Sch A, Cohort 50mg/m20
Sch A, Cohort 70mg/m21
Sch A, Cohort 80mg/m21
Sch A, Cohort 90mg/m22
Sch B, Cohort 70 mg/m20
Sch B, Cohort 80mg/m21
Sch B, Cohort 90mg/m21

Leukemia-free Survival (LFS)

Leukemia-free survival is censored at the last known alive date without report of relapse. (NCT00541866)
Timeframe: From time of the start of CR or CRp to the earliest date of relapse, commencement of reinduction therapy, or death, assessed monthly up to 2 years after the end of study visit.

Interventionmonths (Median)
Group 1 (Sch A, 10 to 90 mg/m2)12.0
Group 2 (Sch B, 70 to 90 mg/m2):4.7
Group 3 (Sch A, First Relapse, 80 mg/m2):7.4
Group 4 (Sch B, First Relapse, 90 mg/m2)25.2
Group 5 (Sch B, Primary Refractory, 90 mg/m2):NA

Overall Survival

Overall survival is censored at the earlier of the cutoff date for analysis and the last date known to be alive for patients not known to have died. (NCT00541866)
Timeframe: Time between the date of first study treatment and the date of death due to any cause for upto 2 years after the end of study visit

Interventionmonths (Median)
Group 1 (Sch A, 10 to 90 mg/m2)4.1
Group 2 (Sch B, 70 to 90 mg/m2):8.0
Group 3 (Sch A, First Relapse, 80 mg/m2):4.1
Group 4 (Sch B, First Relapse, 90 mg/m2)7.1
Group 5 (Sch B, Primary Refractory, 90 mg/m2):5.9

Remission Rates (CR+CRp)

"Complete remission (CR) plus CR with incomplete platelet recovery (CRp) per The IWG criteria for remission modified by Sunesis, assessed by investigator.~CR is defined as >1000 Neutrophils (ul), >100,000 Platelets (uL) and <5 BM Blasts (%); CRp is defined as >1000 Neutrophils (ul), <=100,000 Platelets (uL) and <5 BM Blasts (%); CRi is defined as >1000 Neutrophils (ul), <100,000 Platelets (uL) and <5 BM Blasts (%); Investigators were to determine a response category for each patient by examination of bone marrow and blood counts at the time of hematologic recovery after induction or reinduction. Investigator assessment categories included CR, CRp, CRi (CR with morphologic CR with incomplete blood count recovery), PR (partial remission), treatment failure, and relapse." (NCT00541866)
Timeframe: Monthly after the end of treatment for the first year, then every 2 months thereafter for upto 2 years

InterventionParticipants (Count of Participants)
Group 1 (Sch A, 10 to 90 mg/m2)9
Group 2 (Sch B, 70 to 90 mg/m2):4
Group 3 (Sch A, First Relapse, 80 mg/m2):7
Group 4 (Sch B, First Relapse, 90 mg/m2)3
Group 5 (Sch B, Primary Refractory, 90 mg/m2):4
Total27

All Cause Mortality

Mortality of those patients enrolled in the study and receiving intervention (NCT00541866)
Timeframe: 30 and 60 days

,,,,
InterventionParticipants (Count of Participants)
30 Days60 Days
Group 1 (Sch A, 10 to 90 mg/m2)810
Group 2 (Sch B, 70 to 90 mg/m2):02
Group 3 (Sch A, First Relapse, 80 mg/m2)12
Group 4 (Sch B, First Relapse, 90 mg/m2)01
Group 5 (Sch B, Primary Refractory, 90 mg/m2)11

Minimal Residual Disease

The presence or absence of tumor cells at the end of induction assessed by studying tissue and/or blood/marrow. Details of methods and criteria used can be found in Shiramizu at al. BJH 153:758-763, 2011 (full citation in the citation section). (NCT00057811)
Timeframe: Not Provided

Interventionpercentage of samples analyzed (Number)
Group B (Chemotherapy, Protective Therapy, Monoclonal Antib.)22
Group C (Chemotherapy, Monoclonal Antibody Therapy)70

Response Rate

Response includes both complete and partial responses. Per protocol, complete Response is defined as the complete disappearance of all clinical evidence of disease by physical examination, by imaging studies, by bone marrow biopsy (where indicated), by CNS evaluation (where indicated) and by biopsy where there is a residual abnormality on an imaging study. Bone marrow must contain <5% blasts. CSF WBC must be <5/μL with no blasts or lymphomatous cells present. Partial response is defined as: at least a 50% reduction in the size of all measurable tumor areas. Each site is to be defined by the product of the maximum length, width and depth (3 dimensions). No lesion may progress. No new lesion may appear. Bone marrow must contain <5% blasts. CSF WBC must be <5/μL with no blasts or lymphomatous cells present.. (NCT00057811)
Timeframe: Up to 5 years

Interventionpercentage of participants analyzed (Number)
Group B (Chemotherapy, Protective Therapy, Monoclonal Antib.)88
Group C (Chemotherapy, Monoclonal Antibody Therapy)83

Grade ≥ 3 Stomatitis

The incidence of grade ≥ 3 stomatitis. Grade 3 stomatitis: Confluent ulcerations or pseudomembranes; bleeding with minor trauma. Grade 4 stomatitis: Tissue necrosis; Significant spontaneous bleeding; life-threatening consequences (NCT00057811)
Timeframe: Up to 1 year

,
Interventionparticipants (Number)
Incidence of grade ≥ 3 stomatitisNo incidence of grade ≥ 3 stomatitis
Group B (Chemotherapy, Protective Therapy, Monoclonal Antib.)441
Group C (Chemotherapy, Monoclonal Antibody Therapy)634

Toxic Death

Implementation of the toxic death rate stopping rule, a death must be possibly, probably or definitely attributable to Rituximab and/or chemotherapy to be considered a toxic death. (NCT00057811)
Timeframe: Up to 1 year

,
Interventionparticipants (Number)
Toxic deathNo toxic death
Group B (Chemotherapy, Protective Therapy, Monoclonal Antib.)045
Group C (Chemotherapy, Monoclonal Antibody Therapy)238

Disease-free Survival, for Only Complete Response Patients

Disease Free Survival (DFS) is defined as the time from a Complete Response (CR) until death or relapse. The date of last clinical assesment will be used as the censor date for patients with no death or relapse. The DFS will be estimated using the Kaplan-Meier method with confidence intervals presented. (NCT00061945)
Timeframe: 9 years 4 months

Interventionmonths (Median)
Phase I - Alemtuzumab and Combination Chemotherapy58.6
Phase II - Alemtuzumab and Combination Chemotherapy19.8

Maximum Tolerated Dose (MTD) of Alemtuzumab (Phase I)

The maximum tolerated dose is defined as the highest alemtuzumab dose at which less than 40% of patients develop the dose limiting toxicity (DLT), where DLT is defined as the inability to proceed (due to medical complications) with the protocol treatment within six weeks of receiving the last dose of alemtuzumab. Groups of six patients will be enrolled into each cohort at the time of re-registration prior to starting Course IV. After a cohort has accrued 6 patients and at least 3 have completed the 2-6 week post alemtuzumab observation period without DLT, the incoming patients will be assigned to the next cohort in the table while the DLT and other toxicities continue to be assessed for the newly closed cohort. If less than 3 out of 6 enrolled patients in a cohort have completed the 2-6 week post alemtuzumab observation period without DLT, additional patients may continue to enroll in that same cohort, i.e., accrual will not be suspended while waiting for patient follow-up data. (NCT00061945)
Timeframe: 6 weeks

Interventionmg (Number)
Phase I - Alemtuzumab and Combination Chemotherapy30

Minimal Residual Disease (MRD) During Treatment With Alemtuzumab (Phase II)

Minimal Residual Disease measures the presence of of circulating leukemia cells in the body. Patients that report a Complete Response (CR) during treatment are further tested to determine the presence of small amounts of circulating leukemia cells. Here we report the number of patients who were MRD negative. (NCT00061945)
Timeframe: 9 years 4 months

InterventionParticipants (Count of Participants)
Phase II - Alemtuzumab and Combination Chemotherapy16

Number of Participants Achieving Complete Remission

A complete remission (CR) requires the following: an absolute neutrophil count (segs and bands) > 1500/μl, no circulating blasts, platelets > 100,000/μl; bone marrow cellularity > 20% with trilineage hematopoiesis, and < 5% marrow blast cells, none of which appear neoplastic. All previous extramedullary manifestations of disease must be absent (e.g., lymphadenopathy, splenomegaly, skin or gum infiltration, testicular masses, or CNS involvement). (NCT00061945)
Timeframe: 9 years

Interventionparticipants (Number)
Phase I - Alemtuzumab and Combination Chemotherapy92
Phase II - Alemtuzumab and Combination Chemotherapy145

Number of Participants Who Proceed to Course V Within 2-6 Weeks of the Last Dose of Alemtuzumab (Phase II)

The primary endpoint is the number of participants who are able to proceed to course V within two - six weeks of completion of course IV. (NCT00061945)
Timeframe: 8 months

Interventionparticipants (Number)
Phase II - Alemtuzumab and Combination Chemotherapy30

Overall Survival

Overall Survival is defines as the time from registration to death due to any cause. It is estimated using the Kaplan-Meier method with confidence intervals presented. (NCT00061945)
Timeframe: 9 years 4 months

Interventionmonths (Median)
Phase I - Alemtuzumab and Combination Chemotherapy33.6
Phase II - Alemtuzumab and Combination Chemotherapy23.1

Number of Patients Experiencing Adverse Events.

Number of patients experiencing adverse events during the course of protocol therapy. (NCT00450801)
Timeframe: Up to 5 years

Interventionparticipants (Number)
R-MACLO Cycles22
R-IVAM Cycles22
Thalidomide Therapy19

Response Rate

Percentage of participants achieving complete response (CR) to protocol therapy according to International Working Group Response Criteria for Non-Hodgkin's Lymphoma (NHL) using the CT imaging method. Patients were classified by best tumor response; CR was defined as normalization of the lactate dehydrogenase (LDH), complete disappearance of disease-related symptoms and lymph nodes, and clearance of lymphoma from involved organs; complete response unconfirmed (CRu) as a residual lymph node greater than 1.5 cm in greatest transverse diameter that had regressed by more than 75% or an indeterminate bone marrow examination; partial response (PR) as greater than 50% reduction in the involved lymph nodes, or disappearance of the involved lymph nodes but persistent bone marrow involvement; relapse/progression as new or increased lymph nodes, organomegaly, or reappearance of bone marrow involvement. (NCT00450801)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
R-MACLO-IVAM-T100

Overall Survival Rate

Percentage of participants who are alive up to five years after receipt of protocol therapy. (NCT00450801)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
1-year rate overall survival Rate2-year overall survival Rate3-year overall survival rate4-year overall survival rate5-year overall survival rate
R-MACLO-IVAM-T9696968787

Progression-free Survival Rate

Percentage of participants achieving progression-free survival at 1, 3 and 5 years after the start of protocol therapy, based upon the International Working Group Response Criteria for Non-Hodgkin's Lymphoma (NHL). Progression is defined as a ≥ 50% increase from nadir in the product of the two largest perpendicular diameters (PPD-size) of any previously identified abnormal node, or appearance of any new lesion. (NCT00450801)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
1 year progression-free survival3 year progression-free survival5-year progression-free survival
R-MACLO-IVAM-T917869

Event-free Survival

Assessed by time to treatment failure, occurrence of second malignant neoplasm, or death from any cause. Statistical analysis will be to estimate the difference in the proportion of patients treated with each therapy who are long-term event-free survivors due either to the difference between the backbone therapy regimens (CCG BFM vs NHL/BFM-95), or due to the intensification. (NCT00004228)
Timeframe: 5 years

Interventionpercentage of particpants (Number)
A0 (Localized Disease Stg I/II) Modified CCG BFM88
A1 (Disseminated, No CNS - CCG Mod BFM w/Out Intens82
A2 (Disseminated, No CNS - CCG Mod BFM w/ Intens80
B2 (CNS+) NHL/BFM-95 w/Intens Delayed Radiation Therapy63
B2 (Disseminated,CNS- (< Amend 7B)) NHL/BFM-95 w/Intens82
B1 (Disseminated CNS-) NHL/BFM-95 w/Out Intens84
B1 (NHL/BFM-95 w/Out Intens) Additional Enrollment90

Percentage of Patients With Overall Survival as Assessed by Time to Death

Overall survival will be computed by measuring the rate of deaths during induction due primarily to treatment toxicity and cumulative incidence of toxic deaths in induction or deaths in remission overall and separately for treatment groups defined by the two design factors. (NCT00004228)
Timeframe: 5 years

Interventionpercentage of participants (Number)
A0 (Localized Disease Stg I/II) Modified CCG BFM96
A1 (Disseminated, No CNS - CCG Mod BFM w/Out Intens84
A2 (Disseminated, No CNS - CCG Mod BFM w/Intens88
B2 (CNS+) NHL/BFM-95 w/Intens Delayed Radiation Therapy81
B2 (Disseminated,CNS- (< Amend 7B)) NHL/BFM-95 w/Intens85
B1 (Disseminated CNS-) NHL/BFM-95 w/Out Intens85
B1 (NHL/BFM-95 w/Out Intens) Additional Enrollment92

Complete Remission

Complete remission (CR) is defined as: <5% marrow aspirate blasts. Blasts can be >=5% if the blasts are found to be myeloid and there is no evidence of lymphoblasts by flow cytometry or immunostaining. Neutrophils >= 1000/mcl; platelets >100,000/mcl; and no blasts in the peripheral blood. C1 Extramedullary disease status as defined in the protocol. Complete remission with incomplete platelet recovery (CRi) is same as CR but platelet count may be <=100,000/mcl and/or ANC may be <1,000/mcl. (NCT00945815)
Timeframe: After induction therapy was completed (1 or 2 months)

Interventionpercentage of participants (Number)
Ara-C + Clofarabine + Epratuzumab52

Number of Patients With Grade 3 Through 5 Treatment-Related Adverse Events

Only adverse events that are possibly, probably or definitely related to study drug are reported. Any CTCAE 4.0 event of Grade 3 (severe), Grade 4 (life threatening), or Grade 5 (fatal) which were deemed to be related to protocol treatment are included. (NCT00945815)
Timeframe: Up to 5 years

InterventionParticipants (Number)
Abdominal painAcute kidney injuryAlanine aminotransferase increasedAlkaline phosphatase increasedAnemiaAnorexiaAspartate aminotransferase increasedCardiac arrestCatheter related infectionDiarrheaElectrocardiogram QT corrected interval prolongedEncephalopathyEnterocolitis infectiousFebrile neutropeniaGum infectionHepatic failureHypercalcemiaHyperglycemiaHyperkalemiaHypertensionHypocalcemiaHypokalemiaHyponatremiaHypophosphatemiaHypotensionHypoxiaInfections and infestations-Gram neg. bacteremiaInvestigations - BacteremiaLeukocytosisLung infectionLymphocyte count decreasedNervous system disorders - subdural hematomaNeutrophil count decreasedOral painPlatelet count decreasedRespiratory failureSepsisTooth infectionTumor lysis syndromeTyphlitisWhite blood cell decreased
Ara-C + Clofarabine + Epratuzumab1151616222122171111111111221114419112141128

Event Free Survival (EFS)

Time from date of treatment start until the date of first objective documentation of disease-relapse. (NCT03634228)
Timeframe: Up to 3 years, 4 months

InterventionMonths (Median)
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 01.9
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 12.4
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 21.8

Maximum Tolerated Dose (MTD) (Phase I)

As determined by dose limiting toxicity (DLT). MTD is defined the highest dose at which no more than one patient out of 6 patients experience DLTs in the first cycle. A 3+3 algorithm will be applied for dose escalation or dose de-escalation. (NCT03634228)
Timeframe: Up to 28 days

InterventionMilligrams (Number)
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b)260

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT03634228)
Timeframe: Up to 3 years, 4 months

InterventionMonths (Median)
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 02.1
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 14.3
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 27.6

Participants With a Response

Response is Complete Response (CR) + Complete Response with Incomplete Blood Count Recovery (CRi) + Partial Response (PR) + Morphologic Leukemia -Free State (MLFS: CR is Bone marrow blasts < 5%; absence of circulating blasts and blasts with Auer rods; absence of extra-medullary disease; ANC >/= 1.0 x 10^9/L; platelet count >/= 100 x 10^9/L. CRi is CR except for ANC < 1.0 x 10^9 or platelet count , 100 x 10^9/L. PR is decreased bone marrow blast % by at least 50% to a value of 5% to 25% and ANC >/= 1.0 x 10^9/L; platelet count >/= 100 x 10^9/L. MLFS is Bone marrow blasts < 5%; abcence of blasts with Auer rods; absence of extra-medullary disease; no hematologic recovery required. (NCT03634228)
Timeframe: Up to 3 years, 4 months

InterventionParticipants (Count of Participants)
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 00
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 11
Phase I (Low Dose Cytarabine, MDM2 Inhibitor DS-3032b) Cohort 21

Disease-free Survival (Consolidation Phase)

Disease-free survival is defined from the time of the confirmation of a complete remission via biopsy to the relapse of the disease. (NCT00049517)
Timeframe: Assessed during the first 4 months, then at least every three months for 2 years. then every six months until five years after study entry, and every 12 months thereafter.

InterventionMonths (Median)
Autologous HCT15.0
Go/Autologous HCT13.6

Overall Survival (Consolidation Phase)

Overall survival is defined as the time from randomization in the consolidation phase to death. (NCT00049517)
Timeframe: Assessed during the first 4 months, then at least every three months for 2 years. then every six months until five years after study entry, and every 12 months thereafter.

InterventionMonths (Median)
Autologous HCT35.5
Go/Autologous HCT27.9

Overall Survival (Induction Phase)

Overall survival is defined as the time from randomization in the induction phase to death. (NCT00049517)
Timeframe: Assessed during the first 4 months, then at least every three months for 2 years. then every six months until 5 years after study entry and every 12 months thereafter.

Interventionmonths (Median)
Standard Daunorubicin15.7
High-dose Daunorubicin23.7

Number of Participants Who Discontinued Study Treatment Due to an AE

An AE was defined as any untoward medical occurrence in a participant administered a study treatment and which does not necessarily have to have a causal relationship with this treatment. An AE can be any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a study treatment, whether or not considered related to this study treatment. The number of participants who discontinued study treatment due to an AE is summarized. (NCT00907517)
Timeframe: Up to 135 days

InterventionParticipants (Number)
MK-8776 10 mg/m^2 + Cytarabine 2 g/m^20
MK-8776 20 mg/m^2 + Cytarabine 2 g/m^20
MK-8776 40 mg/m^2 + Cytarabine 2 g/m^20
MK-8776 56 mg/m^2 + Cytarabine 2 g/m^20
MK-8776 140 mg + Cytarabine 2 g/m^20

Number of Participants Who Experienced a Dose-limiting Toxicity (DLT)

Toxicity was assessed according to the Common Terminology Criteria for Adverse Events version 3.0 (CTCAE v 3.0). DLTs in Cycle 1 consisted of any of the following: 1) Selected Grade 4 drug-related nonhematologic toxicities, 2) Selected Grade 3 drug-related nonhematologic toxicities that do not resolve to ≤ Grade 2 within 48 hours: Neurotoxicity of any duration, Nephrotoxicity of any duration, QT interval corrected by Fridericia (QTcF) prolongation of any duration, 3) Inability to administer Day 10 cytarabine therapy due to ongoing, uncontrolled serious or life-threatening toxicity. The number of participants who experienced a DLT during Cycle 1 is summarized. (NCT00907517)
Timeframe: Throughout Cycle 1 (Up to 6 weeks)

InterventionParticipants (Number)
MK-8776 10 mg/m^2 + Cytarabine 2 g/m^20
MK-8776 20 mg/m^2 + Cytarabine 2 g/m^20
MK-8776 40 mg/m^2 + Cytarabine 2 g/m^20
MK-8776 56 mg/m^2 + Cytarabine 2 g/m^20
MK-8776 140 mg + Cytarabine 2 g/m^23

Number of Participants Who Experienced an Adverse Event (AE)

An AE was defined as any untoward medical occurrence in a participant administered a study treatment and which does not necessarily have to have a causal relationship with this treatment. An AE can be any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of a study treatment, whether or not considered related to this study treatment. The number of participants who experienced an AE is summarized. (NCT00907517)
Timeframe: Up to 45 days after last dose of study treatment (Up to 180 days)

InterventionParticipants (Number)
MK-8776 10 mg/m^2 + Cytarabine 2 g/m^23
MK-8776 20 mg/m^2 + Cytarabine 2 g/m^23
MK-8776 40 mg/m^2 + Cytarabine 2 g/m^26
MK-8776 56 mg/m^2 + Cytarabine 2 g/m^26
MK-8776 140 mg + Cytarabine 2 g/m^26

Complete Remission Rate (CR) + CR With Incomplete Recovery (CRi)

Expansion from phase II pilot to a phase II trial will depend on demonstration of CR/CRi amongst 4 of the initial 10 treated patients. Investigators will measure remission rate at day 28 to address the primary endpoint of complete remission (with or without complete hematologic recovery), as defined by Cheson Criteria (ref 27). For those with extramedullary disease, Lugano criteria will be used to assess response. This is a standard assessment of drug efficacy for phase 2 clinical trial design in acute leukemias, as response correlates closely with progression free- and overall survival (PFS and OS). (NCT03575325)
Timeframe: At day 28

InterventionPercent of participants (Number)
CPX-351 Treatment30

Overall Survival (OS)

Overall Survival as defined by Cheson and Lygano Criteria, namely death due to any cause as assessed from time of first treatment. Overall Survival will be reported as per Cheson Criteria, and analyzed using a standard Kaplan-Meier approach. Patients will undergo bone marrow biopsy evaluation after each cycle of therapy per standard of care to facilitated assessment. (NCT03575325)
Timeframe: 12 months

InterventionDays (Median)
CPX-351 Treatment218

Progression Free Survival (PFS)

Progression Free Survival as defined by Cheson and Lugano Criteria, namely progression, failure to respond, or death, as assessed from time of first treatment. Progression Free Survival will be reported as per Cheson Criteria, and analyzed using a standard Kaplan-Meier approach. Patients will undergo bone marrow biopsy evaluation after each cycle of therapy per standard of care to facilitated assessment. (NCT03575325)
Timeframe: 12 months

InterventionDays (Median)
CPX-351 Treatment57

Event Free Survival

Percentage of patients who were event free at 4 months (NCT00873093)
Timeframe: 4 months after enrollment

InterventionPercentage of participants (Number)
Pre-B ALL Relapse 18-36 Mths From Dx (Chemotherapy)Age<=21 Yrs68.5
Pre-B ALL Relapse <18 Mths From Dx (Chemotherapy) Age <=21 Yrs37.8

Rate of Minimal Residual Disease (MRD) < 0.01% at End Block 1

Percentage of eligible and evaluable patients with MRD < 0.01% among those who had successful MRD determination at the end of Block 1. (NCT00873093)
Timeframe: End of Block 1 (Day 36 of Block 1) of re-induction therapy

Interventionpercentage of participants (Number)
Pre-B ALL Relapse 18-36 Mths From Dx (Chemotherapy)Age<=21 Yrs35.4
Pre-B ALL Relapse <18 Mths From Dx (Chemotherapy) Age <=21 Yrs25

Rate of Minimal Residual Disease (MRD) < 0.01% at End Block 2

Percentage of eligible and evaluable patients with MRD < 0.01% among those who had successful MRD determination at the end of Block 2. (NCT00873093)
Timeframe: End of Block 2 (Day 36 of Block 2) of re-induction therapy

Interventionpercentage of participants (Number)
Pre-B ALL Relapse 18-36 Mths From Dx (Chemotherapy)Age<=21 Yrs66.7
Pre-B ALL Relapse <18 Mths From Dx (Chemotherapy) Age <=21 Yrs42.1

Rate of Minimal Residual Disease (MRD) < 0.01% at End Block 3

Percentage of eligible and evaluable patients with MRD < 0.01% among those who had successful MRD determination at the end of Block 3. (NCT00873093)
Timeframe: End of Block 3 (Day 36 of Block 3) of re-induction therapy

InterventionPercentage of participants (Number)
Pre-B ALL Relapse 18-36 Mths From Dx (Chemotherapy)Age<=21 Yrs80
Pre-B ALL Relapse <18 Mths From Dx (Chemotherapy) Age <=21 Yrs63.6

Second Complete Remission Rate at the End of Block 1 Reinduction Chemotherapy

The percentage of eligible and evaluable patients who have achieved complete response at the end Block 1 of re-induction therapy. (NCT00873093)
Timeframe: The outcome is measured the end of Block 1 (Day 36 of Block 1) of re-induction therapy.

InterventionPercentage of participants (Number)
Pre-B ALL Relapse 18-36 Mths From Dx (Chemotherapy)Age<=21 Yrs72.2
Pre-B ALL Relapse <18 Mths From Dx (Chemotherapy) Age <=21 Yrs63

Severe Adverse Events (SAE) Rate.

The proportion of SAE rate among all eligible patients (NCT00873093)
Timeframe: 4 months

Interventionpercentage of participants (Number)
Overall8.2

Toxic Death Rate

The proportion of toxic death rate among all eligible patients. (NCT00873093)
Timeframe: 4 months

Interventionpercentage of participants (Number)
Overall2.1

Area Under the Curve of LY2181308 Over the Dosing Interval

Area under the curve of LY2181308 over the dosing interval (NCT00620321)
Timeframe: Day 3: 0,12,24,36,48,60,72,84,96,108,120,132 hours

Interventionnanograms*hour/milliliter (ng*h/mL) (Geometric Mean)
LY2181308216947
LY2181308 + Idarubicin + Cytarabine185734

Change From Baseline in Survivin Index at Day 2

Data presented are the ratio of Day 2 survivin index to the baseline survivin index. Survivin is a protein expressed in tumor cells, including acute myeloid leukemia (AML), which regulates mitosis and prevents tumor cell death. Survivin index was calculated as ([blast survivin mean equivalent fluorochrome (MEFL) - blast isotypic control MEFL]/blast isotypic control MEFL). (NCT00620321)
Timeframe: Baseline, Day 2

Interventionratio (Least Squares Mean)
LY21813080.43

Percentage of Participants With Response to LY2181308 Sodium in Combination With Idarubicin and Cytarabine (Remission Rates)

Response is complete remission (CR) + CR with incomplete blood count recovery (CRi) + partial remission (PR) + cytoreduction. CR: fewer than 5% blasts based on a cell count of at least 200 cells from a bone marrow aspirate containing bone marrow spicules, in the setting of peripheral blood recovery to: platelets ≥100x10⁹/liter (L), neutrophils ≥10⁹/L. PR: defined as a decrease of at least 50% in blast count on the bone marrow aspirate; or cytoreduction (defined as a decrease in blast count not meeting the criteria for a PR or CR). Response rate is calculated as a total number of participants with CR or CRi or PR or cytoreduction divided by the total number of participants treated multiplied by 100. (NCT00620321)
Timeframe: Baseline to progression of disease or death up to 6 months

Interventionpercentage of participants (Number)
LY21813080
LY2181308 + Idarubicin + Cytarabine56.3

Pharmacodynamics: Number of Participants With Survivin Protein Expression

Pharmacodynamics: Number of participants with Survivin Protein Expression. (NCT00620321)
Timeframe: 6 Months

InterventionParticipants (Count of Participants)
LY21813082

Number of Participants Who Died Due to Progressive Disease or Unknown Cause During the 21 Days Post Study Treatment Follow-Up

Deaths due to progressive disease (PD) and unknown cause are not considered adverse events. Deaths due to PD and unknown cause occurring during the 21-day follow-up period after treatment discontinuation are reported here and for those occurring while participants were on treatment are reported in the Participant Flow. Deaths due to serious adverse events occurred during the study including the 21-day follow-up period are reported in the Reported Adverse Events section. (NCT00620321)
Timeframe: Study treatment discontinuation up to 21 days post study treatment discontinuation

,
InterventionParticipants (Count of Participants)
Death due to progressive diseaseDeath due to unknown cause
LY218130811
LY2181308 + Idarubicin + Cytarabine00

Number of Participants With Adverse Events (Safety Profile)

Data are presented as number of participants who experienced serious adverse events (SAE) and possibly drug-related treatment-emergent adverse events (TEAE) during the study including the 21-day follow-up period. A summary of serious adverse events and other nonserious adverse events regardless of causality is located in the Reported Adverse Events section. (NCT00620321)
Timeframe: Start of treatment to study completion up to 6.7 months

,
InterventionParticipants (Count of Participants)
SAEDrug-related TEAE
LY218130862
LY2181308 + Idarubicin + Cytarabine1311

Disease Free Survival(DFS) Rate

Disease free survival time is defined for all evaluable patients who have achieved a CR or CRi as the time from registration to relapse or death due to any cause. The distribution of disease-free survival will be estimated using the method of Kaplan-Meier. (NCT01806571)
Timeframe: 35 months

Interventionpercentage of patients (Number)
Treatment (Nilotinib, Daunorubicin Hydrochloride, Cytarabine)56.4

Duration of Complete Response

The distribution of duration of complete response will be estimated using the method of Kaplan-Meier. (NCT01806571)
Timeframe: 2 years

InterventionMonths (Median)
Treatment (Nilotinib, Daunorubicin Hydrochloride, Cytarabine)NA

Incidence of Adverse Events as Assessed by NCI CTCAE Version 4.0

The maximum grade for each type of adverse event will be recorded for each patient, and frequency tables will be reviewed to determine patterns. Additionally, the relationship of the adverse event(s) to the study treatment will be taken into consideration. This data will be reported in the Adverse Events section of the results. (NCT01806571)
Timeframe: 35 months

InterventionParticipants (Count of Participants)
Treatment (Nilotinib, Daunorubicin Hydrochloride, Cytarabine)34

Overall Survival(OS) Rate

Overall survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier(Kaplan E 1958). (NCT01806571)
Timeframe: 39 Months

Interventionpercentage of patients alive (Number)
Treatment (Nilotinib, Daunorubicin Hydrochloride, Cytarabine)70.6

Proportion of Complete Responses (CR or CRi) During Induction Therapy

The proportion of successes will be estimated by the number of successes divided by the total number of evaluable patients. Confidence intervals for the true success proportion will be calculated according to the approach of Duffy and Santner. (NCT01806571)
Timeframe: Up to 56 days

Interventionproportion of participants (Number)
Treatment (Nilotinib, Daunorubicin Hydrochloride, Cytarabine)0.618

Days to Engraftment of Lymphocytes

Lymphocyte recovery: designated by the first of 3 consecutive days with absolute lymphocyte count (ALC) above 500/mm(3). (NCT00520130)
Timeframe: 2 years

InterventionDays (Median)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm16
B - Cyclosporine (AC Arm)76

Days to Engraftment of Neutrophils

Days to engraftment is defined as neutrophil recovery: designated by the first of 3 consecutive days with an absolute neutrophil count (ANC) above 500/mm(3). (NCT00520130)
Timeframe: 2 years

InterventionDays to neutrophil engraftment (Median)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm11
B - Cyclosporine (AC Arm)9

Days to Engraftment of Platelets

Platelet recovery: designated by the first of 7 days where the platelet count remains above 20,000/mm(3) without transfusion support (NCT00520130)
Timeframe: 2 years

InterventionDays (Median)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm19
B - Cyclosporine (AC Arm)14

Early Treatment Related Mortality

Any death occurring within 28 days after transplantation in a patient in continuous remission. (NCT00520130)
Timeframe: Less than or equal to 28 days after transplantation

Interventionparticipants (Number)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm1
B - Cyclosporine (C) Arm0

Overall Survival

Time between the first day of transplant to the day of death. (NCT00520130)
Timeframe: Patients were followed for an average of up to 5 years.

InterventionMonths (Median)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm41.7
B - Cyclosporine (AC) Arm18.8

Percentage of Participants With Grade II-IV Acute Graft Versus Host Disease (GVHD)

Acute GVHD is assessed by the 1994 Consensus Conference on Acute GVHD Grading criteria. See Przepiorka D, Weisdorf D, Martin P, et al. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995; 15:825-8., for grading criteria. (NCT00520130)
Timeframe: 6 months

Interventionpercentage of participants (Number)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm42
B - Cyclosporine (AC) Arm38

Percentage of Participants With Grade III-IV Acute Graft Versus Host Disease (GVHD)

Acute GVHD is assessed by the 1994 Consensus Conference on acute GVHD Grading criteria. See Przepiorka D, Weisdorf D, Martin P, et al. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995; 15:825-8., for grading criteria. (NCT00520130)
Timeframe: 6 months

Interventionpercentage of participants (Number)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm13
B - Cyclosporine (AC) Arm21

Percentage of Participants With Late Treatment Related Mortality

Any death occurring 28 days or more after transplantation in a patient in continuous remission. (NCT00520130)
Timeframe: Greater than 28 days after transplantation

Interventionpercentage of participants (Number)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm28
B - Cyclosporine (AC) Arm29

Toxicities

Here are the number of participants with adverse events. For a detailed list of adverse events, see the adverse event module. (NCT00520130)
Timeframe: 103 months and 22 days

Interventionparticipants (Number)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm43
B - Cyclosporine (AC) Arm42

Changes in CD8 T Cell Receptor Vbeta Repertoire

Ribonucleic acid (RNA) was extracted from sorted CD4 and cluster of differentiation 8 (CD8) T cells and analyzed for Vbeta repertoire by nested polymerase chain reaction (PCR) analysis using Vbeta family specific primers and a labeled constant region primer (spectratyping). The receptor repertoire diversity was calculated from spectratyping data by creating a normal standard for repertoire diversity from healthy normal controls and assessing the divergence of individual patient's T cell receptor repertoire from these standard normal donor values. In this Vbeta repertoire divergence index, lower numbers are consistent with a more normal highly diverse repertoire, and high numbers represent a highly skewed, oligoclonal repertoire. The assay is described in Memon SA et al, J Immunol Methods, 2012, 375: 84-92. The repertoire diversity of the CD4 and CD8 T cells of the donor infusion is shown for comparison. (NCT00520130)
Timeframe: Donor at time of collection and recipient at 1, 3, 6 and 12 months post transplant

,
InterventionDivergence index (Median)
1 month3 months6 mo (TMS=7; AC= 9)12 mo (TMS=7; AC= 9)Donor CD8 cells (TMS=8; AC= 8)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm6254625547
B - Cyclosporine (AC) Arm7881848943

Changes in Cluster of Differentiation 4 (CD4) T Cell Receptor Vbeta Repertoire

Ribonucleic acid (RNA) was extracted from sorted CD4 and cluster of differentiation 8 (CD8) T cells and analyzed for Vbeta repertoire by nested polymerase chain reaction (PCR) analysis using Vbeta family specific primers and a labeled constant region primer (spectratyping). The receptor repertoire diversity was calculated from spectratyping data by creating a normal standard for repertoire diversity from healthy normal controls and assessing the divergence of individual patient's T cell receptor repertoire from these standard normal donor values. In this Vbeta repertoire divergence index, lower numbers are consistent with a more normal highly diverse repertoire, and high numbers represent a highly skewed, oligoclonal repertoire. The assay is described in Memon SA et al, J Immunol Methods, 2012, 375: 84-92. The repertoire diversity of the CD4 and CD8 T cells of the donor infusion is shown for comparison. (NCT00520130)
Timeframe: Donor at time of collection and recipient at 1, 3, 6 and 12 months post transplant

,
InterventionDivergence index (Median)
1 month3 months6 mo (TMS=7; AC= 9)12 mo (TMS=7; AC=9)Donor CD4 cells (TMS=8; AC=8)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm4831353023
B - Cyclosporine (AC) Arm9066756722

Immune Reconstitution of Cluster of Differentiation 4 (CD4) T Cell Populations

Cluster of Differentiation 3 (CD3)+CD4+ and CD3+Cluster of Differentiation 8 (CD8)+ T cells within the lymphocyte population were determined by flow cytometry. The absolute numbers of cells/µl were calculated from the absolute lymphocyte count. (NCT00520130)
Timeframe: 2 weeks, and 1, 3, 6, 12 and 24 months post transplant

,
InterventionCells/µl (Median)
2 weeks1 month3 months6 months12 months24 months
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm171285297387447451
B - Cyclosporine (AC) Arm02161121132373

Immune Reconstitution of Cluster of Differentiation 8 (CD8) T Cell Populations

Cluster of differentiation 3 (CD3)+cluster of differentiation 4 (CD4)+ and CD3+CD8+ T cells within the lymphocyte population were determined by flow cytometry. The absolute numbers of cells/µl were calculated from the absolute lymphocyte count. (NCT00520130)
Timeframe: 2 weeks, 1, 3, 6, 12 and 24 months post transplant

,
InterventionCells/µl (Median)
2 weeks1 month3 months6 months12 months24 months
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm72204334429485434
B - Cyclosporine (AC) Arm1654158243502

Immune Reconstitution of Normal Killer (NK) Cells

Cluster of differentiation 3 (CD3) - cluster of differentiation 56 (CD56) + Natural Killer (NK) cells within the lymphocyte population were determined by flow cytometry. The absolute numbers of cells/µl were calculated from the absolute lymphocyte count. (NCT00520130)
Timeframe: 2 weeks, and 1, 3, 6, 12, and 24 months post transplant

,
InterventionCells/µl (Median)
2 weeks1 month3 months6 months12 months24 months
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm123270134136134133
B - Cyclosporine (AC) Arm1531124202150307

Percentage of Participants With Chronic Graft Versus Host Disease (cGVHD)

Chronic GVHD is assessed by the 2005 Chronic GVHD Consensus Project. First the individual organ scoring is done, and then based on that the Global score is determined (mild-moderate-severe). See Citation: Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report. Biol Blood Marrow Transplant. 2005; 11:945-56., for grading criteria. (NCT00520130)
Timeframe: 2 years post transplant

,
Interventionpercentage of participants (Number)
Moderate or Severe cGVHDSevere cGVHD
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm5028
B - Cyclosporine (AC) Arm125

Recovery of Naïve Cluster of Differentiation 4 (CD4) T Cells

The percentage of C-C motif chemokine receptor 7 (CCR7)+CD45RA+ naïve T cells within the CD4 T cell populations was determined by flow cytometry. (NCT00520130)
Timeframe: Recipient recovery at 6, 12 and 24 months post transplant

,
Intervention% of naive (CCR7+CD45RA+) CD4 Cells (Median)
6 mo (TMS=28; AC= 28)12 mo (TMS=25; AC= 21)24 mo (TMS=18; AC= 13)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm242220
B - Cyclosporine (AC) Arm1725

Recovery of Naïve Cluster of Differentiation 8 (CD8) T Cells

The percentage of CCR7+CD45RA+ naïve T cells within the CD4 and CD8 T cell populations was determined by flow cytometry. (NCT00520130)
Timeframe: Recipient recovery at 6, 12 and 24 months post transplant

,
Intervention% of naive (CCR7+CD45RA+) CD8 Cells (Median)
6 mo (TMS=28; AC= 28)12 mo (TMS=25; AC= 21)24 mo (TMS=18; AC= 13)
A - Tacrolimus, Methotrexate, Sirolimus (TMS) Arm201716
B - Cyclosporine (AC) Arm366

Overall Survival

(NCT00512252)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Phase II Dose Treatment37

Phase I Only: Optimal Dose of AMD3100 Plus MEC in Patients With Relapsed or Refractory AML

A standard 3+3 design was used in the Phase I portion starting with the AMD3100 dose of 80 mcg/kg and escalating by 80 mcg/kg for each successive cohort up to a maximum of 240 mcg/kg/d. The optimal dose was defined as the highest dose of AMD3100 <= 240 mcg/kg at which 0-1 of 6 patients experienced a dose limiting toxicity. (NCT00512252)
Timeframe: Completion of all patients in Phase I portion (232 days)

Interventionmcg/kg (Number)
Phase I Dose Escalation240

Relapse-free Survival

"This is determined only for patients achieving a complete remission. Defined as the interval from the date of the first documentation of a leukemia free state to date of recurrence or death due to any cause.~Kaplain-Meier estimate was used." (NCT00512252)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Phase II Dose Treatment42.9

Time to Neutrophil Recovery

Defined as the date of the first dose of AMD3100 to the date that the absolute neutrophil count >1,000 cells/mm^3. (NCT00512252)
Timeframe: 42 days

Interventiondays (Median)
Phase II Dose Treatment28

Time to Platelet Recovery

Defined as the date of the first dose of AMD3100 to the date that the platelet count is >100,000/mm3 in the absence of platelet transfusions. (NCT00512252)
Timeframe: 42 days

Interventiondays (Median)
Phase II Dose Treatment28.5

Characterize the Mobilization of Leukemic Cells With AMD3100 by Measuring the Peak Mobilization of AML Blasts (Phase I)

Measured at 0 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, and 24 hours after AMD3100 dose on Day 0. (NCT00512252)
Timeframe: Day 0

,,
Interventionpercentage of AML blasts (Median)
AML blasts at 0 hoursAML blasts at 1 hourAML blasts at 2 hoursAML blasts at 4 hoursAML blasts at 6 hoursAML blasts at 8 hoursAML blasts at 12 hoursAML blasts at 24 hours
Phase I Dose Escalation - Dose Level 146.026.026.037.031.032.027.035.0
Phase I Dose Escalation - Dose Level 24.09.037.516.014.019.045.023
Phase I Dose Escalation - Dose Level 343.530.532.530.027.026.035.055

Characterize the Mobilization of Leukemic Cells With AMD3100 by Measuring the Peak Mobilization of Total Leukocytes (Phase I)

"Measured at 0 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, and 24 hours after AMD3100 dose on Day 0.~Characterization of the mobilized cells as well as the kinetics of mobilization will be determined by analyzing the surface expression of mobilized cells by flow cytometry at the specified time points in conjunction with their total leukocyte count from the patient's CBC." (NCT00512252)
Timeframe: Day 0

,,
Interventioncells x 10^3/microliter (Median)
Total leukocytes at 0 hoursTotal leukocytes at 1 hourTotal leukocytes at 2 hoursTotal leukocytes at 4 hoursTotal leukocytes at 6 hoursTotal leukocytes at 8 hoursTotal leukocytes at 12 hoursTotal leukocytes at 24 hours
Phase I Dose Escalation - Dose Level 12.54.344.74.84.55.14.3
Phase I Dose Escalation - Dose Level 24.77.08.59.411.312.012.17.9
Phase I Dose Escalation - Dose Level 33.56.47.58.39.58.97.85.8

Phase II Only: Complete Response Rate of AMD3100 + MEC

"Responses were assessed according to the International Working Group Criteria for AML. All patients who received at least one dose of AMD3100 were considered evaluable for response.~Response rate was the rate of complete remission plus complete remission with incomplete blood count recovery (CR + CRi)." (NCT00512252)
Timeframe: 42 days

,,,
Interventionpercentage of participants (Number)
CR + CRiCR only
>= Second Relapse/Salvage2525
First Relapse, First Salvage5647
Primary Refractory2020
Total4639

Safety and Tolerability of AMD3100 + MEC.

Treatment related mortality (deaths occurring during treatment) (NCT00512252)
Timeframe: 42 days

Interventionparticipants (Number)
SepsisAdverse transfusion reaction w/febrile neutropenia
Phase II Dose Treatment21

Treatment Failure

Treatment failures includes those patients for whom treatment has failed to achieve a CR or a CRi. (NCT00512252)
Timeframe: 42 days

,,,
Interventionparticipants (Number)
Persistent leukemiaDeath during aplasiaUnknown
>= Second Relapse/Salvage300
First Relapse, First Salvage1211
Primary Refractory620
Total2131

Number of Patients With Complete Remission

Complete remission is defined as: less than 5% bone marrow blasts, neutrophils greater or equal to 1,000 per microliter, platelets greater than 100,000 per microliter, no blasts in the peripheral blood, and no extramedullary disease (NCT00337168)
Timeframe: Between day 28 and day 35 inclusive

Interventionparticipants (Number)
Induction3

Number of Patients With Very Poor Risk Cytogenetics

(NCT00337168)
Timeframe: On average, 2 weeks before treatment started

Interventionparticipants (Number)
Induction10

Expression of Nucleoside Transporters

Expression was examined in paraffin-embedded tissue by immunohistochemistry. Intensities were scored on a 0-2+ scale. High expression was a score of 2+. (NCT00337168)
Timeframe: On average, two weeks before treatment started

Interventionparticipants (Number)
High expression of hENT1High expression of hCNT3High expression of dCK cytoplasmicHigh expression of dCK nuclear
Induction7644

Toxicity

Number of patients with Grade 3-5 adverse events that are related to study drug by given type of adverse event (NCT00337168)
Timeframe: Patients were assess for adverse events after each induction cycle (up to two cycles) and after the one consolidation cycle

InterventionParticipants with a given type of AE (Number)
ALT, SGPT (serum glutamic pyruvic transaminase)AST, SGOT (serum glut oxaloacetic transaminase)Albumin, serum-low (hypoalbuminemia)AnorexiaAscites (non-malignant)Bilirubin (hyperbilirubinemia)Calcium, serum-low (hypocalcemia)ColitisColitis, infectious (e.g., Clostridium difficile)ConfusionCreatinineDIC (disseminated intravascular coagulation)Death not assoc with CTCAE term-Multi-organ failDermatology/Skin-Other (Specify)DiarrheaDyspnea (shortness of breath)Edema: limbFatigue (asthenia, lethargy, malaise)Febrile neutropeniaGlucose, serum-high (hyperglycemia)HemoglobinHypotensionHypoxiaINRInfec with Gr 34 neutrophils - Bladder (urinIInfec with Gr 34 neutrophils - BloodInfec with Gr 34 neutrophils - Catheter-relaInfec with Gr 34 neutrophils - ColonInfec with Gr 34 neutrophils - ConjunctivaInfec with Gr 34 neutrophils - LarynxInfec with Gr 34 neutrophils - Lung (pneumonInfec with Gr 34 neutrophils - Skin (celluliInfec with Gr 34 neutrophils - Urinary tractInfec with Gr 34 neutrophils - WoundInfection with unknown ANC - Lung (pneumonia)Infection-Other (Specify)Leukocytes (total WBC)Liver dysfunction/failure (clinical)LymphopeniaMental statusNeutrophils/granulocytes (ANC/AGC)PTT (Partial thromboplastin time)Pain - Abdomen NOSPain - BackPain - BonePlateletsPleural effusion (non-malignant)Pneumonitis/pulmonary infiltratesPotassium, serum-high (hyperkalemia)Potassium, serum-low (hypokalemia)Pruritus/itchingRenal failureRestrictive cardiomyopathySodium, serum-high (hypernatremia)Sodium, serum-low (hyponatremia)Supraventricular and nodal arrhythmiaTumor lysis syndromeTyphlitis (cecal inflammation)Uric acid, serum-high (hyperuricemia)
Induction5731231121411121121411331119121121111111171181111201114131121111

Early Post BMT Toxicities

Number of participants who experience organ failure or severe infections (defined as Grade III/IV by NCI CTC for Adverse Events (CTCAE), version 2.0) (NCT00368355)
Timeframe: 100 Days

InterventionParticipants (Count of Participants)
CLINIMACS Device14
ISOLEX Device14

Engraftment Rate After Transplant

Percentage of participants with hematopoietic engraftment post-transplant. Engraftment is defined as the first day absolute neutrophil counts exceeded 0.5 X 10^9/ml. (NCT00368355)
Timeframe: 28 days

Interventionpercentage of participants (Number)
CLINIMACS Device100
ISOLEX Device100

Severe GVHD Rate

Percentage of participants with Grade III/IV acute GVHD. Severe GVHD is defined as Grade III/IV acute GVHD. (NCT00368355)
Timeframe: 100 Days

Interventionpercentage of participants (Number)
CLINIMACS Device0
ISOLEX Device4.3

Disease-free Survival (DFS) Rate at 1 Year

"For participants who achieved a complete remission (CR), this is the percentage of participants who were alive and relapse free at 1 year. The 1 year rate, with 95% confidence interval, was estimated using the Kaplan-Meier method~A CR is defined as those with > 20% cellularity of bone marrow biopsy, no presence of extramedullary leukemia for AML, <5 % myeloblast cells for bone marrow with peripheral blood and normal complete blood count (absolute neutrophils > 1000 mL and platelets >= 100,000 mL)." (NCT00416598)
Timeframe: At 1 year

Interventionpercentage of participants (Number)
Decatibine Maintenance80

Number of Participants Who Completed Maintenance Decitabine.

To determine feasibility of decitabine maintenance, this outcome measures the number of participants who completed all 8 planned cycles of decitabine maintenance as per protocol. (NCT00416598)
Timeframe: Up to 5 years

Interventionparticipants (Number)
Decatibine Maintenance62

Event-free Survival (EFS)

EFS - time from study entry until failure to achieve complete remission during consolidation, relapse, or death. For further clarification see definitions provided in the protocol. (NCT00866918)
Timeframe: At 3 years from study entry

InterventionPercentage of participants (Number)
Standard Risk95.4
High Risk82.9

Hematologic Remission Rate

Proportion of patients in hematologic remission at end of consolidation, course 1 are reported. (NCT00866918)
Timeframe: End of consolidation, course 1: up to 5 months

InterventionProportion of participants (Number)
Standard Risk1.0000
High Risk0.8824

Hematologic, Molecular, and Cytogenetic Remission Rate

Proportion of patients in hematologic, molecular, and cytogenetic remission at end of consolidation, course 3 and 4 are reported. Patients were determined to be in remission by all three criteria. (NCT00866918)
Timeframe: End of consolidation, course 3; up to 7 months (for Standard Risk) or end of consolidation, course 4; up to 9 months (for High Risk)

InterventionProportion of participants (Number)
Standard Risk0.8095
High Risk0.5882

Overall Survival (OS)

OS - time from study entry to death. (NCT00866918)
Timeframe: At 3 years from study entry

InterventionPercentage of participants (Number)
Standard Risk98.4
High Risk85.7

Number of Participants With a 2-Year Progression-Free Survival (PFS)

Response evaluated using the standard criteria response for lymphoma through CT scan. (NCT00591630)
Timeframe: 2 years (beginning day 30 after treatment)

InterventionParticipants (Count of Participants)
Group 1 - Zevalin + BEAM + Rituximab +Stem Cell Transplant6
Group 1 - Zevalin + BEAM + Stem Cell Transplant8
Group 2 - BEAM + Rituximab + Stem Cell Transplant8
Group 2 - BEAM + Stem Cell Transplant5

Progression Free Survival

Overall Progression Free Survival at 2 years (NCT00594815)
Timeframe: 2 Years

Interventionpercentage of participants (Number)
Immunocompetent Pts With Newly Diagnosed Primary CNS Lymphoma57

Total Number of Participants Who Experienced Acute Treatment Related Adverse Events

The toxicity of this combined regimen will be measured using the NCI CTC version 2.0. (NCT00594815)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Immunocompetent Pts With Newly Diagnosed Primary CNS Lymphoma52

1 Year Progression-free Survival Rate

Progression-free survival is measured from the first day of induction chemotherapy to the date of progression, relapse or death. Definitions of response criteria are as described by Cheson. Progressive Disease: >50% increase from nadir in the sum of the products of the greatest diameters (SPD) of any previously identified abnormal node for PDs or nonresponders, appearance of any new lesion during or at the end of therapy. (NCT00577629)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Experimental: Induction + Consolidation + Bexxar0.74

Disease-free Survival

Disease-free survival is measured from the date of CR or CRu to date of relapse or death (NCT00577629)
Timeframe: 10 years

Interventionmonths (Mean)
Experimental: Induction + Consolidation + Bexxar54.10

Overall Response

"Percent of subjects who achieved a complete response (CR) or partial response (PR) any time during the treatment period.~CR = complete disappearance of all detectable clinical evidence of disease and disease-related symptoms if present before therapy.~PR =~>/= 50% decrease in sum of the product of the diameters (SPD) of up to six of the largest dominant nodes or nodal masses.~No increase should be observed in the size of other nodes, liver, or spleen.~Splenic and hepatic nodules must regress by ≥ 50% in their SPD or, for single nodules, in the greatest transverse diameter.~Except splenic and hepatic nodules, involvement of other organs is usually assessable and no measurable disease should be present.~Patients who achieve a CR by the above criteria, but who have persistent morphologic bone marrow involvement will be considered partial responders.~No new sites of disease should be observed." (NCT00577629)
Timeframe: up to 1 year

Interventionpercentage of participants (Number)
Experimental: Induction + Consolidation + Bexxar92

Overall Survival

Overall Survival is measured from the first day of chemotherapy until death from any cause. (NCT00577629)
Timeframe: 10 years

Interventionpercentage of participants (Number)
Experimental: Induction + Consolidation + Bexxar82

Secondary Malignancies

The number of patients who develop secondary malignancies including solid tumors, acute leukemia and myelodysplasia or other bone marrow failure syndromes. (NCT00577629)
Timeframe: 10 years

InterventionParticipants (Count of Participants)
Myelodysplastic Syndrome (MDS)Acute Myeloid Leukemia (AML)Pancreatic CancerHodgkins LymphomaMelanomaRenal Cell Carcinoma
Experimental: Induction + Consolidation + Bexxar311111

Maximum Tolerated Dose [MTD] Determination by Number of Participants With Dose Limiting Toxicity (DLT)

"The Maximum tolerated dose (MTD) was the highest dose level at which fewer than 2 of 6 patients developed a dose limiting toxicity (DLT) in the first two cycles of therapy. A 3 by 3 design was used for dose escalation in the phase I portion of the study.~A dose-limiting toxic effect (DLT) was defined as a clinically significant adverse event or abnormal laboratory value directly attributable to everolimus and assessed as unrelated to disease progression, intercurrent illness, or concomitant medications, occurring during the first or second cycle of therapy, that met any of the following criteria: CTCAE version 3.0 grade 3 increased AST or ALT for 7 days, CTCAE grade 4 increased AST or ALT of any duration, or any other clinically significant CTCAE grade 3 or 4 toxic effect. Electrolyte abnormalities (changes in glucose, chemistries, liver enzymes, pancreatic enzymes) correctable by optimal therapy and without clinical impact were not considered DLTs." (NCT00968253)
Timeframe: Following first two dose cycles (21 days/each), up to 42 days

InterventionParticipants (Count of Participants)
Phase I: RAD001 5 mg + Combination Chemo0
Phase I: RAD001 10 mg + Combination Chemo1

Overall Response Rate (OR) Where OR = CR + CRp + CRi

Number of participants out of total treated who experienced a complete response response according to RECIST criteria either (CR + CRp) CR Without Platelet Recovery. Response (CR + CRp) defined as Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x10^9/L, platelet count > 100 x10^9/L, and blasts < 5% in a normocellular or hypercellular marrow. Complete remission without platelet recovery (CRp): Peripheral blood and marrow parameters as for CR, but with platelet count > 20 x 10^9/L and < 100 x 10^9/L in the absence of platelet transfusions. CR with incomplete blood count recovery (CRi): Same as CR but platelets ≤ 100,000/mcl and/or neutrophils ≤ 1,000/mcl. (NCT00968253)
Timeframe: 8 courses of treatment, up to 24 weeks

Interventionpercentage of participants (Number)
Phase I: RAD001 5 mg + Combination Chemo33
Phase I: RAD001 10 mg + Combination Chemo33
Phase II: MTD RAD001 + Combination Chemo33

Participant Responses by Daily Dose Level Assignment (RAD001 5 mg, 10 mg and MTD 5 mg)

Response defined as Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x10^9/L, platelet count > 100 x10^9/L, and blasts < 5% in a normocellular or hypercellular marrow. Complete remission without platelet recovery (CRp): Peripheral blood and marrow parameters as for CR, but with platelet count > 20 x 10^9/L and < 100 x 10^9/L in the absence of platelet transfusions. CR with incomplete blood count recovery (CRi): Same as CR but platelets ≤ 100,000/mcl and/or neutrophils ≤ 1,000/mcl. Partial remission (PR): Peripheral blood count recovery as for CR, with decrease in marrow blasts by > 50% from pretreatment values with no more than 25% leukemia/lymphoma cells in the marrow. Nonresponder, Other: All other responses will be considered failures. (NCT00968253)
Timeframe: Up to 20 cycles of study drugs (21 day cycles) or till disease progression

,,
Interventionparticipants (Number)
Complete RemissionComplete Remission without platelet recoveryCR with incomplete blood count recoveryPartial RemissionNonresponder
Phase I: RAD001 10 mg + Combination Chemo20115
Phase I: RAD001 5 mg + Combination Chemo10011
Phase II: MTD RAD001 + Combination Chemo31008

Number of Participants With Adverse Events (AEs)

Safety and tolerability of sorafenib with DepoCyt. Toxicities were to be reported using tables and descriptive statistics by type and grade. All patients were to be followed up until death. (NCT00964743)
Timeframe: 6 Months

Interventionparticipants (Number)
Intrathecal DepoCyt and Oral Sorafenib2

Complete Response Rate

Percentage of participants who achieved a complete response (CR). A CR was defined as normalization of blood counts and a marrow showing less than 5% blasts occurring on or before day 60. (NCT00651261)
Timeframe: Induction therapy (up to 60 days)

Interventionpercentage of participants (Number)
Induction and Consolidation Chemotherapy Plus Midostaurin59
Induction and Consolidation Chemotherapy Plus Placebo54

Disease-free Survival (DFS)

Disease free survival (DFS) is defined as the time from documentation of first CR at any time to the first of relapse or death from any cause in participants who achieved a CR. (NCT00651261)
Timeframe: Duration of study (Up to 10 years)

Interventionmonths (Median)
Induction and Consolidation Chemotherapy Plus Midostaurin26.7
Induction and Consolidation Chemotherapy Plus Placebo15.5

Event- Free Survival

"Event free survival (EFS) was defined as the time from randomization until the earliest qualifying event, including: failure to obtain a CR on or before 60 days of initiation of protocol therapy; relapse; or death from any cause. Patients alive and event free at the time of analysis were censored on the date of last clinical assessment. The median EFS with 95% CI was estimated using the Kaplan-Meier method.~Due to a higher than expected transplant rate, EFS was promoted to be a key secondary endpoint." (NCT00651261)
Timeframe: Duration of study (Up to 10 years)

Interventionmonths (Median)
Induction and Consolidation Chemotherapy Plus Midostaurin8.2
Induction and Consolidation Chemotherapy Plus Placebo3.0

Overall Survival (OS)

Overall survival (OS) was defined as the time interval from randomization to death from any cause. The median OS with 95% CI was estimated using the Kaplan-Meier method. (NCT00651261)
Timeframe: Duration of study (Up to 10 years)

Interventionmonths (Median)
Induction and Consolidation Chemotherapy Plus Midostaurin74.7
Induction and Consolidation Chemotherapy Plus Placebo25.6

Overall Survival, Censoring Participants Who Receive a Stem Cell Transplant at the Time of the Transplant

Overall survival (OS) was defined as the time interval from randomization to death from any cause. Any participants who received a stem cell transplant were censored at the time of transplant. The median OS with 95% CI was estimated using the Kaplan-Meier method. (NCT00651261)
Timeframe: Duration of study (Up to 10 years)

Interventionmonths (Median)
Induction and Consolidation Chemotherapy Plus MidostaurinNA
Induction and Consolidation Chemotherapy Plus PlaceboNA

Overall Response (CR for ALL Patients), (CR + CRp for AML Patients)

"Overall response for ALL patients: CR - complete remission (attainment of an M1 bone marrow (< 5% blasts) with no evidence of circulating blasts or extramedullary disease and with recovery of peripheral counts (absolute neutrophil count (ANC) > 750/μL and platelet count > 75,000/μL).~Overall response for AML patients: (CR + CRp), defined as:~CR - complete remission (attainment of an M1 bone marrow (<5% blasts) with no evidence of circulating blasts or extramedullary disease and with recovery of peripheral blood counts (absolute neutrophil count (ANC) > 1000/uL and platelet count > 100,000/uL)) or CRp - remission without platelet recovery (Attainment of an M1 bone marrow (<5% blasts) with no evidence of circulating blasts or extramedullary disease and with recovery of absolute neutrophil count (ANC) > 1000/uL and platelet transfusion independence (defined as: no platelet transfusions x 1 week))." (NCT00372619)
Timeframe: 2 cycles or up to 84 days

Interventionparticipants (Number)
Clofarabine 40 mg/m² to Assess Feasibility in ALL Patients.1
Clofarabine 40 mg/m² to Assess Feasibility in AML Patients.2
Clofarabine 52 mg/m² to Assess Feasibility in ALL Patients.0
Clofarabine 52 mg/m² to Assess Efficacy in ALL Patients.2
Clofarabine 52 mg/m² to Assess Feasibility in AML Patients.2
Clofarabine 52 mg/m² to Assess Efficacy in AML Patients19
Clofarabine 52 mg/m² to Assess Efficacy - Ambiguous Lineage pt2

Safety and Tolerability as Measured by CTCAE v3.0

Number of participants with at least one grade 3 or higher adverse event during therapy. (NCT00372619)
Timeframe: End of therapy

Interventionnumber participants (Number)
Clofarabine 40 mg/m² to Assess Feasibility in ALL Patients.7
Clofarabine 40 mg/m² to Assess Feasibility in AML Patients.2
Clofarabine 52 mg/m² to Assess Feasibility in ALL Patients.3
Clofarabine 52 mg/m² to Assess Efficacy in ALL Patients.10
Clofarabine 52 mg/m² to Assess Feasibility in AML Patients.7
Clofarabine 52 mg/m² to Assess Efficacy in AML Patients35
Clofarabine 52 mg/m² to Assess Efficacy - Ambiguous Lineage pt2

Median Event-Free Survival (EFS)

Event-free survival (EFS) defined as time from start of treatment to first documentation of disease relapse or death. Bayesian time-to-event model will be used to monitor progression free survival. (NCT01025154)
Timeframe: 2 years

InterventionMonths (Median)
Clofarabine, Cytarabine + Idarubicin13.5

Overall Response: Number of Participants With Complete Remission or Complete Remission Without Platelet Recovery

Overall Response (CR+CRp) defined as Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x 10^9/L and platelet count > 100 x 10^9/L, and normal bone marrow differential (< 5% blasts); and, Complete Remission without Platelet Recovery (CRp): Peripheral blood and bone marrow results as for CR, but with platelet counts of < 100 x 10^9/L. Response evaluated within 8 weeks after induction therapy. (NCT01025154)
Timeframe: 8 weeks after Induction therapy (induction cycle 4-6 weeks)

Interventionparticipants (Number)
Complete RemissionComplete Remission without Platelet Recovery
Clofarabine, Cytarabine + Idarubicin423

Intensive Study: Overall Survival (OS)

OS is defined as the time from the date of randomization to the date of death due to any cause. Participants last known to be alive were to be censored at the date of last contact. (NCT03416179)
Timeframe: Baseline up to 25 months

Interventionmonths (Median)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin17.3
Intensive Study: Placebo + Cytarabine + Daunorubicin20.4

Intensive Study: Percentage of Participants Who Improved in Fatigue Score Measured by the MD Anderson Symptom Inventory -Acute Myelogenous Leukemia/Myelodysplastic Syndrome (MDASI-AML/MDS) Questionnaire

"MDASI-AML/MDS: consists of 23 items, 13-item core cancer symptoms (pain, fatigue, nausea, disturbed sleep, distress, shortness of breath, problem remembering, lack of appetite, drowsiness, dry mouth, sadness, vomiting, and numbness), 4-item AML/MDS specific symptoms (malaise, diarrhea, muscle weakness, and skin problems), and 6 areas of interference (general activity, mood, work, walking, relations with other people, and enjoyment of life). Fatigue was measured at the participants' worst level in last 24 hours by asking participants to respond to each item on an 0-10 numeric rating scale (NRS), where 0 = not present and 10 = as bad as you can imagine. Percentage of participants who had improvement in fatigue symptoms reported in this outcome measure." (NCT03416179)
Timeframe: Post-baseline up to Week 8

InterventionPercentage of participants (Number)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin17.41
Intensive Study: Placebo + Cytarabine + Daunorubicin17.24

Intensive Study: Percentage of Participants With Complete Remission Including Negative Minimal Residual Disease (CRMRD-neg)

CR was defined based on 2017 ELN recommendations. CR: Bone marrow blasts <5%; absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; ANC >=1.0*10^9/L; platelet count >=100*10^9/L. CRMRDneg: CR with negativity for a genetic marker by RT-qPCR, or CR with negativity by MFC. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

InterventionPercentage of participants (Number)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin49.3
Intensive Study: Placebo + Cytarabine + Daunorubicin47.3

Intensive Study: Percentage of Participants With Complete Remission With Incomplete Hematologic Recovery (CRi)

CR was defined based on 2017 ELN recommendations. CR: MRD (positive or unknown), bone marrow blasts <5%; absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; ANC less than (<) 1.0*10^9/L; platelet count <100 × 10^9/L. CRi (included CR [includes CRMRD-neg]): not qualifying for CR, neutropenia <1.0*10^9/L or platelets <100*10^9, absence of extramedullary disease, and absence of blasts with Auer rods. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

InterventionPercentage of participants (Number)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin1.5
Intensive Study: Placebo + Cytarabine + Daunorubicin5.4

Intensive Study: Percentage of Participants With Complete Remission Without Negative Minimal Residual Disease (CRMRD-neg)

Complete remission (CR) was defined based on 2017 European LeukemiaNet (ELN) recommendations. CR: Bone marrow blasts <5 percentage (%); absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; absolute neutrophil count (ANC) greater than equal to (>=) 1.0*10^9/Liter (L); platelet count >=100*10^9/L. CRMRD-neg: CR with negativity for a genetic marker by reverse transcription quantitative polymerase chain reaction (RT-qPCR), or CR with negativity by Multiparameter Flow Cytometry (MFC). (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

InterventionPercentage of participants (Number)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin5.0
Intensive Study: Placebo + Cytarabine + Daunorubicin5.4

Intensive Study: Percentage of Participants With Morphologic Leukemia-free State (MLFS)

MLFS was defined based on 2017 ELN recommendations. MLFS: MRD (positive or unknown), bone marrow blasts <5%, no hematologic recovery required, marrow should not be aplastic, at least 200 cells enumerated or cellularity absence of extramedullary disease >=10%, and absence of blasts with Auer rods. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

InterventionPercentage of participants (Number)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin1.5
Intensive Study: Placebo + Cytarabine + Daunorubicin2.0

Intensive Study: Percentage of Participants With Partial Remission (PR)

PR was defined based on 2017 ELN recommendations. PR: MRD (positive or unknown); bone marrow blasts - decrease to 5-25% and decrease of pre-treatment bone marrow blast percentage by at least 50%; neutrophil count >=1.0*10^9/L; and platelets count >=100*10^9/L. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

InterventionPercentage of participants (Number)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin5.0
Intensive Study: Placebo + Cytarabine + Daunorubicin4.4

Non-intensive Study: Overall Survival (OS)

OS is defined as the time from the date of randomization to the date of death due to any cause. Participants last known to be alive were to be censored at the date of last contact. (NCT03416179)
Timeframe: Baseline up to 25 months

Interventionmonths (Median)
Non-intensive Study: Glasdegib + Azacitidine10.3
Non-intensive Study: Placebo + Azacitidine10.6

Non-intensive Study: Percentage of Participants Who Improved in Fatigue Score Measured by the MDASI-AML/MDS Questionnaire at Week 12

"MDASI-AML/MDS: consists of 23 items, 13-item core cancer symptoms (pain, fatigue, nausea, disturbed sleep, distress, shortness of breath, problem remembering, lack of appetite, drowsiness, dry mouth, sadness, vomiting, and numbness), 4-item AML/MDS specific symptoms (malaise, diarrhea, muscle weakness, and skin problems), and 6 areas of interference (general activity, mood, work, walking, relations with other people, and enjoyment of life). Fatigue was measured at the participants' worst level in last 24 hours by asking participants to respond to each item on an 0-10 NRS, where 0 = not present and 10 = as bad as you can imagine. Percentage of participants who had improvement in fatigue symptoms reported in this outcome measure." (NCT03416179)
Timeframe: Post-baseline up to Week 12

InterventionPercentage of participants (Number)
Non-intensive Study: Glasdegib + Azacitidine11.66
Non-intensive Study: Placebo + Azacitidine15.43

Non-intensive Study: Percentage of Participants With Complete Remission (CR) Including Negative Minimal Residual Disease (CRMRD-neg)

CR was defined based on 2017 ELN recommendations. CR: Bone marrow blasts <5%; absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; ANC >=1.0*10^9/L; platelet count >=100*10^9/L. CRMRDneg: CR with negativity for a genetic marker by RT-qPCR, or CR with negativity by MFC. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

InterventionPercentage of participants (Number)
Non-intensive Study: Glasdegib + Azacitidine19.6
Non-intensive Study: Placebo + Azacitidine13.0

Non-intensive Study: Percentage of Participants With Complete Remission With Incomplete Hematologic Recovery (CRi)

CR was defined based on 2017 ELN recommendations. CR: MRD (positive or unknown), bone marrow blasts <5%; absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; ANC <1.0*10^9/L; platelet count <100 × 10^9/L. CRi (included CR [includes CRMRD-neg]): not qualifying for CR, neutrophil <0.5*10^9/L or platelets <50*10^9, absence of extramedullary disease, and absence of blasts with Auer rods. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

InterventionPercentage of participants (Number)
Non-intensive Study: Glasdegib + Azacitidine2.5
Non-intensive Study: Placebo + Azacitidine4.9

Non-intensive Study: Percentage of Participants With Complete Remission With Partial Hematologic Recovery (CRh)

CRh: MRD (positive or unknown); bone marrow blasts <5%; assessed in non-intensive chemotherapy study only, not qualifying for CR, ie, both neutrophil >=0.5*10^9/L and platelets >=50*10^9/L must be met, but does not satisfy both neutrophils >=1*10^9/L and platelets >=100*10^9/L at the same time; absence of extramedullary disease; and absence of blasts with Auer rods. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

InterventionPercentage of participants (Number)
Non-intensive Study: Glasdegib + Azacitidine3.1
Non-intensive Study: Placebo + Azacitidine3.1

Non-intensive Study: Percentage of Participants With Complete Remission Without Negative Minimal Residual Disease (CRMRD-neg)

CR was defined based on 2017 ELN recommendations. CR: Bone marrow blasts <5%; absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; ANC >=1.0*10^9/L; platelet count >=100*10^9/L. CRMRDneg: CR with negativity for a genetic marker by RT-qPCR, or CR with negativity by MFC. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

InterventionPercentage of participants (Number)
Non-intensive Study: Glasdegib + Azacitidine1.8
Non-intensive Study: Placebo + Azacitidine0.6

Non-intensive Study: Percentage of Participants With Morphologic Leukemia-free State (MLFS)

MLFS was defined based on 2017 ELN recommendations. MLFS: MRD (positive or unknown), bone marrow blasts <5%, no hematologic recovery required, marrow should not be aplastic, at least 200 cells enumerated or cellularity absence of extramedullary disease >=10%, and absence of blasts with Auer rods. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

InterventionPercentage of participants (Number)
Non-intensive Study: Glasdegib + Azacitidine3.1
Non-intensive Study: Placebo + Azacitidine0.6

Non-intensive Study: Percentage of Participants With Partial Remission (PR)

PR was defined based on 2017 ELN recommendations. PR: MRD (positive or unknown); bone marrow blasts - decrease to 5-25% and decrease of pre-treatment bone marrow blast percentage by at least 50%; neutrophil count >=1.0*10^9/L; and platelets count >=100*10^9/L. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

InterventionPercentage of participants (Number)
Non-intensive Study: Glasdegib + Azacitidine2.5
Non-intensive Study: Placebo + Azacitidine4.9

Intensive Study: Number of Participants With Adverse Events (AEs),Serious Adverse Events (SAEs) and According to Severity AEs Graded by NCI CTCAE v.4.03

AE: any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. SAE: any AE, regardless of dose, that led to death; was life-threatening; required hospitalization or prolonged hospitalization; led to persistent or significant incapacity or led to congenital anomaly or birth defect. AEs included SAEs and all non-SAEs. NCI CTCAE v.4.03, Grade 3=severe adverse event, Grade 4= life threatening consequences; urgent intervention indicated, Grade 5= death related to adverse event. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

,
InterventionParticipants (Count of Participants)
AEsSAEsGrade 3 or 4 AEGrade 5 AE
Intensive Study: Glasdegib + Cytarabine + Daunorubicin1968617316
Intensive Study: Placebo + Cytarabine + Daunorubicin1989216920

Intensive Study: Number of Participants With Shift From Baseline in Chemistry Laboratory Abnormalities Graded by NCI CTCAE v.4.03

Chemistry laboratory test included: alanine aminotransferase increased, alkaline phosphatase increased, aspartate aminotransferase increased, blood bilirubin increased, chronic kidney disease, creatine phosphokinase (CPK) increased, creatinine increased, gamma glutamyl transferase (GGT) increased, hypercalcemia, hyperglycemia, hyperkalemia, hypermagnesemia, hypernatremia, hypoalbuminemia, hypocalcemia, hypoglycemia, hypokalemia, hypomagnesemia, hyponatremia, and hypophosphatemia. Laboratory results were categorically summarized according to the NCI-CTCAE criteria v4.03. Grade 1= mild; Grade 2= moderate; Grade 3= severe and Grade 4= life-threatening or disabling. Number of participants with shift from baseline for chemistry laboratory test were assessed. Only those laboratory test parameters in which at least 1 participant had data were reported. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

,
InterventionParticipants (Count of Participants)
Alanine aminotransferase increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Alanine aminotransferase increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Alanine aminotransferase increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Alanine aminotransferase increased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Alkaline phosphatase increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Alkaline phosphatase increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Alkaline phosphatase increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Aspartate aminotransferase increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Aspartate aminotransferase increased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Aspartate aminotransferase increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Aspartate aminotransferase increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Blood bilirubin increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Blood bilirubin increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Blood bilirubin increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Blood bilirubin increased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Chronic kidney disease: Missing (baseline grade) to Grade <=2 (CTCAE grade)Chronic kidney disease: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Chronic kidney disease: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Chronic kidney disease: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Chronic kidney disease: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)CPK increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)CPK increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)CPK increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Creatinine increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Creatinine increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)GGT increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)GGT increased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)GGT increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypercalcemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypercalcemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hyperglycemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hyperglycemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hyperglycemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hyperglycemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hyperglycemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hyperkalemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hyperkalemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hyperkalemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hypermagnesemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypermagnesemia: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Hypermagnesemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypermagnesemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypermagnesemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hypernatremia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypernatremia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypernatremia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hypoalbuminemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypoalbuminemia: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Hypoalbuminemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypoalbuminemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypoalbuminemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hypocalcemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypocalcemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypocalcemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypoglycemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypoglycemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypoglycemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypokalemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypokalemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypokalemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hypomagnesemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypomagnesemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypomagnesemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hyponatremia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hyponatremia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hyponatremia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hyponatremia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hypophosphatemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypophosphatemia: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Hypophosphatemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypophosphatemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE gradeHypophosphatemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hypophosphatemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin1175161218725017315018480418281020161218875801191117415221942020190101960001185511190111930155410219011771711311533303
Intensive Study: Placebo + Cytarabine + Daunorubicin118413031923211869318951518840119166019267112519231831011194314118421196113118581518933194115937251870188811311533321

Intensive Study: Number of Participants With Shift From Baseline in Coagulation Laboratory Abnormalities Graded by NCI CTCAE v.4.03

Coagulation laboratory test included: activated partial thromboplastin time prolonged, and INR increased. Laboratory results were categorically summarized according to the NCI-CTCAE criteria v4.03. Grade 0= no abnormality; Grade 1= mild; Grade 2= moderate; Grade 3= severe and Grade 4= life-threatening or disabling. Number of participants with shift from baseline for coagulation were assessed. Only those laboratory test parameters in which at least 1 participant had data were reported. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

,
InterventionParticipants (Count of Participants)
Activated partial thromboplastin time prolonged: Missing (baseline grade) to Grade 0 (CTCAE grade)Activated partial thromboplastin time prolonged: Missing (baseline grade) to Grade 1 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 0 (baseline grade) to Grade 0 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 0 (baseline grade) to Grade 1 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 0 (baseline grade) to Grade 2 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 0 (baseline grade) to Grade 3 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 1 (baseline grade) to Grade 1 (CTCAE grade)INR increased: Missing (baseline grade) to Grade 0 (CTCAE grade)INR increased: Missing (baseline grade) to Grade 1 (CTCAE grade)INR increased: Missing (baseline grade) to Grade 2 (CTCAE grade)INR increased: Grade 0 (baseline grade) to Grade 0 (CTCAE grade)INR increased: Grade 0 (baseline grade) to Grade 1 (CTCAE grade)INR increased: Grade 0 (baseline grade) to Grade 2 (CTCAE grade)INR increased: Grade 0 (baseline grade) to Grade 3 (CTCAE grade)INR increased: Grade 1 (baseline grade) to Grade 0 (CTCAE grade)INR increased: Grade 1 (baseline grade) to Grade 1 (CTCAE grade)INR increased: Grade 1 (baseline grade) to Grade 2 (CTCAE grade)INR increased: Grade 1 (baseline grade) to Grade 3 (CTCAE grade)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin100101110101100011
Intensive Study: Placebo + Cytarabine + Daunorubicin014210111020111310

Intensive Study: Number of Participants With Shift From Baseline in Corrected QT (QTc) Interval

Triplicate 12-lead ECG measurements (each recording separated by approximately 2 minutes) were performed and average was calculated. The time corresponding to beginning of depolarization to repolarization of the ventricles (QT interval) was adjusted for RR interval using QT and RR from each ECG by Fridericia's formula (QTcF = QT divided by cube root of RR) and by Bazette's formula (QTcB = QT divided by square root of RR). Participants with maximum increase from baseline of <=450 to >500 msec (post-baseline) were summarized. Number of participants with shift from baseline for QTc were assessed. Only those QTc parameters in which at least 1 participant had data were reported. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

,
InterventionParticipants (Count of Participants)
QTcB: <=450 msec (baseline) to <=450 msec (post-baseline)QTcB: <=450 msec (baseline) to >450-<=480 msec (post-baseline)QTcB: <=450 msec (baseline) to >480-<=500 msec (post-baseline)QTcB: <=450 msec (baseline) to >500 msec (post-baseline)QTcB: >450-<=480 msec (baseline) to <=450 msec (post-baseline)QTcB: >450-<=480 msec (baseline) to >450-<=480 msec (post-baseline)QTcB: >450-<=480 msec (baseline) to >480-<=500 msec (post-baseline)QTcB: >450-<=480 msec (baseline) to >500 msec (post-baseline)QTcB: >480-<=500 msec (baseline) to >450-<=480 msec (post-baseline)QTcB: >480-<=500 msec (baseline) to >480-<=500 msec (post-baseline)QTcB: >480-<=500 msec (baseline) to >500 msec (post-baseline)QTcB: >500 msec (baseline) to >500 msec (post-baseline)QTcF: <=450 msec (baseline) to <=450 msec (post-baseline)QTcF: <=450 msec (baseline) to >450-<=480 msec (post-baseline)QTcF: <=450 msec (baseline) to >480-<=500 msec (post-baseline)QTcF: <=450 msec (baseline) to >500 msec (post-baseline)QTcF: >450-<=480 msec (baseline) to <=450 msec (post-baseline)QTcF: >450-<=480 msec (baseline) to >450-<=480 msec (post-baseline)QTcF: >450-<=480 msec (baseline) to >500 msec (post-baseline)QTcF: >480-<=500 msec (baseline) to >480-<=500 msec (post-baseline)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin51641811220161121111650852510
Intensive Study: Placebo + Cytarabine + Daunorubicin71631163171050101132391001511

Intensive Study: Number of Participants With Shift From Baseline in Hematological Laboratory Abnormalities Graded by NCI CTCAE v.4.03

Hematology laboratory test included: anemia, hemoglobin increased, international normalized ratio (INR) increased, lymphocyte count decreased, lymphocyte count increased, neutrophil count decreased, platelet count decreased, and white blood cell decreased. Laboratory results were categorically summarized according to the NCI-CTCAE criteria v4.03. Grade 1= mild; Grade 2= moderate; Grade 3= severe and Grade 4= life-threatening or disabling. Number of participants with shift from baseline for hematology laboratory test were assessed. Only those laboratory test parameters in which at least 1 participant had data were reported. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

,
InterventionParticipants (Count of Participants)
Anemia: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Anemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Anemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Anemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Anemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hemoglobin increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hemoglobin increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hemoglobin increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)INR increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count decreased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count decreased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count decreased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count decreased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count decreased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count decreased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count increased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count increased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Neutrophil count decreased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Neutrophil count decreased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Neutrophil count decreased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Neutrophil count decreased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Neutrophil count decreased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Platelet count decreased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Platelet count decreased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Platelet count decreased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)White blood cell decreased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)White blood cell decreased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)White blood cell decreased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)White blood cell decreased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)
Intensive Study: Glasdegib + Cytarabine + Daunorubicin0271134520194230021160111018561104850105198971315537
Intensive Study: Placebo + Cytarabine + Daunorubicin2141035732194112426152096180430427921062100954215635

Intensive Study: Number of Participants With Treatment Related Adverse Events (AEs) and Serious Adverse Events (SAEs) Graded by NCI CTCAE v.4.03

A treatment-related AE was any untoward medical occurrence attributed to the study drug in a participant who received study drug. SAE: any AE, regardless of dose, that led to death; was life-threatening; required hospitalization or prolonged hospitalization; led to persistent or significant incapacity or led to congenital anomaly or birth defect. NCI CTCAE v.4.03, Grade 3=severe adverse event, Grade 4= life threatening consequences; urgent intervention indicated, Grade 5= death related to adverse event. (NCT03416179)
Timeframe: Day 1 up to maximum of 2 years

,
InterventionParticipants (Count of Participants)
Treatment related AEsTreatment related SAEsGrade 3 or 4 AEGrade 5 AE
Intensive Study: Glasdegib + Cytarabine + Daunorubicin181481615
Intensive Study: Placebo + Cytarabine + Daunorubicin188601498

Intensive Study: Plasma Trough Concentration (Ctrough) of Glasdegib

Ctrough of Glasdegib was measured in nanogram per milliliter (ng/mL). (NCT03416179)
Timeframe: Induction, Day 10+/-1: pre-dose, 1, 4 hour (hr); Consolidation phase, Day 1: pre-dose, 1, 4 hr

Interventionng/ml (Geometric Mean)
Induction Day 10Consolidation 1, Day 1Consolidation 2, Day 1
Intensive Study: Glasdegib + Cytarabine + Daunorubicin413.54245.48259.79

Non-intensive Study: Number of Participants With Adverse Events (AEs),Serious Adverse Events (SAEs) and According to Severity AEs Graded by NCI CTCAE v.4.03

AE: any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. SAE: any AE, regardless of dose, that led to death; was life-threatening; required hospitalization or prolonged hospitalization; led to persistent or significant incapacity or led to congenital anomaly or birth defect. AEs included SAEs and all non-SAEs. NCI CTCAE v.4.03, Grade 3=severe adverse event, Grade 4= life threatening consequences; urgent intervention indicated, Grade 5= death related to adverse event. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

,
InterventionParticipants (Count of Participants)
AEsSAEsGrade 3 or 4 AEGrade 5 AE
Non-intensive Study: Glasdegib + Azacitidine16111710650
Non-intensive Study: Placebo + Azacitidine15812410052

Non-intensive Study: Number of Participants With Shift From Baseline in Chemistry Laboratory Abnormalities Graded by NCI CTCAE v.4.03

Chemistry laboratory test included: alanine aminotransferase increased, alkaline phosphatase increased, aspartate aminotransferase increased, blood bilirubin increased, chronic kidney disease, CPK increased, creatinine increased, GGT increased, hypercalcemia, hyperglycemia, hyperkalemia, hypermagnesemia, hypernatremia, hypoalbuminemia, hypocalcemia, hypoglycemia, hypokalemia, hypomagnesemia, hyponatremia, and hypophosphatemia. Laboratory results were categorically summarized according to the NCI-CTCAE criteria v4.03. Grade 1= mild; Grade 2= moderate; Grade 3= severe and Grade 4= life-threatening or disabling. Number of participants with shift from baseline for chemistry laboratory test were assessed. Only those laboratory test parameters in which at least 1 participant had data were reported. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

,
InterventionParticipants (Count of Participants)
Alanine aminotransferase increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Alanine aminotransferase increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Alanine aminotransferase increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Alanine aminotransferase increased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Alkaline phosphatase increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Alkaline phosphatase increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Alkaline phosphatase increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Aspartate aminotransferase increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Aspartate aminotransferase increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Aspartate aminotransferase increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Blood bilirubin increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Blood bilirubin increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Chronic kidney disease: Missing (baseline grade) to Grade <=2 (CTCAE grade)Chronic kidney disease: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Chronic kidney disease: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Chronic kidney disease: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)CPK increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)CPK increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)CPK increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Creatinine increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Creatinine increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)GGT increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)GGT increased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)GGT increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypercalcemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypercalcemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hyperglycemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hyperglycemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hyperglycemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hyperglycemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hyperkalemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hyperkalemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hyperkalemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hyperkalemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hyperkalemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hypermagnesemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypermagnesemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypermagnesemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypernatremia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypernatremia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypoalbuminemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypoalbuminemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypoalbuminemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypocalcemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypocalcemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypocalcemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypocalcemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hypoglycemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypoglycemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypoglycemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypokalemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypokalemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypokalemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypokalemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hypokalemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hypomagnesemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypomagnesemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypomagnesemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hypomagnesemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hyponatremia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hyponatremia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Hyponatremia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hyponatremia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hypophosphatemia: Missing (baseline grade) to Grade <=2 (CTCAE grade)Hypophosphatemia: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Hypophosphatemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hypophosphatemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE gradeHypophosphatemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Hypophosphatemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)
Non-intensive Study: Glasdegib + Azacitidine215260015903152415732130280914711537341215621478221552002155215822151621541021553212923142151601352401311411201
Non-intensive Study: Placebo + Azacitidine0154601157101564159111342328147215555302158413614201573004154215910157321553041560013915244153211421512201381712

Non-intensive Study: Number of Participants With Shift From Baseline in Coagulation Laboratory Abnormalities Graded by NCI CTCAE v.4.03

Coagulation laboratory test included: activated partial thromboplastin time prolonged, and INR increased. Laboratory results were categorically summarized according to the NCI-CTCAE criteria v4.03. Grade 0= no abnormality; Grade 1= mild; Grade 2= moderate; Grade 3= severe and Grade 4= life-threatening or disabling. Number of participants with shift from baseline for coagulation were assessed. Only those laboratory test parameters in which at least 1 participant had data were reported. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

,
InterventionParticipants (Count of Participants)
Activated partial thromboplastin time prolonged: Missing (baseline grade) to Grade 0 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 0 (baseline grade) to Grade 0 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 0 (baseline grade) to Grade 1 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 1 (baseline grade) to Grade 0 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 1 (baseline grade) to Grade 1 (CTCAE grade)Activated partial thromboplastin time prolonged: Grade 2 (baseline grade) to Grade 1 (CTCAE grade)INR increased: Missing (baseline grade) to Grade 0 (CTCAE grade)INR increased: Grade 0 (baseline grade) to Grade 0 (CTCAE grade)INR increased: Grade 0 (baseline grade) to Grade 1 (CTCAE grade)INR increased: Grade 0 (baseline grade) to Grade 2 (CTCAE grade)INR increased: Grade 0 (baseline grade) to Grade 3 (CTCAE grade)INR increased: Grade 1 (baseline grade) to Grade 1 (CTCAE grade)INR increased: Grade 1 (baseline grade) to Grade 2 (CTCAE grade)INR increased: Grade 3 (baseline grade) to Grade 1 (CTCAE grade)
Non-intensive Study: Glasdegib + Azacitidine18511114611121
Non-intensive Study: Placebo + Azacitidine05700005420101

Non-intensive Study: Number of Participants With Shift From Baseline in Corrected QT (QTc) Interval

Triplicate 12-lead ECG measurements (each recording separated by approximately 2 minutes) were performed and average was calculated. The time corresponding to beginning of depolarization to repolarization of the ventricles (QT interval) was adjusted for RR interval using QT and RR from each ECG by Fridericia's formula (QTcF = QT divided by cube root of RR) and by Bazette's formula (QTcB = QT divided by square root of RR). Participants with maximum increase from baseline of <=450 to >500 msec (post-baseline) were summarized. Number of participants with shift from baseline for QTc were assessed. Only those QTc parameters in which at least 1 participant had data were reported. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

,
InterventionParticipants (Count of Participants)
QTcB: <=450 msec (baseline) to <=450 msec (post-baseline)QTcB: <=450 msec (baseline) to >450-<=480 msec (post-baseline)QTcB: <=450 msec (baseline) to >480-<=500 msec (post-baseline)QTcB: <=450 msec (baseline) to >500 msec (post-baseline)QTcB: >450-<=480 msec (baseline) to <=450 msec (post-baseline)QTcB: >450-<=480 msec (baseline) to >450-<=480 msec (post-baseline)QTcB: >450-<=480 msec (baseline) to >480-<=500 msec (post-baseline)QTcB: >450-<=480 msec (baseline) to >500 msec (post-baseline)QTcB: >480-<=500 msec (baseline) to >450-<=480 msec (post-baseline)QTcB: >480-<=500 msec (baseline) to >480-<=500 msec (post-baseline)QTcB: >480-<=500 msec (baseline) to >500 msec (post-baseline)QTcF: <=450 msec (baseline) to <=450 msec (post-baseline)QTcF: <=450 msec (baseline) to >450-<=480 msec (post-baseline)QTcF: <=450 msec (baseline) to >480-<=500 msec (post-baseline)QTcF: >450 - <=480 (baseline) to <=450msec (post-baseline)QTcF: >450 - <=480 msec (baseline) to >450 - <=480msec(post baseline)QTcF: >450 - <=480 msec (baseline) to >480 - <=500 msec (post baseline)QTcF: >480 msec (baseline) to <=500 msec (post baseline)QTcF: >450 - <=480 (baseline) to >500 msec (post baseline)QTcF: >480 - <=500msec (baseline) to >480 - <=500 msec (post baseline)
Non-intensive Study: Glasdegib + Azacitidine485367020135132100397105111
Non-intensive Study: Placebo + Azacitidine584311312283213104360118410

Non-intensive Study: Number of Participants With Shift From Baseline in Hematological Laboratory Abnormalities Graded by NCI CTCAE v.4.03

Hematology laboratory test included: anemia, hemoglobin increased, INR increased, lymphocyte count decreased, lymphocyte count increased, neutrophil count decreased, platelet count decreased, and white blood cell decreased. Laboratory results were categorically summarized according to the NCI-CTCAE criteria v4.03. Grade 1= mild; Grade 2= moderate; Grade 3= severe and Grade 4= life-threatening or disabling. Number of participants with shift from baseline for hematology laboratory test were assessed. Only those laboratory test parameters in which at least 1 participant had data were reported. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

,
InterventionParticipants (Count of Participants)
Anemia: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Anemia: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Anemia: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Anemia: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Hemoglobin increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Hemoglobin increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count decreased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count decreased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count decreased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count decreased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count decreased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count decreased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count increased: Missing (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count increased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Lymphocyte count increased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Lymphocyte count increased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Neutrophil count decreased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)Neutrophil count decreased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Neutrophil count decreased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Neutrophil count decreased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Neutrophil count decreased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)Platelet count decreased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)Platelet count decreased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)Platelet count decreased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)Platelet count decreased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)White blood cell decreased: Missing (baseline grade) to Grade <=2 (CTCAE grade)White blood cell decreased: Missing (baseline grade) to Grade 3/4 (CTCAE grade)White blood cell decreased: Grade <=2 (baseline grade) to Grade <=2 (CTCAE grade)White blood cell decreased: Grade <=2 (baseline grade) to Grade 3/4 (CTCAE grade)White blood cell decreased: Grade 3/4 (baseline grade) to Grade <=2 (CTCAE grade)White blood cell decreased: Grade 3/4 (baseline grade) to Grade 3/4 (CTCAE grade)
Non-intensive Study: Glasdegib + Azacitidine2997529160021965127314772413482922155183114370045
Non-intensive Study: Placebo + Azacitidine41871311591008954413015730022401972864068004862148

Non-intensive Study: Number of Participants With Treatment Related Adverse Events (AEs) and Serious Adverse Events (SAEs) Graded by NCI CTCAE v.4.03

A treatment-related AE was any untoward medical occurrence attributed to the study drug in a participant who received study drug. SAE: any AE, regardless of dose, that led to death; was life-threatening; required hospitalization or prolonged hospitalization; led to persistent or significant incapacity or led to congenital anomaly or birth defect. NCI CTCAE v4.03, Grade 3=severe adverse event, Grade 4= life threatening consequences; urgent intervention indicated, Grade 5= death related to adverse event. (NCT03416179)
Timeframe: Day 1 up to maximum of 3 years

,
InterventionParticipants (Count of Participants)
Treatment related AEsTreatment related SAEsGrade 3 or 4 AEGrade 5 AE
Non-intensive Study: Glasdegib + Azacitidine13345974
Non-intensive Study: Placebo + Azacitidine12337724

Non-intensive Study: Plasma Trough Concentration (Ctrough) of Glasdegib

Ctrough of Glasdegib was measured in ng/mL. (NCT03416179)
Timeframe: Pre-dose: Cycle 1 Day 15 and Cycle 2 Day 1

Interventionng/ml (Geometric Mean)
Cycle 1 Day 15Cycle 2 Day 1
Non-intensive Study: Glasdegib + Azacitidine565.44472.42

Non-intensive Study: Time to Response

TTRi:Participants who achieved CRi or better, as the time from date of randomization to first documentation of response(CRi or better).TTRh:Participants who achieved CRh or better, as the time from date of randomization to first documentation of response(CRh or better). CRi:not qualifying for CR, neutrophil<0.5*10^9/L or platelets<50*10^9, absence of extramedullary disease,absence of blasts with Auer rods. CR was defined based on 2017 ELN recommendations. CR: Bone marrow blasts <5 %; absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; ANC >=1.0*10^9/L; platelet count >=100*10^9/L. CRh: MRD (positive or unknown); bone marrow blasts <5%; assessed in non-intensive chemotherapy study only, not qualifying for CR, ie, both neutrophil >=0.5*10^9/L and platelets >=50*10^9/L must be met, but does not satisfy both neutrophils >=1*10^9/L and platelets >=100*10^9/L at the same time; absence of extramedullary disease; and absence of blasts with Auer rods. (NCT03416179)
Timeframe: From the date of randomization to the first documentation of response (CRi/CRh or better) (maximum up to 3 years)

,
InterventionMonths (Mean)
TTRiTTRh
Non-intensive Study: Glasdegib + Azacitidine4.0574.334
Non-intensive Study: Placebo + Azacitidine4.0934.146

Overall Survival

Overall survival time is defined as time from randomization to the date of death from any cause or last known follow-up (censored). Overall survival rates are estimated by the Kaplan-Meier method. (NCT01399372)
Timeframe: From randomization to last follow-up. Maximum follow-up at time of analysis was 7.3 years.

Interventionyears (Median)
ChemotherapyNA
Chemotherapy + Low-Dose WBRTNA

Percentage of Participants Experiencing Partial Response or Complete Response

Response was evaluated using the international criteria proposed by the International Primary Central Nervous System Lymphoma (PCNSL) Study Group criteria. Complete Response was defined as no contrast brain enhancement on magnetic resonance imaging (MRI), no corticosteroid use for at least 2 weeks, a normal eye exam, and negative CSF cytology. Partial response was defined as at least 50% decrease in enhancing tumor on MRI compared with baseline imaging, no or minor retinal pigment epithelium (RPE) abnormality, and negative CSF cytology. (NCT01399372)
Timeframe: After 4th cycle of chemotherapy, approximately 4 months after randomization.

Interventionpercentage of participants (Number)
Chemotherapy83.3
Chemotherapy + Low-Dose WBRT80.6

Percentage of Participants With Neurocognitive Failure

Neurocognitive failure is defined as cognitive failure on two or more of the following tests: Hopkins Verbal Learning Test - Revised (HVLT-R) Free Recall, HVLT-R Delayed Recall, HVLT-R Delayed Recognition, Trail Making Test Part A, Trail Making Test Part B, and Controlled Oral Word Association. Cognitive failure for each test is defined as a change from baseline in raw score (post baseline score - baseline score) at or exceeding the minimally important difference reported in the literature [determined by the reliable change index (RCI) method] of -5, -3, -2,12, 26, and -12, respectively, indicating a worsening of neurocognitive function. Time to neurocognitive failure is defined as time from randomization to date of first failure, last known follow-up (censored), or death without failure (competing risk). Failure rates are estimated using the cumulative incidence method and the distributions of failure times are compared between the arms. Two-year estimates are provided here. (NCT01399372)
Timeframe: Neurocognitive function tests were administered at baseline, after cycle 4 of treatment (4 months), then every six months after end of treatment (7 months) for five years. Two-year rates are provided here.

Interventionpercentage of participants (Number)
Chemotherapy31.6
Chemotherapy + Low-Dose WBRT17.9

Progression-free Survival

Progression is defined as any of the following: more than 25% increase in the contrast-enhancing lesion seen on magnetic resonance imaging (MRI) compared with baseline or best response; new site of disease (central nervous system or systemic); recurrent or new ocular disease; recurrent or positive cerebrospinal fluid (CSF) cytology. Progression-free survival time is defined as time from randomization to the date of first progression, death, or last known follow-up (censored). Progression-free survival rates are estimated using the Kaplan-Meier method. (NCT01399372)
Timeframe: From randomization to last follow-up. Maximum follow-up at time of analysis was 7.3 years.

Interventionyears (Median)
Chemotherapy2.1
Chemotherapy + Low-Dose WBRTNA

Global Heath Status (GHS) Score of the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30)

"Global Health Status is calculated from two questions on the EORTC QLQ-C30. The question responses range from 1 very poor to 7 excellent such that a higher response indicates better quality of life (QOL). The mean of these responses is linearly transformed to a range of 0 (worst) to 100 (best). Data through five years after end of protocol treatment is included in the analysis, while summary data is provided only through three years due to very few participants after that timepoint." (NCT01399372)
Timeframe: EORTC QLQ-C30 was administered at baseline, after cycle 4 of treatment (4 months), then every six months after end of treatment (7 months) for five years.

,
Interventionscore on a scale (Mean)
BaselineEnd of cycle 46 Months Post-Treatment12 Months Post-Treatment18 Months Post-Treatment24 Months Post-Treatment30 Months Post-Treatment36 Months Post-Treatment42 Months Post-Treatment48 Months Post-Treatment54 Months Post-Treatment60 Months Post-Treatment
Chemotherapy62.7867.6774.5478.4776.7970.8377.0875.0075.0079.7670.2480.56
Chemotherapy + Low-Dose WBRT54.9064.5875.3578.9976.6783.3379.6385.0077.5684.3891.6783.33

Number of Participants With a Response

Response - Complete remission (CR): Normalization peripheral blood & bone marrow 5% or & platelet count >100x10^9/L; CR with incomplete platelet recovery (CRp): CR but platelet count <100x10^9/L. CR with incomplete recovery (CRi): CR but platelet count <100x10^9/L or absolute neutrophil count < 1x10^9/L. Partial response (PR): As above except for presence of 6-25% marrow blasts. Lymphoblastic lymphoma (& ALL subtypes with extramedullary disease): CR - disappearance all known disease. PR - >50% decrease in tumor size using sum of product, includes 50% volume decrease in lesions measurable in 3 dimensions. No Response (NR) - No significant change (includes stable disease). Lesions decreased size but <50% or lesions with slight enlargement <25% increase in size. Progressive Disease (PD): Appearance new lesions, 25% or > increase in size existing lesions (>50% if 1 lesion & <2). (NCT00671658)
Timeframe: Response assessed following first 21 day course up to end of treatment with 8 cycles, up to 210 days

InterventionParticipants (Number)
Complete Response (CR)Complete Response without Platelet RecoveryPartial Response (PR)
HYPER-CVAD19834

Immunogenicity

Number of Patients with Positive Immunogenicity tests (NCT00671034)
Timeframe: 25 Days Post-dose (Day 29)

InterventionParticipants (Count of Participants)
Arm I (Calaspargase Pegol 2100)2
Arm II (Calaspargase Pegol 2500)2
Arm III (Pegaspargase 2500)4

Percentage of Participants With Complete Remission at the End of Induction

Complete Remission (CR) rate; where CR is defined as M1 marrow (< 5% lymphoblasts in the bone marrow) (NCT00671034)
Timeframe: End of induction (Day 29)

InterventionPercentage of participants (Number)
Arm I (Calaspargase Pegol 2100)92.4
Arm II (Calaspargase Pegol 2500)97.6
Arm III (Pegaspargase 2500)94.1

Percentage of Participants With Event-free Survival (EFS)

Percentage of participants who were event free. Event Free Probability defined as time from randomization at study entry to first event (induction failure, induction death, relapse, second malignant neoplasm, remission death) or date of last contact for subjects who are event-free. (NCT00671034)
Timeframe: 5 Years

InterventionPercentage of participants (Number)
Arm I (Calaspargase Pegol 2100)72.35
Arm II (Calaspargase Pegol 2500)80.8
Arm III (Pegaspargase 2500)79.34

Percentage of Participants With Minimal Residual Disease (MRD)<0.01% at the End of Induction

Percentage of participants with Negative MRD (MRD<0.01%). (NCT00671034)
Timeframe: End of induction (Day 29)

InterventionPercentage of participants (Number)
Arm I (Calaspargase Pegol 2100)65.2
Arm II (Calaspargase Pegol 2500)81
Arm III (Pegaspargase 2500)72.5

Asparaginase Level

The proportion of patients with an asparaginase level of at least 0.1 IU/mL and the proportion with at least 0.4 IU/mL on Days 4, 15, 22 and 29 of Induction compared to Oncaspar (NCT00671034)
Timeframe: Days 4, 15, 22 and 29 of Induction

,,
Interventionpercentage of patients (Number)
Level at least 0.1 IU/mL day 4Level at least 0.1 IU/mL day 15Level at least 0.1 IU/mL day 22Level at least 0.1 IU/mL day 29Level at least 0.4 IU/mL day 4Level at least 0.4 IU/mL day 15Level at least 0.4 IU/mL day 22Level at least 0.4 IU/mL day 29
Arm I (Calaspargase Pegol 2100)098.498.294.9075.837.513.6
Arm II (Calaspargase Pegol 2500)010010095.0095.062.527.5
Arm III (Pegaspargase 2500)010095.128.6093.014.60

Pharmacodynamics (PD)

Plasma Asparaginase Concentration During consolidation and induction. (NCT00671034)
Timeframe: Day 29 of consolidation and induction

,,
InterventionmIU/mL (Median)
Plasma Asparaginase Concentration- ConsolidationPlasma Asparaginase Concentration- Induction
Arm I (Calaspargase Pegol 2100)575.9271.6
Arm II (Calaspargase Pegol 2500)617.2339.6
Arm III (Pegaspargase 2500)562.172.8

Pharmacokinetics (PK) (Half-life of SC-PEG E. Coli L-asparaginase (EZN-2285) Compared to Pegaspargase During Induction and Consolidation Therapy)

Mean half-life of plasma asparaginase during consolidation and Induction; half-life is defined as the time taken for drug concentration to decrease by half. (NCT00671034)
Timeframe: Post Day 29 of Induction and Post Day 22 of Consolidation

,,
Interventionhours (Mean)
Asparaginase half-life during ConsolidationAsparaginase half-life during Induction
Arm I (Calaspargase Pegol 2100)415.8305.1
Arm II ( Calaspargase Pegol 2500)355.9321.5
Arm III (Pegaspargase 2500)117.2126.9

Plasma and CSF Concentrations of Asparagine in ug/ml

The plasma and CSF concentrations of asparagine in ug/ml after administration of EZN-2285 compared to Oncaspar. (NCT00671034)
Timeframe: 25 Days Post-dose (Day 29)

,,
Interventionug/mL (Mean)
CSF asparagine concentration (ug/mL)Plasma asparagine concentration (ug/mL)
Arm I (Calaspargase Pegol 2100)0.20.2
Arm II (Calaspargase Pegol 2500)0.190.25
Arm III (Pegaspargase 2500)0.260.83

Toxicities During Post Induction Intensification Therapy (All Grades)

The calculation of AE incidence will be based on the number of patients per AE category. For each patient who has multiple AEs classified to the same category, that patient will be tabulated under the worst toxicity grade for that AE category. The incidence of AEs will be tabulated by treatment arm and by organ class. Special attention will be paid to hypersensitivity, pancreatitis, coagulopathy, infection, neurologic dysfunction and thromboembolic events. (NCT00671034)
Timeframe: Up to 5 years

,,
InterventionPercentage of participants (Number)
Alergic Reaction - ConsolidationAlergic Reaction - Delayed Intensification IAlergic Reaction - Interim Maintenance ICNS - ConsolidationCNS - Delayed Intensification ICNS - Interim Maintenance IHyperbilirubinemia - ConsolidationHyperbilirubinemia - Delayed Intensification IHyperbilirubinemia - Interim Maintenance IHyperglycemia - ConsolidationHyperglycemia - Delayed Intensification IHyperglycemia - Interim Maintenance IHyperlipidemia - ConsolidationHyperlipidemia - Delayed Intensification IHyperlipidemia - Interim Maintenance I% patients w/INR increase - Consolidation% pts w/INR increase - Delayed Intensification I% patients w/INR increase - Interim Maintenance IPancreatitis - ConsolidationPancreatitis -Delayed Intensification IPancreatitis - Interim Maintenance I% pts w/prolongation of APT time - Consolidation% pts w/prolongation APT time -Delayed Intension I%pts w/prolongation APT time-Interim maintenance IThrombosis - ConsolidationThrombosis - Delayed Intensification IThrombosis - Interim Maintenance I
Arm I (Calaspargase Pegol 2100)20.44.40.00.00.00.053.128.941.344.944.434.82.02.22.26.16.72.210.22.22.28.28.96.50.02.20.0
Arm II (Calaspargase Pegol 2500)27.30.00.00.03.80.045.538.527.642.461.544.83.00.03.43.03.80.06.17.73.49.126.96.90.00.00.0
Arm III (Pegaspargase 2500)23.30.02.10.02.60.030.210.533.346.536.833.37.00.02.67.00.02.67.00.02.67.018.47.72.30.00.0

Dose Limiting Toxicity

Number of participants with dose limiting toxicity. (NCT00666588)
Timeframe: During Course 1

Interventionparticipants (Number)
Bortezomib 1.3mg/m2-assess Efficacy-low Anthracycline Exposure0
Bortezomib 1.0mg/m2-assess Feasibility High Anthracycline Exp1
Bortezomib 1.3 mg/m2-assess Feasibility High Anthracycline Exp0
Bortezomib 1.3 mg/m2-assess Efficacy High Anthracycline Exp0

Overall Response (Complete Remission [CR] and CR With Partial Recovery [CRp]) During Course 1

Overall response (complete remission [CR] and CR with partial recovery [CRp]) during course 1. (NCT00666588)
Timeframe: After course 1

Interventionparticipants (Number)
Bortezomib 1.3mg/m2-assess Efficacy-low Anthracycline Exposure4
Bortezomib 1.0mg/m2-assess Feasibility High Anthracycline Exp2
Bortezomib 1.3 mg/m2-assess Feasibility High Anthracycline Exp2
Bortezomib 1.3 mg/m2-assess Efficacy High Anthracycline Exp9

Feasibility of Stem Cell Quantitation: Percentage of Leukemia Initiation Cells (LIC) Depletion

Descriptive statistics to assess mean +/- standard deviation for the percentage leukemia initiation cells (LIC) depletion. (NCT00666588)
Timeframe: At baseline and after completion of course 1

,,
Interventionpercentage of LIC depletion (Mean)
Prior to TreatmentPost Treatment
Bortezomib 1.3 mg/m2-assess Efficacy High Anthracycline Exp1.63850.0435
Bortezomib 1.3 mg/m2-assess Feasibility High Anthracycline Exp0.20.43
Bortezomib 1.3mg/m2-assess Efficacy-low Anthracycline Exposure0.0136670.021

NF-kB Activity by Enzyme-linked Immunosorbent Assay (ELISA)

NF-kB activity will be measured as a continuous variable (ng NF-kB/Mg protein). Differences in NF-κB activity between time points will be assessed using summary statistics such as mean, standard deviation, and range. We may perform exploratory analyses to determine if single time point measurements, or the difference between time points, correlate with treatment response. (NCT00666588)
Timeframe: At baseline, prior to and up to 24 hours after bortezomib treatment

,,,
Interventionng/Mg protein (Mean)
Baseline24 Hrs after treatment
Bortezomib 1.0mg/m2-assess Feasibility High Anthracycline Exp655.4345.46
Bortezomib 1.3 mg/m2-assess Efficacy High Anthracycline Exp408.144497.31
Bortezomib 1.3 mg/m2-assess Feasibility High Anthracycline Exp974.66855.96
Bortezomib 1.3mg/m2-assess Efficacy-low Anthracycline Exposure718.218774.1925

Proteasome Inhibition Activity

Mean and standard deviation of β1 and β5- Results are ratios (proteasome subunit/β-actin based on loading of 15 µg total protein, normalized to CEM). (NCT00666588)
Timeframe: At baseline

,,
Interventionratio (Mean)
Baseline β1Baseline β5
Bortezomib 1.0mg/m2-assess Feasibility High Anthracycline Exp0.78940.2576
Bortezomib 1.3 mg/m2-assess Efficacy High Anthracycline Exp0.28951430.3721286
Bortezomib 1.3mg/m2-assess Efficacy-low Anthracycline Exposure0.081150.121575

Complete Remission (CR) Rate for First 60 Participants

All responses were defined as per IWG criteria (2003) where CR Rate defined as number of participants with CR out of total treated participants. Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 times 10^9/L and platelet count > 100 times 10^9/L, and normal bone marrow differential (< 5% blasts). Bone marrow aspirate and/or biopsy starting on day 21 (+/- 7 days) of therapy. (NCT00778375)
Timeframe: Evaluation following two 10 day cycles on day 21 of therapy, continuing up to 210 days

InterventionPercentage of Participants (Number)
Clofarabine + Cytarabine + Decitabine58

Disease-free (DFS) or Relapse-free Survival (RFS) Time

Disease (DFS) or Relapse-free survival (RFS): Time from date of treatment start until the date of first objective documentation of disease-relapse; Bone marrow aspirate and/or biopsy starting on day 21 (+/- 7 days) of therapy and then every 2 weeks (+/- 7 days) as required by leukemia evolution until remission or non-response. Among participants who achieved CR or CRp, RFS was defined as the time interval between the date of response (ie CR or CRp) and the date of relapse or date of death, whichever occurs first. CR or CRp participants who were alive and relapse-free were censored at the off-study date. Full range reflects time to disease progression only, therefore does not reflect a lesser survival time due to other reasons than disease progression/relapse. (NCT00778375)
Timeframe: Evaluated from treatment date until date of disease progression/relapse, followed for 5 years/60 months.

