Page last updated: 2024-12-11

belinostat

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

Cross-References

ID SourceID
PubMed CID53437714
CHEMBL ID3392649
SCHEMBL ID375656
MeSH IDM0519759
PubMed CID6918638
CHEMBL ID408513
CHEBI ID61076
CHEBI ID94531
MeSH IDM0519759

Synonyms (99)

Synonym
2-propenamide, n-hydroxy-3-(3-((phenylamino)sulfonyl)phenyl)-
(e)-n-hydroxy-3-[3-(phenylsulfamoyl)phenyl]prop-2-enamide
px 105684
pdx101
belinostat (non-stereospecific)
pxd 101
NCGC00263155-01
SCHEMBL375656
2-propenamide, n-hydroxy-3-[3-[(phenylamino)sulfonyl]phenyl]-
CHEMBL3392649
AKOS026750234
n-hydroxy-3-[3-(phenylsulfamoyl)phenyl]-2-propenamide
HMS3654I17
FT-0774029
Q23766508
pxd101;px105684
SB16466
HMS3426E07
belinostat (pxd101)
n-hydroxy-3-(3-(phenylsulfamoyl)phenyl)prop-2-enamide
(2e)-n-hydroxy-3-[3-(phenylsulfamoyl)phenyl]acrylamide
belinostat ,
px-105684
pxd-101
pxd101
(2e)-n-hydroxy-3-[3-(phenylsulfamoyl)phenyl]prop-2-enamide
bdbm25150
n-hydroxy-3-(3-phenylsulphamoylphenyl)acrylamide
nsc-726630
414864-00-9
nsc726630
beleodaq
chebi:61076 ,
CHEMBL408513
D08870
belinostat (usan/inn)
beleodaq (tn)
EC-000.2286
n-hydroxy-3-[3-[(phenylamino)sulfonyl]phenyl]-2-propenamide
A25012
px105684
BELINOSTAT - PXD101
BCP9000386
866323-14-0
2-propenamide, n-hydroxy-3-[3-[(phenylamino)sulfonyl]phenyl]-, (2e)-
BCPP000351
belinostat,pxd101, px105684
(e)-n-hydroxy-3-(3-(n-phenylsulfamoyl)phenyl)acrylamide
S1085
n-hydroxy-3-(3-phenylsulfamoylphenyl)acrylamide
BRD-K17743125-001-01-9
2-propenamide, n-hydroxy-3-(3-((phenylamino)sulfonyl)phenyl)-, (2e)-
belinostat [mi]
belinostat [orange book]
(2e)-n-hydroxy-3-(3-(phenylsulfamoyl)phenyl)prop-2-enamide
belinostat [usan]
n-hydroxy-3-(3-((phenylamino)sulfonyl)phenyl)-2-propenamide
F4H96P17NZ ,
belinostat [vandf]
n-hydroxy-3-[3-(phenylsulfamoyl)phenyl]prop-2-enamide
belinostat [who-dd]
belinostat [inn]
CCG-208758
smr004702879
MLS006011091
NCNRHFGMJRPRSK-MDZDMXLPSA-N
(e)-n-hydroxy-3-(3-phenylsulfamoyl-phenyl)-acrylamide
gtpl7496
(e)-3-[3-(phenylsulfamoyl)phenyl]prop-2-enehydroxamic acid
mfcd08064035
e-belinostat
unii-f4h96p17nz
belinostat [usan:inn]
n-hydroxy-3-(3-(n-phenylsulfamoyl)phenyl)acrylamide
AC-35365
B5888
AC-25046
5og ,
AKOS025401741
J-523584
DTXSID60194378 ,
EX-A180
CHEBI:94531
NCGC00263155-05
SW219445-1
DB05015
(e)-n-hydroxy-3-[3-(phenylsulfamoyl)phenyl]prop-2-enamide.
AS-17068
BCP01741
Q4882925
AMY1792
nsc758774
nsc-758774
NCGC00263155-02
(2e)-n-hydroxy-3-(3-(phenylsulfamoyl)phenyl)acrylamide
belinostatum
l01xx49
dtxcid70116869
EN300-7361191

Research Excerpts

Overview

Belinostat is a pan-histone deacetylase (HDAC) inhibitor which recently approved for the treatment of relapsed/refractory Peripheral T-cell lymphomas (PTCL) BelinostAt is a second-generation histone de acetylase inhibitor (HDI) predominantly metabolized by UGT1A1-mediated glucuronidation.

ExcerptReferenceRelevance
"Belinostat is an intravenous histone deacetylase inhibitor with approval for T-cell lymphomas. "( Phase 1 study of belinostat and adavosertib in patients with relapsed or refractory myeloid malignancies.
Bandyopadhyay, D; Grant, S; Hudson, D; Kagan, AB; Kmieciak, M; Lancet, JE; Rudek, MA; Sankala, H; Shafer, D; Shrader, E; Tombes, MB; Weir, C, 2023
)
2.69
"Belinostat is a pan-histone deacetylase (HDAC) inhibitor which recently approved for the treatment of relapsed/refractory Peripheral T-cell lymphomas (PTCL). "( Inhibition of human UDP-glucuronosyltransferase enzyme by belinostat: Implications for drug-drug interactions.
Cao, J; Chen, X; Jiang, L; Liu, Y; Wang, X; Wang, Z; Yin, H, 2021
)
2.31
"Belinostat is a second-generation histone deacetylase inhibitor (HDI) predominantly metabolized by UGT1A1-mediated glucuronidation. "( A population pharmacokinetic/toxicity model for the reduction of platelets during a 48-h continuous intravenous infusion of the histone deacetylase inhibitor belinostat.
Balasubramaniam, S; Bates, SE; Figg, WD; Hall, OM; Peer, CJ; Piekarz, R; Sissung, TM, 2018
)
2.12
"Belinostat is a histone deacetylase inhibitor with anti-tumor effect in several pre-clinical tumor models and clinical trials. "( [18F]FDG and [18F]FLT positron emission tomography imaging following treatment with belinostat in human ovary cancer xenografts in mice.
Björkling, F; Erichsen, KD; Højgaard, L; Jensen, MM; Jensen, PB; Johnbeck, CB; Kjær, A; Madsen, J; Sehested, M, 2013
)
2.06
"Belinostat (PXD101) is a hydroxamate-type HDAC inhibitor tested in phase I and II clinical trials in solid tumors and hematological cancers."( Epigenetic and molecular mechanisms underlying the antileukemic activity of the histone deacetylase inhibitor belinostat in human acute promyelocytic leukemia cells.
Navakauskiene, R; Savickiene, J; Stirblyte, I; Treigyte, G; Valiuliene, G, 2014
)
1.34
"Belinostat is a novel histone deacetylase (HDAC) inhibitor that is being developed in various solid tumors and hematologic malignancies. "( Belinostat for the treatment of peripheral T-cell lymphomas.
Jimeno, A; McDermott, J, 2014
)
3.29
"Belinostat is a pan-histone deacetylase inhibitor with antitumour and anti-angiogenic properties. "( A Phase II trial of Belinostat (PXD101) in patients with relapsed or refractory peripheral or cutaneous T-cell lymphoma.
Advani, R; Allen, LF; Belt, RJ; Ben-Yehuda, D; Beylot-Barry, M; Bhat, G; Chawla, S; Duvic, M; Foss, F; Hillen, U; Hymes, KB; Intragumtornchai, T; Jacobsen, ED; Knoblauch, P; Laurent, G; Lekhakula, A; Lerner, A; Pohlman, B; Shpilberg, O, 2015
)
2.18
"Belinostat is a well-tolerated HDAC inhibitor that has shown activity in heavily pretreated patients with relapsed/refractory PTCL."( Belinostat for the treatment of relapsed or refractory peripheral T-cell lymphoma.
Cashen, AF; Rashidi, A, 2015
)
2.58
"Belinostat is a second-generation zinc-binding histone deacetylase inhibitor that is approved for peripheral T-cell lymphoma and is currently being studied in small cell lung cancer and other advanced carcinomas as a 48-hour continuous intravenous infusion. "( UGT1A1 genotype-dependent dose adjustment of belinostat in patients with advanced cancers using population pharmacokinetic modeling and simulation.
Balasubramaniam, S; Bates, SE; Erlich, S; Figg, WD; Goey, AK; Lee, MJ; Peer, CJ; Piekarz, R; Sissung, TM; Tomita, Y; Trepel, JB, 2016
)
2.14
"Belinostat is a potent small molecule inhibitor that exerts its antitumor effect through inhibition of histone deacetylase. "( A phase I clinical trial of the effect of belinostat on the pharmacokinetics and pharmacodynamics of warfarin.
Agarwal, N; Bailey, EB; Bailey, H; Bhat, G; Gupta, S; McPherson, JP; Reddy, G; Sharma, S; Werner, TL, 2016
)
2.14
"Belinostat is a safe and effective treatment option for relapsed and refractory peripheral T-cell lymphoma, with many future applications currently being investigated."( Belinostat for the treatment of relapsed or refractory peripheral T-cell lymphoma.
Campbell, P; Thomas, CM, 2017
)
2.62
"1. Belinostat is a histone deacetylase inhibitor that has been approved for the treatment of peripheral T-cell lymphoma. "( In vitro characterization of belinostat glucuronidation: demonstration of both UGT1A1 and UGT2B7 as the main contributing isozymes.
Dong, D; Liu, J; Lu, D; Wu, B; Zhang, T, 2017
)
1.37
"Belinostat is an inhibitor of histone deacetylase enzymes, resulting in DNA repair inhibition and apoptosis. "( A phase I study to determine the pharmacokinetics and urinary excretion of belinostat and metabolites in patients with advanced solid tumors.
Agarwal, N; Bailey, EB; Bailey, H; Batten, J; Bhat, G; Gupta, S; McPherson, JP; Reddy, G; Sharma, S; Werner, TL, 2016
)
2.11
"Belinostat (PXD 101) is a novel inhibitor of class I and II histone deacetylases. "( Phase II study of belinostat (PXD101), a histone deacetylase inhibitor, for second line therapy of advanced malignant pleural mesothelioma.
Belani, CP; Espinoza-Delgado, I; Frankel, P; Gandara, D; Gitlitz, B; Koczywas, M; Ramalingam, SS; Ruel, C, 2009
)
2.13
"Belinostat is a potent hydroxamate-type histone deacetylase inhibitor with a broad antineoplastic activity in a spectrum of preclinical tumor models and with demonstrated clinical efficacy in the still very early clinical trial program. "( Belinostat: a new broad acting antineoplastic histone deacetylase inhibitor.
Gimsing, P, 2009
)
3.24
"Belinostat is a hydroxamate-type histone deactylase inhibitor (HDACi), which has recently entered phase I and II clinical trials. "( Gene expression-signature of belinostat in cell lines is specific for histone deacetylase inhibitor treatment, with a corresponding signature in xenografts.
Hose, CD; Kondapaka, S; Monforte, J; Monks, A; Petersen, KD; Pezzoli, P; Sehested, M; Shoemaker, RH; Vansant, G, 2009
)
2.09
"Belinostat is a hydroxamate class HDAC inhibitor that has demonstrated activity in peripheral T-cell lymphoma and is undergoing clinical trials for non-hematologic malignancies. "( Glucuronidation by UGT1A1 is the dominant pathway of the metabolic disposition of belinostat in liver cancer patients.
Ansher, S; Chan, A; Chan, MY; Goh, BC; Ho, PC; Lau, JY; Lee, HS; Lee, SC; Lim, R; Ramírez, J; Ratain, MJ; Thuya, WL; Wan, SC; Wang, LZ; Wong, AL; Yeo, W; Zee, YK, 2013
)
2.06
"Belinostat is a novel, potent, pan-HDAC inhibitor with antiproliferative activity on a wide variety of tumor cell lines."( Plasma and cerebrospinal fluid pharmacokinetics of the histone deacetylase inhibitor, belinostat (PXD101), in non-human primates.
Balis, FM; Dvinge, H; Lichenstein, HS; McCully, C; Sehested, M; Tjørnelund, J; Warren, KE, 2008
)
1.29

Effects

Belinostat's efficacy has been evaluated in a clinical trial showing an overall response rate (ORR) of 25.8% and a median duration of response of 8.4 months. The drug has been relatively well tolerated following both i.v. and IV therapy.

ExcerptReferenceRelevance
"Belinostat's efficacy has been evaluated in a clinical trial showing an overall response rate (ORR) of 25.8% and a median duration of response of 8.4 months."( Belinostat for the treatment of relapsed or refractory peripheral T-cell lymphoma.
Campbell, P; Thomas, CM, 2017
)
2.62
"Belinostat has been relatively well tolerated following both i.v."( Belinostat: a new broad acting antineoplastic histone deacetylase inhibitor.
Gimsing, P, 2009
)
2.52

Treatment

Belinostat treatment significantly suppressed the activation of M1 microglia and the proinflammatory cytokine expression. Co-treatment with bel inostat effectively eliminated a vincristine-induced, actively cycling polyploid cell population.