InterventionMonths (Median)
Clofarabine + Cytarabine + Decitabine15.9

Event Free Survival (EFS)

EFS is defined as length of time after primary treatment for a cancer ends that the participant remains free of certain complications or events that the treatment was intended to prevent or delay, for example but not exclusive of hematologic and non-hematologic toxicities, cumulative toxicities with consolidation courses, or emergence of resistance to the chemotherapy component of treatment. (NCT00778375)
Timeframe: Follow up up to 5 years/60 months.

InterventionMonths (Median)
Clofarabine + Cytarabine + Decitabine7.7

Overall Response Rate (CR, CRp/CRi and PR)

IWG Response criteria (2003): Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 times 10^9/L and platelet count > 100 times 10^9/L, and normal bone marrow differential (< 5% blasts). Complete Remission without Platelet Recovery (CRp): Peripheral blood and bone marrow results as for CR, but with platelet counts of < 100 x 10^9/L or Complete remission with incomplete marrow recovery (CRi), defined as CR above, but without normal blood counts; Partial Remission (PR): Blood count recovery as for CR, but with both a decrease in marrow blasts of at least 50% and not more than 6 to 25% abnormal cells in the marrow. Participants not achieving a complete remission following first induction course, can receive a second induction course at least 28 days following first to optimize response if possible. (NCT00778375)
Timeframe: Beginning assessment following two 10-day induction cycles through an additional re-induction cycle, up to 40 days

InterventionPercentage of Participants (Number)
Clofarabine + Cytarabine + Decitabine73

Median Overall Survival (OS)

Overall survival (OS): Time from date of treatment start until date of death due to any cause. (NCT00778375)
Timeframe: Evaluated from treatment date until date of death, followed for 5 years/60 months.

InterventionMonths (Median)
Overall SurvivalOverall Survival among Responders ((n=86)
Clofarabine + Cytarabine + Decitabine11.121.1

Number of Participants With Complete Remission [Complete Response (CR), Complete Response With Platelet Recover (CRp) or Complete Response With Incomplete Marrow Recovery (CRi)]

All responses were defined as per IWG criteria (2003) where CR Rate defined as number of participants with CR out of total treated participants. Complete remission (CR): Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 times 10^9/L and platelet count > 100 times 10^9/L, and normal bone marrow differential (< 5% blasts). Complete response with incomplete marrow recovery (CRi), defined as CR above, but without normal blood counts. Bone marrow aspirate and/or biopsy starting on day 21 (+/- 7 days) of therapy. (NCT00778375)
Timeframe: Beginning assessment following two 10-day induction cycles through an additional re-induction cycle, up to 40 days

,
InterventionParticipants (Number)
CRCRp/CRi
Induction Clofarabine + Cytarabine + Decitabine609
Re-Induction124

Cohort 1: Percentage of Participants With Complete Response (CR) for B-cell Acute Lymphoblastic Leukemia (ALL)

Complete response based on the modified National Comprehensive Cancer Network (NCCN) criteria was defined as: less than 5 percent (%) blasts in the bone marrow; no evidence of circulating blasts or extramedullary disease; full recovery of peripheral blood counts: platelets greater than (>)100*10^9 cells/liter (L) and absolute neutrophil count (ANC) >1.0*10^9 cells/L. This outcome measure was planned to be analyzed for specified arm only. (NCT03384654)
Timeframe: Up to 2 cycles, that is, up to 56 days (each cycle of 28-days)

InterventionPercentage of participants (Number)
Cohort 1: B-cell ALL (1-17 Years)0

Cohort 2: Percentage of Participants With Complete Response (CR) for T-cell ALL

Complete response based on the modified NCCN criteria was defined as: less than 5% blasts in the bone marrow; no evidence of circulating blasts or extramedullary disease; full recovery of peripheral blood counts: platelets >100*10^9 cells/L and ANC >1.0*10^9 cells/L. This outcome measure was planned to be analyzed for specified arms only. (NCT03384654)
Timeframe: End of Cycle 1 (that is, up to 28 days)

InterventionPercentage of participants (Number)
Cohort 2: T-Cell ALL (1-17 Years)41.7
Cohort 2: T-Cell ALL (18-30 Years)60.0
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)30.0

Event-free Survival (EFS)

EFS was defined as the time (in months) from the date of first treatment to the first documented treatment failure (that is [ie], disease progression) or date of relapse from CR or death due to any cause, whichever occurred first. Per NCCN criteria, relapse from CR is defined as: reappearance of leukemia blasts in the peripheral blood or >5% blasts in the bone marrow; reappearance of extramedullary disease or new extramedullary disease. Progressive disease: increase of at least 25% in the absolute number of circulating peripheral or bone marrow blasts, or development of new extramedullary disease. Kaplan-Meier method was used for the analysis. (NCT03384654)
Timeframe: Up to 4 years 4 months

InterventionMonths (Median)
Cohort 1: B-cell ALL (1-17 Years)1.1
Cohort 2: T-Cell ALL (1-17 Years)8.9
Cohort 2: T-Cell ALL (18-30 Years)10.3
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)2.9

Maximum Observed Serum Concentration (Cmax) of Daratumumab

Cmax was defined as maximum observed serum concentration of daratumumab. (NCT03384654)
Timeframe: Cohort 1: B-cell ALL: End of infusion (EOI) on Day 1 of Cycle 2; Cohort 2: T-cell ALL (1-17 years): EOI on Day 22 of Cycle 2; Cohort 2: T-cell ALL (18-30 years): EOI on Day 1 of Cycle 2; Cohort 2: T-cell LL (1-30 years): EOI on Day 22 of Cycle 2

InterventionMicrograms per milliliter (mcg/mL) (Mean)
Cohort 1: B-cell ALL (1-17 Years)494
Cohort 2: T-Cell ALL (1-17 Years)763
Cohort 2: T-Cell ALL (18-30 Years)501
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)758

Minimal Residual Disease (MRD) Negative Rate

MRD negative rate was defined as the percentage of participants who were considered MRD negative after MRD testing by bone marrow aspirate at any timepoint after first study treatment administration and before disease progression or starting subsequent anti-cancer therapy or allogeneic hematopoietic stem cell transplant (HSCT). MRD negative was defined as less than (<) 0.01% abnormal population counts to nucleated mononuclear cells when measured by flow cytometry. (NCT03384654)
Timeframe: Up to 4 years 4 months

InterventionPercentage of participants (Number)
Cohort 1: B-cell ALL (1-17 Years)0
Cohort 2: T-Cell ALL (1-17 Years)45.8
Cohort 2: T-Cell ALL (18-30 Years)20.0
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)50.0

Minimum Observed Serum Concentration (Cmin) of Daratumumab

Cmin was defined as minimum observed serum concentration of daratumumab. (NCT03384654)
Timeframe: Cohort 1: B-cell ALL: Predose on Day 1 of Cycle 2; Cohort 2: T-cell ALL (1-17 years): Predose on Day 22 of Cycle 2; Cohort 2: T-cell ALL (18-30 years): Predose on Day 1 of Cycle 2; Cohort 2: T-cell LL (1-30 years): Predose on Day 22 of Cycle 2

Interventionmcg/mL (Mean)
Cohort 1: B-cell ALL (1-17 Years)172
Cohort 2: T-Cell ALL (1-17 Years)369
Cohort 2: T-Cell ALL (18-30 Years)172
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)365

Number of Participants With Anti-daratumumab Antibodies

Number of participants with anti-daratumumab antibodies was reported. (NCT03384654)
Timeframe: Up to 4 years 4 months

InterventionParticipants (Count of Participants)
Cohort 1: B-cell ALL (1-17 Years)0
Cohort 2: T-Cell ALL (1-17 Years)0
Cohort 2: T-Cell ALL (18-30 Years)0
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)0

Overall Response Rate (ORR)

For ALL participants, ORR was defined as percentage of participants who achieved CR or CR with only partial hematological recovery(CRi) as per NCCN criteria. CR for ALL: less than 5% blasts in bone marrow; no evidence of circulating blasts or extramedullary disease; full recovery of peripheral blood counts: platelets >100*10^9 cells/L and ANC >1.0*10^9 cells/L. CRi for ALL: less than 5% blasts in bone marrow; no evidence of circulating blasts or extramedullary disease; partial recovery of peripheral blood counts not meeting criteria for CR. For LL participants, ORR was defined as percentage of participants who had CR or PR during or after treatment administration but prior to start of subsequent anti-cancer therapy or allogeneic hematopoietic stem cell transplant(HSCT). CR for LL: disappearance of all evidence of disease from all sites. PR for LL: decrease of >=50% in sum of products of diameter of lesions of up to 6 of largest dominant nodes or nodal masses with no new lesions. (NCT03384654)
Timeframe: Up to 4 years 4 months

InterventionPercentage of participants (Number)
Cohort 1: B-cell ALL (1-17 Years)14.3
Cohort 2: T-Cell ALL (1-17 Years)83.3
Cohort 2: T-Cell ALL (18-30 Years)80.0
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)50.0

Overall Survival (OS)

OS was defined as the time (in months) from the date of first study drug administration to the date of death due to any cause. Kaplan-Meier method was used for the analysis. Participants who died after consent withdrawal were considered as having an OS event. (NCT03384654)
Timeframe: Up to 4 years 4 months

InterventionMonths (Median)
Cohort 1: B-cell ALL (1-17 Years)3.2
Cohort 2: T-Cell ALL (1-17 Years)10.9
Cohort 2: T-Cell ALL (18-30 Years)12.0
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)4.2

Percentage of Participants Who Received an Allogeneic Hematopoietic Stem Cell Transplant (HSCT)

Percentage of participants who received an allogeneic HSCT after treatment with daratumumab were reported. (NCT03384654)
Timeframe: Up to 4 years 4 months

InterventionPercentage of participants (Number)
Cohort 1: B-cell ALL (1-17 Years)14.3
Cohort 2: T-Cell ALL (1-17 Years)75.0
Cohort 2: T-Cell ALL (18-30 Years)60.0
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)30.0

Relapse-free Survival (RFS)

RFS was defined as the time (in months) from CR to relapse from CR, or disease progression or death due to any cause, whichever occurred first. For ALL, as per NCCN criteria, relapse from CR is defined as: reappearance of leukemia blasts in the peripheral blood or >5% blasts in the bone marrow, or reappearance of extramedullary disease or new extramedullary disease. Progressive disease: increase of at least 25% in absolute number of circulating peripheral or bone marrow blasts, or development of new extramedullary disease. Kaplan-Meier method was used for analysis. (NCT03384654)
Timeframe: Up to 4 years 4 months

InterventionMonths (Median)
Cohort 2: T-Cell ALL (1-17 Years)19.4
Cohort 2: T-Cell ALL (18-30 Years)9.4
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)NA

Concentration of Daratumumab in Cerebrospinal Fluid (CSF)

Concentration of daratumumab in CSF was reported. '0' in the number analyzed field signifies that none of the participants were available for the analysis at the specified time points. (NCT03384654)
Timeframe: Pre-dose on Cohort 1: Cycles 1 and 2: Day 1; Cohort 2: Cycle 1 Day 1 and Day 15 and Cycle 2 Day 2 and Day 15

Interventionmcg/mL (Mean)
Cycle 1 Day 1 predoseCycle 2 Day 1 predose
Cohort 1: B-cell ALL (1-17 Years)NA0.573

Concentration of Daratumumab in Cerebrospinal Fluid (CSF)

Concentration of daratumumab in CSF was reported. '0' in the number analyzed field signifies that none of the participants were available for the analysis at the specified time points. (NCT03384654)
Timeframe: Pre-dose on Cohort 1: Cycles 1 and 2: Day 1; Cohort 2: Cycle 1 Day 1 and Day 15 and Cycle 2 Day 2 and Day 15

,,
Interventionmcg/mL (Mean)
Cycle 1 Day 1 predoseCycle 1 Day 15 predoseCycle 2 Day 2 predoseCycle 2 Day 15 predose
Cohort 2: T-Cell ALL (1-17 Years)NA0.9070.9150.934
Cohort 2: T-Cell ALL (18-30 Years)NA0.3190.2960.163
Cohort 2: T-Cell Lymphoblastic Leukemia (LL) (1-30 Years)NA0.4561.231.06

Dose Limiting Toxicity

To determine the dose limiting toxicity (DLT) of Plitidepsin (Aplidin®) when administered daily x 5 days with a fixed dose of Cytarabine for 5 days every four weeks in patients with relapsed/refractory leukemia. (NCT00780143)
Timeframe: Until disease progression or unacceptable toxicity

InterventionParticipants (Count of Participants)
Aplidin®0

Contribution of Dasatinib on Minimal Residual Disease (MRD) After Induction Therapy

Percent of patients MRD Positive (MRD > 0.01%) at End of Induction. (NCT00720109)
Timeframe: At the end of induction therapy (at 5 weeks)

InterventionPercentage of participants (Number)
Treatment Induction (Enzyme Inhibitor Therapy & Chemotherapy)40.7
Standard-risk29.2
High-risk100.0

Event-Free Survival (EFS) of Patients With Standard-risk Disease Treated With Dasatinib in Combination With Intensified Chemotherapy

Event-Free Survival (EFS) curves will be constructed using the Kaplan-Meier life table method with standard errors computed using the method of Peto and Peto. A 1-sided 95% confidence interval for EFS will be constructed. (NCT00720109)
Timeframe: At 3 years

InterventionPercent probability (Number)
Treatment Induction (Enzyme Inhibitor Therapy & Chemotherapy)79.8
Standard-risk83.2
High-risk66.7

Feasibility and Toxicity of an Intensified Chemotherapeutic Regimen Incorporating Dasatinib for Treatment of Children and Adolescents With Ph+ ALL Assessed by Examining Adverse Events

Number of patients in safety cohort with dose limiting toxicity (DLT)(including treatment delay) (NCT00720109)
Timeframe: Weeks 3 through 23 of treatment (From week 3 Induction through Intensification Block 1)

InterventionPts with DLTs (Number)
Treatment Induction (Enzyme Inhibitor Therapy & Chemotherapy)1

Overall EFS Rate for the Combined Cohort of Standard- and High-Risk Patients (Who Receive the Final Chosen Dose of Dasatinib)

An event is defined as: Induction failure, relapse at any site, secondary malignancy, or death. (NCT00720109)
Timeframe: From the time entry on study to first event or date of last follow-up, assessed up to 7 years

Interventionpercentage of patients (Number)
Treatment Induction (Enzyme Inhibitor Therapy & Chemotherapy)79.8
Standard-risk83.2
High-risk66.7

Percent of Patients MRD Positive (MRD > 0.01%) at End of Consolidation

A 1-sample Z-test of proportions (alpha=5%, 1-sided test) will be used. (NCT00720109)
Timeframe: At end of consolidation (at 11 weeks)

InterventionPercentage of participants (Number)
Treatment Induction (Enzyme Inhibitor Therapy & Chemotherapy)10.5
Standard-risk0
High-risk66.7

Progression-free Survival Rate

"after autologous stem cell transplantation (ASCT). Disease progression is defined using International Workshop Criteria for non-Hodgkin lymphoma37 and is defined as:~≥ 50% increase in products of diameters of any previously identified abnormal node or nodule AND/OR~appearance of any new lesions" (NCT00521014)
Timeframe: up to 3 years

Interventionyears (Mean)
Relapsed Follicular Lymphoma Patients1.759

Maximum Tolerated Dose (MTD) of Clofarabine

Maximum tolerated dose (MTD) defined as the highest dose schedule in which 6 patients were treated with at most 1 experiencing a dose-limiting toxicity (DLT). Clofarabine 15 mg/m2 IV over approximately 1 hour daily (number of days selected based on Phase I portion). (NCT01794702)
Timeframe: After second, 33 day cycle

Interventionmg/m^2 x 4 days (6-9) (Number)
Period 115

Number of Participants With a Response

Primary endpoint is overall response defined as the best response either complete response, complete remission without platelet recovery, or complete remission without incomplete blood count recovery within 56 days. (NCT01794702)
Timeframe: 56 days

InterventionParticipants (Count of Participants)
Phase II Clofarabine + Cytarabine + Decitabine + Idarubicin20

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT01794702)
Timeframe: Up to 2 years after participants off study date

InterventionMonths (Median)
Phase II Clofarabine + Cytarabine + Decitabine + Idarubicin7.7

To Determine the Disease-free Survival (DFS).

Time from date of treatment start until the date of first objective documentation of return of disease. (NCT01794702)
Timeframe: Up to 2 years after participants off study date

Interventionmonths (Median)
Phase II Clofarabine + Cytarabine + Decitabine + Idarubicin17.9

Disease-free Survival

Disease-free survival (DFS) was measured as the interval from achievement of CR until relapse or death, regardless of cause. DFS was estimated using the Kaplan Meier method. (NCT00742625)
Timeframe: Duration of study (up to 10 years)

Interventionmonths (Median)
Bortezomib + Daunorubicin + Cytarabine8

Overall Survival

Overall survival (OS) as the interval from the on-study date until death. OS was estimated using the Kaplan Meier method. (NCT00742625)
Timeframe: Duration of study (up to 10 years)

Interventionmonths (Median)
Bortezomib + Daunorubicin + Cytarabine12

Participants Experiencing a Dose-limiting Toxicity (DLT) of Bortezomib When Administered in Combination With Intermediate-dose Cytarabine

"DLTs were considered only during the first cycle of consolidation therapy and included grade 3 or 4 sensory or autonomic neuropathy, persistent grade 4 thrombocytopenia or neutropenia at day 42 in the absence of AML,any grade 4 or 5 nonhematologic toxicity, and any grade 3 nonhematologic toxicity (excluding neuropathy and toxicities secondary to neutropenia and sepsis) that did not resolve to grade 2 by day 42 unless attributable to persistent or recurrent AML. Grade 4 anorexia (requiring total parenteral nutrition) and grade 4 fatigue (requiring bed rest) were not considered DLTs.~Toxicity was graded per National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. Grading scale is as follows: grade 1: mild; grade 2: moderate; grade 3: Severe; grade 4: Life Threatening; grade 5: Death." (NCT00742625)
Timeframe: during consolidation cycle 1 (42 days)

Interventionparticipants (Number)
Bortezomib (0.7 mg/m^2) + Int-DAC0
Bortezomib (1.0 mg/m^2) + Int-DAC0
Bortezomib (1.3 mg/m^2) + Int-DAC1

Remission Induction Response

"Response was calculated according to Revised International Working Group (IWG) criteria for Acute myeloid leukemia (AML)~A response was defined as the portion of participants who achieved a complete response (CR) or CR with incomplete platelet recovery(CRp) during induction.~A CR is defined as those with > 20% cellularity of bone marrow biopsy, no presence of extramedullary leukemia for AML, <5 % myeloblast cells for bone marrow with peripheral blood and normal complete blood count (absolute neutrophils > 1000 mL and platelets >= 100,000 mL).~A CRp is defined as a CR except platelets < 100,000 mL without need for transfusion." (NCT00742625)
Timeframe: 2 months

Interventionparticipants (Number)
Complete responseComplete response with incomplete platelet recover
Bortezomib + Daunorubicin + Cytarabine624

Overall Survival at One Year

The number of participants alive one year after baseline. (NCT00973752)
Timeframe: 1 years

InterventionParticipants (Count of Participants)
Experimental19

Disease-free Survival

To determine DFS at 1 year post transplant. (NCT00439556)
Timeframe: At 1 year

InterventionParticipants (Count of Participants)
Velcade Dose Level 116
Velcade Dose Level 20
Velcade Dose Level 30

Number of Participants With Dose Limiting Toxicity (DLT)

To determine the maximum tolerated dose(MTD) of velcade and dose limiting toxicity(DLT). A dose limiting toxicity (DLT) was defined as a grade 3-4 neurological toxicity, graft failure, or death due to GvHD. The Commom Terminlogy Criteria for Adverse Events v3.0 was used. (NCT00439556)
Timeframe: From start of treatment to 90 days after the start of treatment

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, Transplant, Filgrastim, Tacrolimus)0
Velcade Dose Level 20
Velcade Dose Level 30

3 Year Progression-Free Survival Rate

Percentage of participants out of total treated alive with disease progression 3 years following registration. Progression-free survival (PFS) was measured from the date of study registration to the date of documented disease progression or death of any cause. (NCT00290433)
Timeframe: From registration to disease progression or death, up to 3 years

Interventionpercentage of participants (Number)
HCVIDDOXIL Regimen30

Overall Survival (OS) of Patients With Burkitt's and Burkitt-like Lymphoma/Leukemia Treated With Modified Magrath Regimen

Overall Survival (OS) of patients with Burkitt's and Burkitt-like Lymphoma/Leukemia treated with modified Magrath regimen. OS is defined from the first dose of study drug to the time of death for any reason. (NCT00392990)
Timeframe: At 2 years from treatment initiation Median follow up 34 months (range 15-45)

Interventionpercentage of patients alive (Number)
High Risk - Treated With Alternating R-CODOX-M/R-IVAC81
Low Risk - Treatment With 3 Cycles of R-CODOX-M100

Progression Free Survival (PFS) of Patients With Burkitt's and Burkitt-like Lymphoma/Leukemia Treated With Modified Magrath Regimen

Progression Free Survival (PFS) will be defined from the first dose of study drug to the first documentation of progressive disease or death for any reason. Patients that are lost to follow-up before progression will be censored at the point of last documentation of not experiencing the event. (NCT00392990)
Timeframe: At 2 years from treatment initiation. Median follow up 34 months (range 15-45)

Interventionpercentage of patients progression free (Number)
High Risk - Treated With Alternating R-CODOX-M/R-IVAC76
Low Risk - Treatment With 3 Cycles of R-CODOX-M100

Overall Response Rate (ORR) of Patients With Burkitt's and Burkitt-like Lymphoma/Leukemia Treated With Modified Magrath Regimen

"Response Rate is defined as combined number of patients with Complete Remission (CR) Complete Remission undetermined (Cru) and Partial Remission (PR) for patients with Burkitt's and Burkitt-like Lymphoma/Leukemia. Response will be measured by CT scans following treatment completion and assessed using Response Criteria for Non-Hodgkin's Lymphoma where:~CR=complete disappearance of all detectable clinical and radiographic evidence of disease and disappearance of all disease-related symptoms.~CRu=same as above but allowing for 75% decrease in lymph node masses and indeterminate or normal bone marrow.~PR=50% decrease in SPD of the six largest dominant nodes or nodal masses with no increase in size or other nodes, liver, spleen and no new sites of disease." (NCT00392990)
Timeframe: After two cycles of treatment and at completion of treatment (4 cycles for high risk and 3 cycles for low risk) up to approximately 20 weeks.

,
Interventionpercentage of patients (Number)
ORR after two cycles of treatmentORR at completion of treatment
High Risk - Treated With Alternating R-CODOX-M/R-IVAC100100
Low Risk - Treatment With 3 Cycles of R-CODOX-M100100

Safety of Patients With Burkitt's and Burkitt-like Lymphoma/Leukemia Treated With Modified Magrath Regimen (Addition of Rituximab, Subsitution of Adriamycin for Doxil and Using Lower Dose of Methotrexate)

"Adverse events (AE) graded as either 3 or 4 and determined to be at least possibly related to study treatment will be collected in patients Burkitt's and Burkitt-like Lymphoma/Leukemia treated with modified Magrath regimen to assess the safety and toxicity profile of the addition of rituximab, subsitution of adriamycin for doxil and lower dose of methotrexate. AEs will be assessed as follows at the following time points: At the end of each cycle or monthly whichever is less frequent and 30 days post last dose of study treatment for a maximum of 3 cycles for regimen A and 4 cycles for regimen B and up to 12 months.~In general AEs will be assessed according to the National Cancer Institute's Common Toxicity Criteria for adverse events version 3.0 (CTCAE v 3.0). In general adverse events (AEs) will be graded according to the following:~Grade 1 Mild AE Grade 2 Moderate AE Grade 3 Severe AE Grade 4 Life-threatening or disabling AE Grade 5 Death related to AE" (NCT00392990)
Timeframe: After each cycle or monthly (whichever is less frequent), 30 days post last dose. Treatment duration was at a median of 13 weeks (range 11-20) for high-risk patients and 10 weeks (range 9-12) for low-risk patients.

,
Interventionparticipants (Number)
Anemia : Grade 3Anemia : Grade 4Neutropenia : Grade 3Neutropenia : Grade 4Thrombocytopenia : Grade 3Thrombocytopenia : Grade 4Mucositis : Grade 3Mucositis : Grade 4Infection : Grade 3Infection : Grade 4Elevated Transaminases : Grade 3Elevated Transaminases : Grade 4Fever (Neutropenic) : Grade 3Fever (Neutropenic) : Grade 4Low Phosphate : Grade 3Low Phosphate : Grade 4Sodium Abnormalities : Grade 3Sodium Abnormalities : Grade 4Hyperglycemia : Grade 3Hyperglycemia : Grade 4Hypoalbuminemia : Grade 3Hypoalbuminemia : Grade 4Hypokalemia : Grade 3Hypokalemia : Grade 4Cardiac : Grade 3Cardiac : Grade 4Diarrhea : Grade 3Diarrhea : Grade 4Elevated Creatinine : Grade 3Elevated Creatinine : Grade 4Nausea/Vomiting : Grade 3Nausea/Vomiting : Grade 4Low Blood Pressure : Grade 3Low Blood Pressure : Grade 4Rash : Grade 3Rash : Grade 4Edema : Grade 3Edema : Grade 4Low Magnesium : Grade 3Low Magnesium : Grade 4
High Risk - Treated With Alternating R-CODOX-M/R-IVAC131561137380604050401540103020201010101010
Low Risk - Treatment With 3 Cycles of R-CODOX-M3022122010203010203000200000000000000000

Relationship of Inhibition of DNA Synthesis and Clinical Response

Clinical response is defined as MRD (minimal residual disease) measured by flow cytometry at day 22. The MRD at day 22 is classified as positive (with MRD) or negative (no detectable MRD). The relation between inhibition of DNA synthesis and MRD was performed by logistic regression. In the model, logit of probability of MRD positive was regressed on inhibition of DNA synthesis. (NCT00136084)
Timeframe: Measurements were assessed in Induction I chemotherapy

InterventionPercent inhibition of DNA Synthesis (Mean)
Overall66.7

To Assess Whether Inhibition of DNA Synthesis is Greater After High-dose Ara-C (HDAC) Than After Low-dose Ara-C (LDAC) Therapy

Inhibition of DNA synthesis is defined as the percentage of DNA synthesis rate at 24-hour post-araC treatment over DNA synthesis rate pre-araC treatment. (NCT00136084)
Timeframe: Measurements were assessed in Induction I chemotherapy

InterventionPercent Inhibition of DNA Synthesis (Mean)
Arm 1: (HDAC)60.6
Arm 2:(LDAC)72.8

To Estimate the Overall Event-free Survival (EFS) of AML Patients Who Undergo Risk-adapted and Genotype-directed Therapy

Overall event-free survival (EFS) was defined as the time from study enrollment to induction failure, relapse, secondary malignancy, death, or study withdrawal for any reason, with event-free patients censored on the date of the last follow-up (NCT00136084)
Timeframe: Five Year

InterventionPercentage of Participants (Number)
Overall62.4

Minimal Residual Disease (MRD).

Detection of Minimal Residual Disease following one course of chemotherapy where positive MRD was defined as one or more leukemic cell per 1000 mononuclear bone-marrow cells (>=0.1%). (NCT00136084)
Timeframe: Day 22 MRD measurement

,
Interventionparticipants (Number)
MRD PositiveMRD Negative
Arm 1: (HDAC)3168
Arm 2:(LDAC)4363

Proportion of Minimal Residual Disease (MRD)+ Patients Who Become MRD- After One Course of Gemtuzumab Ozogamicin (GO)

To estimate the proportion of minimal residual disease (MRD)+ patients who become MRD- after one course of gemtuzumab ozogamicin (GO) (NCT00136084)
Timeframe: Consolidation I

InterventionParticipants (Number)
NegativePositive
Overall114

Proportion of MRD Reduction After One Course of Cytarabine + Daunomycin + Etoposide (ADE) + GO

To estimate proportion of patients with MRD reduction after one course of Induction II (cytarabine + daunomycin + etoposide (ADE) + GO), who had no response to first course of induction therapy. (NCT00136084)
Timeframe: Induction II

InterventionParticipants (Number)
DecreaseIncrease or no change
Overall272

Proportion of Patients Experienced Toxicity of Cytarabine + Daunomycin + Etoposide (ADE) + GO.

To estimate proportion of patients experiencing CTC Grade 3 or 4 toxicity during Induction II (Cytarabine + Daunomycin + Etoposide (ADE) + GO), who had no response to first course of induction therapy (NCT00136084)
Timeframe: Induction II

InterventionParticipants (Number)
Experienced Grade 3 or 4 toxicitiesDid not experience Grade 3 or 4 toxicities
Overall273

Number of Participants Who Died or Were Censored by 24 Months

This measure (time to death or censor) was defined for all enrolled subjects from the date of randomization until death from any cause. If a subject was not known to have died by the end of the followup period, observation of overall survival was censored on the date the patient was last known to be alive. The number of participants who died or were censored is presented here. (Note: all subjects participating in this study had either died or were censored by 24 months.) (NCT00513305)
Timeframe: From Baseline through 24 months following Baseline

InterventionParticipants (Number)
Low-dose Cytarabine Plus Arsenic Trioxide33
Low-dose Cytarabine Alone34

Number of Participants Who Experienced Early Death

Early death is defined as death from any cause within the first 14 days after start of study drug treatment. The number of patients in each study group who experienced early death is presented here. (NCT00513305)
Timeframe: 14 days from start of study drug treatment

InterventionParticipants (Number)
Low-dose Cytarabine Plus Arsenic Trioxide0
Low-dose Cytarabine Alone2

Number of Participants Who Experienced Induction (Thirty-Day) Mortality

The number of subjects who died from any cause within the first 30 days after the start of study drug treatment is presented here. (NCT00513305)
Timeframe: Up to 30 days following start of study drug treatment

InterventionParticipants (Number)
Low-dose Cytarabine Plus Arsenic Trioxide3
Low-dose Cytarabine Alone2

Percentage of Participants in Complete Remission (CR)

The primary efficacy variable was the percentage of subjects in each treatment group who achieved complete remission after treatment with study drug. Complete remission was defined as: 1) Less than 5% blasts in normocellular bone marrow sample. 2) No blasts in bone marrow sample containing Auer rods. 3)Clearance of previous extramedullary disease. 4)Normal values for absolute neutrophil count (at least 1000/microliter), platelet count (at least 100,000/microliter), without platelet transfusions, and in absence of peripheral myeloblasts. (NCT00513305)
Timeframe: From baseline through Day 70. Assessments were performed on Day 21 in cycle 1, no later than Day 56 of cycle 1 or 2 (if applicable), and no later than Day 70 of cycle 1 or 2 (if applicable) or at any other time at the discretion of the investigator

InterventionPercentage of participants in CR (Number)
Low-dose Cytarabine Plus Arsenic Trioxide15.2
Low-dose Cytarabine Alone8.8

Proportion of Participants Surviving at 6 Months and 12 Months Using the Kaplan Meier Estimate

The duration of overall survival was defined for all patients and was measured from the date of randomization until death from any cause. For a patient who was not known to have died by the end of the follow-up period, observation of overall survival was censored on the date the patient was last known to be alive. The estimate of likelihood of survival at 6 and 12 months after Baseline using the Kaplan Meier Estimate is presented here. (NCT00513305)
Timeframe: Baseline through 12 months

,
InterventionProportion of Participants (Number)
Month 6 (95% Confidence Interval)Month 12 (95% Confidence Interval)
Low-dose Cytarabine Alone0.6498.51
Low-dose Cytarabine Plus Arsenic Trioxide0.62612.22

Proportion of Participants With Relapse-Free Survival (RFS)Using the Kaplan-Meier Estimate

RFS is defined for patients who achieved a complete remission (CR), complete remission with incomplete platelet recovery (CRp), or partial remission(PR), and was measured from the date of attaining CR, CRp, or PR until the date of AML relapse or death from any cause, whichever occurred first. For a patient not known to have relapsed or died by the end of the study followup, observation of relapse free survival was censored on the date of the last followup examination. The Kaplan Meier Estimate of relapse-free survival at Month 3 and Month 6 is presented here. (NCT00513305)
Timeframe: From Baseline (randomization) through 24 months following Baseline

,
InterventionProportion of participants (Number)
Month 3 (95% Confidence Interval)Month 6 (95% Confidence Interval)
Low-dose Cytarabine Alone0.8000.800
Low-dose Cytarabine Plus Arsenic Trioxide0.8180.818

Event Free Survival. EFS

Percentage of patients who were event free at 3 years among those on Standard VCR dosing who did not undergo Hematopoietic Stem Cell Transplant (SCT). (NCT00381680)
Timeframe: 3 years after enrollment

Interventionpercentage of participants EFS at 3 yrs3 (Number)
Regimen A: Standard Vincristine Dosing66.0

Rate of Minimal Residual Disease (MRD) < 0.01% at End Block 1

Percentage of patients who had minimal residual disease (MRD) < 0.01% among those with isolated BM or combined BM relapse >= 36 months and had successful MRD determinations at End Block 1 (NCT00381680)
Timeframe: End of Block 1 (35 days) of Induction therapy

Interventionpercentage of participants (Number)
Regimen A50.8
Regimen B41.5

Rate of Minimal Residual Disease (MRD) < 0.01% at End Block 3

Percentage of patients who had minimal residual disease (MRD) < 0.01% among those with isolated BM or combined BM relapse >= 36 months and had successful MRD determinations at End Block 3. (NCT00381680)
Timeframe: End of Block 3 (105 days) of Induction therapy

Interventionpercentage of participants (Number)
Regimen A81.4
Regimen B88.9

Adjusted Event Free Survival

Adjusted percentage of patients who were event free at 3 years. For patients who received matched donor SCT, EFS was adjusted to start from the actual SCT date. For patients who did not undergo SCT, EFS was adjusted to start from median time to SCT based on patients who received matched related SCT (where patients who had events prior to SCT date were excluded from the calculation of median time to SCT). (NCT00381680)
Timeframe: 3 years

Interventionadjusted percentage of participants (Number)
Received SCTDid not receive SCT
Regimen A: Standard Vincristine Dosing82.264.2

Event Free Survival (EFS)

Percentage of patients who were event free at 3 years among those with isolated BM or combined BM relapse >= 36 months. (NCT00381680)
Timeframe: 3 years

,
Interventionpercentage of participants (Number)
MRD < 0.01% BL1MRD >= 0.01% BL1MRD < 0.01% BL3MRD >= 0.01% BL3
Regimen A88.560.083.861.5
Regimen B77.346.283.333.3

Frequency and Severity of Adverse Effects

Percentage of patients who developed at least 1 episode of grade 2 to 4 neuropathy. (NCT00381680)
Timeframe: Up to 107 weeks

Interventionpercentage of participants (Number)
CC or CT genotypeHigh-risk CEP72 genotype (TT at rs924607)
All Patients17.344.4

Number of Participants With Serious Adverse Events as a Measure of Safety (First-line Treatment)

A serious adverse event is any experience that suggests a significant hazard, contraindication, side effect or precaution that: results in death, is life-threatening, requires in-patient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or is medically significant (NCT00333840)
Timeframe: 144 months

InterventionParticipants (Number)
Imatinib (STI571)226
IFN-a + Ara-C163

Number of Participants With Serious Adverse Events as a Measure of Safety (Second-line Treatment)

A serious adverse event is any experience that suggests a significant hazard, contraindication, side effect or precaution that: results in death, is life-threatening, requires in-patient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or is medically significant (NCT00333840)
Timeframe: 144 months

InterventionParticipants (Number)
Imatinib to IFN-a + Ara-C2
IFN-a + Ara-C to Imatinib160

Kaplan Meier Estimates of Event Free Survival (All Randomized Participants)

"Event-free survival is defined as the time between randomization and the earliest of any of the following events on treatment:~progression to Accelerated Phase (AP) or Blast Crisis (BC)~loss of Complete Hematological Response (CHR)~loss of Major Cytogenetic Response (MCyR) confirmed~loss of Major Cytogenetic Response (MCyR) unconfirmed~increase in white blood cell count (WBC) if approved by the Study Management Committee (SMC)~death (due to any cause when reported as primary reason for discontinuation of treatment).~Kaplan Meier estimates of the percentage of participants with Event Free Survival at the given time point was calculated. This outcome was measured in all randomized patients, regardless of whether crossover occurred, i.e., events that occurred in patients, who had crossed over, were attributed following crossover to the original randomized treatment." (NCT00333840)
Timeframe: 12,24,36,48,60,72,84,96,108,120,132 and 144 months

,
InterventionPercentage of participants (Number)
12 months (n = 553, 553)24 months (n = 505, 332)36 months (n = 448, 263)48 months (n = 415, 241)60 months (n = 395, 223)72 months (n = 376, 207)84 months (n = 352, 181)96 months (n = 334, 166)108 months (n = 301, 141)120 months (n = 269, 130)132 months (n = 257, 124)144 months (n = 143, 76)
IFN-a + Ara-C79.470.067.063.761.960.159.058.356.656.656.651.8
Imatinib (STI571)96.689.685.383.983.283.082.081.079.979.679.679.6

Kaplan Meier Estimates of Time to Progression to Accelerated Phase (AP) or Blast Crisis (BC) (All Randomized Participants)

Time to progression to AP/BC is defined as the time between randomization and either of the following events on treatment: death (due to CML when reported as primary reason for discontinuation of treatment) or progression to Accelerated Phase or Blast Crisis and is censored at last examination date for patients without event. No data after discontinuation of study treatment was included. The Kaplan Meier estimates of the percentage of participants with survival without progression to AP/BC at the given time point was calculated. This outcome was measured in all randomized patients, regardless of whether crossover occurred, i.e., events that occurred in patients, who had crossed over, were attributed following crossover to the original randomized treatment. (NCT00333840)
Timeframe: 12,24,36,48,60,72,84,96,108,120,132 and 144 months

,
InterventionPercentage of participants (Number)
12 months (n = 553, 553)24 months (n = 513, 388)36 months (n = 461, 337)48 months (n = 431, 311)60 months (n = 409, 289 )72 months (n = 384, 265)84 months (n = 358, 236)96 months (n = 338, 220)108 months (n = 305, 189)120 months (n = 272, 175)132 months (n = 259, 165)144 months (n = 145, 94)
IFN-a + Ara-C92.990.087.984.783.583.182.482.482.482.482.482.4
Imatinib (STI571)98.595.794.093.192.792.792.792.492.192.192.192.1

Kaplan-Meier Estimates of Overall Survival (All Randomized Participants)

Overall survival was defined as the time between date of randomization and death due to any cause. The time was censored at last examination date for patients who were still being treated and at date of last contact for patients who discontinued treatment. Kaplan-Meier estimates of the percentage of participants at each time point was calculated. This outcome was measured in all randomized patients, regardless of whether crossover occurred, i.e., events that occurred in patients, who had crossed over, were attributed following crossover to the original randomized treatment. (NCT00333840)
Timeframe: 12,24,36,48,60,72,84,96,108,120,132 and 144 months

,
InterventionPercentage of participants (Number)
12 months (n = 553, 553)24 months (n = 542, 512)36 months (n = 518, 471)48 months (n = 492, 441)60 months (n = 475, 411)72 months (n = 461, 388)84 months (n = 446, 373)96 months (n = 430, 358)108 months (n = 391, 315)120 months (n = 368, 299)132 months (n = 356, 288)144 months (n = 250, 199)
IFN-a + Ara-C97.793.689.887.785.483.883.181.479.978.877.775.4
Imatinib (STI571)98.996.092.490.489.488.286.685.484.383.382.881.3

Percentage of Participants With Best Cytogenetic Response (First-line Treatment)

"Bone marrow aspirate was performed to evaluate cytogenetic results (percentage of Philadelphia chromosome positive (Ph+) metaphases) and amount of blasts and promyelocytes in bone marrow to establish cytogenetic response.~Major Cytogenetic Response= Complete Response or Partial Response. Complete Cytogenetic Response= 0 % of Ph+ metaphases (out of 20 metaphases). Partial Cytogenetic Response= > 0 and ≤ 35 % of Ph+ metaphases (out of 20 metaphases). The percentage of participants with cytogenetic response in each category was calculated." (NCT00333840)
Timeframe: 144 months

,
InterventionPercentage of participants (Number)
Major Cytogenic ResponseComplete Cytogenic ResponsePartial Cytogenic Response
IFN-a + Ara-C23.311.611.8
Imatinib (STI571)89.082.86.1

Percentage of Participants With Best Cytogenetic Response (Second-line Treatment)

"Bone marrow aspirate was performed to evaluate cytogenetic results (percentage of Ph chromosome (Ph+) containing metaphases) and the amount of blasts and promyelocytes in bone marrow to establish cytogenetic response.~Major Cytogenetic Response= Complete Response or Partial Response. Complete Cytogenetic Response= 0 % of Ph+ metaphases (out of 20 metaphases). Partial Cytogenetic Response= > 0 and ≤ 35 % Ph+ metaphases (out of 20 metaphases). The percentage of participants with cytogenetic response in each category was calculated." (NCT00333840)
Timeframe: 144 months

,
InterventionPercentage of participants (Number)
Major Cytogenic ResponseComplete Cytogenic ResponsePartial Cytogenic Response
IFN-a + Ara-C to Imatinib86.081.05.0
Imatinib to IFN-a + Ara-C14.37.17.1

Percentage of Participants With Event Free Survival Events (All Randomized Participants)

"Event-free survival is defined as the time between randomization and the earliest of any of the following events on treatment:~progression to Accelerated Phase (AP) or Blast Crisis (BC)~loss of Complete Hematological Response (CHR)~loss of Major Cytogenic Response (MCyR) confirmed~loss of Major Cytogenic Response (MCyR) unconfirmed~increase in white blood cell count (WBC) if approved by the Study Management Committee (SMC)~death (due to any cause when reported as primary reason for discontinuation of treatment).~The percentage of participants with Event Free Survival events in each category was calculated. This outcome was measured in all randomized patients, regardless of whether crossover occurred, i.e., events that occurred in patients, who had crossed over, were attributed following crossover to the original randomized treatment." (NCT00333840)
Timeframe: 144 months

,
InterventionPercentage of participants (Number)
Participants with eventsProgression to AP or BCLoss of CHRLoss of MCyR (confirmed)Loss of MCyR (unconfirmed)Increase of WBCDeath
IFN-a + Ara-C32.212.87.65.41.13.41.8
Imatinib (STI571)17.96.92.74.20.90.23.1

Percentage of Participants With Major Molecular Response (First-line Treatment)

Major Molecular Response was determined using a quantitative polymerase chain reaction (PCR) laboratory test and was defined as BCR-ABL protein transcripts of ≤ 0.1% according to the international scale. (NCT00333840)
Timeframe: 12,24,36,48,60,72,84,96,108,120,132 and 144 months

,
InterventionPercentage of participants (Number)
12 months (n= 305, 83)24 months (n= 102, 12)36 months (n= 68, 7)48 months (n= 305, 12)60 months (n= 316, 10)72 months (n= 292, 9)84 months (n= 247, 8)96 months (n= 228, 4)108 months (n= 233, 0)120 months (n= 204, 0)132 months (n= 168, 0)144 months (n= 1, 0)
IFN-a + Ara-C9.625.028.658.3100.088.987.5100.0NANANANA
Imatinib (STI571)50.269.676.577.088.088.091.992.593.693.192.3100.0

Percentage of Participants With Major Molecular Response (Second-line Treatment)

Major Molecular Response was determined using a quantitative polymerase chain reaction (PCR) laboratory test and was defined as BCR-ABL protein transcripts of ≤ 0.1% according to the international scale. (NCT00333840)
Timeframe: 12,24,36,48,60,72,84,96,108,120,132 and 144 months

InterventionPercentage of participants (Number)
12 months (n = 86)24 months (n = 62)36 months (n = 130)48 months (n = 216)60 months (n = 174)72 months (n = 146)84 months (n = 139)96 months (n = 138)108 months (n = 120)120 months (n = 84)132 months (n = 31)144 months
IFN-a + Ara-C to Imatinib51.269.478.581.986.291.186.387.090.085.790.3NA

Disease-free Survival by IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)

"Disease-free survival was defined as the time from first complete remission (CR) or complete remission with incomplete peripheral blood count recovery (CRi) until the date of first objective documentation of disease recurrence or death due to any cause, whichever occurred first.~See Outcome #3 for definition of CR and CRi.~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 12 to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine8.1
Placebo and Cytarabine7.0
Clofarabine and Cytarabine - In Stratum < 6 Months5.7
Placebo and Cytarabine - In Stratum < 6 Months6.7
Clofarabine and Cytarabine In Stratum >= 6 Months10.3
Placebo and Cytarabine In Stratum >= 6 Months9.1

Disease-free Survival by IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)

"Disease-free survival was defined as the time from first complete remission (CR) or complete remission with incomplete peripheral blood count recovery (CRi) until the date of first objective documentation of disease recurrence or death due to any cause, whichever occurred first.~See Outcome #3 for definition of CR and CRi.~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 12 to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine9.5
Placebo and Cytarabine7.0
Clofarabine and Cytarabine - In Stratum < 6 Months6.7
Placebo and Cytarabine - In Stratum < 6 Months6.7
Clofarabine and Cytarabine In Stratum >= 6 Months15.4
Placebo and Cytarabine In Stratum >= 6 Months9.2

Duration of Remission (DOR) Per IRRP Assessment-Overall and by Calculated Strata (CSR 7-April-11)

"DOR was defined as the time from first CR or CRi to the date of first objective documentation of disease recurrence, initiation of alternative antileukemic therapy [including hematopoietic stem cell transplant] while in remission, or death due to any cause, whichever occurred first.~CR is defined on morphologic criteria at a single response assessment:~a bone marrow aspirate or biopsy of <5% blasts, with evidence of normal hematopoiesis;~absence of Auer rods in the blasts that are present;~absence of extramedullary disease;~absence of a unique phenotype determined at the pretreatment specimen, as assessed by immunophenotyping;~only rare evidence of circulating blasts. If present, evidence of a regenerating bone marrow;~recovery of peripheral counts (platelets ≥100*10^9/L and absolute neutrophil count (ANC) ≥1.0*10^9/L).~CRi met all criteria for CR except for either residual neutropenia (ANC <1.0*10^9/L) or thrombocytopenia (platelet count <100*10^9/L)." (NCT00317642)
Timeframe: Day 12 to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine7.6
Placebo and Cytarabine3.8
Clofarabine and Cytarabine - In Stratum < 6 Months5.7
Placebo and Cytarabine - In Stratum < 6 Months6.3
Clofarabine and Cytarabine In Stratum >= 6 Months11.5
Placebo and Cytarabine In Stratum >= 6 Months3.8

Duration of Remission (DOR) Per IRRP Assessment-Overall and by Randomized Strata (CSR 9-July-12)

"DOR was defined as the time from first CR or CRi to the date of first objective documentation of disease recurrence, initiation of alternative antileukemic therapy [including hematopoietic stem cell transplant] while in remission, or death due to any cause, whichever occurred first.~CR is defined on morphologic criteria at a single response assessment:~a bone marrow aspirate or biopsy of <5% blasts, with evidence of normal hematopoiesis;~absence of Auer rods in the blasts that are present;~absence of extramedullary disease;~absence of a unique phenotype determined at the pretreatment specimen, as assessed by immunophenotyping;~only rare evidence of circulating blasts. If present, evidence of a regenerating bone marrow;~recovery of peripheral counts (platelets ≥100*10^9/L and absolute neutrophil count (ANC) ≥1.0*10^9/L).~CRi met all criteria for CR except for either residual neutropenia (ANC <1.0*10^9/L) or thrombocytopenia (platelet count <100*10^9/L)." (NCT00317642)
Timeframe: Day 12 to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine7.7
Placebo and Cytarabine3.8
Clofarabine and Cytarabine - In Stratum < 6 Months6.7
Placebo and Cytarabine - In Stratum < 6 Months6.3
Clofarabine and Cytarabine In Stratum >= 6 Months10.2
Placebo and Cytarabine In Stratum >= 6 Months3.8

Event-free Survival by IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)

"Event-free survival (EFS) was defined as the time from randomization to the date of treatment failure, first disease recurrence (for participants who achieved remission), or death due to any cause, whichever occurred first.~Treatment Failure - ≥5% leukemic blasts by bone marrow exam, with no evidence of hematologic response (ie, <30% decrease in % leukemic blasts).~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 1 (randomization) up to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine1.9
Placebo and Cytarabine1.0
Clofarabine and Cytarabine - In Stratum < 6 Months1.4
Placebo and Cytarabine - In Stratum < 6 Months1.0
Clofarabine and Cytarabine In Stratum >= 6 Months2.0
Placebo and Cytarabine In Stratum >= 6 Months1.0

Event-free Survival by IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)

"Event-free survival (EFS) was defined as the time from randomization to the date of treatment failure, first disease recurrence (for participants who achieved remission), or death due to any cause, whichever occurred first.~Treatment Failure - ≥5% leukemic blasts by bone marrow exam, with no evidence of hematologic response (ie, <30% decrease in % leukemic blasts).~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 1 (randomization) up to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine1.9
Placebo and Cytarabine1.0
Clofarabine and Cytarabine - In Stratum < 6 Months1.1
Placebo and Cytarabine - In Stratum < 6 Months1.0
Clofarabine and Cytarabine In Stratum >= 6 Months2.8
Placebo and Cytarabine In Stratum >= 6 Months1.0

Four-Month Event-free Survival Per IRRP Assessment - Overall and by Calculated Strata (CSR 7-April-11)

"Four-month event-free survival (EFS) was defined as achieving an EFS of at least 122 days, where EFS is defined as the time from randomization to the date of treatment failure, first disease recurrence (for participants who achieved remission), or death due to any cause, whichever occurred first.~Treatment Failure - ≥5% leukemic blasts by bone marrow exam, with no evidence of hematologic response (ie, <30% decrease in % leukemic blasts).~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 1 (randomization) to Day 122

Interventionpercentage of participants (Number)
Clofarabine (IV Formulation) and Cytarabine37.7
Placebo and Cytarabine16.6
Clofarabine and Cytarabine - In Stratum < 6 Months35.2
Placebo and Cytarabine - In Stratum < 6 Months16.9
Clofarabine and Cytarabine In Stratum >= 6 Months40.5
Placebo and Cytarabine In Stratum >= 6 Months16.2

Four-Month Event-free Survival Per IRRP Assessment - Overall and by Randomized Strata (CSR 9-July-12)

"Four-month event-free survival (EFS) was defined as achieving an EFS of at least 122 days, where EFS is defined as the time from randomization to the date of treatment failure, first disease recurrence (for participants who achieved remission), or death due to any cause, whichever occurred first.~Treatment Failure - ≥5% leukemic blasts by bone marrow exam, with no evidence of hematologic response (ie, <30% decrease in % leukemic blasts).~Disease recurrence - reappearance of leukemic blasts in the peripheral blood, confirmed by ≥5% blasts in the bone marrow, and reappearance or development of pathologically proven extramedullary disease." (NCT00317642)
Timeframe: Day 1 (randomization) to Day 122

Interventionpercentage of participants (Number)
Clofarabine (IV Formulation) and Cytarabine38.9
Placebo and Cytarabine17.1
Clofarabine and Cytarabine - In Stratum < 6 Months31.4
Placebo and Cytarabine - In Stratum < 6 Months17.9
Clofarabine and Cytarabine In Stratum >= 6 Months47.4
Placebo and Cytarabine In Stratum >= 6 Months16.2

Overall Survival - Overall and by Calculated Strata (CSR 7-April-11)

Overall survival (OS) for the Full Analysis Set (FAS) and for the 2 calculated strata. OS was defined as the number of months from date of randomization until date of death due to any cause. (NCT00317642)
Timeframe: Day 1 (randomization) up to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine (FAS)6.6
Placebo and Cytarabine (FAS)6.4
Clofarabine and Cytarabine - In Stratum < 6 Months5.1
Placebo and Cytarabine - In Stratum < 6 Months5.5
Clofarabine and Cytarabine In Stratum >= 6 Months8.7
Placebo and Cytarabine In Stratum >= 6 Months7.2

Overall Survival - Overall and by Randomized Strata (CSR 9-July-12)

Overall survival (OS) for the Full Analysis Set (FAS) and for the 2 randomized strata. OS was defined as the number of months from date of randomization until date of death due to any cause. (NCT00317642)
Timeframe: Day 1 (randomization) up to approximately 4 years

Interventionmonths (Median)
Clofarabine (IV Formulation) and Cytarabine (FAS)6.6
Placebo and Cytarabine (FAS)6.3
Clofarabine and Cytarabine - In Stratum < 6 Months4.8
Placebo and Cytarabine - In Stratum < 6 Months6.3
Clofarabine and Cytarabine In Stratum >= 6 Months9.7
Placebo and Cytarabine In Stratum >= 6 Months6.6

Best Response Per Independent Response Review Panel (IRRP) Assessment - Overall and by Calculated Strata (CSR 7-April-11)

"Percentage of participants whose best response was assessed by the IRRP as complete remission (CR) or complete remission with incomplete peripheral blood count recovery (CRi) using the revised International Working Group for Response Criteria (Cheson 2003).~CR is defined on morphologic criteria at a single response assessment:~a bone marrow aspirate or biopsy of <5% blasts, with evidence of normal hematopoiesis;~absence of Auer rods in the blasts that are present;~absence of extramedullary disease;~absence of a unique phenotype determined at the pretreatment specimen, as assessed by immunophenotyping;~only rare evidence of circulating blasts. If present, evidence of a regenerating bone marrow;~recovery of peripheral counts (platelets ≥100*10^9/L and absolute neutrophil count (ANC) ≥1.0*10^9/L).~CRi met all criteria for CR except for either residual neutropenia (ANC <1.0*10^9/L) or thrombocytopenia (platelet count <100*10^9/L)." (NCT00317642)
Timeframe: Day 12 up to approximately 6 months

,,,,,
Interventionpercentage of participants (Number)
Overall Remission (CR + CRi)Complete Remission (CR)CR with incomplete blood count recovery (CRi)
Clofarabine (IV Formulation) and Cytarabine46.935.211.7
Clofarabine and Cytarabine - In Stratum < 6 Months45.533.012.5
Clofarabine and Cytarabine In Stratum >= 6 Months48.637.810.8
Placebo and Cytarabine22.917.85.1
Placebo and Cytarabine - In Stratum < 6 Months22.918.14.8
Placebo and Cytarabine In Stratum >= 6 Months23.017.65.4

Participants With Adverse Events (CSR 7-April-11)

"Number of participants with treatment emergent adverse events (TEAEs) or death due to related AE. Related AEs for the combination arm can be related to either clofarabine or cytarabine.~Grade 1 = Mild AE, Grade 2 = Moderate AE, Grade 3 = Severe AE, Grade 4 = Life Threatening AE, Grade 5 = Death" (NCT00317642)
Timeframe: Day 1 up to a maximum of 4 years (includes up to a maximum of 3 cycles of therapy plus 45 days follow up. Related AEs are followed to resolution.)

,
Interventionparticipants (Number)
Any Treatment Emergent AEAny Related Treatment Emergent AEAny Treatment Emergent Grade >=3 AEAny Related Treatment Related Grade >=3 AEDiscontinue of study medication due to AEDiscontinue of study medication due to related AE
Clofarabine (IV Formulation) and Cytarabine1611571571271714
Placebo and Cytarabine1551331338353

>80% Inhibition of FLT3 Phosphorylation in a Majority of Post-treatment Trough Time Points

FLT3 inhibition is determined in patients receiving lestaurtinib by measuring FLT3 plasma inhibitory activity (PIA). For PIA predictive modeling, a random effects linear regression model will be used to describe the relationship between PIA and Pharmacokinetic (PK) levels for each of the 5 trough plasma samples collected for each patient (NCT00469859)
Timeframe: Course 1 day 7, day 14, day 21, and day 28.

Interventionparticipants (Number)
Group 1 (Lestaurtinib Dose 50 mg/m25
Group 2 (Lestaurtinib: Dose 62.5 mg/m25

Dose-limiting Toxicity

Number of patients with dose-limiting toxicity (DLT) (NCT00469859)
Timeframe: 28 days

Interventionparticipants (Number)
Group 1 (Lestaurtinib Dose 50 mg/m20
Group 2 (Lestaurtinib: Dose 62.5 mg/m20

Event-Free Survival Probability During the 3 Years After Transplant

Event-free survival (EFS) is survival without death or disease activity from any one of the following criteria: 1) loss of neurological function, defined as a change in pretransplant Extended Disability Status Scale (EDSS) of > 0.5. 2) Relapse, defined as the development of a new neurological sign and corresponding symptom, or worsening of an existing neurological sign and symptom, localized to central nervous system white matter, resulting in neurological deficit/disability, and lasting over 48 hours. 3) New lesions on magnetic resonance imaging (MRI), defined as presence of 2 or more independent multiple sclerosis brain lesions detected on MRI 1 year or more after stem cell transplant. Kaplan-Meier estimates of survival probability, with 90% confidence interval based on Greenwood's formula for standard error. (NCT00288626)
Timeframe: 3 years

InterventionProbability (Number)
HDIT and HCT0.784

Event-Free Survival Probability During the 5 Years After Transplant

Event-free survival (EFS) is survival without death or disease activity from any one of the following criteria: 1) loss of neurological function, defined as a change in pretransplant Extended Disability Status Scale (EDSS) of > 0.5. 2) Relapse, defined as the development of a new neurological sign and corresponding symptom, or worsening of an existing neurological sign and symptom, localized to central nervous system white matter, resulting in neurological deficit/disability, and lasting over 48 hours. 3) New lesions on magnetic resonance imaging (MRI), defined as presence of 2 or more independent multiple sclerosis brain lesions detected on MRI 1 year or more after stem cell transplant. Kaplan-Meier estimates of survival probability, with 90% confidence interval based on Greenwood's formula for standard error. (NCT00288626)
Timeframe: 5 years

InterventionProbability (Number)
HDIT and HCT0.692

Percent of Participants Who Experienced All-Cause Morbidity Within 12 Months of Post-HCT

Morbidity is the occurrence of NCI Common Terminology Criteria for Adverse Events (CTCAE) v3.0 adverse event grade 3 or higher. (NCT00288626)
Timeframe: From the time of Autologous CD34+ Hematopoietic Stem Cell Transplant (HCT) to 1 year after HCT.

InterventionPercentage of Participants (Number)
HDIT and HCT95.8

Percent of Participants Who Experienced All-Cause Morbidity

Morbidity is the occurrence of NCI Common Terminology Criteria for Adverse Events (CTCAE) v3.0 adverse event grade 3 or higher. (NCT00288626)
Timeframe: From the time of enrollment until completion of the 5-year follow-up, an average of 6 years.

Interventionpercentage of participants (Number)
HDIT and HCT100

Time to Neutrophil Engraftment

Neutrophil engraftment, or neutrophil count recovery, is defined as an Absolute Neutrophil Count (ANC) > 500/ μL for 2 consecutive measurements on different days. Normal range is 1500 to 8000/μL. Reference: http://www.medicinenet.com (NCT00288626)
Timeframe: From time of graft infusion to time of engraftment, up to 6 years

InterventionDays (Mean)
HDIT and HCT10.8

Time to Platelet Engraftment

Platelet engraftment, or platelet count recovery, is defined as Platelets > 20,000/μL for two consecutive measurements on different days with no platelet transfusions in the preceding 7 days. Normal range is 150,000-450,000/μL. Reference: http://www.hopkinsmedicine.org/heart_vascular_institute/clinical_services/centers_excellence/womens_cardiovascular_health_center/patient_information/health_topics/platelets.html. (NCT00288626)
Timeframe: From time of graft infusion to time of engraftment, up to 6 years

InterventionDays (Mean)
HDIT and HCT18.5

Change From Baseline in Extended Disability Status Scale (EDSS)

Kurtzke's Expanded Disability Status Scale (EDSS) assesses disability in Multiple Sclerosis patients. Eight functional systems are evaluated: visual, brain stem, pyramidal, cerebellar, sensory, bowel and bladder, cerebral, and ambulation. The overall score ranges from 0 (normal neurological exam) to 10 (death due to MS). Change from baseline was computed as the value at the time point minus the baseline value. A negative value in change from baseline indicates an improvement and a positive value indicates worsening. A change of > 0.5 in EDSS was a treatment-failure criterion. (NCT00288626)
Timeframe: 6 months to 5 years after HCT

Interventionunits on a scale (Mean)
6 Months Post Transplant1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT-0.3-0.7-0.8-0.8-0.6-0.9

Change From Baseline in Number of Gadolinium-Enhanced Lesions

Multiple sclerosis disease-related lesions were assessed by gadolinium-enhanced magnetic resonance imaging (MRI). Change from baseline was computed as the value at the time point minus the baseline value. A negative value in change from baseline indicates an improvement and a positive value indicates worsening. (NCT00288626)
Timeframe: 8 weeks to 5 years after HCT

InterventionLesions per scan (Mean)
8 Weeks Post Transplant6 Months Post Transplant1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT-2.1-2.3-2.5-2.5-1.3-2.2-2.7

Change From Baseline in T1-Weighted Lesion Volume

A T1-weighted magnetic resonance imaging (MRI) scan was used to assess the volume of T1 lesions in the brain. Change from baseline was computed as the value at the time point minus the baseline value. (NCT00288626)
Timeframe: 8 weeks to 5 years after HCT

Interventionmilliliter (Mean)
8 Weeks Post Transplant6 Months Post Transplant1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT-0.10.00.20.30.40.40.3

Change From Baseline in T2-Weighted Lesion Volume

A T2-weighted magnetic resonance imaging (MRI) scan was used to assess the volume of T2 lesions in the brain. Change from baseline was computed as the value at the time point minus the baseline value. (NCT00288626)
Timeframe: 8 weeks to 5 years after HCT

Interventionmilliliters (Mean)
8 Weeks Post Transplant6 Months Post Transplant1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT-0.8-0.7-1.0-1.6-1.9-1.9-2.3

Disease-Modifying Therapy Survival Probability After Transplant

Treatment with disease-modifying therapy was measured by the number of days from transplant to the first treatment with an additional disease-modifying therapy. Examples of therapy include interferon beta-1a, glatiramer acetate, natalizumab, alemtuzumab, other immunosuppressive medications, or experimental therapies directed against MS activity. Kaplan-Meier estimates of survival probability, with 90% confidence interval based on Greenwood's formula for standard error. (NCT00288626)
Timeframe: 1 to 5 years after HCT

InterventionProbability (Number)
1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT1.01.01.01.00.950

Event-Free Survival Probability After Transplant

Event-free survival (EFS) is survival without death or disease activity from any one of the following criteria: 1) loss of neurological function, defined as a change in pretransplant Extended Disability Status Scale (EDSS) of > 0.5. 2) Relapse, defined as the development of a new neurological sign and corresponding symptom, or worsening of an existing neurological sign and symptom, localized to central nervous system white matter, resulting in neurological deficit/disability, and lasting over 48 hours. 3) New lesions on magnetic resonance imaging (MRI), defined as presence of 2 or more independent multiple sclerosis brain lesions detected on MRI 1 year or more after stem cell transplant. Kaplan-Meier estimates of survival probability, with 90% confidence intervals based on Greenwood's formula for standard error. (NCT00288626)
Timeframe: 1, 2, and 4 years after HCT

InterventionProbability (Number)
1 Year Post Transplant2 Years Post Transplant4 Years Post Transplant
HDIT and HCT0.9580.8280.738

MRI Activity-Free Survival Probability After Transplant

MS disease activity is measured as days from transplant to first occurrence of >= 2 new MS lesions on Magnetic resonance imaging (MRI) relative to baseline. Kaplan-Meier estimates of survival probability, with 90% confidence intervals based on Greenwood's formula for standard error. (NCT00288626)
Timeframe: 1 to 5 years after HCT

InterventionProbability (Number)
1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT0.9580.9580.9580.9100.863

MS Progression-Free Survival Probability After Transplant

"MS progression is measured as number of days from transplant to first Kurtzke's Expanded Disability Status Scale (EDSS) increase of more than 0.5 relative to the baseline measurement. EDSS assesses disability in Multiple Sclerosis patients. Eight functional systems are evaluated: visual, brain stem, pyramidal, cerebellar, sensory, bowel and bladder, cerebral, and ambulation. The overall score ranges from 0 (normal neurological exam) to 10 (death due to MS).~Kaplan-Meier estimates of survival probability, with 90% confidence intervals based on Greenwood's formula for standard error." (NCT00288626)
Timeframe: 1 to 5 years after HCT

InterventionProbability (Number)
1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT1.00.9130.9130.9130.913

MS Relapse-Free Survival Probability After Transplant

"MS clinical relapse is defined as the development of a new neurological sign and corresponding symptom, or worsening of an existing neurological sign and symptom, localized to central nervous system white matter, resulting in neurological deficit or disability, and lasting over 48 hours. Clinical relapse was determined by the participant's neurologist and was measured as days from transplant to new or worsening neurological symptom relative to baseline.~Kaplan-Meier estimates of survival probability, with 90% confidence interval based on Greenwood's formula for standard error." (NCT00288626)
Timeframe: 1 to 5 years after HCT

InterventionProbability (Number)
1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT0.9580.9150.8690.8690.869

Number of New T2-Weighted Lesions From Baseline

A T2-weighted magnetic resonance imaging (MRI) scan was used to determine the number of new T2 lesions in the brain relative to Baseline. A value of 0 means that the participant didn't worsen. Values greater than 0 indicate an increase in disease activity from baseline. (NCT00288626)
Timeframe: 6 Months to 5 years after HCT

InterventionLesions per scan (Mean)
6 Months Post Transplant1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT0.20.20.20.10.40.1

Overall Survival

The probability that a participant did not experienced a death estimated at 1, 2, 3, 4, and 5 years following transplant via the Kaplan-Meier Method. Greenwood's formula for standard error was used to calculate 90% confidence intervals. Participants that did not die were censored at the time of last follow-up. (NCT00288626)
Timeframe: From study entry to death, loss to follow-up, or the end of the study, whichever came first, up to 6 years

InterventionProbability (Number)
1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT1.01.00.9570.9110.863

Percent Change From Screening in Brain Volume

Magnetic resonance imaging (MRI) scan techniques measured ventricular volumes and grey and white matter brain volumes. Change from screening was computed as the value at the time point minus the screening value. (NCT00288626)
Timeframe: 8 weeks to 5 years after HCT

InterventionPercent Change (Mean)
8 Weeks Post Transplant6 Months Post Transplant1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT-0.8-1.1-1.2-1.6-2.2-2.0-2.3

Survival From MS-Related Mortality

The probability that a participant did not experienced a MS-related death estimated at 1, 2, 3, 4, and 5 years following transplant via the Kaplan-Meier Method. Greenwood's formula for standard error was used to calculate 90% confidence intervals. Participants that did not experience a MS-related death were censored at the time of last follow-up. A MS-related death was defined as death that occurred at any time after study entry and that was possibly, probably, or definitely related to disease progression. (NCT00288626)
Timeframe: From study entry to death, loss to follow-up, or the end of the study, whichever came first, up to 6 years

InterventionProbability (Number)
1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT1.01.00.9570.9570.906

Survival From Treatment-Related Mortality

The probability that a participant did not experienced a treatment-related death estimated at 1, 2, 3, 4, and 5 years following transplant via the Kaplan-Meier Method. Greenwood's formula for standard error was used to calculate 90% confidence intervals. Participants that did not experience a treatment-related death were censored at the time of last follow-up. A treatment-related death was defined as death that occurred at any time after study entry and that was possibly, probably, or definitely related to the cellular product or possibly, probably, or definitely related to mobilization of autologous peripheral blood hematopoietic progenitor cells with G-CSF and prednisone or to the high-dose immunosuppressive therapy. There were no treatment-related mortality events in the study. (NCT00288626)
Timeframe: From study entry to death, loss to follow-up, or the end of the study, whichever came first, up to 6 years

InterventionProbability (Number)
1 Year Post Transplant2 Years Post Transplant3 Years Post Transplant4 Years Post Transplant5 Years Post Transplant
HDIT and HCT1.01.01.01.01.0

Response

Any patient who is enrolled and receives at least one dose of cytarabine will be considered evaluable for response if (1) the patient demonstrates progressive disease while on protocol therapy or (2) the patient is observed on protocol therapy for at least one cycle. Patients who achieve a complete or partial response according to the RECIST (Response Evaluation Criteria In Solid Tumors) criteria will be considered responders for the study design. All other patients who are evaluable for response will be considered non-responders for the study. (NCT00470275)
Timeframe: the first six cycles of study chemotherapy (126 days)

Interventionparticipants (Number)
Non ResponderResponder
Cytarabine100

Maximum Tolerated Dose of Bortezomib (Phase I)

Determine the safety and the maximum tolerated dose (MTD) of bortezomib when added to the combination of rituximab, methotrexate, and cytarabine alternating with bortezomib, rituximab-hyperCVAD in patients with untreated aggressive mantle cell lymphoma. (NCT00477412)
Timeframe: Cycle 1 Day 1 - End of Cycle 2 up to 60 days.

Interventionmg/m^2 (Number)
Phase 1 Maximum Tolerated Dose (MTD)1.3

Number of Participants With Overall Response Rate

Response rate is determined by CT scans of the chest, abdomen, and pelvis, unilateral BM biopsy and aspirate with lymphoma markers (if initially positive) and colonoscopy/endoscopy if applicable. (NCT00477412)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Phase II- Treatment (Combination Chemotherapy)87

Overall Survival

Overall survival is the time in number of months from start of study treatment to date of death due to any cause. (NCT00477412)
Timeframe: Date of diagnosis to last known date of survival, up to 5 years

Interventionmonths (Median)
Phase II- Treatment (Combination Chemotherapy)44

Time to Failure (Phase II)

Failure will be defined as recurrence/progression of disease or death from either disease or toxicity. Bayesian toxicity monitoring schema will be used to monitor severe toxicity profile in combined therapy. Severe toxicity is defined as at least two episodes of neutropenic fever during treatment courses. (NCT00477412)
Timeframe: First date of diagnosis up to 5 years

Interventionmonths (Median)
Phase II- Treatment (Combination Chemotherapy)55

Number of Patients With Complete or Partial Response at Day 30

Definition of disease status is based on the article of Revised Response Criteria for Malignant Lymphoma Response Definitions for Clinical Trials (Cheson et al, 2007). Tests used for evaluation of disease status would be physical examination, laboratory testing, bone marrow testing, bone marrow biopsy and aspirate, PET scan, and CT scans of neck, chest, abdomen and pelvis as indicated. (NCT02366663)
Timeframe: Day 0 to Day +30 post-HCT

InterventionParticipants (Count of Participants)
Arm I (ZBEAM)1
Arm II (BEAM)1

Time to Neutrophil Engraftment

Time to neutrophil engraftment will be compared between treatment arms using a log-rank test, and the cumulative incidence curves will be estimated. (NCT02366663)
Timeframe: Day 0 to Day 100 post-HCT

Interventiondays (Median)
Arm I (ZBEAM)11
Arm II (BEAM)10

Time to Platelet Engraftment

Time to platelet engraftment will be compared between treatment arms using a log-rank test, and the cumulative incidence curves will be estimated. (NCT02366663)
Timeframe: Day 0 to Day 100 post-HCT

Interventiondays (Median)
Arm I (ZBEAM)22.5
Arm II (BEAM)26

Number of Patients With Grade 3-5 Toxicities Graded by the NCI CTCAE Version 4.0

Observed toxicities will be summarized in terms of type and severity. In accordance with the secondary study objectives, descriptive analyses on these data will be performed. (NCT02366663)
Timeframe: Day -21 to Day +100 post-HCT

,
InterventionParticipants (Count of Participants)
AnemiaFebrile neutropeniaHyperglycemiaHyponatremiaINR increasedLymphocyte count decreasedMucositis oralNeutrophil count decreasedObesityOral painPharyngeal mucositisPlatelet count decreasedSore ThroatWhite blood cell decreased
Arm I (ZBEAM)22111222100202
Arm II (BEAM)11000111111111

Clinical Response to Crenolanib With Standard Salvage Chemotherapy

To determine the response rate to crenolanib. Complete remission (CR) response criteria include a post-baseline bone marrow (BM) biopsy or aspiration % blasts <5%, absolute neutrophil count (ANC) >1×10^9/L and platelet count >100×10^9/L. CRi response included all CR criteria met, except participant did not have either platelet recovery or ANC recovery. CRh response included all CR criteria met, except subject only has partial platelet recovery and ANC recovery. Complete CR (CRc) response includes all subjects who achieve a CR, CRi and CRh. Partial Response (PR) response included a decrease of ≥50% in % blasts in the BM aspirate or biopsy from baseline but >5%. Morphologic Leukemia-Free State (MLFS) response included ≤5% in % blasts in the BM aspirate or biopsy. (NCT02626338)
Timeframe: 1 year

,,,
InterventionParticipants (Count of Participants)
Composite complete remission (CR+CRh+CRi)MLFSClinical benefit (CRc+PR+MLFS)
All Subjects737
Arm A: HAM Chemotherapy323
Arm B: FLAG-Ida Chemotherapy404
Arm C: MEC Chemotherapy010

Event-free Survival (EFS)

"Event-free survival is defined as the time from randomization to the date of progressive disease (PD), confirmed morphologic relapse from CR or CRi, treatment failure (failure to achieve CR, CRi or morphologic leukemia free state (MLFS) after at least 6 cycles of study treatment), or death from any cause, assessed by the investigator according to the modified IWG criteria.~PD:~> 50% increase in marrow blasts (minimum 15% increase required if blasts < 30% at baseline); or persistent marrow blast > 70% for ≥ 3 months; without at least a 100% improvement in ANC to an absolute level > 0.5 × 10⁹/L, and/or platelets to > 50 × 10⁹/L non-transfused; or~50% increase in peripheral blasts to > 25 × 10⁹/L; or~New extramedullary disease~Participants with no events prior to the cut-off date were censored at their last assessment date; participants with no events prior to post-treatment therapy initiated before the cut-off date were censored on the start date of post-treatment therapy." (NCT03069352)
Timeframe: From randomization until the primary analysis cut-off date of February 15, 2019; the median follow-up time was 12.0 months (range: 0.2-17.0) in the placebo arm and 12.0 months (range: 0.1-17.6) in the venetoclax arm.

Interventionmonths (Median)
Placebo + Low Dose Cytarabine (LDAC)2.0
Venetoclax + Low Dose Cytarabine (LDAC)4.7

Overall Survival (OS)

Overall survival is defined as the time from the date of randomization to the date of death. Participants who had not died were censored at the date they were last known to be alive on or before the cutoff date. Overall survival was analyzed using Kaplan-Meier methodology. (NCT03069352)
Timeframe: From randomization until the primary analysis cut-off date of February 15 2019; the median follow-up time was 12.0 months (range: 0.2-17.0) in the placebo arm and 12.0 months (range: 0.1-17.6) in the venetoclax arm.

Interventionmonths (Median)
Placebo + Low Dose Cytarabine (LDAC)4.1
Venetoclax + Low Dose Cytarabine (LDAC)7.2

Overall Survival After an Additional 6-Months Follow-up

Overall survival is defined as the time from the date of randomization to the date of death. Participants who had not died were censored at the date they were last known to be alive on or before the cutoff date. Overall survival was analyzed using Kaplan-Meier methodology. (NCT03069352)
Timeframe: From randomization until the 6-month follow-up analysis cut-off date of August 15, 2019; the median follow-up time was 17.7 months (range: 0.2-20.8) in the placebo arm and 17.5 months (range: 0.1-23.5) in the venetoclax arm.

Interventionmonths (Median)
Placebo + Low Dose Cytarabine (LDAC)4.1
Venetoclax + Low Dose Cytarabine (LDAC)8.4

Percentage of Participants With Complete Remission and Complete Remission With Partial Hematologic Recovery (CR + CRh) by Initiation of Cycle 2

"The percentage of participants who achieved a complete remission (CR) or complete remission with partial hematologic recovery (CRh) before initiation of Cycle 2 assessed by the investigator.~CR is defined according to the revised guidelines by the IWG for AML as no morphologic evidence of AML, ANC count ≥ 10³/μL, platelets ≥ 10⁵/μL, RBC transfusion independence, and bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~CRh is a derived response based on bone marrow blast and hematology lab values, achieved when when the following criteria are met:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count of > 0.5 × 10³/μL and~Peripheral blood platelet count of > 0.5 × 10⁵/μL and~A 1-week platelet transfusion-free period prior to the hematology lab collection.~Participants with no disease assessments were considered to be non-responders." (NCT03069352)
Timeframe: Cycle 1, 28 days

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)4.4
Venetoclax + Low Dose Cytarabine (LDAC)30.8

Percentage of Participants With Complete Remission or Complete Remission With Incomplete Blood Count Recovery (CR + CRi) and Minimal Residual Disease (MRD) Response

"The percentage of participants with complete remission or complete remission with incomplete blood count recovery AND minimal residual disease (MRD) as assessed by the investigator. Response was based on the modified IWG response criteria for AML:~CR: No morphologic evidence of AML and ANC ≥ 10³/μL, platelets ≥ 10⁵/μL, red blood cell (RBC) transfusion independence, and bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~CRi: All criteria as CR except for residual neutropenia < 10³/μL or thrombocytopenia < 10⁵/μL. If all criteria for CR are met except RBC transfusion independence, CRi criteria are met.~MRD response (at lowest-point MRD value) was defined as having less than 10-³ residual blasts per leukocyte measured in the bone marrow, per European LeukemiaNet (ELN) recommendations.~Participants who had no disease or MRD assessments were considered to be non-responders." (NCT03069352)
Timeframe: Response was assessed at the end of Cycle 1 and every 3 cycles thereafter to the end of treatment. Median treatment duration at the 15 February 2019 cut-off date was 1.7 months (range: 0.1-14.2) and 3.9 months (range: 0.0-17.1) in each group respectively.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)1.5
Venetoclax + Low Dose Cytarabine (LDAC)5.6

Percentage of Participants With Complete Remission or Complete Remission With Incomplete Blood Count Recovery (CR + CRi) by Initiation of Cycle 2

"The composite complete remission rate is defined as the percentage of participants with complete remission (CR) or complete remission with incomplete blood count recovery (CRi) before initiation of Cycle 2, assessed by the investigator. Response was based on physical examination, bone marrow results and hematology values according to the revised guidelines by the IWG for AML:~CR: No morphologic evidence of AML and absolute neutrophil count ≥ 10³/μL, platelets ≥ 10⁵/μL, red cell transfusion independence, and bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~CRi: All criteria as CR except for residual neutropenia < 10³/μL or thrombocytopenia < 10⁵/μL. If all criteria for CR are met except for RBC transfusion independence, CRi criteria are met.~Participants who had no IWG disease assessments were considered to be non-responders." (NCT03069352)
Timeframe: Cycle 1, 28 days

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)2.9
Venetoclax + Low Dose Cytarabine (LDAC)34.3

Percentage of Participants With Complete Remission or Complete Remission With Incomplete Blood Count Recovery (CR + CRi)

"The composite complete remission rate is defined as the percentage of participants with complete remission (CR) or complete remission with incomplete blood count recovery (CRi) at any time during the study as assessed by the investigator. Response was based on physical examination, bone marrow results and hematology values according to the revised guidelines by the International Working Group (IWG) for AML:~CR: No morphologic evidence of AML and absolute neutrophil count (ANC) ≥ 10³/μL (1,000/μL), platelets ≥ 10⁵/μL (100,000/μL), red blood cell (RBC) transfusion independence, and bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~CRi: All criteria as CR except for residual neutropenia < 10³/μL or thrombocytopenia < 10⁵/μL. If all criteria for CR are met except RBC transfusion independence, the CRi criteria are met.~Participants who had no IWG disease assessments were considered to be non-responders." (NCT03069352)
Timeframe: Response was assessed at the end of Cycle 1 and every 3 cycles thereafter to the end of treatment. Median treatment duration at the 15 February 2019 cut-off date was 1.7 months (range: 0.1-14.2) and 3.9 months (range: 0.0-17.1) in each group respectively.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)13.2
Venetoclax + Low Dose Cytarabine (LDAC)47.6

Percentage of Participants With Complete Remission or Complete Remission With Partial Hematologic Recovery (CR + CRh) and Minimal Residual Disease (MRD) Response

"The percentage of participants with complete remission or complete remission with partial hematologic recovery (CRh) AND MRD response as assessed by the investigator.~CR is defined according to the modified IWG response criteria for AML as no morphologic evidence of AML, ANC ≥ 10³/μL, platelets ≥ 10⁵/μL, RBC transfusion independence, and bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~CRh is achieved when the following criteria are met:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count of > 0.5 × 10³/μL and~Peripheral blood platelet count of > 0.5 × 10⁵/μL and~A 1 week platelet transfusion-free period prior to the hematology lab collection.~MRD response (at lowest-point MRD value) was defined as less than 10-³ residual blasts per leukocyte measured in the bone marrow, per ELN recommendations.~Participants who had no disease or MRD assessments were considered to be non-responders." (NCT03069352)
Timeframe: Response was assessed at the end of Cycle 1 and every 3 cycles thereafter to the end of treatment. Median treatment duration at the 15 February 2019 cut-off date was 1.7 months (range: 0.1-14.2) and 3.9 months (range: 0.0-17.1) in each group respectively.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)1.5
Venetoclax + Low Dose Cytarabine (LDAC)5.6

Percentage of Participants With Complete Remission or Complete Remission With Partial Hematologic Recovery (CR + CRh)

"The percentage of participants who achieved a complete remission (CR) or complete remission with partial hematologic recovery (CRh) at any time point during the study assessed by the investigator.~CR is defined according to the revised guidelines by the IWG for AML as no morphologic evidence of AML, ANC count ≥ 10³/μL, platelets ≥ 10⁵/μL, RBC transfusion independence, and bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~CRh is a derived response based on bone marrow blast and hematology lab values, achieved when the following criteria are met:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count of > 0.5 × 10³/μL and~Peripheral blood platelet count of > 0.5 × 10⁵/μL and~A 1 week platelet transfusion-free period prior to the hematology lab collection.~Participants with no disease assessments were considered to be non-responders." (NCT03069352)
Timeframe: Response was assessed at the end of Cycle 1 and every 3 cycles thereafter to the end of treatment. Median treatment duration at the 15 February 2019 cut-off date was 1.7 months (range: 0.1-14.2) and 3.9 months (range: 0.0-17.1) in each group respectively.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)14.7
Venetoclax + Low Dose Cytarabine (LDAC)46.9

Percentage of Participants With Complete Remission

"The complete remission rate is defined as the percentage of participants with complete remission (CR) at any time during the study as assessed by the investigator. Response was based on physical examination, bone marrow results and hematology values according to the revised guidelines by the International Working Group (IWG) for AML. CR is defined as no morphologic evidence of AML and absolute neutrophil count ≥ 10³/μL, platelets ≥ 10⁵/μL, red blood cell transfusion independence, and bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~Participants who had no IWG disease assessments were considered to be non-responders." (NCT03069352)
Timeframe: Response was assessed at the end of Cycle 1 and every 3 cycles thereafter to the end of treatment. Median treatment duration at the 15 February 2019 cut-off date was 1.7 months (range: 0.1-14.2) and 3.9 months (range: 0.0-17.1) in each group respectively.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)7.4
Venetoclax + Low Dose Cytarabine (LDAC)27.3

Percentage of Participants With Platelet Transfusion Independence Among Those Who Were Transfusion Dependent at Baseline

The rate of conversion was calculated as the percentage of participants who were post-baseline platelet transfusion independent among participants who had a platelet transfusion within 8 weeks prior to the first dose of study drug (i.e. were transfusion dependent at Baseline). Platelet transfusion independence is defined as a period of at least 56 days with no platelet transfusion after the first dose of study drug and on or before the last dose of study drug plus 30 days, or disease progression, or confirmed morphological relapse, or death, or the data cut-off date, whichever occurred earlier. (NCT03069352)
Timeframe: From first dose of study drug until 30 days after last dose up to the data cut-off date of 15 February 2019; Median time on treatment was 1.7 months (range: 0.1-14.2) in the placebo arm and 3.9 months (range: 0.0-17.1) in the venetoclax arm.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)12.5
Venetoclax + Low Dose Cytarabine (LDAC)30.2

Percentage of Participants With Post Baseline Platelet Transfusion Independence

The post baseline platelet transfusion independence rate was calculated as the percentage of participants who achieved platelet transfusion independence post Baseline. Platelet transfusion independence is defined as a period of at least 56 consecutive days with no platelet transfusion after the first dose of study drug and on or before the last dose of study drug plus 30 days, or disease progression, or confirmed morphological relapse, or death, or the data cut--off date, whichever occurred earlier. (NCT03069352)
Timeframe: From first dose of study drug until 30 days after last dose up to the data cut-off date of 15 February 2019; Median time on treatment was 1.7 months (range: 0.1-14.2) in the placebo arm and 3.9 months (range: 0.0-17.1) in the venetoclax arm.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)32.4
Venetoclax + Low Dose Cytarabine (LDAC)47.6

Percentage of Participants With Post Baseline Red Blood Cell (RBC) Transfusion Independence

The post baseline red blood cell (RBC) transfusion independence rate was calculated as the percentage of participants who achieved RBC transfusion independence post baseline. RBC transfusion independence is defined as a period of at least 56 consecutive days with no RBC transfusion after the first dose of study drug and on or before the last dose of study drug plus 30 days, or disease progression, or confirmed morphological relapse, or death, or the data cut-off date, whichever occurred earlier. (NCT03069352)
Timeframe: From first dose of study drug until 30 days after last dose up to the data cut-off date of 15 February 2019; Median time on treatment was 1.7 months (range: 0.1-14.2) in the placebo arm and 3.9 months (range: 0.0-17.1) in the venetoclax arm.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)17.6
Venetoclax + Low Dose Cytarabine (LDAC)40.6

Percentage of Participants With RBC Transfusion Independence Among Those Who Were Transfusion Dependent at Baseline

The rate of conversion was calculated as the percentage of participants who were post-baseline RBC transfusion independent among participants who had an RBC transfusion within 8 weeks prior to the first dose of study drug (i.e. were transfusion dependent at Baseline). RBC transfusion independence is defined as a period of at least 56 days with no RBC transfusion after the first dose of study drug and on or before the last dose of study drug plus 30 days, or disease progression, or confirmed morphological relapse, or death, or the data cut-off date, whichever occurred earlier. (NCT03069352)
Timeframe: From first dose of study drug until 30 days after last dose up to the data cut-off date of 15 February 2019; Median time on treatment was 1.7 months (range: 0.1-14.2) in the placebo arm and 3.9 months (range: 0.0-17.1) in the venetoclax arm.

Interventionpercentage of participants (Number)
Placebo + Low Dose Cytarabine (LDAC)15.1
Venetoclax + Low Dose Cytarabine (LDAC)37.5

Change From Baseline in Global Health Status / Quality of Life

"The European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core (EORTC QLQ-C30) consists of a Global Health Status/Quality of Life (GHS/QoL) scale, a Financial Difficulties scale, 5 functional scales (Cognitive, Social, Physical, Emotional, and Role Functioning), and 8 symptom scales/items (Fatigue, Insomnia, Appetite Loss, Pain, Constipation, Diarrhea, Dyspnea, and Nausea and Vomiting).~The GHS/QoL scale includes 2 questions in which participants were asked to rate their overall health and overall quality of life during the past week on a scale from 1 (very poor) to 7 (excellent). The 2 scores were averaged and transformed to a scale from 0 to 100, where a high score represents a high QoL. A positive change from baseline indicates better quality of life." (NCT03069352)
Timeframe: Baseline and Day 1 of Cycles 3, 5, 7, and 9

,
Interventionscores on a scale (Least Squares Mean)
Cycle 3 Day 1Cycle 5 Day 1Cycle 7 Day 1Cycle 9 day 1
Placebo + Low Dose Cytarabine (LDAC)1.9412.6273.4816.918
Venetoclax + Low Dose Cytarabine (LDAC)4.85716.01510.59913.299

Change From Baseline in Patient Reported Outcomes Measurement Information System (PROMIS) Fatigue Short Form (SF) 7a

PROMIS Fatigue SF 7a is a seven-item questionnaire that assesses the impact and experience of fatigue over the past 7 days. All questions employ the following five response options: 1 = Never, 2 = Rarely, 3 = Sometimes, 4 = Often, and 5 = Always. The PROMIS Fatigue 7a score is calculated as a T-score, which is a standardized score with a mean of 50 (based on the average for the United States general population) and a standard deviation (SD) of 10. Higher scores indicate higher levels of fatigue. A decrease in score (negative change from Baseline) indicates improvement in fatigue; the minimum important difference used in this study was 3 points. (NCT03069352)
Timeframe: Baseline and Day 1 of Cycles 3, 5, 7, and 9

,
Interventionscore on a scale (Least Squares Mean)
Cycle 3 Day 1Cycle 5 Day 1Cycle 7 Day 1Cycle 9 day 1
Placebo + Low Dose Cytarabine (LDAC)1.567-0.336-3.818-3.453
Venetoclax + Low Dose Cytarabine (LDAC)-2.940-5.259-4.625-5.101

Overall Survival (OS) by Mutation Subgroups

"Overall survival is defined as the time from the date of randomization to the date of death. Participants who had not died were censored at the date they were last known to be alive on or before the cutoff date. Overall survival was analyzed using Kaplan-Meier methodology.~Overall survival was analyzed in participants with the following molecular markers:~Isocitrate dehydrogenase 1 and 2 (IDH1/2) mutation~FMS (Feline McDonough Sarcoma)-like tyrosine kinase 3 (FLT3) mutation" (NCT03069352)
Timeframe: From randomization until the primary analysis cut-off date of February 15, 2019; the median follow-up time was 12.0 months (range: 0.2-17.0) in the placebo arm and 12.0 months (range: 0.1-17.6) in the venetoclax arm.

,
Interventionmonths (Median)
IDH1/2 mutationFLT3 mutation
Placebo + Low Dose Cytarabine (LDAC)9.09.8
Venetoclax + Low Dose Cytarabine (LDAC)10.85.9

Percentage of Participants With Complete Remission or Complete Remission With Incomplete Blood Count Recovery (CR + CRi) by Mutation Subgroup

"Response was based on physical examination, bone marrow results and hematology values according to the revised guidelines by the IWG for AML:~CR: No morphologic evidence of AML and ANC ≥ 10³/μL, platelets ≥ 10⁵/μL, RBC transfusion independence, and bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~CRi: All criteria as CR except for residual neutropenia < 10³/μL or thrombocytopenia < 10⁵/μL. If all criteria for CR are met except RBC transfusion independence, CRi criteria is met.~Participants who had no IWG disease assessments were considered to be non-responders.~Response was analyzed in participants with the following mutations:~Isocitrate dehydrogenase 1 and 2 (IDH1/2) mutation~FMS-like tyrosine kinase 3 (FLT3) mutation" (NCT03069352)
Timeframe: Response was assessed at the end of Cycle 1 and every 3 cycles thereafter to the end of treatment. Median treatment duration at the 15 February 2019 cut-off date was 1.7 months (range: 0.1-14.2) and 3.9 months (range: 0.0-17.1) in each group respectively.

,
Interventionpercentage of participants (Number)
IDH1/2 mutationFLT3 mutation
Placebo + Low Dose Cytarabine (LDAC)33.344.4
Venetoclax + Low Dose Cytarabine (LDAC)57.145.0

Percentage of Participants With Complete Remission or Complete Remission With Partial Hematologic Recovery (CR + CRh) by Mutation Subgroup

"The percentage of participants who achieved a complete remission or complete remission with partial hematologic recovery assessed by the investigator for participants with the following mutations:~Isocitrate dehydrogenase 1 and 2 (IDH1/2) mutation~FMS-like tyrosine kinase 3 (FLT3) mutation~CR is defined according to the modified IWG criteria for AML as no morphologic evidence of AML, ANC ≥ 10³/μL, platelets ≥ 10⁵/μL, RBC transfusion independence, bone marrow with < 5% blasts, absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease.~CRh is achieved when the following criteria are met:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count > 0.5 × 10³/μL and~Peripheral blood platelet count > 0.5 × 10⁵/μL and~A 1 week platelet transfusion-free period prior to hematology lab collection. Participants with no disease assessments were considered non-responders" (NCT03069352)
Timeframe: Response was assessed at the end of Cycle 1 and every 3 cycles thereafter to the end of treatment. Median treatment duration at the 15 February 2019 cut-off date was 1.7 months (range: 0.1-14.2) and 3.9 months (range: 0.0-17.1) in each group respectively.

,
Interventionpercentage of participants (Number)
IDH1/2 mutationFLT3 mutation
Placebo + Low Dose Cytarabine (LDAC)33.344.4
Venetoclax + Low Dose Cytarabine (LDAC)57.145.0

Disease-free Survival for Maintenance

DFS for maintenance comparison is defined as the time from maintenance randomization until relapse or death of any cause. The censored follow-up time for patients without relapse and death information is the date of last contact. Only patients who remained in CR or CRi after completion of consolidation therapy that were randomized to either observation or decitabine in the maintenance Step were included in this analysis. (NCT02085408)
Timeframe: Assessed every 3 months for 4 years and then every 6 months for 1 year

Interventionmonths (Median)
Maintenance : Observation8.2
Maintenance : Decitabine16.3

Overall Survival by Donor Status

Overall survival is defined as time between achieving leukemia-free state after induction therapy to death from any cause or date last known alive. The association between overall survival and donor status was evaluated regardless of assigned treatment arms. Patients with transplant donor information (either had donor or did not have a donor) reported after achieving CR/Cri/leukemia-free state post induction therapy were included in this analysis. (NCT02085408)
Timeframe: Assessed every 3 months for 4 years and then every 6 months for 1 year

Interventionmonths (Median)
With Transplant Donor27.2
Without Transplant Donor12.9

Overall Survival

Overall survival is defined as the time from randomization to death or date last known alive. (NCT02085408)
Timeframe: Assessed every 3 months for 4 years and then every 6 months for 1 year

Interventionmonths (Median)
A (Step 1:Daunorubicin/Cytarabine; Step 2:Cytarabine; Step 3: Observation/Decitabine/Transplant)12.9
B (Step 1: Clofarabine; Step 2: Clofarabine; Step 3: Observation/Decitabine/Transplant)11.6

Proportion of Patients With Complete Remission

"Patients are required to have all of the following to be considered as having a completion remission (CR).~Peripheral Blood Counts~Neutrophil count > 1.0 x 10^9 /L~Platelet count ≥ 100 x 10^9 /L~Reduced hemoglobin concentration or hematocrit has no bearing on remission status~Leukemic blasts must not be present in the peripheral blood~Bone Marrow Aspirate and Biopsy~Cellularity of bone marrow biopsy must be > 20% with maturation of all cell lines~< 5% blasts by morphologic review~Auer rods must not be detectable~Extramedullary leukemia, such as CNS or soft tissue involvement, must not be present" (NCT02085408)
Timeframe: Assessed every 3 months for 4 years and then every 6 months for 1 year

Interventionproportion of participants (Number)
A (Step 1:Daunorubicin/Cytarabine; Step 2:Cytarabine; Step 3: Observation/Decitabine/Transplant)0.446
B (Step 1: Clofarabine; Step 2: Clofarabine; Step 3: Observation/Decitabine/Transplant)0.453

Complete Response Rate - Locally Reviewed

"Assessed by the ECOG-ACRIN data manager based upon local review of images and data sent by the local sites.~Treatment response was determined by calculating the sum of the maximal cross section in 2 separate axes using enhancing lesion(s) on CT or MRI imaging. The same imaging modality was to be used throughout assessment. Complete response was defined as the disappearance of all contrast enhancing tumor size on CT or MRI, patient was off all glucocorticoids, and resolution of all meningeal and vitreous involvement if present. Response must have lasted at least 4 weeks." (NCT00335140)
Timeframe: For the primary endpoint, complete response will be based on disease status at three weeks post the end of therapy (week 17).