ExcerptReferenceRelevance
"Belinostat treatment significantly suppressed the activation of M1 microglia and the proinflammatory cytokine expression; but it had no effects on the M2 microglial polarization."( The histone deacetylase inhibitor belinostat ameliorates experimental autoimmune encephalomyelitis in mice by inhibiting TLR2/MyD88 and HDAC3/ NF-κB p65-mediated neuroinflammation.
Chen, H; Chen, M; Chen, S; Du, G; Ji, B; Li, L; Liu, D; Shen, Y; Yang, R; Zhao, J, 2022
)
1.72
"Co-treatment with belinostat effectively eliminated a vincristine-induced, actively cycling polyploid cell population."( Belinostat and vincristine demonstrate mutually synergistic cytotoxicity associated with mitotic arrest and inhibition of polyploidy in a preclinical model of aggressive diffuse large B cell lymphoma.
Bhakta, A; Demirjian, JA; Hahn, S; Havas, AP; Rodrigues, KB; Scavello, M; Schmelz, M; Smith, CL; Tran, J; Tula-Sanchez, AA; Zeng, Y, 2016
)
2.2
"Treatment with belinostat resulted in significant in vitro and in vivo growth inhibition of PDAC cells. "( Experimental in vivo and in vitro treatment with a new histone deacetylase inhibitor belinostat inhibits the growth of pancreatic cancer.
Arnold, SM; Dovzhanskiy, DI; Felix, K; Giese, N; Hackert, T; Oehme, I; Werner, J; Witt, O, 2012
)
0.96

Toxicity

ExcerptReferenceRelevance
" Grade III/IV adverse events were (n; %): leucopenia (5; 22%), neutropenia (7; 30%), thrombocytopenia (3; 13%) anaemia (1; 4%), peripheral sensory neuropathy (2; 9%), fatigue (1; 4%), vomiting (1; 4%) and myalgia (1; 4%)."( A phase I study of the safety and pharmacokinetics of the histone deacetylase inhibitor belinostat administered in combination with carboplatin and/or paclitaxel in patients with solid tumours.
Buhl-Jensen, P; Crowley, E; de Bono, JS; Engelholm, SA; Knoblauch, P; Lassen, U; Molife, LR; Penson, RT; Sinha, R; Sorensen, M; Tjornelund, J; Vidal, L, 2010
)
0.58
" These agents are associated with a range of class-related and agent-specific serious and/or severe adverse effects, notably myelosuppression, diarrhoea and various cardiac effects."( Safety and Tolerability of Histone Deacetylase (HDAC) Inhibitors in Oncology.
Shah, RR, 2019
)
0.51
"Current melphalan-based regimens for intravitreal chemotherapy for retinoblastoma vitreous seeds are effective but toxic to the retina."( Intravitreal HDAC Inhibitor Belinostat Effectively Eradicates Vitreous Seeds Without Retinal Toxicity In Vivo in a Rabbit Retinoblastoma Model.
Bogan, CM; Boyd, KL; Bridges, TM; Calcutt, MW; Chen, SC; Daniels, AB; Friedman, DL; Kaczmarek, JV; Lindsley, CW; Liu, Q; Liu, X; Pierce, JM; Richmond, A; Tao, YK, 2021
)
0.92

Pharmacokinetics

Belinostat was cleared rapidly from plasma with a half-life of 1. Part B was designed to evaluate the relative contribution of belinost at to the combination based on analysis of pharmacodynamic markers.

ExcerptReferenceRelevance
" Furthermore, the ability to measure histone acetylation in blood samples could provide a suitable pharmacodynamic end point to monitor drug activity."( Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101.
Bandara, MJ; Brown, R; Finn, PW; La Thangue, NB; Plumb, JA; Romero, MR; Watkins, CJ; Williams, RJ, 2003
)
0.32
" Pharmacokinetic parameters were calculated using non-compartmental methods, and CSF penetration is expressed as the ratio of the area under the concentration-time curve (AUC) in CSF to the AUC in plasma."( Plasma and cerebrospinal fluid pharmacokinetics of the histone deacetylase inhibitor, belinostat (PXD101), in non-human primates.
Balis, FM; Dvinge, H; Lichenstein, HS; McCully, C; Sehested, M; Tjørnelund, J; Warren, KE, 2008
)
0.57
"To determine the safety, dose-limiting toxicity, maximum tolerated dose, and pharmacokinetic and pharmacodynamic profiles of the novel hydroxamate histone deacetylase inhibitor belinostat (previously named PXD101) in patients with advanced refractory solid tumors."( A phase 1 pharmacokinetic and pharmacodynamic study of the histone deacetylase inhibitor belinostat in patients with advanced solid tumors.
Brown, R; Buhl-Jensen, P; DeBono, JS; Evans, TR; Knoblauch, P; Ooi, CE; Plumb, JA; Rasmussen, A; Steele, NL; Tjørnelund, J; Vidal, L, 2008
)
0.76
" Pharmacokinetic variables were evaluated at all dose levels."( A phase 1 pharmacokinetic and pharmacodynamic study of the histone deacetylase inhibitor belinostat in patients with advanced solid tumors.
Brown, R; Buhl-Jensen, P; DeBono, JS; Evans, TR; Knoblauch, P; Ooi, CE; Plumb, JA; Rasmussen, A; Steele, NL; Tjørnelund, J; Vidal, L, 2008
)
0.57
" belinostat is well tolerated, exhibits dose-dependent pharmacodynamic effects, and has promising antitumor activity."( A phase 1 pharmacokinetic and pharmacodynamic study of the histone deacetylase inhibitor belinostat in patients with advanced solid tumors.
Brown, R; Buhl-Jensen, P; DeBono, JS; Evans, TR; Knoblauch, P; Ooi, CE; Plumb, JA; Rasmussen, A; Steele, NL; Tjørnelund, J; Vidal, L, 2008
)
1.48
"The combination was well tolerated, with no evidence of pharmacokinetic interaction."( A phase I study of the safety and pharmacokinetics of the histone deacetylase inhibitor belinostat administered in combination with carboplatin and/or paclitaxel in patients with solid tumours.
Buhl-Jensen, P; Crowley, E; de Bono, JS; Engelholm, SA; Knoblauch, P; Lassen, U; Molife, LR; Penson, RT; Sinha, R; Sorensen, M; Tjornelund, J; Vidal, L, 2010
)
0.58
" Preliminary pharmacodynamic (PD) studies were also performed to enable comparison of the biological effects of the oral and intravenous formulations."( Pharmacokinetic and pharmacodynamic properties of an oral formulation of the histone deacetylase inhibitor Belinostat (PXD101).
Brown, R; Buhl-Jensen, P; de Bono, JS; Evans, TR; Knoblauch, P; Molife, R; Plumb, JA; Steele, NL; Tjørnelund, J; Vidal, L, 2011
)
0.58
" Part B was designed to evaluate the relative contribution of belinostat to the combination based on analysis of pharmacodynamic markers, and incorporated a design in which patients were randomized during cycle 1 to AZA alone, or the combination, at the maximally tolerated dose of belinostat."( A phase I and pharmacodynamic study of the histone deacetylase inhibitor belinostat plus azacitidine in advanced myeloid neoplasia.
Bennett, M; Fulton, N; Godley, LA; Green, M; Halpern, A; Karrison, T; Koval, G; Larson, RA; Madzo, J; Malnassy, G; Mattison, RJ; Odenike, O; Ratain, MJ; Stock, W; Yee, KW, 2015
)
0.89
" A 2-compartment population pharmacokinetic (PK) model was developed and validated that incorporated the UGT1A1 genotype, albumin, and creatinine clearance on the clearance parameter; body weight was a significant covariate on volume."( UGT1A1 genotype-dependent dose adjustment of belinostat in patients with advanced cancers using population pharmacokinetic modeling and simulation.
Balasubramaniam, S; Bates, SE; Erlich, S; Figg, WD; Goey, AK; Lee, MJ; Peer, CJ; Piekarz, R; Sissung, TM; Tomita, Y; Trepel, JB, 2016
)
0.69
" It was the intention of this brief report to expand a population pharmacokinetic (PPK) model to include a pharmacodynamic (PD) model describing the change in platelet levels in patients with cancer administered belinostat as a 48-h continuous intravenous infusion, along with cisplatin and etoposide."( A population pharmacokinetic/toxicity model for the reduction of platelets during a 48-h continuous intravenous infusion of the histone deacetylase inhibitor belinostat.
Balasubramaniam, S; Bates, SE; Figg, WD; Hall, OM; Peer, CJ; Piekarz, R; Sissung, TM, 2018
)
0.86

Compound-Compound Interactions

This study identified the MTD of belinostat in combination with PAC and indicates that the combination is active and feasible in TETs. To assess drug-drug interactions (DDIs) potential of belInostat via inhibition of UDP-glucuronosyltransferases (UGTs), the effects of bel Inostat on UGTs activities were investigated.

ExcerptReferenceRelevance
"This phase I/II study sought to determine the safety and maximum tolerated dose (MTD) of a novel schedule of belinostat, a histone deacetylase inhibitor (HDAC) administered before and in combination with cisplatin (P), doxorubicin (A), and cyclophosphamide (C) in thymic epithelial tumors (TET)."( A phase I/II trial of belinostat in combination with cisplatin, doxorubicin, and cyclophosphamide in thymic epithelial tumors: a clinical and translational study.
Carter, CA; Chen, Y; Figg, WD; Frosch, A; Giaccone, G; Lee, MJ; Lopez-Chavez, A; Peer, CJ; Piekarz, R; Rajan, A; Redon, CE; Scepura, B; Schrump, DS; Steinberg, SM; Szabo, E; Thomas, A; Tomita, Y; Trepel, JB, 2014
)
0.93
"This study identified the MTD of belinostat in combination with PAC and indicates that the combination is active and feasible in TETs."( A phase I/II trial of belinostat in combination with cisplatin, doxorubicin, and cyclophosphamide in thymic epithelial tumors: a clinical and translational study.
Carter, CA; Chen, Y; Figg, WD; Frosch, A; Giaccone, G; Lee, MJ; Lopez-Chavez, A; Peer, CJ; Piekarz, R; Rajan, A; Redon, CE; Scepura, B; Schrump, DS; Steinberg, SM; Szabo, E; Thomas, A; Tomita, Y; Trepel, JB, 2014
)
1
" Our aim was to study the therapeutic efficacy of checkpoint inhibitors (anti-CTLA-4 and anti-PD-1 antibodies) in combination with the histone deacetylase inhibitor (HDACi) Belinostat."( Enhanced anti-tumor efficacy of checkpoint inhibitors in combination with the histone deacetylase inhibitor Belinostat in a murine hepatocellular carcinoma model.
Dixon, G; Egea, J; Iglesias, T; Lasarte, JJ; Llopiz, D; Pivette, P; Ruiz, M; Sangro, B; Sarobe, P; Silva, L; Trochon-Joseph, V; Vasseur, B; Villanueva, L, 2019
)
0.92
" To assess drug-drug interactions (DDIs) potential of belinostat via inhibition of UDP-glucuronosyltransferases (UGTs), the effects of belinostat on UGTs activities were investigated using the non-selective probe substrate 4-methylumbelliferone (4-MU) and trifluoperazine (TFP) by UPLC-MS/MS."( Inhibition of human UDP-glucuronosyltransferase enzyme by belinostat: Implications for drug-drug interactions.
Cao, J; Chen, X; Jiang, L; Liu, Y; Wang, X; Wang, Z; Yin, H, 2021
)
1.11
"This study was conducted to identify novel therapeutic targets that can be combined with HDACis for treating CTCL."( Kinome profiling analysis identified Src pathway as a novel therapeutic target in combination with histone deacetylase inhibitors for cutaneous T-cell lymphoma.
Fujii, K; Jimura, N; Kanekura, T; Kondo, T; Qiao, Z; Tsuchiya, R; Yoshimatsu, Y, 2021
)
0.62

Bioavailability

ExcerptReferenceRelevance
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51
" This work resulted in the discovery of spirocycle 30d that shows good oral bioavailability and tumor growth inhibition in an HCT-116 murine xenograft model."( Discovery, synthesis, and pharmacological evaluation of spiropiperidine hydroxamic acid based derivatives as structurally novel histone deacetylase (HDAC) inhibitors.
Abate, A; Bigogno, C; Boggio, R; Carenzi, G; Cataudella, T; Dal Zuffo, R; Dondio, G; Fulco, MC; Mai, A; Mercurio, C; Minucci, S; Rozio, MG; Thaler, F; Varasi, M, 2011
)
0.37
"Spray-dried dispersions (SDDs) are an important technology for enhancing the oral bioavailability of poorly water-soluble drugs."( Mechanistic Study of Belinostat Oral Absorption From Spray-Dried Dispersions.
Goodwin, A; Morgen, M; Mudie, D; Pivette, P; Sarmiento, A; Stewart, A; Vodak, D; Winter, M; Yates, I, 2019
)
0.83
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51

Dosage Studied

Belinostat was dosed at 1000 mg/m(2) daily for five days with carboplatin AUC 5 on day three of 21-day cycles. There was no clear evidence of drug accumulation on twice daily dosing (bid), but a trend towards accumulation was apparent when given three times daily (tid)