Interventionpercentage of participants (Number)
Rituximab + Standard Chemotherapy64

Percentage of Patients With ALL at High Risk of Relapse (Arm 2) Who Were Relapse-free at 5 Years

This measure looks at the percentage of patients on Arm 2 who did not experience a relapse at 5 years, where relapse is defined as the presence of progressive disease after the achievement of a complete remission. (NCT00176462)
Timeframe: 5 years

Interventionpercentage of participants (Number)
Arm 2 High Risk64.9

Burden of Therapy in AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Maintenance Cycle 4: Emotional

Age standardized Quality of life, measured by the emotional subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with means and standard deviation reported. (NCT01190930)
Timeframe: 1.7 years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.80
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.89

Burden of Therapy in AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Maintenance Cycle 4: Physical

Age standardized Quality of life, measured by the physical subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with means and standard deviation reported. (NCT01190930)
Timeframe: 1.7 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.62
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.64

Burden of Therapy in AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Maintenance Cycle 4: Social Functioning

Age standardized Quality of life, measured by the social functioning subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with means and standard deviation reported. (NCT01190930)
Timeframe: 1.7 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.16
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.25

Burden of Therapy in AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Emotional

Age standardized Quality of life, measured by the emotional subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with mean and standard deviation reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.72
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.77

Burden of Therapy in AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Physical

Age standardized Quality of life, measured by the physical subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with mean and standard deviation reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.64
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.67

Burden of Therapy in AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Social Functioning

Age standardized Quality of life, measured by the social functioning subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with mean and standard deviation reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.27
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.34

Burden of Therapy in AR Patients Overall at End of Consolidation Therapy: Emotional

Age standardized Quality of life, measured by the emotional subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 2 Months

InterventionZ-Score (Mean)
B-ALL Average Risk-1.21

Burden of Therapy in AR Patients Overall at End of Consolidation Therapy: Physical

Age and gender standardized Quality of life, measured by the physical subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 2 Months

InterventionZ-Score (Mean)
B-ALL Average Risk-1.44

Burden of Therapy in AR Patients Overall at End of Consolidation Therapy: Social Functioning

Age standardized Quality of life, measured by the social functioning subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 2 Months

InterventionZ-Score (Mean)
B-ALL Average Risk-0.42

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 1: Emotional

Age standardized Quality of life, measured by the emotional subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 1 year

InterventionZ-Score (Mean)
B-ALL Average Risk-0.86

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 1: Physical

Age standardized Quality of life, measured by the physical subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 1 Year

InterventionZ-Score (Mean)
B-ALL Average Risk-0.59

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 1: Social Functioning

Age standardized Quality of life, measured by the social functioning subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 1 Year

InterventionZ-Score (Mean)
B-ALL Average Risk-0.19

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 4: Emotional

Age standardized Quality of life, measured by the emotional subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 1.7 years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.84

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 4: Physical

Age standardized Quality of life, measured by the physical subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 1.7 years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.63

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 4: Social Functioning

Age standardized Quality of life, measured by the social functioning subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 1.7 Years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.20

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Emotional

Age standardized Quality of life, measured by the emotional subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 2.5 years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.74

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Physical

Age standardized Quality of life, measured by the physical subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.66

Burden of Therapy in AR Patients Overall at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Social Functioning

Age standardized Quality of life, measured by the social functioning subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.30

Burden of Therapy in Boy AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Therapy: Emotional

Age standardized Quality of life, measured by the emotional subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with mean and standard deviation reported. (NCT01190930)
Timeframe: 3.2 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.71
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.51

Burden of Therapy in Boy AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Therapy: Physical

Age standardized Quality of life, measured by the physical subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with mean and standard deviation reported. (NCT01190930)
Timeframe: 3.2 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.85
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.47

Burden of Therapy in Boy AR Patients by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Therapy: Social Functioning

Age standardized Quality of life, measured by the social functioning subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated for each group with mean and standard deviation reported. (NCT01190930)
Timeframe: 3.2 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-0.46
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.33

Burden of Therapy in Boy AR Patients Overall at End of Therapy: Emotional

Age standardized Quality of life, measured by the emotional subscale of Pediatric Quality of Life Inventory 4.0 Genetic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 3.2 years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.62

Burden of Therapy in Boy AR Patients Overall at End of Therapy: Physical

Age standardized Quality of life, measured by the physical subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 3.2 years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.67

Burden of Therapy in Boy AR Patients Overall at End of Therapy: Social Functioning

Age standardized Quality of life, measured by the Social functioning subscale of Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL), will be calculated with mean and standard deviation reported. (NCT01190930)
Timeframe: 3.2 Years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.40

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL 12 Months Post Therapy: Left

Strength in the left ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for the cohort will be reported. (NCT01190930)
Timeframe: 4.2 Years

InterventionZ-Score (Mean)
B-ALL Average Risk-1.19

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL 12 Months Post Therapy: Right

Strength in the right ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for the cohort will be reported. (NCT01190930)
Timeframe: 4.2 Years

InterventionZ-Score (Mean)
B-ALL Average Risk-1.12

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL at End of Consolidation Therapy-Left

Strength in the left ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for the cohort will be reported. (NCT01190930)
Timeframe: 2 Months

InterventionZ-Score (Mean)
B-ALL Average Risk-0.84

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL at End of Consolidation Therapy-Right

Strength in the right ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for the cohort will be reported. (NCT01190930)
Timeframe: 2 Months

InterventionZ-Score (Mean)
B-ALL Average Risk-0.87

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL at End of Maintenance Cycle 1: Left

Strength in the left ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for the cohort will be reported. (NCT01190930)
Timeframe: 1 Year

InterventionZ-Score (Mean)
B-ALL Average Risk-0.36

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL at End of Maintenance Cycle 1: Right

Strength in the right ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for the cohort will be reported. (NCT01190930)
Timeframe: 1 Year

InterventionZ-Score (Mean)
B-ALL Average Risk-0.39

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Left

Strength in the left ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for the cohort will be reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.28

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Right

Strength in the right ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for the cohort will be reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk-0.27

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Left

Strength in the left ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for each randomization group will be reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-1.19
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks0.21

Characterize Vincristine-associated Neuropathy in Children Undergoing Therapy for Average Risk (AR) ALL by Vincristine Pulse Frequency Randomization Groups (4 Week vs. 12 Week) at End of Maintenance Cycle 7 (Boys)/End of Therapy (Girls): Right

Strength in the right ankle dorsiflexors averaged over two measurements. Age and gender standardized mean and standard deviation for each randomization group will be reported. (NCT01190930)
Timeframe: 2.4 Years

InterventionZ-Score (Mean)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks-1.12
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks-0.02

DFS for SR Down Syndrome Patients With Standardized Treatment and Enhanced Supportive Care

DFS is calculated as the time from end of Induction to first event (relapse, second malignancy, remission death) or date of last contact. The 5-year DFS and 95% confidence interval for these patients will be estimated. (NCT01190930)
Timeframe: 5.1 years

Interventionpercent probability (Number)
Standard Risk With Down Syndrome89.77

DFS in Average Risk (AR) Patients Based on the Pulse Frequency Randomization

DFS is calculated as the time from randomization at the end of interim maintenance II to first event (relapse, second malignancy, remission death) or date of last contact. Five year DFS estimates will be calculated from the point of randomization for both groups. Two-sided 95% confidence intervals will be calculated. (NCT01190930)
Timeframe: 5.7 years

Interventionpercent probability (Number)
B-ALL Average Risk: VCR/DEX Pulse Every 4 Weeks94.10
B-ALL Average Risk: VCR/DEX Pulse Every 12 Weeks95.13

DFS in Low Risk (LR) Patients Based on Randomization to 1 of 2 Low-intensity Regimens

DFS is calculated as the time from randomization at the end of Induction to first event (relapse, second malignancy, remission death) or date of last contact. Five year DFS estimates will be calculated from the point of randomization for both groups. Two-sided 95% confidence intervals will be calculated. (NCT01190930)
Timeframe: 5.1 years

Interventionpercent probability (Number)
B-ALL Low Risk Arm I (LR-M)98.75
B-ALL Low Risk Arm II (LR-C)98.50

Disease Free Survival (DFS) in Average Risk (AR) Patients Based on the Methotrexate Dose Randomization

DFS is calculated as the time from randomization at the end of interim maintenance II to first event (relapse, second malignancy, remission death) or date of last contact. Five year DFS estimates will be calculated from the point of randomization for both groups. Two-sided 95% confidence intervals will be calculated. (NCT01190930)
Timeframe: 5.7 years

Interventionpercent probability (Number)
B-ALL Average Risk: MTX 20 mg/m^2/Week Starting Dose95.05
B-ALL Average Risk: MTX 40 mg/m^2/Week Starting Dose94.17

Event Free Survival (EFS) for B-LLy Patients

EFS is calculated as the Time from study enrollment to first event (induction failure, relapse, second malignancy, remission death) or date of last contact. The 5-year EFS and 95% confidence interval for these patients will be estimated. (NCT01190930)
Timeframe: 5 years

Interventionpercent probability (Number)
B-LLy94.54

Overall Survival (OS) for B-LLy Patients

OS is calculated as the time from study enrollment to death or date of last contact. The 5-year OS and 95% confidence interval for these patients will be estimated. (NCT01190930)
Timeframe: 5 years

Interventionpercent probability (Number)
B-LLy93.97

Sample Collection of Central Path Review Slides in B-LLy Patients

Percent of B-LLy patients who had adequate/usable samples of samples collected will be reported. (NCT01190930)
Timeframe: Up to 1 month

Interventionpercentage of patients (Number)
B-LLy89.7

Blast Response

Blast response was defined as a greater than 50% decrease in bone marrow blast count and 2 log reduction in peripheral blood blast count, sustained for at least 28 days. (NCT01056523)
Timeframe: 2-3 years

InterventionParticipants (Count of Participants)
Ribavirin and Cytarabine2

Complete Response Rate

Defined as <5% blasts in the bone marrow and a hgb 100 g/L, platelets 100,000/uL, neutrophils 1000/uL. (NCT01056523)
Timeframe: 2-3 years

InterventionParticipants (Count of Participants)
Ribavirin and Cytarabine2

Overall Response Rate

Overall response rate comprises complete response (<5% blasts in the bone marrow, and in the peripheral blood Hgb more than or equal to 100 g/L, platelets more than or equal to 100x10-9/L, and neutrophils more than or equal to 1x10-9/L), partial response (5 to 25% blasts in the bone marrow and same peripheral blood parameters) and blast response (a greater than 50% decrease in bone marrow blast count and 2 log reduction in peripheral blood blast count, sustained for at least 28 days). (NCT01056523)
Timeframe: 2-3 years

InterventionParticipants (Count of Participants)
Ribavirin and Cytarabine5

Partial Response

Partial response was defined as 5 to 25% blasts in the bone marrow and Hgb >100g/L, platelets >100,000/ul and neutrophils >1000/ul. (NCT01056523)
Timeframe: 2-3 years

InterventionParticipants (Count of Participants)
Ribavirin and Cytarabine1

Recommended Phase II Dose (RP2D) of Ribavirin When Given in Combination With Low-dose Ara-C

This 3+3 designed aimed to determine recommended phase II dose (RP2D) based on pharmacokinetics (PK) and maximum tolerated dose (MTD). For the dose to be selected, a target steady state level of ribavirin 20 uM was needed for all patients and no more than 1 of 6 patients could have had dose limiting toxicity at that dose. (NCT01056523)
Timeframe: 56 days

Interventionmg (Number)
Ribavirin po bid for 28 dayscytarabine arabinoside sc bid for 10 days
Ribavirin-Cytarabine140010

AALL08P1 Feasibility Outcome

Percentage of Group B (High Risk-High) patients that tolerate at least 8 of the 12-14 total doses of pegaspargase during Consolidation, Interim Maintenance, and Delayed Intensification periods. Only Grp B analyzed since this is prespecified in protocol. (NCT00866307)
Timeframe: Consolidation through Delayed Intensification

Interventionpercentage of participants (Number)
Group B (High Risk-High)53.3

AALL08P1 Safety Outcome

Percentage of Group B (High Risk-High) patients taking less than 49 weeks from day 1 of consolidation to day 1 of maintenance therapy. Only Group B analyzed since this is prespecified in protocol. (NCT00866307)
Timeframe: Consolidation through Delayed Intensification

Interventionpercentage of participants (Number)
Group B (High Risk-High)50.0

Participant Progression Free Survival at 2 Years

Progression-free survival defined as the number of participants without evidence of progression or death after 2 years from stem cell transplant. (NCT00505921)
Timeframe: 2 years

Interventionparticipants (Number)
Campath-1H15

Median Time to Disease Progression

median days from transplant to relapse/progression (NCT00992446)
Timeframe: time post ASCT to progression

Interventionyears (Median)
Treatment (Chemotherapy, ASCT, Bortezomib, Vorinostat))1.05

Toxicity of Vorinostat Bortezomib Maintenance Therapy After Autologous Transplant

Number of patients on maintenance therapy post-transplant who experienced grade 3 or higher toxicity per NCI-Common Terminology Criteria for Adverse Events, version 3. The first three months of bortezomib and vorinostat therapy will be used as the time period to evaluate toxicity for stopping rules of the study. Toxicity that meets stopping rules will be determined based on the number of patients that are withdrawn from study for significant toxicity (grade IV, non-hematological, non-metabolic, non-peripheral neuropathy). (NCT00992446)
Timeframe: 3 months after start of maintenance therapy

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, ASCT, Bortezomib, Vorinostat))19

Event-free Survival

Number of patients alive without disease progression/relapse (NCT00992446)
Timeframe: 6.64 Years Post-Transplant

InterventionParticipants (Count of Participants)
Alive without disease porgressionalive with disease progression
Treatment (Chemotherapy, ASCT, Bortezomib, Vorinostat))145

Overall Survival

Number of patients alive who received maintenance therapy (NCT00992446)
Timeframe: 6.64 Years Post-Transplant

Interventionparticipants (Number)
AliveDead
Treatment (Chemotherapy, ASCT, Bortezomib, Vorinostat))163

Characterize the Effects of Plerixafor Plus G-CSF on Fold Change in CXCR4 Clone 12G5 Relative Mean Fluorescent Intensity

(NCT00906945)
Timeframe: 6 hours after plerixafor

Interventionfold change in CXCR4 clone 1D9 (Mean)
Phase I and Phase II Participants0.9

Characterize the Effects of Plerixafor Plus G-CSF on Fold Change in CXCR4 Clone 1D9 Relative Mean Fluorescent Intensity

(NCT00906945)
Timeframe: 6 hours after plerixafor

Interventionfold change in CXCR4 clone 1D9 (Mean)
Phase I and Phase II Participants8.0

Characterize the Mobilization of Leukemic Cells With Plerixafor Plus G-CSF as Measured by Fold Change in AML Blast Count

(NCT00906945)
Timeframe: 6 hours after plerixafor

Interventionfold change in AML blast count (Mean)
Phase I and Phase II Participants9.4

Characterize the Mobilization of Leukemic Cells With Plerixafor Plus G-CSF as Measured by Fold Change in White Blood Cells

(NCT00906945)
Timeframe: 6 hours after plerixafor

Interventionfold change in white blood cells (Mean)
Phase I and Phase II Participants4.6

Overall Survival

Overall survival: Defined as the date of first dose of study drug to the date of death from any cause. (NCT00906945)
Timeframe: Median follow-up was 34.6 months

Interventiondays (Median)
Phase I and Phase II Participants227

Phase I: Maximum Tolerated Dose of Plerixafor Plus G-CSF When Combined With MEC

(NCT00906945)
Timeframe: Completion of Phase I enrollment (17 months)

Interventionmcg/kg/day (Number)
Phase I (Includes Levels 1-5)750

Phase II: Complete Response Rate (CR+CRi)

"Morphologic complete remission (CR): Defined as morphologic leukemia-free state, including <5% blasts in BM aspirate with marrow spicules and a count of > 200 nucleated cells and no blasts with Auer rods, no persistent extramedullary disease, ANC > 1,000/mm3, platelet count > 100,000/mm3.~Morphologic complete remission with incomplete blood count recovery (CRi): Defined as CR with the exception of neutropenia <1,000/mm3 or thrombocytopenia <100,000/mm3." (NCT00906945)
Timeframe: 45 days

Interventionpercentage of participants (Number)
Phase II (MTD)30

Relapse Free-survival Rate

(NCT00906945)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Phase I and Phase II Participants75

Time to Hematologic Recovery as Measured by Time to Neutrophil Recovery

-Neutrophil recovery is defined as absolute neutrophil count (ANC) >= 500/mm^3 (NCT00906945)
Timeframe: Up to 62 days after treatment

Interventiondays (Median)
Phase I and Phase II Participants38

Time to Hematologic Recovery as Measured by Time to Neutrophil Recovery

-Neutrophil recovery is defined as absolute neutrophil count >= 1000/mm^3 (NCT00906945)
Timeframe: Up to 62 days after treatment

Interventiondays (Median)
Phase I and Phase II Participants40

Time to Hematologic Recovery as Measured by Time to Platelet Recovery

-Platelet recovery is defined as platelets >= 100,000/mm3 (NCT00906945)
Timeframe: Up to 62 days after treatment

Interventiondays (Median)
Phase I and Phase II Participants32

Time to Hematologic Recovery as Measured by Time to Platelet Recovery

-Platelet recovery is defined as platelets >= 50,000/mm^3 (NCT00906945)
Timeframe: Up to 62 days after treatment

Interventiondays (Median)
Phase I and Phase II Participants32

Phase I and Phase II: Safety and Tolerability of Regimen as Measured by Grade and Frequency of Adverse Events Exceeding 10% in Total Frequency

(NCT00906945)
Timeframe: 30 days following end of treatment

,,,,
Interventionnumber of events (Number)
AnemiaFebrile neutropeniaAbdominal painConstipationDiarrheaGastroesophageal reflux diseaseMucositis oralNauseaVomitingChillsEdema-limbsFatigueFeverNon-cardiac chest painPainBacteremiaLung infectionSepsisActivated partial thromboplastin time prolongedAlanine aminotransferase increasedAlkaline phosphatase increasedAspartate aminotransferase increasedBlood bilirubin increasedINR increasedNeutrophil count decreasedPlatelet count decreasedWhite blood cell count decreasedAnorexiaHypoalbuminemiaHypocalcemiaHypokalemiaHypomagnesemiaHyponatremiaBone painDizzinessHeadacheParesthesiaInsomniaProteinuriaCoughDyspneaPneumonitisRash maculo-papularHypotension
Grade 10065712211255961200053342700043257444745333134
Grade 2400265330012422010001030000261120021612123334
Grade 3920100010100101045001001000000020120100000002
Grade 40000000000000000040000007151600020000000000000
Grade 500000000000000002400000000000000000000000100

Number of Participants Who Experienced Dose Limiting Toxicities (DLT)

Dose-limiting toxicities were defined as events related to onvansertib that were considered an adverse reaction or suspected adverse reaction during the first cycle of therapy and that fulfilled one of the following: Hematologic (persistent pancytopenia resistant to current standards of care that continues for ≥42 days and is not related to leukemic infiltration or another cause unrelated to study therapy) or Non-Hematologic (any Grade 3 abnormalities that persist >7 days without decreasing in severity despite standards of care, are clinically significant, or that are Grade 4 and symptomatic). (NCT03303339)
Timeframe: Up to Day 28 of Cycle 1

InterventionParticipants (Count of Participants)
Phase 1b: Onvansertib 12 mg/m^2 + Low-dose Cytarabine0
Phase 1b: Onvansertib 18 mg/m^2 + Low-dose Cytarabine0
Phase 1b: Onvansertib 27 mg/m^2 + Low-dose Cytarabine0
Phase 1b: Onvansertib 40 mg/m^2 + Low-dose Cytarabine0
Phase 1b: Onvansertib 60 mg/m^2 + Low-dose Cytarabine0
Phase 1b: Onvansertib 12 mg/m^2 + Decitabine0
Phase 1b: Onvansertib 18 mg/m^2 + Decitabine0
Phase 1b: Onvansertib 27 mg/m^2 + Decitabine0
Phase 1b: Onvansertib 40 mg/m^2 + Decitabine0
Phase 1b: Onvansertib 60 mg/m^2 + Decitabine0
Phase 1b: Onvansertib 90 mg/m^2 + Decitabine2

Number of Participants With Change From Baseline in Eastern Co-operative Oncology Group (ECOG) Performance Status

ECOG performance status was determined using 6-point scale from 0-5, with 0 meaning a participant was fully active/able to carry on all pre-disease activities without restriction and 5 meaning the participant was deceased. (NCT03303339)
Timeframe: Baseline and end of study (approximately up to up to 27 months)

InterventionParticipants (Count of Participants)
Phase 1b: Onvansertib + Low-dose Cytarabine11
Phase 1b: Onvansertib + Decitabine14
Phase 2: Onvansertib + Decitabine22

Phase 2: Duration of Response (DOR)

Duration of Response (DOR) is the time (in months) from the first response of CR, CRi or PR until recurrence of or progression of disease (or death). MLF State is also included as a response when calculating DOR. Responding subjects without death or progression will be censored at the date of their last evaluable disease assessment. (NCT03303339)
Timeframe: Up to 27 months

Interventionmonths (Median)
Phase 2: Onvansertib + Decitabine5.2

Phase 2: Event-free Survival (EFS)

EFS is defined as the time from enrollment until disease progression or death from any cause and reported as the proportion of participants event free at 12 months. (NCT03303339)
Timeframe: 12 Months

InterventionProportion of Participants (Number)
Phase 2: Onvansertib + Decitabine0.1

Phase 2: Number of Participants Who Achieved a Complete Response (CR)

"Complete Response also includes Complete Response with Incomplete Blood Count Recovery (CRi). Complete response is defined by the following criteria:~Morphologic leukemia-free state plus:~Subject is independent of transfusions~Absolute neutrophil count of >1000/mm3~Platelets of ≥100,000/mm3~Complete response with incomplete blood count recovery meets all criteria for CR except for either neutropenia (ANC <1000/mm3) or thrombocytopenia (<100,000/mm3) but must include transfusion independence." (NCT03303339)
Timeframe: Up to 27 months

InterventionParticipants (Count of Participants)
Phase 2: Onvansertib + Decitabine3

Phase 2: Number of Participants Who Achieved a Morphologic Leukemia-free (MLF) State

Defined as bone marrow (BM) <5% blasts in an aspirate with spicules and no blasts with Auer rods or persistence of extramedullary disease. (NCT03303339)
Timeframe: Up to 27 months

InterventionParticipants (Count of Participants)
Phase 2: Onvansertib + Decitabine1

Phase 2: Number of Participants With Partial Response (PR)

PR criteria includes all of the hematologic values for a CR but with a decrease of at least 50% in the percentage of blasts to 5% to 25% in the bone marrow aspirate and a normalization of blood counts. (NCT03303339)
Timeframe: Up to 27 months

InterventionParticipants (Count of Participants)
Phase 2: Onvansertib + Decitabine0

Phase 2: Overall Survival (OS)

OS is defined as the time from enrollment until death from any cause and reported as the proportion of participants alive at 12 months. (NCT03303339)
Timeframe: 12 Months

InterventionProportion of Participants (Number)
Phase 2: Onvansertib + Decitabine0.2

Number of Participants With Adverse Events (AEs)

Any clinically significant change in electrocardiogram (ECG), physical examination findings, body weight, vital signs, and laboratory parameters were recorded as Adverse Events. (NCT03303339)
Timeframe: Baseline up to 30 days after last dose of study drug (up to 27 months)

,,,,,,,,,,,
InterventionParticipants (Count of Participants)
Any Adverse EventsAny Serious Adverse EventsAny Treatment-Related Serious Adverse Events
Phase 1b: Onvansertib 12 mg/m^2 + Decitabine420
Phase 1b: Onvansertib 12 mg/m^2 + Low-dose Cytarabine320
Phase 1b: Onvansertib 18 mg/m^2 + Decitabine330
Phase 1b: Onvansertib 18 mg/m^2 + Low-dose Cytarabine331
Phase 1b: Onvansertib 27 mg/m^2 + Decitabine331
Phase 1b: Onvansertib 27 mg/m^2 + Low-dose Cytarabine310
Phase 1b: Onvansertib 40 mg/m^2 + Decitabine440
Phase 1b: Onvansertib 40 mg/m^2 + Low-dose Cytarabine321
Phase 1b: Onvansertib 60 mg/m^2 + Decitabine321
Phase 1b: Onvansertib 60 mg/m^2 + Low-dose Cytarabine530
Phase 1b: Onvansertib 90 mg/m^2 + Decitabine653
Phase 2: Onvansertib 60 mg/m^2 + Decitabine32275

Pharmacokinetic Parameter: Area Under the Curve Over the First 24 Hours AUC(0-24) for Onvansertib

(NCT03303339)
Timeframe: Cycle 1: Days 1 and 5

,,,,,,,,,,,
Interventionh*ng/mL (Geometric Least Squares Mean)
Cycle 1 Day 1Cycle 1 Day 5
Phase 1b: Onvansertib 12 mg/m^2 + Decitabine1099.431868.69
Phase 1b: Onvansertib 12 mg/m^2 + Low-dose Cytarabine1197.931868.69
Phase 1b: Onvansertib 18 mg/m^2 + Decitabine2789.813626.44
Phase 1b: Onvansertib 18 mg/m^2 + Low-dose Cytarabine772.683626.44
Phase 1b: Onvansertib 27 mg/m^2 + Decitabine3257.575343.01
Phase 1b: Onvansertib 27 mg/m^2 + Low-dose Cytarabine2534.225343.01
Phase 1b: Onvansertib 40 mg/m^2 + Decitabine4084.187415.04
Phase 1b: Onvansertib 40 mg/m^2 + Low-dose Cytarabine2761.117415.04
Phase 1b: Onvansertib 60 mg/m^2 + Decitabine9599.1919470.17
Phase 1b: Onvansertib 60 mg/m^2 + Low-dose Cytarabine5386.6919470.17
Phase 1b: Onvansertib 90 mg/m^2 + Decitabine9087.7712981.17
Phase 2: Onvansertib 60 mg/m^2 + Decitabine6280.2811767.29

Pharmacokinetic Parameter: Maximum Observed Plasma Concentration (Cmax) for Onvansertib

(NCT03303339)
Timeframe: Cycle 1: Days 1 and 5

,,,,,,,,,,,
Interventionng/mL (Geometric Mean)
Cycle 1 Day 1Cycle 1 Day 5
Phase 1b: Onvansertib 12 mg/m^2 + Decitabine80.15146.10
Phase 1b: Onvansertib 12 mg/m^2 + Low-dose Cytarabine92.57139.22
Phase 1b: Onvansertib 18 mg/m^2 + Decitabine129.18197.67
Phase 1b: Onvansertib 18 mg/m^2 + Low-dose Cytarabine69.56103.27
Phase 1b: Onvansertib 27 mg/m^2 + Decitabine309.86398.38
Phase 1b: Onvansertib 27 mg/m^2 + Low-dose Cytarabine251.16298.63
Phase 1b: Onvansertib 40 mg/m^2 + Decitabine410.42496.24
Phase 1b: Onvansertib 40 mg/m^2 + Low-dose Cytarabine256.11339.52
Phase 1b: Onvansertib 60 mg/m^2 + Decitabine724.76955.59
Phase 1b: Onvansertib 60 mg/m^2 + Low-dose Cytarabine626.18857.29
Phase 1b: Onvansertib 90 mg/m^2 + Decitabine737.211137.16
Phase 2: Onvansertib 60 mg/m^2 + Decitabine515.63861.86

Pharmacokinetic Parameter: Plasma Terminal Elimination Half-life (t1/2) for Onvansertib

(NCT03303339)
Timeframe: Cycle 1: Days 1 and 5

,,,,,,,,,,,
InterventionHours (Geometric Least Squares Mean)
Cycle 1 Day 1Cycle 1 Day 5
Phase 1b: Onvansertib 12 mg/m^2 + Decitabine16.96013.282
Phase 1b: Onvansertib 12 mg/m^2 + Low-dose Cytarabine11.29416.413
Phase 1b: Onvansertib 18 mg/m^2 + Decitabine13.7959.082
Phase 1b: Onvansertib 18 mg/m^2 + Low-dose Cytarabine7.99312.561
Phase 1b: Onvansertib 27 mg/m^2 + Decitabine9.39410.713
Phase 1b: Onvansertib 27 mg/m^2 + Low-dose Cytarabine12.19712.246
Phase 1b: Onvansertib 40 mg/m^2 + Decitabine11.38916.337
Phase 1b: Onvansertib 40 mg/m^2 + Low-dose Cytarabine11.7477.321
Phase 1b: Onvansertib 60 mg/m^2 + Decitabine12.91744.018
Phase 1b: Onvansertib 60 mg/m^2 + Low-dose Cytarabine7.30914.484
Phase 1b: Onvansertib 90 mg/m^2 + Decitabine12.63312.378
Phase 2: Onvansertib 60 mg/m^2 + Decitabine11.90414.060

Pharmacokinetic Parameter: Time to Reach the Maximum Observed Plasma Concentration (Tmax) for Onvansertib

(NCT03303339)
Timeframe: Cycle 1: Days 1 and 5

,,,,,,,,,,,
InterventionHours (Geometric Least Squares Mean)
Cycle 1 Day 1Cycle 1 Day 5
Phase 1b: Onvansertib 12 mg/m^2 + Decitabine3.1022.438
Phase 1b: Onvansertib 12 mg/m^2 + Low-dose Cytarabine1.9611.844
Phase 1b: Onvansertib 18 mg/m^2 + Decitabine2.3651.583
Phase 1b: Onvansertib 18 mg/m^2 + Low-dose Cytarabine2.5482.892
Phase 1b: Onvansertib 27 mg/m^2 + Decitabine1.2883.145
Phase 1b: Onvansertib 27 mg/m^2 + Low-dose Cytarabine2.3012.267
Phase 1b: Onvansertib 40 mg/m^2 + Decitabine1.3733.161
Phase 1b: Onvansertib 40 mg/m^2 + Low-dose Cytarabine1.3841.474
Phase 1b: Onvansertib 60 mg/m^2 + Decitabine2.8933.161
Phase 1b: Onvansertib 60 mg/m^2 + Low-dose Cytarabine2.0712.148
Phase 1b: Onvansertib 90 mg/m^2 + Decitabine2.4353.189
Phase 2: Onvansertib 60 mg/m^2 + Decitabine2.7252.665

Median of Serum Complement CD20 Levels

Median serum C20 MFI (mean fluorescence intensity) (NCT01527149)
Timeframe: Baseline

Interventionmean fluorescence intensity (Median)
Treatment (Monoclonal Antibody and Combination Chemotherapy)186.8

Median Overall Survival (OS)

Estimated distributions obtained using the Kaplan-Meier method. Estimates of quantities such as median survival will be obtained. Corresponding confidence intervals using the methodology of Brookmeyer and Crowley will be computed. (NCT01527149)
Timeframe: From baseline through study completion, an average of 5 years

Interventionmonths (Median)
Treatment (Monoclonal Antibody and Combination Chemotherapy)56.0

Median Progression-free Survival (PFS)

Estimated distributions obtained using the Kaplan-Meier method. Estimates of quantities such as median survival will be obtained. Corresponding confidence intervals using the methodology of Brookmeyer and Crowley will be computed. (NCT01527149)
Timeframe: From baseline through study completion, an average of 5 years

Interventionmonths (Median)
Treatment (Monoclonal Antibody and Combination Chemotherapy)45.5

Number of Participants With at Least One Serious Adverse Event

Evaluated using National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. (NCT01527149)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
Treatment (Monoclonal Antibody and Combination Chemotherapy)19

Percentage of Participants With Autologous Stem Cell Transplantation

Estimated using simple relative frequencies. The corresponding 95% confidence intervals will be computed using the method proposed in Clopper and Pearson. (NCT01527149)
Timeframe: Up to 6 weeks after the last dose of ofatumumab-chemotherapy, an average of 4 month

Interventionpercentage of participants (Number)
Treatment (Monoclonal Antibody and Combination Chemotherapy)73

Proportion of Patients Experiencing a Complete Response

"Evaluated according to the International Working Group Response criteria as reported by Cheson et al. and the revised Cheson criteria.~Complete response is defined as complete disappearance of all detectable clinical evidence of disease and disease-related symptoms if present before therapy. Typically FDG-avid lymphoma: in patients with PET scan positive before therapy, a posttreatment residual mass of any size is permitted as long as it is PET negative. Variably FDG-avid lymphomas/FDG avidity unknown: in patients with a negative pretreatment PET scan, all lymph nodes and nodal masses must have regressed on CT to normal size (1.5 cm in their greatest transverse diameter for nodes 1.5 cm before therapy). Previously involved nodes that were 1.1 cm to 1.5 cm in their long axis and more than 1)" (NCT01527149)
Timeframe: 22 weeks

InterventionProportion of participants (Number)
Treatment (Monoclonal Antibody and Combination Chemotherapy).62

Proportion of Patients Who Experience Complete Remission as Assessed by HSFCM

Established when all CR criteria are met and negative flow cytometry examination of peripheral blood and bone marrow biopsy/aspiration collected at baseline, before courses 3 and 5, within 3 weeks after course 6, on day 100 (if HDC-ASCT eligible), and then every 6 months for 3 years. (NCT01527149)
Timeframe: Up to 3 years

InterventionProportion of participants (Number)
Treatment (Monoclonal Antibody and Combination Chemotherapy).84

Time-to-tumor Progression (TTP) at 3 Years

Estimated percentage of patients that progressed at 3 years. Time to Progression distributions obtained using the Kaplan-Meier method. Corresponding confidence intervals using the methodology of Brookmeyer and Crowley will be computed. (NCT01527149)
Timeframe: From baseline until objective tumor progression, as assessed up to 3 years

Interventionpercentage of participants (Number)
Treatment (Monoclonal Antibody and Combination Chemotherapy)76

Change From Baseline in Percentage of Cells Positive for Ki67

Mean change from baseline in Percentage of Cells Positive for Ki67 by patient response. (NCT01527149)
Timeframe: Baseline and up to 3 years

Interventionpercentage of cells positive for Ki-67 (Mean)
Not CRCR
Treatment (Monoclonal Antibody and Combination Chemotherapy)-2.5NA

Number of Biomarker-positive Participants With Clinical Responses

"Biomarkers: FLT3-ITD positive, NPM1 positive, CEBPA. A good CR is defined as <5% blasts in the marrow by morphologic evaluation along with the absence of any MRD by flow cytometry or cytogenetics and recovery of blood counts (platelets >100,00 and absolute neutrophil count >1,000) by day 35 after induction. Cheson AML Response Criteria is used for Morphologic Leukemia Free State, Morphologic Complete Remission, Cytogenetic Complete Remission (CRc), Molecular Complete Remission (CRm), Morphologic Complete Remission with Incomplete Blood Count Recovery (CRi), Partial Remission (PR), Treatment Failure, Recurrence (Progressive Disease)." (NCT01831232)
Timeframe: 38 days after dosing

Interventionparticipants with clinical responses (Number)
Treatment (Pravastatin Sodium, Idarubicin, and Cytarabine)0

Number of Participants With Good Complete Remission (CR)

"A Bayesian design intended to simultaneously monitor efficacy and toxicity will be used.~Definition of Good CR: Conventional criteria for CR (absolute neutrophil count > 1,000/uL, platelet count > 100,000/uL, marrow with <5% morphologic blasts) and additionally the requirements that marrow Minimal Residual Disease (MRD) - detected by 10-color flow cytometry or conventional cytogenetic evaluation - be absent and that the above blood counts be obtained." (NCT01831232)
Timeframe: 35 days

InterventionParticipants (Count of Participants)
Treatment (Pravastatin Sodium, Idarubicin, and Cytarabine)12

Number of Participants With TRM.

"A Bayesian design intended to simultaneously monitor efficacy and toxicity will be used.~TRM: Treatment Related Mortality" (NCT01831232)
Timeframe: 28 days

Interventionparticipants (Number)
Treatment (Pravastatin Sodium, Idarubicin, and Cytarabine)2

Overall Survival

Amount of time a patient lives after treatment with IAP (NCT01831232)
Timeframe: 1 year after treatment with IAP

Interventionpercentage of patients surviving (Number)
Treatment (Pravastatin Sodium, Idarubicin, and Cytarabine)53.5

Progression Free Survival (PFS)

(NCT01831232)
Timeframe: 1 year after treatment with IAP

Interventionpercentage of patients with PFS (Number)
Treatment (Pravastatin Sodium, Idarubicin, and Cytarabine)52.6

Rate of Complete Remission (CR), Remission With Incomplete Blood Count Recovery (CRi) and Partial Remission (PR)

Complete remission (CR) - includes patients with good CR and CR with minimal residual disease (MRD); remission with incomplete blood count recovery (CRi), partial remission (PR) (NCT01831232)
Timeframe: 35 days

InterventionParticipants (Count of Participants)
CRCRiPR
Treatment (Pravastatin Sodium, Idarubicin, and Cytarabine)1520

Disease-free Survival in Patients Undergoing Autologous Stem Cell Transplant

(NCT00774046)
Timeframe: Up to 883 days

InterventionDays (Median)
All Patients367

Feasibility of Stem Cell Collection

Feasibility is the ability to cryopreserve >=2.0 x 10^6 CD34+ cells/kg (NCT00774046)
Timeframe: 1-5 days from initiation of stem cell collection

Interventionpercentage of participants (Number)
All Patients70

Numbers of Stem Cells Collected

(NCT00774046)
Timeframe: 1-5 days from initiation of stem cell collection

Intervention10^6 CD34+ cells/kg (Median)
All Patients4.50

Overall Survival in Patients Undergoing Autologous Stem Cell Transplant

(NCT00774046)
Timeframe: Up to 817 days

InterventionDays (Median)
All Patients294

Overall Survival

(NCT00774046)
Timeframe: Up to 2000 days

InterventionDays (Median)
All Patients399

Relapse-free Survival

Relapse is defined as bone marrow blasts >5% if the patient had achieved a complete remission, or the recurrence of any clonal cytogenetic abnormality. (NCT00774046)
Timeframe: Up to 2000 days

InterventionDays (Median)
All Patients415

Response to Induction Chemotherapy (CR or PR)

Complete remission (CR): <5% bone marrow blasts with recovery of peripheral blood counts; complete cytogenetic remission, the disappearance of any pre-existing cytogenetic abnormality Partial remission (PR): >5% bone marrow blasts, but less than the pre-treatment blast percentage within the bone marrow Resistant disease (RD): no significant cytoreduction in bone marrow leukemic cells from pre-treatment levels Not evaluable (NE): patients who died during induction chemotherapy or who withdrew from follow-up before assessment could be made (NCT00774046)
Timeframe: Day 28-40

Interventionpercentage of participants (Number)
All Patients81.2

Number of Participants Who Experienced a Dose Limiting Toxicity (DLT)

To evaluate the side effects of giving Azacytidine before and during chemotherapy using the standard drugs Fludarabine, Cytarabine, IT Cytarabine (AML patients) and IT methotrexate (ALL patients) (NCT01861002)
Timeframe: From Day 1 to Day 42 (Cycle 1)

,
InterventionParticipants (Count of Participants)
# of patients with DLT# of patients without DLT# of patients not evaluable
Dose 75 mg/m2/Day Azacytidine Diagnosed With ALL020
Dose 75 mg/m2/Day Azacytidine Diagnosed With AML0121

5-Year Disease-Free Survival by Bone Marrow Day 18 Status

Disease-free survival (DFS) in a landmark analysis is defined as the duration of time from asparaginase randomization (which occurred after patients achieved complete remission and were assigned to a final risk group) to documented relapse, death during remission or second malignant neoplasm. DFS is estimated based on the Kaplan-Meier method and 5-year DFS is the probability of patients remaining alive, relapse-free and without occurrence of second malignant neoplasm 5 years from asparaginase randomization. Disease relapse is defined as >25% lymphoblasts identified morphologically in bone marrow aspirate/biopsy, or identification of lymphoblasts in marrow (any percentage) identified to be leukemic by flow cytometry, cytogenetics, FISH, immunohistochemistry, or other tests. Appearance of leukemic cells at any extramedullary site (a single, unequivocal lymphoblast in the CSF may qualify as CNS leukemia) also qualifies if confirmed by the PI. (NCT00400946)
Timeframe: Disease evaluations occurred continuously on treatment. Suggested long-term follow-up was monthly for 6m, bi-monthly for 6m, every 4 months for 1y, semi-annually for 1y, then annually. Median follow-up in this study cohort is 6 yrs, up to 10y.

Interventionprobability (Number)
M1 Day 18 Bone Marrow Status.89
M2/M3 Day 18 Bone Marrow Status.78
Hypocellular Day 18 Bone Marrow Status.88

5-year Disease-Free Survival by CNS Directed Treatment Group

Disease-free survival (DFS) in a landmark analysis is defined as the duration of time from asparaginase randomization (which occurred after patients achieved complete remission and were assigned to a final risk group) to documented relapse, death during remission or second malignant neoplasm. DFS is estimated based on the Kaplan-Meier method and 5-year DFS is the probability of patients remaining alive, relapse-free and without occurrence of second malignant neoplasm 5 years from asparaginase randomization. Disease relapse is defined as >25% lymphoblasts identified morphologically in bone marrow aspirate/biopsy, or identification of lymphoblasts in marrow (any percentage) identified to be leukemic by flow cytometry, cytogenetics, FISH, immunohistochemistry, or other tests. Appearance of leukemic cells at any extramedullary site (a single, unequivocal lymphoblast in the CSF may qualify as CNS leukemia) also qualifies if confirmed by the PI. (NCT00400946)
Timeframe: Disease evaluations occurred continuously on treatment. Suggested long-term follow-up was monthly for 6m, bi-monthly for 6m, every 4 months for 1y, semi-annually for 1y, then annually. Median follow-up in this study cohort is 6 yrs, up to 10y.

Interventionprobability (Number)
CNS-1.89
CNS-2.89
CNS-31.00
Traumatic Tap With Blasts.84
Traumatic Tap Without Blasts.87

5-Year Disease-Free Survival by MRD Day 32 Status

Disease-free survival (DFS) in a landmark analysis is defined as the duration of time from asparaginase randomization (which occurred after patients achieved complete remission and were assigned to a final risk group) to documented relapse, death during remission or second malignant neoplasm. DFS is estimated based on the Kaplan-Meier method and 5-year DFS is the probability of patients remaining alive, relapse-free and without occurrence of second malignant neoplasm 5 years from asparaginase randomization. Disease relapse is defined as >25% lymphoblasts identified morphologically in bone marrow aspirate/biopsy, or identification of lymphoblasts in marrow (any percentage) identified to be leukemic by flow cytometry, cytogenetics, FISH, immunohistochemistry, or other tests. Appearance of leukemic cells at any extramedullary site (a single, unequivocal lymphoblast in the CSF may qualify as CNS leukemia) also qualifies if confirmed by the PI. (NCT00400946)
Timeframe: Disease evaluations occurred continuously on treatment. Suggested long-term follow-up was monthly for 6m, bi-monthly for 6m, every 4 months for 1y, semi-annually for 1y, then annually. Median follow-up in this study cohort is 6 yrs, up to 10y.

Interventionprobability (Number)
Low Day 32 MRD Level.79
High Day 32 MRD Level.90

5-Year Disease-Free Survival

Disease-free survival (DFS) in a landmark analysis is defined as the duration of time from asparaginase randomization (which occurred after patients achieved complete remission and were assigned to a final risk group) to documented relapse, death during remission or second malignant neoplasm. DFS is estimated based on the Kaplan-Meier method and 5-year DFS is the probability of patients remaining alive, relapse-free and without occurrence of second malignant neoplasm 5 years from asparaginase randomization. Disease relapse is defined as >25% lymphoblasts identified morphologically in bone marrow aspirate/biopsy, or identification of lymphoblasts in marrow (any percentage) identified to be leukemic by flow cytometry, cytogenetics, FISH, immunohistochemistry, or other tests. Appearance of leukemic cells at any extramedullary site (a single, unequivocal lymphoblast in the CSF may qualify as CNS leukemia) also qualifies if confirmed by the PI. (NCT00400946)
Timeframe: Disease evaluations occurred continuously on treatment. Suggested long-term follow-up was monthly for 6m, bi-monthly for 6m, every 4 months for 1y, semi-annually for 1y, then annually. Median follow-up in this study cohort is 6 yrs, up to 10y.

Interventionprobability (Number)
Intramuscular Native E Coli L-asparaginase (IM-EC).89
Intravenous PEG-asparaginase (IV-PEG).90

Asparaginase-Related Toxicity Rate

Asparaginase-related toxicity rate is defined as the percentage of patients who experience allergy (all grades), symptomatic pancreatitis (grade 2 or worse), thrombotic or bleeding complications requiring intervention (grade 2 or worse) with treatment attribution of possibly, probably or definite based on CTCAEv3. (NCT00400946)
Timeframe: 30-week post-induction asparaginase treatment period

Interventionpercentage of participants (Number)
Intramuscular Native E Coli L-asparaginase (IM-EC)26
Intravenous PEG-asparaginase (IV-PEG)28

Induction Infection Toxicity Rate

Infection toxicity rate is defined as the percentage of patients who experience bacterial or fungal infection of grade 3 or higher with treatment attribution of possibly, probably or definite based on CTCAEv3 during remission induction phase of combination chemotherapy. (NCT00400946)
Timeframe: Assessed daily during remission induction days 4-32.

Interventionpercentage of participants (Number)
Overall26

Post-Induction Therapeutic Nadir Serum Asparaginase Activity Rate

Nadir serum asparaginase activity (NSAA) levels were estimated based on established methods. Post-Induction therapeutic NSAA rate is defined as the percentage of patients achieving a NSAA level above 0.1 IU/mL ever during post-induction therapy. (NCT00400946)
Timeframe: Samples for nadir serum asparaginase activity analyses were obtained before doses administered at weeks 5, 11, 17, 23 and 29 of post-induction asparaginase treatment.

Interventionpercentage of participants (Number)
Intramuscular Native E Coli L-asparaginase (IM-EC)71
Intravenous PEG-asparaginase (IV-PEG)99

Induction Serum Asparaginase Activity Level

Serum asparaginase activity (NSAA) levels were estimated based on established methods. (NCT00400946)
Timeframe: Samples for serum asparaginase activity analyses were obtained days 4, 11, 18 and 25 post one-dose of IV-PEG on day 7 of the induction phase.

InterventionIU/mL (Median)
Day 4 NSAA LevelDay 11 NSAA LevelDay 18 NSAA LevelDay 25 NSAA Level
Overall.694.505.211.048

Induction Therapeutic Nadir Serum Asparaginase Activity Rate

Nadir serum asparaginase activity (NSAA) levels were estimated based on established methods. Induction therapeutic NSAA rate is defined as the percentage of patients achieving a NSAA level above 0.1 IU/mL at a given timepoint. (NCT00400946)
Timeframe: Samples for serum asparaginase activity analyses were obtained days 4, 11, 18 and 25 post one-dose of IV-PEG on day 7 of the induction phase.

Interventionpercentage of participants (Number)
Day 4 NSAA RateDay 11 NSAA RateDay 18 NSAA RateDay 25 NSAA Rate
Overall97968712

Post-Induction Nadir Serum Asparaginase Activity Level

Nadir serum asparaginase activity (NSAA) levels were estimated based on established methods. (NCT00400946)
Timeframe: Samples for nadir serum asparaginase activity analyses were obtained before doses administered at weeks 5, 11, 17, 23 and 29 of post-induction asparaginase treatment.

,
InterventionIU/mL (Mean)
Week 5 NSAA LevelWeek 11 NSAA LevelWeek 17 NSAA LevelWeek 23 NSAA LevelWeek 29 NSAA Level
Intramuscular Native E Coli L-asparaginase (IM-EC)0.1290.1430.1590.1800.123
Intravenous PEG-asparaginase (IV-PEG)0.7260.7730.7870.7570.806

Event-Free Survival

Event-free survival defined as the time interval from date of treatment start until the date of death, disease progression or relapse. (NCT03488225)
Timeframe: Start of treatment up to 2 years

InterventionMonths (Median)
Treatment (Hyper-CVAD, Inotuzumab Ozogamicin)24

Number of Participants With Minimal Residual Disease (MRD) Negativity

MRD levels continuously assessed during induction and consolidation therapy by 6-color multiparameter flow. MRD negativity defined by a value of at least 10-4 and confirmed on a second bone marrow aspiration/biopsy performed after a subsequent cycle. (NCT03488225)
Timeframe: Start of treatment up to 2 years

InterventionParticipants (Count of Participants)
Treatment (Hyper-CVAD, Inotuzumab Ozogamicin)4

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT03488225)
Timeframe: Start of treatment up to 2 years

InterventionMonths (Median)
Treatment (Hyper-CVAD, Inotuzumab Ozogamicin)24

Participants to Achieve Complete Remission (CR):

Complete Remission (CR) is defined as - Normalization of the peripheral blood and bone marrow blasts /= 100X10^9/L and complete resolution of all sites of extramedullary disease. (NCT03488225)
Timeframe: Start of treatment up to 2 years

InterventionParticipants (Count of Participants)
Treatment (Hyper-CVAD, Inotuzumab Ozogamicin)4

Number of Participants With Adverse Events

For the purpose of toxicity monitoring, toxicities are defined as any treatment -related grade 3 or 4 non-hematologic AEs occurred any time during the trial.NCI Common Terminology Criteria for Adverse Events (CTCAE) version 4.03 utilized for adverse event reporting. (NCT03488225)
Timeframe: Start of treatment up to 30 days after last dose received.

InterventionParticipants (Count of Participants)
Neutropenic FeverPeripheral Sensory NeuropathyAllergic ReactionMuscle Weakness
Treatment (Hyper-CVAD, Inotuzumab Ozogamicin)2111

Biologic Activity, Defined as Global Deoxyribonucleic Acid (DNA) Methylation Change in Peripheral Blood Mononuclear Cells (PBMC)s; Day 1 Prior to First Course of Azacitidine

Will calculate the percentage of CpG site methylation for all patients before the first course of azacitidine. Mean and standard deviation will be reported. (NCT02828358)
Timeframe: Week 6, Day 1 (Following the induction phase (35 days), the first course of Azacitidine began around Week 6 of therapy)

InterventionPercentage of CpG methylation (Mean)
KMT2A-Rearranged78.17

Biologic Activity, Defined as Global Deoxyribonucleic Acid (DNA) Methylation Change in Peripheral Blood Mononuclear Cells (PBMC)s; Day 1 Prior to Second Course of Azacitidine

Will calculate the percentage of CpG site methylation for all patients before the second course of azacitidine. Mean and standard deviation will be reported. (NCT02828358)
Timeframe: Week 13, Day 1 (Following induction phase (5 weeks), the first course of Azacitidine (1 week), and consolidation (6 weeks), the second course of Azacitidine began around Week 13 of therapy)

InterventionPercentage of CpG methylation (Mean)
KMT2A-Rearranged76.5

Biologic Activity, Defined as Global Deoxyribonucleic Acid (DNA) Methylation Change in Peripheral Blood Mononuclear Cells (PBMC)s; Day 5 of First Course of Azacitidine

Will calculate the percentage of CpG site methylation for all patients after the first course of azacitidine. Mean and standard deviation will be reported. (NCT02828358)
Timeframe: Week 6, Day 5 (Following the induction phase (35 days), the first course of Azacitidine began around Week 6 of therapy)

InterventionPercentage of CpG methylation (Mean)
KMT2A-Rearranged75.54

Biologic Activity, Defined as Global Deoxyribonucleic Acid (DNA) Methylation Change in Peripheral Blood Mononuclear Cells (PBMC)s; Day 5 of Second Course of Azacitidine

Will calculate the percentage of CpG site methylation for all patients after the second course of azacitidine. Mean and standard deviation will be reported. (NCT02828358)
Timeframe: Week 13, Day 5 (Following induction phase (5 weeks), the first course of Azacitidine (1 week), and consolidation (6 weeks), the second course of Azacitidine began around Week 13 of therapy)

InterventionPercentage of CpG methylation (Mean)
KMT2A-Rearranged74.52

Tolerability of Azacitidine in Combination With Interfant-06 Standard Chemotherapy in Evaluable Infant Patients With Newly Diagnosed ALL With KMT2A Gene Rearrangement (KMT2A-R). KMT2A Gene Rearrangement (KMT2A-R)

Proportion of KMT2A-Rearranged patients treated with azacitidine with Dose Limiting Toxicities (DLTs) from the first course of azacitidine administration up to fourth course of azacitidine administration. (NCT02828358)
Timeframe: 6 months

Interventionpercentage of participants (Number)
KMT2A-Rearranged6.45

Number of Participants Who Experienced Dose-limiting Toxicity (DLT)

Defined as induction mortality (death occurring on or before day 60), grade 3 or 4 non-hematologic toxicity, or dose limiting hematologic toxicity at least possibly related to the study drug occurring during the first 28 days from the start of therapy. Estimated for each arm with 95% confidence intervals. Fisher's exact test will be used to compare the toxicity rate between the two dose levels. (NCT02286726)
Timeframe: Up to day 60

InterventionParticipants (Count of Participants)
Arm I (Lower-dose (50 Units/m^2) CPX-351)5
Arm II (Intermediate-dose (75 Units/m^2) CPX-351)3
Arm III (Standard-dose (100 Units/m^2) CPX-351)3

Number of Participants With a Response

Response is defined as Complete response (CR) or CR with incomplete blood count recovery (CRi) rate: Complete Remission (CR) is Bone marrow blasts <5%; absence of blasts with Auer rods; absence of extramedullary disease; absolute neutrophil count >1.0 x 10^9/L (1000/μL); platelet count >100 x 10^9/L (100,000/μL). Complete Response with incomplete blood count recovery (CRi) is All CR criteria except for residual neutropenia (<1.0 x 10^9/L [1000/μL]) and/or thrombocytopenia (<100 x 10^9/L [100,000/μL]). (NCT02286726)
Timeframe: Up to 8 weeks (after induction therapy)

,,
InterventionParticipants (Count of Participants)
Complete Response (CR)Complete Response with incomplete blood count recovery (CRi)
Arm I (Lower-dose (50 Units/m^2) CPX-351)30
Arm II (Intermediate-dose (75 Units/m^2) CPX-351)63
Arm III (Standard-dose (100 Units/m^2) CPX-351)70

Describe FLT3 Protein Expression as a Molecular Mechanism of Acquired Resistance to Lestaurtinib in Leukemic Blasts

Described via means and standard deviations in available Arm C relapse samples (NCT00557193)
Timeframe: At relapse (up to 3 years)

InterventionqPCR fold expression ratio (Mean)
Arm C (Safety/Efficacy Dose Level 2)5.73

Describe FLT3 Protein Expression as a Molecular Mechanism of Primary Resistance to Lestaurtinib in Leukemic Blasts

Described via mean and standard deviation by group. (NCT00557193)
Timeframe: Sampled at the start of induction

InterventionqPCR fold expression ratio (Mean)
Arm A (Standard Risk MLL-G)1.25
Arm B (IR/HR MLL-R Chemotherapy)7.85
Arm C (IR/HR MLL-R Chemotherapy and Lestaurtinib)5.83

Describe in Vitro Sensitivity as a Molecular Mechanism of Acquired Resistance to Lestaurtinib in Leukemic Blasts

Described via means and standard deviations in samples which have acquired resistance to lestaurtinib (NCT00557193)
Timeframe: At relapse (up to 3 years)

InterventionProportion of cells that are viable (Mean)
Arm C (IR/HR MLL-R Chemotherapy and Lestaurtinib)0.69

Describe in Vitro Sensitivity as a Molecular Mechanism of Primary Resistance to Lestaurtinib in Leukemic Blasts

Described via means and standard deviations in samples which have primary resistance to lestaurtinib (NCT00557193)
Timeframe: Sampled at the start of induction

InterventionProportion of cells that are viable (Median)
Arm A (Standard Risk MLL-G)0.75
Arm B (IR/HR MLL-R Chemotherapy)0.48
Arm C (IR/HR MLL-R Chemotherapy and Lestaurtinib)0.47

Number of Patients Who Experienced Lestaurtinib-related Dose Limiting Toxicity (DLT)

Lestaurtinib-related dose-limiting toxicity proportions, as measured by NCI Common Terminology Criteria for Adverse Events (CTCAE) version 4.0, will by summarized by dose level for Safety phase patients. (NCT00557193)
Timeframe: Up to 12 weeks from start of induction

InterventionParticipants (Count of Participants)
Arm C (IR/HR MLL-R Chemotherapy and Lestaurtinib-Dose Level 1)0
Arm C (IR/HR MLL-R Chemotherapy and Lestaurtinib-Dose Level 2)1

Percent Probability for Event-free Survival (EFS) for Patients on Arm A

EFS time is defined as time from treatment assignment to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. EFS is constructed using the Kaplan-Meier life table method with confidence interval based on standard errors computed using the method of Peto and Peto. (NCT00557193)
Timeframe: From start of post-induction therapy for up to 10 years

Interventionpercentage probability (Number)
Arm A (Standard Risk MLL-G)86.67

Percent Probability for Event-free Survival (EFS) for Patients on Arm C at Dose Level 2 (DL2)

EFS time is defined as time from randomization to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. EFS is constructed using the Kaplan-Meier life table method with confidence interval based on standard errors computed using the method of Peto and Peto. (NCT00557193)
Timeframe: From start of post-induction therapy for up to 10 years

Interventionpercentage probability (Number)
Arm C (Safety/Efficacy Dose Level 2)35.82

Percent Probability for Event-free Survival (EFS) of MLL-R Infants Treated With Combination Chemotherapy With or Without Lestaurtinib at DL2

Event Free Probability where EFS time is defined as time from randomization to first event (relapse, second malignant neoplasm, death) or date of last contact for patients who are event-free. EFS is constructed using the Kaplan-Meier life table method with confidence interval based on standard errors computed using the method of Peto and Peto. EFS will be compared between patients on treatment Arm C at DL2 to those on Arm B. (NCT00557193)
Timeframe: From start of post-induction therapy for up to 10 years.

Interventionpercent probability (Number)
Arm B (IR/HR MLL-R Chemotherapy)38.89
Arm C (IR/HR MLL-R Chemotherapy and Lestaurtinib)35.82

Percent Probability of Event Free Survival (EFS) by MRD Status and Treatment Arm

Three-year EFS estimates and 90% CI will be reported by treatment arm and end-induction MRD status. (NCT00557193)
Timeframe: 3 Years from end of Induction)

Interventionpercent probability (Number)
Arm A (MRD Negative)86.05
Arm A (MRD Positive)87.5
Arm B (MRD Negative)47.37
Arm B (MRD Positive)22.73
Arm C (MRD Negative)51.85
Arm C (MRD Positive)27.03

Pharmacodynamics PIA Levels in Infants Given Lestaurtinib at DL2 in Combination With Chemotherapy

Summarized with mean and standard deviation for those with available data in Arm C (NCT00557193)
Timeframe: Sampled between weeks 6-12 from start of induction

InterventionActivity percentage (Mean)
Arm C (Safety/Efficacy Dose Level 2)69.00

Dose Escalation Phase: Number of Participants With Dose-Limiting Toxicities (DLTs) During the First Cycle of Induction Treatment

Sponsor's decision to prematurely terminate the study was made at the end of the Phase 1. The planned Phase 2 was not initiated. This decision was not based on safety concerns, but rather due to the overall company strategy about the adult AML (NCT03850535)
Timeframe: Cycle 1 of induction treatment (1 cycle is 28 days)

InterventionParticipants (Number)
Dose-Escalation Cohort 12
Dose Escalation Cohort 24
Dose Escalation Cohort 30
Post Consolidation Cohort0

Number of Participants With at Least One Adverse Event

Sponsor's decision to prematurely terminate the study was made at the end of the Phase 1. The planned Phase 2 was not initiated. This decision was not based on safety concerns, but rather due to the overall company strategy about the adult AML. (NCT03850535)
Timeframe: From Baseline until 28 days after the final dose of study drug (up to 2 years)

InterventionPercentage of Participants (Number)
Dose-Escalation Cohort 14
Dose Escalation Cohort 29
Dose Escalation Cohort 36
Post Consolidation Cohort4

Number of Participants With Grade ≥3 Adverse Events, Severity Determined According to National Cancer Institute Common Terminology Criteria for Adverse Events, Version 5.0 (NCI CTCAE v5.0)

Sponsor's decision to prematurely terminate the study was made at the end of the Phase 1. The planned Phase 2 was not initiated. This decision was not based on safety concerns, but rather due to the overall company strategy about the adult AML (NCT03850535)
Timeframe: From Baseline until 28 days after the final dose of study drug (up to 2 years)

InterventionParticipants (Number)
Dose-Escalation Cohort 14
Dose Escalation Cohort 29
Dose Escalation Cohort 35
Post Consolidation Cohort4

Complete Response Rate

The complete response rate will be defined as the total number of patients who have defined complete response using study regimen (intensive induction therapy/progressive chemotherapy/stem cell rescue), divided by the number of patients entered in the trial using response-evaluable patients. (NCT00632827)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
Treatment Plan14

Median Time to Response

The time to response is measured from the time measurement criteria are met for complete response or partial response (whichever is first recorded) until the first date that recurrent or progressive disease is objectively documented (taking as reference for progressive disease the smallest measurements recorded since the treatment started) in months. (NCT00632827)
Timeframe: Up to 2 years

Interventionmonths (Median)
Treatment Plan3.0

Overall Survival Rate

Overall Survival will be defined the percentage of participants alive at median follow up of 25 months. If the patient is lost to follow-up, survival will be censored on the last date the patient was known to be alive. The analysis is expected to occur up to 60 months after the first patient is entered the trial. (NCT00632827)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Treatment Plan75

Progression Free Survival

Progression-Free Survival will be defined the percentage of participants alive and progression-free at median follow up of 25 months. Patients will be routinely followed for disease progression and those who die without a reported prior progression will be considered to have progressed on the day of their death. Patients who did not progress or die will be censored at the day of their last treatment assessment. Patients who have not received study regimen or did not have on-study treatment assessments will be censored on the day they entered the trial. Patients who receive chemotherapy for reasons other than documented progression of disease or clinical progression without documented progression will be censored on the earliest date of subsequent therapy (NCT00632827)
Timeframe: Up to 3 years

Interventionpercentage of partcipants (Number)
Treatment Plan65

Median Time to Progression

Time to progression (TTP) is defined as the time from treatment after OVA until documented lymphoma progression or receipt of anti-lymphoma therapy (except for planned post-ASCT radiotherapy) or death due to lymphoma. Patients are to be censored at the time of last followup or death due to another cause (NCT01555541)
Timeframe: Up to 48 months

Interventionmonths (Median)
Treatment13.2

Number of Participants With Successful Neutrophil Engraftments

Neutrophil engraftment is defined as the first day of 3 consecutive days with absolute neutrophil count of >500 cells/microlitre (uL). Response evaluation will occur at day +90 after ASCT, and at 6, 12 and 24 months thereafter (NCT01555541)
Timeframe: Up to 24 months after ASCT

Interventionparticipants (Number)
Treatment12

Number of Participants With Successful Platelet Engraftments

Platelet engraftment is defined as the first of three consecutive measurements for which the platelet count was > 20,000/uL, and must be at least 24 hours following the last platelet transfusion. Response evaluation will occur at day +90 after ASCT, and at 6, 12 and 24 months after ASCT (NCT01555541)
Timeframe: Up to 24 months after ASCT

Interventionparticipants (Number)
Treatment12

Number of Patients Achieving Complete Response (CR) to the Treatment Upon Successful Stem Cell Mobilization

CR will be calculated based on the PET/CT scan following 2 cycles of salvage therapy using the revised International Working Group (IWG) Criteria for a lymphoma response. This includes: Complete disappearance of all detectable clinical evidence of disease and disease-related symptoms if present before therapy, Post-treatment residual mass of any size is permitted as long as it is PET-, Spleen and/or liver, if enlarged before therapy based on physical exam or CT scan, should not be palpable on physical exam and should be considered normal size by imaging studies, and nodules related to lymphoma should disappear, If bone marrow was involved by lymphoma before treatment, infiltrate must have cleared on repeat bone marrow biopsy. Biopsy sample must be adequate (with goal of > 20 mm unilateral core). If sample is indeterminate by morphology, it should be negative by immunohistochemistry. A sample that is negative by immunohistochemistry but demonstrates small population of clonal l (NCT01555541)
Timeframe: Day 42

InterventionParticipants (Count of Participants)
Treatment10

Number of Patients Achieving Mobilization-adjusted Complete Response (maCR)

Number of patients achieving maCR to the treatment upon successful stem cell mobilization, defined as at least 2 x10^6 cluster of differentiation 34 (CD34)+cells/Kg of actual body weight. Patients who require use of plerixafor or an autologous bone marrow harvest are considered mobilization failures and will be treated as non-responders. (NCT01555541)
Timeframe: Day 42

InterventionParticipants (Count of Participants)
Treatment10

Number of Patients Who Advance From Partial Response (PR) to Complete (CR)

Fluorodeoxyglucose-positron emission tomography (FDG-PET) conversion rate was used determined the number of participants who improved following OVA Treatment. (NCT01555541)
Timeframe: Up to 5 months

InterventionParticipants (Count of Participants)
Treatment2

Number of Patients Who Received OVA and Then Met Criteria to Proceed to ASCT and Achieved a CR/Partial Response (PR) Post-ASCT

Determination of CR or PR must meet the revised International Working Group (IWG) Criteria for lymphoma response (NCT01555541)
Timeframe: Up to 5 months

InterventionParticipants (Count of Participants)
Treatment11

Overall Survival Rate (OS)

The percentage of participants in the study still alive at time of censoring. The time frame is defined as the time from day 0 of OVA treatment until death as a result of any cause. Patients will be censored at the time of last followup (NCT01555541)
Timeframe: Up to 24 months

Interventionpercentage of participants (Number)
Treatment59

Progression Free Survival Rate

The percentage of participants in the study still alive at time of censoring. The time frame defined as the time from day 0 of OVA treatment until lymphoma progression, receipt of anti-lymphoma therapy (except for planned post-ASCT radiotherapy), or death as a result of any cause. Patients will be censored at the time of median follow-up. (NCT01555541)
Timeframe: Up to 24 months

Interventionpercentage of participants (Number)
Treatment47

Overall Response Rate (ORR)

Percentage of participants with complete response following treatment out of all participants in that particular treatment group. Response assessed by blood test or bone marrow aspirate following day 21 of induction and then every 2 weeks thereafter until remission or non-response. Complete remission (CR): Disappearance all clinical and/or radiologic evidence of disease; Neutrophil count > 1.0 x10^9/L and platelet count >100x10^9/L, and normal bone marrow differential (< 5% blasts). Complete remission without platelet recovery (CRp): Peripheral blood and bone marrow results as for CR, but with platelet counts of > 20 x 10^9/L and < 100 x 10^9/L. Partial remission (PR): Peripheral blood count recovery as for CR, but with decrease in marrow blasts of > 50% and not more than 6-25% abnormal cells in the marrow. All other responses considered as failures. (NCT00067028)
Timeframe: Up to 6 years

InterventionPercentage of Participants (Number)
Clofarabine + Ara-C36
Clofarabine + Idarubicin44
Clofarabine + Idarubicin + Ara-C24

Participants With a Response

Response assessed by blood test or bone marrow aspirate following day 21 of induction and then every 2 weeks thereafter until remission or non-response. Complete remission (CR): Disappearance all clinical and/or radiologic evidence of disease; Neutrophil count > 1.0 x10^9/L and platelet count >100x10^9/L, and normal bone marrow differential (< 5% blasts). Complete remission without platelet recovery (CRp): Peripheral blood and bone marrow results as for CR, but with platelet counts of > 20 x 10^9/L and < 100 x 10^9/L. Partial remission (PR): Peripheral blood count recovery as for CR, but with decrease in marrow blasts of > 50% and not more than 6-25% abnormal cells in the marrow. All other responses considered as failures. (NCT00067028)
Timeframe: Up to 6 years

,,
Interventionparticipants (Number)
CRCRpPR
Clofarabine + Ara-C420
Clofarabine + Idarubicin951
Clofarabine + Idarubicin + Ara-C1671

Overall Remission Rate (CR+CRp)

Overall Remission Rate for the CR/CR with incomplete platelet count recovery (CRp) rate after up to 4 cycles of induction/re-induction therapy. (NCT01804101)
Timeframe: Up to day 28

InterventionParticipants (Count of Participants)
Arm I (Lower-dose CPX-351)10
Arm II (Higher Dose COX-351)1

Treatment-related Mortality Rate. (TRM)

Estimate whether the 32 units/m2 or the 64 units/m2 or both dose levels of CPX-351 are likely to improve treatment-related mortality (TRM) rate while keeping the CR rate constant in patients with untreated high-risk MDS or non-APL AML at high risk of TRM. (NCT01804101)
Timeframe: Up to 1 month after completion of study treatment

InterventionParticipants (Count of Participants)
Arm I (Lower-dose Liposomal Cytarabine-daunorubicin CPX-351)6
Arm II (Closed to Accrual Effective 4/21/14)2

Event-free Survival

The Kaplan-Meier method will be used to estimate the event-free survival distribution. (NCT01035463)
Timeframe: 1 year

Interventionpercentage of participants (Number)
All Phase I Participants84
All Phase II Participants87

Maximum Tolerated Dose of Lenalidomide (Phase I)

The Maximum Tolerated Dose (MTD) is defined to be the dose cohort below which 3 out of 6 subjects experience dose limiting toxicities during cycle 1. Dose limiting toxicities graded using the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0 (NCT01035463)
Timeframe: Cycle 1, 28 days

Interventionmilligrams PO daily (Number)
Treatment (Stem Cell Transplantation)10

Overall Survival

The Kaplan-Meier method will be used to estimate the overall survival distribution. This outcome only reports data as it pertains to overall survival at one year. All-cause mortality includes survival for follow up for all subjects on the study. (NCT01035463)
Timeframe: 1 year

Interventionpercentage of participants (Number)
All Phase I Participants100
All Phase II Participants95

2 Year Event Free Survival

Percentage of patients who were event free at 2 years. The 2-year event free rate was estimated using the Kaplan Meier method. An event is defined as death, progression or treatment failure. (NCT00039130)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Rituximab With High Intensity Chemotherapy78

2 Year Overall Survival

Percentage of participants who were alive at 2 years. The 2 year survival, with 95% confidence interval, was estimated using the Kaplan Meier method. (NCT00039130)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Rituximab With High Intensity Chemotherapy80

Complete Response Rate

Response is assessed by investigator according to Revised Response Criteria for Malignant Lymphoma. Complete response requires disappearance of all evidence of disease. (NCT00039130)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Rituximab With High Intensity Chemotherapy83

Number of Participants With Response

Patient response defined by: Death, Resistant to Therapy [no major hematologic improvement using International Myelodysplastic Syndromes (MDS) Working Group (Cheson B, Bennett J, Kantarjian H et al, Blood 2006) criteria after a maximum of 4 courses], or Relapse. (NCT00382590)
Timeframe: Evaluated every 3 weeks, following 4 courses (16/24 weeks ) and till study end

,
InterventionParticipants (Number)
DeathResistant to TherapyRelapse
5-Aza + VPA040
Ara-C050

Disease-free Survival (DFS)

DFS defined as time from transplantation to disease relapse, disease progression, death during remission, or last follow-up. Evaluation at 3 months and 6 months after transplantation, then every 6 months for 3 years, and then once a year up to 5 years from the transplant date. (NCT00472056)
Timeframe: Up to 5 years from transplant date.

Interventionmonths (Mean)
Standard Dose Rituximab11.33
High Dose Rituximab9.635

Number of Patients That Achieved Complete Response to ABT-751

Complete response (CR) is the occurrence of all of the following on approximately Day 29: less than 5% leukemic blasts in the bone marrow aspirate with no evidence of leukemic blasts in the CSF or peripheral blood and recovery of peripheral blood counts of an Absolute neutrophil count (ANC) > 750/μL and Platelet count > 75,000 μL. (NCT00439296)
Timeframe: Day 29 of Course 1

,
InterventionParticipants (Count of Participants)
# of patients not achieving complete response# of patients who achieved complete response
Dose Level 013
Dose Level 141

Number of Patients That Experienced Dose Limiting Toxicity From ABT-751

"ABT-751 was given daily for 21 days for a period of 28 day course in combination with dexamethasone, PEG-asparaginase, and doxorubicin. The occurrence of a dose limiting toxicity (DLT) was evaluated at the end of the 28 day course.~DLT will be defined as any of the following events that are deemed by the investigator as probably or definitely attributable to ABT-751. Toxicity grade follows the CTCAE criteria, version 3.0. A copy of the CTCAE can be downloaded from the CTEP home page (http://ctep.cancer.gov).~Grade 3 or 4 Ileus~Grade 3 or 4 Constipation~Grade 3 or 4 Gastrointestinal obstruction, any location~Grade 3 or 4 Sensory Neuropathy~Grade 3 or 4 Motor Neuropathy~Grade 3 or 4 Neuropathic pain lasting longer than 24 hours despite medical intervention~Grade 3 or 4 Hypoxia in the absence of anemia or infection~Grade 4 Alanine aminotransferase (ALT) which does not return to" (NCT00439296)
Timeframe: Each dose level is evaluated

,
InterventionParticipants (Count of Participants)
# of patients with DLT# of patients without DLT
Dose Level 004
Dose Level 123

Number of Patients With Occurrence of Toxic Death

The occurrence of toxic death at anytime that is definitely, probably or possibly related to the treatment. (NCT00439296)
Timeframe: From the first dose of study therapy until 30 days after last therapy dose. Last dose protocol therapy is on day 21.

,
InterventionParticipants (Count of Participants)
# of patients that experienced toxic death# of patients that did not experience toxic death
Dose Level 004
Dose Level 105

Number of Participants With Complete Remission

Clinical response is determined by achievement of a complete remission (CR) as judged by morphological criteria (< 1% blasts in bone marrow with neutrophil recovery) according to International Working Group (IWG) criteria. (NCT00569010)
Timeframe: 6 weeks

Interventionparticipants (Number)
Low-Dose Ara-C + AZA-Level 00
Low-Dose Ara-C + AZA-Level 10
High-Dose Ara-C + AZA-Level 00
High-Dose Ara-C + AZA-Level 12

Aplasia Rate

Bone marrow aplasia was defined as <20% cellularity and 5% blasts in the bone marrow aspiration evaluation. (NCT00788892)
Timeframe: Day 14 (1st Induction)

InterventionParticipants (Count of Participants)
Arm A: CPX-35155
Arm B: Cytarabine + Daunorubicin15

Event Free Survival

Event-free survival begins from randomization to the date persistent disease is documented or date of relapse after CR, or death, whichever comes first. (NCT00788892)
Timeframe: Up to 1 year from randomization

Interventiondays (Median)
Arm A: CPX-351161
Arm B: Cytarabine + Daunorubicin55

Number of Participants With Complete Remission

Response was defined according to International Working Group Criteria (Cheson, et al. 2003) which requires peripheral blood neutrophils of >1000/µL and peripheral blood platelets of >100,000/µL in the absence of bone marrow blasts. (NCT00788892)
Timeframe: Within 6 weeks of the last induction treatment

InterventionParticipants (Count of Participants)
Arm A: CPX-35141
Arm B: Cytarabine + Daunorubicin20

Overall Survival Rate at 1 Year

Survival defined as the time from randomization to death. (NCT00788892)
Timeframe: 1 year

Interventionparticipants (Number)
Arm A: CPX-35139
Arm B: Cytarabine + Daunorubicin18

Rate of Stem Cell Transplant

The rate of patients who underwent stem cell transplant. (NCT00788892)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Arm A: CPX-35113
Arm B: Cytarabine + Daunorubicin10

Remission Duration/Time to Remission

"Remission Duration was assessed from the time measurement criteria for CR were met until the first date that disease relapse was objectively documented or the subject died.~Time to remission was measured from the date of randomization to the time measurement criteria for CR were first met." (NCT00788892)
Timeframe: Following achievement of CR over the study period

,
Interventiondays (Median)
Remission DurationTime to Remission
Arm A: CPX-35127549
Arm B: Cytarabine + Daunorubicin23540

Disease Free Survival

Median disease-free survival (NCT00839982)
Timeframe: Up to 5 years

Interventionmedian months (Median)
Treatment (Chemotherapy)7.4

Maximum Tolerated Dose

We identified 20 mg/d for 5 d as the maximum tolerated dose (MTD) of oral clofarabine. (NCT00839982)
Timeframe: up to 5 years

Interventionmg/day (Number)
Treatment (Chemotherapy)20

Number of Patients With Dose Limiting Toxicity

Dose limiting toxicity (DLT) consists of grade 3-4 non-hematologic toxicity at least possibly related to study drug. Exceptions include neutropenic fever; drug-related fever; alopecia; anorexia; inadequately treated nausea, vomiting and/or diarrhea; and grade 3/4 increase in ALT, AST, or bilirubin recovering to < grade 2 by 7 days. Prolonged grade 2 myelosuppression lasting longer than 49 days in patients who don't proceed to additional cytotoxic therapy is considered a DLT. The MTD or recommended phase II dose is the highest dose level at which no more than 1 patient out of 6 experiences DLT. (NCT00839982)
Timeframe: Outcomes by day 30

InterventionParticipants (Count of Participants)
Dose Level 10
Dose Level 24
Dose Level 32
Dose Level 42

Overall Survival

Median overall survival (NCT00839982)
Timeframe: Up to 5 years

Interventionmedian months (Median)
Treatment (Chemotherapy)6.8

Occurrence of a Dose-Limiting Toxicity

The MTD in each stratum will be the highest dose at which 1 or fewer of six patients experience DLT during cycle 1 of therapy. (NCT00928200)
Timeframe: Beginning with the first dose of investigational product until 30 days following the last dose of Erwinase

InterventionParticipants (Count of Participants)
Single Arm0

Event-Free Survival (EFS)

Number of patients surviving without relapsed/progressive disease (NCT01008462)
Timeframe: 1 Year post-autograft

InterventionParticipants (Count of Participants)
Treatment (Autologous HCT, Donor HCT)9

Number of Patients Who Engrafted

Number of patients with donor engraftment. (NCT01008462)
Timeframe: Day 84 post-allograft

InterventionParticipants (Count of Participants)
Treatment (Autologous HCT, Donor HCT)13

Number of Patients Who Had Infections

Number of patients who had infections. (NCT01008462)
Timeframe: 1 Year post-autograft

InterventionParticipants (Count of Participants)
Treatment (Autologous HCT, Donor HCT)16

Number of Patients With Relapsed/Progressive Disease

"Relapse/Progression defined as:~Nodes, liver, and/or spleen ≥50% increased or new by physical exam / imaging studies.~Circulating lymphocytes ≥50% increased by morphology and/or flow cytometry. Richter's transformation by lymph node biopsy ." (NCT01008462)
Timeframe: 1 year post-autograft

InterventionParticipants (Count of Participants)
Treatment (Autologous HCT, Donor HCT)6

Overall Survival

Number of patients surviving one year post-autograft (NCT01008462)
Timeframe: 1 year post-autograft

InterventionParticipants (Count of Participants)
Treatment (Autologous HCT, Donor HCT)10

Non-relapse Mortality (NRM)

Number of patients with non-relapse mortalities. (NCT01008462)
Timeframe: 200 days and 1 Year post-allograft

InterventionParticipants (Count of Participants)
200 days1 Year
Treatment (Autologous HCT, Donor HCT)01

Number of Patients With Grade II-IV Acute Graft-versus-Host-Disease and/or Chronic Extensive Graft-versus-Host-Disease

"aGVHD The diagnosis of aGVHD is identified through various stages and grading of the disease related to Skin (Rash), Gut (Diarrhea, Nausea/vomiting and/or anorexia) and the liver (Bilirubin) assessed by severity and grading scale outlined in the section Grafts vs Hosts by Sullivan (1999).~GVHD Grades Grade I: 1-2 Skin Rash; No gut or liver involvement Grade II: Stage 1-3 Skin rash; Stage 1 gut and/or stage 1 liver involvement Grade III: Stage 2-4 gut involvement and/or stage 2-4 liver involvement with or without rash Grade IV: Pattern and severity of GVHD similar to grade 3 with extreme constitutional symptoms or death~CGVHD The diagnosis of cGVHD requires at least one manifestation that is distinctive for chronic GVHD as opposed to acute GVHD. In all cases, infection and others causes must be ruled out in the differential diagnosis of chronic GVHD." (NCT01008462)
Timeframe: 1 year post-allograft,

InterventionParticipants (Count of Participants)
Acute Graft-versus-Host-DiseaseChronic extensive Graft-versus-Host-Disease
Treatment (Autologous HCT, Donor HCT)81

Complete Response in the Absence of Platelet Recovery

"Complete response in the absence of platelet recovery is defined as:~- Bone marrow (<5% blasts) with adequate bone marrow cellularity, no evidence of circulating blasts or extramedullary disease and normalization of peripheral blood counts except for platelets (neutrophil count =1,000/µL)" (NCT01184898)
Timeframe: Within one week of peripheral count recovery but no later than day 42

Interventionparticipants (Number)
Sirolimus and MEC2

Complete Response

"Complete response is defined as:~Peripheral Blood Counts -Neutrophil count >1 x 109/L.~Platelet count ≥ 100 x 109/L.~Reduced hemoglobin concentration or hematocrit has no bearing on remission status.~Leukemic blasts must not be present in the peripheral blood.~Cellularity of bone marrow biopsy must be > 20% with maturation of all cell lines with < 5% blasts and no Auer rods.~Extramedullary leukemia, such as CNS or soft tissue involvement, must not be present" (NCT01184898)
Timeframe: Within one week of peripheral count recovery but no later than day 42

Interventionparticipants (Number)
Sirolimus and MEC11

Partial Response

"Partial response is defined as:~Requires that all of the criteria for complete remission be satisfied except that the bone marrow may contain ≥ 5% blasts but < 25% blasts.~A marrow with <5% blasts that contain Auer rods will also be considered a PR" (NCT01184898)
Timeframe: Within one week of peripheral count recovery but no later than day 42

Interventionparticipants (Number)
Sirolimus and MEC3

Association Between the Magnitude of mTOR Target Inhibition Post-treatment in Leukemic Blasts and Clinical Response in Patients With High Risk AML Treated With Sirolimus MEC

"Percent change compared between response groups (responder vs nonresponder).~This outcome measure only includes patients who survived to outcome assessment." (NCT01184898)
Timeframe: From pre- to post-treatment

Interventionpercentage change in leukemic blasts (Mean)
Responders (17 pts)Nonresponders (10 pts)
Sirolimus and MEC69-36

4-year Event Free Survival

Event Free Survival defined as the time from the start of therapy to time of primary refractory disease, relapse from CR, death from any cause or last follow-up. Kaplan-Meier method will be utilized to analyze event free survival for the 4 year percent alive and in CR. (NCT01363128)
Timeframe: Up to 4 years

InterventionParticipants (Count of Participants)
Treatment (Hyper-CVAD, Ofatumumab)41

4-Year Overall Survival

Overall Survival is defined as time from date of treatment start until date of death due to any cause or last Follow-up. For continuous data, summary statistics including n, mean, standard deviation, median, minimum and maximum will be computed. The posterior median time to event and it 95% credible interval will be estimated. Kaplan-Meier method, Log rank test and Cox proportional hazards regression modeling will be utilized to analyze survival at 4 years. (NCT01363128)
Timeframe: Up to 4 years

InterventionParticipants (Count of Participants)
Treatment (Hyper-CVAD, Ofatumumab)47

Number of Participants With Complete Remission (CR)

Complete Remission is Normalization of the peripheral blood and bone marrow with 5% or less blasts in marrow with a granulocyte count of 1 x 109/L or above and a platelet count of 100x 109/L or above. Complete resolution of all sites of extramedullary disease is required for CR. (NCT01363128)
Timeframe: Up to 8 years

InterventionParticipants (Count of Participants)
Treatment (Hyper-CVAD, Ofatumumab)68

Maximal Tolerated Dose (MTD) of Ruxolitinib in Combination With Chemotherapy Defined as the Highest Dose Level at Which no More Than 1 Out of 6 Patients Experience a Dose Limiting Toxicity (Phase I)

The method of Thall, Simon and Estey will be used for toxicity monitoring for this study. The severity of the toxicities will be graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0 whenever possible. Safety data will be summarized by category, severity and frequency. (NCT02420717)
Timeframe: 42 days

InterventionMilligrams (mg) (Number)
Phase I Ruxolitinib 15mg25

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT02420717)
Timeframe: Up to 4 years 7 months

InterventionMonths (Median)
Phase I Ruxolitinib 15mg4.8
Phase I Ruxolitinib 20mg5.4
Phase I Ruxolitinib 25mg38.5

Participants With Complete Response (Complete Response [CR]/CR With Incomplete Marrow Recovery [CRi]) (Phase II)

Complete Response (CR) is disappearance of all clinical and/or radiologic evidence of disease, Neutrophil count ≥ 1.0 x 10^9/L, Platelet count ≥ 100 x 10^9/L, Normal bone marrow differential (≤ 5% blasts), No extra-medullary leukemia. Complete Remission with Incomplete Blood Count Recovery (CRi) is CR except for ANC < 1.0 x 10^9/L and/or platelets < 100 x 10^9/L. (NCT02420717)
Timeframe: 42 days

InterventionParticipants (Count of Participants)
Phase I Ruxolitinib 15mg0
Phase I Ruxolitinib 20mg1
Phase I Ruxolitinib 25mg0

Progression-free Survival

Time from date of treatment start until the date of first objective documentation of disease-relapse. (NCT02420717)
Timeframe: Up to 4 years 7 months

InterventionMonths (Median)
Phase I Ruxolitinib 15mg2.3
Phase I Ruxolitinib 20mg1.8
Phase I Ruxolitinib 25mg1.9

Duration of Relapse-free Survival (for Patients Achieving CR or CRp)

Categorized according to criteria recommended by International Working Groups. (NCT01729845)
Timeframe: Up to 5 years

InterventionDays (Median)
Dose Level 2: 7-Days of Decitabine-MEC150

Overall Survival

Survival measured as of day of last contact. Categorized according to criteria recommended by International Working Groups. (NCT01729845)
Timeframe: Up to 5 years

Interventiondays (Median)
Dose Level 2: 7-Days of Decitabine-MEC564

Remission Rate Including CR and CRp

"Complete remission (CR) and Complete remission with incomplete platelet recovery (CRp) categorized according to criteria recommended by International Working Groups:~Complete resolution of disease-related symptoms and signs including palpable hepatosplenomegaly; hemoglobin level at least 110 g/L, platelet count at least 100x10^9/L, and absolute neutrophil count at least 1.0 x10^9/L. In addition, all 3 blood counts should be no higher than the upper normal limit; Normal leukocyte differential; Bone marrow histologic remission defined as the presence of age-adjusted normocellularity, no more than 5% myeloblasts, and an osteomyelofibrosis grade no higher than 1." (NCT01729845)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Dose Level 2: 7-Days of Decitabine-MEC11

Most Efficacious and Tolerated Dosage of Decitabine (Period 1)

MTD (most tolerated dose) of decitabine, measured in number of dose limiting toxicities. MTD defined as the highest dose in which the incidence of dose limiting toxicity is < 33%, graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0 (Phase I) (NCT01729845)
Timeframe: through day 45

,,
InterventionIncidents (Number)
Dose-limiting toxiticiesComplete RemissionComplete Remission, incomplete PLT recoveryComplete Remission, incomplete blood count recoverMorphologic leukemia-free stateResistant DiseaseDeath (among those who received MEC)
Dose Level 1: 5-Days of Decitabine-MEC0120012
Dose Level 2: 7-Days of Decitabine-MEC0511031
Dose Level 3: 10-Days of Decitabine-MEC0320340

3-year Event Free Survival (EFS)

We will use the Kaplan-Meier method to describe event-free survival. EFS will be defined as the time from enrollment to death, relapse, or refractory disease with event-free subjects' time censored at the date of last follow-up. (NCT02433483)
Timeframe: 3 years after enrollment of the last participant

InterventionPercentage of participants (Number)
Myeloid Malignancies0

3-year Overall Survival (OS)

We will use the Kaplan-Meier method to describe overall survival. Overall survival will be defined as the time from enrollment to death, with living subjects' time censored at the date of last follow-up (NCT02433483)
Timeframe: 3 years after enrollment of the last participant

InterventionPercentage of participants (Number)
Myeloid Malignancies0

Median Time to Neutrophil Recovery

The time to neutrophil recovery will be summarized using descriptive statistics. If there are no deaths prior to recovery of neutrophils, nonparametric confidence intervals for the median time to recovery will be computed by inverting the sign test. Otherwise, we will compute cumulative incidence curves to describe the time to platelet and neutrophil recovery while adjusting for competing events. (NCT02433483)
Timeframe: From start of therapy to completion of therapy (approximately 1 year)

InterventionDays (Median)
Myeloid Malignancies14.5

Number of Participants by Stratum Who Complete 2 Cycles of Therapy

If two or more patients die from causes other than leukemia progression or experience ≥ Grade 3 GVHD that is associated with detectable donor chimerism due to this protocol, or demonstrate persistent engraftment defined as >5% donor chimerism at the time of count recovery (ANC > 0.3 x 10^9/L and platelet count > 30 x 10^/L), then the cohort will close due to intolerability. Any subject who transfers to transplant prior to completion of two courses without experiencing an unacceptable toxicity is considered inevaluable for purposes of evaluating tolerability. Accrual will be halted for intolerability if there are two or more failures in tolerability among the first six subjects who are evaluable for tolerability. (NCT02433483)
Timeframe: At the end of therapy cycle 2 (approximately 2-3 months)

InterventionParticipants (Count of Participants)
Myeloid Malignancies0

Proportion of Participants Who Experience Therapeutic Success

"All patients will be counted towards this two-stage design. Therapeutic success for patients at time of enrollment is defined as:~Patients with fewer than 5% blasts, a ≥ 10-fold decrease in level of minimal residual disease after completion of 1 or 2 cycles of therapy.~Patients with greater than 5% in leukemic blasts in the marrow, achieving CR or CRi after completion of 1 or 2 cycles of therapy.~In terms of efficacy, patients who die before achieving therapeutic success will be counted as a failure, and all patients who receive ≥ 1 dose of protocol chemotherapy will be counted as a failure or success. Only subjects who withdraw or die prior to receiving the first dose of protocol chemotherapy will be considered inevaluable and replaced. The evaluation of tolerability and this phase II design will be performed concurrently, i.e., the first enrollees will be counted for both tolerability and efficacy." (NCT02433483)
Timeframe: At the end of therapy cycle 2 (approximately 2-3 months)

Interventionproportion (Number)
Myeloid Malignancies0

Time to Platelet Recovery

"The time to platelet recovery will be summarized using descriptive statistics. If there are no deaths prior to recovery of platelets, nonparametric confidence intervals for the median time to recovery will be computed by inverting the sign test. Otherwise, we will compute cumulative incidence curves to describe the time to platelet and neutrophil recovery while adjusting for competing events.~Due to the small number of patients enrolled, the data is presented by patient." (NCT02433483)
Timeframe: From start of therapy to completion of therapy (approximately 1 year)

Interventiondays (Number)
Myeloid Malignancies15

1-year Cumulative Incidence of Acute Graft Versus Host Disease (GVHD)

"Children's Oncology Group (COG) Stem Cell Committee Consensus Guidelines for Establishing Organ Stage and Overall Grade of Acute Graft Versus Host Disease (GVHD) were used. Overall clinical grade was based on the highest stage obtained:~Grade 0: no stage 1-4 of any organ~Grade I: stage 1-2 skin and no liver or gut involvement~Grade II: stage 3 skin, or stage 1 liver involvement, or stage 1 GI~Grade III: stage 0-3 skin, with stage 2-3 liver, or stage 2-3 GI~Grade IV: stage 4 skin, liver or GI involvement" (NCT02433483)
Timeframe: From start of therapy through completion of therapy (approximately 1 year)

InterventionParticipants (Count of Participants)
Stage 0Stage IStage IIStage IIIStage IV
Myeloid Malignancies00020

Percent Donor Chimerism

Percent donor chimerism in blood and bone marrow. (NCT02433483)
Timeframe: At weeks 1, 2, 3, and 4 after infusion of HPC-A

InterventionPercentage of donor chimerism (Mean)
Week 1Week 2Week 3Week 4
Myeloid Malignancies79.59999.5100

Complete Response Duration

The complete remission (CR) is the total number of patients who are in CR at one year divided by the total number of patients who received at least one dose of HCVAD with liposomal vincristine. CR was defined as the presence of 1x10^9/L neutrophils, >100x10^9/L platelets in the perpherial blood, and no extramedullary disease. Response date to loss of response or last follow up. Remission duration will be measured by the estimated median remission duration computed by Kaplan-Meier (K-M) analysis, which is the time point at which the cumulative remission duration drops below 50%, if present. If not present then median remission duration is not reached and not available (NA) as there are an insufficient number of participants with events. In either case ranges are provided for observed survival intervals used in the K-M analysis.. (NCT01319981)
Timeframe: Up to 7 years, 8 months

InterventionMonths (Median)
Hyper-CMAD + RituximabNA
Hyper-CMADNA

Number of Patients With Complete Remission at One Year

The complete remission (CR) is the total number of patients who are in CR at one year divided by the total number of patients who received at least one dose of HCVAD with liposomal vincristine. CR was defined as the presence of 1x10^9/L neutrophils, >100x10^9/L platelets in the perpherial blood, and no extramedullary disease. (NCT01319981)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Hyper-CMAD + Rituximab12
Hyper-CMAD13

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. Survival will be measured by the estimated median survival computed by Kaplan-Meier (K-M) analysis, which is the time point at which the cumulative survival drops below 50%, if present. If not present then the median Overall Survival is not reached and not available (NA) as there are an insufficient number of participants with events. In either case ranges are provided for observed survival intervals used in the K-M analysis. (NCT01319981)
Timeframe: Up to 7 years, 8 months

InterventionMonths (Median)
Hyper-CMAD + RituximabNA
Hyper-CMADNA

Complete Response (CR)

"Complete response (CR) was determined the number of participants who achieved CR by Day 42 after induction treatment. The outcome is reported as the total number of participants without dispersion.~• CR = less than 5% blasts; no blasts with auer rods; and no persistence of extramedullary disease, with blood count recovery to platelets ≥ 100,000/uL and ANC > 1000/uL, with transfusion independence." (NCT02019069)
Timeframe: Day 42

InterventionParticipants (Count of Participants)
Liposomal Cytarabine-daunorubicin CPX-3512

Complete Response With Incomplete Count Recovery (CRi)

"Complete response (CR) with incomplete count recovery (CRi) was determined as the number of participants who achieved CRi after induction therapy. The outcome is reported as the total number or participants without dispersion.~CR = less than 5% blasts; no blasts with auer rods; and no persistence of extramedullary disease, with blood count recovery to platelets ≥ 100,000/uL and ANC > 1000/uL, with transfusion independence.~CRi = all the parameters for CR, but platelets < 100,000/uL and/or ANC ≤ 1000/uL." (NCT02019069)
Timeframe: Day 42

InterventionParticipants (Count of Participants)
Liposomal Cytarabine-daunorubicin CPX-3511

Duration of Remission (DOR) Following Induction With CPX-351

"Duration of remission (DOR) was assessed as the length of time from documented complete response (CR) or complete response with incomplete count recovery (CRi) until documented lost of response, relapse, or death. The outcome is reported as the median with full range.~CR = less than 5% blasts; no blasts with auer rods; and no persistence of extramedullary disease, with blood count recovery to platelets ≥ 100,000/uL and ANC > 1000/uL, with transfusion independence.~CRi = all the parameters for CR, but platelets < 100,000/uL and/or ANC ≤ 1000/uL.~For patients remaining alive, duration of remission (DOR) is reported as the length of time from documented complete response (CR) or complete response with incomplete count recovery (CRi) until the most recent assessment." (NCT02019069)
Timeframe: Up to 1 year

Interventiondays (Median)
Liposomal Cytarabine-daunorubicin CPX-351185

Early Induction Mortality (Day 30 After 1st Induction)

Early induction mortality was assessed as the number of participants who died within 30 days of completing the 1st cycle of CPX-351 (1st induction). The outcome is reported as the number of participants without dispersion. (NCT02019069)
Timeframe: 30 days

InterventionParticipants (Count of Participants)
Liposomal Cytarabine-daunorubicin CPX-3512

Mortality at Day 60 After 1st Induction

Mortality at Day 60 after 1st induction was assessed as the number of participants who died within 60 days of completing the 1st cycle of CPX-351 (1st induction). The outcome is reported as the number of participants without dispersion. (NCT02019069)
Timeframe: 60 days

InterventionParticipants (Count of Participants)
Liposomal Cytarabine-daunorubicin CPX-3513

Overall Survival (OS)

Overall survival (OS) was assessed as the number of participants remaining alive 12 months, starting from date of entry into trial. The outcome is reported as the number of participants (without dispersion). (NCT02019069)
Timeframe: At 12 months

InterventionParticipants (Count of Participants)
Liposomal Cytarabine-daunorubicin CPX-3511

Participants Experiencing of Serious Adverse Events

Serious adverse events per participant were assessed as serious adverse events per 21CFR§312.32 that were Grade 3 or greater, and independent of relationship to CPX-351. The outcome is reported as the number of participants that experienced any defined SAE, a number without dispersion. (NCT02019069)
Timeframe: Up to 4 weeks after completion of treatment

InterventionParticipants (Count of Participants)
Liposomal Cytarabine-daunorubicin CPX-3515

Response Rate (RR)

"The response rate was determined as the sum of complete response calculated by adding the total complete response (CR) and complete response with incomplete count recovery (CRi). The outcome is reported as the total number without dispersion.~CR = less than 5% blasts; no blasts with auer rods; and no persistence of extramedullary disease, with blood count recovery to platelets ≥ 100,000/uL and ANC > 1000/uL, with transfusion independence.~CRi = all the parameters for CR, but platelets < 100,000/uL and/or ANC ≤ 1000/uL." (NCT02019069)
Timeframe: Day 42

InterventionParticipants (Count of Participants)
Liposomal Cytarabine-daunorubicin CPX-3513

Serious Adverse Events

Serious adverse events were assessed as serious adverse events per 21CFR§312.32 that were Grade 3 or greater, and independent of relationship to CPX-351. The outcome is reported as the total number of the defined SAEs, a number without dispersion. (NCT02019069)
Timeframe: Up to 4 weeks after completion of treatment

InterventionAdverse events (Number)
Liposomal Cytarabine-daunorubicin CPX-3518

Number of Participants With 1 Year Overall Survival

(NCT00345865)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
NHL With Irradiation139
HL Without Irradiation144
NHL - HIV Infected With Irradiation1
NHL - HIV Infected Without Irradiation5
NHL Without Radiation and Cyclophosphamide128

Number of Participants With 1 Year Progression Free Survival

"Progression is defined using the Response Criteria for Non-Hodgkin's Lymphoma given by NCI Sponsored International Working Group.The definition is as follows:~At least a 50% increase from nadir of any previously identified abnormal node. Appearance of any new lesion during or at the end of therapy." (NCT00345865)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
NHL With Irradiation112
HL Without Irradiation102
NHL - HIV Infected With Irradiation1
NHL - HIV Infected Without Irradiation2
NHL Without Radiation and Cyclophosphamide116

Number of Participants With 2 Years Overall Survival

(NCT00345865)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
NHL With Irradiation124
HL Without Irradiation140
NHL - HIV Infected With Irradiation1
NHL - HIV Infected Without Irradiation4
NHL Without Radiation and Cyclophosphamide121

Number of Participants With 2 Years Progression Free Survival

"Progression is determined using Response Criteria for Non-Hodgkin's Lymphoma given by NCI Sponsored International Working Group.~Definition is as follows:~At least a 50% increase from nadir of any previously identified abnormal node. Appearance of any new lesion during or at the end of therapy." (NCT00345865)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
NHL With Irradiation96
HL Without Irradiation91
NHL - HIV Infected With Irradiation1
NHL - HIV Infected Without Irradiation1
NHL Without Radiation and Cyclophosphamide106

Number of Participants With Hematopoietic Recovery After Transplantation

return to ANC (absolute neutrophil count) more than 500 cells/milliliter. (NCT00345865)
Timeframe: Day 42

InterventionParticipants (Count of Participants)
NHL With Irradiation171
HL Without Irradiation147
NHL - HIV Infected With Irradiation2
NHL - HIV Infected Without Irradiation5
NHL Without Radiation and Cyclophosphamide145

Number of Participants Who Survived

Number of Participants Who Survived at day 180. (NCT03019640)
Timeframe: From the time of transplant, assessed up to day 180

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, NK Infusion, Stem Cell Transplant)16

Treatment-related Mortality Within 30 Days (TRM30)

Participants that had Treatment-related mortality within 30 days. (NCT03019640)
Timeframe: Up to 30 days

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy, NK Infusion, Stem Cell Transplant)0

Maximum Tolerated Dose (MTD)

MTD is dose level where grade 3-4 sorafenib-attributable toxicity in <2 of 6 participants. Dose-Limiting Toxicity graded according to the NCI Common Toxicity Criteria version 3.0. (NCT00542971)
Timeframe: Twice a week for first two 28 day cycles

Interventionmilligrams/twice a day (BID) (Number)
Sorafenib + Idarubicin + Ara-C400

Number of Participants With Complete Response

Complete response was defined by the presence of < 5% blasts in the bone marrow (BM) with > 1 x 10^9/L platelets in the peripheral blood (PB). (NCT00542971)
Timeframe: Baseline to 2 years or disease progression.

InterventionParticipants (Number)
Complete ResponseNo Complete Response
Sorafenib + Idarubicin + Ara-C5421

Complete Response Rate

Complete response rate is defined as the percentage of patients who achieve bone marrow response (defined using the M bone marrow criteria for acute lymphoblastic leukemia (ALL); if M0 to M1 status (blast cells ,5%) was achieved by the end of induction or extended induction, the patient was considered a responder) at the end of induction therapy. (NCT00558519)
Timeframe: Up to 8 years post-registration

Interventionpercentage of patients (Number)
Treatment (Pediatric Regimen)89

Disease-free Survival

DFS was defined as time from bone marrow response in this study to the earliest occurrence of any of the following: failure to achieve bone marrow response (defined using the M bone marrow criteria for acute lymphoblastic leukemia (ALL); if M0 to M1 status (blast cells ,5%) was achieved by the end of induction or extended induction, the patient was considered a responder) by day 60, death, relapse at any site, or development of second malignant disease. (NCT00558519)
Timeframe: Up to 8 years post-registration

Interventionmonths (Median)
Treatment (Pediatric Regimen)81.7

Event-free Survival

EFS was defined as time from registration in this study to the earliest occurrence of any of the following: failure to achieve bone marrow response (defined using the M bone marrow criteria for acute lymphoblastic leukemia (ALL); if M0 to M1 status (blast cells ,5%) was achieved by the end of induction or extended induction, the patient was considered a responder) by day 60, death, relapse at any site, or development of second malignant disease. (NCT00558519)
Timeframe: Up to 8 years post-registration

Interventionmonths (Median)
Treatment (Pediatric Regimen)78.1

Number of Participants Who Experienced at Least One Grade 3 or Higher Adverse Event at Least Possibly Related to Treatment (Toxicity)

The number of participants who experienced toxicity (defined as at least one grade 3 or higher adverse event at least possibly related to treatment) is reported below. (NCT00558519)
Timeframe: Up to 10 years post-registration

InterventionParticipants (Count of Participants)
Treatment (Pediatric Regimen)286

Overall Survival

OS was defined from registration to death resulting from any cause. (NCT00558519)
Timeframe: Up to 8 years post-registration

Interventionmonths (Median)
Treatment (Pediatric Regimen)NA

Maximum Total Tolerated Dose (MTD) of Daily Combination Fludarabine 30 mg/m^2 and Cytarabine 500 mg/m^2 Among 3 Dose Levels (Dose Level 1: 2 Days, Dose Level 2: 3 Days or Dose Level 3: 4 Days)

Maximum dose levels for Phase I determined among three possible dose levels of Fludarabine and Cytarabine in combination with fixed doses of Oxaliplatin and Rituximab. Fludarabine and Cytarabine Dose Level 1: Days 2-3 (2 Days); Dose Level 2: Days 2-4 (3 Days); and Dose 3: Days 2-5 (4 Days). MTD is dose level at which less than 2/3 or 2/6 participants experience dose limiting toxicities (DLTs). The number of days of fludarabine and cytarabine administration increased simultaneously. Participants received a subsequent cycle of treatment with 1 additional day of fludarabine and cytarabine treatment no less than 4 weeks from the initiation of the previous cycle if no drug-related grade 3 or 4 non-hematologic life-threatening adverse events, and drug-related non-hematologic toxicity resolved to baseline or < grade 2. A maximum of 6 cycles were administered. (NCT00472849)
Timeframe: Up to 36 weeks (6 cycles each 4-6 weeks)

Interventionmg/m^2 (Number)
Fludarabine MTD (Total 3 Day Dose)Cytarabine MTD (Total 3 Day Dose)
OFAR (Phase I)901500

Overall Response: Number of Participants With Complete Remission, Nodular Partial Remission, and Partial Remission

Overall Response includes Complete remission (CR), nodular partial remission (nPR), and partial remission (PR) in high-risk, previously untreated participants with Chronic Lymphocytic Leukemia treated with CFAR using National Cancer Institute - Working Group response criteria. CR defined as zero nodes, Liver/spleen not palpable, zero symptoms, polymorphonuclear leukocyte (PMN)>1,500/uL, Platelets >100,000uL, Hemoglobin (untransfused) >11.0g/dL, Lymphocytes <4,000/uL and Bone Marrow Aspirate biopsy <30% lymphocytes with no lymphocyte infiltrate; PR defined as nodes >/= 50% decrease,Liver/spleen >/= 50% decrease, symptoms not applicable, PMN >1,500/uL or >50% improvement from baseline, Platelets 100,000uL or >/=50% decrease improvement from baseline, Hemoglobin (untransfused) >11.0g/dL or >50% improvement from baseline, Lymphocytes >50% decrease and Bone Marrow Aspirate biopsy Not Applicable for PR; with nPR defined same as PR but with <30% lymphocytes with residual disease on biopsy. (NCT00472849)
Timeframe: Up to 36 weeks (6 cycles each 4-6 weeks)

Interventionparticipants (Number)
Complete RemissionPartial RemissionNodular Partial Remission
OFAR MTD (Phase II)3279

Complete Response Rate to 1.3mg/m^2 of Bortezomib With Mitoxantrone and Etoposide in Phase II

"Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions.~The percentage of participants that experienced complete response will be reported." (NCT00410423)
Timeframe: Up to 7 years

Interventionpercentage of participants (Number)
Phase 2 - Bortezomib 1.3mg/m^227

Number of Participants With Dose Limiting Toxicity in Phase I

Dose limiting toxicity (DLT) is defined as grade-3 toxicity definitely related to bortezomib or grade-4 toxicity probably or definitely related to bortezomib. (NCT00410423)
Timeframe: 30-90 days

InterventionParticipants (Count of Participants)
Phase 1 - Bortezomib 0.7mg/m^20
Phase 1 - Bortezomib 1.0 mg/m^20
Phase 1 - Bortezomib 1.3mg/m^20

Disease-free Survival

Number of patients who were free of disease and alive at 1 year. (NCT00609739)
Timeframe: 1 year

InterventionParticipants (Number)
Cytarabine + Mitoxantrone0

Patients Who Relapsed

Number of patients whose disease relapsed. (NCT00609739)
Timeframe: 1 Year

Interventionparticipants (Number)
Cytarabine + Mitoxantrone1

Patients With Graft-Versus-Host-Disease

Number of patients who exhibited acute and/or chronic graft-versus-host disease. (NCT00609739)
Timeframe: Up to 30 Days Post Study Treatment

Interventionparticipants (Number)
Cytarabine + Mitoxantrone1

Patients With Regimen-Related Toxicity

Number of patients with adverse events related to treatment. (NCT00609739)
Timeframe: Up to 30 Days Post Study Treatment

Interventionparticipants (Number)
Cytarabine + Mitoxantrone0

Median Percentage of Treg Cells at 1 Year Post Transplant

The investigative intent is to determine the changes in numbers and function of the regulatory cell population using the best methods to measure this cell population. The frequency of T cells will be summarized at baseline and each time point of follow-up. (NCT00578461)
Timeframe: 1 Year

Interventionpercentage of total CD4+ cells (Median)
Stem Cell Transplant4.1

Median Percentage of Treg Cells at 1 Year Post Transplant

To define the biologic recovery and behavior of T regulatory cells for patients undergoing stem cell transplantation as specified in this protocol (NCT00578539)
Timeframe: 1 year

Interventionpercentage of total CD4+ cells (Median)
Stem Cell Transplant6.9

Disease-free Survival

Disease-free survival is defined as the length of time after treatment ends that the participant survives without any signs or symptoms of that cancer. (NCT00593645)
Timeframe: 5 years from time of restaging

Interventionparticipants (Number)
Arm 1: Non-myeloablative Conditioning Regimen0

Median Time to Progression

Time to progression is defined as the length of time from the start of treatment until the disease starts to get worse or spread to other parts of the body. (NCT00593645)
Timeframe: 5 years from time of restaging

Interventiondays (Median)
Arm 1: Non-myeloablative Conditioning Regimen152

Overall Survival

(NCT00593645)
Timeframe: 5 years from time of restaging

Interventiondays (Median)
Arm 1: Non-myeloablative Conditioning Regimen237

Rate of Acute Graft-versus-host Disease (GVHD)

Acute GVHD occurs within 100 days of transplant. (NCT00593645)
Timeframe: Up to 100 days after transplant

Interventionpercentage of participants (Number)
Arm 1: Non-myeloablative Conditioning Regimen0

Rate of Chronic Graft-versus-host Disease (GVHD)

(NCT00593645)
Timeframe: 100 days-1 year after transplant

Interventionpercentage of participants (Number)
Arm 1: Non-myeloablative Conditioning Regimen0

Engraftment as Measured by Percent Donor Chimerism

(NCT00593645)
Timeframe: Day +30

Interventionparticipants (Number)
Not done50% donor (bone marrow)100% donor (bone marrow)23.5% donor (FISH)
Arm 1: Non-myeloablative Conditioning Regimen2111

Engraftment as Measured by Percent Donor Chimerism

(NCT00593645)
Timeframe: Day +40-+60

Interventionparticipants (Number)
60% donor (bone marrow)70% donor (bone marrow)75% donor (bone marrow)100% donor (peripheral blood)
Arm 1: Non-myeloablative Conditioning Regimen1111

Engraftment as Measured by Percent Donor Chimerism

(NCT00593645)
Timeframe: Day +80-+90

Interventionparticipants (Number)
0% donor33% donor - myeloid (peripheral blood)67% donor (bone marrow)100% donor (bone marrow)
Arm 1: Non-myeloablative Conditioning Regimen1111

Inhibition of NF-kB Target Transcripts and/or Inhibition of Drug Efflux in at Least 50% of Patients

The clinical trial will be based on a sequential monitoring so that we will have a 90% confidence that choline magnesium trisalicylate (CMT) based modulation of NF-kB transcriptional targets and/or drug efflux occurs in at least 50% of patients. (NCT02144675)
Timeframe: 24 hours

InterventionParticipants (Count of Participants)
Arm I (Choline Magnesium Trisalicylate and Chemotherapy)13
Arm II (Chemotherapy)14

3-Year Event-Free Survival (EFS)

3-year EFS was calculated based on the participants with a complete response (CR). Study regimen considered successful if it exhibits a 3-year EFS rate greater than 60% and response rate no less than 90% with Grade III-IV infectious toxicity rate in induction no more than 33%. (NCT00866749)
Timeframe: 3 Years

InterventionParticipants (Count of Participants)
Augmented BFM Therapy68

Overall Survival

Overall Survival defined: Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT00866749)
Timeframe: Up to 12 years

InterventionMonths (Median)
Augmented BFM Therapy121

Participants With a Complete Response (CR)

Complete Response defined as: Bone Marrow blasts /= 100 and an Absolute Neutrophil Count (ANC) >/= 1000 (NCT00866749)
Timeframe: Up to 1 year

Interventionparticipants (Number)
Augmented BFM Therapy108

Participants Achieving Negative Minimal Residual Disease (MRD)

To evaluate the prognostic significance of minimal residual disease (MRD) in bone marrow samples of participants who achieved a complete response (CR) at the end of induction (day 29) and at the end of consolidation (day 84) in this group of patients. (NCT00866749)
Timeframe: up to 3 months

Interventionparticipants (Number)
Participants with CR and MRD negative on Day 29Participants with CR and MRD negative on day 84
Augmented BFM Therapy6087

Acute Graft Versus Host Disease Among Patients Who Received Early Transplant

Acute graft versus host disease (graded II, III, or IV) among patients who received early allogeneic HCT on study. (NCT02756572)
Timeframe: At day 100

InterventionParticipants (Count of Participants)
Received Allogeneic HCT on Study0

Demonstrate the Feasibility of Collecting Resource Utilization Data for Trial Participants

The amount of days of hospitalization (the major driver of costs within the first year) will be collected for resource utilization. (NCT02756572)
Timeframe: Within the first year of induction chemotherapy on study

Interventiondays (Median)
Treatment (Chemotherapy, HCT)49

Event-free Survival (EFS) Among Patients Who Received Early Transplant

Event-free survival among patients who received early allogeneic HCT on study as estimated with the Kaplan-Meier method. Events included death, relapse, and grade 3-4 acute graft vs host disease. (NCT02756572)
Timeframe: Up to 100 days post-transplant

InterventionPercentage of participants (Number)
Received Allogeneic HCT on Study91

Event-free Survival (EFS) Among Patients Who Received Early Transplant

Event-free survival among patients who received early allogeneic HCT on study as estimated with the Kaplan-Meier method. Events included death, relapse, and grade 3-4 acute graft vs host disease. (NCT02756572)
Timeframe: Up to 6 months post-transplant

InterventionPercentage of participants (Number)
Received Allogeneic HCT on Study82

Factors That Distinguish Patients Who Received Early Transplant From Those Who Did Not - AGE

Patients who receive early transplant will be compared to those that don't using the Wilcoxon rank sum test for the quantitative variable of age. (NCT02756572)
Timeframe: From time of subject's study enrollment to time of subject's feasibility success assessment (i.e. when subject received transplant within 60 days or when it was determined subject would not proceed with transplant on study)

Interventionyears (Median)
Received Allogeneic HCT on Study55
Did Not Receive Allogeneic HCT on Study57

Factors That Distinguish Patients Who Received Early Transplant From Those Who Did Not - TREATMENT RELATED MORTALITY

"Patients who receive early transplant will be compared to those that don't using the Wilcoxon rank sum test for the quantitative variable of treatment-related mortality (TRM). This outcome measure is intended to report a predicted TRM score assessed at the time of feasibility evaluation. The TRM score is used to measure treatment-related mortality, or likelihood of death within 28 days of initiation of induction chemotherapy for patients with AML. The score is normalized from 0 to 100, so that a score of 0 demonstrates the patient has a very low likelihood of TRM and a score of 100 a very high likelihood of death. A calculation is used to predict TRM using age, performance status, if they have secondary AML, albumin, creatinine, platelet count, white blood cell count, and peripheral blood blast percentage. The higher the TRM score, the higher the risk of TRM. Calculator and table of relationship between TRM score and TRM probability found here: https://trmcalculator.fredhutch.org/." (NCT02756572)
Timeframe: From time of subject's study enrollment to time of subject's feasibility success assessment (i.e. when subject received transplant within 60 days or when it was determined subject would not proceed with transplant on study)

Interventionunits on a scale (Median)
Received Allogeneic HCT on Study2.15
Did Not Receive Allogeneic HCT on Study3.045

Overall Survival (OS) Among Patients Who Did Not Receive Early Transplant

Overall survival among patients who did not receive early allogeneic HCT on study as estimated with the Kaplan-Meier method. (NCT02756572)
Timeframe: Up to 100 days after induction day 1

InterventionPercentage of participants (Number)
Did Not Receive Allogeneic HCT on Study75

Overall Survival (OS) Among Patients Who Did Not Receive Early Transplant

Overall survival among patients who did not receive early allogeneic HCT on study as estimated with the Kaplan-Meier method. (NCT02756572)
Timeframe: Up to 6 months after induction day 1

InterventionPercentage of participants (Number)
Did Not Receive Allogeneic HCT on Study62

Overall Survival (OS) Among Patients Who Received Early Transplant.

Overall survival among patients who received early allogeneic HCT on study as estimated with the Kaplan-Meier method. (NCT02756572)
Timeframe: Up to 100 days post-transplant

InterventionPercentage of participants (Number)
Received Allogeneic HCT on Study91

Overall Survival (OS) Among Patients Who Received Early Transplant.