ExcerptRelevanceReference
" An LC-MSMS-based method was applied in a quantitative proteomic study of the dose-response effect of the histone deacetylase inhibitor (HDACi) PXD101 on histone acetylation in human cell cultures."( Quantitative proteomic analysis of post-translational modifications of human histones.
Beck, HC; Finn, P; Grauslund, M; Hansen, AM; Jensen, LH; Jensen, ON; Matthiesen, R; Nielsen, EC; Sehested, M, 2006
)
0.33
" Evaluation of combination strategies or alternate dosing schedules may be necessary for further development of this novel agent in mesothelioma."( Phase II study of belinostat (PXD101), a histone deacetylase inhibitor, for second line therapy of advanced malignant pleural mesothelioma.
Belani, CP; Espinoza-Delgado, I; Frankel, P; Gandara, D; Gitlitz, B; Koczywas, M; Ramalingam, SS; Ruel, C, 2009
)
0.69
" There was no clear evidence of drug accumulation on twice daily dosing (bid); however, a trend towards accumulation was apparent when belinostat was given three times daily (tid)."( Pharmacokinetic and pharmacodynamic properties of an oral formulation of the histone deacetylase inhibitor Belinostat (PXD101).
Brown, R; Buhl-Jensen, P; de Bono, JS; Evans, TR; Knoblauch, P; Molife, R; Plumb, JA; Steele, NL; Tjørnelund, J; Vidal, L, 2011
)
0.79
" When orally dosed to tumor-bearing mice, 3 is enriched in tumor tissue which may contribute to its potent antitumor activity and prolonged duration of action."( Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
Bonday, Z; Chang, JW; Chen, D; Deng, W; Dymock, BW; Fang, L; Goh, KC; Goh, KL; Goh, SK; Hu, C; Kantharaj, E; Khng, HH; Khoo, ML; Lee, KC; Liu, X; Lu, T; Lye, PL; Ng, MC; Poulsen, A; Sangthongpitag, K; Sun, ET; Wang, H; Wang, X; Wood, JM; Wu, X; Yeo, P; Yu, N, 2011
)
0.37
" Belinostat was dosed at 1000 mg/m(2) daily for five days with carboplatin AUC 5 on day three of 21-day cycles."( A phase II evaluation of belinostat and carboplatin in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube, or primary peritoneal carcinoma: a Gynecologic Oncology Group study.
Blessing, JA; Disilvestro, PA; Dizon, DS; Drake, RD; Fader, AN; Johnston, CM; Penson, RT; Walker, JL, 2012
)
1.59
" Belinostat as monotherapy has minimal single-agent effect in AML on this dosing schedule."( A phase 2 study of belinostat (PXD101) in patients with relapsed or refractory acute myeloid leukemia or patients over the age of 60 with newly diagnosed acute myeloid leukemia: a California Cancer Consortium Study.
Foon, KA; Frankel, P; Kirschbaum, MH; Newman, EM; Pulone, B; Ruel, C; Tuscano, JM, 2014
)
1.64
" We found that treatment with Bel, depending on the dosage used, inhibits cell proliferation, whereas in combination with RA enhances and accelerates granulocytic leukaemia cell differentiation."( Belinostat, a potent HDACi, exerts antileukaemic effect in human acute promyelocytic leukaemia cells via chromatin remodelling.
Cicenaite, D; Kaupinis, A; Navakauskiene, R; Stirblyte, I; Valiuliene, G; Valius, M, 2015
)
1.86
" Clinical effects of this genotype-based dosing recommendation is currently prospectively being investigated."( UGT genotyping in belinostat dosing.
Figg, WD; Goey, AK, 2016
)
0.77
" Belinostat is metabolized hepatically through cytochrome P-450 enzymes 3A4, 2C9, and 2A6; however, no empiric dosage adjustments of belinostat are recommended during concurrent use of inhibitors or inducers of these enzymes."( Belinostat for the treatment of relapsed or refractory peripheral T-cell lymphoma.
Campbell, P; Thomas, CM, 2017
)
2.81
" Present data are lacking to provide dosing recommendations in renal insufficiency."( A phase I study to determine the pharmacokinetics and urinary excretion of belinostat and metabolites in patients with advanced solid tumors.
Agarwal, N; Bailey, EB; Bailey, H; Batten, J; Bhat, G; Gupta, S; McPherson, JP; Reddy, G; Sharma, S; Werner, TL, 2016
)
0.66
" Since these metabolites are primarily inactive, belinostat may not require dosage adjustment in renal dysfunction."( A phase I study to determine the pharmacokinetics and urinary excretion of belinostat and metabolites in patients with advanced solid tumors.
Agarwal, N; Bailey, EB; Bailey, H; Batten, J; Bhat, G; Gupta, S; McPherson, JP; Reddy, G; Sharma, S; Werner, TL, 2016
)
0.92
" Thus, genotype-directed dosing could improve pharmacotherapy by reducing the risk of toxicities or preventing suboptimal treatment."( Pharmacogenomics and histone deacetylase inhibitors.
Figg, WD; Goey, AK; Peer, CJ; Sissung, TM, 2016
)
0.43
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (2)

RoleDescription
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
EC 3.5.1.98 (histone deacetylase) inhibitorAn EC 3.5.1.* (non-peptide linear amide C-N hydrolase) inhibitor that interferes with the function of histone deacetylase (EC 3.5.1.98).
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (3)