Overall survival among patients who received early allogeneic HCT on study as estimated with the Kaplan-Meier method. (NCT02756572)
Timeframe: Up to 6 months post-transplant

InterventionPercentage of participants (Number)
Received Allogeneic HCT on Study82

Relapse-free Survival (RFS) Among Patients Who Received Early Transplant

Relapse-free survival among patients who received early allogeneic HCT on study as estimated with the Kaplan-Meier method. (NCT02756572)
Timeframe: Up to 100 days post-transplant

InterventionPercentage of participants (Number)
Received Allogeneic HCT on Study91

Relapse-free Survival (RFS) Among Patients Who Received Early Transplant

Relapse-free survival among patients who received early allogeneic HCT on study as estimated with the Kaplan-Meier method. (NCT02756572)
Timeframe: Up to 6 months post-transplant

InterventionPercentage of participants (Number)
Received Allogeneic HCT on Study82

Treatment Related Mortality Among Patients Who Received Early Transplant vs Patients Who Did Not Receive Early Transplant

Treatment related mortality calculated among patients who received early allogeneic HCT on study vs patients who did not receive early transplant on study. (NCT02756572)
Timeframe: At day 100

InterventionPercentage of participants (Number)
Received Allogeneic HCT on Study9
Did Not Receive Allogeneic HCT on Study23.8

Demonstrate the Feasibility of Collecting Patient-reported Outcomes for Trial Participants

The amount of returned patient-reported outcome questionnaires will be summarized for each collection timepoint using percent collection from surviving patients for PRO timepoints. (NCT02756572)
Timeframe: Up to 12 months post-HCT

InterventionParticipants (Count of Participants)
Enrollment PROs returnedPost G-CLAM PROs returnedPre-HCT PROs returned6 months post-HCT PROs returned12 months post-HCT PROs returned
Treatment (Chemotherapy, HCT)2723843

Factors That Distinguish Patients Who Received Early Transplant From Those Who Did Not - GENDER

Patients who receive early transplant will be compared to those that don't using the Fisher's exact test for the categorical variables of gender. (NCT02756572)
Timeframe: From time of subject's study enrollment to time of subject's feasibility success assessment (i.e. when subject received transplant within 60 days or when it was determined subject would not proceed with transplant on study)

,
InterventionParticipants (Count of Participants)
FemaleMale
Did Not Receive Allogeneic HCT on Study1011
Received Allogeneic HCT on Study81

Feasibility of Early Allogeneic Hematopoietic Cell Transplant Assessed by Enrollment and Incidence of Early Transplant

Success will be defined as enrollment of at least 30 patients with transplants occurring in 15 of these 30 (50%) within 60 days of enrollment or start of induction chemotherapy with G-CLAM, whichever is earlier. (NCT02756572)
Timeframe: Up to 60 days after start of chemotherapy

InterventionParticipants (Count of Participants)
Received allogeneic HCT on study within 60 days (feasibility success)Did not receive allogeneic HCT on study within 60 days (feasibility failure)
Treatment (Chemotherapy, HCT)921

Feasibility of Early Allogeneic Hematopoietic Cell Transplant Assessed by Relapsed-free Survival 6 Months After Transplant

Success will be defined as observing a 40% of higher 6-month relapse-free survival among patients who received early transplant on study. (NCT02756572)
Timeframe: 6 months after early allogeneic HCT on study

InterventionParticipants (Count of Participants)
No relapse within 6 months post-HCT (feasibility success)Relapse within 6 months post-HCT (feasibility failure)
Received Early Allogeneic HCT on Study62

Response Assessments After Early Allogeneic Hematopoietic Cell Transplant, Day 28

"Complete Remission (CR), defined as <5% blasts in bone marrow with hematologic recovery (ANC>1000/ul and platelets >100,000/ml).~CRi, defined as complete remission with insufficient hematologic recovery (ANC<1000/ul or platelets<100,000/ul).~CRp, defined as complete remission but platelets <100,000/ul. Minimal residual disease (MRD), defined as any detectable disease by flow cytometry, cytogenetics, FISH or PCR in a patient otherwise fulfilling remission criteria.~Morphologic leukemia-free state (MLFS), defined as <5% blasts in bone marrow with no hematologic recovery.~Relapse, defined as >5% blasts in bone marrow, flow cytometry, or manual differential; OR treatment for active relapsed disease." (NCT02756572)
Timeframe: Approximately 28 days after early allogeneic HCT

InterventionParticipants (Count of Participants)
CR with MRDCR without MRDCRi with MRDCRi without MRDMLFS with MRDMLFS without MRDRelapse
Received Allogeneic HCT on Study0701000

Response Assessments After Early Allogeneic Hematopoietic Cell Transplant, Day 84

"Complete Remission (CR), defined as <5% blasts in bone marrow with hematologic recovery (ANC>1000/ul and platelets >100,000/ml).~CRi, defined as complete remission with insufficient hematologic recovery (ANC<1000/ul or platelets<100,000/ul).~CRp, defined as complete remission but platelets <100,000/ul. Minimal residual disease (MRD), defined as any detectable disease by flow cytometry, cytogenetics, FISH or PCR in a patient otherwise fulfilling remission criteria.~Morphologic leukemia-free state (MLFS), defined as <5% blasts in bone marrow with no hematologic recovery.~Relapse, defined as >5% blasts in bone marrow, flow cytometry, or manual differential; OR treatment for active relapsed disease." (NCT02756572)
Timeframe: Approximately 84 days after early allogeneic HCT

InterventionParticipants (Count of Participants)
CR with MRDCR without MRDCRi with MRDCRi without MRDMLFS with MRDMLFS without MRDRelapse
Received Allogeneic HCT on Study0402002

Phase I - Dose Level With Best Kinetics of Platelet Count Recovery

To describe the kinetics of platelet count recovery in acute myeloid leukemia patients in complete remission receiving intensive consolidation chemotherapy who will be receiving eltrombopag. This is assessed graphically by plotting platelet count vs. days relative to start of cytarabine for each patient. (NCT01656252)
Timeframe: 13 months

Interventionmg (Number)
Phase I- Cytarabine & Eltrombopag150

Phase I- Optimal Tolerated Dose of Eltrombopag

To determine the safety, tolerability and optimal dose of eltrombopag in acute myeloid leukemia patients in complete remission receiving intensive consolidation chemotherapy. The optimal dose was based on rules involving observation of Dose Limiting Toxicity (DLT events), defined as a CTCAE Version 4 non-hematologic adverse event of grade 3 or higher occurring within 30 days of the last dose of eltrombopag judged by the investigator to be at least possibly related to eltrombopag administration. (NCT01656252)
Timeframe: 13 months

Interventionmg (Number)
Phase I- Cytarabine & Eltrombopag300

Response Rate(CR/CRi) Rate

For descriptive purposes, the CR/CRi (complete response/Complete response with incomplete blood count recovery) rate will be reported at the end of the study separately for Arm A and Arm B. Responses are following definitions consistent with those published by Dohner H, Estey EH, Amadori S, et al. CR is defined as Bone marrow showing less than 5% myeloblasts with normal maturation of all cell lines, an ANC of at least 1000/μL and a platelet count of 100,000/μL, absence of blasts in peripheral blood, absence of identifiable leukemic cells in the bone marrow, clearance of disease-associated cytogenetic abnormalities, and clearance of any previously existing extramedullary disease. CRi: All CR criteria except for residual neutropenia (ANC < 1000/μL) (NCT01870596)
Timeframe: Up to 3 years

Interventionparticipants (Number)
Arm A (Cytarabine, Chk1 Inhibitor SCH 900776)6
Arm B (Cytarabine)9

Measure Patient Response to High-dose Cytarabine Followed by Clofarabine in Adult Patients With Relapsed or Refractory AML

Response to the therapy is measured by a defined improvement in Neutrophil and platlet counts, along with improved cellularity of bone marrow biopsy (>20% with maturation of all cell lines), <5% blasts, auer rods must not be detectable and extramedullary leukemia or soft tissue involvment must not be present. (NCT01656031)
Timeframe: 5 weeks

Interventionparticipants (Number)
High-Dose Cytarabine and Clofarabine17

Disease-free Survival

Number of participants who survived and were disease-free at 5 years (NCT00602225)
Timeframe: At five years after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Arm I: Filgrastim + Clofarabine + Cytarabine (GCLAC)11

Dose-limiting Toxicity as Assessed by NCI CTCAE v3.0

(NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Arm I: Filgrastim + Clofarabine + Cytarabine (GCLAC)2

Efficacy

Number of Patients Surviving at Five Years (NCT00602225)
Timeframe: At five years after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Arm I: Filgrastim + Clofarabine + Cytarabine (GCLAC)12

Maximum Tolerated Dose of Clofarabine

(NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

Interventionmg/m^2 of clofarabine (Number)
Arm I: Filgrastim + Clofarabine + Cytarabine (GCLAC)25

Overall Survival

(NCT00602225)
Timeframe: At five years after the last dose of clofarabine

Interventionmonths (Median)
Arm I9

Response Rates by Cytogenetic Risk Category and Clofarabine Dose

Number of participants under each Cytogenetic Risk Category and Clofarabine dose who achieve CR (Complete Remission = less than 5% blasts in the marrow and count recovery of Absolute Neutrophil Count to 1,000/microL and Platelet Count to 100,000/microL) or CRp (Complete Remission, but with a platelet count of less than 100,000/microL). (NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Favorable Risk + 25 mg/m^2 achieve CRIntermediate Risk + 15 mg/m^2 achieve CRIntermediate Risk + 20 mg/m^2 achieve CRIntermediate Risk + 25 mg/m^2 achieve CRIntermediate Risk + 25 mg/m^2 achieve CRpUnfavorable Risk + 15 mg/m^2 achieve CRUnfavorable Risk + 20 mg/m^2 achieve CRUnfavorable Risk + 25 mg/m^2 achieve CRUnfavorable Risk + 25 mg/mg^2 achieve CRp
Arm I223542163

Response Rates by Cytogenetic Risk Category

Number of participants who achieved Complete Remission (less than 5% blasts in the marrow and count recovery of Absolute Neutrophil Count to 1,000/microL and Platelet Count to 100,000/microL) under each cytogenetic risk category. (NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Favorable risk Complete RemissionIntermediate risk Complete remissionUnfavorable risk Complete remission
Arm I3109

Response Rates by Duration First Complete Remission (CR1)

Number of participants whose first Complete Remission lasted 0, 1-6, 6-12, or greater than 12 months. Only those participant who had a first CR are included in this data. (NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Duration CR1 (months): 0Duration CR1 (months): 1-6Duration CR1 (months): 6-12Duration CR1 (months): greater than 12
Arm I12423

Response Rates by Salvage Number

Number of participants in each Salvage number category who achieved a Complete Remission. Salvage number refers to whether treatment with GCLAC on this study was the pariticipant's first salvage regimen (1), second salvage regimen (2), or third or greater salvage regimen (3 or greater). (NCT00602225)
Timeframe: 45 days after the last dose of clofarabine

InterventionParticipants (Count of Participants)
Salvage number 1Salvage number 2Salvage number 3 or greater
Arm I1650

Complete Remission (CR) Rate at the End of Induction in Participants With Newly Diagnosed FLT3-ITD (+) Acute Myeloid Leukemia

Complete remission (CR) rate is defined as the percentage of participants achieving CR, defined as a disappearance of all target lesions, after Induction. (NCT02668653)
Timeframe: Approximately Cycle 1 Day 21 (Induction) to end of Induction, up to approximately 120 days (each Induction cycle is up to 60 days)

InterventionParticipants (Count of Participants)
Quizartinib147
Placebo150

Composite CR Rate at the End of Induction in Participants With Newly Diagnosed FLT3-ITD (+) Acute Myeloid Leukemia

Composite complete remission (CRc) rate is defined as the percentage of participants whose best response is complete remission (CR), defined as a disappearance of all target lesions, or CR with incomplete neutrophil or platelet recovery (CRi) at the end of first Induction cycle. (NCT02668653)
Timeframe: Approximately Cycle 1 Day 21 (Induction) to end of Induction, up to approximately 120 days (each Induction cycle is up to 60 days)

InterventionParticipants (Count of Participants)
Quizartinib192
Placebo176

Event-free Survival in Participants With Newly Diagnosed FLT3-ITD (+) Acute Myeloid Leukemia

Event-free survival (EFS) is the time from randomization to the earliest date of either refractory disease (or treatment failure [TF]), relapse, or death from any cause. Refractory disease is defined as complete remission never achieved during Induction (CR: >1000 neutrophils, >100,000 platelets, <5% blasts, and other [defined as absence of extramedullary disease [EMD], blasts with rods, and leukemic blasts]). For refractory disease, EFS event date is Day 1 (randomization). Relapse after CR is defined as ≥5% blasts, leukemic blasts, extramedullary leukemia, and presence of rods. This analysis is based on a response assessment with TF defined as not achieving response of CR, using a 42- day window from the start of the last cycle in Induction for CR evaluation. (NCT02668653)
Timeframe: Date of randomization to the date of refractory disease, relapse, or death, up to approximately 3 years after enrollment

Interventionmonths (Median)
Quizartinib0.03
Placebo0.71

Overall Survival in Participants With Newly Diagnosed FLT3-ITD (+) Acute Myeloid Leukemia

Overall survival is defined as the time from randomization until death from any cause. (NCT02668653)
Timeframe: Date of randomization to the date of death due to any cause, up to approximately 3 years after enrollment

Interventionmonths (Median)
Quizartinib31.9
Placebo15.1

Number of Participants With Treatment-emergent Adverse Events Occurring in ≥10% Participants With Newly Diagnosed FLT3-ITD (+) Acute Myeloid Leukemia

A treatment-emergent adverse event (TEAE) is defined as an adverse event that occur, having been absent before first dose of quizartinib or placebo, or have worsened in severity after initiating quizartinib or placebo. Adverse events collected more than 30 days after the last dose of quizartinib/placebo will not be considered TEAEs unless they are considered drug-related. (NCT02668653)
Timeframe: Date of first dose up to 30 days after last dose, up to 36 cycles following continuation (approximately 6 years 11 months, each cycle is 28 days)

,
InterventionParticipants (Count of Participants)
Any TEAEGastrointestinal disordersDiarrhoeaNauseaVomitingStomatitisConstipationAbdominal painDyspepsiaAbdominal pain upperInfections and infestationsPneumoniaSepsisGeneral disorders and administration site disordersPyrexiaOedema peripheralFatigueBlood and lymphatic system disordersFebrile neutropeniaNeutropeniaThrombocytopeniaAnaemiaMetabolism and nutrition disordersHypokalaemiaDecreased appetiteHypomagnesaemiaHypophosphataemiaHypocalcaemiaSkin and subcutaneous tissue disordersRashPruritusInvestigationsAlanine aminotransferase increasedElectrocardiogram QT prolongedAspartate aminotransferase increasedNeutrophil count decreasedRespiratory, thoracic, and mediastinal disordersCoughEpistaxisOropharyngeal painNervous system disordersHeadacheMusculoskeletal and connective tissue disordersArthralgiaBack painVascular disordersHypertensionPsychiatric disordersInsomnia
Placebo26520994845356693823251884128173109372314311327301915396363024291586640105271119121154429189753108352870335030
Quizartinib26421598906557564630292043915177112302916811754302916593463027261526935140423628271235040271037391291971295737

Maximum Tolerated Dose of Dasatinib (Phase I)

Maximum tolerated dose of dasatinib, determined according to incidence of dose limiting toxicity (DLT), graded using the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (Phase I) (NCT01876953)
Timeframe: From the first dose of Dasatinib through the DLT observation period (Day +28)

Interventionmg/m2 (Number)
Dasatinib 100mg/Day + Cytarabine 200mg/m2/Day + Idarubicin 12mg/m2/Day100

CRi Defined as Meeting All Criteria for a Morphologic CR But ANC Remains Less Than 1,000/μL and/or Platelet Count Less Than 100,000/μL

(NCT01607645)
Timeframe: Assessed for up to 5 years

InterventionParticipants (Count of Participants)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)0
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)0

CRiMRD+ Defined as Meeting All Criteria for a CRi But With Evidence of Minimal Residual Disease by Flow Cytometry, Cytogenetics, or Other Known Molecular Biomarkers

(NCT01607645)
Timeframe: Assessed for up to 5 years

InterventionParticipants (Count of Participants)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)1
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)0

CRMRD- Defined as Morphologic CR Without Evidence of Minimal Residual Disease by Flow Cytometry, Cytogenetics, or Other Known Molecular Biomarkers

(NCT01607645)
Timeframe: Assessed for up to 5 years

InterventionParticipants (Count of Participants)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)2
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)0

CRMRD+ Defined as a Morphologic CR But With Minimal Residual Disease by Flow Cytometry, Cytogenetics, or Other Known Molecular Biomarkers

(NCT01607645)
Timeframe: Assessed for up to 5 years

InterventionParticipants (Count of Participants)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)0
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)0

Cytogenetic Response Defined as no Detectable Cytogenetic Abnormality in a Subsequent BM Specimen After Induction or Re-induction

(NCT01607645)
Timeframe: Assessed for up to 5 years

InterventionParticipants (Count of Participants)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)3
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)0

Duration of Moderate Neutropenia Defined as an ANC Less Than 1000

(NCT01607645)
Timeframe: Assessed for up to 5 years

Interventiondays (Mean)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)67

Duration of Severe Neutropenia Defined as an ANC Less Than 500

(NCT01607645)
Timeframe: Assessed for up to 5 years

Interventiondays (Mean)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)65

Duration of Thrombocytopenia Defined as Platelet Count Less Than 100,000

(NCT01607645)
Timeframe: Assessed for up to 5 years

Interventiondays (Mean)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)51

Resistant Disease Defined as Patient Survives at Least 14 Days After Completion of the Last Dose of Induction or Re-induction But Has Persistent Leukemia in Peripheral Blood (PB) or BM

(NCT01607645)
Timeframe: Assessed for up to 90 days

InterventionParticipants (Count of Participants)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)1
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)3

Severe Prolonged Aplasia

(NCT01607645)
Timeframe: Assessed for up to 45 days

InterventionParticipants (Count of Participants)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)0
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)0

TRM With Each Course of Decitabine-priming, Idarubicin, and Cytarabine

(NCT01607645)
Timeframe: Assessed for up to Day 30

InterventionParticipants (Count of Participants)
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)0
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)0

Frequency and Severity of Grade 3, 4, and 5 Toxicities With Each Course of Decitabine-priming, Idarubicin, and Cytarabine According to NCI Common Terminology Criteria for Adverse Events Version (CTCAE) 4.0

(NCT01607645)
Timeframe: Assessed for up to 3 months after completion study treatment

,
InterventionParticipants (Count of Participants)
CYCLE 1 : Infection Grade 3CYCLE 1 : Infection Grade 4CYCLE 1 : Hepatobiliary Grade 3CYCLE 1 : Blood and Lymphatic Grade 3CYCLE 1 : Gastrointenstinal Grade 3CYCLE 2 : Infection Grade 3CYCLE 2 : Infection Grade 4CYCLE 2 : Hepatobiliary Grade 3CYCLE 2 : Blood and Lymphatic Grade 3CYCLE 2 : Gastrointenstinal Grade 3
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)2003000011
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)0111000000

Number of Participants Who Achieved Morphologic CR

Morphologic complete remission (CR): Absolute Neutrophil Count (ANC)≥1,000/uL, platelet count ≥100,000/uL, <5% Bone Marrow (BM) blasts, no Auer rods (cytoplasmic inclusions which result from an abnormal fusion of the primary (azurophilic) granules), no morphologic dysplasia, and no evidence of extramedullary disease (NCT01607645)
Timeframe: Participants were monitored up until the point when they went off study following completion of the treatment (3 months)

,
Interventionparticipants (Number)
CRCRi-MRD (Minimal Residual Disease)Refractory
Arm I (Decitabine Day -4 to Day 0), Idarubicin, Cytarabine)211
Arm II (Decitabine (Day -9 to Day -5), Idarubicin, Cytarabine)003

3-year Event-free Survival Rates in Patients With Relapsed ALL

Estimate the 3-year event-free survival rate of participants with first relapse or primary refractory precursor B-cell ALL treated with risk-directed therapy. (NCT01700946)
Timeframe: 3 years of follow-up since the on-study date

Interventionpercentage of participants (Number)
STANDARD RISK83.3
HIGH RISK55.7
All Patients Enrolled on the Study67.83

3-year Overall Survival Rate of Patients With Relapsed ALL

Estimate the 3-year survival rate of participants with first relapse or primary refractory precursor B-cell ALL treated with risk-directed therapy. (NCT01700946)
Timeframe: 3 years of follow-up since the on-study date

Interventionpercentage of participants (Number)
STANDARD RISK94.4
HIGH RISK55.5
All Patients Enrolled on the Study72.63

Mean of CD20 Expression Levels

To estimate mean levels of CD20 expression at baseline, during treatment with dexamethasone-containing chemotherapy and following rituximab treatment in Block I of remission induction therapy for relapsed precursor B-cell ALL. (NCT01700946)
Timeframe: Baseline and at the end of Block I (approximately 5 weeks after the on-study date)

,,
Interventionpercentage of CD20 Antigen (Mean)
At BaselineAt Block I
All Patients Enrolled on the Study36.2319.43
HIGH RISK39.8220.10
STANDARD RISK31.1018.54

Median CD20 Expression Levels

To estimate median levels of CD20 expression at baseline, during treatment with dexamethasone-containing chemotherapy and following rituximab treatment in Block I of remission induction therapy for relapsed precursor B-cell ALL. (NCT01700946)
Timeframe: Baseline and at the end of Block I (approximately 5 weeks after the on-study date)

,,
Interventionpercentage of CD20 Antigen (Median)
At BaselineAt Block 1
All Patients Enrolled on the Study22.015.58
HIGH RISK23.1319.58
STANDARD RISK16.4011.83

Cycle 2: Daunorubicin Dose-normalized Plasma: AUC(0-24)

Cycle 2 Daunorubicin AUC(0-24). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 2 Day 1 to Day 2 (0 to 24 post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD10.315
Placebo QD8.1146

Cycle 2: Daunorubicin Dose-normalized Plasma: Cmax

Cycle 2 Daunorubicin Cmax. PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 2 Day 1 to Day 2 (0 to 24 post-dose)

Intervention(ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD11.141
Placebo QD3.8905

Cycle 2: Daunorubicinol Dose-normalized Plasma: AUC(0-24)

Cycle 2 Daunorubicinol AUC(0-24). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 2 Day 1 to Day 2 (0 to 24 post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD34.067
Placebo QD30.820

Cycle 2: Daunorubicinol Dose-normalized Plasma: Cmax

Cycle 2 Daunorubicinol Cmax. PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 2 Day 1 to Day 2 (0 to 24 post-dose)

Intervention(ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD4.0200
Placebo QD1.9868

Daunorubicin Dose-normalized Plasma: AUC(0-∞)

Daunorubicin AUC(0-∞). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 3 to Day 9 (0 to 144 hrs post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD8.0807
Placebo QD8.7880

Daunorubicin Dose-normalized Plasma: AUC(0-t)

Daunorubicin AUC(0-t). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 3 to Day 9 (0 to 144 hrs post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD7.9523
Placebo QD8.6723

Daunorubicin Dose-normalized Plasma: AUC(24-∞)

Daunorubicin AUC(24-∞). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 4 to Day 9 (24 to 144 hrs post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD0.87496
Placebo QD0.72315

Daunorubicin Dose-normalized Plasma: AUC(24-t)

Daunorubicin AUC(24-t). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 4 to Day 9 (24 to 144 hrs post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD0.76524
Placebo QD0.59660

Daunorubicin Dose-normalized Plasma: Cmax

Daunorubicin Cmax. PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 3 to Day 9 (0 to 144 hrs post-dose)

Intervention(ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD5.1527
Placebo QD6.4113

Daunorubicinol Dose-normalized Plasma: AUC(0-∞)

Daunorubicinol AUC(0-∞). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 3 to Day 9 (0 to 144 hrs post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD63.997
Placebo QD62.835

Daunorubicinol Dose-normalized Plasma: AUC(0-t)

daunorubicinol AUC(0-t). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 3 to Day 9 (0 to 144 hrs post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD62.463
Placebo QD61.608

Daunorubicinol Dose-normalized Plasma: AUC(24-∞)

Daunorubicinol AUC(24-∞). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 4 to Day 9 (24 to 144 hrs post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD24.537
Placebo QD23.039

Daunorubicinol Dose-normalized Plasma: AUC(24-t)

Daunorubicinol AUC(24-t). PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 4 to Day 9 (24 to 144 hrs post-dose)

Intervention(h*ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD22.963
Placebo QD21.821

Daunorubicinol Dose-normalized Plasma: Cmax

Daunorubicinol Cmax. PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 3 to Day 9 (0 to 144 hrs post-dose)

Intervention(ug/ml)/(mg/m2) (Geometric Mean)
Eltrombopag (ELQ) QD3.5770
Placebo QD3.3640

Maximum Duration (Days) of Platelet Transfusion Independence

Maximum time period (in days) during which the patient did not receive any platelet transfusion (NCT01890746)
Timeframe: At differnt time points from start of treatment and up to end of study year 2 assessment

InterventionDays (Median)
Eltrombopag (ELQ) QD29.0
Placebo QD29.5

Number of Participants Who Achieved Platelet Count Recovery by Day 21

Number of participants with platelet counts 20 Gi/L for 3 consecutive days, unaided by transfusions, in patients with < 20 Gi/L after chemotherapy. (NCT01890746)
Timeframe: By Day 21

InterventionCount of participants (Number)
Eltrombopag (ELQ) QD4
Placebo QD7

Number of Participants With Liver Events.

The number of participants with liver enzyme (ALT, AST, ALP, Total bilirubin) abnormalities while receiving study treatment in each arm are presented. (NCT01890746)
Timeframe: 8 weeks

InterventionParticipants (Number)
Eltrombopag (ELQ) QD2
Placebo QD6

Number of Platelet Transfusions Per Week Within Cycles

This was the average number of platelet transfusions per week within cycles. (NCT01890746)
Timeframe: Post-Base line up to Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

InterventionPlatelet transfusions per week (Median)
Eltrombopag (ELQ) QD1.5
Placebo QD1.4

Overall Survival (OS)

Overall survival defined as the time form randomization until the date of death due to any cause. (NCT01890746)
Timeframe: From randomization to end of 2-year follow-up

InterventionCount of participants (Number)
Eltrombopag (ELQ) QD39
Placebo QD30

Percentage of Patients Who Achieved Platelet Transfusion Independence ≥ 28 Days

Percentage of patients who achieved platelet transfusion independence ≥ 28 days. (NCT01890746)
Timeframe: From start of treatment and up to end of study year 2 assessment

InterventionPercentage of participants (Number)
Eltrombopag (ELQ) QD55
Placebo QD53

Plasma Pharmacokinetics (PK) Parameter of Daunorubicin: Half-life (t1/2)

Daunorubicin half-life. PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 3 to Day 9 (0 to 144 hrs post-dose)

Interventionhour (h) (Geometric Mean)
Eltrombopag (ELQ) QD15.754
Placebo QD13.709

Plasma Pharmacokinetics (PK) Parameter of Daunorubicinol: Half-life (t1/2)

Daunorubicinol half-life. PK analyses used actual relative time and actual dosing information in mg/m2. All parameter values were divided by daunorubicin dose in mg/m2 except t1/2. (NCT01890746)
Timeframe: Cycle 1 Day 3 to Day 9 (0 to 144 hrs post-dose)

Interventionhour (h) (Geometric Mean)
Eltrombopag (ELQ) QD22.735
Placebo QD21.603

Time to Neutrophil Engraftment

Time to absolute neutrophil count (ANC) >= 0.5 Gi/L for 3 consecutive days in participants with ANC < 0.5 Gi/L after chemotherapy (NCT01890746)
Timeframe: At different time points from last dose of chemotherapy up to end of study year 2 assessment

InterventionMonths (Median)
Eltrombopag (ELQ) QDNA
Placebo QDNA

Time to Platelet Count Recovery >=20 Gi/L

Time to Platelet counts >= 20 Gi/L for 3 consecutive days, unaided by transfusions in patients with < 20 Gi/L after chemotherapy. For this endpoint, the event required platelet count to be >= 20 Gi/L for 3 consecutive days. Hematology was assessed daily during hospital stay but only weekly after hospital discharge and thus, platelet count was not always available for 3 consecutive days to confirm the achievement of platelet count recovery. (NCT01890746)
Timeframe: From last dose of chemotherapy to up to end of study year 2 assessment

InterventionMonths (Median)
Eltrombopag (ELQ) QDNA
Placebo QDNA

Time to Platelet Recovery >=100 Gi/L

Time to platelet counts >= 100 Gi/L unaided by transfusions in participants with < 100 Gi/L after chemotherapy. (NCT01890746)
Timeframe: From last dose of chemotherapy to up to end of study year 2 assessment

InterventionMonths (Median)
Eltrombopag (ELQ) QD0.69
Placebo QD0.69

Change From Baseline in the Left Ventricular Ejection Fraction (LVEF).

LVEF is a measurement of the percentage of blood leaving heart each time it contracts. LVEF was assessed by an echocardiogram (ECHO) or Multiple Gated Acquisition scan (MUGA). Baseline was defined as the most recent, non-missing value prior to or on the first study treatment dose date. Change from Baseline was calculated as the Day 42 value minus the Baseline value. (NCT01890746)
Timeframe: Baseline and Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
InterventionLVEF percent (Mean)
chnge from baseline (BL) to end of studychange from BL to worse post-BL case
Eltrombopag (ELQ) QD-2.5-4.1
Placebo QD-4.3-5.7

Incidence of Hemorrhagic Events

Incidence of bleeding events using WHO bleeding grade (G0=No bleeding, G1=Petechiae, G2=Mild blood loss, G3=Gross blood loss, G4=Debilitating blood loss) by week and cycle (NCT01890746)
Timeframe: Baseline, weekly within induction and re-induction cycles, end of therapy

InterventionParticipants (Number)
C1D7 - GRADE 0C1D7 - GRADE 1C1D7 - GRADE 2C1D7 - GRADE 3C1D14 - GRADE 0C1D14 - GRADE 1C1D14 - GRADE 2C1D14 - GRADE 3C1D21 - GRADE 0C1D21 - GRADE 1C1D21 - GRADE 2C1D21 - GRADE 3C1D28 - GRADE 0C1D28 - GRADE 1C1D28 - GRADE 2C1D28 - GRADE 3C1D35 - GRADE 0C1D35 - GRADE 1C1D35 - GRADE 2C1D35 - GRADE 3C2D1 - GRADE 0C2D1 - GRADE 1C2D1 - GRADE 2C2D1 - GRADE 3C2D7 - GRADE 0C2D7 - GRADE 1C2D7 - GRADE 2C2D7 - GRADE 3C2D14 - GRADE 0C2D14 - GRADE 1C2D14 - GRADE 2C2D14 - GRADE 3C2D21 - GRADE 0C2D21 - GRADE 1C2D21 - GRADE 2C2D21 - GRADE 3C2D28 - GRADE 0C2D28 - GRADE 1C2D28 - GRADE 2C2D28 - GRADE 3C2D35 - GRADE 0C2D35 - GRADE 1C2D35 - GRADE 2C2D35 - GRADE 3Remission visit GRADE 0Remission visit GRADE 1Remission visit GRADE 2Remission visit GRADE 3
Eltrombopag (ELQ) QD58951421651361330314201220081109010800071005000300056231

Incidence of Hemorrhagic Events

Incidence of bleeding events using WHO bleeding grade (G0=No bleeding, G1=Petechiae, G2=Mild blood loss, G3=Gross blood loss, G4=Debilitating blood loss) by week and cycle (NCT01890746)
Timeframe: Baseline, weekly within induction and re-induction cycles, end of therapy

InterventionParticipants (Number)
C1D7 - GRADE 0C1D7 - GRADE 1C1D7 - GRADE 2C1D7 - GRADE 3C1D14 - GRADE 0C1D14 - GRADE 1C1D14 - GRADE 2C1D14 - GRADE 3C1D21 - GRADE 0C1D21 - GRADE 1C1D21 - GRADE 2C1D21 - GRADE 3C1D28 - GRADE 0C1D28 - GRADE 1C1D28 - GRADE 2C1D28 - GRADE 3C1D35 - GRADE 0C1D35 - GRADE 1C1D35 - GRADE 2C1D35 - GRADE 3C1D42 - GRADE 0C1D42 - GRADE 1C1D42 - GRADE 2C1D42 - GRADE 3C2D1 - GRADE 0C2D1 - GRADE 1C2D1 - GRADE 2C2D1 - GRADE 3C2D7 - GRADE 0C2D7 - GRADE 1C2D7 - GRADE 2C2D7 - GRADE 3C2D14 - GRADE 0C2D14 - GRADE 1C2D14 - GRADE 2C2D14 - GRADE 3C2D21 - GRADE 0C2D21 - GRADE 1C2D21 - GRADE 2C2D21 - GRADE 3C2D28 - GRADE 0C2D28 - GRADE 1C2D28 - GRADE 2C2D28 - GRADE 3C2D35 - GRADE 0C2D35 - GRADE 1C2D35 - GRADE 2C2D35 - GRADE 3C2D42 - GRADE 0C2D42 - GRADE 1C2D42 - GRADE 2C2D42 - GRADE 3Remission visit GRADE 0Remission visit GRADE 1Remission visit GRADE 2Remission visit GRADE 3
Placebo QD4716604313604371125300112000100840083007300720071002100010058400

Number of Participants Who Required Medical Resource Utilization

Medical Resource Utilization pertained to unscheduled hospitalizations, unscheduled office visits, unscheduled laboratory tests, and unscheduled procedures. (NCT01890746)
Timeframe: At screening and from start of treatment to end of therapy/remission assessment visit (Day 42 of the latest chemotherapy cycle)

,
InterventionCount of participants (Number)
In-patient hospitalizations/ admissions?Diagnostic imaging procedures performed?Health care resources use or emergency visits?Out-patient lab tests performed?
Eltrombopag (ELQ) QD3386
Placebo QD4466

Number of Participants With Any Adverse Events (AE) and Any Serious Adverse Events (SAE) as a Measure of Safety and Tolerability.

An AE is defined as any untoward medical occurrence in a participant or clinical investigation participant, temporally associated with the use of a medicinal product, whether or not considered related to the medicinal product. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom, or disease (new or exacerbated) temporally associated with the use of a medicinal product. An SAE is defined as any untoward medical occurrence that, at any dose, results in death, is life threatening, requires hospitalization or prolongation of existing hospitalization, results in disability/incapacity, is a congenital anomaly/birth defect, is an important medical event that jeopardizes the participant or may require medical or surgical intervention to prevent one of the other outcomes listed in the above definition, or is associated with liver injury and impaired liver function. (NCT01890746)
Timeframe: From the time the first dose of study treatment was administered until 30 days following discontinuation of investigational product regardless of initiation of a new cancer therapy or transfer to hospice

,
InterventionParticipants (Number)
Any AEAny SAE
Eltrombopag (ELQ) QD7224
Placebo QD6614

Number of Participants With Worst-case Changes From Baseline in Electrocardiogram (ECG) Values

The number of participants with worst case post-baseline changes (normal, abnormal - not clinically significant [NCS], abnormal - clinically significant [NS]) in ECG QT prolonged values are presented. The protocol does not define the criteria for normal, abnormal-NCS and abnormal CS ECG. The outcome was based solely on the investigator interpretation of ECG tracings. (NCT01890746)
Timeframe: Baseline and Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
InterventionParticipants (Number)
NormalAbnormal - NCSAbnormal - CS
Eltrombopag (ELQ) QD34232
Placebo QD33291

Number of Participants With Worst-case Changes From Baseline in the Eastern Cooperative Oncology Group (ECOG) Performance Status

The number of participants with worst case post-baseline changes (improved, no change, deteriorated) are presented. (NCT01890746)
Timeframe: Baseline and Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
InterventionParticipants (Number)
DeterioratedImprovedNo Change
Eltrombopag (ELQ) QD36037
Placebo QD36134

Number of Participants With Worst-case Grade Changes From Baseline in the Clinical Chemistry Parameters

The number of participants with a maximum post-baseline grade increase of Grade 3 or Grade 4 from their baseline grade are presented. Clinical Clinical Chemistry parameters included only lab tests that are gradable by CTCAE v4.0. (NCT01890746)
Timeframe: Baseline and up to Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
InterventionParticipants (Number)
Alanine Aminotransferase, G3Albumin, G3Aspartate Aminotransferase, G3Bilirubin, G3Bilirubin, G4Calcium Low, G3Creatinine, G3Creatinine, G4Glucose High, G3Glucose High, G4Magnesium Low, G3Magnesium High, G3Phosphate, G3Phosphate, G4Potassium Low, G3Potassium Low, G4Potassium High, G3Potassium High, G4Sodium Low, G3Urate, G4
Eltrombopag (ELQ) QD160100016003101804133
Placebo QD5414111031101901020140

Number of Participants With Worst-case Grade Changes From Baseline in the Hematology Parameters

The number of participants with a maximum post-baseline grade increase of Grade 3 (G3) or Grade 4 (G4) from their baseline grade are presented. Hematology parameters included only lab tests that are gradable by Common Terminology Criteria for Adverse Events (CTCAE) v4.0. (NCT01890746)
Timeframe: Baseline and up to Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
InterventionParticipants (Number)
Hemoglobin Low, G3Leukocytes, G3Leukocytes, G4Lymphocytes Low, G3Lymphocytes Low, G4Neutrophils, G4Platelets, G3Platelets, G4
Eltrombopag (ELQ) QD53109313544163
Placebo QD46105263839056

Percentage of Participants With Disease Response Rate and Type of Response

"Disease response as assessed by the investigator using the AML International Working Group Response Assessment at the end of therapy/remission assessment visit; Complete remission (CR): defined as transfusion independence, blood count recovery (Abs. neutrophil count > 1.0 Gi/L and Platelet count > 100.0 Gi/L), no leukemic blast in peripheral blood, Bone Marrow (BM) blasts < 5%, maturation of all cell lines, Auer rods not detectable, and no extramedullary disease.~Partial remission (PR): defined as CR except that for BM blasts where a decrease of at least 50% of BM blasts to 5-25% in BM aspirate is sufficient or BM blasts < 5% with Auer rods present.~Overall response (OR) = CR + PR." (NCT01890746)
Timeframe: Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
InterventionPercentage of participants (Number)
Overall responseComplete Remission (CR)Partial Remission (PR)
Eltrombopag (ELQ) QD70655
Placebo QD73703

Summary of Absolute Neutrophil Counts (ANC)

Absolute neutrophil counts over time (NCT01890746)
Timeframe: Baseline, daily then weekly within cycle up to 42 days after last chemotherapy dose, end of therapy /remission assessment visit

InterventionGi/L (Median)
BaselineC1D1C1D2C1D3C1D4C1D5C1D6C1D7C1D8C1D9C1D14C1D21C1D28C1D35C2D1C2D2C2D3C2D4C2D5C2D6C2D7C2D14
Eltrombopag (ELQ) QD0.80.80.60.60.40.30.20.10.10.00.00.64.32.20.10.10.20.30.20.10.00.0

Summary of Absolute Neutrophil Counts (ANC)

Absolute neutrophil counts over time (NCT01890746)
Timeframe: Baseline, daily then weekly within cycle up to 42 days after last chemotherapy dose, end of therapy /remission assessment visit

InterventionGi/L (Median)
BaselineC1D1C1D2C1D3C1D4C1D5C1D6C1D7C1D8C1D9C1D14C1D21C1D28C1D35C1D42C2D1C2D2C2D3C2D4C2D5C2D6C2D7C2D14
Placebo QD0.50.60.50.40.20.20.10.10.00.00.00.32.71.73.10.00.20.50.50.10.10.10.0

Summary of Hemoglobin

Hemoglobin level over time (NCT01890746)
Timeframe: Baseline, daily then weekly within cycle up to 42 days after last chemotherapy dose, end of therapy /remission assessment visit

Interventiong/L (Median)
BaselineC1D1C1D2C1D3C1D4C1D5C1D6C1D7C1D8C1D9C1D14C1D21C1D28C1D35C2D1C2D2C2D3C2D4C2D5C2D6C2D7C2D14C2D21C2D28C2D35
Eltrombopag (ELQ) QD87.688.088.086.083.084.086.085.085.384.585.088.099.099.094.588.586.589.088.084.084.577.586.089.0104.0

Summary of Hemoglobin

Hemoglobin level over time (NCT01890746)
Timeframe: Baseline, daily then weekly within cycle up to 42 days after last chemotherapy dose, end of therapy /remission assessment visit

Interventiong/L (Median)
BaselineC1D1C1D2C1D3C1D4C1D5C1D6C1D7C1D8C1D9C1D14C1D21C1D28C1D35C1D42C2D1C2D2C2D3C2D4C2D5C2D6C2D7C2D14C2D21C2D28C2D35C2D42
Placebo QD87.086.083.082.081.583.082.083.084.081.584.088.098.094.098.082.586.580.086.584.085.083.087.091.080.087.090.5

Summary of Platelet Counts Over Time

Platelet counts over time (NCT01890746)
Timeframe: Baseline, daily then weekly within cycle up to 42 days after last chemotherapy dose, end of therapy /remission assessment visit

InterventionGi/L (Median)
BaselineC1D1C1D2C1D3C1D4C1D5C1D6C1D7C1D8C1D9C1D14C1D21C1D28C1D35C2D1C2D2C2D3C2D4C2D5C2D6C2D7C2D14C2D21C2D28C2D35
Eltrombopag (ELQ) QD51.552.043.535.536.533.032.027.024.020.516.539.0484.5547.031.026.025.532.037.027.024.010.527.068.0515.0

Summary of Platelet Counts Over Time

Platelet counts over time (NCT01890746)
Timeframe: Baseline, daily then weekly within cycle up to 42 days after last chemotherapy dose, end of therapy /remission assessment visit

InterventionGi/L (Median)
BaselineC1D1C1D2C1D3C1D4C1D5C1D6C1D7C1D8C1D9C1D14C1D21C1D28C1D35C1D42C2D1C2D2C2D3C2D4C2D5C2D6C2D7C2D14C2D21C2D28C2D35C2D42
Placebo QD50.048.542.037.029.029.030.027.022.019.018.025.0121.0181.0304.030.528.529.035.522.033.028.516.038.0173.0272.0147.5

Worst-case Change From Baseline in Pulse Rate Values

The worst-case post Baseline high and low changes in pulse rate values from Baseline are presented. Baseline was defined as the most recent, non-missing value prior to or on the first study treatment dose date. Post Baseline is defined as the highest and lowest non-missing post Baseline value respectively. Change from Baseline was calculated as the post Baseline value minus the Baseline value. (NCT01890746)
Timeframe: Baseline and up to Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
InterventionBeats/minute (Mean)
HighLow
Eltrombopag (ELQ) QD18.48-10.36
Placebo QD17.73-11.24

Worst-case Post Baseline Change in Blood Pressure Values From Baseline

The worst-case post Baseline high changes in systolic blood pressure (SBP) and diastolic blood pressure (DBP) values from Baseline are presented. Baseline was defined as the most recent, non-missing value prior to or on the first study treatment dose date. Change from Baseline was calculated as the visit value minus the Baseline value. (NCT01890746)
Timeframe: Baseline and up to Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
Interventionmillimeter of mercury (mmHg) (Mean)
SBPDBP
Eltrombopag (ELQ) QD14.599.38
Placebo QD14.3412.61

Worst-case Post Baseline Change in Temperature Values From Baseline

The worst-case post Baseline high and low changes in temperature values from Baseline are presented. Baseline was defined as the most recent, non-missing value prior to or on the first study treatment dose date. Post Baseline was defined as the highest and lowest non-missing post Baseline value respectively. Change from Baseline was calculated as the post Baseline value minus the Baseline value. (NCT01890746)
Timeframe: Baseline and up to Day 42 of the latest chemotherapy cycle (Up to 8 weeks)

,
InterventionDegrees Celsius (Mean)
HighLow
Eltrombopag (ELQ) QD0.62-0.44
Placebo QD0.77-0.63

100-Day Treatment-Related Mortality

The cumulative incidence was estimated after taking into account the competing risk of relapse post-ASCT. (NCT00695409)
Timeframe: From peripheral stem cell infusion (Day0 ASCT) to death due to any couse, assessed up to 5 years

InterventionPercentage of Participants (%) (Number)
Treatment (RIT, ZBEAM, ASCT)0.9

2-Year Cumulative Incidence of Progression

The cumulative incidence was estimated after taking into account the competing risk of early death. (NCT00695409)
Timeframe: From peripheral stem cell infusion (Day0 ASCT) to date of first observation of progressive disease or relapsed disease, assessed up to 5 years

InterventionPercentage of Participants (%) (Number)
Treatment (RIT, ZBEAM, ASCT)28

2-Year Overall Survival

Overall survival (OS) was measured from peripheral stem cell infusion to death from any cause. It was estimated using the Kaplan-Meier method; the 95% confidence interval was calculated using Greenwood's formula. [Breslow NE, Day NE. Statistical methods in cancer research: volume II, the design and analysis of cohort studies. IARC Sci Publ 1987;82:1-406.] (NCT00695409)
Timeframe: From peripheral stem cell infusion (Day0 ASCT) to death due to any cause, assessed up to 5 years

InterventionPercentage of Participants (%) (Number)
Treatment (RIT, ZBEAM, ASCT)89

2-Year Progression-Free Survival

Progression-free survival (PFS) was defined as time from peripheral stem cell infusion to recurrence, progression or death. In a clinical trial, measuring the progression-free survival is one way to see how well a new treatment works. Progression-free survival was estimated using the Kaplan-Meier method; the 95% confidence interval was calculated using Greenwood's formula [Breslow NE, Day NE. Statistical methods in cancer research: volume II, the design and analysis of cohort studies. IARC Sci Publ 1987;82:1-406.] (NCT00695409)
Timeframe: From peripheral stem cell infusion (Day0 ASCT) to first observation of progressive disease or death due to any cause, whichever comes first, assessed up to 5 years

InterventionPercentage of Participants (%) (Number)
Treatment (RIT, ZBEAM, ASCT)71

Number of Patients With Active Disease at ASCT Achieving CR/PR by Day 100 After ASCT

Responses are assessed using the Revised Criteria for Malignant Lymphoma Response Definitions for Clinical Trials (Cheson et al. 2007). Complete Response (CR) defined as disappearance of all evidence of disease. Partial Response (PR) defined as regression of measurable disease and no new sites. (NCT00695409)
Timeframe: Up to Day 100 post-ASCT

InterventionParticipants (Count of Participants)
Patients With Active Disease at ASCT53

Number of Patients With RIT/ZBEAM Developing Therapy Induced MDS and AML

Patient receiving the full treatment of RIT/ZBEAM developed therapy induced MDS or AML. (NCT00695409)
Timeframe: From peripheral stem cell infusion (Day0 ASCT) to onset of therapy induced MDS/AML, assessed up to 5 years

InterventionParticipants (Count of Participants)
Treatment (RIT, ZBEAM, ASCT)0

Time to Neutrophil Recovery

Neutrophil recovery was defined as the first of 3 consecutive days of an absolute neutrophil count ≥ 500/µL. (NCT00695409)
Timeframe: From peripheral stem cell infusion (Day0 ASCT) till the first of 3 consecutive days of an absolute neutrophil count ≥ 500/µL.)

InterventionDays (Median)
Treatment (RIT, ZBEAM, ASCT)10

Time to Platelet Recovery

Platelet recovery was defined as the first of 7 consecutive days with a platelet count ≥ 20,000/µL with no transfusions. (NCT00695409)
Timeframe: From peripheral stem cell infusion (Day0 ASCT) till the first of 7 consecutive days with a platelet count ≥ 20,000/µL with no transfusions

InterventionDays (Median)
Treatment (RIT, ZBEAM, ASCT)12

2-yr Overall Survival

Overall survival is defined as the time from date of diagnosis to death or date of last follow-up. 2-year overall survival is the probability of patients remaining alive at 2-years from study entry estimated using Kaplan-Meier (KM) methods which censors patients at date of last follow-up. Precision of this conditional probability estimate was measured in terms of standard error. Median OS, the original primary endpoint, was not estimable based on the Kaplan-Meier method because of insufficient follow-up. (NCT00084838)
Timeframe: Patients are followed for survival up to 5 yrs post-therapy completion or death; As of this analysis, median follow-up among survivors was 31 months with the longest follow-up being 40 months.

Interventionprobability (Number)
Multi-agent Intrathecal and Systemic CT With RT (Modified IRS0.70

Grade 3-4 Allergy/Immunology

All Grade 3-4 Allergy/Immunology events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)1

Grade 3-4 Auditory/Hearing Events

All Grade 3-4 Auditory/Hearing events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)8

Grade 3-4 Blood/Bone Marrow Events

"All Grade 3-4 Blood/Bone Marrow events based on CTCAEv2 as reported on case report forms.~Arm Name" (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)564

Grade 3-4 Cardiovascular Events

All Grade 3-4 Cardiovascular events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)6

Grade 3-4 Constitutional Events

All Grade 3-4 Constitutional events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)22

Grade 3-4 Dermatology Events

All Grade 3-4 Dermatology events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)3

Grade 3-4 Gastrointestinal Events

All Grade 3-4 Gastrointestinal events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)139

Grade 3-4 Hemorrhage Events

All Grade 3-4 Hemorrhage events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)1

Grade 3-4 Hepatic Events

All Grade 3-4 Hepatic events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)8

Grade 3-4 Infection/Febrile Neutropenia Events

All Grade 3-4 Infection/Febrile Neutropenia events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)49

Grade 3-4 Metabolic/Laboratory Events

All Grade 3-4 Metabolic/Laboratory events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)128

Grade 3-4 Muscloskeletal Events

All Grade 3-4 Muscloskeletal events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)8

Grade 3-4 Neurology Events

All Grade 3-4 Neurology events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)45

Grade 3-4 Pain Events

All Grade 3-4 Pain events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)31

Grade 3-4 Pulmonary Events

All Grade 3-4 Pulmonary events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)4

Grade 3-4 Renal/Genitourinary Events

All Grade 3-4 Renal/Genitourinary events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)4

Grade 3/4 Events

All Grade 3-4 events based on CTCAEv2 as reported on case report forms. (NCT00084838)
Timeframe: Assessed during therapy up to 30 days post-therapy completion which is approximately 55 weeks for patients who completed therapy.

Interventionadverse events (Number)
Multi-agent Intrathecal and Systemic CT With RT (Mod IRS III)1021

Pre-Radiation Therapy Chemotherapeutic Response

"Response pre-RT/post-CT was defined as follows with overall response defined as achieving PR or CR.~Complete Response (CR): Complete resolution of all initially demonstrable tumor on MRI or CT evaluation w/o appearance of any new areas of disease; negative CSF cytology. Partial Response (PR): >/= 50% decrease in the sum of the products of the maximum perpendicular diameters of the tumor (sum LD) relative to baseline w/o appearance of any new areas of disease; CSF cytology unchanged from that at diagnosis or clearing after being initially positive Stable Disease (SD): <50% decrease in the sum LD w/o appearance of any new areas of disease; CSF cytology unchanged from that at diagnosis or clearing after being initially positive Progressive Disease (PD): >/= 25% increase in the sum LD relative to baseline, or the appearance of any new areas of disease or appearance of positive cytology after two consecutive negative samples." (NCT00084838)
Timeframe: Assessed at study entry and pre-RT/post-CT at week 7.

Interventionproportion of evaluable patients (Number)
Multi-agent Intrathecal and Systemic CT With RT (Modified IRS0.58

Complete Response Rate

Bone marrow showing less than 5% myeloblasts with normal maturation of all cell lines, an ANC of at least 1000/cu mm and a platelet count of 100,000/cu mm, absence of blast in peripheral blood, absence of identifiable leukemic cells in the bone marrow, clearance of disease-associated cytogenetic abnormalities, and clearance of any previously existing extramedullary disease. These criteria are taken from Dohner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 2010;115:453-474 (NCT01349972)
Timeframe: 3 years

Interventionparticipants (Number)
Arm I (Alvocidib, Cytarabine, Mitoxantrone Hydrochloride)76
Arm II (Cytarabine, Daunorubicin Hydrochloride)24

Disease-free Survival

Probabilities will be estimated with the Kaplan-Meier estimate. Survival estimates at two years will be estimated. Disease-free survival Overall survival was defined from date of randomization to death or last known follow-up. Event free survival was defined as date of randomization to the first occurrence of persistent AML after 1 cycle of induction, relapse or death. Patients were censored for event free survival if they had received non-protocol therapy or a stem cell transplant. (NCT01349972)
Timeframe: Time from randomization until death from any cause or relapse or recurrence, assessed up to 2 years

Interventionyears (Median)
Arm I (Alvocidib, Cytarabine, Mitoxantrone Hydrochloride)1.46
Arm II (Cytarabine, Daunorubicin Hydrochloride)1.85

Incidence of Toxicities, Characterized by Number of Events by Treatment and Grade

The descriptions and grading scales found in the revised NCI Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 will be utilized for AE reporting. (NCT01349972)
Timeframe: Up to 14 days after completion of study treatment

InterventionNumber of events (Number)
Arm I (Alvocidib, Cytarabine, Mitoxantrone Hydrochloride)156
Arm II (Cytarabine, Daunorubicin Hydrochloride)77

Number of Patients With Minimal Residual Disease

Comparisons of the treatments with respect to MRD will be based on the number of patients with MRD at day 14 after the start of treatment. (NCT01349972)
Timeframe: From study start to 14 days after the start of treatment

InterventionParticipants (Count of Participants)
Arm I (Alvocidib, Cytarabine, Mitoxantrone Hydrochloride)26
Arm II (Cytarabine, Daunorubicin Hydrochloride)24

Overall Survival

Probabilities will be estimated with the Kaplan-Meier estimate. Survival estimates at two years will be estimated. (NCT01349972)
Timeframe: 4 years

Interventionyears (Median)
Arm I (Alvocidib, Cytarabine, Mitoxantrone Hydrochloride)1.46
Arm II (Cytarabine, Daunorubicin Hydrochloride)1.850

Progression-free Survival

Probabilities will be estimated with the Kaplan-Meier estimate. Survival estimates at two years will be estimated. (NCT01349972)
Timeframe: 4 years

Interventionyears (Median)
Arm I (Alvocidib, Cytarabine, Mitoxantrone Hydrochloride)0.81
Arm II (Cytarabine, Daunorubicin Hydrochloride)0.28

Number of Participants Experiencing Dose Limiting Toxicities (DLTs) at the Maximum Tolerated Dose (MTD) for Decitabine When Given Together With G-CLAM Toxicities (DLTs) (Phase I)

"Evaluated according to National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. MTD is defined as the highest dose studied in which the incidence of DLT is < 33%. Each participant was monitored for DLTs in the first 20 days of treatment. DLTs monitored included~any grade 3 non-hematologic toxicity lasting more than 48 hours that resulted in more than a 7 day delay of the subsequent treatment cycle (except for febrile neutropenia or infection)~any grade 4 or greater non-hematologic toxicity (except febrile neutropenia and infection unless a direct consequence of a treatment-related toxicity, and except constitutional symptoms if they recovered to grade 2 or less within 14 days)~lack of recovery of the absolute neutrophil count to 500/microliter or greater and lack of self-sustained platelet count of at least 50,000/microliter by treatment day +49 with no evidence of residual leukemia" (NCT02921061)
Timeframe: Up to 49 days

InterventionParticipants (Count of Participants)
Treatment (Decitabine 20 mg/m2 and G-CLAM)3

Number of Participants Who Achieved Remission (Complete Remission [CR]/CR With Incomplete Peripheral Blood Count Recovery [CRi])

(NCT02921061)
Timeframe: Up to 1 year

Interventionparticipants (Number)
Treatment (Decitabine 20 mg/m2 and G-CLAM)13

Number of Participants With Event-free Survival

(NCT02921061)
Timeframe: Up to 1 year

Interventionparticipants (Number)
Treatment (Decitabine 20 mg/m2 and G-CLAM)8

Number of Participants With Minimal Residual Disease Negative (MRDneg) Complete Remission (Phase II)

Compared to historical controls of filgrastim, cladribine, cytarabine, and mitoxantrone hydrochloride (G-CLAM) alone. A Simon Minimax two-stage design will be used. (NCT02921061)
Timeframe: Up to 1 year

Interventionparticipants (Number)
Treatment (Decitabine 20 mg/m2 and G-CLAM)13

Number of Participants With Overall Survival

(NCT02921061)
Timeframe: Up to 1 year

Interventionparticipants (Number)
Treatment (Decitabine 20 mg/m2 and G-CLAM)9

Number of Participants With Relapse-free Survival

(NCT02921061)
Timeframe: Up to 1 year

Interventionparticipants (Number)
Treatment (Decitabine 20 mg/m2 and G-CLAM)8

AUCtau of Glasdegib in Participants Receiving Glasdegib and LDAC at Phase 2 Unfit on Cycle 1/Day 10

(NCT01546038)
Timeframe: Pre-dose, 1, 2, 4, and 6 hour post-dose on Cycle 1/Day 10

Interventionng*hr/mL (Geometric Mean)
Phase 2 Unfit: Glasdegib 100 mg + LDAC17210

Baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 1B

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. The data of analytes for which the serum level showed statistically significant correlation with clinical response in Arm C are reported. Baseline levels statistically associated with best overall response was only seen in SDF-1 (stromal cell-derived factor 1) in glasdegib+cytarabine/daunorubicin arm. (NCT01546038)
Timeframe: Baseline (Cycle 1/Day 1 pre-dose for Glasdegib + LDAC and Glasdegib + Decitabine Arms; Induction Cycle 1/Day -3 pre-dose for Glasdegib +Cytarabine/Daunorubicin Arm)

Interventionpg/mL (Median)
Phase 1B: Glasdegib + Cytarabine/Dauno (Biomarker, Responder)2275.00
Phase 1B: Glasdegib+Cytarabine/Dauno(Biomarker,Non-Responder)3275.00

Baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 2 Fit

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. Selected data of analyte for which the serum level showed statistically significant correlation with clinical response are reported. (NCT01546038)
Timeframe: Baseline (Induction Cycle 1/Day -3 pre-dose)

Interventionpg/mL (Median)
Phase 2 Fit (Biomarker, Responder)323.00
Phase 2 Fit (Biomarker, Non-Responder)362.00

Baseline mRNA Levels Associated With Best Overall Response at Phase 2 Fit

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. Whole blood mRNA analyses were performed on 21 mRNA candidates. Baseline mRNA level showing statistically significant correlation with clinical response are reported. Baseline mRNA levels statistically significant associated with best overall response was only seen for CCND2 (G1/S-Specific Cyclin D2). (NCT01546038)
Timeframe: Baseline (Induction Cycle 1/Day -3 pre-dose)

InterventionNormalized expression units (Median)
Phase 2 Fit (Biomarker, Responder)10.9
Phase 2 Fit (Biomarker, Non-Responder)14.80

Cmax of Glasdegib in Participants Receiving Glasdegib and LDAC at Phase 2 Unfit on Cycle 1/Day 10

(NCT01546038)
Timeframe: Pre-dose, 1, 2, 4, and 6 hour post-dose on Cycle 1/Day 10

Interventionng/mL (Geometric Mean)
Phase 2 Unfit: Glasdegib 100 mg + LDAC1252

Number of Participants With Dose-limiting Toxicities (DLTs) at Phase 1B

A DLT was any of the following adverse events (AEs) in Cycle 1 and considered by the investigator possibly related to glasdegib in combination with chemotherapy: (1) Grade >= 3 non-hematologic toxicity, excluding Grade >= 3 infection, fever (including febrile neutropenia), infusion related AEs, electrolyte abnormalities and ALT/AST elevation that returned to Grade <= 1 or baseline within 7 days; (2) prolonged myelosuppression that lasted longer than 42 days from the point of detection, defined as absolute neutrophil count (ANC) < 500/microliter(mcL) or platelet count < 10 *10^9/L with a normal bone marrow (<5% blasts and no evidence of disease or dysplasia); (3) inability to deliver at least 80% of the planned study doses for all agents in a combination due to non-hematologic toxicities; (4) Delay of >28 days in receiving the next scheduled cycle due to persisting non-hematologic toxicities. Arm A: Glasdegib+LDAC; Arm B: Glasdegib+Decitabine; Arm C: Glasdegib+Cytarabine/Daunorubicin. (NCT01546038)
Timeframe: Arms A and B: Cycle 1, Day 1 to Day 28; Arm C: Cycle 1, Day -3 to Day 21 or to Day 28 depending on when the next chemotherapy cycle was started

InterventionParticipants (Number)
Phase 1B: Glasdegib 100 mg + LDAC0
Phase 1B: Glasdegib 200 mg + LDAC0
Phase 1B: Glasdegib 100 mg + Decitabine0
Phase 1B: Glasdegib 200 mg + Decitabine0
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin1
Phase 1B: Glasdegib 200 mg + Cytarabine/Daunorubicin0

Overall Survival (OS) at Phase 1B

OS was defined as duration from the date of randomization to the date of death from any cause. Kaplan-Meier (KM) method was used to estimate median OS. In this method, every participant had a follow-up time which was associated with an indicator, 1=event (death in our case), and 0 =censored. If the participants were not known to have died, time to date of last known to be alive was used as to calculate the follow-up time and indicator was 0 for these participants. KM method estimates the median OS based on the K-M curve. The K-M curve only drops when we had an event and censor data are the ticks in the graph. To estimate median OS, the K-M curve usually will be smoothed first and a line will be drawn at 50%. The median OS is the point when K-M curve and the horizontal hit. Survival status was collected every month for the first 2 months after discontinuation of study treatment and thereafter every 2 months until death or 4 years from each participant's first dose. (NCT01546038)
Timeframe: First dose to Follow-up (4 years)

InterventionMonths (Median)
Phase 1B: Glasdegib + LDAC4.4
Phase 1B: Glasdegib + Decitabine11.5
Phase 1B: Glasdegib + Cytarabine/Daunorubicin37.8

Overall Survival (OS) at Phase 2 Unfit

OS was defined as duration from the date of randomization to the date of death from any cause. Kaplan-Meier (KM) method was used to estimate median OS. In this method, every participant had a follow-up time which was associated with an indicator, 1=event (death in our case), and 0 =censored. If the participants were not known to have died, time to date of last known to be alive was used as to calculate the follow-up time and indicator was 0 for these participants. KM method estimates the median OS based on the K-M curve. The K-M curve only drops when we had an event and censor data are the ticks in the graph. To estimate median OS, the K-M curve usually will be smoothed first and a line will be drawn at 50%. The median OS is the point when K-M curve and the horizontal hit. Survival status was collected every month for the first 2 months after discontinuation of study treatment and thereafter every 2 months until death or 4 years from time of randomization for each participant. (NCT01546038)
Timeframe: Randomization to Follow-up (4 years)

InterventionMonths (Median)
Phase 2 Unfit: Glasdegib 100 mg + LDAC8.8
Phase 2 Unfit: LDAC Alone4.9

Percentage of Participants With Complete Response (CR) at Phase 2 Unfit

For AML participants:CR were those with repeat bone marrow showing <5% myeloblasts,spicules present and no Auer rods, peripheral blood showing neutrophils>=1000/mcL and platelets>=100,000/mcL, transfusion independent and no extramedullary disease. For MDS participants:CR were those with repeat bone marrow showing <=5% myeloblasts, peripheral blood showing neutrophils>=1000/mcL, platelets>=100,000/mcL, 0% blast and hemoglobin (Hgb)>= 11 g/dL, normal maturation of all cell lines. (NCT01546038)
Timeframe: 4 years

InterventionPercentage of participants (Number)
Phase 2 Unfit: Glasdegib 100 mg + LDAC18.2
Phase 2 Unfit: LDAC Alone2.3

Percentage of Participants With CR / Complete Response With Incomplete Blood Count Recovery (CRi) at Phase 1B

For AML participants:CR were those with repeat bone marrow showing <5% myeloblasts,spicules present and no Auer rods, peripheral blood showing neutrophils>=1000/mcL and platelets>=100,000/mcL, transfusion independent and no extramedullary disease. For MDS participants:CR were those with repeat bone marrow showing <=5% myeloblasts, peripheral blood showing neutrophils>=1000/mcL, platelets>=100,000/mcL, 0% blast and hemoglobin (Hgb)>= 11 g/dL, normal maturation of all cell lines.For AML and MDS participants, complete response with incomplete blood count recovery(CRi)were those with repeat bone marrow showing <5% myeloblasts with either platelets or neutrophils not recovered (platelets <100,000/mcL or neutrophils <1000/mcL). (NCT01546038)
Timeframe: 4 years

InterventionPercentage of participants (Number)
Phase 1B: Glasdegib + LDAC8.7
Phase 1B: Glasdegib + Decitabine28.6
Phase 1B: Glasdegib + Cytarabine/Daunorubicin54.5

Post-baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 1B - Induction Cycle 1/Day 3

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. The data of analytes for which the serum level showed statistically significant correlation with clinical response are reported. Post-baseline levels statistically significant associated with best overall response was only seen for SDF-1 (Stromal cell-derived factor 1) at Induction Cycle 1/Day 3. (NCT01546038)
Timeframe: Induction Cycle 1/Day 3, 1 Hour Post dose

Interventionpg/mL (Median)
Phase 1B: Glasdegib + Cytarabine/Dauno (Biomarker, Responder)2510.00
Phase 1B: Glasdegib+Cytarabine/Dauno(Biomarker,Non-Responder)3260.00

Post-baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 1B - Induction Cycle 1/Lead-In

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. The data of analytes for which the serum level showed statistically significant correlation with clinical response are reported. Post-baseline levels statistically significant associated with best overall response was only seen for MMP-3 (Matrix metalloproteinase-3) at Induction Cycle 1/Lead-in. (NCT01546038)
Timeframe: Induction Cycle 1/Lead-in, 1 Hour Post dose

Interventionng/mL (Median)
Phase 1B: Glasdegib + Cytarabine/Dauno (Biomarker, Responder)8.90
Phase 1B: Glasdegib+Cytarabine/Dauno(Biomarker,Non-Responder)10.50

Post-baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 2 Fit - Induction Cycle 1/Day 10

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. Selected data of analyte for which the serum level showed statistically significant correlation with clinical response are reported. (NCT01546038)
Timeframe: Induction Cycle 1/Day 10, 1 Hour Post dose

Interventionpg/mL (Mean)
Phase 2 Fit (Biomarker, Responder)1.20
Phase 2 Fit (Biomarker, Non-Responder)6.60

Post-baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 2 Fit - Induction Cycle 1/Day 3

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. Selected data of analyte for which the serum level showed statistically significant correlation with clinical response are reported. (NCT01546038)
Timeframe: Induction Cycle 1/Day 3, 1 Hour Post dose

Interventionpg/mL (Mean)
Phase 2 Fit (Biomarker, Responder)3.20
Phase 2 Fit (Biomarker, Non-Responder)10.90

Post-baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 2 Unfit - End of Treatment

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. Selected data of analyte for which the serum level showed statistically significant correlation with clinical response are reported. (NCT01546038)
Timeframe: End of Treatment (maximum of 12 cycles from start of therapy or until disease progression or relapse, participant refusal or unacceptable toxicity occurred, whichever came first, an average of 1 year)

Interventionpg/mL (Median)
Phase 2 Unfit: Glasdegib 100 mg + LDAC (Biomarker, Responder)0.00
Phase 2 Unfit: Glasdegib 100 mg+LDAC(Biomarker, Non-Responder)9.40

Pre-dose Plasma Concentration (Ctrough) of Glasdegib in Phase 2 Fit on Induction Cycle 1/Day 10

(NCT01546038)
Timeframe: Pre-dose, 1 and 4 hours post-dose on Induction Cycle 1/Day 10

Interventionng/mL (Geometric Mean)
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin308.7

Ratios of mRNA Levels Associated With Best Overall Response at Phase 2 Unfit

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. Whole blood mRNA analyses were performed on 21 mRNA candidates. Ratios of mRNA level to baseline showing statistically significant correlation with clinical response are reported. Ratios of mRNA levels to baseline statistically significant associated with best overall response was only seen for MYCN (Neuroblastoma Myc oncogene) at Cycle 1/Day 1. (NCT01546038)
Timeframe: Baseline (Cycle 1/Day 1 pre-dose); Cycle 1/Day 1, 1 Hour Post dose

Interventionratio (Median)
Phase 2 Unfit: Glasdegib 100 mg + LDAC (Biomarker, Responder)1.60
Phase 2 Unfit: Glasdegib 100 mg+LDAC(Biomarker, Non-Responder)0.50

Serum Levels of Circulating Protein Analytes at Phase 1B - Induction Cycle 1/Day 3

Serum levels were determined for 38 circulating proteins. Values showing statistically significant, ≥2-fold difference compared with baseline are reported here. Statistically significant, >=2-fold baseline difference was only seen for MMP-3 (Matrix metalloproteinase-3) at Induction Cycle 1/Day 3. (NCT01546038)
Timeframe: Induction Cycle 1/Day 3, 1 Hour Post dose

Interventionpg/mL (Median)
Phase 1B: Glasdegib + Cytarabine/Daunorubicin20000

Serum Levels of Circulating Protein Analytes at Phase 2 Unfit - Cycle 1/Day 1

Serum levels were determined for 38 circulating proteins. Selected value showing statistically significant difference compared with baseline is reported here. (NCT01546038)
Timeframe: Cycle 1/Day 1, 1 Hour Post dose

Interventionpg/mL (Median)
Phase 2 Unfit: Glasdegib 100 mg + LDAC483.00

Tmax of Glasdegib in Participants Receiving Glasdegib and LDAC at Phase 2 Unfit on Cycle 1/Day 10

(NCT01546038)
Timeframe: Pre-dose, 1, 2, 4, and 6 hour post-dose on Cycle 1/Day 10

InterventionHours (Median)
Phase 2 Unfit: Glasdegib 100 mg + LDAC1.67

Area Under the Plasma Concentration-time Profile From Time 0 to Dosing Interval (AUCtau) of Glasdegib in Participants Receiving Glasdegib and LDAC at Phase 1B on Cycle 1/Day 10 and Cycle 1/Day 21

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 2, 4, 6 and 24 hours post-dose on Cycle 1/Day 10 and Cycle 1/Day 21

,
Interventionng*hr/mL (Geometric Mean)
Cycle 1/Day 10Cycle 1/Day 21
Phase 1B: Glasdegib 100 mg + LDAC1502016660
Phase 1B: Glasdegib 200 mg + LDAC2860031400

Area Under the Plasma Concentration-time Profile From Time 0 to Infinity (AUCinf) of LDAC in Participants Receiving Glasdegib and LDAC at Phase 1B on Cycle 1/Day 2 and Cycle 1/Day 10

(NCT01546038)
Timeframe: Pre-dose, 0.25, 0.5, 1, 2, 4 and 6 hours post-dose on Cycle 1/Day 2 and Cycle 1/Day 10

,
Interventionng*hr/mL (Geometric Mean)
LDAC Cycle 1/Day 2LDAC Cycle 1/Day 10
Phase 1B: Glasdegib 100 mg + LDAC71.1092.28
Phase 1B: Glasdegib 200 mg + LDAC89.35143.9

Area Under the Plasma Concentration-time Profile From Time 0 to the Time of the Last Quantifiable Concentration (AUClast) of LDAC and Ara-U in Participants Receiving Glasdegib and LDAC at Phase 1B on Cycle 1/Day 2 and Cycle 1/Day 10

Ara-U is the major metabolite of cytarabine. LDAC (low dose cytarabine) is rapidly degraded to the stable metabolite Ara-U. Area under the plasma concentration-time profile from time 0 to the time of the last quantifiable concentration (AUClast) levels of both LDAC and Ara-U were reported. (NCT01546038)
Timeframe: Pre-dose, 0.25, 0.5, 1, 2, 4 and 6 hours post-dose on Cycle 1/Day 2 and Cycle 1/Day 10

,
Interventionng*hr/mL (Geometric Mean)
LDAC Cycle 1/Day 2LDAC Cycle 1/Day 10Ara-U Cycle 1/Day 2Ara-U Cycle 1/Day 10
Phase 1B: Glasdegib 100 mg + LDAC62.5565.5620362283
Phase 1B: Glasdegib 200 mg + LDAC87.49134.830503528

AUCinf of Decitabine in Participants Receiving Glasdegib and Decitabine at Phase 1B on Cycle 1/Day 1 and Cycle 1/Day 2

(NCT01546038)
Timeframe: Pre-dose, 0.5 hour from start of infusion, 1 hour (at end of infusion) and 2, 3 and 4 hours from start of infusion on Cycle 1/Day 1 and Cycle 1/Day 2

,
Interventionng*hr/mL (Geometric Mean)
Cycle 1/Day 1Cycle 1/Day 2
Phase 1B: Glasdegib 100 mg + Decitabine133.4NA
Phase 1B: Glasdegib 200 mg + Decitabine251.5NA

AUCtau of Cytarabine and Ara-U in Participants Receiving Glasdegib and Cytarabine/Daunorubicin at Phase 1B on Induction Cycle 1/Day 3

Ara-U is the major metabolite of cytarabine. LDAC (low dose cytarabine) is rapidly degraded to the stable metabolite Ara-U, levels of both cytarabine and Ara-U were reported. (NCT01546038)
Timeframe: Pre-dose, 6 and 24 hours post start of cytarabine infusion on Induction Cycle 1/Day 3

,
Interventionng*hr/mL (Geometric Mean)
CytarabineAra-U
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin107028420
Phase 1B: Glasdegib 200 mg + Cytarabine/DaunorubicinNANA

AUCtau of Daunorubicin and Daunorubicinol in Participants Receiving Glasdegib and Cytarabine/Daunorubicin at Phase 1B on Induction Cycle 1/Day 3

Daunorubicinol is the major metabolite of daunorubicin, which has anti-neoplastic activity. AUCtau values of daunorubicin and daunorubicinol are reported. (NCT01546038)
Timeframe: Pre-dose, 0.25, 0.5, 1, 4, 6, 24 hours post administration of daunorubicin on Induction Cycle 1/Day 3

,
Interventionng*hr/mL (Geometric Mean)
DaunorubicinDaunorubicinol
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin499.32152
Phase 1B: Glasdegib 200 mg + Cytarabine/Daunorubicin424.92712

AUCtau of Glasdegib in Participants Receiving Glasdegib and Cytarabine/Daunorubicin at Phase 1B on Induction Cycle 1/Day 3 and Day 10

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 6 and 24 hours post-dose on Induction Cycle 1/Day 3; pre-dose, 0.5, 1, 4, 6 and 24 hours post-dose on Induction Cycle 1/Day 10

,
Interventionng*hr/mL (Geometric Mean)
Induction Cycle 1/Day 3Induction Cycle 1/Day 10
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin933216300
Phase 1B: Glasdegib 200 mg + Cytarabine/Daunorubicin2284026370

AUCtau of Glasdegib in Participants Receiving Glasdegib and Decitabine at Phase 1B on Cycle 1/Day 10 and Cycle 2/Day 1

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 2, 4, 6 and 24 hours post-dose on Cycle 1/Day 10; pre-dose, 0.5, 1, 2, 6 and 24 hours post-dose on Cycle 2/Day 1

,
Interventionng*hr/mL (Geometric Mean)
Cycle 1/Day 10Cycle 2/Day 1
Phase 1B: Glasdegib 100 mg + DecitabineNA17060
Phase 1B: Glasdegib 200 mg + Decitabine28380NA

Baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 2 Unfit

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. The data of analytes for which the serum level showed statistically significant correlation with clinical response are reported. (NCT01546038)
Timeframe: Baseline (Cycle 1/Day 1 pre-dose)

,
Interventionpg/mL (Median)
BDNFICAM-1 (Intercellular cell adhesion molecule-1)6CKINEBAFF (B-cell activating factor)MIP-3βEotaxin-1 (C-C motif chemokine 11)
Phase 2 Unfit: Glasdegib 100 mg + LDAC (Biomarker, Responder)2000128000223.50704.50275.00169.00
Phase 2 Unfit: Glasdegib 100 mg+LDAC(Biomarker, Non-Responder)900161000318.001295.00414.500.00

Baseline mRNA Levels Associated With Best Overall Response at Phase 2 Unfit

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. Whole blood mRNA analyses were performed on 21 mRNA candidates. Baseline mRNA level showing statistically significant correlation with clinical response are reported. FOXM1: Forkhead box M1; PTCH1: Patched 1. (NCT01546038)
Timeframe: Baseline (Cycle 1/Day 1 pre-dose)

,
InterventionNormalized expression units (Median)
FOXM1PTCH1
Phase 2 Unfit: Glasdegib 100 mg + LDAC (Biomarker, Responder)0.200.20
Phase 2 Unfit: Glasdegib 100 mg+LDAC(Biomarker, Non-Responder)0.400.10

Cmax of Daunorubicin and Daunorubicinol in Participants Receiving Glasdegib and Cytarabine/Daunorubicin at Phase 1B on Induction Cycle 1/Day 3

Daunorubicinol is the major metabolite of daunorubicin, which has anti-neoplastic activity. Cmax values of daunorubicin and daunorubicinol are reported. (NCT01546038)
Timeframe: Pre-dose, 0.25, 0.5, 1, 4, 6, 24 hours post administration of daunorubicin on Induction Cycle 1/Day 3

,
Interventionng/mL (Geometric Mean)
DaunorubicinDaunorubicinol
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin275.3195.4
Phase 1B: Glasdegib 200 mg + Cytarabine/Daunorubicin341.0233.4

Cmax of Decitabine in Participants Receiving Glasdegib and Decitabine at Phase 1B on Cycle 1/Day 1 and Cycle 1/Day 2

(NCT01546038)
Timeframe: Pre-dose, 0.5 hour from start of infusion, 1 hour (at end of infusion) and 2, 3 and 4 hours from start of infusion on Cycle 1/Day 1 and Cycle 1/Day 2

,
Interventionng/mL (Geometric Mean)
Cycle 1/Day 1Cycle 1/Day 2
Phase 1B: Glasdegib 100 mg + Decitabine113.4127.9
Phase 1B: Glasdegib 200 mg + Decitabine174.2121.7

Cmax of Glasdegib in Participants Receiving Glasdegib and Cytarabine/Daunorubicin at Phase 1B on Induction Cycle 1/Day 3 and Day 10

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 6 and 24 hours post-dose on Induction Cycle 1/Day 3; pre-dose, 0.5, 1, 4, 6 and 24 hours post-dose on Induction Cycle 1/Day 10

,
Interventionng/mL (Geometric Mean)
Induction Cycle 1/Day 3Induction Cycle 1/Day 10
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin674.21135
Phase 1B: Glasdegib 200 mg + Cytarabine/Daunorubicin16222371

Cmax of Glasdegib in Participants Receiving Glasdegib and Decitabine at Phase 1B on Cycle 1/Day 10 and Cycle 2/Day 1

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 2, 4, 6 and 24 hours post-dose on Cycle 1/Day 10; pre-dose, 0.5, 1, 2, 6 and 24 hours post-dose on Cycle 2/Day 1

,
Interventionng/mL (Geometric Mean)
Cycle 1/Day 10Cycle 2/Day 1
Phase 1B: Glasdegib 100 mg + Decitabine17181826
Phase 1B: Glasdegib 200 mg + Decitabine2381NA

Cmax of LDAC and Ara-U in Participants Receiving Glasdegib and LDAC at Phase 1B on Cycle 1/Day 2 and Cycle 1/Day 10

Ara-U is the major metabolite of cytarabine. LDAC (low dose cytarabine) is rapidly degraded to the stable metabolite Ara-U, Cmax levels of both LDAC and Ara-U were reported. (NCT01546038)
Timeframe: Pre-dose, 0.25, 0.5, 1, 2, 4 and 6 hours post-dose on Cycle 1/Day 2 and Cycle 1/Day 10

,
Interventionng/mL (Geometric Mean)
LDAC Cycle 1/Day 2LDAC Cycle 1/Day 10Ara-U Cycle 1/Day 2Ara-U Cycle 1/Day 10
Phase 1B: Glasdegib 100 mg + LDAC58.5063.01379.5452.2
Phase 1B: Glasdegib 200 mg + LDAC100.1132.5569.7652.0

Maximum Observed Plasma Concentration (Cmax) of Glasdegib in Participants Receiving Glasdegib and LDAC at Phase 1B on Cycle 1/Day 10 and Cycle 1/Day 21

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 2, 4, 6 and 24 hours post-dose on Cycle 1/Day 10 and Cycle 1/Day 21

,
Interventionng/mL (Geometric Mean)
Cycle 1/Day 10Cycle 1/Day 21
Phase 1B: Glasdegib 100 mg + LDAC10741242
Phase 1B: Glasdegib 200 mg + LDAC19422577

Number of Participants With Corrected QT Interval Using Fridericia's Formula (QTcF) Values Meeting Predefined Criteria at Phase 1B

Maximum absolute values and increases from baseline were summarized for QTcF interval (time from the beginning of Q wave to the end of T wave corresponding to electrical systole corrected for heart rate using Fridericia's formula). Number of participants with QTcF meeting the following criteria is presented: QTcF interval:<450 msec; QTcF interval: 450 to <480 msec; QTcF interval: 480 to <500 msec; QTcF interval >=500 msec; QTcF interval increase from baseline: <30 msec; QTcF interval increase from baseline: 30 to <60 msec; QTcF interval increase from baseline >=60 msec. Arms in the time frame description are defined as: Arm A, Glasdegib +LDAC; Arm B, Glasdegib + Decitabine; Arm C, Glasdegib + Cytarabine/Daunorubicin. End of treatment in the time frame were defined as: maximum of 12 cycles from start of therapy or until disease progression or relapse, participant refusal or unacceptable toxicity occurred, whichever came first. (NCT01546038)
Timeframe: 1 year

,,
InterventionParticipants (Number)
QTcF interval increase < 30 msecQTcF interval increase: 30 to < 60 msecQTcF interval increase >= 60 msecMaximum QTcF interval < 450 msecMaximum QTcF interval: 450 to < 480 msecMaximum QTcF interval: 480 to < 500 msecMaximum QTcF interval >= 500 msec
Phase 1B: Glasdegib + Cytarabine/Daunorubicin1462101011
Phase 1B: Glasdegib + Decitabine2324201
Phase 1B: Glasdegib + LDAC1650101100

Number of Participants With Corrected QT Interval Using Fridericia's Formula (QTcF) Values Meeting Predefined Criteria at Phase 2 Fit and Unfit

Maximum absolute values and increases from baseline were summarized for QTcF interval (time from the beginning of Q wave to the end of T wave corresponding to electrical systole corrected for heart rate using Fridericia's formula). Number of participants with QTcF meeting the following criteria is presented:QTcF interval:<450 msec; QTcF interval: 450 to <480 msec; QTcF interval: 480 to <500 msec; QTcF interval >=500 msec; QTcF interval increase from baseline: <30 msec; QTcF interval increase from baseline: 30 to <60 msec; QTcF interval increase from baseline >=60 msec. End of treatment in the time frame were defined as: maximum of 12 cycles from start of therapy or until disease progression or relapse, participant refusal or unacceptable toxicity occurred, whichever came first. (NCT01546038)
Timeframe: 1 year

,,
InterventionParticipants (Number)
QTcF interval increase < 30 msecQTcF interval increase: 30 to < 60 msecQTcF interval increase >= 60 msecMaximum QTcF interval < 450 msecMaximum QTcF interval: 450 to < 480 msecMaximum QTcF interval: 480 to < 500 msecMaximum QTcF interval >= 500 msec
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin41216461831
Phase 2 Unfit: Glasdegib 100 mg + LDAC60194462935
Phase 2 Unfit: LDAC Alone12418432

Number of Participants With Disease-related Gene Mutations at Phase 1B

Peripheral blood and bone marrow aspirate were collected for baseline mutational analyses. Genetic abnormalities frequently associated with AML were analyzed. These genetic abnormalities included known mutations in the genes NPM1, CEBPA, FLT3, RUNX1, IDH1, IDH2, KIT, K Ras, N Ras and WT1. Additional genes with mutations known to be associated with AML and MDS such as TET2 and DNMT3A were also evaluated. (NCT01546038)
Timeframe: Baseline (Cycle 1/Day 1 pre-dose for Glasdegib + LDAC and Glasdegib + Decitabine Arms; Induction Cycle 1/Day -3 pre-dose for Glasdegib +Cytarabine/Daunorubicin Arm)

,,,
InterventionParticipants (Number)
CEBPA (CCAAT/enhancer-binding protein alpha)DNMT3A (DNA [cytosine-5]-methyltransferase 3A)FLT3 (Fms-like tyrosine kinase 3)FLT3-ITD (FLT3 internal tandem duplications)IDH1 (Isocitrate dehydrogenase 1)IDH2 (Isocitrate dehydrogenase 2)KIT(Tyrosine-protein kinase Kit)KRAS(Kirsten rat sarcoma 2 viral oncogene homolog)NPM1 (Nucleophosmin)NRAS(Neuroblastoma RAS viral oncogene homolog)RUNX1 (Runt related transcription factor 1)TET2 (Tet methylcytosine dioxygenase 2)WT1 (Wilm's tumour tumor suppressor gene1)
Phase 1B: Glasdegib + Cytarabine/Dauno (Biomarker, Responder)2021020041110
Phase 1B: Glasdegib + Decitabine (Biomaker,Non-Responder)0000001000000
Phase 1B: Glasdegib + LDAC (Biomarker, Non-Responder)3210100105130
Phase 1B: Glasdegib+Cytarabine/Dauno(Biomarker,Non-Responder)0000000000000

Number of Participants With Disease-related Gene Mutations at Phase 2 Fit and Unfit

Peripheral blood and bone marrow aspirate were collected for baseline mutational analyses. Genetic abnormalities frequently associated with AML were analyzed. These genetic abnormalities included known mutations in the genes NPM1, CEBPA, FLT3, RUNX1, IDH1, IDH2, KIT, K Ras, N Ras and WT1. Additional genes with mutations known to be associated with AML and MDS such as TET2 and DNMT3A were also evaluated. (NCT01546038)
Timeframe: Baseline (Induction Cycle 1/Day -3 pre-dose for Phase 2 Fit; Cycle 1/Day 1 pre-dose for Phase 2 Unfit)

,,,,,
InterventionParticipants (Number)
CEBPADNMT3AFLT3FLT3-ITDIDH1IDH2KITKRASNPM1NRASRUNX1TET2WT1
Phase 2 Fit (Biomarker, Responder)612322520125770
Phase 2 Fit (Biomarker,Non-Responder)3621141131751
Phase 2 Unfit: Glasdegib 100 mg + LDAC (Biomarker, Responder)32115210211071
Phase 2 Unfit: Glasdegib 100 mg+LDAC(Biomarker, Non-Responder)5134251022341882
Phase 2 Unfit: LDAC Alone (Biomarker, Non-Responder)3602251213781
Phase 2 Unfit: LDAC Alone (Biomarker, Responder)0000000000010

Number of Participants With Treatment-emergent Adverse Events (AEs) at Phase 1B (All Causality)

An adverse event (AE) was any untoward medical occurrence in a clinical investigation participant administered a product or medical device; the event did not necessarily had a causal relationship with the treatment or usage. Treatment Emergent AEs were those with initial onset or increasing in severity after the first dose of study medication and occurred within 28 days post last dose. AEs were graded by the investigator according to the Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 : Grade 1: mild AE; Grade 2: moderate AE; Grade 3: severe AE; Grade 4: life-threatening consequences, urgent intervention indicated; Grade 5: death related to AE. (NCT01546038)
Timeframe: 4 years

,,
InterventionParticipants (Number)
Grade 1 AEsGrade 2 AEsGrade 3 AEsGrade 4 AEsGrade 5 AEsMissing or unknown AEs
Phase 1B: Glasdegib + Cytarabine/Daunorubicin0381010
Phase 1B: Glasdegib + Decitabine101410
Phase 1B: Glasdegib + LDAC1231070

Number of Participants With Treatment-emergent AEs at Phase 1B (Treatment-related)

An adverse event (AE) was any untoward medical occurrence in a clinical investigation participant administered a product or medical device; the event did not necessarily had a causal relationship with the treatment or usage. Treatment Emergent AEs were those with initial onset or increasing in severity after the first dose of study medication and occurred within 28 days post last dose. Treatment-related AEs were AEs related to glasdegib and/or backbone chemotherapy. AEs were graded by the investigator according to the Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 : Grade 1: mild AE; Grade 2: moderate AE; Grade 3: severe AE; Grade 4: life-threatening consequences, urgent intervention indicated; Grade 5: death related to AE. (NCT01546038)
Timeframe: 4 years

,,
InterventionParticipants (Number)
Grade 1 AEsGrade 2 AEsGrade 3 AEsGrade 4 AEsGrade 5 AEsMissing or unknown AEs
Phase 1B: Glasdegib + Cytarabine/Daunorubicin2731000
Phase 1B: Glasdegib + Decitabine200400
Phase 1B: Glasdegib + LDAC327630

Number of Participants With Treatment-emergent AEs at Phase 2 Fit and Unfit (All Causality)

An adverse event (AE) was any untoward medical occurrence in a clinical investigation participant administered a product or medical device; the event did not necessarily had a causal relationship with the treatment or usage. Treatment Emergent AEs were those with initial onset or increasing in severity after the first dose of study medication and occurred within 28 days post last dose. AEs were graded by the investigator according to the Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 : Grade 1: mild AE; Grade 2: moderate AE; Grade 3: severe AE; Grade 4: life-threatening consequences, urgent intervention indicated; Grade 5: death related to AE. (NCT01546038)
Timeframe: 4 years

,,
InterventionParticipants (Number)
Grade 1 AEsGrade 2 AEsGrade 3 AEsGrade 4 AEsGrade 5 AEsMissing or unknown AEs
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin01115250
Phase 2 Unfit: Glasdegib 100 mg + LDAC241539240
Phase 2 Unfit: LDAC Alone01815170

Number of Participants With Treatment-emergent AEs at Phase 2 Fit and Unfit (Treatment-related)

An adverse event (AE) was any untoward medical occurrence in a clinical investigation participant administered a product or medical device; the event did not necessarily had a causal relationship with the treatment or usage. Treatment Emergent AEs were those with initial onset or increasing in severity after the first dose of study medication and occurred within 28 days post last dose. Treatment-related AEs were AEs related to glasdegib and/or backbone chemotherapy. AEs were graded by the investigator according to the Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 : Grade 1: mild AE; Grade 2: moderate AE; Grade 3: severe AE; Grade 4: life-threatening consequences, urgent intervention indicated; Grade 5: death related to AE. (NCT01546038)
Timeframe: 4 years

,,
InterventionParticipants (Number)
Grade 1 AEsGrade 2 AEsGrade 3 AEsGrade 4 AEsGrade 5 AEsMissing or unknown AEs
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin04154610
Phase 2 Unfit: Glasdegib 100 mg + LDAC49203410
Phase 2 Unfit: LDAC Alone4631010

Number of Participants With Treatment-emergent AEs Categorized by Seriousness at Phase 1B

An adverse event (AE) was any untoward medical occurrence in a clinical investigation participant administered a product or medical device; the event did not necessarily had a causal relationship with the treatment or usage. Treatment Emergent AEs were those with initial onset or increasing in severity after the first dose of study medication and occurred within 28 days post last dose. An serious adverse event (SAE) was any untoward medical occurrence at any dose that: resulted in death; was life threatening (immediate risk of death); required inpatient hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity (substantial disruption of the ability to conduct normal life functions); resulted in congenital anomaly/birth defect. (NCT01546038)
Timeframe: 4 years

,,,,,
InterventionParticipants (Number)
AEsSAEs
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin1610
Phase 1B: Glasdegib 100 mg + Decitabine44
Phase 1B: Glasdegib 100 mg + LDAC1713
Phase 1B: Glasdegib 200 mg + Cytarabine/Daunorubicin63
Phase 1B: Glasdegib 200 mg + Decitabine32
Phase 1B: Glasdegib 200 mg + LDAC65

Number of Participants With Treatment-emergent AEs Categorized by Seriousness at Phase 2 Fit and Unfit

An adverse event (AE) was any untoward medical occurrence in a clinical investigation participant administered a product or medical device; the event did not necessarily had a causal relationship with the treatment or usage. Treatment Emergent AEs were those with initial onset or increasing in severity after the first dose of study medication and occurred within 28 days post last dose. An serious adverse event (SAE) was any untoward medical occurrence at any dose that: resulted in death; was life threatening (immediate risk of death); required inpatient hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity (substantial disruption of the ability to conduct normal life functions); resulted in congenital anomaly/birth defect. (NCT01546038)
Timeframe: 4 years

,,
InterventionParticipants (Number)
AEsSAEs
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin6935
Phase 2 Unfit: Glasdegib 100 mg + LDAC8468
Phase 2 Unfit: LDAC Alone4132

Overall Survival (OS) at Phase 2 Fit

OS was defined as duration from the date of randomization to the date of death from any cause. Kaplan-Meier (KM) method was used to estimate median OS. In this method, every participant had a follow-up time which was associated with an indicator, 1=event (death in our case), and 0 =censored. If the participants were not known to have died, time to date of last known to be alive was used as to calculate the follow-up time and indicator was 0 for these participants. KM method estimates the median OS based on the K-M curve. The K-M curve only drops when we had an event and censor data are the ticks in the graph. To estimate median OS, the K-M curve usually will be smoothed first and a line will be drawn at 50%. The median OS is the point when K-M curve and the horizontal hit. Survival status was collected every month for the first 2 months after discontinuation of study treatment and thereafter every 2 months until death or 4 years from each participant's first dose. (NCT01546038)
Timeframe: First dose to Follow-up (4 years)

InterventionMonths (Median)
Total participantsParticipants >= 55 years oldParticipants < 55 years old
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin14.914.7NA

Percentage of Participants With Complete Response (CR) at Phase 2 Fit

For AML participants:CR were those with repeat bone marrow showing <5% myeloblasts,spicules present and no Auer rods, peripheral blood showing neutrophils>=1000/mcL and platelets>=100,000/mcL, transfusion independent and no extramedullary disease. For MDS participants:CR were those with repeat bone marrow showing <=5% myeloblasts, peripheral blood showing neutrophils>=1000/mcL, platelets>=100,000/mcL, 0% blast and hemoglobin (Hgb)>= 11 g/dL, normal maturation of all cell lines. (NCT01546038)
Timeframe: 4 years

InterventionPercentage of participants (Number)
Total participantsParticipants >= 55 years oldParticipants < 55 years old
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin42.036.777.8

Percentage of Participants With Disease-specific Efficacy for Acute Myeloid Leukemia (AML) at Phase 2 Fit and Unfit

AML participants,disease specific efficacy measures included:CRi;Morphologic Leukemia Free State(MLFS)(bone marrow<5%myeloblasts with spicules and no blasts with auer rods,neutrophils<1000/mcL and platelets<100,000/mcL);partial remission(PR)(bone marrow myeloblasts decrease to 5-25&>=50%decrease from start, neutrophils>=1000/mcL, platelets>=100,000/mcL);PR with incomplete blood count recovery(PRi)(bone marrow myeloblasts decrease to 5-25&>=50%decrease from start,neutrophils<1000/mcL or platelets<100,000/mcL);minor response(MR)(bone marrow myeloblasts decrease to>=25% from start);stable disease(SD)(bone marrow myeloblasts stable+/-25% from screening value);cytogenetic complete response(CRc)(bone marrow<5%myeloblasts, neutrophils>1000/mcL, platelets>100,000/mcL and normal cytogenetics),molecular complete response(CRm)(bone marrow<5%myeloblasts, neutrophils>1000/mcL, platelets>100,000/mcL and molecular-negative). (NCT01546038)
Timeframe: 4 years

,,
InterventionPercentage of participants (Number)
CRiMLFSPRPRiMRSDCRcCRm
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin10.97.81.61.610.96.335.937.5
Phase 2 Unfit: Glasdegib 100 mg + LDAC5.12.66.41.36.416.711.516.7
Phase 2 Unfit: LDAC Alone2.60.02.60.010.521.10.02.6

Percentage of Participants With Disease-specific Efficacy for Myelodysplastic Syndrome (MDS) at Phase 2 Fit and Unfit

For all MDS participants, disease specific efficacy measures included: CRi (bone marrow showing <5% myeloblasts with platelets <100,000/mcL or neutrophils <1000/mcL, including confirmed and unconfirmed responses); PR (repeat bone marrow myeloblasts showing decreased by >= 50% decrease but still >5%, peripheral blood showing neutrophils >= 1,000/mcL, platelets >= 100,000/mcL and Hgb>=11g/dL; including confirmed and unconfirmed responses); SD (including confirmed and unconfirmed responses, failure to achieve PR and no evidence of progression for >8 weeks); marrow complete response (mCR) (bone marrow showing <=5% myeloblasts and decreased by >= 50%), partial cytogenetic response (>=50% reduction of chromosomal abnormality) and complete cytogenetic response (CRc) (disappearance of chromosomal abnormality with no appearance of now ones). (NCT01546038)
Timeframe: 4 years

,,
InterventionPercentage of participants (Number)
mCRPRSDCRiUnconfirmed SDUnconfirmed CRimCR (CRi not included)CRc
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin0.00.00.020.00.00.00.060.0
Phase 2 Unfit: Glasdegib 100 mg + LDAC10.00.00.010.010.010.010.010.0
Phase 2 Unfit: LDAC Alone0.00.033.30.00.00.00.00.0

Post-baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 2 Fit - End of Treatment

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. Selected data of analytes for which the serum level showed statistically significant correlation with clinical response are reported. (NCT01546038)
Timeframe: End of Treatment (maximum of 12 cycles from start of therapy or until disease progression or relapse, participant refusal or unacceptable toxicity occurred, whichever came first, an average of 1 year)

,
Interventionpg/mL (Median)
IL-1βIL-15 (Interleukin-15)
Phase 2 Fit (Biomarker, Non-Responder)6.70600
Phase 2 Fit (Biomarker, Responder)9.70700

Post-baseline Levels of Serum Circulating Protein Analytes Associated With Best Overall Response at Phase 2 Unfit - Cycle 1/Day 1

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. A total of 38 proteins were analyzed. Selected data of analytes for which the serum level showed statistically significant correlation with clinical response are reported. (NCT01546038)
Timeframe: Cycle 1/Day 1, 1 Hour Post-dose

,
Interventionpg/mL (Median)
Factor VII:activated blood coagulation factor VIIIL-6 (Interleukin-6)
Phase 2 Unfit: Glasdegib 100 mg + LDAC (Biomarker, Responder)3115000.00
Phase 2 Unfit: Glasdegib 100 mg+LDAC(Biomarker, Non-Responder)2345006.80

Ratios of mRNA Levels to Baseline Associated With Best Overall Response at Phase 2 Fit

Responders were AML participants who achieved CR, CRi, MLFS, PR or PRi based on investigator-reported best overall response and MDS participants who achieved CR, mCR, PR or SD based on investigator-reported best overall response. Whole blood mRNA analyses were performed on 21 mRNA candidates. Ratios of mRNA level to baseline showing statistically significant correlation with clinical response are reported. (NCT01546038)
Timeframe: Baseline (Induction Cycle 1/Day -3 pre-dose); End of Treatment (maximum of 12 cycles from start of therapy or until disease progression or relapse, participant refusal or unacceptable toxicity occurred, whichever came first, an average of 1 year)

,
Interventionratio (Median)
CCNE1MSI2
Phase 2 Fit (Biomarker, Non-Responder)1.100.50
Phase 2 Fit (Biomarker, Responder)0.600.90

Ratios of mRNA Levels to Baseline at Phase 2 Fit - End of Treatment

Whole blood mRNA analyses were performed on 21 mRNA candidates. Selected values showing statistically significant differences compared with baseline are reported here. CCND2:G1/S-Specific Cyclin D2; MSI2: Musashi RNA Binding Protein 2; PTCH2: Patched 2. (NCT01546038)
Timeframe: Baseline (Induction Cycle 1/Day -3 pre-dose); End of Treatment (maximum of 12 cycles from start of therapy or until disease progression or relapse, participant refusal or unacceptable toxicity occurred, whichever came first, an average of 1 year)

Interventionratio (Median)
CCND2MSI2PTCH2
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin0.800.800.70

Ratios of mRNA Levels to Baseline at Phase 2 Fit - Induction Cycle 1/Day 3

Whole blood mRNA analyses were performed on 21 mRNA candidates. Values showing statistically significant, ≥2-fold differences compared with baseline are reported here. CDKN1A: cyclin-dependent kinase inhibitor 1A; SMO: mRNA encoding the glasdegib target Smoothened; PTCH2: Patched 2; MYCN: Neuroblastoma Myc oncogene. (NCT01546038)
Timeframe: Baseline (Induction Cycle 1/Day -3 pre-dose); Induction Cycle 1/Day 3, 1 Hour Post dose

Interventionratio (Median)
CDKN1ASMOPTCH2MYCN
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin2.404.800.600.20

Ratios of mRNA Levels to Baseline at Phase 2 Unfit - End of Treatment

Whole blood mRNA analyses were performed on 21 mRNA candidates. Only the analytes showing statistically significant change from baseline are reported here. (NCT01546038)
Timeframe: Baseline (Cycle 1/Day 1 pre-dose); End of Treatment (maximum of 12 cycles from start of therapy or until disease progression or relapse, participant refusal or unacceptable toxicity occurred, whichever came first, an average of 1 year)

Interventionratio (Median)
CCND2SMOCCND1
Phase 2 Unfit: Glasdegib 100 mg + LDAC0.700.400.40

Serum Levels of Circulating Protein Analytes at Phase 1B - Baseline

Serum levels were determined for 38 circulating proteins. Values showing statistically significant, ≥2-fold difference compared with baseline are reported here. (NCT01546038)
Timeframe: Baseline (Induction Cycle 1/Day -3 pre-dose)

Interventionpg/mL (Median)
MMP-3 (Matrix metalloproteinase-3)IL-8 (Interleukin-8)BDNF (Brain-derived neurotrophic factor)IL-5 (Interleukin-5)VEGF (Vascular endothelial growth factor)MCP-1 (Monocyte chemotactic protein-1)ITAC:Interferon-inducible T-cell α chemoattractant
Phase 1B: Glasdegib + Cytarabine/Daunorubicin1020010.712000.0088.00180.50.00

Serum Levels of Circulating Protein Analytes at Phase 1B - Induction Cycle 1/Day 10

Serum levels were determined for 38 circulating proteins. Values showing statistically significant, ≥2-fold difference compared with baseline are reported here. (NCT01546038)
Timeframe: Induction Cycle 1/Day 10, 1 Hour Post dose

Interventionpg/mL (Median)
IL-8BDNFIL-5VEGFMCP-1ITAC
Phase 1B: Glasdegib + Cytarabine/Daunorubicin37.0020099.0051.00684.000.00

Serum Levels of Circulating Protein Analytes at Phase 2 Fit - Consolidation Cycle 1/Day 1

Serum levels were determined for 38 circulating proteins. Selected values showing statistically significant difference compared with baseline are reported here. (NCT01546038)
Timeframe: Consolidation Cycle 1/Day 1, 1 Hour Post dose

Interventionpg/mL (Median)
MIP-1β (Macrophage Inflammatory Protein-1β)BDNFVEGFIL-8ITAC
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin226.007000232.509.9041.50

Serum Levels of Circulating Protein Analytes at Phase 2 Fit - Consolidation Cycle 1/Day 10

Serum levels were determined for 38 circulating proteins. Selected values showing statistically significant difference compared with baseline are reported here. (NCT01546038)
Timeframe: Consolidation Cycle 1/Day 10, Pre-dose

Interventionpg/mL (Median)
MIP-1βMCP-1MMP-3IL-8ITAC
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin239.50581.001200011.004.10

Serum Levels of Circulating Protein Analytes at Phase 2 Fit - End of Treatment

Serum levels were determined for 38 circulating proteins. Selected values showing statistically significant difference compared with baseline are reported here. (NCT01546038)
Timeframe: End of Treatment (maximum of 12 cycles from start of therapy or until disease progression or relapse, participant refusal or unacceptable toxicity occurred, whichever came first, an average of 1 year)

Interventionpg/mL (Median)
MIP-1βVEGFMCP-1
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin338.00133.00277.00

Serum Levels of Circulating Protein Analytes at Phase 2 Fit - Induction Cycle 1/Day 10

Serum levels were determined for 38 circulating proteins. Selected values showing statistically significant difference compared with baseline are reported here. (NCT01546038)
Timeframe: Induction Cycle 1/Day 10, 1 Hour Post dose

Interventionpg/mL (Median)
IL-1β (Interleukin-1β)IL-6Factor VIIBDNFVEGFMCP-1MMP-3IL-8IL-5ITAC
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin8.5017.0029250030069.00594.001200055.0085.000.00

Serum Levels of Circulating Protein Analytes at Phase 2 Fit - Induction Cycle 1/Day 3

Serum levels were determined for 38 circulating proteins. Selected values showing statistically significant difference compared with baseline are reported here. (NCT01546038)
Timeframe: Induction Cycle 1/Day 3, 1 Hour Post dose

Interventionpg/mL (Median)
Factor VII(activated blood coagulation factor VII)BDNFMMP-3IL-8ITAC
Phase 2 Fit: Glasdegib 100 mg + Cytarabine/Daunorubicin3180007002100028.0014.00

Serum Levels of Circulating Protein Analytes at Phase 2 Unfit - Cycle 1/Day 10

Serum levels were determined for 38 circulating proteins. Selected values showing statistically significant differences compared with baseline are reported here. ITAC (Interferon-inducible T-cell α chemoattractant) level in LDAC alone arm at Cycle 1/Day 10 exhibited non-significant change from baseline but similar trends as in Glasdegib 100 mg+LDAC arm. (NCT01546038)
Timeframe: Cycle 1/Day 10, Pre-dose

,
Interventionpg/mL (Median)
BDNFITAC
Phase 2 Unfit: Glasdegib 100 mg + LDAC5007.5
Phase 2 Unfit: LDAC Alone2000.00

Time to Cmax (Tmax) of Glasdegib in Participants Receiving Glasdegib and LDAC at Phase 1B on Cycle 1/Day 10 and Cycle 1/Day 21

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 2, 4, 6 and 24 hours post-dose on Cycle 1/Day 10 and Cycle 1/Day 21

,
InterventionHours (Median)
Cycle 1/Day 10Cycle 1/Day 21
Phase 1B: Glasdegib 100 mg + LDAC1.751.34
Phase 1B: Glasdegib 200 mg + LDAC4.004.00

Tmax of Daunorubicin and Daunorubicinol in Participants Receiving Glasdegib and Cytarabine/Daunorubicin at Phase 1B on Induction Cycle 1/Day 3

Daunorubicinol is the major metabolite of daunorubicin, which has anti-neoplastic activity. Tmax values of daunorubicin and daunorubicinol are reported. (NCT01546038)
Timeframe: Pre-dose, 0.25, 0.5, 1, 4, 6, 24 hours post administration of daunorubicin on Induction Cycle 1/Day 3

,
InterventionHours (Median)
DaunorubicinDaunorubicinol
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin0.5001.00
Phase 1B: Glasdegib 200 mg + Cytarabine/Daunorubicin0.4920.642

Tmax of Decitabine in Participants Receiving Glasdegib and Decitabine at Phase 1B on Cycle 1/Day 1 and Cycle 1/Day 2

(NCT01546038)
Timeframe: Pre-dose, 0.5 hour from start of infusion, 1 hour (at end of infusion) and 2, 3 and 4 hours from start of infusion on Cycle 1/Day 1 and Cycle 1/Day 2

,
InterventionHours (Median)
Cycle 1/Day 1Cycle 1/Day 2
Phase 1B: Glasdegib 100 mg + Decitabine0.750.58
Phase 1B: Glasdegib 200 mg + Decitabine0.530.53

Tmax of Glasdegib in Participants Receiving Glasdegib and Cytarabine/Daunorubicin at Phase 1B on Induction Cycle 1/Day 3 and Day 10

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 6 and 24 hours post-dose on Induction Cycle 1/Day 3; pre-dose, 0.5, 1, 4, 6 and 24 hours post-dose on Induction Cycle 1/Day 10

,
InterventionHours (Median)
Induction Cycle 1/Day 3Induction Cycle 1/Day 10
Phase 1B: Glasdegib 100 mg + Cytarabine/Daunorubicin5.994.08
Phase 1B: Glasdegib 200 mg + Cytarabine/Daunorubicin6.001.04

Tmax of Glasdegib in Participants Receiving Glasdegib and Decitabine at Phase 1B on Cycle 1/Day 10 and Cycle 2/Day 1

(NCT01546038)
Timeframe: Pre-dose, 0.5, 1, 2, 4, 6 and 24 hours post-dose on Cycle 1/Day 10; pre-dose, 0.5, 1, 2, 6 and 24 hours post-dose on Cycle 2/Day 1

,
InterventionHours (Median)
Cycle 1/Day 10Cycle 2/Day 1
Phase 1B: Glasdegib 100 mg + Decitabine2.001.03
Phase 1B: Glasdegib 200 mg + Decitabine2.05NA

Tmax of LDAC and Ara-U in Participants Receiving Glasdegib and LDAC at Phase 1B on Cycle 1/Day 2 and Cycle 1/Day 10

Ara-U is the major metabolite of cytarabine. LDAC (low dose cytarabine) is rapidly degraded to the stable metabolite Ara-U, Tmax levels of both LDAC and Ara-U were reported. (NCT01546038)
Timeframe: Pre-dose, 0.25, 0.5, 1, 2, 4 and 6 hours post-dose on Cycle 1/Day 2 and Cycle 1/Day 10

,
InterventionHours (Median)
LDAC Cycle 1/Day 2LDAC Cycle 1/Day 10Ara-U Cycle 1/Day 2Ara-U Cycle 1/Day 10
Phase 1B: Glasdegib 100 mg + LDAC0.2500.3253.972.00
Phase 1B: Glasdegib 200 mg + LDAC0.2500.2504.001.99

Disease-Free Survival (DFS)

Estimate the DFS at one-year post-transplantation. The event is defined as relapse or death due to relapse. The number of participants who did not relapse up to one year post transplant is reported. (NCT01621477)
Timeframe: one year post transplant

Interventionparticipants (Number)
Treatment7

Event-Free Survival (EFS)

Estimate the EFS at one-year post-transplantation. The event is defined as relapse or death due to any cause. The number of participants who were alive without relapse at one year post-transplant is reported. (NCT01621477)
Timeframe: one year post transplant

Interventionparticipants (Number)
Treatment4

Incidence of Malignant Relapse

Estimate the incidence of malignant relapse at one year post-transplant. The number of participants with malignant relapse or progressive disease is given. Relapse was evaluated using standard WHO criteria for each disease. (NCT01621477)
Timeframe: One year post transplantation.

Interventionparticipants (Number)
Treatment10

Number of Participants With Transplant Related Mortality (TRM)

The number of participants who died due to TRM in the first 100 days post-transplant is given. (NCT01621477)
Timeframe: 100 days post transplant

Interventionparticipants (Number)
Treatment2

One-year Survival (OS)

Evaluate the number of participants alive at 1 year. The number of participants surviving to one-year post-transplantation is given. (NCT01621477)
Timeframe: One year post transplant

Interventionparticipants (Number)
Treatment7

Incidence and Severity of Acute Graft Versus Host Disease (GVHD)

The number of participants with acute GVHD is given, organized by grade. Participants are graded on a scale from 1 to 4, with 1 being mild and 4 being severe. (NCT01621477)
Timeframe: 100 days post transplant

Interventionparticipants (Number)
No Acute GVHDGrade IGrade IIGrade IIIGrade IV
Treatment93131

Incidence and Severity of Chronic Graft Versus Host Disease (GVHD)

The severity of chronic GVHD will be described. Chronic GVHD was evaluated using NIH Consensus Global Severity Scoring. The number of participants with chronic GVHD is given, organized by severity. (NCT01621477)
Timeframe: 100 days post transplant

Interventionparticipants (Number)
No Chronic GVHDMildModerateSevere
Treatment15020

Overall Survival

(NCT00992602)
Timeframe: Time from start of therapy until death, assessed up to 4 years

Interventionmonths (Median)
Treatment (Liposomal Cytarabine, High-dose Methotrexate)8.2

Survival Free of Neurological Progression, Measured in Weeks

Neurological progression defined by either clinical impression (measured by Karnofsky Performance Status), radiographical response (using Macdonald criteria), or cytologic response (measured by CSF cytology). (NCT00992602)
Timeframe: Time from start of therapy, assessed up to 4 years

Interventionweeks (Median)
Treatment (Liposomal Cytarabine, High-dose Methotrexate)6

Minimum Residual Disease (MRD) Levels Present at End of Cycle 1 Therapy in Patients

"MRD positive is defined as > or = to 0.1% MRD MRD negative is define as < 0.1% MRD~All these analyses will be descriptive and exploratory and hypotheses generating in nature." (NCT01614197)
Timeframe: Cycle 1 (a minimum of 4 weeks and a max of 8 weeks)

,,,
InterventionParticipants (Count of Participants)
MRD positiveMRD negative
Dose Level 130
Dose Level 221
Dose Level 351
Dose Level 421

Maximum Tolerated Dose

The maximum tolerated dose of bismuth Bi 213 monoclonal antibody M195 following cytarabine in patients with advanced myeloid malignancies. (NCT00014495)
Timeframe: 2 years

InterventionmCi/kg (Number)
Bismuth Bi 213 Monoclonal Antibody M195 & Cytarabine1

Overall Response Rate (ORR = Complete Response (CR) + Partial Response (PR)) of Subjects

-For definitions of CR and PR please refer to the Recommendations for Initial Evaluation, Staging and Response Assessment of Hodgkin and Non-Hodgkin Lymphoma: The Lugano Classification (NCT03623373)
Timeframe: Through completion of treatment (estimated to be 6 months)

InterventionParticipants (Count of Participants)
Bendamustine/Rituximab/Acalabrutinib/Cytarabine10

Pre-transplant Complete Response Rate

-For definitions of CR, please refer to the Recommendations for Initial Evaluation, Staging and Response Assessment of Hodgkin and Non-Hodgkin Lymphoma: The Lugano Classification (NCT03623373)
Timeframe: Through completion of treatment (estimated to be 6 months)

InterventionParticipants (Count of Participants)
Bendamustine/Rituximab/Acalabrutinib/Cytarabine9

Stem Cell Mobilization Success Rate With Cytarabine and Rituximab

-Stem cell mobilization success is defined as a yield of >2x10^6 CD34+ stem cells/kg with a maximum of 5 courses of apheresis (NCT03623373)
Timeframe: Through 5 courses of apheresis (up to 5 days)

InterventionParticipants (Count of Participants)
Bendamustine/Rituximab/Acalabrutinib/Cytarabine4

Safety and Tolerability of Regimen in Subjects With MCL as Measured by Treatment Related Non-hematologic Toxicity of Grade 3 or Higher

-Toxicity is measured using NCI Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 (NCT03623373)
Timeframe: 30 days following completion of treatment (estimated to be 7 months)

InterventionParticipants (Count of Participants)
DiarrheaInfusion related reactionUpper respiratory infectionSkin infectionPerirectal abscessPeritoneal infectionBlood bilirubin increasedEjection fraction decreasedAlanine aminotransferase increased
Bendamustine/Rituximab/Acalabrutinib/Cytarabine111111111

The Rate of Complete Remission (CR+CRi)

CR requires: 1. peripheral blood counts: neutrophil count ≥ 1.0 x 10^9/L, platelet count ≥ 100 x 10^9/L, reduced hemoglobin concentration or hematocrit has no bearing on remission status, and leukemic blasts must not be present in the peripheral blood. 2. bone marrow aspirate and biopsy: maturation of all cell lines must be present, ≤ 5% blasts, auer rods must not be detectable. 3. extramedullary leukemia, such as central nervous system (CNS) or soft tissue involvement, must not be present. CRi requires that all criteria for complete remission be satisfied except patients can have residual neutropenia (<1 x 10^9/L) or thrombocytopenia (<100 x 10^9/L). (NCT00634244)
Timeframe: Assessed every 3 months for the first 2 years and then every 6 months until relapse or death up to 3 years from registration.

Interventionproportion of participants (Number)
Arm A (Carboplatin+Topotecan Hydrochloride)0.143
Arm B (Alvocidib+Cytarabine+Mitoxantrone)0.278
Arm C (Sirolimus+Mitoxantrone+Etoposide+Cytarabine)0.15

The Rate of Treatment Failure

"The definition of treatment failure will include:~≥ 5% leukemic blasts at the time of pre-consolidation marrow~Death during/following induction chemotherapy (pre-consolidation)~Persisting marrow hypoplasia and pancytopenia for ≥ 2 months after chemotherapy~CNS or extramedullary disease at the time of pre-consolidation~Leukemia persistence after completion of induction treatment. Leukemia persistence is defined as greater than 10% residual blasts on marrow biopsy done 5-7 days after completion of induction chemotherapy" (NCT00634244)
Timeframe: Assessed every 3 months for the first 2 years and then every 6 months until relapse or death up to 3 years from registration.

Interventionproportion of participants (Number)
Arm A (Carboplatin+Topotecan Hydrochloride)0.86
Arm B (Alvocidib+Cytarabine+Mitoxantrone)0.72
Arm C (Sirolimus+Mitoxantrone+Etoposide+Cytarabine)0.84

Achievement of Complete Remission (CR) at Reinduction

Disease response assessed after chemotherapy from bone marrow aspirates/biopsies and complete blood count. (NCT00939653)
Timeframe: Between Days 22-36 or on Day 43 and weekly thereafter if peripheral counts haven't recovered

InterventionParticipants (Count of Participants)
Enrolled Patients1

Death

Number of participants who died. (NCT00939653)
Timeframe: From the first dose of study therapy until 30 days after last therapy dose

InterventionParticipants (Count of Participants)
Enrolled Patients4

Complete Remission Rate (CR + CRi)

"Morphologic complete remission (CR): neutrophil count > 1.0 x 109 /L, platelet count ≥ 100 x 109/L, < 5% bone marrow blasts by morphologic review, no Auer rods, no evidence of extramedullary disease. (No requirements for marrow cellularity, hemoglobin concentration).~Morphologic complete remission with incomplete blood count recovery (CRi): same as CR but ANC may be <1000/mcl or platelet count <100,000/mcl~Participants in the phase I portion of the study, treated at the MTD will count towards the phase II accrual goal for evaluation of the primary endpoint." (NCT02416908)
Timeframe: Median follow-up of 34 days

InterventionParticipants (Count of Participants)
Phase I Schedule A and Phase II18

Duration of Remission

-Duration of remission (DOR): Defined as the interval from the date complete remission is documented to the date of recurrence. (NCT02416908)
Timeframe: Up to 2 years

Interventionmonths (Median)
Phase I Schedule A and Phase II9.1

Event-free Survival

Event-free survival (EFS): Defined as the interval from the date of first dose of study drug to date of treatment failure including progressive disease, recurrence, or discontinuation for any reason (including toxicity, patient preference, initiation of new treatment without documented progression, or death due to any cause). (NCT02416908)
Timeframe: Up to 2 years (median follow-up of 307 days)

Interventionmonths (Median)
Phase I Schedule A and Phase II6.1

Number of Participants Who Were Able to Undergo Hematopoietic Stem Cell Transplantation

Allogeneic stem cell transplant utilization: the number of patients proceeding to allogeneic transplant within 2 months following end of study without any additional salvage therapy following study treatment. (NCT02416908)
Timeframe: Up to 2 years (median follow-up of 307 days)

InterventionParticipants (Count of Participants)
Phase I Schedule A and Phase II24

Overall Survival

Overall survival (OS): Defined as the date of first dose of study drug to the date of death from any cause. OS will be evaluated at 3 month intervals for at least 12 months and up to a maximum of 2 years. (NCT02416908)
Timeframe: Up to 2 years (median follow-up of 307 days)

Interventionmonths (Median)
Phase I Schedule A and Phase II7.8

Relapse-free Survival

Relapse-free survival (RFS): For patients achieving a complete remission, defined as the interval from the date of first documentation of a leukemia free state to date of recurrence or death due to any cause. (NCT02416908)
Timeframe: Median follow-up of 307 days

Interventiondays (Median)
Phase I Schedule A and Phase II152

Time to Neutrophil Engraftment

-Time to neutrophil engraftment: Defined as the date of the first dose of study drug to the date that the absolute neutrophil count is >1,000/mm3 (NCT02416908)
Timeframe: Up to 2 years

Interventiondays (Median)
Phase I Schedule A and Phase II28

Time to Platelet Engraftment

-Time to platelet engraftment: Defined as the date of the first dose of study drug to the date that the platelet count is >100,000/mm^3 in the absence of platelet transfusions. (NCT02416908)
Timeframe: 56 days

Interventiondays (Median)
Phase I Schedule A and Phase II38

Safety and Tolerability of Treatment as Measured by Incidence of Grade 3-4 Adverse Events Occurring in >5% of Participants

-All adverse events will be classified using the descriptions and grading scales found in the revised NCI Common Terminology Criteria for Adverse Events (CTCAE) v4.0 (NCT02416908)
Timeframe: From start of treatment until 30 days following last day of study treatment or until the start of a subsequent treatment for AML, whichever came first (41 days)

InterventionParticipants (Count of Participants)
Lymphocyte count decreasedWhite blood cell decreasedHypophosphatemiaPlatelet count decreasedNeutrophil count decreasedHyponatremiaAnemiaHyperglycemiaSkin infectionFebrile neutropeniaSepsisHypokalemiaLung infectionAlanine aminotransferase increasedOral thrushHypoxiaHypertensionDiarrheaNauseaEdema limbsCatheter-related infectionHematuriaRespiratory failure
Phase I Schedule A and Phase II32282622211814111088875444333333

Number of Participants With Complete Response

Complete Response (CR) is required bone marrow blasts ≤5% and recovery of normal hematopoiesis with an absolute neutrophil count (ANC) of 1*10^9/L or more and platelet count of 100*10^9/L or more; and a complete response without platelets (CRp) is the same criteria as CR but with platelet counts from 20*10^9/L to less than 100*10^9/L. (NCT00096122)
Timeframe: 21 Day Cycle

InterventionParticipants (Number)
CRCRp
Idarubicin, Cytarabine + Tipifarnib619

The Rate of Major Cytogenetic Response at 6 Months

Cytogenetic response is defined in terms of the percentage of Philadelphia (Ph) chromosome. Major cytogenetic response is defined as 0-34% Ph-positive cells. (NCT00022490)
Timeframe: 6 months

InterventionParticipants (Number)
Cytarabine/ Imatinib Mesylate5

2-year Disease-free Survival (DFS)

Measured from data of randomization to post-consolidation therapy until relapse from complete response or death from any cause, with observations censored at the date of last contact for patients last known to be alive without report of relapse. (NCT00085709)
Timeframe: After completing any treatment, every 6 months for 2 years, than annually for years 3-5

InterventionPercentage of population (Number)
Post-consolidation GO39
Post-consolidation Observation50

Complete Remission

(NCT00085709)
Timeframe: After induction therapy was completed (1 or 2 months)

Interventionparticipants (Number)
Ara-C+Daunomycin203
Ara-C+Daunomycin+Mylotarg207

Toxicity

Number of patients with Grade 3-5 adverse events that are related to study drug by given type of adverse event (NCT00085709)
Timeframe: For induction, daily for the first 10 days, then twice weekly until consolidation treatment. Weekly during consolidation treatment. Weekly if randomized to post-consolidation G.O.

,,,
InterventionParticipants with a given type of AE (Number)
ALT, SGPT (serum glutamic pyruvic transaminase)AST, SGOT (serum glut. oxaloacetic transaminase)Acidosis (metabolic or respiratory)Adult respiratory distress syndrome (ARDS)Albumin, serum-low (hypoalbuminemia)Alkaline phosphataseAlkalosis (metabolic or respiratory)Allergic reaction/hypersens. (inc drug fever)AmylaseAnorexiaApneaAtaxia (incoordination)Bicarbonate, serum-lowBilirubin (hyperbilirubinemia)Blood/Bone Marrow-Other (Specify)Bronchospasm, wheezingCalcium, serum-low (hypocalcemia)Cardiac Arrhythmia-Other (Specify)Cardiac General-Other (Specify)Cardiac troponin I (cTnI)Cardiac troponin T (cTnT)Cardiac-ischemia/infarctionColitisColitis, infectious (e.g., Clostridium difficile)ConfusionConstipationCoughCreatinineDIC (disseminated intravascular coagulation)Death not assoc with CTCAE term-Multi-organ failuDehydrationDiarrheaDistention/bloating, abdominalDizzinessDysphagia (difficulty swallowing)Dyspnea (shortness of breath)Edema, larynxEdema: limbEdema: visceraEnteritis (inflammation of the small bowel)EsophagitisFatigue (asthenia, lethargy, malaise)Febrile neutropeniaFever (in the absence of neutropenia)Fistula, GI - RectumFistula, GU - VaginaFlu-like syndromeGGT (gamma-glutamyl transpeptidase)Glucose, serum-high (hyperglycemia)Glucose, serum-low (hypoglycemia)HematomaHemoglobinHemolysisHemorrhage, CNSHemorrhage, GI - Abdomen NOSHemorrhage, GI - JejunumHemorrhage, GI - Lower GI NOSHemorrhage, GI - Oral cavityHemorrhage, GI - RectumHemorrhage, GU - BladderHemorrhage, GU - KidneyHemorrhage, GU - Urinary NOSHemorrhage, GU - UterusHemorrhage, GU - VaginaHemorrhage, pulmonary/upper respiratory - LungHemorrhage, pulmonary/upper respiratory - NoseHemorrhage/Bleeding-Other (Specify)Hepatobiliary/Pancreas-Other (Specify)Hiccoughs (hiccups, singultus)HypertensionHypotensionHypoxiaInfec with Grade 3 or 4 neut - Abdomen NOSInfec with Grade 3 or 4 neut - Anal/perianalInfec with Grade 3 or 4 neut - AppendixInfec with Grade 3 or 4 neut - Bladder (urinInfec with Grade 3 or 4 neut - BloodInfec with Grade 3 or 4 neut - Bone (osteomyInfec with Grade 3 or 4 neut - Brain + SpinaInfec with Grade 3 or 4 neut - BronchusInfec with Grade 3 or 4 neut - Catheter-relaInfec with Grade 3 or 4 neut - CecumInfec with Grade 3 or 4 neut - ColonInfec with Grade 3 or 4 neut - Dental-toothInfec with Grade 3 or 4 neut - Heart (endocaInfec with Grade 3 or 4 neut - Lip/perioralInfec with Grade 3 or 4 neut - LiverInfec with Grade 3 or 4 neut - Lung (pneumonInfec with Grade 3 or 4 neut - Meninges (menInfect with Grade 3 or 4 neut - MucosaInfec with Grade 3 or 4 neut - Nerve-peripheInfec with Grade 3 or 4 neut - Oral cavity-gInfec with Grade 3 or 4 neut - Pelvis NOSInfec with Grade 3 or 4 neut- PharynxInfec with Grade 3 or 4 neut - RectumInfec with Grade 3 or 4 neut - SinusInfec with Grade 3 or 4 neut - Skin (celluliInfec with Grade 3 or 4 neut - Small bowel NInfec with Grade 3 or 4 neut - Upper airwayInfection with Grade 3 or 4 neut - Urinary tractInfec with Grade 3 or 4 neut - VulvaInfec with Grade 3 or 4 neut - WoundInfec with normal ANC or Grade 1/2 neut - BloodInfec with norm ANC or Gr 1/2 neut, Catheter-relInfec with norm ANC or Grade 1/2 neut - ColonInfec with norm ANC or Gr 1/2 neutr- Dental-toothInfec with norm ANC or Gr 1/2 neut - HeartInfec with norm ANC or Gr 1/2 neutrophils - LiverInfec with norm ANC or Gr 1/2 neut-Lung (pneumoni)Infec with norm ANC or Gr 1/2 neut - MuscleInf with norm ANC or Gr 1/2 neut-Oral cavity-gumsInfec with norm ANC or Gr 1/2 neut - ScrotumInfec with norm ANC or Gr 1/2 neut-SkinInfec with norm ANC or Gr 1/2 neut-Urinary tractInfection with unknown ANC - BloodInfection with unknown ANC - Catheter-relatedInfection with unknown ANC - Dental-toothInfection with unknown ANC - JointInfection with unknown ANC - LiverInfection with unknown ANC - Lung (pneumonia)Infection with unknown ANC - MucosaInfection with unknown ANC - SinusInfection with unknown ANC - Skin (cellulitis)Infection with unknown ANC - Urinary tract NOSInfection with unknown ANC - WoundInfection-Other (Specify)Left ventricular diastolic dysfunctionLeft ventricular systolic dysfunctionLeukocytes (total WBC)Leukoencephalopathy (radiolographic findings)Liver dysfunction/failure (clinical)LymphopeniaMagnesium, serum-low (hypomagnesemia)Metabolic/Laboratory-Other (Specify)Mood alteration - anxietyMood alteration - depressionMucositis/stomatitis (clinical exam) - EsophagusMucositis/stomatitis (clinical exam) - Large bowelMucositis/stomatitis (clinical exam) - LarynxMucositis/stomatitis (clinical exam) - Oral cavityMucositis/stomatitis (clinical exam) - PharynxMucositis/stomatitis (func/sympt) - EsophagusMucositis/stomatitis (func/sympt)- Oral cavityMucositis/stomatitis (func/sympt) - PharynxMuscle weakness (func/sym, Whole body/generalizedNasal cavity/paranasal sinus reactionsNauseaNecrosis, GI - RectumNeurology-Other (Specify)Neuropathy: motorNeuropathy: sensoryNeutrophils/granulocytes (ANC/AGC)NystagmusObstruction/stenosis of airway - TracheaOcular/Visual-Other (Specify)Opportunistic infec assoc with Gr 2 lymphopeniaPTT (Partial thromboplastin time)Pain - Abdomen NOSPain - AnusPain - BackPain - BonePain - Chest/thorax NOSPain - Extremity-limbPain - EyePain - Head/headachePain - JointPain - MusclePain - Oral-gumsPain - RectumPain - Throat/pharynx/larynxPain-Other (Specify)Pancreatic endocrine: glucose intolerancePerforation, GI - ColonPericardial effusion (non-malignant)PericarditisPetechiae/purpuraPhosphate, serum-low (hypophosphatemia)PlateletsPleural effusion (non-malignant)Pneumonitis/pulmonary infiltratesPortal vein flowPotassium, serum-high (hyperkalemia)Potassium, serum-low (hypokalemia)ProctitisPulmonary/Upper Respiratory-Other (Specify)Rash/desquamationRenal failureRigors/chillsSeromaSerum sicknessSodium, serum-high (hypernatremia)Sodium, serum-low (hyponatremia)Somnolence/depressed level of consciousnessAtrial fibrillationSinus tachycardiaSupraventricular arrhythmia NOSSyncope (fainting)Syndromes-Other (Specify)Thrombosis/embolism (vascular access-related)Thrombosis/thrombus/embolismThrombotic microangiopathyTumor lysis syndromeTyphlitis (cecal inflammation)Uric acid, serum-high (hyperuricemia)Vasovagal episodeVentricular arrhythmia - Ventricular fibrillationVision-blurred visionVomiting
Ara-C Consolidation22172232001212042031003414001000141301001000221831600141501144100012000001041200246040447101131230012211020102910610641010900100710212103004121511050141000020011207112119301001311421180100000000362263500190110100040000421400111005
Ara-C+Daunomycin1111001021001300011107021016101002000190016100032519680000120187011000100021015013120211250116022101270203011331060001000000100131100201311401960123150111111229210130100110101205000110100101001002913011021510960000150100301412400015
Ara-C+Daunomycin+Mylotarg2932271531601400114011300002283103210502110101112417861101221110105010112110153830258100230000505111022011210024016010001016100200300000141040110604211310000620410170101111000008010200810000110116101451324210168010281011100402310102
Post-consolidation G.O.22000000000000000000000000000010000000000422100002002000000000000000000100000400020000000000001001000000000000000000000000000000041004000000000000001000043000100110010100010000001140000030000001000000000000000001

Number of Patients With Dose-Limiting Toxicity (DLT)

Treatment related toxicities that preclude proceeding to HSCT by day 56 of the treatment course. (NCT01158885)
Timeframe: Beginning with the first dose of investigational product until day 56 of treatment course, an average of 1 year

InterventionParticipants (Count of Participants)
Single Arm0

AUC (0-inf, ss)(Area Under the Concentration-time Curve of BI 811283 in Plasma Over the Time Interval From 0 Extrapolated to Infinity) at Steady State

AUC (0-inf, ss)(area under the concentration-time curve of BI 811283 in plasma over the time interval from 0 extrapolated to infinity) at steady state during Cycle 1 (NCT00632749)
Timeframe: -0.05 hours before and 1:00, 4:00, 6:00, 24:00, 25:00, 26:00, 28:00, 32:00, 36:00, 48:00 hours after administration of BI 811283

Interventionnmol·h/L (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A188
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A615
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule ANA
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A4460
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A5140
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A4750

AUC (0-inf) (Area Under the Concentration-time Curve of Cytarabine in Plasma Over the Time Interval From 0 Extrapolated to Infinity)

AUC (0-inf) of Cytarabine after a 20 mg Subcutaneous Dose on the First Day of BI 811283 (NCT00632749)
Timeframe: -0.05 hours before and 0:30, 1:00, 1:30, 2:00, 3:00, 4:00, 6:00 hours after administration of Cytarabine

Interventionng·h/mL (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A104
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A122
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A102
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A85.4
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A65.9
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A68.1
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B62.0
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule BNA
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule BNA
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B71.2
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B72.7
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B75.5
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B76.9
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B76.8

AUC (0-tz,ss) (Area Under the Concentration-time Curve of BI 811283 in Plasma Over the Time Interval From 0 to the Time of the Last Quantifiable Data Point) at Steady State

AUC (0-tz,ss) (area under the concentration-time curve of BI 811283 in plasma over the time interval from 0 to the time of the last quantifiable data point) at steady state during Cycle 1 (NCT00632749)
Timeframe: -0.05 hours before and 1:00, 4:00, 6:00, 24:00, 25:00, 26:00, 28:00, 32:00, 36:00, 48:00 hours after administration of BI 811283

Interventionnmol·h/L (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A183
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A597
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A1620
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A4330
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A4340
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A4820

AUC (0-tz) (Area Under the Concentration-time Curve of Cytarabine in Plasma Over the Time Interval From 0 to the Time of the Last Quantifiable Data Point)

AUC (0-tz) of Cytarabine after a 20 mg Subcutaneous Dose on the First Day of BI 811283 (NCT00632749)
Timeframe: -0.05 hours before and 0:30, 1:00, 1:30, 2:00, 3:00, 4:00, 6:00 hours after administration of Cytarabine

Interventionng·h/L (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A45.0
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A66.9
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A66.8
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A71.8
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A46.6
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A42.3
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B45.9
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B33.9
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B57.0
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B60.2
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B52.5
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B52.1
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B41.3
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B51.6

AUC(0-inf) (Area Under the Concentration-time Curve of BI 811283 in Plasma Over the Time Interval From 0 Extrapolated to Infinity)

AUC(0-inf) (area under the concentration-time curve of BI 811283 in plasma over the time interval from 0 extrapolated to infinity) during Cycle 1 (NCT00632749)
Timeframe: -0.05 hours before and 1:00, 4:00, 6:00, 24:00, 25:00, 26:00, 28:00, 32:00, 36:00, 48:00 hours after administration of BI 811283

Interventionnmol·h/L (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A177
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A985
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A1000
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A2790
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A4400
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A5620
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B192
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1490
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B2840
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B8870
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B9770
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B14100
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B12800
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B23200

AUC0-tz (Area Under the Concentration-time Curve of BI 811283 in Plasma Over the Time Interval From 0 to the Time of the Last Quantifiable Data Point)

AUC0-tz (area under the concentration-time curve of BI 811283 in plasma over the time interval from 0 to the time of the last quantifiable data point) during Cycle 1 (NCT00632749)
Timeframe: -0.05 hours before and 1:00, 4:00, 6:00, 24:00, 25:00, 26:00, 28:00, 32:00, 36:00, 48:00 hours after administration of BI 811283

Interventionnmol·h/L (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A169
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A939
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A2010
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A2350
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A2810
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A5340
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B156
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1450
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B2730
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B8680
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B9260
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B13700
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B12100
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B21700

Cmax (Maximum Measured Concentration of BI 811283 in Plasma)

Cmax (maximum measured concentration of BI 811283 in plasma) during Cycle 1 (NCT00632749)
Timeframe: -0.05 hours before and 1:00, 4:00, 6:00, 24:00, 25:00, 26:00, 28:00, 32:00, 36:00, 48:00 hours after administration of BI 811283

Interventionnmol/L (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A8.03
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A64.10
30 mg BI 811283 + 20 mg Cytarabine- Treatment Schedule A104.00
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A153.00
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A214.00
120mg (BI 811283+ Cytarabine)- Treatment Schedule A272.00
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B7.06
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B66.10
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B139.00
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B499.00
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B445.00
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B631.00
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B528.00
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1130.00

Cmax (Maximum Measured Concentration of Cytarabine in Plasma)

Cmax of Cytarabine after a 20 mg Subcutaneous Dose on the First Day of BI 811283 (NCT00632749)
Timeframe: -0.05 hours before and 0:30, 1:00, 1:30, 2:00, 3:00, 4:00, 6:00 hours after administration of Cytarabine

Interventionng/mL (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A53.2
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A72.6
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A49.0
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A64.9
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A61.9
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A46.0
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B46.3
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B49.8
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B55.0
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B65.1
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B49.4
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B48.6
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B49.0
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B68.7

Cmax,ss (Maximum Measured Concentration of BI 811283 in Plasma at Steady State)

Cmax (maximum measured concentration of BI 811283 in plasma at steady state) during Cycle 1 (NCT00632749)
Timeframe: -0.05 hours before and 1:00, 4:00, 6:00, 24:00, 25:00, 26:00, 28:00, 32:00, 36:00, 48:00 hours after administration of BI 811283

Interventionnmol/L (Geometric Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A8.02
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A24.7
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A43.3
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A141
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A213
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A229

Event Free Survival (EFS)

EFS was defined as the duration of time from randomisation to time of treatment failure (i.e. PD), relapse from CR, or death from any cause, whichever came first. (NCT00632749)
Timeframe: Data collected up to cut-off date 20 Oct 2011, Up to 1239 days

Interventiondays (Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A35.0
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A122.3
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A32.0
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A117.5
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A60.4
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A62.0
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B209.0
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B65.0
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B168.8
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B62.7
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B138.7
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B48.5
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B174.0
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B30.0

Incidence of Dose Limiting Toxicity (DLT)

Number of participants with DLT in the first cycle (28 days) for the determination of the maximum tolerated dose (MTD) (NCT00632749)
Timeframe: up to 28 days of treatment

Interventionparticipants (Number)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A2
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0

Overall Survival (OS)

OS was defined for all patients that entered the trial, and measured from the date of randomization until death from any cause. (NCT00632749)
Timeframe: Data collected up to cut-off date 20 Oct 2011, Up to 1239 days

Interventiondays (Mean)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A122.8
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A212.0
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A71.3
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A236.8
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A148.7
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A86.6
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B236.8
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B137.3
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B198.2
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B339.7
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B175.7
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B73.0
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B294.7
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B48.7

Partial Remission

"Response to treatment was evaluated according to the following criteria (modified from the National Cancer Institute/Cancer and Leukemia Group B criteria; The best overall response was defined as the best overall response recorded during the time period from the start of the treatment until the end of the treatment period, progression or death (whichever was earlier). Possible categories for best overall response were CR, CRi, Partial remission (PR), no change (NC), Progressive disease (PD) and no assessment.~Partial remission (PR). All of the criteria for CR had to be met, except that the bone marrow had to contain ≥ 5% but less than 25% blasts (or ≤ 50% of initial blast count), or < 5% blasts in the presence of Auer rods or abnormal morphology." (NCT00632749)
Timeframe: Data collected up to cut-off date 20 Oct 2011, Up to 1239 days

Interventionparticipants (Number)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0

Relapse Free Survival

"Relapse-free survival was defined only for patients who achieved CR/CRi and was measured from the date of attaining CR/CRi until the date of recurrence or death from any cause, whichever occurred first.~Number of patients having relapse free survival are presented." (NCT00632749)
Timeframe: Data collected up to cut-off date 20 Oct 2011, Up to 1239 days

Interventionparticipants (Number)
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A1
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A1
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B1
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B2

Remission Duration

Remission duration analysis was defined only for patients who achieved CR, and was measured from the date of attaining CR until the date of disease recurrence (relapse). For patients who died without report of relapse, remission duration was censored on the date of death, regardless of the cause. (NCT00632749)
Timeframe: Data collected up to cut-off date 20 Oct 2011, Up to 1239 days

Interventiondays (Mean)
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A263.0
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A28.0
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B337.0
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B455.0
240 mg BI 811283+ 20 mg Cytarabine - Treatment Schedule B128.0
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B15.0

The Maximum Tolerated Dose (MTD) of 2 Schedules of BI 811283 in Combination With Cytarabine.

"The MTD was defined as the highest dose at which 6 patients were treated and less than 2 patients who experienced a dose limiting toxicities (DLT) within the first cycle of treatment.The MTD was defined based on safety data from the first cycle only.~It was determined using a standard 3 + 3 design with de-escalation." (NCT00632749)
Timeframe: up to 28 days of treatment

Interventionmg (Number)
Treatment Schedule A100
Treatment Schedule BNA

Tmax (Time From Dosing to Maximum Measured Concentration of BI 811283 in Plasma)

tmax (time from dosing to maximum measured concentration of BI 811283 in plasma) during Cycle 1 (NCT00632749)
Timeframe: -0.05 hours before and 1:00, 4:00, 6:00, 24:00, 25:00, 26:00, 28:00, 32:00, 36:00, 48:00 hours after administration of BI 811283

Interventionhours (Median)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A24.0
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A26.0
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A25.0
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A22.7
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A6.0
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A23.6
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B23.9
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B6.0
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B24.1
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B24.9
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B6.0
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B23.9
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B5.9
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B14.8

Tmax (Time From Dosing to Maximum Measured Concentration of Cytarabine in Plasma)

Tmax of Cytarabine after a 20 mg Subcutaneous Dose on the First Day of BI 811283 (NCT00632749)
Timeframe: -0.05 hours before and 0:30, 1:00, 1:30, 2:00, 3:00, 4:00, 6:00 hours after administration of Cytarabine

Interventionhours (Median)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0.50
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0.50
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0.92
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0.83
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0.48
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A0.55
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0.50
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0.50
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0.58
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0.50
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0.50
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0.50
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0.48
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B0.49

Tmax,ss (Time From Dosing to Maximum Measured Concentration of BI 811283 in Plasma at Steady State)

tmax,ss (time from dosing to maximum measured concentration of BI 811283 in plasma at steady state) during Cycle 1 (NCT00632749)
Timeframe: -0.05 hours before and 1:00, 4:00, 6:00, 24:00, 25:00, 26:00, 28:00, 32:00, 36:00, 48:00 hours after administration of BI 811283

Interventionhours (Median)
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A23.9
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A24.0
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A6.0
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A6.0
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A6.0
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A6.0

Incidence and Intensity of AEs Graded According to CTCAE (Version 3.0)

"The severity and timing of AEs indicates how well the treatment regimen was tolerated.~Toxicities were evaluated using the common terminology criteria for adverse events (CTCAE) grading scheme." (NCT00632749)
Timeframe: Data from first treatment administration until cut-off date of 20 October 2011; up to 1239 days

,,,,,,,,,,,,,
Interventionparticipants (Number)
CTCAE Grade 1CTCAE Grade 2CTCAE Grade 3CTCAE Grade 4CTCAE Grade 5
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00142
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00034
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00120
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00111
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00232
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A01020
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00112
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00021
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00022
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00024
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00103
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00112
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00121
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00032

Response (Complete Remission [CR], Complete Remission With Incomplete Blood Count Recovery [CRi])

"Response to treatment was evaluated according to the following criteria (modified from the National Cancer Institute/Cancer and Leukemia Group B criteria:~The best overall response was defined as the best overall response recorded during the time period from the start of the treatment until the end of the treatment period, progression or death (whichever was earlier). Possible categories for best overall response were CR, CRi, Partial remission (PR), no change (NC), Progressive disease (PD) and no assessment.~Complete remission (CR): morphologically leukaemia free state (i.e. bone marrow with < 5% blasts by morphologic criteria and no Auer rods, no evidence of extramedullary leukaemia) and absolute neutrophil count ≥ 1,000/μL and platelets > 100,000/μL.~Complete remission with incomplete blood count recovery (incomplete CR, CRi).All of the above criteria for CR had to be met, except that neutrophils < 1,000/μL or platelets < 100,000/μL in the blood." (NCT00632749)
Timeframe: Data collected up to cut-off date 20Oct2011, Up to 1239 days

,,,,,,,,,,,,,
Interventionparticipants (Number)
CRCRi
100 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00
120 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A10
15 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00
160 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B10
240 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B10
30 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00
300 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00
360 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B11
40 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00
420 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B00
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A00
5 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B10
60 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule A10
80 mg BI 811283 + 20 mg Cytarabine - Treatment Schedule B10

Event-Free Survival (EFS) at 2 Years

Comparison of the event-free survival (EFS) between treatment CIA and FLAI, where an event is defined to be resistance to treatment, relapse (after response) or death, whichever occurred first. (NCT01289457)
Timeframe: Up to 2 years or until relapse/death

InterventionMonths (Median)
Group 1 CIA7.1
Group 2 FLAI8.4

Maximum Tolerated Dose (MTD) of Clofarabine, Idarubicin, and Cytarabine

MTD is highest dose level in which <2 patients of 6 develop first cycle dose limiting toxicities (DLT). Toxicity defined as any treatment-related grade 3 or greater non-hematological toxicities. (NCT01289457)
Timeframe: 28 days

Interventionmg/m^2 (Number)
Clofarabine + Idarubicin + Cytarabine15

Overall Survival

Time from date of treatment start until date of death due to any cause or last Follow-up. (NCT01289457)
Timeframe: up to 2 years

InterventionMonths (Median)
Group 1 CIA14.5
Group 2 FLAI15.1

Response Rates of Clofarabine, Idarubicin, and Cytarabine (CIA) Versus Fludarabine, Idarubicin, and Cytarabine (FLAI)

NCI & Myelodysplastic syndromes (MDS) International Working Group (IWG) Definitions: Complete Response (CR): Neutrophil count ≥1.0 ×10^9/L, Platelet count ≥100 ×10^9/L, Bone marrow aspirate 10% after initial response. Response assessed Day 28 of every 2-3 cycles during treatment. (NCT01289457)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
Group 1 CIA107
Group 2 FLAI76

Event Free Survival

the median time point at which a participants experienced and event or toxicity or progression (NCT00073957)
Timeframe: 12 months

Interventionmonths (Median)
Yttrium Y 90 Ibritumomab2.5

Best Response

This data is the best overall response achieved by patients by the 12 month period. (NCT00073957)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
CRPRSDPD
Yttrium Y 90 Ibritumomab Tiuxetan and Rituximab81115

Response Rate = Complete and Partial Response at 12 Weeks.

Definition Nodal Masses Spleen, Liver Bone Marrow CR Disappearance of all evidence of disease Partial response Regression and no new sites ≥ 50% decrease in sum of the perpendicular dimension of up to 6 largest dominant masses; no increase in size of other nodes Stable disease Failure to attain CR/PR or Progressive disease or Relapsed disease : the appearance of any new lesion or the (a) FDG-avid or PET positive prior to therapy; PET positive at prior sites of disease and no new sites on CT or PET Any new lesion or increase by ≥ 50% of previously involved sites from nadir Appearance of a new lesion(s) > 1.5 cm in any axis, ≥ 50% increase in SPD of more than one node, or ≥ 50% increase in longest diameter of a previously identified node > 1 cm in short axis > 50% increase from nadir in the SPD of any previous lesions New or recurrent involvement Lesions PET positive if FDG-avid lymphoma or PET positive prior to therapy (NCT00073957)
Timeframe: 12 weeks

Interventionparticipants (Number)
CRPRSDPD
Yttrium Y 90 Ibritumomab Tiuxetan and Rituximab53215

Overall Survival

Overall Survival at six years after initiating protocol therapy (NCT00083551)
Timeframe: 6 Years

Interventionpercentage of participants (Number)
Thalidomide65
No Thalidomide58

Overall Survival (OS)

OS is measured from the start of on-study to the date of death or to the last date of follow-up. Measurement is determined by Kaplan-Meyer estimate. The probability of survival at 5 years after diagnosis is given. (NCT00186875)
Timeframe: 2 years after last patient completes therapy (approximately 4 years after enrollment)

Interventionprobability (Mean)
Standard Risk0.654
High Risk0.357

Minimal Residual Disease (MRD) Compared With Historical Data From TOTXV Protocol (NCT00137111)

The prevalence of MRD in children undergoing treatment for relapsed ALL and to compare the results to those obtained in children with newly diagnosed ALL. MRD is considered as positive (i.e., prevalent) if its level is >=0.01%. The prevalence of MRD after Block B is defined as the proportion of MRD positives. (NCT00186875)
Timeframe: End of Block B therapy (Day 19)

,,
InterventionParticipants (Count of Participants)
Negative <0.01%Positive ≥0.01%
High Risk210
Standard Risk1111
TOTXV Participants191297

Minimal Residual Disease (MRD) Compared With Historical Data From TOTXV Protocol (NCT00137111)

The prevalence of MRD in children undergoing treatment for relapsed ALL and to compare the results to those obtained in children with newly diagnosed ALL. MRD is considered as positive (i.e., prevalent) if its level is >=0.01%. The prevalence of MRD after Block C is defined as the proportion of MRD positives. (NCT00186875)
Timeframe: End of Block Block C therapy (Day 46)

,,
InterventionParticipants (Count of Participants)
Negative <0.01%Positive ≥0.01%
High Risk18
Standard Risk119
TOTXV Participants390102

Response Rate

"The response rate is defined as the proportion of participants who attain morphological complete remission after the re-induction Block C, inclusive of all patients who begin re-induction. Morphological complete remission was defined as <5% blasts in bone marrow by morphology." (NCT00186875)
Timeframe: End of re-induction Block C (approximately 1 month after the start of therapy)

,
Interventionproportion of participants (Number)
Complete remissionFailure to reach complete remission
High Risk0.7860.214
Standard Risk0.8460.154

Number of Participants With Complete Response (CR)

Complete Response Criteria (CR must last for at least 4 weeks): Marrow: /= 11 g/dL (untransfused, patient not on EPO); Neutrophils >/= 1x109/L (not on myeloid growth factor); Platelets >/= 100 * 109/L (not on thrombopoietic agent); No blasts. Bone marrow aspirate and/or biopsy at the end of course 1 (day 28 +/- 7 days). The method of Thall, Simon, and Estey used to monitor response. (NCT01444742)
Timeframe: 4 weeks after first cycle

InterventionParticipants (Count of Participants)
Clofarabine + Cytarabine19

Overall Survival (OS)

Overall survival defined as the time interval from study entry date to the date of death due to any cause, measured in days/months. Bayesian time-to-event model used to monitor overall survival. (NCT01444742)
Timeframe: 5 years

InterventionMonths (Median)
Clofarabine + Cytarabine10.3

Disease-free Survival

Time from date of treatment start until the date of first objective documentation of disease-relapse. (NCT01692197)
Timeframe: Up to 5 years

Interventionmonths (Median)
E7070 + Idarubicin + Cytarabine1.7

Duration of Response

Response date to loss of response or last follow up. (NCT01692197)
Timeframe: Up to 5 years

Interventionmonths (Median)
E7070 and Idarubicin and Cytarabine6.7

Overall Response

Efficacy measured by overall response - complete response plus complete response with incomplete platelet recovery, plus partial response (Complete remission (CR) + Complete remission without platelet recovery (CRp) + Partial Remission (PR)+ marrow clearance of blast) during cycle 1. (NCT01692197)
Timeframe: 2 cycles (60 days)

InterventionParticipants (Count of Participants)
E7070 and Idarubicin and Cytarabine11

Overall Survival

Time from date of treatment start until date of death due to any cause or last follow-up. (NCT01692197)
Timeframe: Up to 5 years

Interventionmonths (Median)
E7070 and Idarubicin and Cytarabine5.1

Procedure or Procedure Related Adverse Events

Number of unique patients who had a procedure or treatment related (possible, probable or definite) adverse events. (graded per NCI CTC v4.0) (NCT02485353)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Vosaroxin and Cytarabine1

Rate of Complete Remission

Percentage of patients who have complete remission as defined by the International Working Group for AML: morphologic complete remission (CR) or morphologic complete remission with incomplete blood count recovery (CRi or CRp) (NCT02485353)
Timeframe: 2 months

Interventionpercentage of patients (Number)
Vosaroxin and Cytarabine0

Duration of Exposure of Entospletinib

(NCT02343939)
Timeframe: First dose date up to approximately 3 years

Interventionweeks (Median)
Group A Phase 1b ENTO 200 mg + Cytarabine + Daunorubicin8.6
Group A Phase 1b/2 ENTO 400 mg + Cytarabine + Daunorubicin7.1
Group B Phase 1b ENTO 200 mg + Decitabine13.7
Group B Phase 1b ENTO 400 mg + Decitabine15.4
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)10.1
Group B Phase 2 ENTO 400 mg + Decitabine (Randomized)13.9
Group B Phase 2 ENTO 400 mg + Azacitidine (Randomized)10.1
Group C Phase 1b/2 ENTO 400 mg4.4
Group C Phase 1b ENTO 800 mg7.6

Event Free Survival (EFS)

EFS was defined as the time interval from the start of the study therapy until the date of treatment failure, acute myeloid leukemia (AML) relapse, or death from any cause, whichever occurred first. Participants who received other anti-cancer therapy (prior to the event if any) were censored. Median EFS was analyzed using Kaplan-Meier (KM) method. (NCT02343939)
Timeframe: First dose date up to approximately 38 months

Interventionmonths (Median)
Group A Phase 1b ENTO 200 mg + Cytarabine + DaunorubicinNA
Group A Phase 1b ENTO 400 mg + Cytarabine + Daunorubicin1.9
Group A Phase 2 ENTO 400 mg + Cytarabine + Daunorubicin9.0
Group B Phase 1b ENTO 200 mg + Decitabine2.2
Group B Phase 1b ENTO 400 mg + Decitabine2.9
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)2.3
Group B Phase 2 ENTO 400 mg + Decitabine (Randomized)3.2
Group B Phase 2 ENTO 400 mg + Azacitidine (Randomized)2.4
Group C Phase 1b ENTO 400 mg1.8
Group C Phase 1b ENTO 800 mg1.8
Group C Phase 2 ENTO 400 mg (Cohort C1A - R/R AML)1.0
Group C Phase 2 ENTO 400 mg (Cohort C2 - MLL)1.0
Group C Phase 2 ENTO 400 mg (Cohort C3 - Untreated AML)1.7

Overall Survival (OS)

OS was defined as the time interval from the start of the study therapy to death from any cause. Median OS was analyzed using KM method. (NCT02343939)
Timeframe: First dose date up to approximately 38 months

Interventionmonths (Median)
Group A Phase 1b ENTO 200 mg + Cytarabine + Daunorubicin37.1
Group A Phase 1b ENTO 400 mg + Cytarabine + Daunorubicin34.1
Group A Phase 2 ENTO 400 mg + Cytarabine + DaunorubicinNA
Group B Phase 1b ENTO 200 mg + Decitabine3.2
Group B Phase 1b ENTO 400 mg + Decitabine5.3
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)6.9
Group B Phase 2 ENTO 400 mg + Decitabine (Randomized)7.3
Group B Phase 2 ENTO 400 mg + Azacitidine (Randomized)6.2
Group C Phase 1b ENTO 400 mg5.9
Group C Phase 1b ENTO 800 mg5.6
Group C Phase 2 ENTO 400 mg (Cohort C1A - R/R AML)8.2
Group C Phase 2 ENTO 400 mg (Cohort C2 - MLL)7.9
Group C Phase 2 ENTO 400 mg (Cohort C3 - Untreated AML)2.2

Percentage of Participants Experiencing Dose Limiting Toxicities (DLTs)

DLTs refer to toxicities experienced during the first 28 days of study treatment that have been judged to be clinically significant and related to study treatment. DLT assessment was applicable only for Phase 1b and Phase 2 safety run-in participants. (NCT02343939)
Timeframe: Group A: Cycle 0 Day 1 to Cycle 2 Day 28; Group B: Cycle 0 Day 1 to Cycle 1 Day 28; Group C: Cycle 1 Day 1 to Cycle 1 Day 28 (Cycle length: for Cycle 0 = 14 days, for all other cycles = 28 days)

Interventionpercentage of participants (Number)
Group A Phase 1b ENTO 200 mg + Cytarabine + Daunorubicin0
Group A Phase 1b/2 ENTO 400 mg + Cytarabine + Daunorubicin0
Group B Phase 1b ENTO 200 mg + Decitabine0
Group B Phase 1b ENTO 400 mg + Decitabine16.7
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)0
Group C Phase 1b/2 ENTO 400 mg0
Group C Phase 1b ENTO 800 mg16.7

Percentage of Participants Experiencing Treatment-Emergent Adverse Events

(NCT02343939)
Timeframe: First dose date up to the last dose date plus 30 days (maximum: 18 months)

Interventionpercentage of participants (Number)
Group A Phase 1b ENTO 200 mg + Cytarabine + Daunorubicin100.0
Group A Phase 1b/2 ENTO 400 mg + Cytarabine + Daunorubicin100.0
Group B Phase 1b ENTO 200 mg + Decitabine100.0
Group B Phase 1b ENTO 400 mg + Decitabine100.0
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)100.0
Group B Phase 2 ENTO 400 mg + Decitabine (Randomized)100.0
Group B Phase 2 ENTO 400 mg + Azacitidine (Randomized)100.0
Group C Phase 1b/2 ENTO 400 mg100.0
Group C Phase 1b ENTO 800 mg100.0

Percentage of Participants With Composite Complete Remission at the End of Induction

Clinical response was assessed according to the International Working Group criteria (Cheson 2003). Composite complete remission included CR, CRc, and morphologic complete remission with incomplete blood count recovery (CRi). CR required all of the following: < 5% blasts in bone marrow aspirate; Neutrophils ≥ 1,000/mcL; Platelets ≥ 100,000/mcL; No extramedullary disease; No blasts with Auer rods detected; and Independent of transfusions. CRc, in addition to CR criteria, required reversion to a normal karyotype with an abnormal karyotype at the time of diagnosis. CRi required all of the CR criteria except the criterion of neutrophils and platelets. (NCT02343939)
Timeframe: At the end of induction (Group A: up to end of Cycle 2; Group B: up to end of Cycle 4) (cycle length = up to 28 days); Group C: From Day 1 until meeting the criteria for study treatment discontinuation (up to approximately 3 years)

Interventionpercentage of participants (Number)
Group A Phase 1b ENTO 200 mg + Cytarabine + Daunorubicin100.0
Group A Phase 1b ENTO 400 mg + Cytarabine + Daunorubicin77.8
Group A Phase 2 ENTO 400 mg + Cytarabine + Daunorubicin65.9
Group B Phase 1b ENTO 200 mg + Decitabine40.0
Group B Phase 1b ENTO 400 mg + Decitabine50.0
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)25.0
Group B Phase 2 ENTO 400 mg + Decitabine (Randomized)23.5
Group B Phase 2 ENTO 400 mg + Azacitidine (Randomized)14.3
Group C Phase 1b ENTO 400 mg14.3
Group C Phase 1b ENTO 800 mg0.0
Group C Phase 2 ENTO 400 mg (Cohort C1A - R/R AML)0.0
Group C Phase 2 ENTO 400 mg (Cohort C2 - MLL)15.4
Group C Phase 2 ENTO 400 mg (Cohort C3 - Untreated AML)11.1

Percentage of Participants With Morphologic Complete Remission (CR) at the End of Induction

Clinical response was assessed according to the International Working Group criteria (Cheson 2003). Morphologic CR included CR and cytogenetic CR (CRc). CR required all of the following: < 5% blasts in bone marrow aspirate; Neutrophils ≥ 1,000/microliter (mcL); Platelets ≥ 100,000/mcL; No extramedullary disease; No blasts with Auer rods detected; and Independent of transfusions. CRc, in addition to CR criteria, required reversion to a normal karyotype with an abnormal karyotype at the time of diagnosis. (NCT02343939)
Timeframe: At the end of induction (Group A: up to end of Cycle 2; Group B: up to end of Cycle 4) (cycle length = up to 28 days); Group C: From Day 1 until meeting the criteria for study treatment discontinuation (up to approximately 3 years)

Interventionpercentage of participants (Number)
Group A Phase 1b ENTO 200 mg + Cytarabine + Daunorubicin66.7
Group A Phase 1b ENTO 400 mg + Cytarabine + Daunorubicin66.7
Group A Phase 2 ENTO 400 mg + Cytarabine + Daunorubicin46.3
Group B Phase 1b ENTO 200 mg + Decitabine0.0
Group B Phase 1b ENTO 400 mg + Decitabine16.7
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)25.0
Group B Phase 2 ENTO 400 mg + Decitabine (Randomized)0.0
Group B Phase 2 ENTO 400 mg + Azacitidine (Randomized)7.1
Group C Phase 1b ENTO 400 mg0.0
Group C Phase 1b ENTO 800 mg0.0
Group C Phase 2 ENTO 400 mg (Cohort C1A - R/R AML)0.0
Group C Phase 2 ENTO 400 mg (Cohort C2 - MLL)15.4
Group C Phase 2 ENTO 400 mg (Cohort C3 - Untreated AML)11.1

Percentage of Participants With Overall Response at the End of Induction

Clinical response was assessed according to the International Working Group criteria (Cheson 2003). Overall response included CR, CRc, CRi, and partial remission (PR). CR required all of the following: < 5% blasts in bone marrow aspirate; Neutrophils ≥ 1,000/mcL; Platelets ≥ 100,000/mcL; No extramedullary disease; No blasts with Auer rods detected; and Independent of transfusions. CRc, in addition to CR criteria, required reversion to a normal karyotype with an abnormal karyotype at the time of diagnosis. CRi required all of the CR criteria except the criterion of neutrophils and platelets. PR required all of the following: ≥ 50% decrease in blasts in bone marrow aspirate to a range of 5% to 25%; Neutrophils ≥ 1,000/mcL; Platelets ≥ 100,000/mcL; Independent of transfusions; and A value of ≤ 5% blasts was also considered a PR if Auer rods were detected. (NCT02343939)
Timeframe: At the end of induction (Group A: up to end of Cycle 2; Group B: up to end of Cycle 4) (cycle length = up to 28 days); Group C: From Day 1 until meeting the criteria for study treatment discontinuation (up to approximately 3 years)

Interventionpercentage of participants (Number)
Group A Phase 1b ENTO 200 mg + Cytarabine + Daunorubicin100.0
Group A Phase 1b ENTO 400 mg + Cytarabine + Daunorubicin77.8
Group A Phase 2 ENTO 400 mg + Cytarabine + Daunorubicin70.7
Group B Phase 1b ENTO 200 mg + Decitabine40.0
Group B Phase 1b ENTO 400 mg + Decitabine50.0
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)25.0
Group B Phase 2 ENTO 400 mg + Decitabine (Randomized)23.5
Group B Phase 2 ENTO 400 mg + Azacitidine (Randomized)14.3
Group C Phase 1b ENTO 400 mg14.3
Group C Phase 1b ENTO 800 mg0.0
Group C Phase 2 ENTO 400 mg (Cohort C1A - R/R AML)0.0
Group C Phase 2 ENTO 400 mg (Cohort C2 - MLL)15.4
Group C Phase 2 ENTO 400 mg (Cohort C3 - Untreated AML)11.1

Percentage of Participants Who Experienced Laboratory Abnormalities

Treatment-emergent laboratory abnormalities were defined as values that increase at least one toxicity grade from baseline. The most severe graded abnormality from all tests was counted for each participant. (NCT02343939)
Timeframe: First dose date up to the last dose date plus 30 days (maximum: 18 months)

,,,,,,,,
Interventionpercentage of participants (Number)
Any Laboratory AbnormalityGrade 3 or 4 Laboratory Abnormalities
Group A Phase 1b ENTO 200 mg + Cytarabine + Daunorubicin100100
Group A Phase 1b/2 ENTO 400 mg + Cytarabine + Daunorubicin10098.0
Group B Phase 1b ENTO 200 mg + Decitabine100100
Group B Phase 1b ENTO 400 mg + Decitabine100100
Group B Phase 2 ENTO 400 mg + Azacitidine (Randomized)10092.9
Group B Phase 2 ENTO 400 mg + Azacitidine (Safety Run-In)10085.7
Group B Phase 2 ENTO 400 mg + Decitabine (Randomized)94.194.1
Group C Phase 1b ENTO 800 mg10085.7
Group C Phase 1b/2 ENTO 400 mg10082.9

Complete Remission Rate

Complete remission rate is defined as number of subjects with complete remission according to the IWG criteria, which is defined by presence of <5 percent of blasts in the bone marrow, absence of blasts with Auer rods, absence of extramedullary disease, absolute neutrophil count >1.0 x 109/L (1000/µL); platelet count >100 x 109/L (100,000/µL); independence of red cell transfusions. (NCT02532010)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Arm A: Pacritinib and Decitiabine0
Arm B: Pacritinib and Cytarabine0

Event-free Survival

Time from entry on study until time at which there is treatment failure, AML relapse, or death from any cause (NCT02532010)
Timeframe: Time from entry on study until time at which there is treatment failure, AML relapse, or death from any cause, assessed throughout the study period up to 2 years

Interventionmonths (Median)
Arm A: Pacritinib and Decitiabine2.5
Arm B: Pacritinib and Cytarabine3

Overall Remission Rate

Overall remission rate is defined as number of subjects with complete remission (CR), Complete Remission with incomplete platelet recovery (CRp), Complete Remission with Incomplete Blood Count Recovery (CRi), Partial Remission (PR) according to the IWG criteria. CRi is defined as All CR criteria except for residual neutropenia (<1.0 x 109/L (1000/µL)) or thrombocytopenia (<100 x 109/L (100,000/µL)), PR is defined as relevant in the setting of phase I and II clinical trials only; all hematologic criteria of CR; decrease of bone marrow blast percentage to 5 to 25 percent; and decrease of pretreatment bone marrow blast percentage by at least 50 percent. (NCT02532010)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Arm A: Pacritinib and Decitiabine0
Arm B: Pacritinib and Cytarabine0

Overall Survival

Survival following treatment to the date of death (NCT02532010)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Arm A: Pacritinib and Decitiabine3
Arm B: Pacritinib and Cytarabine3

Relapse-free Survival

Time from complete remission documentation to either AML relapse or death from any cause. (NCT02532010)
Timeframe: From date of complete remission until either AML relapse or death from any cause, whichever came first, assessed throughout the study period up to 2 years

Interventionmonths (Median)
Arm A: Pacritinib and DecitiabineNA
Arm B: Pacritinib and CytarabineNA

Remission Duration

Time from CR documentation to AML relapse (NCT02532010)
Timeframe: time from complete remission to AML relapse, assessed throughout the study period up to 2 years.

Interventionmonths (Median)
Arm A: Pacritinib and DecitiabineNA
Arm B: Pacritinib and CytarabineNA

Time to Complete Response

Time from entry on study until documentation of complete remission (CR) (NCT02532010)
Timeframe: From entry on study until complete remission, assessed throughout the study period up to 2 years

Interventionmonths (Median)
Arm A: Pacritinib and DecitiabineNA
Arm B: Pacritinib and CytarabineNA

Level of Methylation

the percentage of methylated DNA (NCT00882206)
Timeframe: Day 0

Interventionpercentage of DNA (Mean)
Decitabine / Vorinostat84.98

Level of Methylation

the percentage of methylated DNA (NCT00882206)
Timeframe: Day 33

Interventionpercentage of DNA (Mean)
Decitabine / Vorinostat86.1

Level of Methylation

the percentage of methylated DNA (NCT00882206)
Timeframe: Day 5

Interventionpercentage of DNA (Mean)
Decitabine / Vorinostat79.82

Response to Treatment

Response includes both complete remission (defined as <5% leukemic blasts in the bone marrow) and partial remission (defined as a greater than 35% reduction in the bone marrow leukemia blast percentage at day 33) (NCT00882206)
Timeframe: Day 33

Interventionparticipants (Number)
Decitabine / Vorinostat6

Event-free Survival (EFS)

EFS was measured from the date of randomisation to the date of progression or relapse, or death from any cause, whichever occurred first. (NCT01721876)
Timeframe: From randomization until disease progression or relapse or death from any cause, up to 1557 days.

InterventionMonths (Median)
Placebo + Low-dose Cytarabine2.8
Volasertib + Low-dose Cytarabine3.3

Objective Response (OR)

OR is the number of patients who achieved complete remission (CR) or complete remission with incomplete blood count recovery (CRi), where OR was based on the best response attained during the treatment period. (NCT01721876)
Timeframe: Response assessment was performed at the end of every 2nd cycle, (i.e. at the end of Cycle 2, 4, 6, 8, etc., and at end of treatment), i.e. up to 52 months.

InterventionParticipants (Number)
Placebo + Low-dose Cytarabine38
Volasertib + Low-dose Cytarabine123

Overall Survival (OS)

OS is the key secondary endpoint and was measured from the date of randomization until death from any cause. Patients who were lost to follow-up were censored on the last date they were known to be alive. (NCT01721876)
Timeframe: From randomization until death due to any cause, up to 1557 days.

InterventionMonths (Median)
Placebo + Low-dose Cytarabine6.5
Volasertib + Low-dose Cytarabine5.6

Relapse-free Survival (RFS)

RFS was defined only for patients who achieved best overall response of CR or CRi; it was measured from the date of achievement of a remission until the date of relapse or death from any cause. Patients not known to have relapsed or died at last follow-up were censored on the date they were last examined. (NCT01721876)
Timeframe: From randomization until disease progression or relapse or death from any cause, up to 1557 days.

InterventionMonths (Median)
Placebo + Low-dose Cytarabine18.7
Volasertib + Low-dose Cytarabine13.1

Event-free Survival

Event-free survival (EFS) was defined as the time for registration to failure to achieve CR during induction, relapse or death. Participants without events were censored at date of last follow-up. The median EFS with 95% CI was estimated using the Kaplan Meier method. (NCT01253070)
Timeframe: Time from registration to death or relapse (up to 10 years)

Interventionmonths (Median)
ITD Mutated Participants8.8
TKD Mutated Participants7.8

OS

OS was defined as the time from registration to death of any cause. Surviving patients were censored at the date of last follow-up. The median OS with 95% confidence interval (CI) was estimated using the Kaplan Meier method. (NCT01253070)
Timeframe: Time from registration to death (up to 10 years)

Interventionmonths (Median)
ITD Mutated Participants15
TKD Mutated Participants16.2

Overall Survival (OS) Rate

Percentage of patients who were alive at 1 year. The analysis was split between patients with having a FLT3 (FMS-like tyrosine kinase-3) ITD (internal tandem duplication) or TKD (tyrosine kinase domain) mutation. The FLT3 mutation testing at baseline was performed centrally for all patients. (NCT01253070)
Timeframe: 1 year

Interventionpercentage of participants (Number)
ITD Mutated Participants62
TKD Mutated Participants71

Disease-Free Survival Rate at 2-year and 5-year.

Disease-Free Survival (DFS) was calculated from the time of complete remission until relapse or death due to any cause. (NCT00038610)
Timeframe: Baseline to 2-year and 5-year

Interventionpercentage of participants (Number)
2-year DFS rate5-year DFS rate
Hyper-CVAD + Imatinib4943

Overall Survival Rate at 2-year and 5-year.

Overall survival (OS) was calculated from the date of initiation of therapy until death. (NCT00038610)
Timeframe: Baseline to 2-year and 5-year

Interventionpercentage of participants (Number)
2-year OS rate5-year OS rate
Hyper-CVAD + Imatinib5743

Response To Induction Therapy With Hyper-CVAD Plus Imatinib Mesylate

"Complete Remission (CR): Defined as the presence of 5% or less blasts in the bone marrow, with a granulocyte count of 1.0 × 109/L or higher and a platelet count of 100 × 109/L and no extramedullary disease.~Partial Response (PR): As above for CR except for the presence of 6-25% marrow blasts.~Molecular CR: Same as for CR with RT-PCR negativity for bcr-abl.~Induction Death: Defined as death occurring after start of therapy without meeting the definition of CR or resistant disease." (NCT00038610)
Timeframe: Baseline to 6 months

Interventionparticipants (Number)
Complete RemissionPartial RemissionMolecular Complete RemissionInduction Death
Hyper-CVAD + Imatinib421171

Number of Participants With Response

"Participant responses are categorized as 'Complete Remission,' Complete Remission, No Platelet Recovery,' 'No Response.'~Complete Remission: Disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x 109/L and platelet count > 100 x 109/L, and normal bone marrow differential (< 5% blasts); Complete Remission, No Platelet Recovery: Peripheral blood and bone marrow results as for CR, but with platelet counts of < 100 x 109/L.~Blood draws once a week until remission then every 2 to 8 weeks during therapy." (NCT00088218)
Timeframe: Every 2 to 8 weeks

,
InterventionParticipants (Number)
Complete RemissionComplete Remission, No Platelet RecoveryNo Response
Clofarabine5011
Clofarabine Plus Ara-C49426

Number of Participants Who Had an Adverse Event While on Treatment With Clofarabine Plus Cytarabine

Patients will be monitored clinically and diagnostically using measures including blood test, bone marrow aspiration and MUGA. Toxicity assessment every week using the NCI Common Terminology Criteria for Adverse Events (CTCAE) version 3.0 will be performed. (NCT00334074)
Timeframe: Up to five months (includes follow up period of 30 days) from the day patient received their first dose of study drug

Interventionparticipants; with adverse events (Number)
Clofarabine and Cytarabine30

Response Rate (Complete Response [CR] Plus Partial Response [PR]) of Clofarabine Plus Cytarabine in Patients With Relapsed/Refractory AML, Untreated MDS, CML in Blast Phase, or in Selected Untreated Patients With High Risk of Anthracycline Toxicity

"Based on International working group for diagnosis, standardization of response criteria, and treatment outcomes for reporting standards for therapeutic trials in Acute myeloid Leukemia:~Complete Response (CR) was defined as normalization of marrow blasts (< 5%), recovery of normal heamtopoiesis (absolute neutrophil count >1 X 10^9/l, platelet count ≥100 X10^9/l, and absence of peripheral blood blasts, independent of transfusions and growth factor support.~Partial response was defined as blood count recovery as for complete response with the exception of leukemic marrow blasts in the range of 6%-25% or a ≥50% decrease in bone marrow blasts.~Treatment failure was defined as a <25% change in marrow blasts within 30 days of starting therapy" (NCT00334074)
Timeframe: Proportion of confirmed responses was estimated by the number of patients who achieved a CR or PR, defined as two consecutive evaluations at least 4 weeks apart, divided by the number of eligible participants in the study.

Interventionparticipants (Number)
Overall Response RateComplete ResponsePartial ResponseNot RespondingNot evaluable
Clofarabine Plus Cytarabine1614286

Duration of Remission

With 50 patients, the rates of these endpoints will be estimated with a standard error of 5 to 7 percentage points, depending on the observed rates. Remission is defined as less than 5% blasts in the bone marrow, no appearance of blasts in the peripheral blood, and no extramedullary disease (appearance of leukemic cells in other tissues). (NCT01101880)
Timeframe: Up to 5 years

Interventionweeks (Median)
Treatment (Chemotherapy and Colony Stimulating Factor)7

Event Free Survival

Number of patients in remission at a median follow up of 15 months. (NCT01101880)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Treatment (Chemotherapy and Colony Stimulating Factor)21

Overall Survival

With 50 patients, the rates of these endpoints will be estimated with a standard error of 5 to 7 percentage points, depending on the observed rates. (NCT01101880)
Timeframe: Up to 5 years

Interventionmonths (Median)
Treatment (Chemotherapy and Colony Stimulating Factor)24.3

Time to Progression

With 50 patients, the rates of these endpoints will be estimated with a standard error of 5 to 7 percentage points, depending on the observed rates. (NCT01101880)
Timeframe: Up to 5 years

Interventionweeks (Median)
Treatment (Chemotherapy and Colony Stimulating Factor)7

Rates of Complete Remission and Complete Remission With Incomplete Recovery of Counts

With 50 patients, the rates of these endpoints will be estimated with a standard error of 5 to 7 percentage points, depending on the observed rates. Complete remission is defined as less than 5% blast cells present in the bone marrow and count recovery (absolute neutrophil count greater than 1000/microL and platelet count greater than 100,000/microL). Complete remission with incomplete recovery of counts is defined as less than 5% blast cells present in the bone marrow without compete count recovery (absolute neutrophil count less than 1000/microL and platelet count less than 100,000/microL). (NCT01101880)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
CR achievedCR achieved with first course of inductionCR + CRp achievedCR achieved with no AHDCR achieved with AHDCR + CRp achieved with AHDCR achieved with FLT3 positiveCR achieved with favorable risk cytogeneticsCR achieved with intermediate risk cytogeneticsCR + CRp achieved with intermediate risk cyto.CR achieved with unfavorable risk cytogeneticsCR + CRp achieved with unfavorable risk cyto.
Treatment (Chemotherapy and Colony Stimulating Factor)383341231518742627810

Complete Response Rate

"Proportion of patients who have achieve CR or CRp after treatment.~Morphologic Complete Remission (CR): Defined as morphologic leukemia-free state, including <5% blasts in Bone Marrow aspirate with marrow spicules, no persistent extramedullary disease, ANC >1000/mm3 and platelet count >100,000/mm3.~Morphologic Complete Remission without platelet recovery (CRp): Defined as CR with the exception of platelet count < 100,000/mm3 (CRp)." (NCT03118466)
Timeframe: up to 45 days

InterventionParticipants (Count of Participants)
Lenalidomide and MEC Chemotherapy19

Number of Patients That Achieved ANC Recovery

The number of patients that achieved a neutrophil count of > 500/mm3 for 3 days within 45 of starting treatment (NCT03118466)
Timeframe: up to 45 days

InterventionParticipants (Count of Participants)
Lenalidomide and MEC Chemotherapy25

Number of Patients That Achieved Platelet Recovery

The number of patients that achieved a stable platelet count > 20,000/mm3 for 3 days within 45 days of starting treatment (NCT03118466)
Timeframe: up to 45 days

InterventionParticipants (Count of Participants)
Lenalidomide and MEC Chemotherapy27

Overall Survival

Overall survival is defined as time from diagnosis of disease until date of death or censored on the last known date alive if patients are still alive. (NCT03118466)
Timeframe: Up to 3 years

InterventionMonths (Median)
Lenalidomide and MEC Chemotherapy16

Treatment-related Mortality

Cumulative number of deaths not related to persistent or relapsed leukemia during treatment within 50 days of the start of treatment. (NCT03118466)
Timeframe: 50 days

InterventionParticipants (Count of Participants)
Lenalidomide and MEC Chemotherapy2

Transfusion Support: Number of Red Blood Cell and Platelet Transfusions

Number of red blood cell and platelet transfusions received within the first 50 days of treatment (NCT03118466)
Timeframe: 50 days

InterventionNumber of Transfusions (Median)
Platelet TransfusionsRed Blood Cell Transfusions
Lenalidomide and MEC Chemotherapy79

Antitumor Activity of Flotetuzumab

Number of response evaluable participants with response (CR/PR) using revised AML International Working Group criteria for response including CR: M1 bone marrow (<5% blasts), ANC at least 1000/mm3 and platelet count at least 100,000/mm3; CRp: platelet transfusion independence; CRi: without platelet transfusion independence, CRc: revision to normal karyotype; PR: decrease at least 50% in percentage of blasts to 5% to 25% in bone marrow aspirate. (NCT04158739)
Timeframe: Up to 2 years

InterventionParticipants (Count of Participants)
Part A Dose Level 1:500 Nanograms/kg/Day1
Part A Dose Level 2: 700 Nanograms/kg/Day1

Dose Limiting Toxicities Due to Flotetuzumab

Number and percentage of patients experiencing dose limiting toxicities during cycle 1 that are possibly, probably, or definitely due to flotetuzumab by dose level and study part. (NCT04158739)
Timeframe: Up to 29 days

InterventionParticipants (Count of Participants)
Part A Dose Level 1: 500 Nanograms/kg/Day1
Part A Dose Level 2: 700 Nanograms/kg/Day1

Maximum Concentration (C Max)

Median with Minimum and Maximum for the maximum (peak) serum concentration measured on days 3-9 post-administration by dose level and study part. (NCT04158739)
Timeframe: Up to 9 days

Interventionpg/ml (Median)
Part A Dose Level 1: 500 Nanograms/kg/Day108.6
Part A Dose Level 2: 700 Nanograms/kg/Day147.6

Maximum Tolerated Dose or Recommended Phase 2 Dose of Flotetuzumab

Estimated maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) of Flotetuzumab defined by the maximum dose at which fewer than one-third of toxicity-evaluable participants experience a DLT. RP2D was established by study committee based off the toxicity profile of DL1 (500 ng/kg/day). (NCT04158739)
Timeframe: Up to 29 days

Interventionnanograms/kg/day (Number)
Treatment (Flotetuzumab, Cytarabine)500

Duration of Disease Control Rate

Duration of DCR calculated for all participants with DCR. CR: < 5% myeloblasts in BM, absence of circulating blasts, hematologic recovery (peripheral blood ANC >1000 cells/microL and platelet count > 100,000/microL, no need for RBC transfusions), absence of extra medullary disease. PR: No circulating blasts, Neutrophil count >=1.0 x 10^9/L, Platelet count >= 100 x 10^9/L, >= 50 % reduction in BM blast to 6% to 25%, or blasts <= 5% if Auer rods are present. CRp: All criteria for CR except for residual neutropenia (<1 x 10^9/L) or thrombocytopenia (<100 x 10^9/L), CRi; < 5% BM blasts with residual neutropenia [ANC < 1000 cells/microL] or thrombocytopenia [platelets < 100,000/microL]. MLFS; morphologic BM blast clearance to < 5% in a marrow sample in which <=200 cells enumerated/cellularity is >= 10%, in absence of blasts with Auer rods, no hematologic recovery required and SD; failure to achieve a response but not meeting criteria for disease progression over period of > 4 weeks. (NCT02088541)
Timeframe: Up to 4 weeks from the date of randomization to the date of progression or recurrence based on IWG criteria

InterventionDays (Median)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)187.0
Physician's Choice 2 (PV >=5)233.0

Duration of Response (DOR)

DOR was calculated from date of response of CR, CRi, CRp, MLFS, or PR to date of progression or recurrence based on IWG criteria. CR: <5% myeloblasts in bone marrow,absence of circulating blasts, hematologic recovery (peripheral blood ANC >1000 cells/microL, platelet count > 100,000/microL, no need for RBC transfusions), absence of extramedullary disease. PR: No circulating blasts, neutrophil count > =1.0 x10^9/L, platelet count >= 100 x10^9/L, >= 50 % reduction in bone marrow blast to 6% to 25%, or blasts less than or equal to (<=) 5% if Auer rods are present. CRp: All criteria for CR except for residual neutropenia (<1x10^9/L) or thrombocytopenia (<100 x10^9/L), CRi; < 5% bone marrow blasts with residual neutropenia [ANC < 1000 cells/microL] or thrombocytopenia [platelets < 100,000/microL]. MLFS: morphologic bone marrow blast clearance to < 5% in marrow sample, <= 200 cells enumerated/cellularity is ≥ 10%, in absence of blasts with Auer rods, no hematologic recovery required. (NCT02088541)
Timeframe: Baseline until disease progression or discontinuation from the study, or death, whichever occurred first (up to a maximum of approximately 104 weeks)

InterventionDays (Median)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)204.0
Physician's Choice 2 (PV >=5)148.0

Median Disease-Free Survival (DFS) for Participants Who Achieved Complete Remission (CR)

DFS for CRR based on IWG criteria, was calculated from the first date of response of CR to the date of progression or recurrence, or date of death if progression or recurrence did not occur. Participants who discontinued prior to disease progression or recurrence or did not progress as of the time of the analysis were censored at the time of last radiologic assessment. CR per IWG 2003 criteria was defined as morphologic presence of < 5 % myeloblasts in bone marrow, the absence of circulating blasts, hematologic recovery (as evidenced by a peripheral blood ANC > 1000 cells/microL and platelet count > 100,000/microL, with no need for RBC transfusions), and the absence of extramedullary disease. (NCT02088541)
Timeframe: Baseline until disease progression or discontinuation from the study, or death, whichever occurred first (up to a maximum of approximately 104 weeks)

InterventionDays (Median)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)121.0

Median Disease-Free Survival (DFS) for Participants Who Achieved Complete Remission or CR With Incomplete Hematologic Recovery (CRi) or Complete Remission With Incomplete Platelet Recovery (CRp)

DFS based on IWG criteria was defined as the duration from start of the complete response achieved until disease progression or death from any cause. Responses defined by IWG 2003 Response Criteria: Morphologic CR: < 5% myeloblasts in bone marrow, the absence of circulating blasts, hematologic recovery (as evidenced by a peripheral blood ANC > 1000 cells/microL and platelet count > 100,000/microL, with no need for RBC transfusions), and the absence of extramedullary disease. Morphologic CRp: All criteria for CR except for residual neutropenia (<1x10^9/L) or thrombocytopenia (<100 x10^9/L), Cri (< 5% bone marrow blasts with residual neutropenia [ANC < 1000 cells/microL] or thrombocytopenia [platelets < 100,000/microL]), normal maturation of all cellular components in the bone marrow, no extramedullary disease and transfusion independent. (NCT02088541)
Timeframe: Baseline until disease progression or discontinuation from the study, or death, whichever occurred first (up to a maximum of approximately 104 weeks)

InterventionDays (Median)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)175.0
Physician's Choice 2 (PV >=5)106.0

Overall Survival

Overall survival was defined as the time (in days) from the date of randomization to the date of death due to any cause. Participants last known to be alive were censored at date of last contact. (NCT02088541)
Timeframe: Baseline until disease progression or discontinuation from the study, or death, whichever occurred first (up to a maximum of approximately 104 weeks)

InterventionDays (Median)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)94.0
Physician's Choice 2 (PV >=5)170.0

Percentage of Participants With Complete Remission Rate (CRR) for Those Who Achieved Complete Remission (CR)

CRR was analyzed using International Working Group (IWG) 2003 criteria, as the difference in the proportions of participants with IWG results of CR. CR per IWG 2003 criteria was defined as morphologic presence of < 5 percentage (%) myeloblasts in bone marrow, the absence of circulating blasts, hematologic recovery (as evidenced by a peripheral blood absolute neutrophil count (ANC) > 1000 cells/microliter (microL) and platelet count > 100,000/microL, with no need for red blood cell (RBC) transfusions), and the absence of extramedullary disease. (NCT02088541)
Timeframe: Baseline until disease progression or discontinuation from the study, or death, whichever occurred first (up to a maximum of approximately 104 weeks)

InterventionPercentage of participants (Number)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)5.1
Physician's Choice 2 (PV >=5)0

Percentage of Participants With Disease Control Rate (DCR)

DCR:Point estimate of % of participants with CR,CRi,CRp,MLFS,PR, or SD for <=4 weeks.CR:<5% myeloblasts in bone marrow (BM),absence of circulating blasts,hematologic recovery(peripheral blood ANC >1000 cells/microL and platelet count >100,000/microL, no need of RBC transfusions),absence of extramedullary disease. PR:No circulating blasts,Neutrophil count >=1.0 x10^9/L, Platelet count >= 100*10^9/L, >=50% reduction in BM blast to 6% to 25%, or blasts <=5% if Auer rods are present.CRp:All criteria for CR except for residual neutropenia (<1*10^9/L) or thrombocytopenia(<100 x10^9/L),CRi;< 5% BM blasts with residual neutropenia [ANC < 1000 cells/microL] or thrombocytopenia [platelets < 100,000/microL]. MLFS:morphologic BM blast clearance to <5% in a marrow sample in which <=200 cells enumerated or cellularity is ≥10%,in absence of blasts with Auer rods,no hematologic recovery required,SD:failure to achieve a response but not meeting criteria for disease progression over period of >4 weeks. (NCT02088541)
Timeframe: Up to 4 weeks from the date of randomization to the date of progression or recurrence based on IWG criteria

InterventionPercentage of participants (Number)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)50.8
Physician's Choice 2 (PV >=5)40.4

Percentage of Participants With Modified Complete Remission Rate (mCRR) for Those Who Achieved Complete Remission (CR) or Complete Remission With Incomplete Hematologic Recovery (Cri)

mCRR was defined as the point estimate of the percentage of participants who had CR, CRi, or CRp. Responses defined as per IWG 2003 response criteria: Morphologic CR:< 5% myeloblasts in bone marrow, the absence of circulating blasts, hematologic recovery (as evidenced by a peripheral blood ANC > 1000 cells/microL and platelet count > 100,000/microL, with no need for RBC transfusions), and the absence of extramedullary disease. Morphologic CRp: All criteria for CR except for residual neutropenia (<1x10^9/L) or thrombocytopenia (<100 x10^9/L), Cri (< 5% bone marrow blasts with residual neutropenia [ANC < 1000 cells/microL] or thrombocytopenia [platelets < 100,000/microL]), normal maturation of all cellular components in the bone marrow, no extramedullary disease and transfusion independent. (NCT02088541)
Timeframe: Baseline until disease progression or discontinuation from the study, or death, whichever occurred first (up to a maximum of approximately 104 weeks)

InterventionPercentage of participants (Number)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)11.9
Physician's Choice 2 (PV >=5)3.5

Percentage of Participants With Overall Response Rate (ORR)

Overall response rate was defined as the point estimate of the percentage of participants who achieved CR (disappearance of all target and non-target lesions), partial response (PR) (>=30 % decrease in sum of longest diameters of target lesions taking as reference baseline sum longest diameters associated to non-progressive disease response for non-target lesions). CRi; < 5% BM blasts with residual neutropenia [ANC < 1000 cells/microL] or thrombocytopenia [platelets < 100,000/microL]. CRp; All criteria for CR except for residual neutropenia (<1x10^9/L) or thrombocytopenia (<100 x10^9/L) and morphologic leukemia-free state (MLFS); morphologic BM blast clearance to <5% in a marrow sample in which <=200 cells enumerated or cellularity is ≥10%, in absence of blasts with Auer rods, no hematologic recovery required. (NCT02088541)
Timeframe: Baseline until disease progression or discontinuation from the study, or death, whichever occurred first (up to a maximum of approximately 104 weeks)

InterventionPercentage of participants (Number)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)13.6
Physician's Choice 2 (PV >=5)8.8

Percentage of Participants With Overall Survival of at Least 3 Months (OS3.0)

Overall survival was defined as the time (in days) from the date of randomization to the date of death due to any cause. Participants last known to be alive were censored at date of last contact. Analysis was performed using Kaplan-Meier method. (NCT02088541)
Timeframe: From randomization (Day 1) up to 3 months

InterventionPercentage of participants (Number)
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)53.49
Physician's Choice 2 (PV >=5)70.73

Change From Baseline in European Quality of Life-5 Dimension (EQ-5D) Health Questionnaire Visual Analogue Scale (VAS)

EQ-5D descriptive system comprises of 5 dimensions: mobility, self-care, usual activities, pain/discomfort and anxiety/depression. Each dimension has 5 levels: 1=no problems, 2=slight problems, 3=moderate problems, 4=severe problems, and 5=extreme problems. EQ-5D-5L is a standardized instrument that measures health-related quality of life for men with prostate cancer. EQ-5D consists of EQ-5D descriptive system and EQ VAS. EQ-5D-5-VAS records participant's self-rated health on a vertical VAS that allows them to indicate their health state that can range from 0 (worst imaginable) to 100 (best imaginable), higher scores indicating a better health state. (NCT02088541)
Timeframe: Baseline, Day 1 of each treatment cycle (a maximum of 20 cycles: 28 days per cycle) up to 30 days after last dose of study drug (final visit)

InterventionUnits on a scale (Mean)
BaselineC2D1C3D1C4D1C5D1C6D1C7D1C8D1C9D1C10D1C11D1C12D1C13D1C14D1C15D1C16D1C17D1
Physician's Choice 2 (PV >=5)63.50.8-5.2-5.0-2.0-1.44.05.05.06.3-5.0-5.010.015.0-10.020.0-5.0

Change From Baseline in European Quality of Life-5 Dimension (EQ-5D) Health Questionnaire Visual Analogue Scale (VAS)

EQ-5D descriptive system comprises of 5 dimensions: mobility, self-care, usual activities, pain/discomfort and anxiety/depression. Each dimension has 5 levels: 1=no problems, 2=slight problems, 3=moderate problems, 4=severe problems, and 5=extreme problems. EQ-5D-5L is a standardized instrument that measures health-related quality of life for men with prostate cancer. EQ-5D consists of EQ-5D descriptive system and EQ VAS. EQ-5D-5-VAS records participant's self-rated health on a vertical VAS that allows them to indicate their health state that can range from 0 (worst imaginable) to 100 (best imaginable), higher scores indicating a better health state. (NCT02088541)
Timeframe: Baseline, Day 1 of each treatment cycle (a maximum of 20 cycles: 28 days per cycle) up to 30 days after last dose of study drug (final visit)

InterventionUnits on a scale (Mean)
BaselineC2D1C3D1C4D1C5D1C6D1C7D1C8D1C9D1C10D1C11D1C12D1C13D1C14D1C15D1C17D1C18D1C19D1C20D1
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)67.9-7.5-13.3-6.1-0.9-11.2-3.70.14.60.70.83.36.76.73.525.025.030.035.0

Change From Baseline in Quality of Life (QoL) and Patient-Reported Outcomes (Functional Assessment of Cancer Therapy -Leukemia [FACT-Leu]

QoL was assessed by the FACT-Leu. FACT-Leu combines the General version of the Functional Assessment of Cancer Therapy (FACT-G) with a leukemia-specific sub-scale (17 items). The sub-scales for the FACT-G are Physical Well-Being (7 items), Social/Family Well-Being (7 items), Emotional Well-Being (6 items), and Functional Well-Being (7 items). The trial outcomes index (TOI; total of 31 items) was the primary measurement of interest, comprising the Physical and Functional sub-scales plus the leukemia - specific sub-scale. Each item was rated on a 5-point Likert scale. Range from 0 = (Not at all) to 4 = (Very much); therefore, the TOI had a score ranging from 0 to 124. Higher scores indicated improvement in well being. The QoL assessment was performed at baseline (prior to first dose of study treatment), Day 1 of each cycle on or after the second, and at the final visit. (NCT02088541)
Timeframe: Baseline, Day 1 of each treatment cycle (a maximum of 20 cycles: 28 days per cycle) up to 30 days after last dose of study drug (final visit)

InterventionUnits on a scale (Mean)
BaselineC2D1C3D1C4D1C5D1C6D1C7D1C8D1C9D1C10D1C11D1C12D1C13D1C14D1C15D1C16D1C17D1Final visit
Physician's Choice 2 (PV >=5)75.4-1.9-5.4-7.41.9-4.7-11.8-10.6-8.0-10.7-10.0-7.0-9.0-15.0-10.0-15.0-7.0-1.7

Change From Baseline in Quality of Life (QoL) and Patient-Reported Outcomes (Functional Assessment of Cancer Therapy -Leukemia [FACT-Leu]

QoL was assessed by the FACT-Leu. FACT-Leu combines the General version of the Functional Assessment of Cancer Therapy (FACT-G) with a leukemia-specific sub-scale (17 items). The sub-scales for the FACT-G are Physical Well-Being (7 items), Social/Family Well-Being (7 items), Emotional Well-Being (6 items), and Functional Well-Being (7 items). The trial outcomes index (TOI; total of 31 items) was the primary measurement of interest, comprising the Physical and Functional sub-scales plus the leukemia - specific sub-scale. Each item was rated on a 5-point Likert scale. Range from 0 = (Not at all) to 4 = (Very much); therefore, the TOI had a score ranging from 0 to 124. Higher scores indicated improvement in well being. The QoL assessment was performed at baseline (prior to first dose of study treatment), Day 1 of each cycle on or after the second, and at the final visit. (NCT02088541)
Timeframe: Baseline, Day 1 of each treatment cycle (a maximum of 20 cycles: 28 days per cycle) up to 30 days after last dose of study drug (final visit)

InterventionUnits on a scale (Mean)
BaselineC2D1C3D1C4D1C5D1C6D1C7D1C8D1C9D1C10D1C11D1C12D1C13D1C14D1C15D1C17D1C18D1C19D1C20D1Final visit
Selinexor 60 mg (PV >=5) (Equivalent to 35 mg/m^2)70.50.5-1.9-1.2-2.4-3.4-5.0-3.9-2.72.4-0.1-3.7-3.3-4.01.5-15.0-1.0-12.0-14.02.5

Early Death Rate

Early death was defined as death before the end of the first induction cycle. (NCT02249091)
Timeframe: 1 induction cycle (4 weeks)

InterventionParticipants (Count of Participants)
Cohort 1 / Selinexor 40 mg/m^2 in Combination With Cytarabine and Idarubicin0
Cohort 2 / Selinexor 60 mg Flat Dose in Combination With Cytarabine and Idarubicin4

Event-Free Survival

Event-free survival (EFS) was calculated from the time of informed consent until death, not achieving CR/CRi or relapse after CR/CRi. (NCT02249091)
Timeframe: Time from registration to event, max 2 years

Interventionmonths (Median)
Cohort 1 / Selinexor 40 mg/m^2 in Combination With Cytarabine and Idarubicin5.6
Cohort 2 / Selinexor 60 mg Flat Dose in Combination With Cytarabine and Idarubicin4.3

Number of Participants With CR/CRi = Overall Reponse Rate

"Efficacy of selinexor in combination with standard chemotherapy in patients with relapsed/refractory AML by determination of rate of complete response (CR) or morphologic complete response with incomplete blood count recovery (CRi), as defined by the recommendations on diagnosis and management of AML in adults from an international expert panel, on behalf of the European LeukemiaNet:~CR: Absolute Neutrophil count (ANC) >1.0x10^9/L, Platelet count >100x10^9/L, Bone marrow blasts <5%, no Auer rods, no evidence of extramedullary disease.~CRi: Same as CR, but ANC may be <1.0x10^9/L and/or Platelet count <100x10^9/L.~Patients with morphologic leukemia free-state (MLFS) were included in the group of responders. MLFS: Bone marrow blasts <5%, no Auer rods, no evidence of extramedullary disease.~The best response after Selinexor treatment was analyzed, thus the best response after the induction cycle(s)." (NCT02249091)
Timeframe: 1-2 induction cycles (4 - 8 weeks)

InterventionParticipants (Count of Participants)
Cohort 1 / Selinexor 40 mg/m^2 in Combination With Cytarabine and Idarubicin15
Cohort 2 / Selinexor 60 mg Flat Dose in Combination With Cytarabine and Idarubicin6

Number of Participants With Partial Remission (PR) = Rate of PR

"Efficacy of selinexor in combination with standard chemotherapy in patients with relapsed/refractory AML by determination of rate of partial remission(PR) as defined by the recommendations on diagnosis and management of AML in adults from an international expert panel, on behalf of the European LeukemiaNet:~PR: Absolute Neutrophil count (ANC) >1.0x10^9/L, Platelet count >100x10^9/L, at least a 50% decrease in the percentage of marrow aspirate blasts to 5-25%, or marrow blasts <5% with persistent Auer rods.~The best response after Selinexor treatment was analyzed, thus the best response after the induction cycle(s)." (NCT02249091)
Timeframe: 1-2 induction cycles (4 - 8 weeks)

InterventionParticipants (Count of Participants)
Cohort 1 / Selinexor 40 mg/m^2 in Combination With Cytarabine and Idarubicin0
Cohort 2 / Selinexor 60 mg Flat Dose in Combination With Cytarabine and Idarubicin0

Overall Survival

Overall survival (OS) was calculated from the date of informed consent to the date of death. Patients still alive at the end of follow-up were censored at the last date of follow-up. (NCT02249091)
Timeframe: Time from registration to event, max 2 years

Interventionmonths (Median)
Cohort 1 / Selinexor 40 mg/m^2 in Combination With Cytarabine and Idarubicin12.6
Cohort 2 / Selinexor 60 mg Flat Dose in Combination With Cytarabine and Idarubicin8.0

Percentage of Patients Transplanted After Induction Therapy (Stem Cell Transplantation)

Percentage of patients being transplanted after induction therapy (stem cell transplantation) (NCT02249091)
Timeframe: 1-2 induction cycles (4 - 8 weeks)

InterventionParticipants (Count of Participants)
Cohort 1 / Selinexor 40 mg/m^2 in Combination With Cytarabine and Idarubicin11
Cohort 2 / Selinexor 60 mg Flat Dose in Combination With Cytarabine and Idarubicin4

Progression-Free Survival

"Progression-free survival (PFS) was calculated from the time of informed consent to the date of recurrence or death, whichever occurred first. Patients were censored at the date of the last follow-up visit if they were alive without relapse.~Disease progression was defined as presence of >50% increase in bone marrow blasts to a level of at least 50% and/or a doubling of the percentage of peripheral blood blasts to a level of at least 50%." (NCT02249091)
Timeframe: Time from registration to event, max 2 years

Interventionmonths (Median)
Cohort 1 / Selinexor 40 mg/m^2 in Combination With Cytarabine and Idarubicin6.3
Cohort 2 / Selinexor 60 mg Flat Dose in Combination With Cytarabine and Idarubicin4.3

Relapse-Free Survival

Relapse-free survival (RFS) was calculated from the date of CR/CRi until death or relapse, whichever occurred first. Patients were censored on the date of the last follow-up if they were alive without relapse. (NCT02249091)
Timeframe: Time from registration to event, max 2 years

Interventionmonths (Median)
Cohort 1 / Selinexor 40 mg/m^2 in Combination With Cytarabine and Idarubicin10.9
Cohort 2 / Selinexor 60 mg Flat Dose in Combination With Cytarabine and IdarubicinNA

Complete Response (CR) Rate in Patients With Relapsed or Refractory AML

"Complete Remission (CR) rate = Percentage of patients achieving CR after Cycle 1 as defined in Stage 1 by the International Working Group (IWG) Criteria and 2010 European LeukemiaNet (EN) criteria in patients with relapsed or refractory AML with MCL-1 dependence >30% and in Stage 2 by the 2017 ELN criteria.~The study was terminated in January 2020 due to a steady and marked reduction in enrollment. Thus, the efficacy endpoints could not be analyzed. As sufficient efficacy results were not available to analyze patients based on the percentage of MCL-1 dependency the treatment efficacy was summarized by distributing the safety population into 6 groups based on whether the patients received the ACM vs CM regimen and their disease stages at study entry." (NCT02520011)
Timeframe: Best response after at least 1 cycle through study completion approximately 4 years

,,,,,
Interventionparticipants (Number)
Complete Remission (CR)CR with Incomplete Neutrophil Recovery (Cri)Partial Remission (PR)Resistant/Relapsed DiseaseNot Evaluated
All Stages and Cohorts (Including Randomized Stage): ACM Total271723413
Stage 1 ACM Relapsed/Refractory85156
Stage 1 Newly Diagnosed ACM62141
Stage 2 ACM Relapsed/Refractory23051
Stage 2 CM Relapsed/Refractory60050
Stages 1 and 2 ACM Relapsed/Refractory211513012

Response to Treatment

To determine if treatment with ACM can induce CR in patients with relapsed or refractory AML with MCL-1 dependence of >30% who failed to achieve CR following 1 cycle of CM (NCT02520011)
Timeframe: Best response after at least 1 cycle through study completion approximately 4 years

Interventionparticipants (Number)
Partial Remission (PR)Not Evaluated
Stage 2 CM Relapsed/Refractory AML11

Evaluate Toxicity by Assessing the Adverse Events of Metformin and Cytarabine

To determine the maximum tolerated dose (MTD) by assessing the adverse events of metformin in combination with cytarabine in evaluating toxicity. Assessments will occur daily during cytarabine administration and at least twice weekly following treatment until blood count recovery. (NCT01849276)
Timeframe: Checked daily during administration of cytarabine and at least 2x weekly following therapy until desired blood counts acheived (maximum 15 days)

InterventionAdverse events related to treatment (Number)
Grade 1Grade 2Grade 3Grade 4
Treatment (Enzyme Inhibitor and Chemotherapy)4620

Event Free Survival (EFS)

Event free survival defined as the time from treatment to relapse of leukemia or death for any reason or lost to follow-up. Study regimen considered successful if it exhibits a 3-year EFS rate greater than 65% and response rate no less than 90% with Grade III-IV infectious toxicity rate in induction no more than 33%. (NCT02419469)
Timeframe: 3 years

InterventionParticipants (Count of Participants)
Ofatumumab Plus ChemotherapyNA

Complete Response (CR)

"Complete response (CR) was determined the number of participants who achieved CR by Day 29 after induction treatment. The outcome is reported as the total number without dispersion. CR is defined as:~CR = No circulating blasts or extramedullary disease; trilineage hematopoiesis; absolute neutrophil count (ANC) > 1,000/microliter; platelets > 100,000/microliter; < 5% blasts in bone marrow.~Not CR = All statuses and conditions if less than or not as defined." (NCT01769209)
Timeframe: Day 29

InterventionParticipants (Count of Participants)
Bortezomib + Chemotherapy11

Complete Response Without Platelet Recovery (CRp)

"Complete response without platelet recovery (CR) was determined as the number of participants who achieved CRp by Day 29 after induction treatment. The outcome is reported as the total number without dispersion. The outcome reflects only those subjects that meet all complete response (CR) criteria except platelet count; participants that meet all criteria including platelet count are not included in this outcome. CR and CRp are defined below.~CR =.No circulating blasts or extramedullary disease; trilineage hematopoiesis; absolute neutrophil count (ANC) > 1,000/microliter; platelets > 100,000/microliter; < 5% blasts in bone marrow.~CRp = Meets all criteria for CR except platelet count." (NCT01769209)
Timeframe: Day 29

InterventionParticipants (Count of Participants)
Bortezomib + Chemotherapy1

Failure-free Survival (FFS)

"Failure-free survival (FFS) was assessed as survival without progression or the addition of another systemic therapy, at or within 2 years. The outcome is reported as the number (without dispersion) of the participants alive without progression. Progression is defined below.~Progression = More than 25% increase in circulating and/or bone marrow blasts, or the development of extramedullary disease." (NCT01769209)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Bortezomib + Chemotherapy3

Overall Survival (OS)

Overall survival (OS) was assessed as participants remaining alive 2 years after induction therapy. The outcome is reported as the number of participants (without dispersion). (NCT01769209)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Bortezomib + Chemotherapy5

Progression-free Survival (PFS)

"Progression-free survival (PFS) was assessed as survival without progression at 2 years. The outcome is reported as the number (without dispersion) of the participants alive without progression.~Progression = More than 25% increase in circulating and/or bone marrow blasts, or the development of extramedullary disease." (NCT01769209)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Bortezomib + Chemotherapy3

Response Rate (RR)

"Response Rate (RR) was determined as the sum of complete response (CR) and partial response (PR). Due to overlap, complete response rate without platelet recovery (CRp) is not included in Response Rate (RR). The outcome is reported as the total number without dispersion.~CR = No circulating blasts or extramedullary disease; trilineage hematopoiesis; absolute neutrophil count (ANC) > 1,000/microliter; platelets > 100,000/microliter; < 5% blasts in bone marrow.~CRp = Meets all criteria for CR except platelet count.~PR = Meets all criteria for CR except bone marrow contains 5 to 25% leukemia cells." (NCT01769209)
Timeframe: Day 29

InterventionParticipants (Count of Participants)
Bortezomib + Chemotherapy11

Related Adverse Events (Grade 3, 4, 5)

Toxicity was assessed as related grade 3, 4, or 5 adverse events (AEs) per National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) v4.03. The outcome is reported as the total numbers of events (without dispersion) by CTCAE Body System, and whether the event was a hematologic toxicity or non-hematologic toxicity. (NCT01769209)
Timeframe: 45 days

InterventionTreatment-related adverse events (Number)
Blood and Lymphatic System DisordersGastrointestinal DisordersHepatobiliary disordersInfections and InfestationsInvestigationsMetabolism and Nutrition DisordersNervous System DisordersRespiratory, Thoracic and Mediastinal DisordersVascular DisordersHematologic ToxicityNon-hematologic Toxicity
Bortezomib + Chemotherapy109921778491126352

Overall Survival (OS) in Participants Who Experienced Complete Response or Complete Response With Incomplete Count Recovery

(NCT01829503)
Timeframe: Up to 38 months (median follow-up = 25.4 months)

Interventionmonths (Median)
Decitabine + Cytarabine15.7

Overall Survival (OS) in Participants Who Experienced Complete Response, Complete Response With Incomplete Count Recovery, or Partial Response

(NCT01829503)
Timeframe: Up to 38 months (median follow-up = 25.4 months)

Interventionmonths (Median)
Decitabine + Cytarabine15.7

Overall Survival (OS) in Participants Who Experienced Complete Response

(NCT01829503)
Timeframe: Up to 38 months (median follow-up = 25.4 months)

Interventionmonths (Median)
Decitabine + Cytarabine18.9

Overall Survival (OS)

(NCT01829503)
Timeframe: Up to 38 months (median follow-up = 25.4 months)

Interventionmonths (Median)
Decitabine + Cytarabine10.8

Relapse-Free Survival in Participants With Complete Response or Complete Response With Incomplete Count Recovery.

(NCT01829503)
Timeframe: Up to 38 months

Interventionmonths (Median)
Decitabine + Cytarabine11.7

Relapse-Free Survival in Participants With Complete Response, Complete Response With Incomplete Count Recovery or Partial Response, and Received Maintenance Therapy

(NCT01829503)
Timeframe: Up to 38 months

Interventionmonths (Median)
Decitabine + Cytarabine + Maintenance Therapy11.5

Demographic Characteristics and Clinical Measures as Potential Predictors of Overall Survival (OS)

Median number of months of survival per individual demographic characteristics and clinical measures. (NCT01829503)
Timeframe: Up to 38 months (median follow-up = 25.4 months)

Interventionmonths (Median)
Age ≤ 75 yearsAge > 75 yearsSex - FemaleSex - MaleECOG status 0ECOG status 1ECOG status 2Charlson co-morbidity status ≤ 6Charlson co-morbidity status > 6FLT and NPM1 Status: favorableFLT and NPM1 Status: intermediateFLT and NPM1 Status: adverseAML Type-Primary AMLAML Type-AML with MDS changesAML Type-Treatment-related AMLWBC ≤ 5.25 µ/LWBC > 5.25 µ/LHematocrit ≤ 27.9%Hematocrit > 27.9%Platelets ≤ 54.5 µ/LPlatelets > 54.5 µ/LAlbumin ≤ 3.4 gm/dLAlbumin > 3.4 gm/dLCreatinine ≤ 0.97 mg/dLCreatinine > 0.97 mg/dLBilirubin ≤ 0.8 mg/dLBilirubin > 0.8 mg/dLLDH ≤ 233 IU/LLDH > 233 IU/LBone marrow blasts ≤ 53.65%Bone marrow blasts > 53.65%Peripheral blasts ≤ 13%Peripheral blasts > 13%
Decitabine + Cytarabine12.49.711.010.616.012.64.214.37.418.97.614.310.311.512.414.97.613.010.510.415.410.813.610.614.910.413.614.46.914.97.614.07.6

Functional Assessment of Cancer Therapy: Health-related Quality of Life (HRQOL) Measure

"The FACT-Leu Health-related Quality of Life (HRQOL) Measure is a 27-item FACT-G scale plus a 17-item leukemia sub-scale. The FACT-G contains uses Likert scale 0-4, with 0 =not at all and 4 =very much. The total score can be 0-108 and includes 7 items related Physical Well-being (PWB), 7 items related to Social Well-being (SWB), 6 items related to Emotional Well-being (EWB) and 7 items related to Functional Well-Being (FWB). Higher scores are better. Responses based on how patients felt in the past 7 days. FACT-Leu uses a Likert scale (0 to 4, with 0= not at all and 4= very much). The total score for the 17 items can be 0-68. Higher scores are better. The FACT-Leu total is the sum of FACT-G and FACT Leu and ranges from 0-176. Higher scores are better. FACT Trial Outcome Index is derived by adding scores on the PWB and FWB sub-scales to the leukemia sub-scales. The total for this index score is from 0-124. Higher scores are better." (NCT01829503)
Timeframe: Baseline to Post-treatment, up to 5 years

Interventionunits on a scale (Mean)
Baseline FACT-LEU SubscaleBaseline FACT Trial Outcome IndexBaseline FACT-LEU TotalBaseline FACT-G TotalBaseline FACT-G - Physical Well-BeingBaseline FACT-G - Social Well-BeingBaseline FACT-G - Emotional Well-BeingBaseline FACT-G - Functional Well-BeingPost-treatment LEU SubscalePost-treatment FACT Trial Outcome IndexPost-treatment FACT-LEU TotalPost-treatment FACT-G TotalPost-treatment FACT-G - Physical Well-BeingPost-treatment FACT-G - Social Well-BeingPost-treatment FACT-G - Emotional Well-BeingPost-treatment FACT-G - Functional Well-Being
Decitabine + Cytarabine + Maintenance Therapy52.0592.90135.4083.5722.7524.7317.3017.1651.7892.50137.2684.2323.2024.7218.0716.62

Number of Participants by Best Clinical Response Experienced

The number of participants who experienced either a Complete Response, Complete Response with Incomplete Count Recovery, Partial Response, or Progressive Disease. Complete response: Less than 5% blasts in an aspirate sample of a patient who has an absolute neutrophil count of >1000µ/L and platelets >100,000µ/L; Complete response with incomplete count recovery: Complete response except for residual neutropenia (<1000µ/L) or thrombocytopenia (<100,000µ/L) Partial response: Decrease of at least 50% in the percentage of blasts to 5-25% in the bone marrow aspirate; Progressive disease: Failure to achieve complete response or partial response (NCT01829503)
Timeframe: Up to 38 months

InterventionParticipants (Number)
Complete ResponseComplete Response with Incomplete Count RecoveryPartial ResponseProgressive Disease
Decitabine + Cytarabine20667

Numbers of Patients (Out of 44) Experiencing Adverse Events With CTCAE Grade ≥ 3 or Adverse Events Grade ≥ 4

The number of participants (out of 44) experiencing adverse events, with CTCAE Grade ≥ 3 or Adverse Events Grade ≥ 4 (NCT01829503)
Timeframe: Up to 38 months

InterventionParticipants (Number)
Adverse Events Grade ≥ 3Adverse Events Grade ≥ 4
Decitabine + Cytarabine4441

Proportion of Participants With Clinical Response (CR)

The number of participants (out of 39) who experienced Clinical Response as Complete Response, or, Complete Response + Complete Response with Incomplete Count Recovery (exact Clopper-Pearson confidence interval). (NCT01829503)
Timeframe: Up to 38 months

InterventionProportion of participants (Number)
CompleteComplete + Complete with Incomplete Count Recovery
Decitabine + Cytarabine0.510.67

Proportion of Participants With Survival to Four and Eight Weeks and One Year

The proportion of all participants experiencing four and eight-week mortality, or, who were alive at one year. (NCT01829503)
Timeframe: Up to one year (4 weeks, 8 weeks, and one year)

InterventionProportion of participants (Number)
Death Within 4 WeeksDeath Within 8 WeeksAlive at One Year
Decitabine + Cytarabine0.0230.0910.48

Complete Clinical Response

Number of patients with newly diagnosed Acute Myeloid Leukemia who achieved Complete Response to therapy as determined by bone marrow biopsy evaluation. A CR designation required that the patient achieved a morphologic leukemia-free state and an absolute neutrophil count greater than or equal to 1.0 x 10^9/l, a platelet count greater than or equal to 100 x 10^9/l, and no evidence of extramedullary disease. (NCT01960387)
Timeframe: Between 14 and 28 days from start of study treatment

Interventionparticipants (Number)
Clofarabine (40mg/m^2/Day) + Cytarabine (1g/m^2/Day)2

Disease-free Survival

Percentage of patients who survive without any signs or symptoms of cancer at 1 year. (NCT01969435)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)70

Disease-free Survival

Percentage of patients who survive without any signs or symptoms of cancer at 2 years. (NCT01969435)
Timeframe: 2 years

Interventionpercentage of participants (Number)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)64

Overall Survival (OS) Rate

(NCT01969435)
Timeframe: Median follow-up 15.4 months (range 4.7-24.6)

Interventionpercentage of participants (Number)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)10

Progression-free Survival (PFS) Rate

PFS - Time from start of treatment to the time of progression or death, whichever occurs first. (NCT01969435)
Timeframe: 6 months

Interventionpercentage of participants (Median)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)84

Progression-free Survival Rate (PFS)

(NCT01969435)
Timeframe: 1 year

Interventionpercentage of participants (Median)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)70

Relapse Free Survival

(NCT01969435)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)0

Time to Engraftment (Neutrophil)

Time from the date of the transplant to the date of neutrophil engraftment. (NCT01969435)
Timeframe: Assessed up to day 30

Interventiondays (Median)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)10

Time to Engraftment (Platelet)

Time from the date of transplant to the date of platelet engraftment. (NCT01969435)
Timeframe: Assessed up to day 100

Interventiondays (Median)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)19

Treatment-related Mortality (TRM)

TRM is defined as death not due to progressive lymphoma prior to Day 100 after transplant (NCT01969435)
Timeframe: 100 days

Interventionpercentage of participants (Number)
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)0

Efficacy as Measured by Response Rates

"The response rates according to each category of response Complete Response (CR), Partial Response (PR), Stable Disease (SD), and Progressive Disease (PD) will be summarized by the proportion of patients meeting each criterion.~Evaluated using PET or CT scan and Revised Response Criteria for Malignant Lymphoma" (NCT01969435)
Timeframe: Up to Day 100

Interventionpercentage of participants (Number)
Complete responsePartial responseStable diseaseProgressive disease
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)844012

Safety and Toxicity as Measured by Treatment Related Non-hematologic Adverse Events

Adverse events will be assessed using the National Cancer Institute (NCI)-CTCAE version 4.0. Number of events, grade 2 or higher, occurring in 10% or greater of participants. Grade 2 diarrhea and Grade 2 nausea/vomiting were not recorded. (NCT01969435)
Timeframe: Day -7 through Day 30

Interventionparticipants (Number)
Fever neutropeniaFeverBacteremiaClostridium difficileRespiratory infectionMucosal infectionSkin infectionGenito-urinary tract infectionHypotensionAbdominal painDiarrheaMucositis oralLiver enzymes increasedBilirubin increasedDehydrationHyperglycemiaHypoalbuminemiaHypocalcemiaHypokalemiaHypophosphatemiaPainHeadacheHypoxiaRash
Melphalan, Carmustine, Etoposide, Cytarabine (BEAM)34673573315582755561012143510798

Minimal Residual Disease Negative Complete Remission Rate in Patients With Newly Diagnosed Disease (Phase II)

Remission Rate defined as Recist Category of Complete Resposne (CR) Disappearance of all non-target lesions and normalization of tumor marker level. All lymph nodes must be non-pathological in size(<10 mm short axis). (NCT02044796)
Timeframe: Up to day 45 after start of second course of induction chemotherapy

InterventionParticipants (Count of Participants)
Newly Diagnosed Group95
Relapsed/Refractory22

Number of Participants With Dose Limiting Toxicities of Mitoxantrone (Phase I, Dose Level 4)

Defined as the highest dose studied in which the incidence of dose-limiting toxicity is < 33%, graded according to NCI Common Terminology Criteria for Adverse Events version 4.0 (NCT02044796)
Timeframe: Up to day 45 after start of induction chemotherapy

InterventionParticipants (Count of Participants)
Newly Diagnosed Group1
Relapsed/Refractory Group2

Overall Survival (Phase II)

Number of subjects that have survived (NCT02044796)
Timeframe: From date of randomization until the date of death from any cause, assessed up to 12 months

InterventionParticipants (Number)
Phase 2 Newly Diagnosed Group 18 mg/m^266
Phase 2 Relapsed/Refractory Group 16 mg/m^213

Remission Rate (Complete Remission and Complete Remission With Incomplete Platelet Count Recovery) of This Regimen in Patients With Relapsed/Refractory Disease (Phase II)

(NCT02044796)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
Phase 2 Relapsed/Refractory Group 16 mg/m^224

30 Day Survival Rate

Percentage of participants who were alive 30 days after starting induction treatment. (NCT01238211)
Timeframe: 30 days

Interventionpercentage of participants (Number)
Treatment (Daunorubicin Hydrochloride, Cytarabine, Dasatinib)97

Complete Response Rate

Percentage of participants who achieve a CR. Complete remission (CR) is defined as: disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x 10^9/L and platelet count > 100 x 10^9/L, and normal bone marrow differential (< 5% blasts). (NCT01238211)
Timeframe: 60 months

Interventionpercentage of patients (Number)
Treatment (Daunorubicin Hydrochloride, Cytarabine, Dasatinib)90

Cumulative Incidence of Death

(NCT01238211)
Timeframe: 36 months

InterventionParticipants (Count of Participants)
Treatment (Daunorubicin Hydrochloride, Cytarabine, Dasatinib)15

Cumulative Incidence of Relapse

(NCT01238211)
Timeframe: 60 months

InterventionParticipants (Count of Participants)
Treatment (Daunorubicin Hydrochloride, Cytarabine, Dasatinib)10

Disease-free Survival

Disease free survival (DFS) is defined as the time from achievement of CR to relapse or death, whichever comes first. The 3 year DFS rate with 95% CI was estimated using the Kaplan-Meier method. (NCT01238211)
Timeframe: 3 years

Interventionpercentage of patients (Number)
Treatment (Daunorubicin Hydrochloride, Cytarabine, Dasatinib)75

Event-free Survival

"Event free survival (EFS) is defined as the time from registration to failure to achieve complete remission (CR), relapse after CR is attained or death, whichever comes first. The 1 year EFS rate with 95% CI was estimated using the Kaplan-Meier method,~Complete remission (CR) is defined as: disappearance of all clinical and/or radiologic evidence of disease. Neutrophil count > 1.0 x 10^9/L and platelet count > 100 x 10^9/L, and normal bone marrow differential (< 5% blasts)." (NCT01238211)
Timeframe: 1 year

Interventionpercentage of patients (Number)
Treatment (Daunorubicin Hydrochloride, Cytarabine, Dasatinib)83

Overall Survival

Overall survival (OS) is defined as time from registration to death. The 3 year OS rate with 95% CI was estimated using the Kaplan-Meier method. (NCT01238211)
Timeframe: 3 years

Interventionpercentage of patients (Number)
Treatment (Daunorubicin Hydrochloride, Cytarabine, Dasatinib)77

Complete Remission (CR)

Complete disappearance of all clinical evidence of disease (NCT03504410)
Timeframe: 12 months

InterventionParticipants (Count of Participants)
CPI-613 + HD Cytarabine and Mitoxantrone20
Control (HAM) and Control Sub-groups (MEC and FLAG)22

Overall Survival

Overall survival will be estimated. (NCT02483000)
Timeframe: Up to 4 years

InterventionParticipants (Count of Participants)
Treatment (PRIT)2

Dosimetry of Yttrium Y 90 DOTA-biotin

Assessed using OLINDA dosimetry software. The estimated dose to normal organs and tumor sites will be described based on the tumor to normal organ ratios derived from dosimetry estimates coupled with the absorbed dose to normal organs based on the administered activity of yttrium Y 90 DOTA-biotin. (NCT02483000)
Timeframe: Up to 7 days after infusion

InterventioncGy/mCi (Median)
LiverSpleenLungsKidneysBone MarrowBrain
Treatment (PRIT)2.53.730.22911.43.750.229

Incidence of Toxicity, Defined According to National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.0

Descriptive statistics on the number and percent toxicities will be calculated. (NCT02483000)
Timeframe: Up to 30 days after transplant

InterventionParticipants (Count of Participants)
Serious Adverse EventsOther (Not Including Serious) Adverse Events
Treatment (PRIT)22

2-year Overall Survival Seen With Using Bortezomib in Combination With the ALL R3 Re-induction Regimen in Pediatric Patients With Relapsed or Refractory ALL or LL.