ClassDescription
olefinic compoundAny organic molecular entity that contains at least one C=C bond.
hydroxamic acidA compound, RkE(=O)lNHOH, derived from an oxoacid RkE(=O)l(OH) (l =/= 0) by replacing -OH with -NHOH, and derivatives thereof. Specific examples of hydroxamic acids are preferably named as N-hydroxy amides.
sulfonamideAn amide of a sulfonic acid RS(=O)2NR'2.
olefinic compoundAny organic molecular entity that contains at least one C=C bond.
[compound class information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Protein Targets (31)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Fumarate hydrataseHomo sapiens (human)Potency17.06190.00308.794948.0869AID1347053
EWS/FLI fusion proteinHomo sapiens (human)Potency2.07180.001310.157742.8575AID1259252; AID1259253; AID1259255; AID1259256
polyproteinZika virusPotency17.06190.00308.794948.0869AID1347053
tyrosine-protein kinase YesHomo sapiens (human)Potency0.00170.00005.018279.2586AID686947
PPM1D proteinHomo sapiens (human)Potency2.33620.00529.466132.9993AID1347411
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency16.93300.01237.983543.2770AID1645841
GVesicular stomatitis virusPotency10.68400.01238.964839.8107AID1645842
cytochrome P450 2D6Homo sapiens (human)Potency21.31740.00108.379861.1304AID1645840
Interferon betaHomo sapiens (human)Potency4.42320.00339.158239.8107AID1347411; AID1645842
HLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)Potency10.68400.01238.964839.8107AID1645842
Spike glycoproteinSevere acute respiratory syndrome-related coronavirusPotency10.00000.009610.525035.4813AID1479145
Inositol hexakisphosphate kinase 1Homo sapiens (human)Potency10.68400.01238.964839.8107AID1645842
cytochrome P450 2C9, partialHomo sapiens (human)Potency10.68400.01238.964839.8107AID1645842
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Inhibition Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Histone deacetylase 3Homo sapiens (human)IC50 (µMol)0.19420.00040.619610.0000AID1187258; AID1545757; AID1589101; AID1639362; AID1759720; AID1759721; AID1759727; AID1759728; AID1798526; AID316875; AID324949; AID750109
Histone deacetylase 3Homo sapiens (human)Ki0.00730.00020.42378.1900AID1236444; AID496803; AID619048
Bromodomain-containing protein 4Homo sapiens (human)IC50 (µMol)0.02700.00040.40329.0500AID1462230
Cytochrome P450 3A4Homo sapiens (human)IC50 (µMol)10.00000.00011.753610.0000AID609494
Leukotriene A-4 hydrolaseHomo sapiens (human)IC50 (µMol)10.00000.00051.28547.6500AID1441630; AID1441631
Cytochrome P450 3A5Homo sapiens (human)IC50 (µMol)0.21470.00330.70736.2000AID1371032
Histone deacetylase 4Homo sapiens (human)IC50 (µMol)4.40380.00061.052610.0000AID1187258; AID1466062; AID1545757; AID1639362; AID1798526; AID1802061; AID1802240; AID316875; AID324950; AID750109
Histone deacetylase 4Homo sapiens (human)Ki0.25830.00021.62559.1242AID1236446; AID496804; AID619049
Aspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)IC50 (µMol)6.64000.02002.66099.4300AID1677972; AID1677973; AID1677974; AID1677975
Potassium voltage-gated channel subfamily H member 2Homo sapiens (human)IC50 (µMol)30.00000.00091.901410.0000AID609493; AID613341
Histone deacetylase 1Homo sapiens (human)IC50 (µMol)3.08320.00010.55439.9000AID1187258; AID1326509; AID1383985; AID1384000; AID1466061; AID1545757; AID1589101; AID1639362; AID1639370; AID1759721; AID1759727; AID1759728; AID1798526; AID1802061; AID1802240; AID316875; AID324948; AID609489; AID619041; AID750109
Histone deacetylase 1Homo sapiens (human)Ki0.00930.00000.49888.1900AID1236442; AID496801; AID619046
Histone deacetylase 7Homo sapiens (human)IC50 (µMol)0.03570.00071.02609.9000AID1187258; AID1545757; AID1639362; AID316875; AID750109
Histone deacetylase 7Homo sapiens (human)Ki0.06700.00022.00059.5000AID1236448; AID496807; AID619052
Histone deacetylase 2Homo sapiens (human)IC50 (µMol)0.24010.00010.72219.9700AID1187258; AID1326520; AID1383985; AID1466055; AID1545757; AID1589101; AID1639362; AID1759721; AID1759727; AID1759728; AID316875; AID750109
Histone deacetylase 2Homo sapiens (human)Ki0.00790.00000.47098.1900AID1236443; AID496802; AID619047
Polyamine deacetylase HDAC10Homo sapiens (human)IC50 (µMol)0.03570.00050.72459.9000AID1187258; AID1545757; AID1639362; AID316875; AID750109
Polyamine deacetylase HDAC10Homo sapiens (human)Ki0.05900.00000.76878.1900AID619055
Histone deacetylase 11 Homo sapiens (human)IC50 (µMol)5.98480.00030.92989.9000AID1187258; AID1545757; AID1639362; AID1802061; AID1802240; AID316875; AID750109
Histone deacetylase 11 Homo sapiens (human)Ki0.02700.00011.21478.1900AID619056
Histone deacetylase 8Homo sapiens (human)IC50 (µMol)4.10220.00070.99479.9000AID1187258; AID1326522; AID1384002; AID1466064; AID1545757; AID1589101; AID1639362; AID1639372; AID1649884; AID1759721; AID1759727; AID1759728; AID1802061; AID1802240; AID316875; AID750109
Histone deacetylase 8Homo sapiens (human)Ki0.02400.00020.75258.1900AID1236445; AID496808; AID619053
Histone deacetylase 6Homo sapiens (human)IC50 (µMol)3.94140.00000.53769.9000AID1187258; AID1326521; AID1384001; AID1466063; AID1525776; AID1545757; AID1639362; AID1639371; AID1798526; AID1802061; AID1802240; AID316875; AID324956; AID750109
Histone deacetylase 6Homo sapiens (human)Ki0.00360.00010.41568.1900AID1236450; AID1802011; AID496806; AID619051
Histone deacetylase 9Homo sapiens (human)IC50 (µMol)0.03570.00050.94139.9000AID1187258; AID1545757; AID1639362; AID316875; AID750109
Histone deacetylase 9Homo sapiens (human)Ki0.17470.00021.85209.0000AID1236449; AID496809; AID619054
Histone deacetylase 5Homo sapiens (human)IC50 (µMol)0.03570.00070.961010.0000AID1187258; AID1545757; AID1639362; AID316875; AID750109
Histone deacetylase 5Homo sapiens (human)Ki0.12500.00021.29939.5000AID1236447; AID496805; AID619050
Histone deacetylase Plasmodium falciparum (malaria parasite P. falciparum)IC50 (µMol)0.21470.00060.16880.9400AID1371032
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Activation Measurements

ProteinTaxonomyMeasurementAverageMin (ref.)Avg (ref.)Max (ref.)Bioassay(s)
Histone deacetylase 3Homo sapiens (human)EC50 (µMol)0.03000.03001.85126.7000AID1404339; AID1845893
Histone deacetylase 4Homo sapiens (human)EC50 (µMol)0.07250.03002.35236.7000AID1404339; AID1845894
Histone deacetylase 1Homo sapiens (human)EC50 (µMol)0.03550.03001.98776.7000AID1404339; AID1845891
Histone deacetylase 7Homo sapiens (human)EC50 (µMol)0.04850.03002.32116.7000AID1404339; AID1845896
Histone deacetylase 2Homo sapiens (human)EC50 (µMol)0.07750.03001.85756.7000AID1404339; AID1845892
Polyamine deacetylase HDAC10Homo sapiens (human)EC50 (µMol)0.03000.03002.54656.7000AID1404339
Histone deacetylase 11 Homo sapiens (human)EC50 (µMol)0.03000.03002.54656.7000AID1404339
Histone deacetylase 8Homo sapiens (human)EC50 (µMol)0.12300.03002.27346.7000AID1404339; AID1845898
Histone deacetylase 6Homo sapiens (human)EC50 (µMol)0.05600.00521.59986.7000AID1404339; AID1845895
Histone deacetylase 9Homo sapiens (human)EC50 (µMol)0.07900.03002.32666.7000AID1404339; AID1845897
Histone deacetylase 5Homo sapiens (human)EC50 (µMol)0.03000.03002.54656.7000AID1404339
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (289)