(NCT02535806)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
Treatment Arm1

Number of Subject With Adverse Events

Toxicities were assessed and graded according to CTCAE v 4.0. (NCT02535806)
Timeframe: 36 days

InterventionParticipants (Count of Participants)
Treatment Arm1

Post-induction Level of Minimal Residual Disease Seen With Using Bortezomib in Combination With the ALL R3 Re-induction Regimen in Pediatric Patients With Relapsed or Refractory ALL or LL.

Percent of Cells Positive for Minimal residual disease measured by multiparameter flow cytometry (NCT02535806)
Timeframe: 36 days

InterventionPercentage of Cells Positive for MRD (Mean)
Treatment Arm0.65

Remission Rate Seen With Using Bortezomib in Combination With the ALL R3 Re-induction Regimen in Pediatric Patients With Relapsed or Refractory ALL or LL.

Number of patients with bone marrow blast percentage <5% after treatment (NCT02535806)
Timeframe: 36 days

InterventionParticipants (Count of Participants)
Treatment Arm2

Event-Free Survival (EFS)

EFS defined as time from the treatment start till treatment failure, relapse, or death whichever comes first. Event Free Survival will be presented by median EFS, which is the time point at which the cumulative survival drops below 50%. If there is no median survival (not reached), it means the cumulative survival was more than 50%. (NCT02464657)
Timeframe: 56 days

InterventionMonths (Median)
Ph 2 Nivolumab (3mg) + Idarubicin + CytarabineNA

Maximum Tolerated Dose (MTD) of Nivolumab

MTD is highest dose level in which <2 patients of 6 develop first cycle dose-limiting toxicity (DLT). (NCT02464657)
Timeframe: 28 days

Interventionmg/kg (Number)
Ph 1 - Nivolumab (1mg) + Idarubicin + CytarabineNA
Ph 1 - Nivolumab (3mg) + Idarubicin + Cytarabine3

Overall Survival

Overall survival was defined as the time from the start of treatment to death or date of last follow-up. (NCT02464657)
Timeframe: Up to 2 years and 10 Months

InterventionMonths (Median)
Ph 2 Nivolumab (3mg) + Idarubicin + Cytarabine18.54

Relapse Free Survival

Relapse-free survival was defined as the time from treatment response to date of relapse or death, whichever occurred first. (NCT02464657)
Timeframe: Up to 2 years and10 Months

InterventionMonths (Median)
Ph 2 - Nivolumab (3mg) + Idarubicin + Cytarabine18.54

Disease-Free Survival at 2-Year Post-Transplant by Cytogenetic Risk

Kaplan-Meier estimates at 2-years post-transplant, with 95% confidence intervals based on logit limit transformation of Greenwood's variance. Outcome is death or relapse, censor is alive in continual complete remission at the date of last clinical disease assessment. (NCT00534469)
Timeframe: Kaplan-Meier estimate 2 years post treatment

InterventionProportion of pts alive in remission (Number)
Unfavorable Risk Cytogenetics0.33
Intermediate Risk Cytogenetics0.75
Favorable Risk Cytogenetics0.75
Unknown Risk: Rejected Cytogenetics Study0.73

Disease-Free Survival at 2-Year Post-Transplant

Kaplan-Meier estimates at 2-years post-transplant, with 95% confidence intervals based on logit limit transformation of Greenwood's variance. Outcome is death or relapse, censor is alive in continual complete remission at the date of last clinical disease assessment. (NCT00534469)
Timeframe: Estimate at 2 years post treatment

InterventionProportion of pts alive in remission (Number)
HD ARA-C Intermediate Cytogenetics0.65

Number of Relevant Toxicities Related to Therapy

"Events were graded using CTCAE v. 4.0. All toxicities will be monitored until the completion of therapy (up to 500 days) for patients that do not go on to bone marrow transplant. If a patient goes on to receive a bone marrow transplant, at that point, they will no longer be monitored for toxicity, as any further toxicities may be secondary to the transplant and not the study regimen.~This outcome reports those toxicities that are that were possibly, probably or definitely related to therapy. Participants were separately monitored for frequency of grade 5 events, grade 4 sepsis, grade 4 hemorrhage, and grade 4 hepatic toxicity across all patients in the stratum. Grade 4 and 5 events that are clearly and incontrovertibly due to extraneous causes or disease progression will be excluded. Higher grade events are considered more severe than lower grade." (NCT02419755)
Timeframe: From on-therapy date up to 18 months

,
Interventionevents (Number)
Grade 5: deathGrade 4: sepsisGrade 4: hemorrhageGrade 4: hepatic toxicity
Stratum 1: Myeloid Malignancies1000
Stratum 2: ALL and MLM3210

Cytogenetic Status (Based on Local Laboratory Analysis) of Participants With CR/CRi (Per EAC Assessment)

Karyotyping was required locally, at screening and at least once during the study in participants who had abnormal cytogenetics at baseline and who achieved CR/CRi. Data presented below are for participants who achieved CR/CRi per EAC and had abnormal karyotype at screening. (NCT01564784)
Timeframe: Up to approximately 4 weeks (EoT) from last dose of study drug

InterventionPercentage of Participants (Number)
Inotuzumab Ozogamicin3.7
Defined Investigator's Choice of Chemotherapy18.2

Duration of Remission (DoR) for Participants Who Achieved CR/CRi (Per Investigator Assessment)

DoR was defined as time from date of first response in responders (CR/CRi per Investigator assessment) to date of PFS event (i.e. death, progressive disease [objective progression, relapse from CR/CRi or treatment discontinuation due to global deterioration of health status] or starting new induction therapy or post-therapy stem cell transplant [SCT] without achieving CR/CRi). Responders without PFS events were censored at the last valid disease assessment including follow-up. (NCT01564784)
Timeframe: Up to 2 years from randomization

InterventionMonths (Median)
Inotuzumab Ozogamicin5.4
Defined Investigator's Choice of Chemotherapy3.5

Overall Survival (OS)

OS was defined as the time from randomization to date of death due to any cause. Participants last known to be alive were censored at date of last contact. (NCT01564784)
Timeframe: Up to 5 years after randomization or 2 years from randomization of the last participant, whichever occurs first.

InterventionMonths (Median)
Inotuzumab Ozogamicin7.7
Defined Investigator's Choice of Chemotherapy6.2

Percentage of Participants Achieving MRD Negativity (Based on Central Laboratory Analysis) in Participants Achieving a CR/CRi (Per EAC Assessment)

MRD analysis was performed at least once in participants with prior assessment of CR or CRi. Bone marrow aspirates, collected at screening and during the study, were sent to the central laboratory and analyzed using multiparametric flow cytometry. The antibody combinations were designed to maximize discrimination between normal and abnormal cells of B-cell lineage and similar maturational stage and included antibodies detecting cluster of differentiation (CD) 9, CD10, CD13, CD19, CD20, CD33, CD34, CD38, CD45, CD58, CD66c, and CD123. A peripheral blood sample was provided if a participant had an inadequate bone marrow aspirate at screening. MRD negativity was considered to have been achieved if the lowest value of MRD from the first date of CR/CRi to EoT was <1 × 10^-4 blasts/nucleated cells. (NCT01564784)
Timeframe: Up to approximately 4 weeks (EoT) from last dose of study drug

InterventionPercentage of Participants (Number)
Inotuzumab Ozogamicin78.4
Defined Investigator's Choice of Chemotherapy28.1

Percentage of Participants Who Had a Hematopoietic Stem-Cell Transplant (HSCT)

HSCT rate was defined as the percentage of participants who underwent SCT following treatment with inotuzumab ozogamicin or Investigator's choice of chemotherapy. (NCT01564784)
Timeframe: Up to 19 weeks from last dose

InterventionPercentage of Participants (Number)
Inotuzumab Ozogamicin42.7
Defined Investigator's Choice of Chemotherapy11.1

Percentage of Participants With Hematologic Remission (Complete Remission [CR]/Complete Remission With Incomplete Hematologic Recovery [CRi]) as Assessed by the Endpoint Adjudication Committee (EAC)

CR was the disappearance of leukemia indicated by less than (<) 5 percent (%) marrow blasts & absence of peripheral blood leukemic blasts, with recovery of hematopoiesis defined by absolute neutrophil count (ANC) greater than or equal to (≥)1000 per microliter (/μL) & platelets ≥100,000/μL. C1 extramedullary disease status (i.e. complete disappearance of measurable & non-measurable extramedullary disease with the following exceptions: for participants with at least 1 measurable lesion, all nodal masses greater than (>) 1.5 centimeters (cm) in greatest transverse diameter (GTD) at baseline must have regressed to less than or equal to (≤) 1.5 cm in GTD; all nodal masses ≥1 cm & ≤1.5 cm in GTD at baseline must have regressed to <1 cm GTD or reduced by 75% in sum of products of greatest diameters, no new lesions, spleen & other previously enlarged organs must have regressed in size & must not be palpable) was required. CRi was defined as CR except ANC <1000/μL &/or platelets <100,000/μL. (NCT01564784)
Timeframe: Screening, Day 16 to 28 of Cycles 1, 2 and 3, then every 1 to 2 cycles (or as clinically indicated) up to approximately 4 weeks (end of treatment [EoT]) from the last dose

InterventionPercentage of Participants (Number)
Inotuzumab Ozogamicin80.7
Defined Investigator's Choice of Chemotherapy29.4

Percentage of Participants With Veno-Occlusive Liver Disease (VOD)/Sinusoidal Obstruction Syndrome (SOS) Following Post Study HSCT

VOD/SOS was defined as the occurrence of 2 out of the following 3 clinical criteria: 1) total serum bilirubin level >34 micromoles per liter (μmol/L) (>2.0 milligrams per deciliter [mg/dL]), 2) an increase in liver size from baseline or development of right upper quadrant pain of liver origin and 3) sudden weight gain >2.5% (eg, within a 72 hour period) because of fluid accumulation in the weeks following infusion of study drug or chemotherapy, or HSCT conditioning/preparative therapy, or development of ascites not present at baseline following such exposures AND the absence of other explanations for these signs and symptoms, OR development of bilirubin elevation, weight gain, or hepatomegaly plus histologic abnormalities on liver biopsy demonstrating hepatocyte necrosis in zone 3 of the liver acinus, sinusoidal fibrosis, and centrilobular hemorrhage, with or without fibrosis of the terminal hepatic venules. (NCT01564784)
Timeframe: Up to 2 years from randomization

InterventionPercentage of Participants (Number)
Inotuzumab Ozogamicin22.8
Defined Investigator's Choice of Chemotherapy8.6

Progression-Free Survival (PFS)

PFS was defined as time from date of randomization to earliest date of the following events: death, progressive disease (objective progression, relapse from CR/CRi or treatment discontinuation due to global deterioration of health status) and starting new induction therapy or post-therapy SCT without achieving CR/CRi. Participants without a PFS event at time of analysis were censored at the last valid disease assessment. In addition, participants with documentation of an event after an unacceptably long interval (>28 weeks if there was post-baseline disease assessment, or >12 weeks if there was no post-baseline assessment) since the previous disease assessment were censored at the time of the previous assessment (date of randomization if no post-baseline assessment). Post-study treatment follow-up disease assessments was included. Kaplan-Meier method used and 2-sided 95% confidence interval (CI) calculated based on the Brookmeyer and Crowley method. (NCT01564784)
Timeframe: Up to 2 years from randomization

InterventionMonths (Median)
Inotuzumab Ozogamicin5.0
Defined Investigator's Choice of Chemotherapy1.7

Change From Baseline in EQ-5D VAS

"The EQ-5D self-report questionnaire is a standardized measure of health status developed by the EuroQoL Group. It consists of the EQ-5D descriptive system and a visual analogue scale (VAS), EQ-VAS. The EQ-5D descriptive system measures a participants' health state on 5 dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension has 3 levels, reflecting no problems, some problems, and extreme problems. The EQ-VAS records the respondent's self-rated health on a scale from 0 (worst imaginable health state) to 100 (best imaginable health state); higher scores indicate a better health state." (NCT01564784)
Timeframe: Day 1 of each cycle prior to dosing and EoT

,
InterventionScore on a scale (Mean)
Cycle 2, Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1EoT
Defined Investigator's Choice of Chemotherapy5.9019.6744.00NANA-0.52
Inotuzumab Ozogamicin5.818.137.137.6215.094.62

Change From Baseline in European Organization for Research and Treatment of Cancer Quality of Life Questionnaire, Core 30 (EORTC QLQ-C30) Score

This questionnaire comprised 30 questions within which are 9 multi-item scales & 6 single-item measures. There are 5 functional scales; physical, role, cognitive, emotional & social, 3 symptom scales; fatigue, pain, & nausea & vomiting, & a global health status/quality of life (QoL) scale. There are 5 single item measures assessing additional symptoms commonly reported by cancer patients (loss of appetite, insomnia, constipation, diarrhea, & dyspnea) & a single item concerning perceived financial impact of the disease. Most questions used a 4 point scale (1='not at all' to 4='very much'); 2 questions used a 7-point scale (1='very poor' to 7='excellent'). Scores were averaged & transformed to a scale ranging from 0 to 100; a higher score indicates a better level of functioning or greater degree of symptoms. (NCT01564784)
Timeframe: Day 1 of each cycle prior to dosing and EoT

,
InterventionScore on a scale (Mean)
Physical Functioning C2D1Physical Functioning C3D1Physical Functioning C4D1Physical Functioning C5D1Physical Functioning C6D1Physical Functioning EoTRole Functioning C2D1Role Functioning C3D1Role Functioning C4D1Role Functioning C5D1Role Functioning C6D1Role Functioning EoTEmotional Functioning C2D1Emotional Functioning C3D1Emotional Functioning C4D1Emotional Functioning C5D1Emotional Functioning C6D1Emotional Functioning EoTCognitive Functioning C2D1Cognitive Functioning C3D1Cognitive Functioning C4D1Cognitive Functioning C5D1Cognitive Functioning C6D1Cognitive Functioning EoTSocial Functioning C2D1Social Functioning C3D1Social Functioning C4D1Social Functioning C5D1Social Functioning C6D1Social Functioning EoTGlobal Health Status C2D1Global Health Status C3D1Global Health Status C4D1Global Health Status C5D1Global Health Status C6D1Global Health Status EoTDyspnoea C2D1Dyspnoea C3D1Dyspnoea C4D1Dyspnoea C5D1Dyspnoea C6D1Dyspnoea EoTInsomnia C2D1Insomnia C3D1Insomnia C4D1Insomnia C5D1Insomnia C6D1Insomnia EoTAppetite Loss C2D1Appetite Loss C3D1Appetite Loss C4D1Appetite Loss C5D1Appetite Loss C6D1Appetite Loss EoTConstipation C2D1Constipation C3D1Constipation C4D1Constipation C5D1Constipation C6D1Constipation EoTDiarrhoea C2D1Diarrhoea C3D1Diarrhoea C4D1Diarrhoea C5D1Diarrhoea C6D1Diarrhoea EoTFinancial Difficulties C2D1Financial Difficulties C3D1Financial Difficulties C4D1Financial Difficulties C5D1Financial Difficulties C6D1Financial Difficulties EoTFatigue C2D1Fatigue C3D1Fatigue C4D1Fatigue C5D1Fatigue C6D1Fatigue EotNausea and Vomiting C2D1Nausea and Vomiting C3D1Nausea and Vomiting C4D1Nausea and Vomiting C5D1Nausea and Vomiting C6D1Nausea and Vomiting EoTPain C2D1Pain C3D1Pain C4D1Pain C5D1Pain C6D1Pain EoT
Defined Investigator's Choice of Chemotherapy0.32-13.330.00NANA-8.17-1.59-11.110.00NANA-12.374.76-19.440.00NANA4.350.00-5.5616.67NANA0.54-2.38-27.780.00NANA-2.150.40-16.678.33NANA-0.40-7.9411.110.00NANA0.541.590.000.00NANA0.003.170.000.00NANA11.830.000.000.00NANA-0.54-1.59-11.110.00NANA3.760.000.000.00NANA2.195.8222.22-11.11NANA5.73-0.790.00-16.67NANA3.49-7.14-5.56-33.33NANA-7.26
Inotuzumab Ozogamicin0.483.155.3311.527.22-3.385.4511.8116.6712.8816.671.325.217.415.696.822.78-0.914.055.794.580.76-8.330.834.205.5610.4212.8816.671.823.989.3811.258.710.690.00-5.41-7.41-12.50-3.03-2.78-2.31-2.40-9.26-7.50-4.55-11.11-2.31-4.20-8.33-11.67-6.062.78-1.32-1.210.47-2.500.005.562.64-3.30-5.09-0.83-9.52-2.782.31-1.800.47-1.67-3.03-5.560.33-4.10-8.33-9.17-7.320.00-0.330.15-4.63-3.332.270.00-0.17-8.86-8.56-4.170.76-2.78-1.98

Change From Baseline in EuroQol 5 Dimension Health Questionnaire (EQ-5D) Index Score

"The EQ-5D self-report questionnaire is a standardized measure of health status developed by the EuroQoL Group. It consists of the EQ-5D descriptive system and a visual analogue scale (VAS), EQ-VAS. The EQ-5D descriptive system measures a participants' health state on 5 dimensions: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension has 3 levels, reflecting no problems, some problems, and extreme problems. The EQ-VAS records the respondent's self-rated health on a scale from 0 (worst imaginable health state) to 100 (best imaginable health state); higher scores indicate a better health state. EQ-5D summary index is obtained with a formula that weights each level of the dimensions. The index-based score is interpreted along a continuum of 0 (death) to 1 (perfect health)." (NCT01564784)
Timeframe: Day 1 of each cycle prior to dosing and EoT

,
InterventionScore on a scale (Mean)
Cycle 2, Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1EoT
Defined Investigator's Choice of Chemotherapy0.02-0.080.00NANA-0.04
Inotuzumab Ozogamicin0.000.010.040.040.03-0.01

Maximum Observed Inotuzumab Ozogamicin Serum Concentration (Cmax) and Pre-Dose Inotuzumab Ozogamicin Serum Concentration (Ctrough) Following Single and Multiple Dosing

Blood samples were collected and analyzed for inotuzumab ozogamicin serum concentrations using a validated high performance liquid chromatography with tandem mass spectrometry (HPLC/MS/MS) method with a lower limit of quantification of 1.0 nanograms per milliliter (ng/mL). Cmax was the maximum observed concentration occurring between 0-8 hours post-dose. Ctrough was the concentration prior to subsequent dose (pre-dose) occurring after 8 hours. n = number of observations (non-missing concentrations). (NCT01564784)
Timeframe: Days 1, 4, 8, and 15 of Cycle 1, Days 1 and 8 of Cycle 2 and Day 1 of Cycle 4

Interventionng/mL (Mean)
Cmax (Cycle 1 Day 1, 1 hour post-dose) (n=128)Ctrough (Cycle 4 Day 1, pre-dose) (n=46)Cmax (Cycle 4 Day 1, 1 hour post-dose) (n=37)
Inotuzumab Ozogamicin21157.9308

Number of Participants With Objective Response

Objective response: Complete Response/Remission (CR) defined as a bone marrow with 5% or fewer blasts and peripheral blood count with an absolute neutrophil count of 10^9/Liters or more and platelet count of 100*10^9/Liters or more; Complete Response with Platelets/remission without platelet recovery (CRp) defined as a complete response except for a platelet less than 100*10^9/Liters and transfusion independent; and Partial Response/Remission defined as peripheral blood count recovery as for CR with decrease in marrow blasts >/= 50% and not more than 6-25% abnormal cells in the marrow. (NCT00480987)
Timeframe: After 2 months

InterventionParticipants (Number)
Complete responseComplete response with plateletsPartial response
Oxaliplatin + Cytarabine + Fludarabine320

Number of Participants Adherent to Readmission Recommendations

This measure will record the number of subjects who complete as 100% compliance with medical provider recommendations to be admitted to the hospital (except for reasons of adopting a palliative or hospice approach to care). Subjects not achieving a 100% rate of compliance will be considered non-adherent to the recommendations. (NCT03988205)
Timeframe: Day 60

InterventionParticipants (Count of Participants)
Intervention3

Number of Participants Adherent to Recommendations

This measure will record the number subjects who adhere to outpatient follow-up appointments and readmission recommendations. Subjects completing 95% of their recommended outpatient visits will be considered to be adherent to the recommendations. (NCT03988205)
Timeframe: Day 60

InterventionParticipants (Count of Participants)
Intervention3

Probability of Event-free Survival (EFS)

"For EFS, relapse and second malignancies are considered as failures in addition to death in complete remission. The time to EFS will be set to 0 for patients who fail to achieve complete remission. Kaplan-Meier estimates of the OS and EFS curves are computed, along with estimates of standard errors by Peto's method.~Please note the unit of measurement of probabilities are percentages." (NCT01451515)
Timeframe: Two years post therapy.

Interventionpercentage of event-free patients (Number)
Stratum 191.7
Stratum 271.4
Stratum 3100
All Enrollments86.96

Probability of Overall Survival (OS)

"For OS, only deaths are considered failures for OS. Kaplan-Meier estimates of the OS curves are computed along with estimates of standard errors by Peto's method.~Please note the unit of measurement of probabilities are percentages." (NCT01451515)
Timeframe: Two years post therapy.

Interventionpercentage of patients alive (Number)
Stratum 191.7
Stratum 271.4
Stratum 3100
All Enrollments86.96

Minimal Disseminated Disease (MDD)

Detectable disease in bone marrow or blood: A binary measure, positive (detectable), negative (non-detectable) (NCT01451515)
Timeframe: At Diagnosis

,,
Interventionparticipants (Number)
NegativePositive
Stratum 141
Stratum 223
Stratum 304

Minimal Residual Disease (MRD)

Detectable disease in bone marrow or blood: A binary measure, positive (detectable), negative (non-detectable) (NCT01451515)
Timeframe: Day 8

,,
Interventionparticipants (Number)
NegativePositive
Stratum 180
Stratum 241
Stratum 304

Area Under the Ulocuplumab Concentration-time Curve From Time Zero to the Last Quantifiable Concentration [AUC(0-T)] - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration.~EOT = end of treatment~AUC(0-T) calculated by log- and linear-trapezoidal summation~Measure type and method of dispersion are Geometric mean and %CV, respectively" (NCT02305563)
Timeframe: Cycle 1 Day 1

Interventionµg.h/mL (Geometric Mean)
ULO 600mg + LDAC - Ph19455.529
ULO 800mg + LDAC - Ph121158.725
ULO 800mg + LDAC - Ph28152.698
ULO 1000mg + LDAC - Ph213744.382
LDAC - Ph216502.463

Area Under the Ulocuplumab Concentration-time Curve in One Dosing Interval [AUC(TAU)] - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration.~EOT = end of treatment~Measure type and method of dispersion are Geometric mean and %CV, respectively" (NCT02305563)
Timeframe: Cycle 1 Day 1

Interventionµg.h/mL (Geometric Mean)
ULO 600mg + LDAC - Ph19455.529
ULO 800mg + LDAC - Ph121158.725
ULO 800mg + LDAC - Ph210082.624
ULO 1000mg + LDAC - Ph213826.833
LDAC - Ph214257.807

Best Overall Response (BOR) - Phase 1

Investigator assessed best overall response prior to the initiation of any alternative therapy for Phase 1 participants. (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionParticipants (Number)
ULO 600mg + LDAC - Ph11
ULO 800mg + LDAC - Ph13

Best Overall Response (BOR) - Phase 2

"The phase 2 primary endpoint was based on the rate of Complete Remission (CR/CRi) prior to the initiation of any alternative therapy (including any subsequent ulocuplumab 800 mg for participants in the LDAC alone arm). The phase 2 primary analysis was conducted after all participants had an opportunity for 6 months of follow-up.~Complete remission rate: CR + CRi, confidence interval based on the Clopper and Pearson method.~CR = complete response CRi = complete response, incomplete blood count" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionPercentage of participants (Number)
ULO 800mg + LDAC - Ph215.4
ULO 1000mg + LDAC - Ph27.1
LDAC - Ph225.0

Duration of Response in Participants With CR/CRi Treated With LDAC Only - Phase 2

This phase 2 secondary endpoint was based on the duration of complete remission prior to the initiation of any alternative therapy (including any subsequent ulocuplumab 800 mg for participants in the LDAC alone arm). The phase 2 analysis was conducted after all participants had an opportunity for 6 months of follow-up. (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionMonths (Median)
LDAC - Ph25.7

Duration of Response in Participants With CR/CRi Treated With Ulocuplumab at Two Different Dose Levels 800 mg and 1000 mg in Combination With LDAC - Phase 2

This phase 2 secondary endpoint was based on the duration of complete remission prior to the initiation of any alternative therapy (including any subsequent ulocuplumab 800 mg for participants in the LDAC alone arm). The phase 2 analysis was conducted after all participants had an opportunity for 6 months of follow-up. (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionMonths (Median)
ULO 800mg + LDAC - Ph22.4
ULO 1000mg + LDAC - Ph24.3

Maximum Observed Serum Concentration (Cmax) - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration~EOT = end of treatment~Measure type and method of dispersion are Geometric mean and %CV, respectively" (NCT02305563)
Timeframe: Cycle 1 Day 1

Interventionµg/mL (Geometric Mean)
ULO 600mg + LDAC - Ph1219.354
ULO 800mg + LDAC - Ph1265.294
ULO 800mg + LDAC - Ph2183.456
ULO 1000mg + LDAC - Ph2256.906
LDAC - Ph2212.564

Number of Deaths - Phase 1

The number of participants who died. (NCT02305563)
Timeframe: From first dose to 30 days post last dose

InterventionParticipants (Number)
ULO 600mg + LDAC - Ph10
ULO 800mg + LDAC - Ph10

Number of Deaths- Phase 2

"The number of participants who died.~Safety data are evaluated for deaths, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionParticipants (Number)
ULO 800mg + LDAC - Ph219
ULO 1000mg + LDAC - Ph210
LDAC - Ph216

Number of Participants With >= Grade 3 AEs - Phase 1

"The number of participants with an on-study adverse event >= Grade level 3.~Safety data are evaluated for >= Grade 3 AEs, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose to 30 days post last dose

InterventionParticipants (Number)
ULO 600mg + LDAC - Ph13
ULO 800mg + LDAC - Ph13

Number of Participants With Adverse Events (AEs) - Phase 1

"The number of participants with an on-study adverse event (AE).~Safety data are evaluated for AEs, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose to 30 days post last dose

InterventionParticipants (Number)
ULO 600mg + LDAC - Ph13
ULO 800mg + LDAC - Ph13

Number of Participants With AEs - Phase 2

"The number of participants with an on-study adverse event (AE).~Safety data are evaluated for AEs, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionParticipants (Number)
ULO 800mg + LDAC - Ph225
ULO 1000mg + LDAC - Ph214
LDAC - Ph222

Number of Participants With AEs Leading to Discontinuation - Phase 1

"The number of participants with an on-study adverse event (AE) leading to discontinuation.~Safety data are evaluated for AEs leading to discontinuation, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose to 30 days post last dose

InterventionParticipants (Number)
ULO 600mg + LDAC - Ph10
ULO 800mg + LDAC - Ph10

Number of Participants With AEs Leading to Discontinuation - Phase 2

"The number of participants with an on-study adverse event (AE) leading to discontinuation.~Safety data are evaluated for AEs leading to discontinuation, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionParticipants (Number)
ULO 800mg + LDAC - Ph27
ULO 1000mg + LDAC - Ph23
LDAC - Ph24

Number of Participants With Anti-drug Antibodies (ADA) Positive for Ulocuplumab - Phases 1 and 2

Serum samples from ulocuplumab treated participants were evaluated for the presence of anti-ulocuplumab antibodies (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionParticipants (Number)
ULO 600mg + LDAC - Ph10
ULO 800mg + LDAC - Ph10
ULO 800mg + LDAC - Ph26
ULO 1000mg + LDAC - Ph20

Number of Participants With Dose-Limiting Toxicities (DLTs) in Treatment Cycle 1 - Phase 1

Safety data evaluated for DLTs. DLTs and all other toxicities were defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03). DLTs were defined based upon events that were considered to be related to ulocuplumab in combination with LDAC and that occurred during the first cycle of drug administration (28 days). (NCT02305563)
Timeframe: From first dose to end of cycle 1 (28 days)

InterventionParticipants (Number)
ULO 600mg + LDAC - Ph10
ULO 800mg + LDAC - Ph10

Number of Participants With SAEs - Phase 2

"The number of participants with an on-study serious adverse event (SAE).~Safety data are evaluated for SAEs, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionParticipants (Number)
ULO 800mg + LDAC - Ph221
ULO 1000mg + LDAC - Ph28
LDAC - Ph215

Number of Participants With Serious Adverse Events (SAEs) - Phase 1

"The number of participants with an on-study serious adverse event (SAE).~Safety data are evaluated for SAEs, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose to 30 days post last dose

InterventionParticipants (Number)
ULO 600mg + LDAC - Ph12
ULO 800mg + LDAC - Ph11

Overall Rate of Remission in Participants Treated With Ulocuplumab at Two Different Dose Levels 800 mg and 1000 mg in Combination With LDAC - Phase 2

"This phase 2 secondary endpoint was based on the rate of Overall Remission (OR=PR+CR +CRi) prior to the initiation of any alternative therapy (including any subsequent ulocuplumab 800 mg for participants in the LDAC alone arm). The phase 2 analysis was conducted after all participants had an opportunity for 6 months of follow-up.~Overall remission rate: CR + CRi, + PR confidence interval based on the Clopper and Pearson method.~CR = complete response CRi = complete response, incomplete blood count PR = partial remission" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionPercentage of participants (Number)
ULO 800mg + LDAC - Ph219.2
ULO 1000mg + LDAC - Ph27.1

Overall Survival (OS) - Phases 1 and 2

OS is defined as the time between the first date of treatment and the date of death due to any cause. A participant who has not died was be censored at the last known alive date. (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionMonths (Median)
ULO 800mg + LDAC - Ph23.3
ULO 1000mg + LDAC - Ph23.0
LDAC - Ph26.9

Time of Maximum Observed Ulocuplumab Serum Concentration (Tmax) - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration.~EOT = end of treatment" (NCT02305563)
Timeframe: Cycle 1 Day 1

Interventionhour (H) (Median)
ULO 600mg + LDAC - Ph11.850
ULO 800mg + LDAC - Ph11.933
ULO 800mg + LDAC - Ph21.500
ULO 1000mg + LDAC - Ph22.117
LDAC - Ph22.03

Change From Baseline of Electrocardiogram (ECG) Endpoints: Heart Rate - Phases 1 and 2

"Change from baseline of ECG endpoints~Heart rate measured in beats per minute (bpm)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline bpm (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 8, Day 1Cycle 10, Day 1Cycle 11, Day 1Cycle 13, Day 1Cycle 15, Day 1
ULO 600mg + LDAC - Ph126.0-2.3-1.0-6.0-6.0-1.0-3.0-12.0-9.0-10.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: Heart Rate - Phases 1 and 2

"Change from baseline of ECG endpoints~Heart rate measured in beats per minute (bpm)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline bpm (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 10, Day 1Cycle 12, Day 1Cycle 13, Day 1Cycle 14, Day 1
ULO 800mg + LDAC - Ph21.85.8-0.14.36.8-3.50.77.09.35.08.014.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: Heart Rate - Phases 1 and 2

"Change from baseline of ECG endpoints~Heart rate measured in beats per minute (bpm)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline bpm (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 10, Day 1Cycle 11, Day 1Cycle 12, Day 1Cycle 13, Day 1Cycle 15, Day 1Cycle 16, Day 1
LDAC - Ph2-3.01.71.38.87.46.50.80.62.0-0.37.04.08.018.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: Heart Rate - Phases 1 and 2

"Change from baseline of ECG endpoints~Heart rate measured in beats per minute (bpm)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline bpm (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 9, Day 1Cycle 10, Day 1
ULO 800mg + LDAC - Ph14.00.0-16.01.5-6.0-11.7-17.0-16.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: Heart Rate - Phases 1 and 2

"Change from baseline of ECG endpoints~Heart rate measured in beats per minute (bpm)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline bpm (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 11, Day 1Cycle 12, Day 1
ULO 1000mg + LDAC - Ph2-0.912.711.010.512.011.03.010.019.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: PR Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~PR interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 8, Day 1Cycle 10, Day 1Cycle 11, Day 1Cycle 13, Day 1Cycle 15, Day 1
ULO 600mg + LDAC - Ph14.08.014.0-2.0-44.04.0-4.04.0-2.00.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: PR Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~PR interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 10, Day 1Cycle 12, Day 1Cycle 13, Day 1Cycle 14, Day 1
ULO 800mg + LDAC - Ph21.1-5.3-16.3-7.6-12.8-6.8-5.3-17.0-11.3-60.0-44.0-54.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: PR Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~PR interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 10, Day 1Cycle 11, Day 1Cycle 12, Day 1Cycle 13, Day 1Cycle 15, Day 1Cycle 16, Day 1
LDAC - Ph21.2-1.9-11.9-24.6-3.4-8.3-9.40.61.0-1.7-5.03.0-10.0-2.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: PR Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~PR interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 9, Day 1Cycle 10, Day 1
ULO 800mg + LDAC - Ph1-14.0-12.0-13.0-16.0-19.3-17.3-10.0-2.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: PR Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~PR interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 11, Day 1Cycle 12, Day 1
ULO 1000mg + LDAC - Ph2-5.1-2.8-10.0-11.0-2.0-6.06.04.0-4.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QRS Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QRS interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 8, Day 1Cycle 10, Day 1Cycle 11, Day 1Cycle 13, Day 1Cycle 15, Day 1
ULO 600mg + LDAC - Ph10.03.30.08.06.02.02.06.02.06.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QRS Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QRS interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 10, Day 1Cycle 12, Day 1Cycle 13, Day 1Cycle 14, Day 1
ULO 800mg + LDAC - Ph25.1-1.32.71.8-1.8-3.30.7-2.0-2.3-6.0-12.0-10.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QRS Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QRS interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 10, Day 1Cycle 11, Day 1Cycle 12, Day 1Cycle 13, Day 1Cycle 15, Day 1Cycle 16, Day 1
LDAC - Ph24.81.46.4-1.45.4-1.52.60.09.59.010.03.58.07.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QRS Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QRS interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 9, Day 1Cycle 10, Day 1
ULO 800mg + LDAC - Ph1-2.0-2.01.0-4.0-2.7-2.7-1.0-4.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QRS Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QRS interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 11, Day 1Cycle 12, Day 1
ULO 1000mg + LDAC - Ph20.52.0-3.32.0-10.0-8.0-6.0-4.0-8.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QT Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QT interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 8, Day 1Cycle 10, Day 1Cycle 11, Day 1Cycle 13, Day 1Cycle 15, Day 1
ULO 600mg + LDAC - Ph1-41.010.0-60.012.02.02.06.016.020.036.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QT Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QT interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 10, Day 1Cycle 12, Day 1Cycle 13, Day 1Cycle 14, Day 1
ULO 800mg + LDAC - Ph24.5-15.410.12.2-11.811.82.0-23.0-18.3-4.0-40.0-44.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QT Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QT interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 10, Day 1Cycle 11, Day 1Cycle 12, Day 1Cycle 13, Day 1Cycle 15, Day 1Cycle 16, Day 1
LDAC - Ph214.213.7-1.4-27.6-3.6-8.80.8-24.4-6.07.0-6.5-15.0-19.0-105.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QT Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QT interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 6, Day 1Cycle 7, Day 1Cycle 9, Day 1Cycle 10, Day 1
ULO 800mg + LDAC - Ph1-5.07.035.013.06.029.337.014.0

Change From Baseline of Electrocardiogram (ECG) Endpoints: QT Interval - Phases 1 and 2

"Change from baseline of ECG endpoints~QT interval measured in milliseconds (msec)" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

Interventionchange from baseline msec (Mean)
Cycle 2 Day 1Cycle 3, Day 1Cycle 4, Day 1Cycle 5, Day 1Cycle 7, Day 1Cycle 8, Day 1Cycle 9, Day 1Cycle 11, Day 1Cycle 12, Day 1
ULO 1000mg + LDAC - Ph210.4-10.7-12.7-5.0-8.0-24.0-14.0-22.0-36.0

Number of Participants With Laboratory Abnormalities - Phase 1

"The number of participants with an on-study laboratory abnormality.~Safety data are evaluated for laboratory abnormalities, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03).~grades 1, 2, 3, 4, 5, unknown, with 5 being the worst outcome" (NCT02305563)
Timeframe: From first dose to 30 days post last dose

,
InterventionParticipants (Number)
ABSOLUTE NEUTROPHIL COUNT - grade 3ABSOLUTE NEUTROPHIL COUNT - grade 4ALANINE AMINOTRANSFERASE - grade 0ALANINE AMINOTRANSFERASE - grade 1ALBUMIN - grade 0ALBUMIN - grade 1ALBUMIN - grade 2ALKALINE PHOSPHATASE - grade 0ALKALINE PHOSPHATASE grade 1ASPARTATE AMINOTRANSFERASE - grade 0ASPARTATE AMINOTRANSFERASE - grade 1BILIRUBIN, TOTAL - grade 0BILIRUBIN, TOTAL - grade 2CALCIUM, TOTAL - grade 0CALCIUM, TOTAL - grade 1CALCIUM, TOTAL - grade 2CREATINE KINASE - grade 0CREATININE - grade 0CREATININE - grade 1FIBRINOGEN - grade 0GLUCOSE, FASTING SERUM - grade 0GLUCOSE, FASTING SERUM - grade 1GLUCOSE, FASTING SERUM - grade 2HEMOGLOBIN - grade 2HEMOGLOBIN - grade 3LEUKOCYTES - grade 0LEUKOCYTES - grade 3LEUKOCYTES - grade 4LIPASE, TOTAL (COLORIMETRIC ASSAY) - grade 0LIPASE, TOTAL (COLORIMETRIC ASSAY) - grade 1LIPASE, TOTAL (COLORIMETRIC ASSAY) - grade 3LYMPHOCYTES (ABSOLUTE) - grade 0LYMPHOCYTES (ABSOLUTE) - grade 1LYMPHOCYTES (ABSOLUTE) - grade 2LYMPHOCYTES (ABSOLUTE) - grade 3NEUTROPHILS (ABSOLUTE) - grade 3NEUTROPHILS (ABSOLUTE) - grade 4PHOSPHORUS, INORGANIC - grade 0PHOSPHORUS, INORGANIC - grade 3PLATELET COUNT - grade 3PLATELET COUNT - grade 4POTASSIUM, SERUM - grade 0POTASSIUM, SERUM - grade 1POTASSIUM, SERUM - grade 3SODIUM, SERUM - grade 0SODIUM, SERUM - grade 1SODIUM, SERUM - grade 3URIC ACID - grade 0URIC ACID - grade 1
ULO 600mg + LDAC - Ph10321102213030102321112012012210002103300310220130
ULO 800mg + LDAC - Ph11221021212121111330010212111102111012211221012021

Number of Participants With Laboratory Abnormalities - Phase 2

"The number of participants with an on-study laboratory abnormality, assessed from Grade 1-4 Serum Chemistry, Electrolytes, and Hematology Laboratory Test results, with grade 4 being the worst.~Safety data are evaluated for laboratory abnormalities, defined and evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events Version 4.03 (NCI CTCAE v4.03)." (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

,,
InterventionParticipants (Number)
ALANINE AMINOTRANSFERASE (ALT), grade 1ALANINE AMINOTRANSFERASE (ALT), grade 2ALANINE AMINOTRANSFERASE (ALT), grade 3ALKALINE PHOSPHATASE (ALP), grade 1ALKALINE PHOSPHATASE (ALP), grade 2ASPARTATE AMINOTRANSFERASE (AST), grade 1ASPARTATE AMINOTRANSFERASE (AST), grade 2ASPARTATE AMINOTRANSFERASE (AST), grade 3BILIRUBIN, TOTAL, grade 1BILIRUBIN, TOTAL, grade 2BILIRUBIN, TOTAL, grade 3CREATININE, grade 1CREATININE, grade 2CALCIUM, TOTAL, grade 1CALCIUM, TOTAL, grade 2CALCIUM, TOTAL, grade 3PHOSPHORUS, INORGANIC, grade 1PHOSPHORUS, INORGANIC, grade 2PHOSPHORUS, INORGANIC, grade 3PHOSPHORUS, INORGANIC, grade 4POTASSIUM, SERUM, grade 1POTASSIUM, SERUM, grade 2POTASSIUM, SERUM, grade 3POTASSIUM, SERUM, grade 4SODIUM, SERUM, grade 1SODIUM, SERUM, grade 2SODIUM, SERUM, grade 3HEMOGLOBIN, grade 2HEMOGLOBIN, grade 3PLATELET COUNT, grade 2PLATELET COUNT, grade 3PLATELET COUNT, grade 4ABSOLUTE NEUTROPHIL COUNT, grade 1ABSOLUTE NEUTROPHIL COUNT, grade 2ABSOLUTE NEUTROPHIL COUNT, grade 3ABSOLUTE NEUTROPHIL COUNT, grade 4LEUKOCYTES, grade 1LEUKOCYTES, grade 2LEUKOCYTES, grade 3LEUKOCYTES, grade 4LYMPHOCYTES (ABSOLUTE), grade 1LYMPHOCYTES (ABSOLUTE), grade 2LYMPHOCYTES (ABSOLUTE), grade 3LYMPHOCYTES (ABSOLUTE), grade 4NEUTROPHILS (ABSOLUTE), grade 1NEUTROPHILS (ABSOLUTE), grade 2NEUTROPHILS (ABSOLUTE), grade 3NEUTROPHILS (ABSOLUTE), grade 4
LDAC - Ph242082420110623701120504070441713170111610510177101116
ULO 1000mg + LDAC - Ph25123580123052351020131213134100113110100441410002010
ULO 800mg + LDAC - Ph25029171034175750032040119002220123203183368375011318

Rate of Complete Remission (CR/CRi) and Overall Rate of Remission in Participants Treated With LDAC Only - Phase 2

"This phase 2 secondary endpoint was based on the rate of Complete Remission (CR/CRi) and rate of Overall Remission (OR=PR+CR +CRi) prior to the initiation of any alternative therapy (including any subsequent ulocuplumab 800 mg for participants in the LDAC alone arm). The phase 2 analysis was conducted after all participants had an opportunity for 6 months of follow-up.~Overall remission rate: CR + CRi, + PR confidence interval based on the Clopper and Pearson method.~CR = complete response CRi = complete response, incomplete blood count PR = partial remission" (NCT02305563)
Timeframe: From first dose until a minimum follow-up of up to 2 months

InterventionPercentage of participants (Number)
CR/CRiOverall remission rate
LDAC - Ph225.025.0

Trough Observed Serum Concentration (Ctrough) - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration.~EOT = end of treatment~Measure type and method of dispersion are Geometric mean and %CV, respectively" (NCT02305563)
Timeframe: Days 1, 8, 15 for cycle 1; Days 8, 15 for cycle 2; Days 1, 8 for cycles 3-5; Day 1 every 4th cycle thereafter; EOT; 30 days post last dose (follow-up)

Interventionµg/mL (Geometric Mean)
Cycle 1 Day 1Cycle 1 Day 8Cycle 1 Day 15Cycle 2 Day 8Cycle 2 Day 15Cycle 3 Day 1Cycle 3 Day 8Cycle 4 Day 1Cycle 4 Day 8Cycle 5 Day 1Cycle 5 Day 8
ULO 800mg + LDAC - Ph10.10075.890125.79796.333126.02978.218129.58080.409133.65032.621124.996

Trough Observed Serum Concentration (Ctrough) - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration.~EOT = end of treatment~Measure type and method of dispersion are Geometric mean and %CV, respectively" (NCT02305563)
Timeframe: Days 1, 8, 15 for cycle 1; Days 8, 15 for cycle 2; Days 1, 8 for cycles 3-5; Day 1 every 4th cycle thereafter; EOT; 30 days post last dose (follow-up)

Interventionµg/mL (Geometric Mean)
Cycle 1 Day 1Cycle 1 Day 8Cycle 1 Day 15Cycle 2 Day 8Cycle 2 Day 15Cycle 3 Day 1Cycle 3 Day 8Cycle 4 Day 1Cycle 4 Day 8Cycle 5 Day 1Cycle 5 Day 8Cycle 9 Day 1
ULO 1000mg + LDAC - Ph20.10017.76661.250103.322143.724153.87182.033212.79377.095224.703202.552187.930

Trough Observed Serum Concentration (Ctrough) - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration.~EOT = end of treatment~Measure type and method of dispersion are Geometric mean and %CV, respectively" (NCT02305563)
Timeframe: Days 1, 8, 15 for cycle 1; Days 8, 15 for cycle 2; Days 1, 8 for cycles 3-5; Day 1 every 4th cycle thereafter; EOT; 30 days post last dose (follow-up)

Interventionµg/mL (Geometric Mean)
Cycle 1 Day 1Cycle 1 Day 8Cycle 1 Day 15Cycle 2 Day 8Cycle 2 Day 15Cycle 3 Day 1Cycle 3 Day 8Cycle 4 Day 1Cycle 4 Day 8Cycle 5 Day 1Cycle 5 Day 8Cycle 9 Day 1EOT
ULO 800mg + LDAC - Ph20.10010.32137.26330.91648.42113.90537.6024.012116.6574.55834.78335.1440.100

Trough Observed Serum Concentration (Ctrough) - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration.~EOT = end of treatment~Measure type and method of dispersion are Geometric mean and %CV, respectively" (NCT02305563)
Timeframe: Days 1, 8, 15 for cycle 1; Days 8, 15 for cycle 2; Days 1, 8 for cycles 3-5; Day 1 every 4th cycle thereafter; EOT; 30 days post last dose (follow-up)

Interventionµg/mL (Geometric Mean)
Cycle 1 Day 1Cycle 1 Day 8Cycle 1 Day 15
LDAC - Ph20.10039.609175.315

Trough Observed Serum Concentration (Ctrough) - Phases 1 and 2

"The Pharmacokinetic (PK) parameters are assessed for ulocuplumab following study drug administration.~EOT = end of treatment~Measure type and method of dispersion are Geometric mean and %CV, respectively" (NCT02305563)
Timeframe: Days 1, 8, 15 for cycle 1; Days 8, 15 for cycle 2; Days 1, 8 for cycles 3-5; Day 1 every 4th cycle thereafter; EOT; 30 days post last dose (follow-up)

Interventionµg/mL (Geometric Mean)
Cycle 1 Day 1Cycle 1 Day 8Cycle 1 Day 15Cycle 2 Day 8Cycle 2 Day 15Cycle 3 Day 8Cycle 4 Day 8Cycle 5 Day 8
ULO 600mg + LDAC - Ph10.1005.30821.74841.63072.10485.993112.048102.751

5-year Overall Survival (OS)

Measured from date of registration to date of death due to any cause. Patients last known to be alive and are censored at date of last contact. (NCT01412879)
Timeframe: Up to 5 years

Interventionpercentage of participants (Number)
Arm 1: R-HCVAD/MTX/Ara-C (Induction Therapy)81
Arm 2: R-Bendamustine (Induction Therapy)79

Progression-Free Survival (PFS) at 2 Years

Disease progression is defined using the 2007 revised Cheson et al. criteria that is at least 50% increase in sum of the product of the diameters (SPD) of target measurable nodal lesions over the smallest sum observed, or >= 50% increase in greatest transverse diameter (GTD) of any nodal > 1 cm in shortest axis, or >= 50% increase in the SPD of other target measurable lesions over the smallest sum observed, any new bone marrow involvement, any new lesion, lymph node with long axis is > 1.5 cm or if both long and short axes are > 1 cm, PET positive if patients with no pretreatment PET scan or when PET scan was positive before therapy. Progression-free survival is measured from date of registration to date of first observation of progressive disease, or death due to any cause. Patients last known to be alive and progression-free are censored at date of last contact. (NCT01412879)
Timeframe: Up to 2 years

Interventionpercentage of participants (Number)
Arm 1: R-HCVAD/MTX/Ara-C (Induction Therapy)82
Arm 2: R-Bendamustine (Induction Therapy)81

Response Rate (Complete and Partial Response)

Complete Response (CR) is a complete disappearance of all disease with the exception of the following. If no PET scan or when the PET scan was positive before therapy, a post-treatment residual mass of any size is permitted if it is PET negative. If the PET scan was negative before therapy, all nodal masses at baseline must have regressed. No new lesions. Previously enlarged organs must have regressed and not be palpable. Bone marrow (BM) must be negative if positive at baseline. Normalization of markers. Partial Response (PR) is a 50% decrease in the sum of products of greatest diameters (SPD) for up to 6 identified dominant lesions, including spleenic and hepatic nodules from baseline. No new lesions and no increase in the size of liver, spleen or other nodes. If PET scan or when the PET scan was positive before therapy, PET should be positive in at least one previously involved site. (NCT01412879)
Timeframe: Up to 9 months

Interventionpercentage of participants (Number)
Arm 1: R-HCVAD/MTX/Ara-C (Induction Therapy)94.1
Arm 2: R-Bendamustine (Induction Therapy)82.9

Number of Patients With Grade 3 Through Grade 5 Adverse Events That Are Related to Study Drug

Adverse Events (AEs) are reported by the NCI Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. For each patient, worst grade of each event type is reported. Grade 3 = Severe, Grade 4 = Life-threatening, Grade 5 = Fatal. (NCT01412879)
Timeframe: Up to 8 months (Assessed at the beginning of each cycle of treatment, at restaging, and at post transplant.)

,,
InterventionParticipants (Number)
AcidosisAdult respiratory distress syndromeAlanine aminotransferase increasedAnemiaAnorexiaArthralgiaAspartate aminotransferase increasedBlood bilirubin increasedCD4 lymphocytes decreasedCatheter related infectionDehydrationDevice related infectionDiarrheaEnterocolitisEnterocolitis infectiousEpistaxisFatigueFebrile neutropeniaGallbladder infectionHyperglycemiaHypoalbuminemiaHypocalcemiaHypokalemiaHypomagnesemiaHyponatremiaHypophosphatemiaHypotensionHypoxiaInfections and infestations - Other, specifyInfusion related reactionLung infectionLymphocyte count decreasedMyalgiaNauseaNeutrophil count decreasedPain in extremityPlatelet count decreasedPruritusRash maculo-papularRectal hemorrhageSmall intestinal obstructionSore throatSyncopeTumor painUrinary tract infectionVascular access complicationVomitingWhite blood cell decreased
R-Bendamustine (Induction Therapy)000301002100001015001020010011027101216110010111014
R-HCVAD/MTX/Ara-C (Induction Therapy)00110001021101001050200500400000100111012010001000010
Stem Cell Transplant (Consolidation Therapy)11091002010141100131002111311001120216019011100000120

Complete Remission Plus CR With Incomplete Blood Count Recovery (CRi) Rate

"The percentage of participants who achieved a CR or CRi at any time point during the study per the modified IWG criteria for AML and investigator assessment.~CR: absolute neutrophil count (ANC) ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, red blood cell (RBC) transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRi: lack of morphologic evidence of leukemia (blasts < 5%), and platelet counts < 10^5 /µL or ANC < 10^3 /µL~Participants who never achieved CR or CRi or had no IWG disease assessment were considered to be non-responders in the calculation of CR + CRi rate." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionpercentage of participants (Number)
Phase 1+2: 600 mg Venetoclax + LDAC53.7

Complete Remission Rate

"Complete remission (CR) rate is defined as the percentage of participants who achieved a complete remission at any time point during the study per the modified IWG criteria for AML and investigator assessment.~CR: absolute neutrophil count (ANC) ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, red blood cell (RBC) transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~Participants who never achieved CR or had no IWG disease assessment were considered to be non-responders in the calculation of CR rate." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionpercentage of participants (Number)
Phase 1+2: 600 mg Venetoclax + LDAC25.6

Complete Remission With Partial Hematologic Recovery (CRh) Rate

"Complete remission with partial hematologic recovery is a derived response based on bone marrow blast and hematology laboratory values. CRh rate is defined as the percentage of participants who achieved CRh as the best response at any time point during the study.~A participant achieved a CRh when meeting the following criteria:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count of > 0.5 × 10^3 /µL and~Peripheral blood platelet count of > 0.5 × 10^5 /µL and~A 1 week (≥ 7 days) platelet transfusion-free period prior to the hematology lab collection.~Participants who never achieved CRh or did not have disease assessment or hematology data were considered to be non-responders in the calculation of CRh rate." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionpercentage of participants (Number)
Phase 1+2: 600 mg Venetoclax + LDAC20.7

CR Plus CRh Rate by Initiation of Cycle 2

"CR + CRh rate by initiation of Cycle 2 is defined as the percentage of participants who achieved CR or CRh by initiation of Cycle 2 of study treatment.~CR: ANC ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, RBC transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRh is a derived response based on bone marrow blast and hematology lab values. A participant achieved a CRh when meeting the following criteria:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count of > 0.5 × 10^3 /µL and~Peripheral blood platelet count of > 0.5 × 10^5 /µL and~A 1 week (≥ 7 days) platelet transfusion-free period prior to the hematology lab collection.~Participants who never achieved CR/CRh or did not have disease assessment by initiation of Cycle 2 were considered to be non-responders in the calculation of CR + CRh rate by initiation of Cycle 2." (NCT02287233)
Timeframe: Cycle 2, Day 1

Interventionpercentage of participants (Number)
Phase 1+2: 600 mg Venetoclax + LDAC30.5

CR Plus CRh Rate

"CR + CRh rate is defined as the percentage of participants who achieved CR or CRh at any time point during the study.~CR: ANC ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, RBC transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRh is a derived response based on bone marrow blast and hematology lab values. A participant achieved a CRh when meeting the following criteria:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count of > 0.5 × 10^3 /µL and~Peripheral blood platelet count of > 0.5 × 10^5 /µL and~A 1 week (≥ 7 days) platelet transfusion-free period prior to the hematology lab collection.~Participants who never achieved CR/CRh or did not have disease assessment or hematology data were considered to be non-responders in the calculation of CR + CRh rate." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionpercentage of participants (Number)
Phase 1+2: 600 mg Venetoclax + LDAC46.3

CR Plus CRi Rate by Initiation of Cycle 2

"The percentage of participants who achieved a CR or CRi by initiation of Cycle 2 of study treatment per the modified IWG criteria for AML and investigator assessment.~CR: ANC ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, RBC transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRi: Lack of morphologic evidence of leukemia (blasts < 5%), and platelet counts < 10^5 /µL or ANC < 10^3 /µL.~Participants who never achieved CR or CRi or had no IWG disease assessment by initiation of Cycle 2 were considered to be non-responders in the calculation of CR + CRi rate by initiation of Cycle 2." (NCT02287233)
Timeframe: Cycle 2, Day 1

Interventionpercentage of participants (Number)
Phase 1+2: 600 mg Venetoclax + LDAC28.0

Duration of Complete Response

Duration of CR is defined as the time from date that a participant achieved CR to the first date of relapse, clinical disease progression or death due to disease progression, whichever occurred earliest. Duration of CR was estimated using Kaplan-Meier methodology. If a participant was still responding at the data cutoff date, then the participant's data were censored at their last disease assessment date. Disease assessment data after the onset of any post-treatment therapy were not included in the duration of CR analysis. (NCT02287233)
Timeframe: Median duration of follow-up was 44.5 months (range: 0.3 to 63.7)

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC14.8

Duration of CR Plus CRh

Duration of CR + CRh is defined as the time from date that a participant achieved CR or CRh to the first date of relapse, clinical disease progression or death due to disease progression, whichever occurred earliest. Duration of CR + CRh was estimated using Kaplan-Meier methodology. If a participant was still responding at the data cutoff date, then the participant's data were censored at their last disease assessment date. Disease assessment data after the onset of any post-treatment therapy were not included in the analysis. (NCT02287233)
Timeframe: Median duration of follow-up was 44.5 months (range: 0.3 to 63.7)

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC11.0

Duration of CR Plus CRi

Duration of CR + CRi is defined as the time from the date that a participant achieved CR or CRi to the first date of relapse, clinical disease progression or death due to disease progression, whichever occurred earliest. Duration of CR + CRi was estimated using Kaplan-Meier methodology. If a participant was still responding at the data cutoff date, then the participant's data were censored at their last disease assessment date. Disease assessment data after the onset of any post-treatment therapy were not included in the analysis. (NCT02287233)
Timeframe: Median duration of follow-up was 44.5 months (range: 0.3 to 63.7)

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC9.8

Duration of CRi

Duration of CRi is defined as the time from date that a participant achieved CRi to the first date of relapse, clinical disease progression or death due to disease progression, whichever occurred earliest. Duration of CRi was estimated using Kaplan-Meier methodology. If a participant was still responding at the data cutoff date, then the participant's data were censored at their last disease assessment date. Disease assessment data after the onset of any post-treatment therapy were not included in the analysis. (NCT02287233)
Timeframe: Median duration of follow-up was 44.5 months (range: 0.3 to 63.7)

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC4.7

Overall Response Rate

"Overall response rate (ORR) is defined as the percentage of participants who achieved a complete remission (CR), complete remission with incomplete marrow recovery (CRi), or partial remission (PR) per the International Working Group (IWG) for AML response criteria, per investigator assessment.~CR: absolute neutrophil count (ANC) ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, red blood cell (RBC) transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRi: lack of morphologic evidence of leukemia (blasts < 5%), and platelet counts < 10^5 /µL or ANC < 10^3 /µL.~PR: all of the hematologic values for a CR but with a decrease of at least 50% in the percentage of blasts to 5% to 25% in the bone marrow aspirate." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionpercentage of participants (Number)
Phase 1+2: 600 mg Venetoclax + LDAC54.9

Overall Survival (OS)

Overall survival is defined as the time from the date of first dose to the date of death. All events of death were included, regardless of whether the event occurred while the participant was still taking study drug, or after the participant discontinued study drug. OS was estimated using Kaplan-Meier methodology. Participants who were still alive were censored at the analysis date. (NCT02287233)
Timeframe: Median duration of follow-up was 44.5 months (range: 0.3 to 63.7)

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC9.7

Phase 1: Number of Participants With Dose-limiting Toxicities

Dose-limiting toxicities (DLTs) were determined during cycle 1 of the dose-escalation phase and defined as Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0 grade 4 (life threatening requiring urgent intervention) or 5 (resulted in death) toxicity, excluding adverse events commonly caused by AML (eg, neutropenia, fever). Hematologic DLT was defined as failure of platelet recovery to 25 × 10^9/L or greater and absolute neutrophil count (ANC) to 0.5 × 10^9/L or greater within 14 days of the last dose of venetoclax in the absence of residual AML. (NCT02287233)
Timeframe: Up to 28 days (Cycle 1)

InterventionParticipants (Count of Participants)
Phase 1: 600 mg Venetoclax + LDAC0
Phase 1: 800 mg Venetoclax + LDAC1

Time to Best Response of CR + CRi

"The time to the best response of CR + CRi is defined as the time from the first date of study drug to the best response of CR or CRi per the IWG AML response criteria assessed by the investigator.~CR: absolute neutrophil count (ANC) ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, RBC transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRi: lack of morphologic evidence of leukemia (blasts < 5%), and platelet counts < 10^5 /µL or ANC < 10^3 /µL." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC2.8

Time to Best Response of CR Plus CRh

"The time to the best response of CR + CRh is defined as the time from the first date of study drug to the best response of CR or CRh.~CR: ANC ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, RBC transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRh is a derived response based on bone marrow blast and hematology lab values. A participant achieved a CRh when meeting the following criteria:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count of > 0.5 × 10^3 /µL and~Peripheral blood platelet count of > 0.5 × 10^5 /µL and~A 1 week (≥ 7 days) platelet transfusion-free period prior to the hematology lab collection." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC2.6

Time to First Response of CR + CRi

"The time to the first response of CR + CRi is defined as the time from the first date of study drug to the first response of CR or CRi per the IWG AML response criteria assessed by the investigator.~CR: absolute neutrophil count (ANC) ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, RBC transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRi: lack of morphologic evidence of leukemia (blasts < 5%), and platelet counts < 10^5 /µL or ANC < 10^3 /µL." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC1.4

Time to First Response of CR Plus CRh

"The time to the first response of CR + CRh is defined as the time from the first date of study drug to the first response of CR or CRh.~CR: ANC ≥ 10^3 /µL, platelet counts ≥ 10^5 /µL, RBC transfusion independence (a period of at least 56 days with no RBC transfusion), and bone marrow with < 5% blasts.~CRh is a derived response based on bone marrow blast and hematology lab values. A participant achieved a CRh when meeting the following criteria:~Bone marrow with < 5% blasts and~Peripheral blood neutrophil count of > 0.5 × 10^3 /µL and~Peripheral blood platelet count of > 0.5 × 10^5 /µL and~A 1 week (≥ 7 days) platelet transfusion-free period prior to the hematology lab collection." (NCT02287233)
Timeframe: Response was assessed at Cycle 2, Day 1, Cycle 4, Day 1, and every 3 cycles thereafter; median duration of treatment was 4.2 months.

Interventionmonths (Median)
Phase 1+2: 600 mg Venetoclax + LDAC1.0

Duration of Post Baseline Transfusion Independence

"The duration of transfusion independence is defined as the first time period that a participant received no RBC/platelet transfusions for at least 56 days during the evaluation period.~Post-baseline transfusion independence is defined as a period of at least 56 days with no RBC or platelet transfusion during the evaluation period. The evaluation period is from the first dose of study drug to the last dose of study drug until the 30 day follow-up visit, disease progression (including clinical progression), or death, whichever was earlier." (NCT02287233)
Timeframe: From the first dose of study drug to the last dose of study drug plus 30 days, disease progression (including clinical progression), or death, whichever was earlier; median duration of treatment was 4.2 months.

Interventiondays (Median)
Duration of RBC and platelet transfusion independenceDuration of RBC transfusion independenceDuration of platelet transfusion independence
Phase 1+2: 600 mg Venetoclax + LDAC150123155.5

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

"An adverse event (AE) is defined as any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. The investigator rated the severity of each AE according to the CTCAE Version 4.0 and the following:~Grade 1: The AE is transient and easily tolerated (mild).~Grade 2: The AE causes discomfort and interrupts usual activities (moderate).~Grade 3: The AE causes considerable interference with usual activities and may be incapacitating (moderate to severe).~Grade 4: The AE is life threatening requiring urgent intervention.~Grade 5: The AE resulted in death.~The investigator assessed each event as either having a reasonable possibility or no reasonable possibility of being related to the use of study drug.~Treatment-emergent events are defined as events that began or worsened in severity after the first dose of study drug." (NCT02287233)
Timeframe: From first dose of study drug until 30 days after last dose of study drug; median (minimum, maximum) duration of treatment was 4.1 (0.2, 62.8) months overall.

,,
InterventionParticipants (Count of Participants)
Any treatment-emergent adverse event (TEAE)TEAE with CTCAE Grade 3 or 4TEAE with CTCAE Grade 3 or aboveVenetoclax-related TEAELDAC-related TEAETEAE leading to hospitalizationTEAE leading to venetoclax discontinuationTEAE leading to LDAC discontinuationTEAE leading to venetoclax interruptionTEAE leading to LDAC interruptionTEAE leading to venetoclax reductionTEAE leading to LDAC reductionTEAE leading to death
Phase 1: 600 mg Venetoclax + LDAC8888873333001
Phase 1: 800 mg Venetoclax + LDAC1010109985543104
Phase 2: 600 mg Venetoclax + LDAC747272667164242645386115

Phase 1: Area Under the Plasma Concentration-Time Curve Over Time From 0 to 24 Hours (AUC0-24) of Venetoclax

(NCT02287233)
Timeframe: Cycle 1, Day 10 at predose and 2, 4, 6, 8, and 24 hours postdose (venetoclax with LDAC); Cycle 1, Day 18 at predose, 2, 4, 6, 8, and 24 hours postdose (venetoclax alone)

,
Interventionµg*h/mL (Mean)
Cycle 1, Day 10 (Venetoclax with LDAC)Cycle 1, Day 18 (Venetoclax Alone)
Phase 1: 600 mg Venetoclax + LDAC33.351.8
Phase 1: 800 mg Venetoclax + LDAC33.435.4

Phase 1: Area Under the Plasma Concentration-Time Curve Over Time From 0 to Last Measurable Concentration (AUCt) of Cytarabine

(NCT02287233)
Timeframe: Cycle 1, Day 1 and Day 10 at pre-dose and at 15 and 30 minutes and 1, 3, 6 hours post-dose.

,
Interventionng*h/mL (Mean)
Cycle 1, Day 1 (LDAC Alone)Cycle 1, Day 10 (LDAC with Venetoclax)
Phase 1: 600 mg Venetoclax + LDAC194231
Phase 1: 800 mg Venetoclax + LDAC204202

Phase 1: Maximum Observed Plasma Concentration (Cmax) of Cytarabine

The highest concentration that a drug achieves in the blood after administration in a dosing interval. (NCT02287233)
Timeframe: Cycle 1, Day 1 and Day 10 at pre-dose and at 15 and 30 minutes and 1, 3, 6 hours post-dose.

,
Interventionng/mL (Mean)
Cycle 1, Day 1 (LDAC Alone)Cycle 1, Day 10 (LDAC with Venetoclax)
Phase 1: 600 mg Venetoclax + LDAC175166
Phase 1: 800 mg Venetoclax + LDAC174175

Phase 1: Maximum Observed Plasma Concentration (Cmax) of Venetoclax

The highest concentration that a drug achieves in the blood after administration in a dosing interval. (NCT02287233)
Timeframe: Cycle 1, Day 10 at predose and 2, 4, 6, 8, and 24 hours postdose (venetoclax with LDAC); Cycle 1, Day 18 at predose, 2, 4, 6, 8, and 24 hours postdose (venetoclax alone)

,
Interventionµg/mL (Mean)
Cycle 1, Day 10 (Venetoclax with LDAC)Cycle 1, Day 18 (Venetoclax alone)
Phase 1: 600 mg Venetoclax + LDAC2.042.92
Phase 1: 800 mg Venetoclax + LDAC2.262.36

Phase 1: Time to Maximum Observed Plasma Concentration (Tmax) of Cytarabine

The time at which the maximum plasma concentration (Cmax) is observed. (NCT02287233)
Timeframe: Cycle 1, Day 1 and Day 10 at pre-dose and at 15 and 30 minutes and 1, 3, 6 hours post-dose.

,
Interventionhours (Median)
Cycle 1, Day 1 (LDAC Alone)Cycle 1, Day 10 (LDAC with Venetoclax)
Phase 1: 600 mg Venetoclax + LDAC0.30.3
Phase 1: 800 mg Venetoclax + LDAC0.30.3

Phase 1: Time to Maximum Observed Plasma Concentration (Tmax) of Venetoclax

The time at which the maximum plasma concentration (Cmax) is observed. (NCT02287233)
Timeframe: Cycle 1, Day 10 at predose and 2, 4, 6, 8, and 24 hours postdose (venetoclax with LDAC); Cycle 1, Day 18 at predose, 2, 4, 6, 8, and 24 hours postdose (venetoclax alone)

,
Interventionhours (Median)
Cycle 1, Day 10 (Venetoclax with LDAC)Cycle 1, Day 18 (Venetoclax Alone)
Phase 1: 600 mg Venetoclax + LDAC4.07.0
Phase 1: 800 mg Venetoclax + LDAC8.06.6

Post Baseline Transfusion Independence Rate Among Participants Transfusion-dependent at Baseline

"Post baseline transfusion independence rate was estimated as the percentage of participants who achieved transfusion independence during the evaluation period. Post-baseline transfusion independence is defined as a period of at least 56 days (≥ 56 days) with no RBC or platelet transfusion during the evaluation period. The evaluation period is from the first dose of study drug to the last dose of study drug until the 30 day follow-up visit, disease progression (including clinical progression), or death, whichever was earlier.~Results are reported for participants who achieved both RBC and platelet transfusion independence and for participants who received RBC transfusion independence and platelet transfusion independence." (NCT02287233)
Timeframe: From the first dose of study drug to the last dose of study drug plus 30 days, disease progression (including clinical progression), or death, whichever was earlier; median duration of treatment was 4.2 months.

Interventionpercentage of participants (Number)
RBC and platelet transfusion independenceRBC transfusion independencePlatelet transfusion independence
Phase 1+2: 600 mg Venetoclax + LDAC45.045.360.9

Post Baseline Transfusion Independence Rate

"Post baseline transfusion independence rate was estimated as the percentage of participants who achieved transfusion independence during the evaluation period. Post-baseline transfusion independence is defined as a period of at least 56 days (≥ 56 days) with no RBC or platelet transfusion during the evaluation period. The evaluation period is from the first dose of study drug to the last dose of study drug until the 30 day follow-up visit, disease progression (including clinical progression), or death, whichever was earlier.~Results are reported for participants who achieved both RBC and platelet transfusion independence and for participants who received RBC transfusion independence and platelet transfusion independence." (NCT02287233)
Timeframe: From the first dose of study drug to the last dose of study drug plus 30 days, disease progression (including clinical progression), or death, whichever was earlier; median duration of treatment was 4.2 months.

Interventionpercentage of participants (Number)
RBC and platelet transfusion independenceRBC transfusion independencePlatelet transfusion independence
Phase 1+2: 600 mg Venetoclax + LDAC45.147.658.5

Median Overall Survival (OS)

Overall Survival is defined as the time from randomization until death from any cause. (NCT00955916)
Timeframe: Up to 3 years

Interventionmonths (Median)
CLAG Regimen With Gleevec®4.9

Median Progression Free Survival (PFS)

Progression Free Survival is defined as the duration of time from start of treatment to time of progression. Leukemia related failure (progressive disease): Failure to induce bone marrow hypoplasia after 2 cycles or regrowth of leukemic blasts ≥ 20%. (NCT00955916)
Timeframe: Up to 3 years

Interventionmonths (Median)
CLAG Regimen With Gleevec®11.1

Overall Response Rate (ORR)

Overall Response Rate: Morphologic Complete Remission (CR) + Morphologic Complete Remission with incomplete blood count recovery (CRi) for evaluable participants. CR - Bone Marrow: < 5% blasts without Auer rods with at least 20% cellularity with maturation of all cell lines, No presence of unique phenotype by flow cytometry identical to what was found in the pretreatment specimen, No persistent dysplasia; Peripheral: normal blood counts, absolute neutrophil count (ANC) > 1.0 k/μl and platelets > 100 k/μl ANC > 1.0 k/μl and platelets > 100 k/μl (Peripheral blood counts documenting recovery can be utilized within 4 weeks of the bone marrow); No evidence of extramedullary leukemia. CRi - All CR criteria are met except for residual Neutropenia <1.0 x 10^9/L platelets < 100 k/μl. (NCT00955916)
Timeframe: 8 weeks per participant

Interventionpercentage of participants (Number)
CLAG Regimen With Gleevec®37

1 Year Overall Survival Rate

The percentage of participants alive at one year (NCT02560025)
Timeframe: 1 Year

Interventionpercentage of participants (Number)
Alisertib / MLN823750

Median Duration of Remission

"The median amount of time from first achieving remission to disease progression (with patients censored at death).~Complete remission (CR): Bone marrow showing less than 5% myeloblasts with normal maturation of all cell lines, an ANC of at least 1000/μL and a platelet count of 100,000/μL, absence of blast in peripheral blood, absence of identifiable leukemic cells in the bone marrow, existing extramedullary disease. If possible, at least one bone marrow biopsy should be performed to confirm CR.~Complete remission with incomplete blood count recovery (CRi): Same as CR but without achievement of specified ANC and/or platelet count~Recurrence/ morphologic relapse: reappearance of leukemic blasts in the peripheral blood or >5% blasts in the bone marrow not attributable to any other cause" (NCT02560025)
Timeframe: From the time of first remission to disease progression or death, median duration of 12.8 months

InterventionMonths (Median)
Alisertib / MLN823712.8

Median Relapse Free Survival

"The median amount of time from achieving a complete remission to the first of disease recurrence or death. RFS applies only to the subset of patients who achieve a CR+CRi at the end of induction therapy.~Complete remission (CR): Bone marrow showing less than 5% myeloblasts with normal maturation of all cell lines, an ANC of at least 1000/μL and a platelet count of 100,000/μL, absence of blast in peripheral blood, absence of identifiable leukemic cells in the bone marrow, existing extramedullary disease. If possible, at least one bone marrow biopsy should be performed to confirm CR.~Complete remission with incomplete blood count recovery (CRi): Same as CR but without achievement of specified ANC and/or platelet count~Recurrence/ morphologic relapse: reappearance of leukemic blasts in the peripheral blood or >5% blasts in the bone marrow not attributable to any other cause" (NCT02560025)
Timeframe: From the time of treatment response until death or disease progression (up to about one year)

InterventionMonths (Median)
Alisertib / MLN8237NA

Number of Participants That Achieved Complete Remission With Incomplete Blood Count Recovery (CRi)

"The number of participants that achieved a best overall response of CRi while on study.~Complete Remission with Incomplete Blood Count Recovery (CRi): Same as for CR but without achievement of ANC at least 1000/uL (CRi) and/or platelet count of 100,000/uL (CRp)." (NCT02560025)
Timeframe: From the start of treatment until the end of study treatment, up to approximately 10 months

InterventionParticipants (Count of Participants)
Alisertib / MLN82375

Number of Participants That Achieved Complete Remission

"The number of participants that achieved a best overall response of complete remission while on study.~Complete remission (CR): Bone marrow showing less than 5% myeloblasts with normal maturation of all cell lines, an ANC of at least 1000/μL and a platelet count of 100,000/μL, absence of blast in peripheral blood, absence of identifiable leukemic cells in the bone marrow, existing extramedullary disease. If possible, at least one bone marrow biopsy should be performed to confirm CR." (NCT02560025)
Timeframe: From the start of treatment until the end of study treatment, up to approximately 10 months

InterventionParticipants (Count of Participants)
Alisertib / MLN823720

Number of Participants With Serious Adverse Events

Adverse events were assessed using Common Terminology Criteria for Adverse Events (CTCAE 4). Adverse events were considered to be Serious Adverse Events (SAE) if they were grade 3 or greater and deemed to be possibly, probably, or definitely related to the study treatment. (NCT02560025)
Timeframe: From the start of treatment until 30 days after the last dose of a study drug is received, up to approximately 11 months

InterventionParticipants (Count of Participants)
Alisertib / MLN823727

Percentage of Treatment-related Mortality (TRM)

"Number of treatment related deaths divided by total number of patients during induction or reinduction therapy.~Presented as percentage" (NCT02553460)
Timeframe: At the end of reinduction (up to 5 months after start of therapy)

InterventionParticipants (Count of Participants)
Stratum 11

Phase 1 and 2: Event-Free Survival

Event free survival is defined as the time from first dose of study drug to relapse from CR, death, or second malignancy for participants who achieved CR. CR is defined as morphologic leukemia-free state, with less than 5 percentage (%) blasts in aspirate sample with marrow spicules and with a count of greater than or equal to (>=) 200 nucleated cells, plus absolute neutrophil count (ANC) greater than (>) 1,000/ microliter platelet count of >100,000/microliter and participant must be independent of transfusions for a minimum of 1 week before each marrow assessment. (NCT01853228)
Timeframe: From enrollment to progression/relapse, death, or withdrawal, whichever comes first, for up to approximately 3 years 10 months

Interventiondays (Median)
Decitabine (Dacogen) + Cytarabine1

Phase 1 and 2: Number of Participants Reporting Treatment-Emergent Adverse Events (TEAEs)

TEAEs are defined as adverse events with onset or worsening on or after date of first dose of study treatment. An adverse event is any untoward medical event that occurs in a participant administered an investigational product, and it does not necessarily indicate only events with clear causal relationship with the relevant investigational product. (NCT01853228)
Timeframe: Approximately 3 years 10 months

InterventionParticipants (Count of Participants)
Decitabine (Dacogen) + Cytarabine17

Phase 1 and 2: Overall Survival (OS)

OS is defined as the time from the date of first dose of study drug to date of death from any cause. If the participant is alive or the vital status is unknown, the participant will be censored at the date the participant will be last known to be alive. (NCT01853228)
Timeframe: From enrollment to death or withdrawal, whichever comes first, for up to approximately 3 years 10 months

Interventionmonths (Median)
Decitabine (Dacogen) + Cytarabine5.1

Phase 1: Maximum Tolerated Dose (MTD) of Cytarabine

The maximum tolerated dose (MTD) for cytarabine was based on the number of participants experiencing a dose-limiting toxicity (DLT) by the end of Cycle 1. A non-hematological DLT was defined as: any Grade >=3 toxicity that persists for greater than (>) 5 days or any Grade 2 toxicity that persists for >7 days and that is intolerable to the participant. A hematological DLT was defined as Grade 4 neutropenia or thrombocytopenia due to a hypoplastic bone marrow at Day 42, in the absence of malignant infiltration. The nominal duration of each cycle was 28 days. However, participants who had not experienced bone marrow recovery at Day 28 were followed up to Day 42. Failure of marrow recovery (improvement to Grade 3) by Day 42 was considered a DLT. The maximum duration of Cycle 1 was therefore 42 days. (NCT01853228)
Timeframe: Cycle 1 (42 days)

Interventiongram per square meter (Number)
Decitabine (Dacogen) + Cytarabine2

Phase 2: Overall Response Rate

Overall response rate is defined as percentage of participants with complete remission (CR) +complete remission with incomplete blood count recovery (CRi)+partial remission (PR) per IWG Criteria. CRi: morphologic CR with residual neutropenia (less than [<] 1,000/microliter) or thrombocytopenia <100,000/microliter). CR is defined as morphologic leukemia-free state, with less than 5 percentage (%) blasts in aspirate sample with marrow spicules and with a count of greater than or equal to (>=) 200 nucleated cells, plus absolute neutrophil count (ANC) greater than (>) 1,000/ microliter platelet count of >100,000/microliter and participant must be independent of transfusions for a minimum of 1 week before each marrow assessment. PR: all the same hematologic values of a CR, but with a decrease of >=50% of the percentage of blasts to 5% to 25% in the bone marrow aspirate. (NCT01853228)
Timeframe: Up to approximately 3 years 10 months

InterventionPercentage of participants (Number)
Decitabine (Dacogen) + Cytarabine37.5

Phase 2: Percentage of Participants Who Achieved CR or CRi at Cycle 1 Day 28

Response rate (percentage of participants who achieved CR or CRi) was measured using international working group (IWG) criteria. Response rate is defined as complete remission (CR) or complete remission with incomplete blood count recovery (CRi) in children with relapsed or refractory acute myeloid leukemia. CRi is defined as morphologic CR with residual neutropenia (less than [<] 1,000/microliter) or thrombocytopenia <100,000/ microliter). CR is defined as morphologic leukemia-free state, with less than 5 percentage (%) blasts in aspirate sample with marrow spicules and with a count of greater than or equal to (>=) 200 nucleated cells, plus absolute neutrophil count (ANC) greater than (>) 1,000/ microliter, platelet count of >100,000/microliter and participant must be independent of transfusions for a minimum of 1 week before each marrow assessment. (NCT01853228)
Timeframe: Cycle 1 (28 days) Day 28

Interventionpercentage of participants (Number)
Decitabine (Dacogen) + Cytarabine20

Phase 2: Percentage of Participants Who Achieved CR or CRi at Cycle 2 Day 28

Response rate (percentage of participants who achieved CR or CRi) was measured using international working group (IWG) criteria. Response rate is defined as complete remission (CR) or complete remission with incomplete blood count recovery (CRi) in children with relapsed or refractory acute myeloid leukemia. CRi is defined as morphologic CR with residual neutropenia (less than [<] 1,000/microliter) or thrombocytopenia <100,000/ microliter). CR is defined as morphologic leukemia-free state, with less than 5 percentage (%) blasts in aspirate sample with marrow spicules and with a count of greater than or equal to (>=) 200 nucleated cells, plus absolute neutrophil count (ANC) greater than (>) 1,000/ microliter, platelet count of >100,000/microliter and participant must be independent of transfusions for a minimum of 1 week before each marrow assessment. (NCT01853228)
Timeframe: Cycle 2 (28 days) Day 28

Interventionpercentage of participants (Number)
Decitabine (Dacogen) + Cytarabine66.7

Phase 2: Percentage of Participants Who Achieved CR or CRi at End of Study Treatment

Response rate (percentage of participants who achieved CR or CRi) was measured using international working group (IWG) criteria. Response rate is defined as complete remission (CR) or complete remission with incomplete blood count recovery (CRi) in children with relapsed or refractory acute myeloid leukemia. CRi is defined as morphologic CR with residual neutropenia (less than [<] 1,000/microliter) or thrombocytopenia <100,000/ microliter). CR is defined as morphologic leukemia-free state, with less than 5 percentage (%) blasts in aspirate sample with marrow spicules and with a count of greater than or equal to (>=) 200 nucleated cells, plus absolute neutrophil count (ANC) greater than (>) 1,000/ microliter, platelet count of >100,000/microliter and participant must be independent of transfusions for a minimum of 1 week before each marrow assessment. (NCT01853228)
Timeframe: End of study treatment (approximately 3 years)

Interventionpercentage of participants (Number)
Decitabine (Dacogen) + Cytarabine12.5

Phase 1 and 2: Area Under the Plasma Concentration-Time Curve (AUC) of Decitabine

AUC is the area under the plasma concentration-time curve of decitabine. (NCT01853228)
Timeframe: Cycle 1 (28 days) Day 5: pre-infusion, 0.5 hour during infusion, end of infusion and at 5 min, 0.5 hour, 1 hour, and 2 hour after end of infusion

Interventionnanogram*hour per milliliter (ng*h/mL) (Median)
> 1 month to <= 2 years> 2 to <= 6 years> 6 to <= 12 years> 12 to <= 16 years
Decitabine (Dacogen) + Cytarabine124.8131.5158.9162.4

Phase 1 and 2: Maximum Plasma Concentration (Cmax) of Decitabine

Cmax is the maximum observed plasma concentration of Decitabine. (NCT01853228)
Timeframe: Cycle 1 (28 days) Day 5: pre-infusion, 0.5 hour during infusion, end of infusion and at 5 min, 0.5 hour, 1 hour, and 2 hour after end of infusion

Interventionnanogram per milliliter (ng/mL) (Median)
> 1 month to <= 2 years> 2 to <= 6 years> 6 to <= 12 years> 12 to <= 16 years
Decitabine (Dacogen) + Cytarabine118.4123.1148.7151.4

Phase 1 and 2: Total Clearance of Decitabine

Total clearance of drug after intravenously administration was calculated as: dose/area under the plasma concentration-time curve. (NCT01853228)
Timeframe: Cycle 1 (28 days) Day 5: pre-infusion, 0.5 hour during infusion, end of infusion and at 5 minute (min), 0.5 hour, 1 hour, and 2 hour after end of infusion

Interventionliter per hour per square meter (Median)
>1 month to less than or equal to (<=) 2 yearsGreater than (>) 2 to <= 6 years> 6 to <= 12 years> 12 to <= 16 years
Decitabine (Dacogen) + Cytarabine160.6152.1125.9123.2

Phase 1 and 2: Volume of Distribution at Steady-State (Vss) of Decitabine

Volume of distribution is defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired blood concentration of a drug. (NCT01853228)
Timeframe: Cycle 1 (28 days) Day 5: pre-infusion, 0.5 hour during infusion, end of infusion and at 5 min, 0.5 hour, 1 hour, and 2 hour after end of infusion

Interventionliter per square meter (Median)
> 1 month to <= 2 years> 2 to <= 6 years> 6 to <= 12 years> 12 to <= 16 years
Decitabine (Dacogen) + Cytarabine35.936.531.835.9

Phase 2: Duration of Response

Duration of response is defined as weeks from date of first response to date of first relapse or date of death. (NCT01853228)
Timeframe: From time of response to relapse, study completion/withdrawal, or death, whichever comes first, for up to approximately 3 years 10 months

Interventionweeks (Number)
Participant 1Participant 2
Decitabine (Dacogen) + Cytarabine44.66.3

Estimated Dose to Tumor Sites Based on the Tumor to Normal Organ Ratios Derived From Dosimetry Estimates Coupled With the Absorbed Dose to Normal Organs Based on the Administered Activity of Yttrium Y 90 Anti-CD45 Monoclonal Antibody BC8

Will be evaluated among all patients and among those treated at the estimated MTD. (NCT01921387)
Timeframe: Up to 5 years

InterventionmCi (Number)
Treatment (90Y-BC8-DOTA, Chemotherapy, PBSC)52.8

Maximum-tolerated Dose (MTD) of Yttrium-90-BC8-DOTA

Single patients will be treated at escalating doses in 2-Gy increments (Table 4) until a DLT is observed. Once a DLT is observed, the second stage will begin at the next lower dose level and patients will be treated in cohorts of 4. (NCT01921387)
Timeframe: Within 30 days post-transplant

InterventionGy - MTD (Number)
Treatment (90Y-BC8-DOTA, Chemotherapy, PBSC)34

Progression-free Survival Following Autologous Stem Cell Transplant (ASCT)

Estimate the 1 year progression-free survival (PFS) rate after ASCT (NCT01921387)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Treatment (90Y-BC8-DOTA, Chemotherapy, PBSC)12

The Lowest Antibody (Yttrium 90-BC8-DOTA) Dose (mg/kg) That is Consistent With a Favorable Biodistribution Rate >= 80% in Lymphoma Patients

(NCT01921387)
Timeframe: Up to 5 years

Interventionmg/kg (Number)
Treatment (90Y-BC8-DOTA, Chemotherapy, PBSC)0.75

Number of Participants With Infusion Reactions (IRs)

An IR was an AE related to isatuximab typically with onset within 24 hours from the start of the isatuximab infusion and was reported by the investigator. (NCT03860844)
Timeframe: From the time of the first treatment administration (Day 1) up to 30 days after the last treatment (maximum duration of exposure of 13.1 weeks for B-ALL cohort, 10.7 weeks for T-ALL cohort and 7.1 weeks for AML cohort)

InterventionParticipants (Count of Participants)
B-cell Acute Lymphoblastic Leukemia (B-ALL)9
T-cell Acute Lymphoblastic Leukemia (T-ALL)5
Acute Myeloid Leukemia (AML)15

Overall Response Rate (ORR)

ORR:Percentage of participants with CR/CRi or partial response (PR) for blood and bone marrow disease based on NCCN guideline. CR: <5% blasts in BMA with spicules; no circulating blasts (ALL)/no blasts with Auer rods (AML) or extramedullary disease, no lymphadenopathy, splenomegaly, skin/gum infiltration/testicular mass/central nervous system involvement (ALL), trilineage hematopoiesis (ALL);ANC >=1000/mcL; platelets >100000/mcL; RBC transfusion independence. If the physician documented transfusion dependency related to study treatment;not to participant's underlying disease, CRi was reported. CRi met the same criteria as CR, except neutrophils and/or platelets recovery (ANC <1000/mcL or platelets <100000/mcL). PR: >50% decrease in the sum of the product of the greatest perpendicular diameters of the mediastinal enlargement. For participants with a previous positive positron emission tomography (PET) scan, a post-treatment PET was to be positive in at least 1 previously involved site. (NCT03860844)
Timeframe: From enrollment until the primary analysis completion date of 12 Sep 2022; the median duration of exposure was approximately 7 weeks

Interventionpercentage of participants (Number)
B-cell Acute Lymphoblastic Leukemia (B-ALL)52.0
T-cell Acute Lymphoblastic Leukemia (T-ALL)54.5
Acute Myeloid Leukemia (AML)65.2

Percentage of Participants With Complete Response (CR) Rate

The complete response rate (CR + CRi [complete response with incomplete peripheral recovery]) was defined as the percentage of participants achieving complete response (CR + CRi) assessed by the investigator per National Comprehensive Cancer Network (NCCN) guidelines version 1.2018 criteria. CR was defined as <5% blasts in a bone marrow aspirate (BMA) with spicules; no circulating blasts (ALL)/no blasts with Auer rods (AML) or extramedullary disease, no lymphadenopathy, splenomegaly, skin/gum infiltration/testicular mass/central nervous system involvement (ALL), trilineage hematopoiesis (ALL); Absolute neutrophil count (ANC) >=1000/microliter (mcL); platelets >100000/mcL; red blood cell transfusion independence. If the physician documented transfusion dependency related to study treatment and not to the participant's underlying disease, CRi was reported. CRi met the same criteria as for CR, except neutrophils and/or platelets recovery (ANC <1000/mcL or platelets <100000/mcL). (NCT03860844)
Timeframe: From enrollment until the primary analysis completion date of 12 Sep 2022; the median duration of exposure was approximately 7 weeks

Interventionpercentage of participants (Number)
B-cell Acute Lymphoblastic Leukemia (B-ALL)52.0
T-cell Acute Lymphoblastic Leukemia (T-ALL)45.5
Acute Myeloid Leukemia (AML)60.9

AML: AUC of Isatuximab

Plasma samples were collected at specified timepoints to determine the AUC of isatuximab. (NCT03860844)
Timeframe: From Week 0 to Week 1, Week 0 to Week 3, and Week 0 to Week 8

Interventionmg*h/L (Mean)
Week 0 to Week 1Week 0 to Week 3Week 0 to Week 8
Acute Myeloid Leukemia (AML)28592130862291962

AML: Ceoi of Isatuximab

Ceoi is the plasma concentration observed at the end of intravenous infusion of isatuximab. (NCT03860844)
Timeframe: At end of infusion on Cycle 1 Days 1 and 15

Interventionmcg/mL (Mean)
Cycle 1: Day 1Cycle 1: Day 15
Acute Myeloid Leukemia (AML)363562

AML: Plasma Concentration Reached by Isatuximab Before Next Dose Administration (Ctrough)

Plasma samples were collected at specified timepoints to determine the Ctrough of isatuximab. (NCT03860844)
Timeframe: Cycle 1 Day 8, Cycle 1 Day 15, Cycle 2 Day 1 and Cycle 2 Day 15

Interventionmcg/mL (Mean)
Cycle 1: Day 8Cycle 1: Day 15Cycle 2: Day 1Cycle 2: Day 15
Acute Myeloid Leukemia (AML)126217115420

B-ALL and T-ALL: Area Under the Concentration Time Curve (AUC) of Isatuximab

Plasma samples were collected at specified timepoints to determine the AUC of isatuximab. (NCT03860844)
Timeframe: From Week 0 to Week 1, Week 0 to Week 5, and Week 0 to Week 10

,
Interventionmg*hour (h)/Liter (L) (Mean)
Week 0 to Week 1Week 0 to Week 5Week 0 to Week 10
B-cell Acute Lymphoblastic Leukemia (B-ALL)31703299071582686
T-cell Acute Lymphoblastic Leukemia (T-ALL)29057289167540375

B-ALL and T-ALL: Concentrations at the End of Infusion (Ceoi) of Isatuximab

Ceoi is the plasma concentration observed at the end of intravenous infusion of isatuximab. (NCT03860844)
Timeframe: At end of infusion on Cycle 1 Days 1 and 29

,
Interventionmcg/mL (Mean)
Cycle 1: Day 1Cycle 1: Day 29
B-cell Acute Lymphoblastic Leukemia (B-ALL)452835
T-cell Acute Lymphoblastic Leukemia (T-ALL)259745

B-ALL and T-ALL: Plasma Concentration Reached by Isatuximab Before Next Dose Administration (Ctrough)

Plasma samples were collected at specified timepoints to determine the Ctrough of isatuximab. (NCT03860844)
Timeframe: Cycle 1 Day 8, Cycle 1 Day 15, Cycle 1 Day 22, Cycle 1 Day 29, Cycle 2 Day 43, Cycle 2 Day 57

,
Interventionmicrogram/milliliter (mcg/mL) (Mean)
Cycle 1: Day 8Cycle 1: Day 15Cycle 1: Day 22Cycle 1: Day 29Cycle 2: Day 43Cycle 2: Day 57
B-cell Acute Lymphoblastic Leukemia (B-ALL)114272388475504531
T-cell Acute Lymphoblastic Leukemia (T-ALL)127263323426357478

CD38 Receptor Occupancy

Blood samples were collected to assess CD38 receptor occupancy as a pharmacodynamics marker. It was assessed across complete responders and non-complete responders. Multicolor flow cytometry assay was validated for CD38 receptor occupancy (CD38RO) quantification, based on the use of two murine monoclonal antibodies (MAbs), one competing with SAR650984 to determine the number of free CD38 receptors (MAb1) and one recognizing a different binding epitope on CD38 to measure the total number of receptors (MAb2) at the cell surface of the cancer cells. Cells were tagged with either MAb1 (Tube #1) or MAb2 (Tube #2). The percentage RO was calculated using the following equation: % CD38RO = [(sABC MAb2 - sABC MAb1)/sABC MAb2] X 100. (NCT03860844)
Timeframe: Pre-dose on Day 15

Interventionpercent receptor occupancy (Mean)
Blood plasma cells: Non CR/CRi;Blood NK cells: CR/CRiBlood NK cells: Non CR/CRi
B-cell Acute Lymphoblastic Leukemia (B-ALL)44.055.661.3

CD38 Receptor Occupancy

Blood samples were collected to assess CD38 receptor occupancy as a pharmacodynamics marker. It was assessed across complete responders and non-complete responders. Multicolor flow cytometry assay was validated for CD38 receptor occupancy (CD38RO) quantification, based on the use of two murine monoclonal antibodies (MAbs), one competing with SAR650984 to determine the number of free CD38 receptors (MAb1) and one recognizing a different binding epitope on CD38 to measure the total number of receptors (MAb2) at the cell surface of the cancer cells. Cells were tagged with either MAb1 (Tube #1) or MAb2 (Tube #2). The percentage RO was calculated using the following equation: % CD38RO = [(sABC MAb2 - sABC MAb1)/sABC MAb2] X 100. (NCT03860844)
Timeframe: Pre-dose on Day 15

Interventionpercent receptor occupancy (Mean)
Blood plasma cells: CR/CRiBlood plasma cells: Non CR/CRi;Blood NK cells: CR/CRiBlood NK cells: Non CR/CRi
T-cell Acute Lymphoblastic Leukemia (T-ALL)40.555.066.770.0

Cluster of Differentiation (CD)38 Receptor Density

"Blood samples were collected to assess CD38 receptor density as a predictive biomarker. It was assessed across complete responders and non-complete responders. The Antibody Binding Capacity (ABC) was calculated using the following equation: ABC = 10^(Logarithm(Mean Fluorescence Intensity)*a+b) where a was the slope and b was the Y-intercept of the calibration curve equation. Specific and absolute quantitative values (specific antibody-binding capacity [sABC]) of binding of the selected antibodies were calculated after subtraction of the negative isotypic immunoglobulin G (IgG) control." (NCT03860844)
Timeframe: Pre-dose on Day 1

,,
InterventionsABC (Mean)
Blood blast cells: CR/CRiBlood blast cells: Non CR/CRi;Blood immune cells (Natural Killer [NK] cells): CR/CRiBlood immune cells (NK cells): Non CR/CRi
Acute Myeloid Leukemia (AML)19502.09815.011220.322530.0
B-cell Acute Lymphoblastic Leukemia (B-ALL)20345.631080.013506.216650.0
T-cell Acute Lymphoblastic Leukemia (T-ALL)12780.022952.022639.033859.0

Number of Participants With Treatment-Emergent Adverse Events (TEAEs) and Treatment-Emergent Serious Adverse Events (TESAEs)

An AE was defined as any untoward medical occurrence in a participant temporally associated with the use of study treatment, whether or not considered related to the study treatment. SAEs were any untoward medical occurrence that at any dose: resulted in death, was life-threatening, required inpatient hospitalization or prolongation of existing hospitalization, resulted in persistent or significant disability/incapacity, was a congenital anomaly/birth defect, was a medically important event. TEAEs were defined as an AE which occurred after the first dose of study treatment administration until the last dose plus 30 days, or until the start of hematological recovery period or a new anti-leukemia/lymphoma therapy, whichever occurred first. (NCT03860844)
Timeframe: From the time of the first treatment administration (Day 1) up to 30 days after the last treatment (maximum duration of exposure of 13.1 weeks for B-ALL cohort, 10.7 weeks for T-ALL cohort and 7.1 weeks for AML cohort)

,,
InterventionParticipants (Count of Participants)
Any TEAEAny TESAE
Acute Myeloid Leukemia (AML)2616
B-cell Acute Lymphoblastic Leukemia (B-ALL)2719
T-cell Acute Lymphoblastic Leukemia (T-ALL)1312

Minimal Residual Disease (MRD) Using Standardized Techniques

Percentage of participants with a CRMRD- response at the end of Cycle 1 (NCT03298984)
Timeframe: During duration of study

Interventionpercentage of participants (Number)
Alvocidib 20/30 mg/m266.7
Alvocidib 30/40 mg/m20
Alvocidib 30/50 mg/m266.7
Alvocidib 30/60 mg/m234.8

Number of Participants Who Experienced Dose Limiting Toxicities (DLTs) of Alvocidib

Determine the safety and tolerability including dose-limiting toxicities (DLTs) of alvocidib when administered over a range of doses on Days 1-3 followed by Ara-c/daunorubicin (7+3) on Days 5-11 in adults with newly diagnosed and previously untreated AML (NCT03298984)
Timeframe: During the first cycle

InterventionParticipants (Count of Participants)
Alvocidib 20/30 mg/m20
Alvocidib 30/40 mg/m20
Alvocidib 30/50 mg/m20
Alvocidib 30/60 mg/m21

Antileukemic Activity of Alvocidib Plus 7+3 - Response to Treatment Based on 2017 ELN Response Criteria

CR: Measurable residual disease is positive or unknown; BM blasts (bls) <5%; no circulating bls and bls w/ Auer rods; no extramedullary disease; ANC >1.0 x 109/L; platelets >100 x 109/L. CRMRD-: CR w/ negativity genetic marker. CRi: CR except residual neutropenia or thrombocytopenia. MLFS: BM bls <5%; no bls with Auer rods; no extramedullary disease; no hematologic recovery required. PR: all hematologic CR criteria; decrease (dec) BM bls % to 5-25%; dec pretreatment BM bls % by >50%. SD: no CRMRD-/CR/CRi/PR/MLFS; PD criteria not met. PD: increase (inc) BM bls % and/or inc absolute bls in blood: 50% inc BM bls over baseline (>15% point inc required in cases w/ <30% bls at baseline or persistent BM bls % of >70% over at least 3 months; without at least 100% improvement in ANC to absolute level [>0.5 x 109/L and/or platelet count to >50 x 109/L non-transfused); or >50% inc in peripheral bls to >25 x 109/L (in the absence of differentiation syndrome); or new extramedullary disease. (NCT03298984)
Timeframe: Best response during duration of study

,,,
Interventionparticipants (Number)
Complete Remission MRD Negative (CRMRD-)Complete Remission MRD Positive or Unknown (CR)Partial Remission (PR)Stable DiseaseProgressive Disease (PD)Not Evaluated
Alvocidib 20/30 mg/m2210000
Alvocidib 30/40 mg/m2021000
Alvocidib 30/50 mg/m2200010
Alvocidib 30/60 mg/m2871241

Maximum Tolerated Dose (MTD) of Alvocidib

Determine the safety and tolerability including the maximum tolerated dose (MTD) of alvocidib when administered over a range of doses on Days 1-3 followed by Ara-c/daunorubicin (7+3) on Days 5-11 in adults with newly diagnosed and previously untreated AML (NCT03298984)
Timeframe: During the first cycle

Interventionmg/m2 (Number)
BolusIV infusion
All Participants3060

Recommended Phase 2 Dose (RP2D) of Alvocidib in Combination With 7+3

The dose at which < 1 of 6 patients experience a DLT during Cycle 1 with the next higher dose having at least 2 of 3 to 6 patients experiencing a DLT during Cycle 1 (NCT03298984)
Timeframe: During Cycle 1 beginning at 1st dose of study drug through Day 50 + or - 3 days

Interventionmg/m2 (Number)
BolusIV infusion
All Participants3060

Change From Baseline in EQ-5D-5L Health Utility Index

The EQ-5D is a standardized instrument for use as a measure of health outcome. The descriptive system comprises 5 dimensions: Mobility, Self-care, Usual Activities, Pain/Discomfort and Anxiety/Depression. Each dimension has 5 levels: no problems, slight problems, moderate problems, severe problems, and extreme problems. Responses from the 5 dimensions are coded so that a '1' indicates no problem on that dimension, and '5' indicates the most serious problem. Results obtained just prior to the start of study treatment on Day 1 of Cycle 1 will serve as the baseline values. If not available, the most recent screening results prior to the start of study treatment on Day 1 of Cycle 1 will be considered the baseline values. The responses for the 5 dimensions can be combined in a 5-digit number describing the respondent's health state. Score was converted to a single index value using the cross-walk method to the EQ-5D-3L value set. (NCT02577406)
Timeframe: From baseline up to cycle 2 day 1 (up to approximately 1 month)

InterventionChange from baseline in EQ-5D score (Mean)
AG-221-0.0738
Conventional Care Regimens (CCRs)-0.0598

Complete Remission Rate

The number of participants with morphologic complete remission (CR) according to modified International Working Group Acute Myeloid Leukemia Response Criteria (IWG AML). CR is defined as less than 5% blasts in a BM aspirate sample with marrow spicules and with a count of at least 200 nucleated cells. There should be no blasts with Auer rods and absence of extramedullary disease; plus the following conditions: absolute neutrophil count (ANC) ≥1,000/μL, Platelet count ≥100,000/μL, and independent of red cell transfusions for ≥1 week before each response assessment. (NCT02577406)
Timeframe: From randomization up to approximately 49 months

InterventionParticipants (Count of Participants)
AG-22111
Conventional Care Regimens (CCRs)1

Duration of Response

Time from first documented MLFS/CR/CRi/CRp/PR to documented morphologic relapse after CR/CRi/CRp/ PD or death due to any cause, whichever occurs first. CR and MLFS are defined as <5% blasts in a BM aspirate sample with marrow spicules + a count of ≥200 nucleated cells with no blasts with Auer rods + no extramedullary disease. CR must also include: ANC ≥ 1,000/μL, Platelet count ≥100,000/μL, + independent of red cell transfusions for ≥1 week before assessment. CRi is all criteria of CR except ANC. CRp is all criteria of CR except platelet count. PR is defined as all hematologic criteria of CR with >50% decrease in BM blasts to 5%-25%. Relapse after CR/CRi/CRp is defined as reappearance of ≥ 5% blasts in the BM not attributable to other cause or development of extramedullary disease. PD is defined as > 50% increase of BM blast count from baseline to ≥ 20% or a doubling of absolute blast count in peripheral blood from baseline to ≥ 10,000/μL or development of new extramedullary disease. (NCT02577406)
Timeframe: From randomization to documented morphologic relapse after CR/CRi/CRp/ PD or death due to any cause, whichever occurs first (up to approximately 49 months)

InterventionMonths (Median)
AG-2213.9
Conventional Care Regimens (CCRs)4.5

Event-Free Survival

Time from randomization to documented morphologic relapse after complete remission/complete remission with incomplete neutrophil recovery/complete remission with incomplete platelet recovery (CR/CRi/CRp), progressive disease (PD) or death from any cause, whichever occurs first. Morphologic Relapse after CR/CRi/CRp is defined as either reappearance of ≥ 5% blasts in the BM not attributable to any other cause or the development of extramedullary disease. PD is defined as a > 50% increase of BM blast count percentage from baseline to ≥ 20% for participants with 5 to 70% BM blasts at baseline or a doubling of absolute blast count in peripheral blood from baseline to ≥ 10 x 109/L (10,000/μL) for participants with > 70% BM blasts at baseline or the development of new extramedullary disease. (NCT02577406)
Timeframe: From randomization to the date of documented morphologic relapse after CR/CRi/CRp, PD or death from any cause, whichever occurs first. (up to approximately 49 months)

InterventionMonths (Median)
AG-2213.2
Conventional Care Regimens (CCRs)2.5

Hematologic Improvement Rate

The number of participants with hematologic improvement neutrophil response (HI-N) + hematologic improvement platelet response (HI-P) + hematologic improvement erythroid response (HI-E) according to the International Working Group for Myelodysplastic Syndromes for Hematologic Improvement (IWG MDS HI) criteria. HI-E is defined as a hemoglobin increase by ≥ 1.5 g/dL and a relevant reduction in units of RBC transfusions by an absolute number of at least 4 RBC transfusions/8 week compared with the pretreatment transfusion number in the previous 8 week. HI-P is defined as an absolute increase of ≥ 30 X 10^9/L for participants starting with > 20 X and an increase from < 20 X 10^9/L to > 20 X 10^9/L and by at least 100%. HI-N is defined as At least 100% increase and an absolute increase > 0.5 X 10^9/L. (NCT02577406)
Timeframe: From randomization up to approximately 49 months

InterventionParticipants (Count of Participants)
AG-22133
Conventional Care Regimens (CCRs)9

One-Year Survival Rate

The proportion of participants alive at 1 year after randomization (NCT02577406)
Timeframe: From randomization to 1 year after randomization

InterventionProportion of participants (Number)
AG-2210.375
Conventional Care Regimens (CCRs)0.261

Overall Remission Rate

The number of participants with CR + CRi + CRp according to modified International Working Group Acute Myeloid Leukemia (IWG AML) response criteria. Complete remission (CR) is defined as <5% blasts in a BM aspirate sample with marrow spicules and a count of ≥200 nucleated cells. There should be no blasts with Auer rods and no extramedullary disease. CR must also include the following conditions: absolute neutrophil count (ANC) ≥1,000/μL, Platelet count ≥100,000/μL, and independent of red cell transfusions for ≥1 week before each response assessment. Complete remission with incomplete neutrophil recovery (CRi) is all criteria of CR except ANC. Complete remission with incomplete platelet recovery (CRp) is all criteria of CR except platelet count. (NCT02577406)
Timeframe: From randomization up to approximately 49 months

InterventionParticipants (Count of Participants)
AG-22117
Conventional Care Regimens (CCRs)7

Overall Response Rate

Number of participants with MLFS + CR + CRi + CRp + PR according to modified International Working Group Acute Myeloid Leukemia (IWG AML) response criteria. Complete response (CR) and morphologic leukemia-free state (MLFS) are defined as <5% blasts in a BM aspirate sample with marrow spicules and a count of ≥200 nucleated cells. There should be no blasts with Auer rods and no extramedullary disease. CR must also include: absolute neutrophil count (ANC) ≥1,000/μL, Platelet count ≥100,000/μL, and independent of red cell transfusions for ≥1 week before each response assessment. Complete remission with incomplete neutrophil recovery (CRi) is all criteria of CR except ANC. Complete remission with incomplete platelet recovery (CRp) is all criteria of CR except platelet count. Partial remission (PR) is defined as all hematologic criteria of CR with a >50% decrease in the percentage of BM blasts to 5% to 25%. (<5% considered if Auer rods are present). (NCT02577406)
Timeframe: From randomization up to approximately 49 months

InterventionParticipants (Count of Participants)
AG-22125
Conventional Care Regimens (CCRs)7

Overall Survival (OS)

The time between randomization and death from any cause. Participants who drop-out or are alive at the end of trial will have their OS times censored at the time of last contact, as appropriate. (NCT02577406)
Timeframe: From randomization to death due to any cause (up to approximately 49 months)

InterventionMonths (Mean)
AG-2216.5
Conventional Care Regimens (CCRs)6.2

The Number of Participants That Underwent Hematopoietic Stem Cell Transplantation (HSCT)

The number of participants that underwent hematopoietic stem cell transplantation during the study. (NCT02577406)
Timeframe: From randomization up to approximately 49 months

InterventionParticipants (Count of Participants)
AG-2217
Conventional Care Regimens (CCRs)2

The Percent of Participants Experiencing Clinically Significant Laboratory Abnormalities - Hematology

The percent of participants with clinically significant hematology laboratory abnormalities. A clinically significant laboratory abnormality is defined as meeting Grade 3 or Grade 4 criteria according to the Common Terminology Criteria for Adverse Events (CTCAE). Grade 3 is defined as severe or medically significant but not immediately life threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self care. Grade 4 is defined as life-threatening consequences; urgent intervention indicated. The hematology panel includes complete blood count (CBC) with differential, including red blood cell (RBC) count, hemoglobin, hematocrit, mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC), white blood cell (WBC) count (with differential), absolute neutrophil count (ANC) and platelet count. (NCT02577406)
Timeframe: From first dose up to approximately 49 months

InterventionPercent of Participants (Number)
AG-22194.3
Conventional Care Regimens (CCRs)89.4

The Percent of Participants Experiencing Clinically Significant Laboratory Abnormalities - Serum Chemistry

The percent of participants with clinically significant serum chemistry laboratory abnormalities. A clinically significant laboratory abnormality is defined as meeting Grade 3 or Grade 4 criteria according to the Common Terminology Criteria for Adverse Events (CTCAE). Grade 3 is defined as severe or medically significant but not immediately life threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self care. Grade 4 is defined as life-threatening consequences; urgent intervention indicated. The chemistry panel includes sodium, potassium, calcium, magnesium, chloride, phosphorus, CO2, bicarbonate, blood urea nitrogen (BUN), creatinine, glucose, albumin, total protein, alkaline phosphatase (ALP), bilirubin (total and direct), uric acid, lactate dehydrogenase (LDH), AST/SGOT, ALT/SGPT, gamma glutamyl transpeptidase (GGT), amylase and lipase. (NCT02577406)
Timeframe: From first dose up to approximately 49 months

InterventionPercent of Participants (Number)
AG-22142.7
Conventional Care Regimens (CCRs)21.3

The Percent of Participants Experiencing Clinically Significant Vital Sign Abnormalities

The percent of participants with clinically significant vital sign abnormalities including weight, temperature, blood pressure, pulse rate, and respiratory rate. (NCT02577406)
Timeframe: From first dose up to approximately 49 months

InterventionPercent of Participants (Number)
AG-2210
Conventional Care Regimens (CCRs)0

The Percentage of Participants Experiencing Clinically Significant Electrocardiogram (ECG) Abnormalities

The percent of participants with clinically significant ECG abnormalities. 12-lead ECG was assessed by a physician trained in ECG interpretation. Intervals including PR, QRS, QT and RR were collected, as well as heart rate and rhythm. (NCT02577406)
Timeframe: From first dose up to approximately 49 months

InterventionPercentage of Participants (Number)
AG-2217.6
Conventional Care Regimens (CCRs)2.1

Time to Response

Time from randomization to first documented MLFS/CR/CRi/CRp/PR. Complete remission (CR) and morphologic leukemia-free state (MLFS) are defined as <5% blasts in a BM aspirate sample with marrow spicules and a count of ≥200 nucleated cells. There should be no blasts with Auer rods and no extramedullary disease. CR must also include the following conditions: absolute neutrophil count (ANC) ≥1,000/μL, Platelet count ≥100,000/μL, and independent of red cell transfusions for ≥1 week before each response assessment. Complete remission with incomplete neutrophil recovery (CRi) is all criteria of CR except ANC. Complete remission with incomplete platelet recovery (CRp) is all criteria of CR except platelet count. partial remission (PR) is defined as all hematologic criteria of CR with a >50% decrease in the percentage of BM blasts to 5% to 25%. (<5% considered if Auer rods are present). (NCT02577406)
Timeframe: From randomization to to first documented MLFS/CR/CRi/CRp/PR (up to approximately 49 months)

InterventionDays (Median)
AG-22158.0
Conventional Care Regimens (CCRs)56.0

Time to Treatment Failure

Time from randomization to discontinuation of study treatment due to any cause (NCT02577406)
Timeframe: From randomization to discontinuation of study treatment due to any cause (up to approximately 49 months)

InterventionMonths (Median)
AG-2214.9
Conventional Care Regimens (CCRs)1.9

Treatment Mortality at 30 Days

The number of participant deaths from any cause within 30 days of initiation of study treatment. (NCT02577406)
Timeframe: From first dose to 30 days after first dose

InterventionParticipants (Count of Participants)
AG-22110
Conventional Care Regimens (CCRs)13

Treatment Mortality at 60 Days

The number of participant deaths from any cause within 60 days of initiation of study treatment. (NCT02577406)
Timeframe: From first dose to 60 days after first dose

InterventionParticipants (Count of Participants)
AG-22127
Conventional Care Regimens (CCRs)30

Change From Baseline in the European Organization for Research and Treatment of Cancer Quality-of-Life Questionnaire (EORTC QLQ-C30)

The change from baseline in the EORTC QLQ-C30 questionnaire. The EORTC QLQ-C30 is composed of 30 items that address general physical symptoms, physical functioning, fatigue and malaise, and social and emotional functioning. Subscale scores are transformed to a 0 to 100 scale, with higher scores on functional scales indicating better function and higher scores on symptom scales indicating worse symptoms. A change of at least 10 points on the standardized domain scores was required for it to be considered meaningful. Results obtained just prior to the start of study treatment on Day 1 of Cycle 1 will serve as the baseline values. If not available, the most recent screening results prior to the start of study treatment on Day 1 of Cycle 1 will be considered the baseline values. (NCT02577406)
Timeframe: From baseline to cycle 2 day 1 (up to approximately 1 month)

,
InterventionChange from baseline in QLQ-C30 score (Mean)
Global QoLPhysical FunctioningRole FunctioningCognitive FunctioningEmotional FunctioningSocial FunctioningFatigueNausea / VomitingPainDyspneaInsomniaAppetite LossConstipationDiarrheaFinancial Difficulties
AG-221-4.7-3.1-3.8-0.60.9-0.65.59.06.0-1.38.510.72.84.1-0.3
Conventional Care Regimens (CCRs)-4.0-6.7-7.8-5.1-0.7-10.27.51.97.00.01.18.61.61.6-3.2

The Number of Participants Experiencing Adverse Events (AEs)

The number of participants experiencing different types of adverse events (AE). An AE is any noxious, unintended, or untoward medical occurrence that may appear or worsen in a participant during the course of a study. A Serious Adverse Event (SAE) is any AE occurring at any dose that: results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect, or constitutes an important medical event. All AEs were coded using the MedDRA Version 22.0. Adverse events were analyzed in terms of treatment-emergent AEs (TEAEs). A treatment-related TEAE was defined as a TEAE that was suspected by the investigator to be related to study treatment. The severity was graded by the study personnel based on NCI National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) Version 4.03. (NCT02577406)
Timeframe: From first dose up to 28 days after last dose (up to approximately 49 months)

,
InterventionParticipants (Count of Participants)
Treatment-Emergent Adverse Event (TEAE)TEAE Related to Study DrugGrade ≥ 3 TEAEGrade ≥ 3 TEAE Related to Study DrugSerious TEAESerious TEAE Related to Study DrugTEAE Leading to Discontinuation of Study DrugTreatment-related TEAE Leading to Discontinuation of Study DrugTEAE Leading to Study Drug Dose Reduction OnlyTreatment-related TEAE Leading to Study Drug Dose Reduction OnlyTEAE Leading to Study Drug Dose Interruption OnlyTreatment-related TEAE Leading to Study Drug Dose Interruption OnlyTEAE Leading to Study Drug Dose Reduction or Dose InterruptionTreatment-related TEAE Leading to Study Drug Dose Reduction or Dose InterruptionTEAE Leading to DeathTreatment-related TEAE Leading to Death
AG-2211571211467413834355221884469256771
Conventional Care Regimens (CCRs)131861124992301663335203621335

30-day All-cause Mortality

"As a summary of adverse events (captured on trial using the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 5.0), 30-day all cause mortality is reported as a percent of patients treated at the MTD/RP2D.~Will be estimated and 95% confidence intervals will be calculated. Regression models (logistic regression for binary endpoints, Cox regression for time-to-event endpoints [Cox models for the hazard of the subdistribution for events with competing risks]) will be used to compare outcomes with patients who have received GCLAM without GO at our institution, controlling for measured prognostic factors." (NCT03531918)
Timeframe: Up to 5 years. 30-day all-cause mortality is reported

Interventionproportion died on/before day 30 (Number)
GO3 Dose Level Includes Phase 1 and Phase 2.03

Maximum Tolerated Dose (MTD) of Gemtuzumab Ozogamicin (GO) When Added to GCLAM (Phase 1)

Defined as the highest dose studied in which the incidence of Dose Limiting Toxicity (DLT) is ≤33% (≤4 of 12 patients experiencing DLT), defined as any Grade 3 non-hematologic toxicity lasting >48 hours that results in >7-day delay of the subsequent treatment cycle, with the exception of febrile neutropenia or infection or toxicities secondary to febrile neutropenia or infection, or any Grade ≥4 non-hematologic toxicity except febrile neutropenia/infection (or toxicities secondary to febrile neutropenia or infection) unless felt to be a direct consequence of treatment-related toxicity (e.g. intestinal infection following mucosal barrier breakdown), and with the exception of constitutional symptoms if recovery to Grade ≤2 within 14 days. The National Cancer Institute Common Terminology Criteria for Adverse Events v5.0 will be used. (NCT03531918)
Timeframe: At time of count recovery, second cycle of treatment, response assessment or removal from protocol (at approximately 1 month).