Processvia Protein(s)Taxonomy
negative regulation of myotube differentiationHistone deacetylase 3Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 3Homo sapiens (human)
establishment of mitotic spindle orientationHistone deacetylase 3Homo sapiens (human)
in utero embryonic developmentHistone deacetylase 3Homo sapiens (human)
positive regulation of protein phosphorylationHistone deacetylase 3Homo sapiens (human)
chromatin organizationHistone deacetylase 3Homo sapiens (human)
transcription by RNA polymerase IIHistone deacetylase 3Homo sapiens (human)
protein deacetylationHistone deacetylase 3Homo sapiens (human)
regulation of mitotic cell cycleHistone deacetylase 3Homo sapiens (human)
positive regulation of protein ubiquitinationHistone deacetylase 3Homo sapiens (human)
regulation of protein stabilityHistone deacetylase 3Homo sapiens (human)
positive regulation of TOR signalingHistone deacetylase 3Homo sapiens (human)
circadian regulation of gene expressionHistone deacetylase 3Homo sapiens (human)
regulation of multicellular organism growthHistone deacetylase 3Homo sapiens (human)
positive regulation of protein import into nucleusHistone deacetylase 3Homo sapiens (human)
regulation of circadian rhythmHistone deacetylase 3Homo sapiens (human)
negative regulation of apoptotic processHistone deacetylase 3Homo sapiens (human)
negative regulation of DNA-templated transcriptionHistone deacetylase 3Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIHistone deacetylase 3Homo sapiens (human)
negative regulation of JNK cascadeHistone deacetylase 3Homo sapiens (human)
spindle assemblyHistone deacetylase 3Homo sapiens (human)
establishment of skin barrierHistone deacetylase 3Homo sapiens (human)
cellular response to fluid shear stressHistone deacetylase 3Homo sapiens (human)
positive regulation of cold-induced thermogenesisHistone deacetylase 3Homo sapiens (human)
DNA repair-dependent chromatin remodelingHistone deacetylase 3Homo sapiens (human)
cornified envelope assemblyHistone deacetylase 3Homo sapiens (human)
negative regulation of cardiac muscle cell differentiationHistone deacetylase 3Homo sapiens (human)
epigenetic regulation of gene expressionHistone deacetylase 3Homo sapiens (human)
regulation of transcription by RNA polymerase IIBromodomain-containing protein 4Homo sapiens (human)
positive regulation of G2/M transition of mitotic cell cycleBromodomain-containing protein 4Homo sapiens (human)
positive regulation of transcription elongation by RNA polymerase IIBromodomain-containing protein 4Homo sapiens (human)
chromatin organizationBromodomain-containing protein 4Homo sapiens (human)
DNA damage responseBromodomain-containing protein 4Homo sapiens (human)
positive regulation of transcription elongation by RNA polymerase IIBromodomain-containing protein 4Homo sapiens (human)
positive regulation of canonical NF-kappaB signal transductionBromodomain-containing protein 4Homo sapiens (human)
positive regulation of DNA-templated transcriptionBromodomain-containing protein 4Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIBromodomain-containing protein 4Homo sapiens (human)
regulation of inflammatory responseBromodomain-containing protein 4Homo sapiens (human)
positive regulation of T-helper 17 cell lineage commitmentBromodomain-containing protein 4Homo 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)
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)
proteolysisLeukotriene A-4 hydrolaseHomo sapiens (human)
lipid metabolic processLeukotriene A-4 hydrolaseHomo sapiens (human)
response to zinc ionLeukotriene A-4 hydrolaseHomo sapiens (human)
leukotriene biosynthetic processLeukotriene A-4 hydrolaseHomo sapiens (human)
protein metabolic processLeukotriene A-4 hydrolaseHomo sapiens (human)
peptide catabolic processLeukotriene A-4 hydrolaseHomo sapiens (human)
response to peptide hormoneLeukotriene A-4 hydrolaseHomo sapiens (human)
type I pneumocyte differentiationLeukotriene A-4 hydrolaseHomo sapiens (human)
lipid hydroxylationCytochrome P450 3A5Homo sapiens (human)
xenobiotic metabolic processCytochrome P450 3A5Homo sapiens (human)
steroid metabolic processCytochrome P450 3A5Homo sapiens (human)
estrogen metabolic processCytochrome P450 3A5Homo sapiens (human)
alkaloid catabolic processCytochrome P450 3A5Homo sapiens (human)
xenobiotic catabolic processCytochrome P450 3A5Homo sapiens (human)
retinol metabolic processCytochrome P450 3A5Homo sapiens (human)
retinoic acid metabolic processCytochrome P450 3A5Homo sapiens (human)
aflatoxin metabolic processCytochrome P450 3A5Homo sapiens (human)
oxidative demethylationCytochrome P450 3A5Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 4Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 4Homo sapiens (human)
chromatin remodelingHistone deacetylase 4Homo sapiens (human)
protein deacetylationHistone deacetylase 4Homo sapiens (human)
inflammatory responseHistone deacetylase 4Homo sapiens (human)
nervous system developmentHistone deacetylase 4Homo sapiens (human)
positive regulation of cell population proliferationHistone deacetylase 4Homo sapiens (human)
negative regulation of myotube differentiationHistone deacetylase 4Homo sapiens (human)
negative regulation of transcription by competitive promoter bindingHistone deacetylase 4Homo sapiens (human)
response to denervation involved in regulation of muscle adaptationHistone deacetylase 4Homo sapiens (human)
cardiac muscle hypertrophy in response to stressHistone deacetylase 4Homo sapiens (human)
protein sumoylationHistone deacetylase 4Homo sapiens (human)
B cell differentiationHistone deacetylase 4Homo sapiens (human)
positive regulation of protein sumoylationHistone deacetylase 4Homo sapiens (human)
peptidyl-lysine deacetylationHistone deacetylase 4Homo sapiens (human)
B cell activationHistone deacetylase 4Homo sapiens (human)
regulation of protein bindingHistone deacetylase 4Homo sapiens (human)
negative regulation of DNA-binding transcription factor activityHistone deacetylase 4Homo sapiens (human)
negative regulation of gene expression, epigeneticHistone deacetylase 4Homo sapiens (human)
negative regulation of glycolytic processHistone deacetylase 4Homo sapiens (human)
positive regulation of DNA-templated transcriptionHistone deacetylase 4Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIHistone deacetylase 4Homo sapiens (human)
positive regulation of DNA-binding transcription factor activityHistone deacetylase 4Homo sapiens (human)
type I interferon-mediated signaling pathwayHistone deacetylase 4Homo sapiens (human)
response to interleukin-1Histone deacetylase 4Homo sapiens (human)
muscle contractionAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
pattern specification processAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
cell population proliferationAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
negative regulation of cell population proliferationAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
regulation of protein stabilityAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
limb morphogenesisAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
positive regulation of proteolysisAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
roof of mouth developmentAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
face morphogenesisAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
activation of cysteine-type endopeptidase activityAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
regulation of protein depolymerizationAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
peptidyl-aspartic acid hydroxylationAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by hormonePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion homeostasisPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cardiac muscle contractionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of ventricular cardiac muscle cell membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cellular response to xenobiotic stimulusPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane depolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of heart rate by cardiac conductionPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
membrane repolarization during ventricular cardiac muscle cell action potentialPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
positive regulation of potassium ion transmembrane transportPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of potassium ion export across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
potassium ion import across plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