InterventionParticipants (Count of Participants)
GO1 Dose Level Phase 1NA
GO3 Dose Level Includes Phase 1 and Phase 2NA

Measurable Residual Disease (MRD) and Remission Rates: Alapasia (MRDneg)

"Will be estimated and 95% confidence intervals will be calculated. Regression models (logistic regression for binary endpoints, Cox regression for time-to-event endpoints [Cox models for the hazard of the subdistribution for events with competing risks]) will be used to compare outcomes with patients who have received GCLAM without GO at our institution, controlling for measured prognostic factors.~o Aplasia rate is defined in this protocol as frequency of patients without blood count recovery after chemotherapy and bone marrow examination showing hypocellularity not meeting cellularity criteria for morphologic leukemia free state (MLFS). MRD negative (MRDneg) status is defined as negative for leukemic markers by multiparameter flow cytometry.~Aplasia was defined as absence of tumoral cells but cellularity not meeting criteria for MLFS.~Statistical significance tests were not performed." (NCT03531918)
Timeframe: 90 days

InterventionParticipants (Count of Participants)
GO1 Dose Level Phase 10
GO3 Dose Level Includes Phase 1 and Phase 22

Measurable Residual Disease (MRD) and Remission Rates: CR/CRi

"Will be estimated and 95% confidence intervals will be calculated. Regression models (logistic regression for binary endpoints, Cox regression for time-to-event endpoints [Cox models for the hazard of the subdistribution for events with competing risks]) will be used to compare outcomes with patients who have received GCLAM without GO at our institution, controlling for measured prognostic factors.~Complete response (CR) + complete response with incomplete hematologic recovery (CRi) rate is defined as the frequency of patients achieving CR or CRi per the European LeukemiaNet 2017 criteria as defined above. MRD negative (MRDneg) status is defined as negative for leukemic markers by multiparameter flow cytometry.~Aplasia was defined as absence of tumoral cells but cellularity not meeting criteria for MLFS.~Statistical significance tests were not performed." (NCT03531918)
Timeframe: 90 days

InterventionParticipants (Count of Participants)
GO1 Dose Level Phase 15
GO3 Dose Level Includes Phase 1 and Phase 252

Measurable Residual Disease (MRD) and Remission Rates: CRi (MRDneg)

"Will be estimated and 95% confidence intervals will be calculated. Regression models (logistic regression for binary endpoints, Cox regression for time-to-event endpoints [Cox models for the hazard of the subdistribution for events with competing risks]) will be used to compare outcomes with patients who have received GCLAM without GO at our institution, controlling for measured prognostic factors.~Complete response with incomplete hematologic recovery (CRi) rate is defined as the frequency of patients achieving CRi, which is defined by the European LeukemiaNet 2017 guidelines as all CR criteria except for residual neutropenia (ANC <1.0 x 109/L [1000/mL]) or thrombocytopenia (platelet count <100 x 109/L [100,000/mL]). MRD negative (MRDneg) status is defined as negative for leukemic markers by multiparameter flow cytometry.~Aplasia was defined as absence of tumoral cells but cellularity not meeting criteria for MLFS.~Statistical significance tests were not performed." (NCT03531918)
Timeframe: 90 days

InterventionParticipants (Count of Participants)
GO1 Dose Level Phase 11
GO3 Dose Level Includes Phase 1 and Phase 27

Measurable Residual Disease (MRD) and Remission Rates: MLFS (MRDneg)

"Will be estimated and 95% confidence intervals will be calculated. Regression models (logistic regression for binary endpoints, Cox regression for time-to-event endpoints [Cox models for the hazard of the subdistribution for events with competing risks]) will be used to compare outcomes with patients who have received GCLAM without GO at our institution, controlling for measured prognostic factors.~Morphologic leukemia free state (MLFS) rate is defined as the frequency of patients achieving MLFS, which is defined by the European LeukemiaNet guidelines as bone marrow blasts <5%; absence of blasts with Auer rods; absence of extramedullary disease; no hematologic recovery required. At least 200 cells should be enumerated or cellularity should be at least 10%. MRD negative (MRDneg) status is defined as negative for leukemic markers by multiparameter flow cytometry.~Aplasia was defined as absence of tumoral cells but cellularity not meeting criteria for MLFS.~Statistical significance" (NCT03531918)
Timeframe: 90 days

InterventionParticipants (Count of Participants)
GO1 Dose Level Phase 10
GO3 Dose Level Includes Phase 1 and Phase 22

Overall Survival

OS was calculated for all participants and measured from initial trial therapy to death from any cause. Patients last known ot be alive were censored at date of last contact. Will be estimated using the Kaplan-Meier method. Regression models (logistic regression for binary endpoints, Cox regression for time-to-event endpoints [Cox models for the hazard of the subdistribution for events with competing risks]) will be used to compare outcomes with patients who have received GCLAM without GO at our institution, controlling for measured prognostic factors. (NCT03531918)
Timeframe: 3 years and 1 month

Interventionpercentage of participants (Number)
GO3 Dose Level60

Relapse-free Survival of GO3 Cohort

RFS was calculated for participants who achieved a complete remission (with or without count recovery; CR or CRi) and measured from the date remission to the first of relapse from CR/CRi or death from any cause. Patients last known to be alive in CR /CRi were censored at date of last contact. Will be estimated using the Kaplan-Meier method. Time to relapse will be estimated using non-parametric estimates of the cumulative incidence curve with death analyzed as a competing event. Regression models (logistic regression for binary endpoints, Cox regression for time-to-event endpoints [Cox models for the hazard of the subdistribution for events with competing risks]) will be used to compare outcomes with patients who have received GCLAM without GO at our institution, controlling for measured prognostic factors. (NCT03531918)
Timeframe: Up to 5 years. 2-year RFS reported.

Interventionpercentage of participants (Number)
GO3 Dose Level Includes Phase 1 and Phase 251

Event-free Survival (EFS) Rate (Phase 2)

A two-stage design will be used to evaluate the EFS. Patients treated at the maximum tolerated dose (MTD) from the phase 1 portion of the trial will be used in the phase 2 analysis. If censoring occurs, secondary analyses analyzing 6-month or 1-year EFS accounting for censoring will be done, including estimating 6-month or 1-year EFS using the Kaplan-Meier method. The first stage of the two-stage phase 2 design will evaluate 30 patients. If 20 or more of the first 30 patients are alive without event at 6-months after study registration, an additional 30 patients will be enrolled. If 46 or more of the 60 patients treated at the MTD are alive and without event at 6-months after study registration, the study will consider the regimen of interest for further investigation. Patients last known to be alive in CR were censored at date of last contact. (NCT03531918)
Timeframe: From the start of study treatment, assessed at 6 months and 1 year

Interventionpercent of participants (Number)
Event-free Survival (6 months)Event-free survival (12 months)
GO3 Dose Level Includes Phase 1 and Phase 27358

Measurable Residual Disease (MRD) Rates and Remission Rates: CR

"Will be estimated and 95% confidence intervals will be calculated. Regression models (logistic regression for binary endpoints, Cox regression for time-to-event endpoints [Cox models for the hazard of the subdistribution for events with competing risks]) will be used to compare outcomes with patients who have received GCLAM without GO at our institution, controlling for measured prognostic factors.~Complete response (CR) rate is defined as the frequency of patients achieving CR, which is defined by the European LeukemiaNet 2017 guidelines as bone marrow blasts <5%; absence of circulating blasts and blasts with Auer rods; absence of extramedullary disease; ANC ≥1.0 x 109/L (1000/mL); platelet count ≥100 x 109/L. (100 000/mL). MRD negative (MRDneg) status is defined as negative for leukemic markers by multiparameter flow cytometry.~Aplasia was defined as absence of tumoral cells but cellularity not meeting criteria for MLFS.~Statistical significance tests were not performed." (NCT03531918)
Timeframe: 90 days

,
InterventionParticipants (Count of Participants)
MRDnegMRDpos
GO1 Dose Level Phase 140
GO3 Dose Level Includes Phase 1 and Phase 2387

Maximum Tolerated Dose (MTD) Oxaliplatin

MTD defined as dose level at which 2/3 or 2/6 participants experience Dose Limiting Toxicity (DLT), where DLTs are any oxaliplatin-related ≥Grade 3 non-hematological toxicity involving a major organ system (brain, heart, kidney, liver, lung) in the National Cancer Institute (NCI) Version 3.0 toxicity scale. (NCT00452374)
Timeframe: From treatment onset to end of each cycle of treatment (every 21 days)

Interventionmg/m^2 (Number)
Oxaliplatin, Fludarabine, Cytarabine + Rituximab25

Number of Participants With a Complete Response or Partial Response

According to International Workshop Response Criteria for Non-Hodgkin's Lymphomas: Complete remission (CR) defined as > 30% lymphocytes in the bone marrow, recovery of blood counts and no clinical symptoms; and Partial remission (PR) defined as > 50% decrease of clinical symptoms from baseline and recovery from blood counts. (NCT00452374)
Timeframe: Evaluation every 3 cycles of treatment (28 days per cycle), approximately 90 days

InterventionParticipants (Number)
Complete response (CR)Partial response (PR)
Oxaliplatin, Fludarabine, Cytarabine + Rituximab1228

Overall Survival of Patients Over Age 60 With Newly Diagnosed AML/High Risk MDS

Compared to historical data of a completed Southwestern Oncology Group (SWOG) trial of azacitidine and gemtuzumab ozogamicin. (NCT02121418)
Timeframe: At 6 months

Interventionparticipants (Number)
Treatment (Decitabine, Cytarabine)7

Response Rate

Rate of Complete Response or Complete Response with Incomplete Count Recovery (NCT02121418)
Timeframe: Up to 2 years

Interventionparticipants (Number)
Complete RespnoseComplete Response with Incomplete Count Recovery
Treatment (Decitabine, Cytarabine)43

Median Overall Survival (OS) Time

OS was defined as the time from start of study drug to death from any cause. OS was estimated using Kaplan-Meier analysis. Participants alive at the last date known to be alive were censored for the analysis. (NCT01880437)
Timeframe: Up to death or 30 days of last dose of study drug (maximum treatment duration = 225 days)

Interventionmonths (Median)
Poor Risk Cytogenetics3.38
FLT-3 Mutation Positive1.43
Neither Poor Risk Cytogenetics Nor FLT-33.65

Percentage of Participants With an Event of Death During the Study

(NCT01880437)
Timeframe: Up to death or 30 days of last dose of study drug (maximum treatment duration = 225 days)

Interventionpercentage of participants (Number)
Poor Risk Cytogenetics80.0
FLT-3 Mutation Positive100.0
Neither Poor Risk Cytogenetics Nor FLT-389.5

Duration of Overall Response (DOR)

DOR is defined as the time from the first occurrence of a documented overall response to the time of relapse, as determined by the investigator using International Working Group (IWG) criteria (Participants not falling under any of the response criteria [CR or CRi or MLFS or PR] described under outcome measure 1 were considered as non-responders) or death from any cause during the study (defined as death within 30 days after the last dose of study drug). (NCT01880437)
Timeframe: Up to 30 days of last dose of study drug (maximum treatment duration = 225 days)

Interventionweeks (Median)
DOR of participants with CRi (n=0,0,1)DOR of participants with PR (n=0,0,1)
Neither Poor Risk Cytogenetics Nor FLT3136.1

Percentage of Participants With CR, CRi, MLFS or PR at Anytime During Study Treatment

CR was defined as achieved if the neutrophils count >1000 cells/µL, platelets count >100000/µL, bone marrow blasts <5%, no Auer rods (clumps of azurophilic granular material that form elongated needles seen in the cytoplasm of leukemic blasts), no transfusion requirements and no signs of EMD. CRi was defined if either of the cell (neutrophil or platelet) lineage was not recovered (neutrophils > 1000 cells/µL or NA or platelets count >100000/µL or NA, bone marrow blasts <5% with no Auer rods and confirmed by flow cytometry with no signs of EMD. MLFS (neutrophil and platelet criteria were NA) was defined as bone marrow blasts <5% with no Auer rods and confirmed by flow cytometry with no signs of EMD. PR was defined as neutrophils count >1000 cells/µL, platelets count >100000/µL, and >50% decrease from baseline to a range of 5-25% of bone marrow blasts or blasts <5% with Auer rods. The 95% confidence intervals (CI) were constructed using Blyth-Still-Cassella method. (NCT01880437)
Timeframe: Up to 30 days of last dose of study drug (maximum treatment duration = 225 days)

,,
Interventionpercentage of participants (Number)
CRCRiMLFSPR
FLT-3 Mutation Positive0000
Neither Poor Risk Cytogenetics Nor FLT306.306.3
Poor Risk Cytogenetics0000

Complete Remission Rate (CR)

CR is calculated as the percentage of patients with complete remission after the therapy. The response criteria is based on International Working Group Criteria. Complete Remission: Neutrophils(cells/μl)>1000, Platelets (plt/μl)≥ 100,000,Bone marrow blasts (%) <5; CR with incomplete count recovery (CRi): meets criteria for CR except for neutrophils ≤1000 or Meets criteria for CR except for platelets< 100,000; Partial Remission: CR except for Bone marrow blasts (%) Decrease of ≥ 50% to value between 5% and 25%; Treatment Failure:Persistent acute myeloid leukemia in blood or bone marrow, or therapy fails to achieve a remission of any category, or death prior to response assessment (NCT02658487)
Timeframe: Up to 3 months

InterventionParticipants (Count of Participants)
Treatment (Vosaroxin, Cytarabine)20

Event-free Survival

Survival distribution will be estimated using the method of Kaplan and Meier. Event-free survival time is defined as the time from start of therapy to progression or death for any reason (which ever comes first), and those alive without progression are censored at the last day of follow up. (NCT02658487)
Timeframe: From start of therapy up to 1 year

Interventionmonths (Median)
Treatment (Vosaroxin, Cytarabine)7.6

Frequency of Grade 3-5 Adverse Event Related to Cytarabine and Vosaroxin (7+V)

Events will be graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Counts of all events are summed up. (NCT02658487)
Timeframe: Up to 3 months

Interventionadverse events (Number)
Treatment (Vosaroxin, Cytarabine)133

Leukemia-free Survival (LFS or DFS)

Survival distribution will be estimated using the method of Kaplan and Meier. LFS time is defined as the time from complete remission to disease progression or death for any reason and those disease free and alive are censored at the last day of follow up. (NCT02658487)
Timeframe: The time from complete remission to disease progression or death for any reason, assessed up to 1 year

Interventionmonths (Median)
Treatment (Vosaroxin, Cytarabine)NA

Overall Survival

Survival distribution will be estimated using the method of Kaplan and Meier. The overall survival time is defined as time from start of therapy to death of any reason. Those alive are censored at the last day of known alive. (NCT02658487)
Timeframe: The time from start of therapy to death, assessed up to 1 year

Interventionmonths (Median)
Treatment (Vosaroxin, Cytarabine)10.7

Rate of CR/CRi

"Frequency of Complete Remission / Complete Remission with Incomplete Count Recovery (CR/CRi) after 7+V induction and/or re-induction" (NCT02658487)
Timeframe: Up to 3 months

InterventionProportion of participants (Number)
Treatment (Vosaroxin, Cytarabine)0.55

Event-free Survival

All randomized subjects were assessed for event-free survival (EFS). EFS was defined as the time from study randomization to the date of induction treatment failure (persistent disease), relapse from CR or CRi or death from any cause, whichever came first. Subjects alive and not known to have any of these events were censored on thee date they were last examined on study. (NCT01696084)
Timeframe: From the date of randomization to the date that persistent disease was documented or the date of relapse after CR or death, whichever came first

Interventionmonths (Median)
Arm A (CPX-351)2.53
Arm B (7+3)1.31

Overall Survival

Overall survival was measured from the date of randomization to death from any cause, subjects not known to have died by the last follow-up were censored on the date they were last known to be alive. (NCT01696084)
Timeframe: From the date of randomization to death from any cause

Interventionmonths (Median)
Arm A (CPX-351)9.56
Arm B (7+3)5.95

Proportion of Subjects Receiving a Stem Cell Transplant

The number and percentage of subjects transferred for HSCT after induction treatment was recorded. (NCT01696084)
Timeframe: Post Induction

InterventionParticipants (Count of Participants)
Arm A (CPX-351)52
Arm B (7+3)39

Proportion of Subjects With a Response

Complete Remission (CR) (NCT01696084)
Timeframe: Post Induction

InterventionParticipants (Count of Participants)
Arm A (CPX-351)57
Arm B (7+3)40

Rate of Achieving Morphologic Leukemia-free State

All randomized subjects with at least 1 evaluable postrandomization bone marrow assessment performed on or after Day 14 after the last induction were assessed for MLFS. (NCT01696084)
Timeframe: Day 14

InterventionParticipants (Count of Participants)
Arm A (CPX-351)87
Arm B (7+3)66

Remission Duration

Only subjects achieving CR or CRi were assessed for remission duration. (NCT01696084)
Timeframe: From the date of achievement of a remission until the date of relapse or death from any cause

Interventionmonths (Median)
Arm A (CPX-351)6.93
Arm B (7+3)6.11

Median Overall Survival (OS)

Overall Survival (OS), defined for those patients who have achieved CR or CRi as the time from study entry to disease progression, relapse or death due to any cause, whichever is earlier, to be analyzed similarly. Descriptive analysis was planned for this measure. (NCT00831766)
Timeframe: Up to 24 Months

Interventionmonths (Median)
All Participants Treated at MTD11.22

Median Progression-Free Survival (PFS)

Progression-free survival (PFS), defined as the time from study entry to disease progression, relapse, or death due to any cause, whichever is earlier, will be summarized with the Kaplan-Meier curve. (NCT00831766)
Timeframe: 24 months

Interventionmonths (Median)
All Participants Treated at MTD7.55

Phase I: Recommended Phase II Dose

For the Phase I component, no formal statistical analysis was planned. The primary endpoint is to determine the maximum tolerated dose (MTD) and recommended Phase II dose of lenalidomide given in combination with standard idarubicin + cytarabine induction therapy. (NCT00831766)
Timeframe: 18 months

Interventionmg/day (Number)
Phase I Participants20

Phase II: Complete Response Rate of Participants Treated at Maximum Tolerated Dose (MTD)

Percentage of participants achieving CR/CRi. Complete Response (CR) plus Complete Response with Incomplete Count Recovery (CRi) rates. Response rates (CR + CRi) of lenalidomide following idarubicin and cytarabine induction therapy in older patients with previously untreated AML. A CR designation requires that the patient achieve the morphologic leukemia-free state and have an absolute neutrophil count of more than 1,000/μL and platelets of 100,000/μL. CRi: After chemotherapy, patients fulfill all of the criteria for CR except for residual neutropenia (1,000/μL) or thrombocytopenia (100,000/μL). (NCT00831766)
Timeframe: 24 months

Interventionpercentage of participants (Number)
All Participants Treated at MTD54

Rate of Lenalidomide Related Toxicity During Maintenance Therapy

Rate of toxicities of lenalidomide as maintenance therapy according to the National Cancer Institute Common Toxicity Criteria (CTC) V3. Adverse Events: Possibly Related; Probably Related, or Definitely Related to study treatment. Events are categorized as Grade 1 or 2, or as Grade 3 or 4. (NCT00831766)
Timeframe: 24 months

InterventionParticipants (Count of Participants)
Leukocytes (total WBC) - low - Grade 1 or 2Leukocytes (total WBC) - low - Grade 3 or 4ANC/AGC - Grade 1 or 2ANC/AGC - Grade 3 or 4Platelets - low - Grade 1 or 2Platelets - low - Grade 3 or 4Fatigue - Grade 1 or 2Fatigue - Grade 3 or 4Dry skin - Grade 1 or 2Dry skin - Grade 3 or 4Pruritus/itching - Grade 1 or 2Pruritus/itching - Grade 3 or 4Rash/desquamation - Grade 1 or 2Rash/desquamation - Grade 3 or 4Constipation - Grade 1 or 2Constipation - Grade 3 or 4Diarrhea - Grade 1 or 2Diarrhea - Grade 3 or 4Nausea - Grade 1 or 2Nausea - Grade 3 or 4Ulcer, GI - Anus - Grade 1 or 2Ulcer, GI - Anus - Grade 3 or 4Hemorrhage, GI - Rectum - Grade 1 or 2Hemorrhage, GI - Rectum - Grade 3 or 4Infection - Skin (cellulitis) Grade 1 or 2Infection - Skin (cellulitis) Grade 3 or 4AST, SGOT - Grade 1 of 2AST, SGOT - Grade 3 or 4Pain - Head/headache - Grade 1 or 2Pain - Head/headache - Grade 3 or 4
Maintenance Phase Participants100130101010201020101010102010

Disease-free Survival

(NCT00863434)
Timeframe: Every 3 months for 2 years, and then annually for 3 years

Interventionmonths (Median)
Treatment (Colony Stimulating Factor and Chemotherapy)6.43

Minimal Residual Disease as Assessed by Bone Marrow Flow Cytometry

Percent of white blood cells that are blasts in the bone marrow post-treatment. (NCT00863434)
Timeframe: Post-treatment

Interventionpercent of white blood cells (Median)
Treatment (Colony Stimulating Factor and Chemotherapy)1.8

Overall Survival

(NCT00863434)
Timeframe: Every 3 months for 2 years, and then annually for 3 years

Interventionmonths (Median)
Treatment (Colony Stimulating Factor and Chemotherapy)9.73

Relapse Free Survival

Percentage of participants alive and without relapsed disease at two years. (NCT01588951)
Timeframe: 2 years

Interventionpercentage of participants (Number)
No Leukemia Stem Cells - Consolidation0
Leukemia Stem Cells - Consolidation0
Leukemia Stem Cells - Transplant0

Overall Survival (OS) at 1 Year

(NCT00392834)
Timeframe: 1 year post treatment

InterventionCumulative proportion surviving at 1 yr (Number)
Regimen A (R-CODOX-M Chemotherapy)1.0
Regimen B (Rituximab and IVAC Chemotherapy)0.82

Complete Response

Bone marrow showing less than 5% myeloblasts with normal maturation of all cell lines, an Absolute Neutrophil Count of at least 1000/mililiter and a platelet count of 100,000 mililiter, absence of blast in peripheral blood, absence of identifiable leukemic cells in the bone marrow, clearance of disease-associated cytogenetic abnormalities, and clearance of any previously existing extramedullary disease. A complete remission must be confirmed 4 to 6 weeks after the initial documentation. If possible, at least one bone marrow biopsy should be performed to confirm the complete remission. (NCT00407966)
Timeframe: 6 months

Interventionparticipants (Number)
Arm A45

Complete Response

Bone marrow showing less than 5% leukemic blasts with normal maturation of all cell lines, an ANC of at least 1000/uL and a platelet count of 100,000/uL, absence of blast in peripheral blood, absence of identifiable leukemic cells in the bone marrow, clearance of disease-associated cytogenetic abnormalities, and clearance of any previously existing extramedullary disease. Repeat marrow confirmation 4-6 weeks following the marrow documenting CR is not required due to the need for continued treatment in CR. (NCT00795002)
Timeframe: 1 year

Interventionparticipants (Number)
Arm I24
Arm II29

Disease-free Survival

This will be defined as the time between study entry and the first date that recurrent or progressive disease is objectively documented, or death from any cause occurs. (NCT00795002)
Timeframe: up to 2 years

Interventionmonths (Median)
Arm I13.6
Arm II12.0

Number of Participants Experiencing Death From Any Cause Within 60 Days of Starting FLAM

Toxicity defined as death from any cause within 60 days of starting FLAM. (NCT00795002)
Timeframe: 60 days

InterventionParticipants (Count of Participants)
Arm I3
Arm II3

Disease-free Survival

Time from date of treatment start until the date of first objective documentation of disease-relapse. (NCT01272245)
Timeframe: Up to 5 years after completion of active treatment and while on study. Participants may receive up to 24 courses of study medication.

InterventionMonths (Median)
Omacetaxine and Cytarabine3.1

Evaluation of CR Duration

The date of Complete Response to the date of loss of response or last follow-up. (NCT01272245)
Timeframe: Up to 5 years after completion of active treatment and while on study. Participants may receive up to 24 courses of study medication.

InterventionMonths (Median)
Omacetaxine and Cytarabine10.6

Induction Mortality

Death within 8 weeks from the start of treatment. (NCT01272245)
Timeframe: Up to 1 year

InterventionParticipants (Count of Participants)
Omacetaxine and Cytarabine4

Overall Survival

Time from date of treatment start until date of death due to any cause (NCT01272245)
Timeframe: Up to 5 years after completion of active treatment and while on study. Participants may receive up to 24 courses of study medication.

InterventionMonths (Median)
Omacetaxine and Cytarabine8.1

Percentage of Participants With Complete Remission (CR)

Complete response (CR) defined as: Peripheral blood counts, no circulating blasts, neutrophil count ≥ 1.0 ×109/L, platelet count ≥ 100 ×109/L, bone marrow aspirate and biopsy, ≤5% blasts, no detectable auer rods, no extramedulary leukemia (NCT01272245)
Timeframe: Up to 4 months

Interventionpercentage of participants (Number)
Omacetaxine and Cytarabine44

Proportion of Patients Achieving Hematologic and Molecular Complete Remission (CR) After Induction Therapy

approximate time: at the recovery of cytopenia (NCT00844298)
Timeframe: 1 month

Interventionpercentage of analyzable subjects (Number)
Nilotinib+mVPD91

Complete Response (CR) Rate

"To compare the complete response rate between patients who receive standard 7+3 therapy or IA to patients who receive IA + vorinostat.~Complete response is defined as: ANC >= 1,000/mcl, platelet count >= 100,000/mcl, < 5% bone marrow blasts, no Auer rods, no evidence of extramedullary disease (no requirements for marrow cellularity or hemoglobin concentration)" (NCT01802333)
Timeframe: Up to 5 years

InterventionPercentage of participants (Number)
Arm I (Standard Dose Cytarabine, Daunorubicin Hydrochloride)75
Arm II (High-dose Cytarabine, Idarubicin)80
Arm III (Vorinostat, High-dose Cytarabine, Idarubicin)77

Disease-free Survival (DFS) Among High Risk Patients

"DFS is calculated for patients who have achieved a CR or CRi (complete response with incomplete blood count recovery). DFS will be measured from the date of CR or CRi until relapse from CR or CRi for death from any cause. Observation is censored at the date of last follow-up for patients last known to be alive without report of relapse.~2-year DFS for high risk patients will be estimated using the Kaplan-Meier method." (NCT01802333)
Timeframe: DFS assessed for up to 5 years, 2 year DFS reported

InterventionProportion of participants (Number)
High Risk Patients0.30

Disease-free Survival (DFS)

"To compare the disease-free survival (DFS) between patients who receive standard 7+3 therapy or IA to patients who receive IA + vorinostat.~DFS is calculated for patients who have achieved a CR or CRi (complete response with incomplete blood count recovery) . DFS will be measured from the date of CR or CRi until relapse from CR or CRi for death from any cause. Observation is censored at the date of last follow-up for patients last known to be alive without report of relapse.~2-year DFS by arm will be estimated using the Kaplan-Meier method." (NCT01802333)
Timeframe: DFS assessed for up to 5 years, 2 year DFS reported

InterventionProportion of participants (Number)
Arm I (Standard Dose Cytarabine, Daunorubicin Hydrochloride)0.48
Arm II (High-dose Cytarabine, Idarubicin)0.51
Arm III (Vorinostat, High-dose Cytarabine, Idarubicin)0.46

EFS of Arm I Compared to Arm II

"EFS is calculated for all patients from the date of initial registration on study until the first of the following: death from any cause, relapse from remission (CR or CRi) or completion of protocol Induction/Re-Induction therapy without documentation of CR or CRi.~A two-sided test of the hazard ratio (HR) of 7:3: IA (versus the null hypothesis of HR =1) will be done using a proportional hazards regression model with the stratification factors included as covariates.~2-year EFS by arm will be estimated using the Kaplan-Meier method." (NCT01802333)
Timeframe: EFS assessed for up to 5 years, 2 year EFS reported

InterventionProportion of participants (Number)
Arm I (Standard Dose Cytarabine, Daunorubicin Hydrochloride)0.36
Arm II (High-dose Cytarabine, Idarubicin)0.41

Event-free Survival (EFS)

"EFS is calculated for all patients from the date of initial registration on study until the first of the following: death from any cause, relapse from remission (CR or CRi) or completion of protocol Induction/Re-Induction therapy without documentation of CR or CRi.~2-year EFS by arm will be estimated using the Kaplan-Meier method. EFS will be compared between Arm I and Arm III and between Arm II and Arm III using Cox proportional hazards regression." (NCT01802333)
Timeframe: EFS assessed for up to 5 years, 2 year EFS reported

InterventionProportion of participants (Number)
Arm I (Standard Dose Cytarabine, Daunorubicin Hydrochloride)0.36
Arm II (High-dose Cytarabine, Idarubicin)0.41
Arm III (Vorinostat, High-dose Cytarabine, Idarubicin)0.37

Overall Survival (OS)

"To compare OS between patients who receive standard 7+3 therapy or IA to patients who receive IA + vorinostat.~OS is calculated for all patients from date of registration to date of death due to any cause. Patients last known to be alive are censored at date of last contact.~2-year OS by arm will be estimated using the Kaplan-Meier method." (NCT01802333)
Timeframe: OS assessed for up to 5 years, 2 year OS reported

InterventionProportion of participants (Number)
Arm I (Standard Dose Cytarabine, Daunorubicin Hydrochloride)0.56
Arm II (High-dose Cytarabine, Idarubicin)0.59
Arm III (Vorinostat, High-dose Cytarabine, Idarubicin)0.58

Prevalence of the Mutation NPM1 in Patients on This Study.

To estimate the prevalence of the mutation NPM1 in this patient population. (NCT01802333)
Timeframe: Baseline

Interventionpercentage of patients (Number)
All Arms Combined33

Rate of Allogeneic HCT

The goal of the transplant objective is to determine whether it is possible to conduct allogeneic HCT on 60% or more of adults with high-risk AML in first complete remission (alternative). If 40% or fewer of high-risk patients in CR can be transplanted, the proposed transplant support system will not be considered feasible. A one-sided binomial test compared to the null transplant rate will be conducted. (NCT01802333)
Timeframe: Up to 5 years

Interventionpercentage of patients (Number)
High Risk Patients in First Complete Remission65

Cytogenetic Risk Distribution of Patients on This Study

To estimate the cytogenetic risk distribution of patients on this study. (NCT01802333)
Timeframe: Baseline

Interventionpercentage of participants (Number)
High riskIntermediate riskLow risk
All Arms Combined22.364.213.5

Frequency and Severity of Toxicities

Number of patients with Grade 3-5 adverse events that were possibly, probably or definitely related to study drug are reported by given type of adverse event. (NCT01802333)
Timeframe: Up to 5 years

,,
InterventionParticipants (Count of Participants)
Abdominal distensionAbdominal infectionAbdominal painAcidosisAcute kidney injuryAdult respiratory distress syndromeAlanine aminotransferase increasedAlkaline phosphatase increasedAlkalosisAllergic reactionAnal hemorrhageAnal painAnal ulcerAnemiaAnorectal infectionAnorexiaAspartate aminotransferase increasedAtelectasisAtrial fibrillationAtrial flutterAtrioventricular block completeBlood and lymphatic system disorders - OtherBlood bilirubin increasedBone infectionBone painBronchopulmonary hemorrhageBullous dermatitisCD4 lymphocytes decreasedCardiac arrestCardiac disorders - Other, specifyCardiac troponin I increasedCatheter related infectionChillsChronic kidney diseaseCognitive disturbanceColitisColonic hemorrhageColonic perforationConduction disorderConfusionConjunctivitisConstipationConstrictive pericarditisCreatinine increasedDeath NOSDehydrationDental cariesDevice related infectionDiarrheaDisseminated intravascular coagulationDry mouthDry skinDuodenal hemorrhageDyspepsiaDysphagiaDyspneaEdema cerebralEdema limbsEjection fraction decreasedElectrocardiogram QT corrected interval prolongedEncephalopathyEnterocolitisEnterocolitis infectiousEpistaxisErythema multiformeErythrodermaEsophageal hemorrhageEsophageal painEsophagitisEye infectionFatigueFebrile neutropeniaFeverGGT increasedGait disturbanceGastric hemorrhageGastritisGastroesophageal reflux diseaseGastrointestinal disorders - Other, specifyGeneral disorders and admin site conditions-OtherGeneralized muscle weaknessGenital edemaGlucose intoleranceGum infectionHeadacheHeart failureHematomaHematuriaHepatic failureHepatic infectionHepatobiliary disorders - Other, specifyHyperglycemiaHyperhidrosisHyperkalemiaHypermagnesemiaHypernatremiaHypertensionHyperuricemiaHypoalbuminemiaHypocalcemiaHypoglycemiaHypokalemiaHypomagnesemiaHyponatremiaHypophosphatemiaHypotensionHypoxiaIleusInfections and infestations - Other, specifyInfective myositisInjury, poison and procedural complications-OtherIntracranial hemorrhageInvestigations - Other, specifyIrregular menstruationJejunal obstructionKidney infectionLaryngeal edemaLaryngeal mucositisLeft ventricular systolic dysfunctionLeukocytosisLipase increasedLower gastrointestinal hemorrhageLung infectionLymphocyte count decreasedMenorrhagiaMetabolism and nutrition disorders-Other, specifyMiddle ear inflammationMucosal infectionMucositis oralMulti-organ failureMusculoskeletal and connective tiss disorder-OtherMyalgiaNauseaNeck edemaNervous system disorders - Other, specifyNeutrophil count decreasedNon-cardiac chest painOral painPainPain in extremityPalmar-plantar erythrodysesthesia syndromePapulopustular rashParesthesiaPericardial effusionPericardial tamponadePeriorbital edemaPeripheral sensory neuropathyPharyngeal mucositisPharyngitisPlatelet count decreasedPleural effusionPneumonitisPruritusPulmonary edemaPurpuraRash acneiformRash maculo-papularRectal hemorrhageRectal painRenal and urinary disorders - Other, specifyResp, thoracic and mediastinal disorders - OtherRespiratory failureRestrictive cardiomyopathySalivary duct inflammationScrotal infectionScrotal painSeizureSepsisSinus bradycardiaSinus tachycardiaSinusitisSkin and subcutaneous tissue disorders - OtherSkin infectionSkin ulcerationSmall intestinal obstructionSoft tissue infectionSore throatStomach painStrokeSupraventricular tachycardiaSyncopeTesticular disorderThromboembolic eventThrombotic thrombocytopenic purpuraTooth infectionTumor lysis syndromeTyphlitisUpper gastrointestinal hemorrhageUpper respiratory infectionUrinary incontinenceUrinary tract infectionUrine output decreasedVaginal hemorrhageVascular access complicationVasovagal reactionVentricular tachycardiaVomitingWeight lossWhite blood cell decreasedWound infection
Arm I (Standard Dose Cytarabine, Daunorubicin Hydrochloride)114121122100001580580000191010111031003201010111111152001113002001121001301415243001110201123001115100070580180202132018110110010210021891111191007001411511000000011175110101111101311001160020501111002013148120500000321561
Arm II (High-dose Cytarabine, Idarubicin)016153172100101640161503111150112131170209000201061003201120017036216410001211816072020054100022120101211106181702012031124121000600100310122107010018310150112805002100010001680412112600138000002602021000201121000814010200211601500
Arm III (Vorinostat, High-dose Cytarabine, Idarubicin)119244201221011064121211100220030021040015110000040300381000008124205420110101311464130011210043000011601026182512429319921400130100101231477000111511510104100110111010012812150010010214001113412014100201130010923121612110421120

Complete Response Rate (Complete Response + Stringent Complete Response)

Defined by the International Myeloma Working Group (IMWG) criteria (NCT01653418)
Timeframe: Day +100

Interventionparticipants (Number)
V-BEAM + Stem Cell Infusion6

Time to Neutrophil Engraftment After V-BEAM.

Time to neutrophil engraftment is defined as duration between Day 0 to the first day of ANC > 0.5x109/L post transplant when it is sustained for more than three consecutive days. (NCT01653418)
Timeframe: Day +100

Interventiondays (Median)
V-BEAM + Stem Cell Infusion10

Treatment Related Mortality (TRM) of V-BEAM

(NCT01653418)
Timeframe: Day +100

Interventionparticipants (Number)
V-BEAM + Stem Cell Infusion2

Very Good Partial Response Rate (VGPR+nCR+sCR+CR)

Response will be assessed per the International Myeloma Working Group (IMWG) Response Criteria. (NCT01653418)
Timeframe: Day +100

Interventionparticipants (Number)
V-BEAM + Stem Cell Infusion8

Number of Participants With Overall Survival (OS)

OS is defined as the duration from the time of transplant to death or last follow-up. (NCT01653418)
Timeframe: Median follow-up of 6 months (range: 6-12 months)

Interventionparticipants (Number)
Expired Day +3Expired Day +18Alive
V-BEAM + Stem Cell Infusion118

Number of Participants With Progression-free Survival (PFS)

"PFS is defined as the duration from transplant to time of first progression, death, relapse after CR, or the date the patient was last known to be in remission.~Response will be assessed per the International Myeloma Working Group (IMWG) Response Criteria." (NCT01653418)
Timeframe: Median follow-up of 6 months (range: 6.0-12.0 months)

Interventionparticipants (Number)
No relapse/progressionRelapse/progression at 12 months
V-BEAM + Stem Cell Infusion71

Overall Response Rate (ORR)

"ORR includes Partial Response (PR) + Very Good Partial Response (VGPR) + Complete Response (CR)~Response will be assessed per the International Myeloma Working Group (IMWG) Response Criteria." (NCT01653418)
Timeframe: 3 months following Day +100 visit

Interventionparticipants (Number)
Partial responseVery good partial responseComplete response
V-BEAM + Stem Cell Infusion026

Time to Platelet Engraftment After V-BEAM.

Time to platelet engraftment is defined as the duration between Day 0 to the first day of platelet count sustained at > 20x109/L without transfusion. The median time to neutrophil and platelet engraftment will be reported. (NCT01653418)
Timeframe: Day +100

Interventiondays (Median)
More than 20 x 10^9/LMore than 50 x 10^9/L
V-BEAM + Stem Cell Infusion22.523

Toxicity of V-BEAM

"Graded per the NCI Common Terminology Criteria for Adverse Events (CTCAE) version 4.0.~Patients are evaluated from first receiving study treatment until a 30-day follow-up after the conclusion of treatment for adverse events not resulting in death. Adverse events resulting in death will be evaluated through Day +100.~This outcomes measures the common toxicities observed. Please refer to the Serious Adverse Event and Other Adverse Event sections of the results for further details." (NCT01653418)
Timeframe: 30 days after end of treatment / Day +100

Interventionparticipants (Number)
Neutropenic feverClostridium difficile colitisNeutropenic colitis without Clostridium difficileSepsisMucositis (grade 1-2)Mucositis (grade 3-4)Diarrhea (grade 3-4)Hepatic toxicity (grade 3-4)Peripheral neuropathy (grade 1-2)Toxic death
V-BEAM + Stem Cell Infusion103338210122

3-Year Overall Survival

Number of participants alive 3 years following treatment. Evaluations done every 3 months for 1 year and then every 6 months to check on the status of the disease. (NCT01538472)
Timeframe: 3 years

Interventionpercentage of participants (Number)
Y Zevalin + BEAM78

Overall Survival Median

Overall survival reported as number of days participants alive following treatment up to 5 years with annual follow up till disease progression. Evaluations done every 3 months for 1 year and then every 6 months for 5 years to check on the status of the disease, with long-term follow up as needed. (NCT01538472)
Timeframe: Participant followed from baseline treatment to 5 years, with study total period 8 years (study duration)

Interventiondays (Median)
Y Zevalin + BEAM1299

Number of Participants With Minimal Residual Disease (MRD)-Negative Complete Remission (CR) at The End of Induction Phase

MRD-negative CR was achieved when a participant met the criteria for both MRD negativity and CR. MRD-negativity: ≤0.01 % breakpoint cluster region-Abelson (BCR-ABL1/ABL1), or undetectable BCR-ABL1 transcripts in complementary deoxyribonucleic acid (cDNA) with ≥ 10,000 ABL1 transcripts. CR: meeting all the following for at least 4 weeks (that is no recurrence):1. No circulating blasts and less than (<) 5% blasts in the bone marrow (BM). 2. Normal maturation of all cellular components in the BM. 3. No extramedullary disease (central nervous system [CNS] involvement, lymphadenopathy, splenomegaly, skin/gum infiltration, testicular mass). 4. Absolute neutrophil count (ANC) > 1000 per microliter (/mcL) (or >1.0*10^9/L). 5. Platelets >100,000/mcL (or >100*10^9/L). (NCT03589326)
Timeframe: From Cycle 1 through Cycle 3 (approximately 3 months) (Cycle length = 28 days)

InterventionParticipants (Count of Participants)
Cohort A: Ponatinib 30 mg53
Cohort B: Imatinib 600 mg13

Event Free Survival

Observed length of time (days) after which patients remained free of recurrence or death. (NCT02440568)
Timeframe: 6 months

InterventionDays (Median)
Patients With Newly Diagnosed AML, Cohort 166
Patients With Newly Diagnosed AML, Cohort 290.5
Patients With Newly Diagnosed AML, Cohort 365.5
Patients With Newly Diagnosed AML, Cohort 433

Optimally Tolerated Dose

"The primary endpoint is determination of the optimally active and safe dose (OD) of Omacetaxine when added to the standard-of-care induction chemotherapy for AML and estimation of the efficacy and response rate.~OD will be defined as a dose level at which fewer than 30% of patients experience hematologic toxicity and greater than 50% of patients achieve a CR (50% is an accepted CR rate in AML when using a single induction)." (NCT02440568)
Timeframe: Within 50 days (duration of hematologic recovery)

InterventionParticipants treated with OD (Number)
Patients With Newly Diagnosed AML, Cohort 10
Patients With Newly Diagnosed AML, Cohort 20
Patients With Newly Diagnosed AML, Cohort 310
Patients With Newly Diagnosed AML, Cohort 40

Overall Participant Survival

Observed overall survival among participants (days) (NCT02440568)
Timeframe: 3 years

InterventionDays (Median)
Patients With Newly Diagnosed AML, Cohort 1726
Patients With Newly Diagnosed AML, Cohort 2162.5
Patients With Newly Diagnosed AML, Cohort 3792.5
Patients With Newly Diagnosed AML, Cohort 433

Progression Free Survival

Among the participants who achieved CR,CRi, progression free survival in number of days, i.e. the number of days participants who achieved CR/CRi remained alive and in a remission. (NCT02440568)
Timeframe: 3 years

InterventionDays (Median)
Patients With Newly Diagnosed AML, Cohort 1852.5
Patients With Newly Diagnosed AML, Cohort 2100
Patients With Newly Diagnosed AML, Cohort 31241.5
Patients With Newly Diagnosed AML, Cohort 41141

Time to Hematologic Recovery

Time to Absolute Neutrophil Count > 1.0 x 10e9/L and Platelet Count > 100 x 10e9/L, whichever was later, for those patients who achieved a remission and achieved these hematologic goals. (NCT02440568)
Timeframe: Within 6 months of last dose of Omacetaxine

InterventionDays (Median)
Patients With Newly Diagnosed AML, Cohort 125.5
Patients With Newly Diagnosed AML, Cohort 235
Patients With Newly Diagnosed AML, Cohort 333.5
Patients With Newly Diagnosed AML, Cohort 436

Toxicity: Describe by the Adverse Events as Assessed by the CTCAE Grading

Describe the adverse events associated with Omacetaxine when administered in combination with cytarabine and Idarubicin as induction therapy for AML, using CTCAE grading (NCT02440568)
Timeframe: Up to 6 months after last dose of Omacetaxine

InterventionTotal Adverse Events (Number)
Patients With Newly Diagnosed AML, Cohort 147
Patients With Newly Diagnosed AML, Cohort 288
Patients With Newly Diagnosed AML, Cohort 3219
Patients With Newly Diagnosed AML, Cohort 437

Engraftment of HLA Identical PBSC Allografts

"Number of patients who engrafted by Day 56 post allogeneic transplant. Failure to engraft is defined as the absence of detectable donor cells in the marrow. The rates and accompanying confidence intervals associated with failure of engraftment at day +56 will be calculated after every 5th patient is enrolled on the study. If the lower limit to the appropriate one-sided 80% confidence interval exceeds 25%, this will be considered sufficient evidence of an excess failure rate and the study will be stopped. For these purposes, all patients will be evaluated together (patients with chemosensitive and chemoresistant disease)." (NCT00005803)
Timeframe: Day 56

InterventionParticipants (Count of Participants)
Treatment (Tandem Transplantation)53

Non-Relapse Mortality

"The rates and accompanying confidence intervals associated with transplant-related mortality will be calculated after every 5th patient is enrolled on the study. If the lower limit to the appropriate one-sided 80% confidence interval exceeds 25%, this will be considered sufficient evidence of an excess failure rate and the study will be stopped. For these purposes, all patients will be evaluated together (patients with chemosensitive and chemoresistant disease)." (NCT00005803)
Timeframe: Day 100 post-non-myeloablative allografting following mobilization and high-dose chemotherapy with autografting

InterventionParticipants (Count of Participants)
Treatment (Tandem Transplantation)3

Overall Survival (OS)

Number of patients surviving by interval. Chemosensitive and chemoresistant subjects will be analyzed separately. (NCT00005803)
Timeframe: From the date of autologous transplant until the time of death, assessed up to 3 years

,,
InterventionParticipants (Count of Participants)
6 Months1 Year1.5 Years2 Years3 Years
Chemoresistant Group1410988
Chemosensitive Group3931272623
Unknown Chemosensitivity Group10000

Progression Free-survival (PFS)

Number of patients surviving without disease by interval. Chemosensitive and chemoresistant subjects will be analyzed separately. (NCT00005803)
Timeframe: From the date of autologous transplant until the time of progression, relapse, death, or the date the patient was last known to be in remission, assessed up to 3 years

,,
InterventionParticipants (Count of Participants)
6 Months1 Year1.5 Years2 Years3 Years
Chemoresistant Group128888
Chemosensitive Group3627252522
Unknown Chemosensitivity Group10000

Clinical Benefit as Measured by the Number of Patients Who Were Not RBC Transfusion-dependent Post-Baseline

Clinical benefit as measured by the number of patients who did not receive a RBC transfusion post-Baseline (NCT02666950)
Timeframe: Up to 17 months

InterventionParticipants (Count of Participants)
Arm A + Arm C (AZD1775 Combined With AraC OR AZD1775 Only)0

Complete Response Rate (CR or CRi) Per the National Comprehensive Cancer Network (NCCN) Guidelines or According to Specific Criteria From Expert Panels

Complete response rate will be evaluated over all courses of study treatment. The proportion of CR/CRi responses will be estimated by the number of CR/CRi responses divided by the total number of evaluable patients. (NCT02666950)
Timeframe: Up to 17 months

Interventionpercentage of patients (Number)
Arm A + Arm C (AZD1775 Combined With AraC OR AZD1775 Only)0

Overall Survival

Overall survival time is defined as the time from registration to death due to any cause. (NCT02666950)
Timeframe: From registration to death due to any cause, assessed up to 17 months

Interventionmonths (Median)
Arm A + Arm C (AZD1775 Combined With AraC OR AZD1775 Only)6

Percentage of Participants With Grade 3 or Higher Adverse Events Considered At Least Possibly Related to Treatment

"The maximum grade for each type of toxicity will be recorded for each patient, and frequency tables will be reviewed to determine toxicity patterns within patient groups. In addition, we will review all adverse event data that is graded as 3, 4, or 5 and classified as either unrelated or unlikely to be related to study treatment in the event of an actual relationship developing. The overall toxicity rates (percentages) for grade 3 or higher adverse events considered at least possibly related to treatment are reported below." (NCT02666950)
Timeframe: Up to 30 days post-treatment

Interventionpercentage of patients (Number)
Arm A + Arm C (AZD1775 Combined With AraC OR AZD1775 Only)100

Time to Progression, Defined as the Time From Registration to the Earliest Date of Documentation of Disease Progression

Time to progression (TTP) is defined to be the length of time from study registration to a) date of disease progression as defined by section 11.0 of the protocol, or b) last follow-up. If a patient dies without documentation of disease progression, the patient will be considered to have had a tumor progression at the time of death unless there is sufficient documented evidence to conclude no progression occurred prior to death. Time to progression curves were compared via the log-rank test. Progression is defined as a >25% increase in product of perpendicular diameters of contrast enhancement or mass or appearance of new lesions or unequivocal increase in size of contrast enhancement or increase in mass effect as agreed upon independently by primary physician and quality control physicians: appearance of new lesions compared to pretreatment MRI and/or CT scan. (NCT02666950)
Timeframe: Up to 17 months

Interventionmonths (Median)
Arm A + Arm C (AZD1775 Combined With AraC OR AZD1775 Only)3.9

Disease Free Survival

1 year disease free survival rate following adoptive transfer (NCT02046122)
Timeframe: one year following adoptive transfer

InterventionParticipants (Count of Participants)
Idarubicin + Cytarabine + DLI3

Number of Participants With Immune Recovery

Immune recovery determined by measurements of cytokine profiles, lymphocyte and natural killer (NK) enumeration and flow-based assays, measured prior to induction, prior to each round of consolidation, 8 weeks after the last cycle of consolidation, and every 3 months after treatment for up to 2 years (NCT02046122)
Timeframe: up to 2 years after completing therapy

InterventionParticipants (Count of Participants)
Idarubicin + Cytarabine + DLI3

Number of Subjects With Unacceptable Toxicity

"Unacceptable toxicity is defined as:~i. Grade III or IV acute GVHD (aGVHD) of the gut or liver or Grade IV aGVHD of the skin lasting > 7 days;~ii. Grade IV Common Terminology Criteria for Adverse Events (CTCAE) toxicity attributable to DLI (e.g. infusion reaction, as opposed to Grade IV CTCAE toxicity from chemotherapy) and lasting >7 days~iii. Treatment-related mortality (TRM)" (NCT02046122)
Timeframe: up to 8 weeks after last cell infusion

InterventionParticipants (Count of Participants)
Idarubicin + Cytarabine + DLI1

Overall Survival

Number of participants alive 2 years after completing adoptive transfer therapy. (NCT02046122)
Timeframe: 2 years after completing therapy

Interventionparticipants (Number)
Idarubicin + Cytarabine + DLI16

Percentage of Subjects With Acute GVHD

Grade III or IV aGVHD of the gut or liver or Grade IV aGVHD of the skin lasting > 7 days (NCT02046122)
Timeframe: 8 weeks after last cell infusion

Interventionpercentage of participants (Number)
Idarubicin + Cytarabine + DLI0

Percentage of Subjects With Unacceptable Toxicity

Grade IV CTCAE toxicity attributable to DLI (e.g. infusion reaction, as opposed to Grade IV CTCAE toxicity from chemotherapy) and lasting >7 days, Grade III or IV aGVHD of the gut or liver or Grade IV aGVHD of the skin lasting > 7 days, or death (NCT02046122)
Timeframe: 8 weeks after the last cell infusion

Interventionpercentage of participants (Number)
Idarubicin + Cytarabine + DLI5.88

Rate of Efficacy

Efficacy as measured by the number of subjects with a complete remission. Responses were evaluated according to standard criteria defined by the National Comprehensive Cancer Network Guidelines for AML (www.nccn.org). (NCT02046122)
Timeframe: 2 years after completing therapy

InterventionParticipants (Count of Participants)
Idarubicin + Cytarabine + DLI10

Number of Days to Hematopoietic Recovery

Hematopoietic recovery as measured by the date of the first of three consecutive laboratory values where the absolute neutrophil count (ANC) ≥ 500/μl , the date of the first of three consecutive laboratory values obtained on different days where the platelet count was > 20,000/μl without transfusion. (NCT02046122)
Timeframe: 2 years after completion of therapy

Interventiondays (Median)
Neutrophil recoveryPlatelet recovery
Idarubicin + Cytarabine + DLI2932

Frequency of Adverse Events, Graded According to NCI CTCAE v4.0

Maximum grade per participant of any AE. (NCT02029417)
Timeframe: Up to 30 days after last dose of study drugs

Interventionparticipants (Number)
Grade 1Grade 2Grade 3Grade 4
Treatment (Cytarabine, Omacetaxine Mepesuccinate, Decitabine)0002

Multiple Dose- Accumulation Ratio (Rac) of PF-04449913: Monotherapy Cohort

Rac was the observed accumulation ratio for AUCtau, determined as ratio of Day 21 AUCtau to Day -5 AUCtau. AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. (NCT02038777)
Timeframe: 0 to 24, 24 to 48, 48 to 72 hours post PF-04449913 dosing on Day -5 and Day 21 of Cycle 1

InterventionRatio (Geometric Mean)
Monotherapy Cohort: PF-04449913 25 mg1.893
Monotherapy Cohort: PF-04449913 50 mg2.076
Monotherapy Cohort: PF-04449913 100 mg1.752

Multiple Dose- AUCtau of Daunorubicinol: Combination Cohort 2

Daunorubicinol was a metabolite of daunorubicin. AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. (NCT02038777)
Timeframe: 0 to 24 hours post daunorubicin dosing on Day 3 of induction Cycle 1

InterventionNanogram*hour per milliliter (Geometric Mean)
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin2800

Multiple Dose- AUCtau of PF-04449913: Monotherapy Cohort

AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. AUClast = area under the curve from time zero to last quantifiable concentration. AUCinf = AUClast + (Clast/kel), where Clast = predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis, and where kel = terminal elimination phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 8, and 24 hours post PF-04449913 dosing Day 21 of Cycle 1

InterventionNanogram*hour per milliliter (Geometric Mean)
Monotherapy Cohort: PF-04449913 25 mg4561
Monotherapy Cohort: PF-04449913 50 mg9299
Monotherapy Cohort: PF-04449913 100 mg15480

Multiple Dose- Clearance (CL/F) of PF-04449913: Monotherapy Cohort

CL/F was defined as apparent total clearance of the drug from plasma after oral administration. CL/F of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. CL/F obtained after oral dose (apparent oral clearance) is influenced by the fraction of the dose absorbed. (NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 8, and 24 hours post PF-04449913 dosing Day 21 of Cycle 1

InterventionLiter per hour (Geometric Mean)
Monotherapy Cohort: PF-04449913 25 mg5.467
Monotherapy Cohort: PF-04449913 50 mg5.369
Monotherapy Cohort: PF-04449913 100 mg6.452

Multiple Dose- Steady State Accumulation Ratio (Rss) of PF-04449913: Monotherapy Cohort

Rss = Ratio of Day 21 AUCtau to Day -5 AUCinf. AUCinf = AUClast + (Clast/kel), where Clast = predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis, and where kel = terminal elimination phase rate constant calculated by a linear regression of the log-linear concentration-time curve. AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. (NCT02038777)
Timeframe: AUCtau: 0 to 24, 24 to 48, 48 to 72 hours post PF-04449913 dosing on Day 21 of Cycle 1; AUCinf: Pre-dose and 0.5, 1, 2, 4, 8, 24, 48, 72 hours post PF-04449913 dosing on Day -5 of Cycle 1

InterventionRatio (Geometric Mean)
Monotherapy Cohort: PF-04449913 25 mg1.355
Monotherapy Cohort: PF-04449913 50 mg1.017
Monotherapy Cohort: PF-04449913 100 mg1.176

Multiple Dose- T1/2 of Daunorubicin: Combination Cohort 2

Terminal plasma half-life is the time required to divide the plasma concentration by two after reaching pseudo-equilibrium (NCT02038777)
Timeframe: Pre-dose, 0.25 (mid-infusion), 0.5 (immediately prior to end of infusion), 1, 4, 6, 24 hours post daunorubicin dosing on Day 3 of at induction Cycle 1

InterventionHours (Mean)
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin7.297

Multiple Dose- Tmax of Daunorubicin: Combination Cohort 2

(NCT02038777)
Timeframe: Pre-dose, 0.25 (mid-infusion), 0.5 (immediately prior to end of infusion), 1, 4, 6, 24 hours post daunorubicin dosing on Day 3 of induction Cycle 1

InterventionHours (Median)
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin0.3585

Multiple Dose- Tmax of Daunorubicinol: Combination Cohort 2

Daunorubicinol was a metabolite of daunorubicin. (NCT02038777)
Timeframe: Pre-dose, 0.25 (mid-infusion), 0.5 (immediately prior to end of infusion), 1, 4, 6, 24 hours post daunorubicin dosing on Day 3 of induction Cycle 1

InterventionHours (Median)
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin0.3585

Multiple Dose- Tmax of PF-04449913: Monotherapy Cohort

(NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 8, and 24 hours post PF-04449913 dosing on Day 21 of Cycle 1

InterventionHours (Median)
Monotherapy Cohort: PF-04449913 25 mg3.970
Monotherapy Cohort: PF-04449913 50 mg4.000
Monotherapy Cohort: PF-04449913 100 mg1.950

Number of Participants With Clinically Significant Changes From Baseline in Vital Signs Abnormalities: Combination Cohort 1

Vital signs included blood pressure (sitting or supine) and heart rate. Clinically significant changes in vital signs were determined by the investigator's discretion. (NCT02038777)
Timeframe: Baseline up to maximum 486 days

InterventionParticipants (Count of Participants)
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg0

Number of Participants With Clinically Significant Changes From Baseline in Vital Signs Abnormalities: Combination Cohort 2

Vital signs included blood pressure (sitting or supine) and heart rate. Clinically significant changes in vital signs were determined by the investigator's discretion. (NCT02038777)
Timeframe: Baseline up to maximum 343 days

InterventionParticipants (Count of Participants)
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin0

Number of Participants With Clinically Significant Changes From Baseline in Vital Signs Abnormalities: Combination Cohort 3

Vital signs included blood pressure (sitting or supine) and heart rate. Clinically significant changes in vital signs were determined by the investigator's discretion. (NCT02038777)
Timeframe: Baseline up to maximum 841 days

InterventionParticipants (Count of Participants)
Combination Cohort 3: PF-04449913 100 mg + Azacitidine0

Number of Participants With Clinically Significant Changes From Baseline in Vital Signs Abnormalities: Monotherapy Cohort

Vital signs included blood pressure (sitting or supine) and heart rate. Clinically significant changes in vital signs were determined by the investigator's discretion. (NCT02038777)
Timeframe: For 25 mg: Baseline up to maximum 108 days; For 50 mg: Baseline up to maximum 151 days; For 100 mg: Baseline up to maximum 444 days

InterventionParticipants (Count of Participants)
Monotherapy Cohort: PF-04449913 25 mg0
Monotherapy Cohort: PF-04449913 50 mg0
Monotherapy Cohort: PF-04449913 100 mg0

Number of Participants With DLTs: Combination Cohort 1

Criteria: Grade >=3 non-hematologic toxicity (nht), except Grade >=3 infection, fever (including febrile neutropenia), infusion related AEs, electrolyte abnormalities, alanine aminotransferase (AT)/aspartate AT elevation that returned to Grade <=1/baseline within 7 days, allergic reactions possibly related to PF-04449913 that led to discontinuation of study drug; Prolonged myelosuppression lasted >42 days from point of detection = absolute neutrophil count < 500/microliter or platelet count <10*10^9/L with a normal bone marrow (<5% blasts and no evidence of disease or dysplasia); Inability to deliver >=80% of the planned study doses for all agents in a combination due to nht; Delay of >28 days in receiving next scheduled cycle due to persisting nht; Asymptomatic participant with Grade >=3 QTc prolongation required repeat testing, re-evaluation by qualified person, and correction of reversible causes for confirmation. Post-correction, if Grade 3 prolongation persisted, event was a DLT. (NCT02038777)
Timeframe: Day 1 up to Day 28 of Cycle 1 (28 days)

InterventionParticipants (Count of Participants)
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg0

Number of Participants With DLTs: Combination Cohort 2

Criteria: Grade >=3 non-hematologic toxicity (nht), except Grade >=3 infection, fever (including febrile neutropenia), infusion related AEs, electrolyte abnormalities, alanine aminotransferase (AT)/aspartate AT elevation that returned to Grade <=1/baseline within 7 days, allergic reactions possibly related to PF-04449913 that led to discontinuation of study drug; Prolonged myelosuppression lasted >42 days from point of detection = absolute neutrophil count < 500/microliter or platelet count <10*10^9/L with a normal bone marrow (<5% blasts and no evidence of disease or dysplasia); Inability to deliver >=80% of the planned study doses for all agents in a combination due to nht; Delay of >28 days in receiving next scheduled cycle due to persisting nht; Asymptomatic participant with Grade >=3 QTc prolongation required repeat testing, re-evaluation by qualified person, and correction of reversible causes for confirmation. Post-correction, if Grade 3 prolongation persisted, event was a DLT. (NCT02038777)
Timeframe: Day -3 up to anytime between Day 21 and Day 28 of first induction cycle (24 to 31 days)

InterventionParticipants (Count of Participants)
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin1

Number of Participants With DLTs: Combination Cohort 3

Criteria: Grade >=3 non-hematologic toxicity (nht), except Grade >=3 infection, fever (including febrile neutropenia), infusion related AEs, electrolyte abnormalities, alanine aminotransferase (AT)/aspartate AT elevation that returned to Grade <=1/baseline within 7 days, allergic reactions possibly related to PF-04449913 that led to discontinuation of study drug; Prolonged myelosuppression lasted >42 days from point of detection = absolute neutrophil count < 500/microliter or platelet count <10*10^9/L with a normal bone marrow (<5% blasts and no evidence of disease or dysplasia); Inability to deliver >=80% of the planned study doses for all agents in a combination due to nht; Delay of >28 days in receiving next scheduled cycle due to persisting nht; Asymptomatic participant with Grade >=3 QTc prolongation required repeat testing, re-evaluation by qualified person, and correction of reversible causes for confirmation. Post-correction, if Grade 3 prolongation persisted, event was a DLT. (NCT02038777)
Timeframe: Day 1 up to Day 28 of Cycle 1 (28 days)

InterventionParticipants (Count of Participants)
Combination Cohort 3: PF-04449913 100 mg + Azacitidine0

Number of Participants With Dose-limiting Toxicities (DLTs): Monotherapy Cohort

Criteria: Grade >=3 non-hematologic toxicity (nht), except Grade >=3 infection, fever (including febrile neutropenia), infusion related AEs, electrolyte abnormalities, alanine aminotransferase (AT)/aspartate AT elevation that returned to Grade <=1/baseline within 7 days, allergic reactions possibly related to PF-04449913 that led to discontinuation of study drug; Prolonged myelosuppression lasted >42 days from point of detection = absolute neutrophil count < 500/microliter or platelet count <10*10^9/L with a normal bone marrow (<5% blasts and no evidence of disease or dysplasia); Inability to deliver >=80% of the planned study doses for all agents in a combination due to nht; Delay of >28 days in receiving next scheduled cycle due to persisting nht; Asymptomatic participant with Grade >=3 QTc prolongation required repeat testing, re-evaluation by qualified person, and correction of reversible causes for confirmation. Post-correction, if Grade 3 prolongation persisted, event was a DLT. (NCT02038777)
Timeframe: Day -5 up to Day 28 of Cycle 1 (33 days)

InterventionParticipants (Count of Participants)
Monotherapy Cohort: PF-04449913 25 mg0
Monotherapy Cohort: PF-04449913 50 mg0
Monotherapy Cohort: PF-04449913 100 mg0

Overall Survival: Combination Cohort 1

Overall survival was defined as the time from the date of first dose of study drug to the date of death due to any cause. Participants last known to be alive were censored at the date of last contact. (NCT02038777)
Timeframe: First dose of study drug up to death or date of last contact (maximum up to 514 days)

InterventionMonths (Median)
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg11.8

Percentage of Participants Achieving Disease Modifying Response (DMR): Expansion Cohort

DMR included complete remission (CR), CR with incomplete blood count recovery (Cri), morphologic leukemia-free state (MLFS), marrow CR (mCR) and partial remission (PR). CR: >=11 gram per deciliter (g/dL) hemoglobin (Hgb), >=1*10^9 neutrophils (L), >=100*10^9 platelets (L), 0% blasts, <=5% bone marrow blasts (BMB), normal maturation of all cell lines, if had persistent dysplasia. . CRi: <1000 neutrophils (mcL), <100000 platelets (mcL), <5% BMB, either neutrophils or platelets not recovered, no extramedullary disease (EMD). MLFS: 1000 neutrophils (mcL) and <100000 platelets (mcL), <5% BMB, neutrophils and platelets not recovered, flow cytometry negative, no EMD. PR: >=1000 neutrophils (mcL), >=100000 platelets (mcL), decrease to 5-25 and >=50% decrease from start, Blasts <=5% if Auer rod positive. mCR: hematologic improvement (HI) response, <=5% and decreased by >=50% BMB. PR: decrease by >=50% but still >5% BMB. (NCT02038777)
Timeframe: Baseline up to maximum 736 days

InterventionPercentage of participants (Number)
Expansion Cohort (Unfit Participants): PF-04449913 100 mg+ LDAC 20 mg46.7

Single Dose- Clearance (CL/F) of PF-04449913: Monotherapy Cohort

CL/F was defined as apparent total clearance of the drug from plasma after oral administration. CL/F of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. CL/F obtained after oral dose (apparent oral clearance) is influenced by the fraction of the dose absorbed. (NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 8, 24, 48, 72 hours post PF-04449913 dosing on Day -5 of Cycle 1

InterventionLiter per hour (Geometric Mean)
Monotherapy Cohort: PF-04449913 25 mg7.415
Monotherapy Cohort: PF-04449913 50 mg5.210
Monotherapy Cohort: PF-04449913 100 mg7.599

Single Dose- Maximum Observed Plasma Concentration (Cmax) of PF-04449913: Monotherapy Cohort

(NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 8, 24, 48, 72 hours post PF-04449913 dosing on Day -5 of Cycle 1

Interventionnanogram per milliliter (Geometric Mean)
Monotherapy Cohort: PF-04449913 25 mg281.5
Monotherapy Cohort: PF-04449913 50 mg321.1
Monotherapy Cohort: PF-04449913 100 mg1019

Single Dose- Terminal Plasma Half-life (T1/2) of PF-04449913: Monotherapy Cohort

Terminal plasma half-life is the time required to divide the plasma concentration by two after reaching pseudo-equilibrium. (NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 8, 24, 48, 72 hours post PF-04449913 dosing on Day -5 of Cycle 1

InterventionHours (Mean)
Monotherapy Cohort: PF-04449913 25 mg17.83
Monotherapy Cohort: PF-04449913 50 mg30.70
Monotherapy Cohort: PF-04449913 100 mg18.67

Single Dose- Time to Reach Maximum Observed Plasma Concentration (Tmax) of PF-04449913: Monotherapy Cohort

(NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 8, 24, 48, 72 hours post PF-04449913 dosing on Day -5 of Cycle 1

InterventionHours (Median)
Monotherapy Cohort: PF-04449913 25 mg1.970
Monotherapy Cohort: PF-04449913 50 mg3.955
Monotherapy Cohort: PF-04449913 100 mg1.950

Single Dose- Volume of Distribution (Vz/F) of PF-04449913: Monotherapy Cohort

Vz/F was defined as the theoretical volume in which the total amount of drug would need to be uniformly distributed to produce the desired blood concentration of a drug. (NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 8, 24, 48, 72 hours post PF-04449913 dosing on Day -5 of Cycle 1

InterventionLiter (Geometric Mean)
Monotherapy Cohort: PF-04449913 25 mg190.5
Monotherapy Cohort: PF-04449913 50 mg227.8
Monotherapy Cohort: PF-04449913 100 mg201.5

Duration of Complete Remission (CR) or CR With Incomplete Blood Count Recovery (CRi) and DMR Response: Combination Cohort 1

Duration of response was the time from the date of first documentation of a CR/CRi and DMR to the date of first documentation of relapse after CR/CRi and DMR or death due to any cause. Duration of response data was censored on the date of the last adequate response assessment for patients who do not have an event (relapse or death). DMR included CR, CRi, MLFS, mCR and PR. CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, bone marrow blasts (BMB) <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. MLFS: neutrophils (mcL) 1000 and platelets (mcL) <100000, BMB <5%. PR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, BMB decrease to 5-25 and >=50% decrease from start. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 486 days)

InterventionMonths (Median)
Duration of CR/CRiDuration of DMR
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg13.915.3

Duration of Complete Remission (CR) or CR With Incomplete Blood Count Recovery (CRi) and DMR Response: Combination Cohort 3

Duration of response was the time from the date of first documentation of a CR/CRi and DMR to the date of first documentation of relapse after CR/CRi and DMR or death due to any cause. Duration of response data was censored on the date of the last adequate response assessment for patients who do not have an event (relapse or death). DMR included CR, CRi, MLFS, mCR and PR. CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, bone marrow blasts (BMB) <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. MLFS: neutrophils (mcL) 1000 and platelets (mcL) <100000, BMB <5%. PR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, BMB decrease to 5-25 and >=50% decrease from start. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 841 days)

InterventionMonths (Median)
Duration of CR/CRiDuration of DMR
Combination Cohort 3: PF-04449913 100 mg + Azacitidine6.66.6

Multiple Dose Cmax, Minimum Observed Plasma Concentration (Cmin), Average Observed Plasma Concentration (Cavg), Trough Plasma Concentration (Ctrough) of PF-04449913: Monotherapy Cohort

Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval, dosing interval was of 24 hours. Ctrough = Pre-dose concentration, observed directly from data. (NCT02038777)
Timeframe: Cmax, Cmin: Pre-dose and 0.5, 1, 2, 4, 8, and 24 hours post PF-04449913 dosing on Day 21 of Cycle 1; Cavg: 0 to 24, 24 to 48, 48 to 72 hours post PF-04449913 dosing on Day 21 of Cycle 1; Ctrough: Pre-dose on Day 21 of Cycle 1

,,
InterventionNanogram per milliliter (Geometric Mean)
CmaxCminCavgCtrough
Monotherapy Cohort: PF-04449913 100 mg1330333.9645.8342.9
Monotherapy Cohort: PF-04449913 25 mg356.984.94190.587.87
Monotherapy Cohort: PF-04449913 50 mg542.2237.2388.1240.0

Multiple Dose- AUCinf and AUCtau of Cytarabine: Combination Cohort 1

LDAC= low dose ara-cytarabine/low dose cytarabine. AUCinf = AUClast + (Clast/kel), where Clast = predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis, and where kel = terminal elimination phase rate constant calculated by a linear regression of the log-linear concentration-time curve. AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 12 hours. (NCT02038777)
Timeframe: AUCinf: Pre-dose and 0.25, 0.5, 1, 2, 4 and 6 hours post LDAC dosing on Day 2 and Day 10 of Cycle 1; AUCtau: 0 to 12 hours post LDAC dosing on Day 2 and Day 10 of Cycle

InterventionNanogram*hour per milliliter (Geometric Mean)
AUCinf: Cycle 1/Day 2AUCtau: Cycle 1/Day 2AUCinf: Cycle 1/Day 10AUCtau: Cycle 1/Day 10
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg78.3777.9797.3497.58

Multiple Dose- AUCinf and AUCtau of Daunorubicin: Combination Cohort 2

AUCinf = AUClast + (Clast/kel), where Clast = predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis, and where kel = terminal elimination phase rate constant calculated by a linear regression of the log-linear concentration-time curve. AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. (NCT02038777)
Timeframe: AUCinf: Pre-dose, 0.25 (mid-infusion), 0.5 (immediately prior to end of infusion), 1, 4, 6, 24 hours post daunorubicin dosing on Day 3 of induction Cycle 1; AUCtau: 0 to 24 hours post daunorubicin dosing on Day 3 of induction Cycle 1

InterventionNanogram*hour per milliliter (Geometric Mean)
AUCinfAUCtau
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin770.4741.6

Multiple Dose- AUCtau and AUCinf of Azacitidine: Combination Cohort 3

AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. AUCinf = AUClast + (Clast/kel), where Clast = predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis, and where kel = terminal elimination phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT02038777)
Timeframe: AUCinf: 0.25, 0.5, 1, 2, and 6 hours post azacitidine dosing at Day 1 of Cycle 1 and pre-dose, 0.25, 0.5, 1, 2, and 6 hours post azacitidine dosing at 7 of Cycle 1; AUCtau: 0 to 24 hours post azacitidine dosing on Day 1 and 7 of Cycle 1

InterventionNanogram*hour per milliliter (Geometric Mean)
AUCtau: Cycle 1/Day 1AUCinf: Cycle 1/Day 1AUCtau: Cycle 1/Day 7AUCinf: Cycle 1/Day 7
Combination Cohort 3: PF-04449913 100 mg + Azacitidine1200910.912411200

Multiple Dose- AUCtau of PF-04449913: Combination Cohort 1

AUCtau was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. (NCT02038777)
Timeframe: 0 to 24 hours post PF-04449913 dose on Day 10 and Day 21 of Cycle 1

InterventionNanogram*hour per milliliter (Geometric Mean)
Cycle 1/Day 10Cycle 1/Day 21
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg1556016070

Multiple Dose- AUCtau of PF-04449913: Combination Cohort 2

AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. (NCT02038777)
Timeframe: Pre-dose, 0.5, 1, 6, and 24 hours post PF-04449913 dosing on Day 3 of induction Cycle 1 and pre-dose, 0.5, 1, 4, 6, and 24 hours post PF-04449913 dosing on Day 10 of induction Cycle 1

InterventionNanogram*hour per milliliter (Geometric Mean)
Cycle 1/Day 3Cycle 1/Day 10
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin1563018120

Multiple Dose- AUCtau of PF-04449913: Combination Cohort 3

AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. (NCT02038777)
Timeframe: 0 to 24 hours post PF-04449913 dosing on Day 7 and Day 21 of Cycle 1

InterventionNanogram*hour per milliliter (Geometric Mean)
Cycle 1/Day 7Cycle 1/Day 21
Combination Cohort 3: PF-04449913 100 mg + Azacitidine2001016860

Multiple Dose- CL/F of PF-04449913: Combination Cohort 1

CL/F was defined as apparent total clearance of the drug from plasma after oral administration. CL/F of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. CL/F obtained after oral dose (apparent oral clearance) is influenced by the fraction of the dose absorbed. (NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 6 and 24 hours post PF-04449913 dose on Day 10 and Day 21 of Cycle 1

InterventionLiter per hour (Geometric Mean)
Cycle 1/Day 10Cycle 1/Day 21
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg6.4286.223

Multiple Dose- CL/F of PF-04449913: Combination Cohort 2

CL/F was defined as apparent total clearance of the drug from plasma after oral administration. CL/F of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. CL/F obtained after oral dose (apparent oral clearance) is influenced by the fraction of the dose absorbed. (NCT02038777)
Timeframe: Pre-dose, 0.5, 1, 6, and 24 hours post PF-04449913 dosing on Day 3 of induction Cycle 1 and pre-dose, 0.5, 1, 4, 6, and 24 hours post PF-04449913 dosing on Day 10 of induction Cycle 1

InterventionLiter per hour (Geometric Mean)
Cycle 1/Day 3Cycle 1/Day 10
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin6.4015.523

Multiple Dose- CL/F of PF-04449913: Combination Cohort 3

CL/F was defined as apparent total clearance of the drug from plasma after oral administration. CL/F of a drug is a measure of the rate at which a drug is metabolized or eliminated by normal biological processes. CL/F obtained after oral dose (apparent oral clearance) is influenced by the fraction of the dose absorbed. (NCT02038777)
Timeframe: Pre-dose, 0.25, 1, 4, 6, 24 hours post PF-04449913 dosing on Day 7 and Day 21 of Cycle 1

InterventionLiter per hour (Geometric Mean)
Cycle 1/Day 7Cycle 1/Day 21
Combination Cohort 3: PF-04449913 100 mg + Azacitidine4.9995.936

Multiple Dose- Cmax, Cmin, and Ctrough of Ara-uridine: Combination Cohort 1

Ara-uridine was a metabolite of cytarabine. Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval, dosing interval was of 12 hours. Ctrough = Pre-dose concentration, observed directly from data. Ara-uridine was a metabolite of cytarabine. (NCT02038777)
Timeframe: Cmax, Cmin: Pre-dose and 0.25, 0.5, 1, 2, 4 and 6 hours post LDAC dosing on Day 2 and Day 10 of Cycle 1; Cavg: 0 to 12 hours post LDAC dosing on Day 2 and Day 10 of Cycle 1; Ctrough: Pre-LDAC dosing on Day 2 and Day 10 of Cycle 1

InterventionNanogram per milliliter (Geometric Mean)
Cmax: Cycle 1/Day 2Cmin: Cycle 1/Day 2Ctrough: Cycle 1/Day 2Cmax: Cycle 1/Day 10Cmin: Cycle 1/Day 10Ctrough: Cycle 1/Day 10
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg371.6141.9141.9454.3201.7201.7

Multiple Dose- Cmax, Cmin, Cavg, and Ctrough of Azacitidine: Combination Cohort 3

Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval of 24 hours. Ctrough = Pre-dose concentration, observed directly from data. (NCT02038777)
Timeframe: Cmax: 0.25, 0.5, 1, 2, 6 hrs post azacitidine dose on Day 1/Cycle 1;Cmin: Pre-dose, 0.25, 0.5, 1, 2, 6 hrs post azacitidine dose on Day 7/Cycle 1;Cavg: 0 to 24 hrs post azacitidine dose on Day 7/Cycle 1;Ctrough:Pre azacitidine dose on Day 7/Cycle 1

InterventionNanogram per milliliter (Geometric Mean)
Cmax: Cycle 1/Day 1Cmax: Cycle 1/Day 7Cmin: Cycle 1/Day 7Cavg: Cycle 1/Day 7Ctrough: Cycle 1/Day 7
Combination Cohort 3: PF-04449913 100 mg + Azacitidine18031717NA51.60NA

Multiple Dose- Cmax, Cmin, Cavg, and Ctrough of Cytarabine: Combination Cohort 1

LDAC= low dose ara-cytarabine/low dose cytarabine. Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval, dosing interval was of 24 hours, dosing interval was of 12 hours. Ctrough = Pre-dose concentration, observed directly from data. (NCT02038777)
Timeframe: Cmax, Cmin: Pre-dose and 0.25, 0.5, 1, 2, 4 and 6 hours post LDAC dosing on Day 2 and Day 10 of Cycle 1; Cavg: 0 to 12 hours post LDAC dosing on Day 2 and Day 10 of Cycle 1; Ctrough: Pre-LDAC dose on Day 2 and Day 10 of Cycle 1

InterventionNanogram per milliliter (Geometric Mean)
Cmax: Cycle 1/Day 2Cmin: Cycle 1/Day 2Cavg: Cycle 1/Day 2Ctrough: Cycle 1/Day 2Cmax: Cycle 1/Day 10Cmin: Cycle 1/Day 10Cavg: Cycle 1/Day 10Ctrough: Cycle 1/Day 10
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg82.88NA6.511NA106.70.59038.1350.5903

Multiple Dose- Cmax, Cmin, Cavg, and Ctrough of Daunorubicin: Combination Cohort 2

Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval, dosing interval was of 24 hours. Ctrough = Pre-dose concentration, observed directly from data. (NCT02038777)
Timeframe: Cmax, Cmin: Pre-dose, 0.25 (mid-infusion), 0.5 (immediately prior to end of infusion), 1, 4, 6, 24 hours post daunorubicin dosing on Day 3 of induction Cycle (IC) 1; Cavg: 0 to 24 hours post dose on Day 3 of IC 1; Ctrough: Pre-dose on Day 3 of IC 1

InterventionNanogram per milliliter (Geometric Mean)
CmaxCminCavgCtrough
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin942.82.58930.892.673

Multiple Dose- Cmax, Cmin, Cavg, and Ctrough of Daunorubicinol: Combination Cohort 2

Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval of 24 hours. Ctrough = Pre-dose concentration, observed directly from data. Daunorubicinol was a metabolite of daunorubicin. (NCT02038777)
Timeframe: Cmax, Cmin: Pre-dose, 0.25 (mid-infusion), 0.5 (immediately prior to end of infusion), 1, 4, 6, 24 hours post daunorubicin dosing on Day 3 of induction Cycle (IC) 1; Cavg: 0 to 24 hours post dose on Day 3 of IC 1; Ctrough: Pre-dose on Day 3 of IC 1

InterventionNanogram per milliliter (Geometric Mean)
CmaxCminCavgCtrough
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin244.466.38116.766.53

Multiple Dose- Cmax, Cmin, Cavg, and Ctrough of PF-04449913: Combination Cohort 1

Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval, dosing interval was of 24 hours. Ctrough = Pre-dose concentration, observed directly from data. (NCT02038777)
Timeframe: Cmax, Cmin: Pre-dose and 0.5, 1, 2, 4, 6 and 24 hours post PF-04449913 dose on Day 10 and 21 of Cycle 1; Cavg: 0 to 24 hours post PF-04449913 dose on Day 10 and 21 of Cycle; Ctrough: Pre-dose on Day 10 and 21 of Cycle 1

InterventionNanogram per milliliter (Geometric Mean)
Cmax: Cycle 1/Day 10Cmin: Cycle 1/Day 10Cavg: Cycle 1/Day 10Ctrough: Cycle 1/Day 10Cmax: Cycle 1/Day 21Cmin: Cycle 1/Day 21Cavg: Cycle 1/Day 21Ctrough: Cycle 1/Day 21
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg1172317.6648.3330.71317341.6670.5376.2

Multiple Dose- Cmax, Cmin, Cavg, and Ctrough of PF-04449913: Combination Cohort 2

Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval, dosing interval was of 24 hours. Ctrough = Pre-dose concentration, observed directly from data. (NCT02038777)
Timeframe: Cmax, Cmin: Pre-dose, 0.5, 1, 6, and 24 hrs post dose on Day 3, 10 of induction Cycle (IC) 1 and 4 hrs post dose on Day 10 of IC 1; Cavg: 0 to 24 hrs post dose on Day 3 and Day 10 of IC 1; Ctrough: Pre dose on Day 3 and Day 10 of IC 1 (PF-04449913 Dose)

InterventionNanogram per milliliter (Geometric Mean)
Cmax: Cycle 1/Day 3Cmin: Cycle 1/Day 3Cavg: Cycle 1/Day 3Ctrough: Cycle 1/Day 3Cmax: Cycle 1/Day 10Cmin: Cycle 1/Day 10Cavg: Cycle 1/Day 10Ctrough: Cycle 1/Day 10
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin1047318.2650.9354.01181356.1755.2359.5

Multiple Dose- Cmax, Cmin, Cavg, and Ctrough of PF-04449913: Combination Cohort 3

Cmax = Maximum plasma concentration, observed directly from data. Cmin = Minimum plasma concentration observed directly from data. Cavg = Average plasma concentration over the dosing interval, dosing interval was of 24 hours. Ctrough = Pre-dose concentration, observed directly from data. (NCT02038777)
Timeframe: Cmax, Cmin: Pre-dose, 0.25, 1, 4, 6, 24 hours post PF-04449913 dosing on Day 7 and Day 21 of Cycle 1; Cavg: 0 to 24 hors post PF-04449913 dosing on Day 7 and Day 21 of Cycle 1; Ctrough: Pre PF-04449913 dosing on Day 7 and Day 21 of Cycle 1

InterventionNanogram per milliliter (Geometric Mean)
Cmax: Cycle 1/Day 7Cmin: Cycle 1/Day 7Cavg: Cycle 1/Day 7Ctrough: Cycle 1/Day 7Cmax: Cycle 1/Day 21Cmin: Cycle 1/Day 21Cavg: Cycle 1/Day 21Ctrough: Cycle 1/Day 21
Combination Cohort 3: PF-04449913 100 mg + Azacitidine1387414.1834.6526.31218323.2701.9347.5

Multiple Dose- T1/2 of Cytarabine: Combination Cohort 1

LDAC= low dose ara-cytarabine/low dose cytarabine. Terminal plasma half-life is the time required to divide the plasma concentration by two after reaching pseudo-equilibrium. (NCT02038777)
Timeframe: Pre-dose and 0.25, 0.5, 1, 2, 4 and 6 hours post LDAC dosing on Day 2 and Day 10 of Cycle 1

InterventionHours (Mean)
Cycle 1/Day 2Cycle 1/Day 10
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg1.0270.8618

Multiple Dose- Tmax of Ara-uridine: Combination Cohort 1

Ara-uridine was a metabolite of cytarabine. (NCT02038777)
Timeframe: Pre-dose and 0.25, 0.5, 1, 2, 4 and 6 hours post LDAC dosing on Day 2 and Day 10 of Cycle 1

InterventionHours (Median)
Cycle 1/Day 2Cycle 1/Day 10
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg1.5152.000

Multiple Dose- Tmax of Azacitidine: Combination Cohort 3

(NCT02038777)
Timeframe: 0.25, 0.5, 1, 2, and 6 hours post azacitidine dosing on Day 1 of Cycle 1 and pre-dose, 0.25, 0.5, 1, 2, and 6 hours post azacitidine dosing on Day 7 of Cycle 1

InterventionHours (Median)
Day 1Day 7
Combination Cohort 3: PF-04449913 100 mg + Azacitidine0.25000.2500

Multiple Dose- Tmax of Cytarabine: Combination Cohort 1

LDAC= low dose ara-cytarabine/low dose cytarabine. (NCT02038777)
Timeframe: Pre-dose and 0.25, 0.5, 1, 2, 4 and 6 hours post LDAC dosing on Day 2 and Day 10 of Cycle 1

InterventionHours (Median)
Cycle 1/Day 2Cycle 1/Day 10
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg0.25000.2500

Multiple Dose- Tmax of PF-04449913: Combination Cohort 1

(NCT02038777)
Timeframe: Pre-dose and 0.5, 1, 2, 4, 6 and 24 hours post PF-04449913 dose on Day 10 and Day 21 of Cycle 1

InterventionHours (Median)
Cycle 1/Day 10Cycle 1/Day 21
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg3.9501.935

Multiple Dose- Tmax of PF-04449913: Combination Cohort 2

(NCT02038777)
Timeframe: Pre-dose, 0.5, 1, 6, and 24 hours post PF-04449913 dosing on Day 3 of Induction Cycle 1 and pre-dose, 0.5, 1, 4, 6, and 24 hours post PF-04449913 dosing on Day 10 of Induction Cycle 1

InterventionHours (Median)
Cycle 1/Day 3Cycle 1/Day 10
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin5.9505.065

Multiple Dose- Tmax of PF-04449913: Combination Cohort 3

(NCT02038777)
Timeframe: Pre-dose, 0.25, 1, 4, 6, 24 hours post PF-04449913 dosing on Day 7 and Day 21 of Cycle 1

InterventionHours (Median)
Cycle 1/Day 7Cycle 1/Day 21
Combination Cohort 3: PF-04449913 100 mg + Azacitidine4.0002.500

Number of Participants With Best Response: Combination Cohort 1

Best response observed for: CR, Cri, MLFS, PR, PRi, CRc, CRm. Response criteria for participants with AML- CR: neutrophils [mcL] >=1000, platelets(pt)[mcL] >=10^5, BMB <5%. CRi: neutrophils (mcL) <1000 or pt (mcL) <100000, BMB <5%. MLFS: neutrophils (mcL) 1000 and pt (mcL) <10^5, BMB <5%. PR: neutrophils (mcL) >=1000, pt (mcL) >=100000, decrease to 5-25 and >=50% decrease from start. PRi: neutrophils (mcL) <1000 or pt (mcL) <10^5, BMB decrease to 5-25 and >=50% decrease from start. Minor Response: BMB >=25% decrease from start. CRc: neutrophils (mcL) >1,000, pt (mcL) >10^6, BMB <5%. CRm: neutrophils (mcL) >1,000, pt (mcL) >100,000, BMB <5%. For participants with myelodysplastic syndrome (MDS), DMR- CR: >=11 Hgb (g/dL), >=1*10^9 neutrophils(L), >=100*10^9 pt(L), 0% blasts, <=5% BMB. mCR: HI response, <=5% and decreased by >=50% BMB. PR: decrease by >=50% but still >5% BMB. Only those responses which had at least 1 participant were reported. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 486 days)

InterventionParticipants (Count of Participants)
Morphologic CR: AMLStable disease: AMLTreatment failure: AMLStable disease: MDS
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg1122

Number of Participants With Best Response: Combination Cohort 2

Best response observed for: CR, Cri, MLFS, PR, PRi, CRc, CRm. Response criteria for AML- CR:neutrophils(nt)[mcL] >=1000, platelets (mcL) >=100000, BMB <5%. CRi: nt(mcL) <1000 or platelets (mcL) <100000, BMB <5%. MLFS: nt(mcL) 1000 and platelets (mcL) <100000, BMB <5%. PR: nt(mcL) >=1000, platelets (mcL) >=100000, decrease to 5-25 and >=50% decrease from start. PRi: nt(mcL) <1000 or platelets (mcL) <100000, BMB decrease to 5-25 and >=50% decrease from start. Minor Response: BMB >=25% decrease from start. Cytogenetic CR (CRc): nt(mcL) >1,000, platelets (mcL) >100,000, BMB <5%. CRm: nt(mcL) >1,000, platelets (mcL) >100,000, BMB <5%. For participants with myelodysplastic syndrome (MDS), DMR was defined as - CR: >=11 Hgb (g/dL), >=1*10^9 nt(L), >=100*10^9 platelets (L), 0% blasts, <=5% BMB. mCR: HI response, <=5% and decreased by >=50% BMB. PR: decrease by >=50% but still >5% BMB. Only those responses which had at least 1 participant were reported. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 343 days)

InterventionParticipants (Count of Participants)
Morphologic CR: AMLMorphologic CRi: AMLMLFs: AMLPR: AMLMR: AMLTreatment failure: AMLRelapse: AMLIndeterminate: AML
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin32212111

Number of Participants With Best Response: Combination Cohort 3

Best response observed for: CR, Cri, MLFS, PR, PRi, CRc, CRm. Response criteria for participants with AML- CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, BMB <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. MLFS: neutrophils (mcL) 1000 and platelets (mcL) <100000, BMB <5%. PR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, decrease to 5-25 and >=50% decrease from start. PRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB decrease to 5-25 and >=50% decrease from start. Minor Response: BMB >=25% decrease from start. CRc: neutrophils (mcL) >1,000, platelets (mcL) >100,000, BMB <5%. CRm: neutrophils (mcL) >1,000, platelets (mcL) >100,000, BMB <5%. Only those responses which had at least 1 participant were reported. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 841 days)

InterventionParticipants (Count of Participants)
Morphologic CRPRiTreatment failure
Combination Cohort 3: PF-04449913 100 mg + Azacitidine312

Number of Participants With Best Response: Monotherapy Cohort

Best response observed for: CR, Cri, MLFS, PR, PRi, CytogeneticCR(CRc), MolecularCR(CRm). For AML-CR:neutrophils(nt) [mcL]>=1000, platelets(pt)[mcL]>=10^5, BMB<5%. CRi:nt(mcL)<1000/pt(mcL)<10^5, BMB<5%. MLFS:nt(mcL)1000 and pt(mcL)<10^5, BMB<5%. PR:nt(mcL)>=1000, pt(mcL)>=10^5, decrease to 5-25 and >=50% decrease from start. PRi: nt<1000, <10^5. CRc: nt(mcL)>1,000, pt(mcL)>10^5, BMB<5%. CRm: nt(mcL)>1,000, pt(uL)>10^5, BMB<5%. For myelodysplasia-CR: hemoglobin(Hgb)[gram per deciliter{g/dL}]>=11, nt(L)>=1*10^9, pt(L)>=100*10^9, blasts0%, BMB<=5%. mCR:<=5% and decreased by >=50% BMB. PR:decrease by>=50% with >5% BMB, CRc: disappearance of chromosomal abnormality, no new appearance, PRc:>=50% reduced chromosomal abnormality. For myleofibrosis-CR: hgb(g/L)>=110, nt(L)>=1*10^9, pt(L)>=100*10^9, All <=ULN, BMB <=5%. PR: hgb>=110, nt(L)>=1*10^9, pt(L)>=100*10^9. CML- PR: 1-35% Philadelphia chromosome(PC) positive(+) cells, CR:0% PC+ cells. Responses with at least 1 participant were reported. (NCT02038777)
Timeframe: Day 1 up to End of Treatment (25 mg: maximum up to 108 days; 50 mg: maximum up to 151 days; 100 mg: maximum up to 444 days)

,,
InterventionParticipants (Count of Participants)
Morphologic CR: AMLMorphologic CRi: AMLMLFs: AMLStable disease: AMLTreatment failure: AMLMarrow complete remission: MDSStable disease: MDSDisease progression: MDSTreatment failure: MDSStable disease: CMLDisease progression: CML
Monotherapy Cohort: PF-04449913 100 mg11123110000
Monotherapy Cohort: PF-04449913 25 mg00011010100
Monotherapy Cohort: PF-04449913 50 mg00011011011

Number of Participants With TEAEs, Serious TEAEs, Treatment Related TEAEs, Grade 3 or 4 TEAEs Based on NCI CTCAE v4.0: Combination Cohort 1

AE: any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. Serious AE: any untoward medical occurrence at any dose that resulted in death; was life threatening; required inpatient hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity; resulted in congenital anomaly/birth defect. TEAEs: events absent before treatment or that worsened relative to pretreatment state. Treatment-related TEAE: any untoward medical occurrence attributed to study drug in a participant who received study drug. Relatedness to drug was assessed by the Investigator. NCI-CTCAE Grade: Grade 3=severe or medically significant but not immediately life-threatening, hospitalization or prolongation of existing hospitalization indicated, disabling, limiting self-care ADL; Grade 4= life-threatening consequence, urgent intervention indicated. (NCT02038777)
Timeframe: Day 1 up to 28 days after last dose of study drug (maximum up to 514 days)

InterventionParticipants (Count of Participants)
TEAEsSerious TEAEsTreatment Related TEAEsGrade 3 or 4 TEAEs
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg6164

Number of Participants With TEAEs, Serious TEAEs, Treatment Related TEAEs, Grade 3 or 4 TEAEs Based on NCI CTCAE v4.0: Combination Cohort 2

AE: any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. Serious AE: any untoward medical occurrence at any dose that resulted in death; was life threatening; required inpatient hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity; resulted in congenital anomaly/birth defect. TEAEs: events absent before treatment or that worsened relative to pretreatment state. Treatment-related TEAE: any untoward medical occurrence attributed to study drug in a participant who received study drug. Relatedness to drug was assessed by the Investigator. NCI-CTCAE Grade: Grade 3=severe or medically significant but not immediately life-threatening, hospitalization or prolongation of existing hospitalization indicated, disabling, limiting self-care ADL; Grade 4= life-threatening consequence, urgent intervention indicated. (NCT02038777)
Timeframe: Day 1 up to 28 days after last dose of study drug (maximum up to 371 days)

InterventionParticipants (Count of Participants)
TEAEsSerious TEAEsTreatment Related TEAEsGrade 3 or 4 TEAEs
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin6464

Number of Participants With TEAEs, Serious TEAEs, Treatment Related TEAEs, Grade 3 or 4 TEAEs Based on NCI CTCAE v4.0: Combination Cohort 3

AE: any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. Serious AE: any untoward medical occurrence at any dose that resulted in death; was life threatening; required inpatient hospitalization or prolongation of existing hospitalization; resulted in persistent or significant disability/incapacity; resulted in congenital anomaly/birth defect. TEAEs: events absent before treatment or that worsened relative to pretreatment state. Treatment-related TEAE: any untoward medical occurrence attributed to study drug in a participant who received study drug. Relatedness to drug was assessed by the Investigator. NCI-CTCAE Grade: Grade 3=severe or medically significant but not immediately life-threatening, hospitalization or prolongation of existing hospitalization indicated, disabling, limiting self-care ADL; Grade 4= life-threatening consequence, urgent intervention indicated. (NCT02038777)
Timeframe: Day 1 up to 28 days after last dose of study drug (maximum up to 869 days)

InterventionParticipants (Count of Participants)
TEAEsSerious TEAEsTreatment related TEAEsGrade 3 or 4 TEAEs
Combination Cohort 3: PF-04449913 100 mg + Azacitidine6165

Number of Participants With Treatment Emergent Adverse Events (TEAEs), Serious TEAEs, Treatment Related TEAEs, Grade 3 or 4 TEAEs Based on NCI CTCAE v4.0: Monotherapy Cohort

AE:any untoward medical occurrence in participant who received study drug without regard to possibility of causal relationship. Serious AE:any untoward medical occurrence at any dose that resulted in death;was life threatening;required inpatient hospitalization or prolongation of existing hospitalization;resulted in persistent or significant disability/incapacity;resulted in congenital anomaly/birth defect. TEAEs:events absent before treatment or that worsened relative to pretreatment state. Treatment-related TEAE:any untoward medical occurrence attributed to study drug in a participant who received study drug. Relatedness to drug was assessed by the Investigator. National cancer institute common terminology criteria (NCI-CTCAE) Grade(G) v4.0:G 3=severe or medically significant but not immediately life-threatening, hospitalization or prolongation of existing hospitalization indicated, disabling, limiting self-care ADL; G 4:life-threatening consequence, urgent intervention indicated. (NCT02038777)
Timeframe: Day 1 up to 28 days after last dose of study drug (For 25 mg: maximum up to 136 days; For 50 mg: maximum up to 179 days; For 100 mg: maximum up to 472 days)

,,
InterventionParticipants (Count of Participants)
TEAEsSerious TEAEsTreatment related TEAEsGrade 3 or 4 TEAEs
Monotherapy Cohort: PF-04449913 100 mg6153
Monotherapy Cohort: PF-04449913 25 mg3112
Monotherapy Cohort: PF-04449913 50 mg4331

Percentage of Participants With Complete Remission (CR) or CR With Incomplete Blood Count Recovery (CRi) and DMR: Combination Cohort 1

CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, bone marrow blasts (BMB) <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. DMR included CR, CRi, MLFS, mCR and PR. CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, bone marrow blasts (BMB) <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. MLFS: neutrophils (mcL) 1000 and platelets (mcL) <100000, BMB <5%. PR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, BMB decrease to 5-25 and >=50% decrease from start. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 486 days)

InterventionPercentage of participants (Number)
CR/CRiDMR
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg16.716.7

Percentage of Participants With CR/CRi and DMR: Combination Cohort 3

CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, bone marrow blasts (BMB) <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. DMR included CR, CRi, MLFS, mCR and PR. CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, bone marrow blasts (BMB) <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. MLFS: neutrophils (mcL) 1000 and platelets (mcL) <100000, BMB <5%. PR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, BMB decrease to 5-25 and >=50% decrease from start. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 841 days)

InterventionPercentage of participants (Number)
CR/CriDMR
Combination Cohort 3: PF-04449913 100 mg + Azacitidine50.050.0

Single Dose- Area Under the Plasma Concentration Curve: From Time Zero to End of Dosing Interval (AUCtau), From Time Zero to Last Quantifiable Concentration (AUClast) and From Time Zero to Infinity (AUCinf) of PF-04449913 for Monotherapy Cohort

AUCtau, was determined by linear/log trapezoidal method. For AUC, tau (dosing interval) was 24 hours. AUClast = area under the curve from time zero to last quantifiable concentration. AUCinf = AUClast + (Clast/kel), where Clast = predicted plasma concentration at the last quantifiable time point estimated from the log-linear regression analysis, and where kel = terminal elimination phase rate constant calculated by a linear regression of the log-linear concentration-time curve. (NCT02038777)
Timeframe: AUCtau: 0 to 24, 24 to 48, 48 to 72 hours post PF-04449913 dosing on Day -5 of Cycle 1; AUClast and AUCinf: Pre-dose and 0.5, 1, 2, 4, 8, 24, 48, 72 hours post PF-04449913 dosing on Day -5 of Cycle 1

,,
InterventionNanogram*hour per milliliter (Geometric Mean)
AUCtauAUClastAUCinf
Monotherapy Cohort: PF-04449913 100 mg88431275013160
Monotherapy Cohort: PF-04449913 25 mg241332553374
Monotherapy Cohort: PF-04449913 50 mg449989119596

Time to Complete Remission (CR) or CR With Incomplete Blood Count Recovery (CRi) and DMR Response: Combination Cohort 3

The time from the date of first dose of study drug to the date of first documentation of a CR or CRi and DMR. DMR included CR, CRi, MLFS, mCR and PR. CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, bone marrow blasts (BMB) <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. MLFS: neutrophils (mcL) 1000 and platelets (mcL) <100000, BMB <5%. PR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, BMB decrease to 5-25 and >=50% decrease from start. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 841 days)

InterventionMonths (Median)
Time to CR/CRiTime to DMR
Combination Cohort 3: PF-04449913 100 mg + Azacitidine5.95.8

Time to Response: Combination Cohort 1

The time from the date of first dose of study drug to the date of first documentation of a CR or CRi and DMR. DMR included CR, CRi, MLFS, mCR and PR. CR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, BMB <5%. CRi: neutrophils (mcL) <1000 or platelets (mcL) <100000, BMB <5%. MLFS: neutrophils (mcL) 1000 and platelets (mcL) <100000, BMB <5%. PR: neutrophils (mcL) >=1000, platelets (mcL) >=100000, BMB decrease to 5-25 and >=50% decrease from start. (NCT02038777)
Timeframe: Day 1 up to end of treatment (maximum up to 486 days)

InterventionMonths (Median)
Time to CR/CRiTime to DMR
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg2.10.8

Number of Participants With Worst On-study Laboratory Abnormalities: Combination Cohort 1

Laboratory parameters included- hematology: lymphocytes/leukocytes (%), neutrophils/leukocytes (%), basophils/leukocytes (%), eosinophils/leukocytes (%), monocytes/leukocytes (%), prothrombin time (sec), blasts/leukocytes (%); clinical chemistry: lactate dehydrogenase (u/l), protein (g/l), BUN (mmol/l), urate (mmol/l), chloride (mmol/l), calcium (mmol/l); urinalysis: specific gravity (scalar), pH (scalar), urine glucose (scalar), ketones (scalar), nitrite, leukocyte esterase, urine erythrocytes (scalar), urine leukocytes (scalar). In this outcome measure participants for each laboratory parameter were evaluated as normal, abnormal low only, abnormal high only or abnormal low and an abnormal high. Laboratory values were as per laboratory normal ranges. Values above normal range = abnormal high and below range = abnormal low. Participants with both an abnormal low and an abnormal high value while on study were reported as 'Abnormal low and Abnormal high'. (NCT02038777)
Timeframe: Baseline up to maximum 514 days

InterventionParticipants (Count of Participants)
Lymphocytes/Leukocytes71912645Neutrophils/Leukocytes71912645Basophils/Leukocytes71912645Eosinophils/Leukocytes71912645Monocytes/Leukocytes71912645Prothrombin time71912645Blasts/Leukocytes71912645Lactate dehydrogenase71912645Protein71912645BUN71912645Urate71912645Chloride71912645Calcium71912645Urine specific gravity71912645Urine pH71912645Urine glucose71912645Urine ketones71912645Urine nitrite71912645Urine leukocyte esterase71912645Urine erythrocytes71912645Urine leukocytes71912645
NormalAbnormal low onlyAbnormal high onlyAbnormal low and abnormal high
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg1
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg0
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg4
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg2
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg3
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg5
Combination Cohort 1 (Unfit Participants): PF-04449913 100 mg + LDAC 20 mg6

Number of Participants With Worst On-study Laboratory Abnormalities: Combination Cohort 2

Laboratory parameters included- hematology: lymphocytes/leukocytes (%), neutrophils/leukocytes (%), basophils/leukocytes (%), eosinophils/leukocytes (%), monocytes/leukocytes (%), prothrombin time (sec), blasts/leukocytes (%); clinical chemistry: lactate dehydrogenase (u/l), protein (g/l), BUN (mmol/l), urate (mmol/l), chloride (mmol/l), calcium (mmol/l); urinalysis: specific gravity (scalar), pH (scalar), urine glucose (scalar), ketones (scalar), urine erythrocytes (scalar), urine leukocytes (scalar). In this outcome measure participants for each laboratory parameter were evaluated as normal, abnormal low only, abnormal high only or abnormal low and an abnormal high. Participants that had both an abnormal low and an abnormal high value while on study were reported as 'Abnormal low and Abnormal high'. (NCT02038777)
Timeframe: Baseline up to maximum 371 days

InterventionParticipants (Count of Participants)
Lymphocytes/Leukocytes71912646Neutrophils/Leukocytes71912646Basophils/Leukocytes71912646Eosinophils/Leukocytes71912646Monocytes/Leukocytes71912646Prothrombin time71912646Blasts/Leukocytes71912646Lactate dehydrogenase71912646Protein71912646BUN71912646Urate71912646Chloride71912646Calcium71912646Urine specific gravity71912646Urine pH71912646Urine glucose71912646Urine ketones71912646Urine erythrocytes71912646Urine leukocytes71912646
Abnormal low onlyAbnormal low and abnormal highNormalAbnormal high only
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin4
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin6
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin3
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin2
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin5
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin0
Combination Cohort 2 (Fit Participants): PF-04449913 100 mg + Cytarabine + Daunorubicin1

Number of Participants With Worst On-study Laboratory Abnormalities: Combination Cohort 3

Laboratory parameters included- hematology: lymphocytes/leukocytes (%), neutrophils/leukocytes (%), basophils/leukocytes (%), eosinophils/leukocytes (%), monocytes/leukocytes (%), blasts/leukocytes (%); clinical chemistry: lactate dehydrogenase (u/l), protein (g/l), BUN (mmol/l), urate (mmol/l), chloride (mmol/l), calcium (mmol/l); urinalysis: specific gravity (scalar), pH (scalar), urine glucose (scalar), ketones (scalar), nitrite, leukocyte esterase, urine erythrocytes (scalar), urine leukocytes (scalar). In this outcome measure participants for each laboratory parameter were evaluated as normal, abnormal low only, abnormal high only or abnormal low and an abnormal high. Participants that had both an abnormal low and an abnormal high value while on study were reported as 'Abnormal low and Abnormal high'. (NCT02038777)
Timeframe: Baseline up to maximum 869 days

InterventionParticipants (Count of Participants)
Lymphocytes/Leukocytes71912648Neutrophils/Leukocytes71912648Basophils/Leukocytes71912648Eosinophils/Leukocytes71912648Monocytes/Leukocytes71912648Blasts/Leukocytes71912648Lactate dehydrogenase71912648Protein71912648BUN71912648Urate71912648Chloride71912648Calcium71912648Urine specific gravity71912648Urine pH71912648Urine glucose71912648Urine ketones71912648Urine nitrite71912648Urine leukocyte esterase71912648Urine erythrocytes71912648Urine leukocytes71912648
Abnormal low onlyAbnormal high onlyAbnormal low and abnormal highNormal
Combination Cohort 3: PF-04449913 100 mg + Azacitidine4
Combination Cohort 3: PF-04449913 100 mg + Azacitidine3
Combination Cohort 3: PF-04449913 100 mg + Azacitidine6
Combination Cohort 3: PF-04449913 100 mg + Azacitidine5
Combination Cohort 3: PF-04449913 100 mg + Azacitidine1
Combination Cohort 3: PF-04449913 100 mg + Azacitidine0
Combination Cohort 3: PF-04449913 100 mg + Azacitidine2

Number of Participants With Worst On-study Laboratory Abnormalities: Monotherapy Cohort

Laboratory parameters included- hematology: lymphocytes/leukocytes (%), neutrophils/leukocytes (%), basophils/leukocytes (%), eosinophils/leukocytes (%), monocytes/leukocytes (%), prothrombin time (sec), blasts/leukocytes (%); clinical chemistry: lactate dehydrogenase (u/l), protein (g/l), blood urea nitrogen (BUN) (mmol/l), urate (mmol/l), chloride (mmol/l), calcium (mmol/l); urinalysis: specific gravity (scalar), pH (scalar), urine glucose (scalar), ketones (scalar), nitrite, leukocyte esterase, urine erythrocytes (scalar), urine leukocytes (scalar). In this outcome measure participants for each laboratory parameter were evaluated as normal, abnormal low only, abnormal high only or abnormal low and an abnormal high. Laboratory values were as per laboratory normal ranges. Values above normal range = abnormal high and below range = abnormal low. Participants with both an abnormal low and an abnormal high value while on study were reported as 'Abnormal low and Abnormal high'. (NCT02038777)
Timeframe: For 25 mg: Baseline up to maximum 136 days; For 50 mg: Baseline up to maximum 179 days; For 100 mg: Baseline up to maximum 472 days

InterventionParticipants (Count of Participants)
Lymphocytes/Leukocytes71912642Lymphocytes/Leukocytes71912643Lymphocytes/Leukocytes71912644Neutrophils/Leukocytes71912642Neutrophils/Leukocytes71912643Neutrophils/Leukocytes71912644Basophils/Leukocytes71912642Basophils/Leukocytes71912643Basophils/Leukocytes71912644Eosinophils/Leukocytes71912643Eosinophils/Leukocytes71912644Eosinophils/Leukocytes71912642Monocytes/Leukocytes71912643Monocytes/Leukocytes71912644Monocytes/Leukocytes71912642Prothrombin time71912643Prothrombin time71912642Prothrombin time71912644Blasts/Leukocytes71912642Blasts/Leukocytes71912643Blasts/Leukocytes71912644Lactate dehydrogenase71912644Lactate dehydrogenase71912642Lactate dehydrogenase71912643Protein71912643Protein71912644Protein71912642BUN71912643BUN71912642BUN71912644Urate71912642Urate71912643Urate71912644Chloride71912644Chloride71912643Chloride71912642Calcium71912642Calcium71912643Calcium71912644Urine specific gravity71912642Urine specific gravity71912644Urine specific gravity71912643Urine pH71912642Urine pH71912644Urine pH71912643Urine glucose71912642Urine glucose71912644Urine glucose71912643Urine ketones71912642Urine ketones71912644Urine ketones71912643Urine nitrite71912642Urine nitrite71912643Urine nitrite71912644Urine leukocyte esterase71912642Urine leukocyte esterase71912643Urine leukocyte esterase71912644Urine erythrocytes71912642Urine erythrocytes71912644Urine erythrocytes71912643Urine leukocytes71912642Urine leukocytes71912643Urine leukocytes71912644
NormalAbnormal low onlyAbnormal high onlyAbnormal low and abnormal high
Monotherapy Cohort: PF-04449913 25 mg2
Monotherapy Cohort: PF-04449913 100 mg2
Monotherapy Cohort: PF-04449913 50 mg2
Monotherapy Cohort: PF-04449913 100 mg4
Monotherapy Cohort: PF-04449913 100 mg0
Monotherapy Cohort: PF-04449913 25 mg1
Monotherapy Cohort: PF-04449913 100 mg1
Monotherapy Cohort: PF-04449913 25 mg3
Monotherapy Cohort: PF-04449913 50 mg4
Monotherapy Cohort: PF-04449913 100 mg5
Monotherapy Cohort: PF-04449913 25 mg0
Monotherapy Cohort: PF-04449913 100 mg3
Monotherapy Cohort: PF-04449913 50 mg3
Monotherapy Cohort: PF-04449913 50 mg0
Monotherapy Cohort: PF-04449913 50 mg1

Median Days to Neutrophil Engraftment

Neutrophil engraftment was recorded as the first day that absolute neutrophil counts (ANC) exceeds 0.5 X 10^9/L for three consecutive readings. (NCT01702961)
Timeframe: 30 days post-transplant

Interventiondays (Median)
BEAM + R: Autologous Stem Cell Transplant11

Disease-free Survival

Disease-free survival at 12 months post-transplant in patients with Hodgkin's disease or non-Hodgkin's lymphomas (NCT01702961)
Timeframe: 12 months post-transplant

Interventionpercentage of participant (Number)
All patientsHodgkin's DiseaseNon-Hodgkin's Lymphomas
BEAM + R: Autologous Stem Cell Transplant768870

Number of Participants With Overall Best Response Achieved After Transplantation

Response was summarized as complete remission (CR): disappearance of all evidence of disease; partial remission (PR): regression of measurable disease (>=50% decrease in sum of the product of the diameters (SPD) of up to six of the largest dominant nodes or nodal masses) and no new sites; stable disease (SD): failure to attain CR/PR/PD; relapsed disease or progressive disease (PD): any new lesion or increase by >= 50% of previously involved sites from nadir. (NCT01702961)
Timeframe: 3 months post-transplant

Interventionparticipants (Number)
Complete Remission (CR)Partial Remission (PR)Stable Disease (SD)Relapsed Disease or Progressive Disease (PD)
BEAM + R: Autologous Stem Cell Transplant63804

Number of Participants Who Successfully Completed the of Quality of Life Form

Subjects receiving outpatient high dose cytarabine or inpatient high dose cytarabine will complete the European Organization for Research and Treatment of Cancer Quality of Life tool on the last day of each cycle of chemotherapy. It encompasses 5 functional scales, 3 symptom scales and a global health measure. Scores range from 0-100 with higher scores associated with improved quality of life. (NCT02101983)
Timeframe: 3 months

InterventionParticipants (Count of Participants)
Outpatient HiDAC Consolidation4
Quality of Life Comparison Group5

Number of Participants With Grades 3 to 5 Non-hematologic Toxicity.

To determine the incidence of number of grades 3 to 5 non-hematologic toxicity of high-dose cytarabine for AML consolidation administered in an outpatient setting. (NCT02101983)
Timeframe: 3 months

InterventionParticipants (Count of Participants)
Outpatient HiDAC Consolidation4
Quality of Life Comparison Group5

Number of Patients Achieving Neutrophil Engraftment

Proportion of patients who successfully achieve neutrophil engraftment after stem cell transplant, defined as an absolute neutrophil count of 500/mm3 or for three consecutive days. (NCT02059239)
Timeframe: 35 Days Post-Transplant

InterventionParticipants (Count of Participants)
Chemo Plus Autologous Transplantation16
Chemo Plus Allogeneic Transplantation13

Number of Patients Achieving Platelet Engraftment

Proportion of patients who successfully achieve platelet engraftment after stem cell transplant, defined as a platelet count of >20k/microL for three consecutive days without transfusion support for seven consecutive days. (NCT02059239)
Timeframe: 74 Days Post-Transplant

InterventionParticipants (Count of Participants)
Chemo Plus Autologous Transplantation16
Chemo Plus Allogeneic Transplantation12

Progression-Free Survival After Stem Cell Transplant

Time elapsed between stem cell transplant (Day 0) and disease progression, as defined by the Cheson Criteria (the appearance of any new lesion >1.5cm in size or a greater than 50% increase in the diameter of a previously identified node of over 1cm in size) (NCT02059239)
Timeframe: Stem cell transplant (Day 0) up to 2 years post-transplant

InterventionMonths (Median)
Chemo Plus Autologous TransplantationNA
Chemo Plus Allogeneic Transplantation8

Transplant-Related Mortality

Death due to any cause other than disease progression within first 100 days post-transplant. (NCT02059239)
Timeframe: From Day 0 until time of death, up to 100 days post-transplant.

InterventionParticipants (Count of Participants)
Chemo Plus Autologous Transplantation0
Chemo Plus Allogeneic Transplantation2

Disease Response 30 Days Post-Transplant

Proportion of patients achieving a complete remission (CR; disappearance of clinically overt disease with no FDG-avid lesions on PET scan), partial remission (PR; reduction in clinical disease buden and >50% bi-dimensional decrease in tumor size on imaging), stable disease (SD; failure to meet the criteria for CR, PR or PD with no new areas of disease involvement) or progressive disease (PD; the appearance of any new lesion >1.5cm in size or a greater than 50% increase in the diameter of a previously identified node of over 1cm in size), as per Cheson Criteria, 30 days following autologous or allogeneic stem cell transplant (NCT02059239)
Timeframe: 30 days after stem cell transplant

,
InterventionParticipants (Count of Participants)
Complete RemissionPartial RemissionStable DiseaseProgressive DiseaseNot Assessed
Chemo Plus Allogeneic Transplantation73111
Chemo Plus Autologous Transplantation122200

Disease Response at 1 Year Post-Transplant

Proportion of patients achieving a complete remission (CR; disappearance of clinically overt disease with no FDG-avid lesions on PET scan), partial remission (PR; reduction in clinical disease buden and >50% bi-dimensional decrease in tumor size on imaging), stable disease (SD; failure to meet the criteria for CR, PR or PD with no new areas of disease involvement) or progressive disease (PD; the appearance of any new lesion >1.5cm in size or a greater than 50% increase in the diameter of a previously identified node of over 1cm in size), as per Cheson Criteria, 1 year following autologous or allogeneic stem cell transplant (NCT02059239)
Timeframe: 1 year after stem cell transplant

,
InterventionParticipants (Count of Participants)
Complete RemissionPartial RemissionStable DiseaseProgressive DiseaseNot AssessedPatient Deceased
Chemo Plus Allogeneic Transplantation400207
Chemo Plus Autologous Transplantation1210300

Disease Response Following Salvage Chemotherapy

Proportion of patients achieving a complete remission (CR; disappearance of clinically overt disease with no FDG-avid lesions on PET scan), partial remission (PR; reduction in clinical disease buden and >50% bi-dimensional decrease in tumor size on imaging), stable disease (SD; failure to meet the criteria for CR, PR or PD with no new areas of disease involvement) or progressive disease (PD; the appearance of any new lesion >1.5cm in size or a greater than 50% increase in the diameter of a previously identified node of over 1cm in size), as per Cheson Criteria, following salvage chemotherapy with Bendamustine. (NCT02059239)
Timeframe: Within 14 days of salvage chemotherapy treatment

,
InterventionParticipants (Count of Participants)
Complete RemissionPartial RemissionStable DiseaseProgressive DiseaseNot Assessed
Chemo Plus Allogeneic Transplantation16261
Chemo Plus Autologous Transplantation48411

Overall Survival at Day 365 Post-Transplant

The time from stem cell infusion (Day 0) to death from any cause. (NCT02059239)
Timeframe: From Day 0 until time of death, assessed up to 365 days post-transplant

,
InterventionParticipants (Count of Participants)
AliveDeceased
Chemo Plus Allogeneic Transplantation67
Chemo Plus Autologous Transplantation160

Research Highlights

Safety/Toxicity (423)

ArticleYear
Lower cardiotoxicity of CPX-351 relative to daunorubicin plus cytarabine free-drug combination in hiPSC-derived cardiomyocytes in vitro.
Scientific reports, Nov-29, Volume: 13, Issue: 1
2023
Safety and efficacy of FLAG-Ida-based therapy combined with venetoclax for the treatment for newly diagnosed and relapsed/refractory patients with AML - A systematic review.
Leukemia research, Volume: 133
2023
A Phase I study of Milademetan (DS3032b) in combination with low dose cytarabine with or without venetoclax in acute myeloid leukemia: Clinical safety, efficacy, and correlative analysis.
Blood cancer journal, 06-29, Volume: 13, Issue: 1
2023
[Efficacy and Safety of Decitabine Combined with Modified CAG Regimen in Patients Aged ≥ 70 Years with Newly Diagnosed Acute Myeloid Leukemia].
Zhongguo shi yan xue ye xue za zhi, Volume: 31, Issue: 3
2023
Efficacy and toxicity of CLAG combined with pegylated liposomal doxorubicin in the treatment of refractory or relapsed acute myeloid leukemia.
Cancer medicine, Volume: 12, Issue: 11
2023
Exposure to cytarabine causes side effects on adult development and physiology and induces intestinal damage via apoptosis in Drosophila.
Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, Volume: 159
2023
Exploring the possible mitoprotective and neuroprotective potency of thymoquinone, betanin, and vitamin D against cytarabine-induced mitochondrial impairment and neurotoxicity in rats' brain.
Journal of biochemical and molecular toxicology, Volume: 37, Issue: 2
2023
Isocitrate dehydrogenase 2 inhibitor enasidenib synergizes daunorubicin cytotoxicity by targeting aldo-keto reductase 1C3 and ATP-binding cassette transporters.
Archives of toxicology, Volume: 96, Issue: 12
2022
Safety and efficacy of a dose-dense short-term therapy in patients with MYC-translocated aggressive lymphoma.
Blood advances, 11-22, Volume: 6, Issue: 22
2022
Efficacy and toxicity of SEAM (semustine, etoposide, cytarabine, and melphalan) conditioning regimen followed by autologous stem cell transplantation in lymphoma.
Hematology (Amsterdam, Netherlands), Volume: 27, Issue: 1
2022
Whole brain radiotherapy combined with intrathecal liposomal cytarabine for leptomeningeal metastasis-a safety analysis and validation of the EANO-ESMO classification.
Strahlentherapie und Onkologie : Organ der Deutschen Rontgengesellschaft ... [et al], Volume: 198, Issue: 5
2022
Retreatment with full-dose cytarabine in a patient with previous neurological toxicity.
Journal of oncology pharmacy practice : official publication of the International Society of Oncology Pharmacy Practitioners, Volume: 28, Issue: 7
2022
Safety and feasibility of outpatient high-dose cytarabine and intermediate-dose cytarabine for consolidation therapy in acute myeloid leukemia.
Journal of oncology pharmacy practice : official publication of the International Society of Oncology Pharmacy Practitioners, Volume: 28, Issue: 8
2022
BuCyE can safely replace BEAM as a conditioning regimen for autologous stem cell transplantation in the treatment of refractory and relapsed lymphomas.
Leukemia research, Volume: 110
2021
Primary mesenchymal stromal cells in co-culture with leukaemic HL-60 cells are sensitised to cytarabine-induced genotoxicity, while leukaemic cells are protected.
Mutagenesis, 11-29, Volume: 36, Issue: 6
2021
Quality-adjusted Time Without Symptoms of disease or Toxicity (Q-TWiST) analysis of CPX-351 versus 7 + 3 in older adults with newly diagnosed high-risk/secondary AML.
Journal of hematology & oncology, 07-13, Volume: 14, Issue: 1
2021
BeEAM conditioning regimen is a safe, efficacious and economical alternative to BEAM chemotherapy.
Scientific reports, 07-07, Volume: 11, Issue: 1
2021
Cytarabine-Induced Corneal Toxicity: Clinical Features and Relief of Symptoms with Loteprednol Etabonate 0.5% in Two Patients
Turkish journal of ophthalmology, 04-29, Volume: 51, Issue: 2
2021
Efficacy and safety of consolidation therapy with intermediate and high dose cytarabine in acute myeloid leukemia patients.
Hematology (Amsterdam, Netherlands), Volume: 26, Issue: 1
2021
Preparation, characterization, and cytotoxicity evaluation of self-assembled nanoparticles of diosgenin-cytarabine conjugate.
Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association, Volume: 151
2021
BL-8040 CXCR4 antagonist is safe and demonstrates antileukemic activity in combination with cytarabine for the treatment of relapsed/refractory acute myelogenous leukemia: An open-label safety and efficacy phase 2a study.
Cancer, 04-15, Volume: 127, Issue: 8
2021
Impact of contemporary regimens on the outcomes and toxicity of primary CNS lymphoma: a single-center retrospective analysis of 73 patients.
Journal of neuro-oncology, Volume: 151, Issue: 2
2021
A dose-dense short-term therapy for human immunodeficiency virus/acquired immunodeficiency syndrome patients with high-risk Burkitt lymphoma or high-grade B-cell lymphoma: safety and efficacy results of the "CARMEN" phase II trial.
British journal of haematology, Volume: 192, Issue: 1
2021
Aprepitant Sensitizes Acute Myeloid Leukemia Cells to the Cytotoxic Effects of Cytosine Arabinoside in vitro and in vivo.
Drug design, development and therapy, Volume: 14
2020
The efficacy and safety of daunorubicin versus idarubicin combined with cytarabine for induction therapy in acute myeloid leukemia: A meta-analysis of randomized clinical trials.
Medicine, Jun-12, Volume: 99, Issue: 24
2020
Linezolid is safe on platelet count for AML patients during myelosuppression after consolidation chemotherapy.
Journal of clinical pharmacy and therapeutics, Volume: 45, Issue: 4
2020
Assessment of Anti-Tumor potential and safety of application of Glutathione stabilized Gold Nanoparticles conjugated with Chemotherapeutics.
International journal of medical sciences, Volume: 17, Issue: 6
2020
Final safety and efficacy results from the CPX-351 early access program for older patients with high-risk or secondary acute myeloid leukemia.
Leukemia & lymphoma, Volume: 61, Issue: 5
2020
Acute cerebellar toxicity induced by high dose of cytarabine (HiDAC): A case report.
Journal of oncology pharmacy practice : official publication of the International Society of Oncology Pharmacy Practitioners, Volume: 26, Issue: 6
2020
HOTAIRM1 knockdown enhances cytarabine-induced cytotoxicity by suppression of glycolysis through the Wnt/β-catenin/PFKP pathway in acute myeloid leukemia cells.
Archives of biochemistry and biophysics, 02-15, Volume: 680
2020
Toxicity of cytarabine constant rate infusion in dogs with high-grade non-Hodgkin lymphoma with bone marrow or central nervous system involvement.
Australian veterinary journal, Volume: 98, Issue: 3
2020
The efficacy and toxicity of the CHG priming regimen (low-dose cytarabine, homoharringtonine, and G-CSF) in higher risk MDS patients relapsed or refractory to decitabine.
Journal of cancer research and clinical oncology, Volume: 145, Issue: 12
2019
Gastrointestinal Adverse Events Observed After Chimeric Antigen Receptor T-Cell Therapy.
American journal of clinical oncology, Volume: 42, Issue: 10
2019
Spinal Cord Toxicity from Intrathecal Chemotherapy: A Case with Clinicopathologic Correlation.
World neurosurgery, Volume: 128
2019
Cytarabine ears - A side effect of cytarabine therapy.
Journal of oncology pharmacy practice : official publication of the International Society of Oncology Pharmacy Practitioners, Volume: 26, Issue: 2
2020
Evaluation of the impact of single-nucleotide polymorphisms on treatment response, survival and toxicity with cytarabine and anthracyclines in patients with acute myeloid leukaemia: a systematic review protocol.
Systematic reviews, May-03, Volume: 8, Issue: 1
2019
[Efficacy and Safety of Decitabine Combined with CAG (Cytarabine, Aclarubicin, G-CSF) for Patients with Intermediate or High Risk Myelodysplastic Syndrome and Acute Myeloid Leukemia: a Meta-Analysis].
Zhongguo shi yan xue ye xue za zhi, Volume: 27, Issue: 2
2019
Chemotherapy-induced skin toxicity and capillary leak syndrome.
International journal of dermatology, Volume: 58, Issue: 7
2019
Homoharringtonine is a safe and effective substitute for anthracyclines in children younger than 2 years old with acute myeloid leukemia.
Frontiers of medicine, Volume: 13, Issue: 3
2019
Case report of neurotoxicity with blinatumomab and concurrent intrathecal chemotherapy in second relapse of acute lymphoblastic leukemia with central nervous system disease.
Journal of oncology pharmacy practice : official publication of the International Society of Oncology Pharmacy Practitioners, Volume: 25, Issue: 8
2019
Successful treatment of cytarabine-related neurotoxicity with corticosteroids, a case series.
International journal of hematology, Volume: 108, Issue: 5
2018
High-dose Bendamustine-EAM followed by autologous stem cell rescue results in long-term remission rates in lymphoma patients, without renal toxicity.
European journal of haematology, Volume: 101, Issue: 3
2018
Neurotoxicity of cytarabine (Ara-C) in dorsal root ganglion neurons originates from impediment of mtDNA synthesis and compromise of mitochondrial function.
Free radical biology & medicine, Volume: 121
2018
Safety and feasibility of a low frame rate protocol for percutaneous coronary intervention to chronic total occlusions: preliminary experience.
EuroIntervention : journal of EuroPCR in collaboration with the Working Group on Interventional Cardiology of the European Society of Cardiology, Aug-03, Volume: 14, Issue: 5
2018
Alternating R-CHOP and R-cytarabine is a safe and effective regimen for transplant-ineligible patients with a newly diagnosed mantle cell lymphoma.
Hematological oncology, Volume: 36, Issue: 1
2018
Total Body Irradiation Is Safe and Similarly Effective as Chemotherapy-Only Conditioning in Autologous Stem Cell Transplantation for Mantle Cell Lymphoma.
Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation, Volume: 24, Issue: 2
2018
Efficacy and safety of G-CSF, low-dose cytarabine and aclarubicin in combination with l-asparaginase, prednisone in the treatment of refractory or relapsed acute lymphoblastic leukemia.
Leukemia research, Volume: 62
2017
Evaluation of standardized triple intrathecal therapy toxicity in oncohematological adult patients.
Farmacia hospitalaria : organo oficial de expresion cientifica de la Sociedad Espanola de Farmacia Hospitalaria, Sep-01, Volume: 41, Issue: 5
2017
Intensive consolidation with G-CSF support: Tolerability, safety, reduced hospitalization, and efficacy in acute myeloid leukemia patients ≥60 years.
American journal of hematology, Volume: 92, Issue: 10
2017
Assessment of neurotoxicity of pharmacological compounds during early neural development of human embryonic stem cells.
Journal of physiology and pharmacology : an official journal of the Polish Physiological Society, Volume: 68, Issue: 2
2017
Impact of novel polymorphisms related to cytotoxicity of cytarabine in the induction treatment of acute myeloid leukemia.
Pharmacogenetics and genomics, Volume: 27, Issue: 7
2017
Incidence and prognostic significance of nephrotoxicity in patients receiving eshap as salvage therapy for lymphoma.
Leukemia research, Volume: 58
2017
Efficacy and Toxicity of Induction Therapy with Cladribine, Idarubicin, and Cytarabine (IAC) for Acute Myeloid Leukemia.
Anticancer research, Volume: 37, Issue: 2
2017
Evaluation of standardized triple intrathecal therapy toxicity in oncohematological pediatric patients.
International journal of clinical pharmacy, Volume: 39, Issue: 1
2017
Optimal AraC-Cytotoxicity to AML Cells Requires ERK5 Activity.
Journal of cellular biochemistry, Volume: 118, Issue: 6
2017
Outpatient administration of BEAM conditioning prior to autologous stem cell transplantation for lymphoma is safe, feasible, and cost-effective.
Cancer medicine, Volume: 5, Issue: 11
2016
[Efficacy and safety analysis of different dose idarubicin plus cytarabine regimen as induction chemotherapy for young patients with de-novo acute myeloid leukemia].
Zhonghua xue ye xue za zhi = Zhonghua xueyexue zazhi, Aug-14, Volume: 37, Issue: 8
2016
Efficacy and Toxicity of Intrathecal Liposomal Cytarabine in First-line Therapy of Childhood Acute Lymphoblastic Leukemia.
Journal of pediatric hematology/oncology, Volume: 38, Issue: 8
2016
[Efficacy and safety analysis of the combination of cladribine, cytarabine, granulocyte colonystimulating factor (CLAG) regime in patients with refractory or relapsed acute myeloid leukemia].
Zhonghua xue ye xue za zhi = Zhonghua xueyexue zazhi, Volume: 37, Issue: 7
2016
Factors Influencing Pulmonary Toxicity in Children Undergoing Allogeneic Hematopoietic Stem Cell Transplantation in the Setting of Total Body Irradiation-Based Myeloablative Conditioning.
International journal of radiation oncology, biology, physics, Feb-01, Volume: 94, Issue: 2
2016
ABCC4 Is a Determinant of Cytarabine-Induced Cytotoxicity and Myelosuppression.
Clinical and translational science, Volume: 9, Issue: 1
2016
LACE versus BEAM conditioning in relapsed and refractory lymphoma transplant: retrospective multicenter analysis of toxicity and efficacy.
International journal of hematology, Volume: 103, Issue: 3
2016
Efficacy and toxicity of carboplatin and cytarabine chemotherapy for dogs with relapsed or refractory lymphoma (2000-2013).
Veterinary and comparative oncology, Volume: 15, Issue: 2
2017
Efficacy and toxicity of decitabine versus CHG regimen (low-dose cytarabine, homoharringtonine and granulocyte colony-stimulating factor) in patients with higher risk myelodysplastic syndrome: a retrospective study.
Leukemia & lymphoma, Volume: 57, Issue: 6
2016
BEAM Conditioning Regimen Has Higher Toxicity Compared With High-Dose Melphalan for Salvage Autologous Hematopoietic Stem Cell Transplantation in Multiple Myeloma.
Clinical lymphoma, myeloma & leukemia, Volume: 15, Issue: 9
2015
AMC 048: modified CODOX-M/IVAC-rituximab is safe and effective for HIV-associated Burkitt lymphoma.
Blood, Jul-09, Volume: 126, Issue: 2
2015
Risk factors for cytarabine-induced cutaneous toxicity in patients with haematological malignancies.
Chemotherapy, Volume: 60, Issue: 3
2014
Safety and Efficacy of Liposomal Cytarabine in the Treatment of Neoplastic Meningitis.
Oncology, Volume: 89, Issue: 3
2015
Efficacy and safety of decitabine in combination with G-CSF, low-dose cytarabine and aclarubicin in newly diagnosed elderly patients with acute myeloid leukemia.
Oncotarget, Mar-20, Volume: 6, Issue: 8
2015
Liposomal cytarabine in the prophylaxis and treatment of CNS lymphoma: toxicity analysis in a retrospective case series study conducted at Polish Lymphoma Research Group Centers.
Medical oncology (Northwood, London, England), Volume: 32, Issue: 4
2015
Impact of single nucleotide polymorphisms of cytarabine metabolic genes on drug toxicity in childhood acute lymphoblastic leukemia.
Pediatric blood & cancer, Volume: 62, Issue: 4
2015
A randomized trial of prophylactic palifermin on gastrointestinal toxicity after intensive induction therapy for acute myeloid leukaemia.
British journal of haematology, Volume: 167, Issue: 5
2014
Safety and clinical activity of 5-aza-2'-deoxycytidine (decitabine) with or without Hyper-CVAD in relapsed/refractory acute lymphocytic leukaemia.
British journal of haematology, Volume: 167, Issue: 3
2014
Safety of intrathecal administration of cytosine arabinoside and methotrexate in dogs and cats.
Veterinary and comparative oncology, Volume: 14, Issue: 3
2016
Enhanced skin toxicity associated with the combination of clofarabine plus cytarabine for the treatment of acute leukemia.
Cancer chemotherapy and pharmacology, Volume: 74, Issue: 2
2014
Toxicity and efficacy of intrathecal liposomal cytarabine in children with leukemia/lymphoma relapsing in the central nervous system: a retrospective multicenter study.
Leukemia & lymphoma, Volume: 56, Issue: 3
2015
Safety of inadvertent administration of overdose of intrathecal Cytarabine in a pediatric patient.
Journal of oncology pharmacy practice : official publication of the International Society of Oncology Pharmacy Practitioners, Volume: 21, Issue: 5
2015
Increasing the dose of aclarubicin in low-dose cytarabine and aclarubicin in combination with granulocyte colony-stimulating factor (CAG regimen) can safely and effectively treat relapsed or refractory acute myeloid leukemia.
International journal of hematology, Volume: 99, Issue: 5
2014
The efficacy and adverse event profile of dexamethasone, melphalan, actinomycin D, and cytosine arabinoside (DMAC) chemotherapy in relapsed canine lymphoma.
The Canadian veterinary journal = La revue veterinaire canadienne, Volume: 55, Issue: 2
2014
Elacytarabine in relapsed/refractory acute myeloid leukaemia: an evaluation of clinical efficacy, pharmacokinetics, cardiac safety and effects on lipid profile.
Leukemia research, Volume: 38, Issue: 3
2014
Prediction of adverse events during intensive induction chemotherapy for acute myeloid leukemia or high-grade myelodysplastic syndromes.
American journal of hematology, Volume: 89, Issue: 4
2014
Fludarabine, cytarabine, granulocyte colony-stimulating factor and melphalan (FALG with L-PAM) as a reduced toxicity conditioning regimen in children with acute leukemia.
Pediatric blood & cancer, Volume: 61, Issue: 4
2014
Safety and tolerability of intrathecal liposomal cytarabine as central nervous system prophylaxis in patients with acute lymphoblastic leukemia.
Leukemia & lymphoma, Volume: 55, Issue: 8
2014
Pharmacokinetics and toxicity of intrathecal liposomal cytarabine in children and adolescents following age-adapted dosing.
Clinical pharmacokinetics, Volume: 53, Issue: 2
2014
Phase II trial to assess the safety and efficacy of clofarabine in combination with low-dose cytarabine in elderly patients with acute myeloid leukemia.
Annals of hematology, Volume: 93, Issue: 1
2014
Cytosine arabinoside promotes cytotoxic effect of T cells on leukemia cells mediated by bispecific antibody.
Human gene therapy, Volume: 24, Issue: 8
2013
A phase I/II trial to evaluate the safety, feasibility and activity of salvage therapy consisting of the mTOR inhibitor Temsirolimus added to standard therapy of Rituximab and DHAP for the treatment of patients with relapsed or refractory diffuse large ce
BMC cancer, Jun-25, Volume: 13
2013
Phase I study assessing the safety and tolerability of barasertib (AZD1152) with low-dose cytosine arabinoside in elderly patients with AML.
Clinical lymphoma, myeloma & leukemia, Volume: 13, Issue: 5
2013
Stage I of a phase 2 study assessing the efficacy, safety, and tolerability of barasertib (AZD1152) versus low-dose cytosine arabinoside in elderly patients with acute myeloid leukemia.
Cancer, Jul-15, Volume: 119, Issue: 14
2013
Aurora B inhibitor barasertib and cytarabine exert a greater-than-additive cytotoxicity in acute myeloid leukemia cells.
Cancer science, Volume: 104, Issue: 7
2013
Cytarabine, aclarubicin and granulocyte colony-stimulating factor regimen represents an effective and safe salvage regimen for patients with acute myeloid leukemia refractory to first course of induction chemotherapy.
Leukemia & lymphoma, Volume: 54, Issue: 11
2013
Safety and pharmacokinetics of the antisense oligonucleotide (ASO) LY2181308 as a single-agent or in combination with idarubicin and cytarabine in patients with refractory or relapsed acute myeloid leukemia (AML).
Investigational new drugs, Volume: 31, Issue: 4
2013
Fucoidan prevents multiple myeloma cell escape from chemotherapy-induced drug cytotoxicity.
Fitoterapia, Volume: 84
2013
Glucose-functionalized multidrug-conjugating nanoparticles based on amphiphilic terpolymer with enhanced anti-tumorous cell cytotoxicity.
International journal of pharmaceutics, Jan-30, Volume: 441, Issue: 1-2
2013
Preclinical strategy to reduce clinical hepatotoxicity using in vitro bioactivation data for >200 compounds.
Chemical research in toxicology, Oct-15, Volume: 25, Issue: 10
2012
Effects of selective checkpoint kinase 1 inhibition on cytarabine cytotoxicity in acute myelogenous leukemia cells in vitro.
Clinical cancer research : an official journal of the American Association for Cancer Research, Oct-01, Volume: 18, Issue: 19
2012
Influence of obesity on efficacy and toxicity of induction chemotherapy in patients with newly diagnosed acute myeloid leukemia.
Leukemia & lymphoma, Volume: 54, Issue: 3
2013
Safety and efficacy of azacitidine in the treatment of elderly patients with myelodysplastic syndrome.
Clinical interventions in aging, Volume: 7
2012
[Clinical efficacies and safety of HAG regimen for patients with high-risk myelodysplastic syndromes: a multicentre study].
Zhonghua yi xue za zhi, Mar-13, Volume: 92, Issue: 10
2012
High-dose cytarabine (24 g/m2) in combination with idarubicin (HiDAC-3) results in high first-cycle response with limited gastrointestinal toxicity in adult acute myeloid leukaemia.
Internal medicine journal, Volume: 43, Issue: 3
2013
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Long-term Use (8)

ArticleYear
Long-Term Outcomes of Imatinib Treatment for Chronic Myeloid Leukemia.
The New England journal of medicine, 03-09, Volume: 376, Issue: 10
2017
Early imatinib-mesylate-induced hepatotoxicity in chronic myelogenous leukaemia.
Acta haematologica, Volume: 118, Issue: 4
2007
In vitro and in vivo pharmacological profile of UFP-512, a novel selective delta-opioid receptor agonist; correlations between desensitization and tolerance.
British journal of pharmacology, Volume: 152, Issue: 8
2007
Neurological complications of antineoplastic therapy.
Acta neurologica Scandinavica. Supplementum, Volume: 100
1984
Effect of cytosine arabinofuranoside (AraC) on reactive gliosis in vivo. An immunohistochemical and morphometric study.
Brain research, Mar-04, Volume: 328, Issue: 2
1985
Study on the long-term toxic efficacy of cytosine-arabinoside in Sprague-Dawley rats.
Cancer letters, Dec-01, Volume: 43, Issue: 1-2
1988
Clinical studies on streptococcal preparation (OK-432) combined with mitomycin-C, 5-FU and cytosine arabinoside in advanced cancer patients.
The Japanese journal of surgery, Volume: 5, Issue: 3
1975
Inhibition of cyclocytidine-induced enlargement of parotid and heart by propranolol or sympathectomy.
Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine (New York, N.Y.), Volume: 150, Issue: 2
1975
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Pharmacokinetics (138)

ArticleYear
Optimization of idarubicin and cytarabine induction regimen with homoharringtonine for newly diagnosed acute myeloid leukemia patients based on the peripheral blast clearance rate: A single-arm, phase 2 trial (RJ-AML 2014).
American journal of hematology, 01-01, Volume: 97, Issue: 1
2022
Intracellular Pharmacokinetics of Pyrimidine Analogues used in Oncology and the Correlation with Drug Action.
Clinical pharmacokinetics, Volume: 59, Issue: 12
2020
Simultaneous determination of cytosine arabinoside and its metabolite uracil arabinoside in human plasma by LC-MS/MS: Application to pharmacokinetics-pharmacogenetics pilot study in AML patients.
Journal of chromatography. B, Analytical technologies in the biomedical and life sciences, Sep-15, Volume: 1126-1127
2019
A phase 2 study to assess the pharmacokinetics and pharmacodynamics of CPX-351 and its effects on cardiac repolarization in patients with acute leukemias.
Cancer chemotherapy and pharmacology, Volume: 84, Issue: 1
2019
Population Pharmacokinetics and Exposure-Response Analyses for CPX-351 in Patients With Hematologic Malignancies.
Journal of clinical pharmacology, Volume: 59, Issue: 5
2019
Clearance of Somatic Mutations at Remission and the Risk of Relapse in Acute Myeloid Leukemia.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 06-20, Volume: 36, Issue: 18
2018
Persistent cytarabine and daunorubicin exposure after administration of novel liposomal formulation CPX-351: population pharmacokinetic assessment.
Cancer chemotherapy and pharmacology, Volume: 81, Issue: 1
2018
Population Pharmacokinetics of Volasertib Administered in Patients with Acute Myeloid Leukaemia as a Single Agent or in Combination with Cytarabine.
Clinical pharmacokinetics, Volume: 57, Issue: 3
2018
Efficacy of Pharmacokinetics-Directed Busulfan, Cyclophosphamide, and Etoposide Conditioning and Autologous Stem Cell Transplantation for Lymphoma: Comparison of a Multicenter Phase II Study and CIBMTR Outcomes.
Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation, Volume: 22, Issue: 7
2016
The pharmacokinetics of high-dose methotrexate in people living with HIV on antiretroviral therapy.
Cancer chemotherapy and pharmacology, Volume: 77, Issue: 3
2016
Etoposide pharmacokinetics impact the outcomes of lymphoma patients treated with BEAM regimen and ASCT: a multicenter study of the LYmphoma Study Association (LYSA).
Cancer chemotherapy and pharmacology, Volume: 76, Issue: 5
2015
Association Between Mutation Clearance After Induction Therapy and Outcomes in Acute Myeloid Leukemia.
JAMA, Aug-25, Volume: 314, Issue: 8
2015
Pharmacodynamics of cytarabine induced leucopenia: a retrospective cohort study.
British journal of clinical pharmacology, Volume: 79, Issue: 4
2015
Simultaneous determination of 1-β-d-Arabinofuranosylcytosine and two metabolites, 1-β-d-Arabinofuranosyluracil and 1-β-d-Arabinofuranosylcytosine triphosphate in leukemic cell by HPLC-MS/MS and the application to cell pharmacokinetics.
Journal of chromatography. B, Analytical technologies in the biomedical and life sciences, Jul-01, Volume: 962
2014
Pharmacokinetics and toxicity of intrathecal liposomal cytarabine in children and adolescents following age-adapted dosing.
Clinical pharmacokinetics, Volume: 53, Issue: 2
2014
Successful large-volume cerebrospinal fluid aspiration for an accidental overdose of intrathecal cytarabine.
Medical oncology (Northwood, London, England), Volume: 30, Issue: 2
2013
Safety and pharmacokinetics of the antisense oligonucleotide (ASO) LY2181308 as a single-agent or in combination with idarubicin and cytarabine in patients with refractory or relapsed acute myeloid leukemia (AML).
Investigational new drugs, Volume: 31, Issue: 4
2013
The pharmacokinetics of cytarabine in dogs when administered via subcutaneous and continuous intravenous infusion routes.
Journal of veterinary pharmacology and therapeutics, Volume: 36, Issue: 4
2013
Pharmacokinetics of CPX-351; a nano-scale liposomal fixed molar ratio formulation of cytarabine:daunorubicin, in patients with advanced leukemia.
Leukemia research, Volume: 36, Issue: 10
2012
Population pharmacokinetics of cytarabine, etoposide, and daunorubicin in the treatment for acute myeloid leukemia.
Cancer chemotherapy and pharmacology, Volume: 69, Issue: 5
2012
Single-cell pharmacodynamic monitoring of S6 ribosomal protein phosphorylation in AML blasts during a clinical trial combining the mTOR inhibitor sirolimus and intensive chemotherapy.
Clinical cancer research : an official journal of the American Association for Cancer Research, Mar-15, Volume: 18, Issue: 6
2012
Phase I pharmacokinetic and pharmacodynamic study of the multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in pediatric relapsed/refractory leukemia.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, Aug-20, Volume: 29, Issue: 24
2011
Effects of interior gelation on pharmacokinetics and biodistribution of liposomes encapsulating an anti-cancer drug cytarabine.
Journal of biomedical nanotechnology, Volume: 6, Issue: 6
2010
Phase 1 and pharmacokinetic study of bolus-infusion flavopiridol followed by cytosine arabinoside and mitoxantrone for acute leukemias.
Blood, Mar-24, Volume: 117, Issue: 12
2011
Effective clearance of Ara-U the major metabolite of cytosine arabinoside (Ara-C) by hemodialysis in a patient with lymphoma and end-stage renal failure.
Cancer chemotherapy and pharmacology, Volume: 67, Issue: 4
2011
Methotrexate area under the curve is an important outcome predictor in patients with primary CNS lymphoma: A pharmacokinetic-pharmacodynamic analysis from the IELSG no. 20 trial.
British journal of cancer, Feb-16, Volume: 102, Issue: 4
2010
In silico prediction of volume of distribution in human using linear and nonlinear models on a 669 compound data set.
Journal of medicinal chemistry, Jul-23, Volume: 52, Issue: 14
2009
Pharmacokinetics and safety of intrathecal liposomal cytarabine in children aged <3 years.
Clinical pharmacokinetics, Volume: 48, Issue: 4
2009
Thioguanine pharmacokinetics in induction therapy of children with acute myeloid leukemia.
Anti-cancer drugs, Volume: 20, Issue: 1
2009
[Relationship between the expression of the genes encoding the key enzymes for cytarabine metabolism and the pharmacokinetics of cytarabine in the treatment of childhood acute leukemia with high-dose cytarabine].
Zhonghua er ke za zhi = Chinese journal of pediatrics, Volume: 46, Issue: 4
2008
Pharmacokinetics of CPX-351 (cytarabine/daunorubicin HCl) liposome injection in the mouse.
Journal of pharmaceutical sciences, Volume: 98, Issue: 7
2009
Phase I and pharmacokinetic study of bortezomib in combination with idarubicin and cytarabine in patients with acute myelogenous leukemia.
Clinical cancer research : an official journal of the American Association for Cancer Research, Mar-01, Volume: 14, Issue: 5
2008
High-dose idarubicin in combination with Ara-C in patients with relapsed or refractory acute lymphoblastic leukemia: a pharmacokinetic and clinical study.
Cancer chemotherapy and pharmacology, Volume: 59, Issue: 6
2007
Pharmacokinetic characteristics of L-valyl-ara-C and its implication on the oral delivery of ara-C.
Acta pharmacologica Sinica, Volume: 28, Issue: 2
2007
A pharmacokinetic study of intra-CSF administered encapsulated cytarabine (DepoCyt) for the treatment of neoplastic meningitis in patients with leukemia, lymphoma, or solid tumors as part of a phase III study.
Journal of neuro-oncology, Volume: 81, Issue: 2
2007
Body mass index does not influence pharmacokinetics or outcome of treatment in children with acute lymphoblastic leukemia.
Blood, Dec-15, Volume: 108, Issue: 13
2006
Clearance of leukaemic blasts from peripheral blood during standard induction treatment predicts the bone marrow response in acute myeloid leukaemia: a pilot study.
British journal of haematology, Volume: 134, Issue: 1
2006
[The pharmacokinetic study on cytarabine nanoparticle lyophilization injection in rabbits].
Sichuan da xue xue bao. Yi xue ban = Journal of Sichuan University. Medical science edition, Volume: 37, Issue: 1
2006
Phase I and pharmacokinetic study of flavopiridol followed by 1-beta-D-arabinofuranosylcytosine and mitoxantrone in relapsed and refractory adult acute leukemias.
Clinical cancer research : an official journal of the American Association for Cancer Research, Dec-01, Volume: 11, Issue: 23
2005
Modeling the pharmacodynamics of highly schedule-dependent agents: exemplified by cytarabine-based regimens in acute myeloid leukemia.
Clinical cancer research : an official journal of the American Association for Cancer Research, Oct-15, Volume: 11, Issue: 20
2005
Clofarabine in adult acute leukemias: clinical success and pharmacokinetics.
Nucleosides, nucleotides & nucleic acids, Volume: 23, Issue: 8-9
2004
Pharmacokinetics of 2-chlorodeoxyadenosine in a child undergoing hemofiltration and hemodialysis for acute renal failure.
Journal of pediatric hematology/oncology, Volume: 24, Issue: 8
2002
Phase 1 and pharmacodynamic studies of G3139, a Bcl-2 antisense oligonucleotide, in combination with chemotherapy in refractory or relapsed acute leukemia.
Blood, Jan-15, Volume: 101, Issue: 2
2003
Clinical pharmacokinetics of cytarabine formulations.
Clinical pharmacokinetics, Volume: 41, Issue: 10
2002
Pharmacodynamic modeling of thrombopoietin, platelet, and megakaryocyte dynamics in patients with acute myeloid leukemia undergoing dose intensive chemotherapy.
Journal of clinical pharmacology, Volume: 42, Issue: 5
2002
Proliferative activity of leukaemic blasts and cytosine arabinoside pharmacodynamics are associated with cytogenetically defined prognostic subgroups in acute myeloid leukaemia.
British journal of haematology, Volume: 113, Issue: 4
2001
Cellular pharmacokinetics and pharmacodynamics of the deoxycytidine analog 2'-C-cyano-2'-deoxy-1-beta-D-arabino-pentofuranosylcytosine (CNDAC).
Biochemical pharmacology, Jun-15, Volume: 61, Issue: 12
2001
Pharmacokinetics of nimustine, cytosine arabinoside, and methotrexate in cerebrospinal fluid during cerebrospinal fluid perfusion chemotherapy.
Biological & pharmaceutical bulletin, Volume: 24, Issue: 4
2001
Cell cycle-related changes in regulatory volume decrease and volume-sensitive chloride conductance in mouse fibroblasts.
Journal of cellular physiology, Volume: 187, Issue: 1
2001
Phase II trial and pharmacokinetic evaluation of cytosine arabinoside for leptomeningeal metastases from breast cancer.
Cancer chemotherapy and pharmacology, Volume: 46, Issue: 5
2000
Pharmacokinetics of cytosine arabinoside, methotrexate, nimustine and valproic acid in cerebrospinal fluid during cerebrospinal fluid perfusion chemotherapy.
Biological & pharmaceutical bulletin, Volume: 23, Issue: 6
2000
In vitro cytotoxic drug activity and in vivo pharmacokinetics in childhood acute myeloid leukemia.
Advances in experimental medicine and biology, Volume: 457
1999
Evaluation of cell-killing effects of 1-beta-D-arabinofuranosylcytosine and daunorubicin by a new computer-controlled in vitro pharmacokinetic simulation system.
Cancer research, Jun-01, Volume: 59, Issue: 11
1999
High single dose of mitoxantrone and cytarabine in acute non-lymphocytic leukemia: a pharmacokinetic and clinical study.
Therapeutic drug monitoring, Volume: 20, Issue: 6
1998
Influence of fludarabine on pharmacokinetics and pharmacodynamics of cytarabine: implications for a continuous infusion schedule.
Clinical cancer research : an official journal of the American Association for Cancer Research, Volume: 2, Issue: 4
1996
Pharmacokinetics of anticancer drugs in cerebrospinal fluid.
The Annals of pharmacotherapy, Volume: 32, Issue: 10
1998
Oral cytarabine ocfosfate in acute myeloid leukemia and non-Hodgkin's lymphoma--phase I/II studies and pharmacokinetics.
Leukemia, Volume: 12, Issue: 10
1998
Pharmacokinetics of nimustine, methotrexate, and cytosine arabinoside during cerebrospinal fluid perfusion chemotherapy in patients with disseminated brain tumors.
European journal of clinical pharmacology, Volume: 54, Issue: 5
1998
[A pharmacokinetic study of the value of oral cytarabine ocfosfate in the treatment of hematological malignancies].
[Rinsho ketsueki] The Japanese journal of clinical hematology, Volume: 39, Issue: 5
1998
Apoptosis- and necrosis-inducing potential of cladribine, cytarabine, cisplatin, and 5-fluorouracil in vitro: a quantitative pharmacodynamic model.
Cancer chemotherapy and pharmacology, Volume: 42, Issue: 1
1998
Pharmacokinetic and pharmacodynamic studies of fludarabine and cytosine arabinoside administered as loading boluses followed by continuous infusions after a phase I/II study in pediatric patients with relapsed leukemias. The Children's Cancer Group.
Clinical cancer research : an official journal of the American Association for Cancer Research, Volume: 4, Issue: 1
1998
Pharmacokinetics of high-dose cytarabine and its deamination product--a reappraisal.
Leukemia & lymphoma, Volume: 27, Issue: 3-4
1997
Pharmacokinetics of N4-octadecyl-1-beta-D-arabinofuranosylcytosine in plasma and whole blood after intravenous and oral administration to mice.
The Journal of pharmacy and pharmacology, Volume: 49, Issue: 11
1997
Intracellular pharmacodynamics of ara-C and flowcytometric analysis of cell cycle progression in leukemia chemotherapy.
Leukemia, Volume: 11 Suppl 3
1997
NONMEM population pharmacokinetic studies of cytosine arabinoside after high-dose and after loading bolus followed by continuous infusion of the drug in pediatric patients with leukemias.
Cancer chemotherapy and pharmacology, Volume: 39, Issue: 1-2
1996
Oral idarubicin pharmacokinetics--correlation of trough level with idarubicin area under curve.
Leukemia, Volume: 10, Issue: 4
1996
Chlorodeoxyadenosine and arabinosylcytosine in patients with acute myelogenous leukemia: pharmacokinetic, pharmacodynamic, and molecular interactions.
Blood, Jan-01, Volume: 87, Issue: 1
1996
Phase I clinical and pharmacokinetic evaluation of high-dose mitoxantrone in combination with cytarabine in patients with acute leukemia.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, Volume: 11, Issue: 10
1993
[Pharmacodynamics and action mechanism of antitumor agents].
Gan to kagaku ryoho. Cancer & chemotherapy, Volume: 20, Issue: 12
1993
Is there a relationship between cytarabine pharmacokinetics and keratitis?--A case report.
International journal of clinical pharmacology, therapy, and toxicology, Volume: 31, Issue: 12
1993
Clinical pharmacokinetics of anti-metabolites.
Cancer surveys, Volume: 17
1993
Plasma and leukemic cell pharmacokinetics of high-dose N4-behenoyl-1-beta-D-arabinofuranosylcytosine in acute leukemia patients.
Journal of clinical pharmacology, Volume: 34, Issue: 1
1994
Pharmacokinetic properties and interactions with blood components of N4-hexadecyl-1-beta-D-arabinofuranosylcytosine (NHAC) incorporated into liposomes.
The Journal of pharmacy and pharmacology, Volume: 47, Issue: 4
1995
Attenuated-dose idarubicin in acute myeloid leukaemia of the elderly: pharmacokinetic study and clinical results.
British journal of haematology, Volume: 90, Issue: 1
1995
Alkylsulphonic acid ion pairing with radial compression columns for determining plasma or cerebrospinal fluid 1-beta-D-arabinofuranosylcytosine in pediatric pharmacokinetic analysis.
Journal of chromatography. A, Feb-10, Volume: 692, Issue: 1-2
1995
Synthesis and pharmacokinetics of a new liver-specific carrier, glycosylated carboxymethyl-dextran, and its application to drug targeting.
Pharmaceutical research, Volume: 10, Issue: 9
1993
Pharmacokinetics of intralumbar DTC-101 for the treatment of leptomeningeal metastases.
Archives of neurology, Volume: 52, Issue: 9
1995
Pharmacokinetics of Ara-CMP-Stearate (YNK01): phase I study of the oral Ara-C derivative.
Leukemia, Volume: 9, Issue: 6
1995
Pharmacokinetics of mitoxantrone, etoposide and cytosine arabinoside in leukemic cells during treatment of acute myelogenous leukemia--relationship to treatment outcome and bone marrow toxicity.
Leukemia research, Volume: 19, Issue: 10
1995
Pharmacokinetic approach to in vitro testing of ovarian cancer cell sensitivity.
Oncology, Volume: 37, Issue: 3
1980
Clinical results and pharmacokinetics of high-dose cytosine arabinoside (HD ARA-C).
Cancer, Oct-01, Volume: 50, Issue: 7
1982
Pharmacokinetics and metabolism of cytosine arabinoside in the central nervous system.
The Journal of pharmacology and experimental therapeutics, Volume: 222, Issue: 1
1982
Pharmacokinetics of continuous intravenous and subcutaneous infusions of cytosine arabinoside.
Blood, Volume: 59, Issue: 6
1982
The pharmacokinetics of subcutaneous cytosine arabinoside in patients with acute myelogenous leukaemia.
British journal of clinical pharmacology, Volume: 12, Issue: 4
1981
Influence of tetrahydrouridine on the pharmacokinetics of intrathecally administered 1-beta-D-arabinofuranosylcytosine.
Cancer research, Volume: 42, Issue: 5
1982
High-dose cytarabine in acute leukemia: toxicity and pharmacokinetics.
Cancer treatment reports, Volume: 67, Issue: 4
1983
Pharmacokinetics of N4-behenoyl-1-beta-D-arabinofuranosylcytosine in patients with acute leukemia.
Cancer research, Volume: 43, Issue: 7
1983
Species similarities in pharmacokinetics.
Federation proceedings, Volume: 39, Issue: 1
1980
Acute doxorubicin toxicity. Relationship to pretreatment liver function, response, and pharmacokinetics in patients with acute nonlymphocytic leukemia.
Cancer, Mar-01, Volume: 53, Issue: 5
1984
Alteration of the pharmacokinetics of high-dose ara-C by its metabolite, high ara-U in patients with acute leukemia.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, Volume: 1, Issue: 12
1983
Cerebrospinal fluid and plasma pharmacokinetics of high doses of 1-beta-D-arabinofuranosylcytosine in nonhuman primates.
Cancer research, Volume: 43, Issue: 11
1983
Effect of dose and schedule on pharmacokinetics of high-dose cytosine arabinoside in plasma and cerebrospinal fluid.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, Volume: 1, Issue: 9
1983
[Pharmacokinetics of intrathecal chemotherapy and clinical problems].
Gan to kagaku ryoho. Cancer & chemotherapy, Volume: 11, Issue: 8
1984
[Chemical modification of anticancer agents from viewpoints of their pharmacokinetics].
Gan to kagaku ryoho. Cancer & chemotherapy, Volume: 11, Issue: 3 Pt 2
1984
Central nervous system pharmacokinetics of high-dose cytosine arabinoside.
Journal of neuro-oncology, Volume: 3, Issue: 2
1985
Pharmacokinetics of 1-beta-D-arabinofuranosylcytosine 5'-triphosphate in leukemic cells after intravenous and subcutaneous administration of 1-beta-D-arabinofuranosylcytosine.
Cancer research, Volume: 45, Issue: 5
1985
[High-dose cytarabine treatment in acute leukemias and leukemic meningiosis: clinical aspects and pharmacokinetics].
Onkologie, Volume: 8, Issue: 1
1985
Pharmacokinetics of low-dose 1-beta-D-arabinofuranosylcytosine given by continuous intravenous infusion over twenty-one days.
Cancer research, Volume: 45, Issue: 12 Pt 1
1985
Pharmacokinetics of the 5'-triphosphates of arabinosylcytosine and 2',2'-difluorodeoxycytidine in L1210 cells.
Nucleic acids symposium series, Issue: 18
1987
Pharmacokinetic studies of intermediate-to high-dose 1-beta-D-arabinofuranosylcytosine in children with acute leukemia and lymphoma.
Journal of clinical pharmacology, Volume: 27, Issue: 4
1987
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Bioavailability (37)

ArticleYear
The first marine dual-drug cocrystal of cytarabine with 5-fluorouracil having synergistic antitumor effects shows superior biopharmaceutical peculiarities by oral administration.
International journal of pharmaceutics, Dec-15, Volume: 629
2022
Venetoclax in combination with nucleoside analogs in acute myelogenous leukemia.
Current opinion in oncology, 09-01, Volume: 34, Issue: 5
2022
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Molecular pharmacology, Volume: 96, Issue: 5
2019
Self-nanoemulsifying system improves oral absorption and enhances anti-acute myeloid leukemia activity of berberine.
Journal of nanobiotechnology, Oct-05, Volume: 16, Issue: 1
2018
Nanosized ethanol based malleable liposomes of cytarabine to accentuate transdermal delivery: formulation optimization, in vitro skin permeation and in vivo bioavailability.
Artificial cells, nanomedicine, and biotechnology, Volume: 46, Issue: sup2
2018
The novel phospholipid mimetic KPC34 is highly active against preclinical models of Philadelphia chromosome positive acute lymphoblastic leukemia.
PloS one, Volume: 12, Issue: 6
2017
Mechanism-Based Drug Combinations with the DNA Strand-Breaking Nucleoside Analog CNDAC.
Molecular cancer therapeutics, Volume: 15, Issue: 10
2016
Transporter-targeted cholic acid-cytarabine conjugates for improved oral absorption.
International journal of pharmaceutics, Sep-10, Volume: 511, Issue: 1
2016
Lipid Nanosystems Enhance the Bioavailability and the Therapeutic Efficacy of FTY720 in Acute Myeloid Leukemia.
Journal of biomedical nanotechnology, Volume: 11, Issue: 4
2015
Entinostat, a novel histone deacetylase inhibitor is active in B-cell lymphoma and enhances the anti-tumour activity of rituximab and chemotherapy agents.
British journal of haematology, Volume: 169, Issue: 4
2015
Anti-leukemic effect of sodium metaarsenite (KML001) in acute myeloid leukemia with breaking-down the resistance of cytosine arabinoside.
International journal of oncology, Volume: 46, Issue: 5
2015
Sapacitabine, the prodrug of CNDAC, is a nucleoside analog with a unique action mechanism of inducing DNA strand breaks.
Chinese journal of cancer, Volume: 31, Issue: 8
2012
Development of cytarabine prodrugs and delivery systems for leukemia treatment.
Expert opinion on drug delivery, Volume: 7, Issue: 12
2010
Intrathecal liposomal cytarabine in relapsed or refractory infant and pediatric leukemias: the Children's Hospital of Philadelphia experience and review of the literature.
Journal of pediatric hematology/oncology, Volume: 32, Issue: 8
2010
Thioguanine pharmacokinetics in induction therapy of children with acute myeloid leukemia.
Anti-cancer drugs, Volume: 20, Issue: 1
2009
Synthesis and evaluation of anti-tumor activities of N4 fatty acyl amino acid derivatives of 1-beta-arabinofuranosylcytosine.
European journal of medicinal chemistry, Volume: 44, Issue: 9
2009
Pharmacokinetic characteristics of L-valyl-ara-C and its implication on the oral delivery of ara-C.
Acta pharmacologica Sinica, Volume: 28, Issue: 2
2007
Therapeutic drug monitoring of antimetabolic cytotoxic drugs.
British journal of clinical pharmacology, Volume: 47, Issue: 2
1999
Oral cytarabine ocfosfate in acute myeloid leukemia and non-Hodgkin's lymphoma--phase I/II studies and pharmacokinetics.
Leukemia, Volume: 12, Issue: 10
1998
Pharmacokinetics of N4-octadecyl-1-beta-D-arabinofuranosylcytosine in plasma and whole blood after intravenous and oral administration to mice.
The Journal of pharmacy and pharmacology, Volume: 49, Issue: 11
1997
Lipophilic 1-beta-D-arabinofuranosyl cytosine derivatives in liposomal formulations for oral and parenteral antileukemic therapy in the murine L1210 leukemia model.
Journal of cancer research and clinical oncology, Volume: 122, Issue: 12
1996
Oral idarubicin pharmacokinetics--correlation of trough level with idarubicin area under curve.
Leukemia, Volume: 10, Issue: 4
1996
Oral antitumour activity in murine L1210 leukaemia and pharmacological properties of liposome formulations of N4-alkyl derivatives of 1-beta-D-arabinofuranosylcytosine.
Journal of cancer research and clinical oncology, Volume: 122, Issue: 2
1996
[Pharmacodynamics and action mechanism of antitumor agents].
Gan to kagaku ryoho. Cancer & chemotherapy, Volume: 20, Issue: 12
1993
Intravenous mercaptopurine: life begins at 40.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, Volume: 11, Issue: 9
1993
Pharmacokinetics of Ara-CMP-Stearate (YNK01): phase I study of the oral Ara-C derivative.
Leukemia, Volume: 9, Issue: 6
1995
Biochemical pharmacological determinants of drug action in cancer therapeutics.
Oncology, Volume: 37 Suppl 1
1980
Cytostatic drugs are without significant effect on digitoxin plasma level and renal excretion.
Clinical pharmacology and therapeutics, Volume: 32, Issue: 5
1982
Acylcarnitines: drug absorption-enhancing agents in the gastrointestinal tract.
The American journal of physiology, Volume: 251, Issue: 3 Pt 1
1986
Enhanced serum concentrations of Ara-C using suppositories containing tetrahydrouridine as a deamination inhibitor of Ara-C.
The Journal of pharmacy and pharmacology, Volume: 38, Issue: 3
1986
Idarubicin: an anthracycline antineoplastic agent.
Clinical pharmacy, Volume: 11, Issue: 2
1992
[Clinical problems of optimum bioavailability, in particular in cytostatic therapy (author's transl)].
Arzneimittel-Forschung, Volume: 26, Issue: 1A
1976
Multiple basis of combination chemotherapy.
Cancer, Volume: 40, Issue: 1 Suppl
1977
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Dosage (430)

ArticleYear
Artesunate reverses cytarabine resistance in acute myeloid leukemia by blocking the JAK/STAT3 signaling.
Hematology (Amsterdam, Netherlands), Volume: 28, Issue: 1
2023
Synergistic interactions of cytarabine-adavosertib in leukemic cell lines proliferation and metabolomic endpoints.
Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, Volume: 166
2023
The first nano-cocrystal formulation of marine drug cytarabine with uracil based on cocrystal nanonization strategy for long-acting injection exhibiting enhanced antitumor activity.
International journal of pharmaceutics, Sep-25, Volume: 644
2023
Obesity in adult acute myeloid leukemia is not associated with inferior response or survival even when dose capping anthracyclines: An ECOG-ACRIN analysis.
Cancer, 08-15, Volume: 129, Issue: 16
2023
Intensification of treatment with vinca alkaloid does not improve outcomes in pediatric patients with Langerhans cell histiocytosis: results from the JPLSG LCH-12 study.
International journal of hematology, Volume: 118, Issue: 1
2023
Retrospective, real-life study of venetoclax plus azacitidine or low-dose cytarabine in French patients with acute myeloid leukemia ineligible for intensive chemotherapy.
Cancer medicine, Volume: 12, Issue: 6
2023
Utility of therapeutic drug monitoring of venetoclax in acute myeloid leukemia.
Medical oncology (Northwood, London, England), Oct-12, Volume: 39, Issue: 12
2022
Ofatumumab with iphosphamide, etoposide and cytarabine for patients with transplantation-ineligible relapsed and refractory diffuse large B-cell lymphoma.
British journal of haematology, Volume: 198, Issue: 1
2022
Clinical observation of low-dose combination chemotherapy in refractory/recurrent paroxysmal nocturnal hemoglobinuria patients: A single-center retrospective analysis.
Journal of clinical laboratory analysis, Volume: 36, Issue: 2
2022
Neutrophil decline rate following autologous transplant for lymphoma is a predictor of patients' outcome.
Leukemia & lymphoma, Volume: 63, Issue: 5
2022
Impact of induction chemotherapy with intermediate-dosed cytarabine and subsequent allogeneic stem cell transplantation on the outcome of high-risk acute myeloid leukemia.
Journal of cancer research and clinical oncology, Volume: 148, Issue: 6
2022
Comprehensive analysis of dose intensity of acute lymphoblastic leukemia chemotherapy.
Haematologica, 02-01, Volume: 107, Issue: 2
2022
A phase I trial of selinexor plus FLAG-Ida for the treatment of refractory/relapsed adult acute myeloid leukemia patients.
Annals of hematology, Volume: 100, Issue: 6
2021
Single daily dosing versus divided dosing intravenous ondansetron to prevent chemotherapy-induced nausea and vomiting among children: A comparative randomized double-blind controlled trial.
Pediatric blood & cancer, Volume: 68, Issue: 6
2021
Impact of high dose cytarabine dosing strategies in obese patients with acute myeloid leukemia.
Leukemia research, Volume: 102
2021
Intracellular Pharmacokinetics of Pyrimidine Analogues used in Oncology and the Correlation with Drug Action.
Clinical pharmacokinetics, Volume: 59, Issue: 12
2020
Midostaurin in acute myeloid leukemia: current evidence and practical considerations in routine clinical use.
Minerva medica, Volume: 111, Issue: 5
2020
Towards better combination regimens of cytarabine and FLT3 inhibitors in acute myeloid leukemia.
Cancer chemotherapy and pharmacology, Volume: 86, Issue: 3
2020
Liposomal encapsulated cytarabine and daunorubicin (CPX-351) for older patients with acute myeloid leukemia.
Leukemia & lymphoma, Volume: 61, Issue: 6
2020
Increased mitochondrial apoptotic priming with targeted therapy predicts clinical response to re-induction chemotherapy.
American journal of hematology, Volume: 95, Issue: 3
2020
Post-remission therapy in acute myeloid leukemia: Are we ready for an individualized approach?
Best practice & research. Clinical haematology, Volume: 32, Issue: 4
2019
Liposomal Cytarabine as Cancer Therapy: From Chemistry to Medicine.
Biomolecules, 11-23, Volume: 9, Issue: 12
2019
Simultaneous determination of cytosine arabinoside and its metabolite uracil arabinoside in human plasma by LC-MS/MS: Application to pharmacokinetics-pharmacogenetics pilot study in AML patients.
Journal of chromatography. B, Analytical technologies in the biomedical and life sciences, Sep-15, Volume: 1126-1127
2019
Cerebrospinal Fluid Drug Concentrations and Clinical Outcome of Patients with Neoplastic Meningitis Treated with Liposomal Cytarabine.
European journal of drug metabolism and pharmacokinetics, Volume: 44, Issue: 6
2019
Film interface for drug testing for delivery to cells in culture and in the brain.
Acta biomaterialia, Volume: 94
2019
Homoharringtonine is a safe and effective substitute for anthracyclines in children younger than 2 years old with acute myeloid leukemia.
Frontiers of medicine, Volume: 13, Issue: 3
2019
Successful nivolumab therapy in an allogeneic stem cell transplant child with post-transplant lymphoproliferative disorder.
Pediatric transplantation, Volume: 22, Issue: 8
2018
Upregulated microRNA-146a expression induced by granulocyte colony-stimulating factor enhanced low-dosage chemotherapy response in aged acute myeloid leukemia patients.
Experimental hematology, Volume: 68
2018
Toward Predicting Acute Myeloid Leukemia Patient Response to 7 + 3 Induction Chemotherapy via Diagnostic Microdosing.
Chemical research in toxicology, 10-15, Volume: 31, Issue: 10
2018
UTX is an escape from X-inactivation tumor-suppressor in B cell lymphoma.
Nature communications, 07-13, Volume: 9, Issue: 1
2018
Analysis of the prolonged infusion of DFP-10917, a deoxycytidine analog, as a therapeutic strategy for the treatment of human tumor xenografts in vivo.
International journal of oncology, Volume: 52, Issue: 3
2018
Liposomal Cytarabine Induces Less Neurocognitive Dysfunction Than Intrathecal Methotrexate in an Animal Model.
Journal of pediatric hematology/oncology, Volume: 40, Issue: 2
2018
Improved outcomes for myeloid leukemia of Down syndrome: a report from the Children's Oncology Group AAML0431 trial.
Blood, 06-22, Volume: 129, Issue: 25
2017
Phase I Trial of the Human Double Minute 2 Inhibitor MK-8242 in Patients With Advanced Solid Tumors.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, Apr-20, Volume: 35, Issue: 12
2017
Combination screening in vitro identifies synergistically acting KP372-1 and cytarabine against acute myeloid leukemia.
Biochemical pharmacology, Oct-15, Volume: 118
2016
A phase I trial of the human double minute 2 inhibitor (MK-8242) in patients with refractory/recurrent acute myelogenous leukemia (AML).
Leukemia research, Volume: 48
2016
Retrospective evaluation of the MEAM regimen as a conditioning regimen before autologous peripheral blood stem cell transplantation for lymphoma in two centers with different dosing schedules of melphalan.
Annals of hematology, Volume: 95, Issue: 9
2016
Effect of a constant rate infusion of cytosine arabinoside on mortality in dogs with meningoencephalitis of unknown origin.
Veterinary journal (London, England : 1997), Volume: 213
2016
Intensified and prolonged therapy comprising cytarabine, vincristine and prednisolone improves outcome in patients with multisystem Langerhans cell histiocytosis: results of the Japan Langerhans Cell Histiocytosis Study Group-02 Protocol Study.
International journal of hematology, Volume: 104, Issue: 1
2016
[Long-term outcomes of homoharringtonine, cytarabine, daunorubicin or idarubicin (HAD/HAI) as induction chemotherapy in de novo acute myeloid leukemia].
Zhonghua xue ye xue za zhi = Zhonghua xueyexue zazhi, Volume: 37, Issue: 2
2016
Synergisitic and Antagonistic AML Cell Type-specific Responses to 5-Aza-2-deoxycitidine and 1-h-D-Arabinofuranoside.
Anticancer research, Volume: 36, Issue: 2
2016
Chemoprotection of murine hematopoietic cells by combined gene transfer of cytidine deaminase (CDD) and multidrug resistance 1 gene (MDR1).
Journal of experimental & clinical cancer research : CR, Dec-12, Volume: 34
2015
Redirection of CD4+ and CD8+ T lymphocytes via an anti-CD3 × anti-CD19 bi-specific antibody combined with cytosine arabinoside and the efficient lysis of patient-derived B-ALL cells.
Journal of hematology & oncology, Oct-06, Volume: 8
2015
Increasing aclarubicin dosage of the conventional CAG (low-dose cytarabine and aclarubicin in combination with granulocyte colony-stimulating factor) regimen is more efficacious as a salvage therapy than CAG for relapsed/refractory acute myeloid leukemia.
Leukemia research, Volume: 39, Issue: 12
2015
Chemotherapy options for previously untreated acute myeloid leukemia.
Expert opinion on pharmacotherapy, Volume: 16, Issue: 14
2015
Outcomes for newly diagnosed patients with acute myeloid leukemia dosed on actual or adjusted body weight.
Cancer chemotherapy and pharmacology, Volume: 76, Issue: 4
2015
Guidelines for the diagnosis and management of acute myeloid leukaemia in pregnancy.
British journal of haematology, Volume: 170, Issue: 4
2015
Randomised Introduction of 2-CDA as Intensification during Consolidation for Children with High-risk AML--results from Study AML-BFM 2004.
Klinische Padiatrie, Volume: 227, Issue: 3
2015
Randomized trial of radiation-free central nervous system prophylaxis comparing intrathecal triple therapy with liposomal cytarabine in acute lymphoblastic leukemia.
Haematologica, Volume: 100, Issue: 6
2015
Myelopathy following intrathecal chemotherapy in adults: a single institution experience.
Journal of neuro-oncology, Volume: 122, Issue: 2
2015
High-dose chemotherapy with carmustine, etoposide, cytarabine and melphalan followed by autologous stem cell transplant is an effective treatment for elderly patients with poor-prognosis lymphoma.
Leukemia & lymphoma, Volume: 56, Issue: 8
2015
Impact of single nucleotide polymorphisms of cytarabine metabolic genes on drug toxicity in childhood acute lymphoblastic leukemia.
Pediatric blood & cancer, Volume: 62, Issue: 4
2015
Minimal residual disease as biomarker for optimal biologic dosing of ARA-C in patients with acute myeloid leukemia.
American journal of hematology, Volume: 90, Issue: 2
2015
Acute promyelocytic leukemia in patients aged >70 years: the cure beyond the age.
Annals of hematology, Volume: 94, Issue: 2
2015
Clinical efficacy of mitoxantrone and Ara-C with or without etoposide salvage chemotherapy in adult patients with relapsed or refractory acute lymphoblastic leukemia: retrospective multicenter study of the Korean Adult ALL Working Party.
Acta haematologica, Volume: 133, Issue: 1
2015
Recipient/donor contradictory genotypes with impact on drug pharmacogenetics after liver transplant: a deadly gift?
Pharmacogenetics and genomics, Volume: 24, Issue: 10
2014
Selection of the best blood compartment to measure cytidine deaminase activity to stratify for optimal gemcitabine or cytarabine treatment.
Nucleosides, nucleotides & nucleic acids, Volume: 33, Issue: 4-6
2014
Toxicity and efficacy of intrathecal liposomal cytarabine in children with leukemia/lymphoma relapsing in the central nervous system: a retrospective multicenter study.
Leukemia & lymphoma, Volume: 56, Issue: 3
2015
Optimal therapy for adult patients with acute myeloid leukemia in first complete remission.
Current treatment options in oncology, Volume: 15, Issue: 2
2014
Combined treatment of rituximab, idarubicin, dexamethasone, cytarabine, methotrexate with radiotherapy for primary central nervous system lymphoma.
Journal of cellular and molecular medicine, Volume: 18, Issue: 6
2014
Dose intensification of methotrexate and cytarabine during intensified continuation chemotherapy for high-risk B-precursor acute lymphoblastic leukemia: POG 9406: a report from the Children's Oncology Group.
Journal of pediatric hematology/oncology, Volume: 36, Issue: 5
2014
Prospective, multicenter, phase II study on reducing the dosage of idarubicin and FLAG for patients younger than 65 years with resistant acute myeloid leukemia: a comparison with a higher dosage trial.
Acta haematologica, Volume: 132, Issue: 1
2014
Phase I study of cladribine, cytarabine, granulocyte colony stimulating factor (CLAG regimen) and midostaurin and all-trans retinoic acid in relapsed/refractory AML.
International journal of hematology, Volume: 99, Issue: 3
2014
Conditioning chemotherapy dose adjustment in obese patients: a review and position statement by the American Society for Blood and Marrow Transplantation practice guideline committee.
Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation, Volume: 20, Issue: 5
2014
High-dose cytarabine in induction treatment improves the outcome of adult patients younger than age 46 years with acute myeloid leukemia: results of the EORTC-GIMEMA AML-12 trial.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, Jan-20, Volume: 32, Issue: 3
2014
Pharmacokinetics and toxicity of intrathecal liposomal cytarabine in children and adolescents following age-adapted dosing.
Clinical pharmacokinetics, Volume: 53, Issue: 2
2014
Outcomes in obese and overweight acute myeloid leukemia patients receiving chemotherapy dosed according to actual body weight.
American journal of hematology, Volume: 88, Issue: 10
2013
A phase I study of CPX-351 in combination with busulfan and fludarabine conditioning and allogeneic stem cell transplantation in adult patients with refractory acute leukemia.
Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation, Volume: 19, Issue: 7
2013
Successful large-volume cerebrospinal fluid aspiration for an accidental overdose of intrathecal cytarabine.
Medical oncology (Northwood, London, England), Volume: 30, Issue: 2
2013
Two dosing regimens of tosedostat in elderly patients with relapsed or refractory acute myeloid leukaemia (OPAL): a randomised open-label phase 2 study.
The Lancet. Oncology, Volume: 14, Issue: 4
2013
Fatal cisplatin overdose in the treatment of mediastinal lymphoma with the ESHAP regimen - analysis of the causes of the adverse drug event.
Onkologie, Volume: 36, Issue: 1-2
2013
Safety and pharmacokinetics of the antisense oligonucleotide (ASO) LY2181308 as a single-agent or in combination with idarubicin and cytarabine in patients with refractory or relapsed acute myeloid leukemia (AML).
Investigational new drugs, Volume: 31, Issue: 4
2013
A phase Ib GOELAMS study of the mTOR inhibitor RAD001 in association with chemotherapy for AML patients in first relapse.
Leukemia, Volume: 27, Issue: 7
2013
Phase I and pharmacologic trial of cytosine arabinoside with the selective checkpoint 1 inhibitor Sch 900776 in refractory acute leukemias.
Clinical cancer research : an official journal of the American Association for Cancer Research, Dec-15, Volume: 18, Issue: 24
2012
Successful importation of cytarabine into the United States during a critical national drug shortage.
American journal of health-system pharmacy : AJHP : official journal of the American Society of Health-System Pharmacists, Aug-15, Volume: 69, Issue: 16
2012
Pharmacokinetics of CPX-351; a nano-scale liposomal fixed molar ratio formulation of cytarabine:daunorubicin, in patients with advanced leukemia.
Leukemia research, Volume: 36, Issue: 10
2012
Dose intensification of daunorubicin and cytarabine during treatment of adult acute lymphoblastic leukemia: results of Cancer and Leukemia Group B Study 19802.
Cancer, Jan-01, Volume: 119, Issue: 1
2013
A facile whole-cell biocatalytic approach to regioselective synthesis of monoacylated 1-β-D-arabinofuranosylcytosine: influence of organic solvents.
Bioresource technology, Volume: 114
2012
Sterilization stability of vesicular phospholipid gels loaded with cytarabine for brain implant.
International journal of pharmaceutics, May-10, Volume: 427, Issue: 2
2012
Therapeutic effects of combination of paeoniflorin and albiflorin from Paeonia radix on radiation and chemotherapy-induced myelosuppression in mice and rabbits.
Asian Pacific journal of cancer prevention : APJCP, Volume: 12, Issue: 8
2011
In vivo enrichment of cytidine deaminase gene-modified hematopoietic cells by prolonged cytosine-arabinoside application.
Cytotherapy, Volume: 14, Issue: 4
2012
Mitoxantrone and etoposide with or without intermediate dose cytarabine for the treatment of primary induction failure or relapsed acute myeloid leukemia.
Leukemia research, Volume: 36, Issue: 4
2012
FDA-approved drug labeling for the study of drug-induced liver injury.
Drug discovery today, Volume: 16, Issue: 15-16
2011
Influence of anticancer therapy on oxidation phenotype and acetylation phenotype in patients with acute myeloblastic leukemia.
Pharmacological reports : PR, Volume: 63, Issue: 1
2011
Cytarabine dose for acute myeloid leukemia.
The New England journal of medicine, Mar-17, Volume: 364, Issue: 11
2011
Treatment of children with B-cell non-Hodgkin lymphoma in a low-income country.
Pediatric blood & cancer, Volume: 56, Issue: 4
2011
Phase 1 and pharmacokinetic study of bolus-infusion flavopiridol followed by cytosine arabinoside and mitoxantrone for acute leukemias.
Blood, Mar-24, Volume: 117, Issue: 12
2011
Development of cytarabine prodrugs and delivery systems for leukemia treatment.
Expert opinion on drug delivery, Volume: 7, Issue: 12
2010
Intrathecal liposomal cytarabine in relapsed or refractory infant and pediatric leukemias: the Children's Hospital of Philadelphia experience and review of the literature.
Journal of pediatric hematology/oncology, Volume: 32, Issue: 8
2010
A phase 1-2 study of a farnesyltransferase inhibitor, tipifarnib, combined with idarubicin and cytarabine for patients with newly diagnosed acute myeloid leukemia and high-risk myelodysplastic syndrome.
Cancer, Mar-15, Volume: 117, Issue: 6
2011
Prodrugs of aza nucleosides based on proton transfer reaction.
Journal of computer-aided molecular design, Volume: 24, Issue: 12
2010
Dose modification of alemtuzumab in combination with dexamethasone, cytarabine, and cisplatin in patients with relapsed or refractory peripheral T-cell lymphoma: analysis of efficacy and toxicity.
Investigational new drugs, Volume: 30, Issue: 1
2012
Effective clearance of Ara-U the major metabolite of cytosine arabinoside (Ara-C) by hemodialysis in a patient with lymphoma and end-stage renal failure.
Cancer chemotherapy and pharmacology, Volume: 67, Issue: 4
2011
Schedule- and dose-dependency of CPX-351, a synergistic fixed ratio cytarabine:daunorubicin formulation, in consolidation treatment against human leukemia xenografts.
Leukemia & lymphoma, Volume: 51, Issue: 8
2010
Pegfilgrastim appears equivalent to daily dosing of filgrastim to treat neutropenia after autologous peripheral blood stem cell transplantation in patients with non-Hodgkin lymphoma.
Clinical lymphoma, myeloma & leukemia, Volume: 10, Issue: 3
2010
[Efficiency of GHA priming therapy on patients with acute monocytic leukemia and its mechanism].
Zhongguo shi yan xue ye xue za zhi, Volume: 18, Issue: 1
2010
Methotrexate area under the curve is an important outcome predictor in patients with primary CNS lymphoma: A pharmacokinetic-pharmacodynamic analysis from the IELSG no. 20 trial.
British journal of cancer, Feb-16, Volume: 102, Issue: 4
2010
Efficacy of escalated imatinib combined with cytarabine in newly diagnosed patients with chronic myeloid leukemia.
Haematologica, Volume: 95, Issue: 6
2010
Retinoblastoma CSF metastasis cured by multimodality chemotherapy without radiation.
Ophthalmic genetics, Volume: 30, Issue: 3
2009
Methylene tetrahydrofolate reductase gene polymorphism in Egyptian children with acute lymphoblastic leukemia.
Blood coagulation & fibrinolysis : an international journal in haemostasis and thrombosis, Volume: 21, Issue: 1
2010
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Interactions (240)

ArticleYear
Safety and efficacy of FLAG-Ida-based therapy combined with venetoclax for the treatment for newly diagnosed and relapsed/refractory patients with AML - A systematic review.
Leukemia research, Volume: 133
2023
[Efficacy and Safety of Decitabine Combined with Modified CAG Regimen in Patients Aged ≥ 70 Years with Newly Diagnosed Acute Myeloid Leukemia].
Zhongguo shi yan xue ye xue za zhi, Volume: 31, Issue: 3
2023
Intensified conditioning regimen with fludarabine combined with post-transplantation cyclophosphamide for haploidentical allogeneic hematopoietic stem cell transplantation in children with high-risk acute leukemia.
Hematology (Amsterdam, Netherlands), Volume: 28, Issue: 1
2023
Cellular Composition and 5hmC Signature Predict the Treatment Response of AML Patients to Azacitidine Combined with Chemotherapy.
Advanced science (Weinheim, Baden-Wurttemberg, Germany), Volume: 10, Issue: 23
2023
Retrospective comparison of survival and responses to Fludarabine, Cytarabine, GCSF (FLAG) in combination with gemtuzumab ozogamicin (GO) or Idarubicin (IDA) in patients with newly diagnosed core binding factor (CBF) acute myelogenous leukemia: MD Anderso
American journal of hematology, Volume: 97, Issue: 11
2022
Venetoclax in combination with nucleoside analogs in acute myelogenous leukemia.
Current opinion in oncology, 09-01, Volume: 34, Issue: 5
2022
Venetoclax in combination with FLAG-IDA-based protocol for patients with acute myeloid leukemia: a real-world analysis.
Annals of hematology, Volume: 101, Issue: 8
2022
Venetoclax combined with FLAG-IDA induction and consolidation in newly diagnosed acute myeloid leukemia.
American journal of hematology, Volume: 97, Issue: 8
2022
Whole brain radiotherapy combined with intrathecal liposomal cytarabine for leptomeningeal metastasis-a safety analysis and validation of the EANO-ESMO classification.
Strahlentherapie und Onkologie : Organ der Deutschen Rontgengesellschaft ... [et al], Volume: 198, Issue: 5
2022
Venetoclax in combination with hypomethylating agents or low dose cytarabine for relapsed and refractory acute myeloid leukemia.
Leukemia & lymphoma, Volume: 63, Issue: 7
2022
Chidamide and Decitabine in Combination with a HAG Priming Regimen for Acute Myeloid Leukemia with TP53 Mutation.
Acta medica Okayama, Volume: 76, Issue: 1
2022
Venetoclax combined with hypomethylating agents or low-dose cytarabine as induction chemotherapy for patients with untreated acute myeloid leukemia ineligible for intensive chemotherapy: a systematic review and meta-analysis.
Clinical and experimental medicine, Volume: 23, Issue: 2
2023
Successful use of trametinib and dasatinib combined with chemotherapy in the treatment of Ph-positive B-cell acute lymphoblastic leukemia: A case report.
Medicine, Jun-25, Volume: 100, Issue: 25
2021
Venetoclax Combined With FLAG-IDA Induction and Consolidation in Newly Diagnosed and Relapsed or Refractory Acute Myeloid Leukemia.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 09-01, Volume: 39, Issue: 25
2021
Role of Roflumilast Combined with ESHAP Chemotherapy in Relapsed/Refractory Patients with Diffuse Large B-Cell Lymphoma.
Cancer research and treatment, Volume: 54, Issue: 1
2022
Long-term results of low-intensity chemotherapy with clofarabine or cladribine combined with low-dose cytarabine alternating with decitabine in older patients with newly diagnosed acute myeloid leukemia.
American journal of hematology, 08-01, Volume: 96, Issue: 8
2021
Decitabine combined with medium-dose cytarabine in the treatment of DEK/CAN-positive acute myeloid leukemia: a case report.
Annals of palliative medicine, Volume: 10, Issue: 4
2021
Iron Oxide Nanoparticles Combined with Cytosine Arabinoside Show Anti-Leukemia Stem Cell Effects on Acute Myeloid Leukemia by Regulating Reactive Oxygen Species.
International journal of nanomedicine, Volume: 16
2021
[Comparison of the Curative Efficacy of Elderly Patients with High-Risk MDS and MDS-Transformed AML between Decitabine Combined with Low-Dose CEG Regimen and Decitabine Combined with Low-Dose CAG Regimen].
Zhongguo shi yan xue ye xue za zhi, Volume: 28, Issue: 6
2020
BL-8040 CXCR4 antagonist is safe and demonstrates antileukemic activity in combination with cytarabine for the treatment of relapsed/refractory acute myelogenous leukemia: An open-label safety and efficacy phase 2a study.
Cancer, 04-15, Volume: 127, Issue: 8
2021
Venetoclax + hypomethylating agents combined with dose-adjusted HAG for relapsed/refractory acute myeloid leukemia: Two case reports.
Medicine, Nov-20, Volume: 99, Issue: 47
2020
The effects of cytarabine combined with ginsenoside compound K synergistically induce DNA damage in acute myeloid leukemia cells.
Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, Volume: 132
2020
[Efficacy of Small Dose HAG Regimen Combined with Decitabine in Treatment of Elderly Patients with Acute Myeloid Leukemia].
Zhongguo shi yan xue ye xue za zhi, Volume: 28, Issue: 3
2020
The efficacy and safety of daunorubicin versus idarubicin combined with cytarabine for induction therapy in acute myeloid leukemia: A meta-analysis of randomized clinical trials.
Medicine, Jun-12, Volume: 99, Issue: 24
2020
Etoposide Combined with FLAG Salvage Therapy Is Effective in Multiple Relapsed/Refractory Acute Myeloid leukemia.
Acta haematologica, Volume: 143, Issue: 5
2020
Phase I Clinical Trial of Selinexor in Combination with Daunorubicin and Cytarabine in Previously Untreated Poor-Risk Acute Myeloid Leukemia.
Clinical cancer research : an official journal of the American Association for Cancer Research, 01-01, Volume: 26, Issue: 1
2020
A novel alkylating deacetylase inhibitor molecule EDO-S101 in combination with cytarabine synergistically enhances apoptosis of acute myeloid leukemia cells.
Medical oncology (Northwood, London, England), Aug-01, Volume: 36, Issue: 9
2019
Optimizing venetoclax dose in combination with low intensive therapies in elderly patients with newly diagnosed acute myeloid leukemia: An exposure-response analysis.
Hematological oncology, Volume: 37, Issue: 4
2019
Clinical Experience With Ibrutinib Alone or in Combination With Either Cytarabine or Azacitidine in Patients With Acute Myeloid Leukemia.
Clinical lymphoma, myeloma & leukemia, Volume: 19, Issue: 8
2019
[Efficacy and Safety of Decitabine Combined with CAG (Cytarabine, Aclarubicin, G-CSF) for Patients with Intermediate or High Risk Myelodysplastic Syndrome and Acute Myeloid Leukemia: a Meta-Analysis].
Zhongguo shi yan xue ye xue za zhi, Volume: 27, Issue: 2
2019
A Phase I/II Trial of MEC (Mitoxantrone, Etoposide, Cytarabine) in Combination with Ixazomib for Relapsed Refractory Acute Myeloid Leukemia.
Clinical cancer research : an official journal of the American Association for Cancer Research, 07-15, Volume: 25, Issue: 14
2019
Venetoclax Combined With Low-Dose Cytarabine for Previously Untreated Patients With Acute Myeloid Leukemia: Results From a Phase Ib/II Study.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 05-20, Volume: 37, Issue: 15
2019
Emodin and Its Combination with Cytarabine Induce Apoptosis in Resistant Acute Myeloid Leukemia Cells in Vitro and in Vivo.
Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology, Volume: 48, Issue: 5
2018
A phase 1 trial evaluating thioridazine in combination with cytarabine in patients with acute myeloid leukemia.
Blood advances, 08-14, Volume: 2, Issue: 15
2018
Comparison of Mitoxantrone in Combination with Intermediate-dose Cytarabine versus High-dose Cytarabine as Consolidation Therapies for Young Non-APL Acute Myeloid Leukemia Patients with Favorable and Intermediate Cytogenetics.
Current medical science, Volume: 38, Issue: 1
2018
Thalidomide in Combination with Chemotherapy in Treating Elderly Patients with Acute Myeloid Leukemia.
Oncology research and treatment, Volume: 41, Issue: 7-8
2018
Preclinical characterization of INCB053914, a novel pan-PIM kinase inhibitor, alone and in combination with anticancer agents, in models of hematologic malignancies.
PloS one, Volume: 13, Issue: 6
2018
Decitabine in combination with G-CSF, low-dose cytarabine and aclarubicin is as effective as standard dose chemotherapy in the induction treatment for patients aged from 55 to 69 years old with newly diagnosed acute myeloid leukemia.
Leukemia & lymphoma, Volume: 59, Issue: 11
2018
Report of the relapsed/refractory cohort of SWOG S0919: A phase 2 study of idarubicin and cytarabine in combination with pravastatin for acute myelogenous leukemia (AML).
Leukemia research, Volume: 67
2018
[Clinical Efficacy of Decitabine Combined with or without Cytarabine-based Low Dose Regimen for Senile patients with Acute Myeloid Leukemia].
Zhongguo shi yan xue ye xue za zhi, Volume: 26, Issue: 1
2018
[Curative Effect of Decitabine Combined with IAG Regimen for Senile Patients with Myelodysplastic Syndrome (MDS) Transformed Acute Myeloid Leukemia].
Zhongguo shi yan xue ye xue za zhi, Volume: 25, Issue: 6
2017
Midostaurin in Combination With Standard Chemotherapy for Treatment of Newly Diagnosed FMS-Like Tyrosine Kinase 3 (FLT3) Mutation-Positive Acute Myeloid Leukemia.
The Annals of pharmacotherapy, Volume: 52, Issue: 4
2018
Targeting BRCA1/2 deficient ovarian cancer with CNDAC-based drug combinations.
Cancer chemotherapy and pharmacology, Volume: 81, Issue: 2
2018
[Clinical Efficacy of Low-dose Decitabine Combined with CAG Regimen in Patients with Myelodysplastic Syndrome-refractory Anemia with Excess Blasts(MDS-RAEB)].
Zhongguo shi yan xue ye xue za zhi, Volume: 25, Issue: 5
2017
A Phase II Study of CLAG Regimen Combined With Imatinib Mesylate for Relapsed or Refractory Acute Myeloid Leukemia.
Clinical lymphoma, myeloma & leukemia, Volume: 17, Issue: 12
2017
Population Pharmacokinetics of Volasertib Administered in Patients with Acute Myeloid Leukaemia as a Single Agent or in Combination with Cytarabine.
Clinical pharmacokinetics, Volume: 57, Issue: 3
2018
A phase 1 study of the CXCR4 antagonist plerixafor in combination with high-dose cytarabine and etoposide in children with relapsed or refractory acute leukemias or myelodysplastic syndrome: A Pediatric Oncology Experimental Therapeutics Investigators' Co
Pediatric blood & cancer, Volume: 64, Issue: 8
2017
Decitabine before Low-Dose Cytarabine-Based Chemotherapy Combined with Human Leukocyte Antigen-Mismatched Stem Cell Microtransplantation Improved Outcomes in Elderly Patients with Newly Diagnosed Acute Myeloid Leukemia.
Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation, Volume: 23, Issue: 5
2017
Fractionated gemtuzumab ozogamicin combined with intermediate-dose cytarabine and daunorubicin as salvage therapy in very high-risk AML patients: a bridge to reduced intensity conditioning transplant?
Annals of hematology, Volume: 96, Issue: 3
2017
A Phase I Dose Escalation Study of the Triple Angiokinase Inhibitor Nintedanib Combined with Low-Dose Cytarabine in Elderly Patients with Acute Myeloid Leukemia.
PloS one, Volume: 11, Issue: 10
2016
Homoharringtonine combined with aclarubicin and cytarabine synergistically induces apoptosis in t(8;21) leukemia cells and triggers caspase-3-mediated cleavage of the AML1-ETO oncoprotein.
Cancer medicine, Volume: 5, Issue: 11
2016
Effect of granulocyte colony-stimulating factor priming combined with low-dose cytarabine and homoharringtonine in higher risk myelodysplastic syndrome patients.
Leukemia research, Volume: 48
2016
Mechanism-Based Drug Combinations with the DNA Strand-Breaking Nucleoside Analog CNDAC.
Molecular cancer therapeutics, Volume: 15, Issue: 10
2016
[Efficacy Comparison of Low dose Thalidomide Combined with Modified VCMP and VAD regimens for Treatment of Aged MM Patients].
Zhongguo shi yan xue ye xue za zhi, Volume: 24, Issue: 3
2016
[Clinical Efficacy of Sorafenib Combined with Low Dose Cytarabine for Treating Patients with FLT3+ Relapsed and Refractory Acute Myeloid Leukemia].
Zhongguo shi yan xue ye xue za zhi, Volume: 24, Issue: 2
2016
Binding of Released Bim to Mcl-1 is a Mechanism of Intrinsic Resistance to ABT-199 which can be Overcome by Combination with Daunorubicin or Cytarabine in AML Cells.
Clinical cancer research : an official journal of the American Association for Cancer Research, Sep-01, Volume: 22, Issue: 17
2016
[Modified Shengma Biejia Decoction Combined with CAG Program for Elderly Acute Myeloid Leuke- mia Patients with Yin Deficiency Toxin Stasis Syndrome].
Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi jiehe zazhi = Chinese journal of integrated traditional and Western medicine, Volume: 36, Issue: 2
2016
A phase I study of intermediate dose cytarabine in combination with lenalidomide in relapsed/refractory acute myeloid leukemia.
Leukemia research, Volume: 43
2016
Clinical assessment of 252Californium neutron intracavitary brachytherapy using a two-channel Y applicator combined with external beam radiotherapy for endometrial cancer.
Clinics (Sao Paulo, Brazil), Volume: 71, Issue: 1
2016
Lenalidomide in combination with R-ESHAP in patients with relapsed or refractory diffuse large B-cell lymphoma: a phase 1b study from GELTAMO group.
British journal of haematology, Volume: 173, Issue: 2
2016
[Outcomes of refractory or relapsed DNMT3A + cytogenetically normal acute myeloid leukemia patients followed the therapy including decitabine combined with CAG or CAG-like regimen].
Zhonghua xue ye xue za zhi = Zhonghua xueyexue zazhi, Volume: 36, Issue: 12
2015
[Clinical Analysis of 4 AML Patients Aged Over 80 Years Treated with Chemotherapy Combined with Haploidentical Hematopoietic Stem Cell Infusion].
Zhongguo shi yan xue ye xue za zhi, Volume: 23, Issue: 6
2015
Redirection of CD4+ and CD8+ T lymphocytes via an anti-CD3 × anti-CD19 bi-specific antibody combined with cytosine arabinoside and the efficient lysis of patient-derived B-ALL cells.
Journal of hematology & oncology, Oct-06, Volume: 8
2015
Decitabine in the Treatment of Acute Myeloid Leukemia and Myelodysplastic Syndromes, Which Combined with Complex Karyotype Respectively.
Asian Pacific journal of cancer prevention : APJCP, Volume: 16, Issue: 15
2015
Increasing aclarubicin dosage of the conventional CAG (low-dose cytarabine and aclarubicin in combination with granulocyte colony-stimulating factor) regimen is more efficacious as a salvage therapy than CAG for relapsed/refractory acute myeloid leukemia.
Leukemia research, Volume: 39, Issue: 12
2015
Idarubicin and cytarabine in combination with gemtuzumab ozogamicin (IAGO) for untreated patients with high-risk MDS or AML evolved from MDS: a phase II study from the EORTC and GIMEMA Leukemia Groups (protocol 06013).
Annals of hematology, Volume: 94, Issue: 12
2015
[Clinical Efficacy of Decitabine Combined with CAG Regimen for Myelodysplastic Syndrome-RAEB and Refractory Acute Myeloid Leukemia].
Zhongguo shi yan xue ye xue za zhi, Volume: 23, Issue: 4
2015
Efficacy of an Fc-modified anti-CD123 antibody (CSL362) combined with chemotherapy in xenograft models of acute myelogenous leukemia in immunodeficient mice.
Haematologica, Volume: 100, Issue: 7
2015
Comparison of consolidation strategies in acute myeloid leukemia: high-dose cytarabine alone versus intermediate-dose cytarabine combined with anthracyclines.
Annals of hematology, Volume: 94, Issue: 9
2015
The HSP90 inhibitor ganetespib: A potential effective agent for Acute Myeloid Leukemia in combination with cytarabine.
Leukemia research, Volume: 39, Issue: 6
2015
Oral Debio1143 (AT406), an antagonist of inhibitor of apoptosis proteins, combined with daunorubicin and cytarabine in patients with poor-risk acute myeloid leukemia--results of a phase I dose-escalation study.
Clinical lymphoma, myeloma & leukemia, Volume: 15, Issue: 7
2015
Efficacy and safety of decitabine in combination with G-CSF, low-dose cytarabine and aclarubicin in newly diagnosed elderly patients with acute myeloid leukemia.
Oncotarget, Mar-20, Volume: 6, Issue: 8
2015
[Clinical resarch of decitabine combined with modified CAG regimen for treatment of relapsed or refractory acute myeloid leukemia].
Zhongguo shi yan xue ye xue za zhi, Volume: 23, Issue: 1
2015
Phase I dose-escalation trial of checkpoint kinase 1 inhibitor MK-8776 as monotherapy and in combination with gemcitabine in patients with advanced solid tumors.
Journal of clinical oncology : official journal of the American Society of Clinical Oncology, Mar-20, Volume: 33, Issue: 9
2015
Retrospective comparison of fludarabine in combination with intermediate-dose cytarabine versus high-dose cytarabine as consolidation therapies for acute myeloid leukemia.
Medicine, Volume: 93, Issue: 27
2014
[The clinical efficacy of the patients of acute myeloid leukemia and myelodysplastic syndromes treated with decitabine alone, combined with half or one couse of CAG regimen].
Zhonghua xue ye xue za zhi = Zhonghua xueyexue zazhi, Volume: 35, Issue: 11
2014
A phase 1b/2 study of vosaroxin in combination with cytarabine in patients with relapsed or refractory acute myeloid leukemia.
Haematologica, Volume: 100, Issue: 2
2015
[Comparison of clinical efficacy between decitabine combined with CAG regimen and CAG regimen alone in patients with intermediate to high-risk myelodysplastic syndromes].
Zhongguo shi yan xue ye xue za zhi, Volume: 22, Issue: 5
2014
[Clinical efficacy of decitabine combined with modified CAG regimen for relapsed-refractory acute myeloid leukemia with AML1-ETO⁺].
Zhongguo shi yan xue ye xue za zhi, Volume: 22, Issue: 5
2014
SWOG0919: a Phase 2 study of idarubicin and cytarabine in combination with pravastatin for relapsed acute myeloid leukaemia.
British journal of haematology, Volume: 167, Issue: 2
2014
A Phase 2 study of bortezomib combined with either idarubicin/cytarabine or cytarabine/etoposide in children with relapsed, refractory or secondary acute myeloid leukemia: a report from the Children's Oncology Group.
Pediatric blood & cancer, Volume: 61, Issue: 10
2014
AAML0523: a report from the Children's Oncology Group on the efficacy of clofarabine in combination with cytarabine in pediatric patients with recurrent acute myeloid leukemia.
Cancer, Aug-15, Volume: 120, Issue: 16
2014
A novel reduced-intensity umbilical cord blood transplantation using a recombinant G-CSF combined with high-dose Ara-C for active myeloid malignancies.
Bone marrow transplantation, Volume: 49, Issue: 7
2014
Low-dose homoharringtonine and cytarabine in combination with granulocyte colony-stimulating factor for elderly patients with de novo acute myeloid leukemia.
Leukemia & lymphoma, Volume: 56, Issue: 1
2015
Clofarabine in combination with a standard remission induction regimen (cytosine arabinoside and idarubicin) in patients with previously untreated intermediate and bad-risk acute myelogenous leukemia (AML) or high-risk myelodysplastic syndrome (HR-MDS): p
Annals of hematology, Volume: 93, Issue: 6
2014
Increasing the dose of aclarubicin in low-dose cytarabine and aclarubicin in combination with granulocyte colony-stimulating factor (CAG regimen) can safely and effectively treat relapsed or refractory acute myeloid leukemia.
International journal of hematology, Volume: 99, Issue: 5
2014
Homoharringtonine combined with cytarabine to treat chronic myelogenous leukemia in myeloid blast crisis and its impact on bone marrow CD34+CD7+ cells.
Acta haematologica, Volume: 132, Issue: 2
2014
Antitumor activity of elacytarabine combined with bevacizumab, cetuximab and trastuzumab in human NSCLC xenografts.
Anticancer research, Volume: 33, Issue: 9
2013
Ofatumumab in combination with ICE or DHAP chemotherapy in relapsed or refractory intermediate grade B-cell lymphoma.
Blood, Jul-25, Volume: 122, Issue: 4
2013
High-dose imatinib versus high-dose imatinib in combination with intermediate-dose cytarabine in patients with first chronic phase myeloid leukemia: a randomized phase III trial of the Dutch-Belgian HOVON study group.
Annals of hematology, Volume: 92, Issue: 8
2013
Long-term follow-up of European APL 2000 trial, evaluating the role of cytarabine combined with ATRA and Daunorubicin in the treatment of nonelderly APL patients.
American journal of hematology, Volume: 88, Issue: 7
2013
Translational phase I trial of vorinostat (suberoylanilide hydroxamic acid) combined with cytarabine and etoposide in patients with relapsed, refractory, or high-risk acute myeloid leukemia.
Clinical cancer research : an official journal of the American Association for Cancer Research, Apr-01, Volume: 19, Issue: 7
2013
Safety and pharmacokinetics of the antisense oligonucleotide (ASO) LY2181308 as a single-agent or in combination with idarubicin and cytarabine in patients with refractory or relapsed acute myeloid leukemia (AML).
Investigational new drugs, Volume: 31, Issue: 4
2013
[Mechanism of apoptosis synergistically induced by bortezomib combined with cytarabine in U937 cell line].
Zhongguo shi yan xue ye xue za zhi, Volume: 20, Issue: 6
2012
Phase 1 trial of gemtuzumab ozogamicin in combination with enocitabine and daunorubicin for elderly patients with relapsed or refractory acute myeloid leukemia: Japan Adult Leukemia Study Group (JALSG)-GML208 study.
International journal of hematology, Volume: 96, Issue: 4
2012
Fractionated doses of gemtuzumab ozogamicin combined with 3 + 7 induction chemotherapy as salvage treatment for young patients with acute myeloid leukemia in first relapse.
Annals of hematology, Volume: 91, Issue: 12
2012
High-dose cytarabine (24 g/m2) in combination with idarubicin (HiDAC-3) results in high first-cycle response with limited gastrointestinal toxicity in adult acute myeloid leukaemia.
Internal medicine journal, Volume: 43, Issue: 3
2013
[Clinical observation of thalidomide combined with VAD regimen for treatment of osteosclerotic myeloma (POEMS syndrome)].
Zhongguo shi yan xue ye xue za zhi, Volume: 20, Issue: 2
2012
The dual kinase inhibitor NVP-BEZ235 in combination with cytotoxic drugs exerts anti-proliferative activity towards acute lymphoblastic leukemia cells.
Anticancer research, Volume: 32, Issue: 2
2012
Valproic acid combined with cytosine arabinoside in elderly patients with acute myeloid leukemia has in vitro but limited clinical activity.
Leukemia & lymphoma, Volume: 53, Issue: 6
2012
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]