negative regulation of myotube differentiationHistone deacetylase 1Homo sapiens (human)
negative regulation of apoptotic processHistone deacetylase 1Homo sapiens (human)
positive regulation of signaling receptor activityHistone deacetylase 1Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 1Homo sapiens (human)
chromatin organizationHistone deacetylase 1Homo sapiens (human)
chromatin remodelingHistone deacetylase 1Homo sapiens (human)
DNA methylation-dependent heterochromatin formationHistone deacetylase 1Homo sapiens (human)
regulation of transcription by RNA polymerase IIHistone deacetylase 1Homo sapiens (human)
protein deacetylationHistone deacetylase 1Homo sapiens (human)
endoderm developmentHistone deacetylase 1Homo sapiens (human)
positive regulation of cell population proliferationHistone deacetylase 1Homo sapiens (human)
epidermal cell differentiationHistone deacetylase 1Homo sapiens (human)
positive regulation of gene expressionHistone deacetylase 1Homo sapiens (human)
negative regulation of gene expressionHistone deacetylase 1Homo sapiens (human)
hippocampus developmentHistone deacetylase 1Homo sapiens (human)
neuron differentiationHistone deacetylase 1Homo sapiens (human)
negative regulation of cell migrationHistone deacetylase 1Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayHistone deacetylase 1Homo sapiens (human)
circadian regulation of gene expressionHistone deacetylase 1Homo sapiens (human)
cellular response to platelet-derived growth factor stimulusHistone deacetylase 1Homo sapiens (human)
odontogenesis of dentin-containing toothHistone deacetylase 1Homo sapiens (human)
regulation of cell fate specificationHistone deacetylase 1Homo sapiens (human)
embryonic digit morphogenesisHistone deacetylase 1Homo sapiens (human)
negative regulation of apoptotic processHistone deacetylase 1Homo sapiens (human)
negative regulation of canonical NF-kappaB signal transductionHistone deacetylase 1Homo sapiens (human)
negative regulation by host of viral transcriptionHistone deacetylase 1Homo sapiens (human)
negative regulation of gene expression, epigeneticHistone deacetylase 1Homo sapiens (human)
negative regulation of DNA-templated transcriptionHistone deacetylase 1Homo sapiens (human)
positive regulation of DNA-templated transcriptionHistone deacetylase 1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIHistone deacetylase 1Homo sapiens (human)
positive regulation of smooth muscle cell proliferationHistone deacetylase 1Homo sapiens (human)
oligodendrocyte differentiationHistone deacetylase 1Homo sapiens (human)
positive regulation of oligodendrocyte differentiationHistone deacetylase 1Homo sapiens (human)
negative regulation of androgen receptor signaling pathwayHistone deacetylase 1Homo sapiens (human)
hair follicle placode formationHistone deacetylase 1Homo sapiens (human)
eyelid development in camera-type eyeHistone deacetylase 1Homo sapiens (human)
fungiform papilla formationHistone deacetylase 1Homo sapiens (human)
negative regulation of canonical Wnt signaling pathwayHistone deacetylase 1Homo sapiens (human)
negative regulation of stem cell population maintenanceHistone deacetylase 1Homo sapiens (human)
positive regulation of stem cell population maintenanceHistone deacetylase 1Homo sapiens (human)
regulation of stem cell differentiationHistone deacetylase 1Homo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathwayHistone deacetylase 1Homo sapiens (human)
heterochromatin formationHistone deacetylase 1Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 7Homo sapiens (human)
vasculogenesisHistone deacetylase 7Homo sapiens (human)
chromatin remodelingHistone deacetylase 7Homo sapiens (human)
protein deacetylationHistone deacetylase 7Homo sapiens (human)
cell-cell junction assemblyHistone deacetylase 7Homo sapiens (human)
protein sumoylationHistone deacetylase 7Homo sapiens (human)
negative regulation of interleukin-2 productionHistone deacetylase 7Homo sapiens (human)
negative regulation of osteoblast differentiationHistone deacetylase 7Homo sapiens (human)
regulation of mRNA processingHistone deacetylase 7Homo sapiens (human)
positive regulation of cell migration involved in sprouting angiogenesisHistone deacetylase 7Homo sapiens (human)
negative regulation of non-canonical NF-kappaB signal transductionHistone deacetylase 7Homo 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)
positive regulation of signaling receptor activityHistone deacetylase 2Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 2Homo sapiens (human)
response to amphetamineHistone deacetylase 2Homo sapiens (human)
cardiac muscle hypertrophyHistone deacetylase 2Homo sapiens (human)
chromatin remodelingHistone deacetylase 2Homo sapiens (human)
positive regulation of cell population proliferationHistone deacetylase 2Homo sapiens (human)
response to xenobiotic stimulusHistone deacetylase 2Homo sapiens (human)
epidermal cell differentiationHistone deacetylase 2Homo sapiens (human)
positive regulation of epithelial to mesenchymal transitionHistone deacetylase 2Homo sapiens (human)
negative regulation of transcription by competitive promoter bindingHistone deacetylase 2Homo sapiens (human)
negative regulation of neuron projection developmentHistone deacetylase 2Homo sapiens (human)
dendrite developmentHistone deacetylase 2Homo sapiens (human)
negative regulation of cell migrationHistone deacetylase 2Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayHistone deacetylase 2Homo sapiens (human)
response to caffeineHistone deacetylase 2Homo sapiens (human)
heterochromatin formationHistone deacetylase 2Homo sapiens (human)
response to lipopolysaccharideHistone deacetylase 2Homo sapiens (human)
positive regulation of interleukin-1 productionHistone deacetylase 2Homo sapiens (human)
positive regulation of tumor necrosis factor productionHistone deacetylase 2Homo sapiens (human)
circadian regulation of gene expressionHistone deacetylase 2Homo sapiens (human)
positive regulation of collagen biosynthetic processHistone deacetylase 2Homo sapiens (human)
cellular response to heatHistone deacetylase 2Homo sapiens (human)
response to nicotineHistone deacetylase 2Homo sapiens (human)
protein modification processHistone deacetylase 2Homo sapiens (human)
response to cocaineHistone deacetylase 2Homo sapiens (human)
odontogenesis of dentin-containing toothHistone deacetylase 2Homo sapiens (human)
positive regulation of tyrosine phosphorylation of STAT proteinHistone deacetylase 2Homo sapiens (human)
regulation of cell fate specificationHistone deacetylase 2Homo sapiens (human)
embryonic digit morphogenesisHistone deacetylase 2Homo sapiens (human)
negative regulation of apoptotic processHistone deacetylase 2Homo sapiens (human)
negative regulation of DNA-binding transcription factor activityHistone deacetylase 2Homo sapiens (human)
negative regulation of MHC class II biosynthetic processHistone deacetylase 2Homo sapiens (human)
positive regulation of proteolysisHistone deacetylase 2Homo sapiens (human)
negative regulation of DNA-templated transcriptionHistone deacetylase 2Homo sapiens (human)
positive regulation of DNA-templated transcriptionHistone deacetylase 2Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIHistone deacetylase 2Homo sapiens (human)
behavioral response to ethanolHistone deacetylase 2Homo sapiens (human)
positive regulation of oligodendrocyte differentiationHistone deacetylase 2Homo sapiens (human)
response to hyperoxiaHistone deacetylase 2Homo sapiens (human)
hair follicle placode formationHistone deacetylase 2Homo sapiens (human)
negative regulation of dendritic spine developmentHistone deacetylase 2Homo sapiens (human)
eyelid development in camera-type eyeHistone deacetylase 2Homo sapiens (human)
fungiform papilla formationHistone deacetylase 2Homo sapiens (human)
cellular response to hydrogen peroxideHistone deacetylase 2Homo sapiens (human)
cellular response to retinoic acidHistone deacetylase 2Homo sapiens (human)
cellular response to transforming growth factor beta stimulusHistone deacetylase 2Homo sapiens (human)
positive regulation of male mating behaviorHistone deacetylase 2Homo sapiens (human)
negative regulation of stem cell population maintenanceHistone deacetylase 2Homo sapiens (human)
positive regulation of stem cell population maintenanceHistone deacetylase 2Homo sapiens (human)
cellular response to dopamineHistone deacetylase 2Homo sapiens (human)
response to amyloid-betaHistone deacetylase 2Homo sapiens (human)
regulation of stem cell differentiationHistone deacetylase 2Homo sapiens (human)
negative regulation of peptidyl-lysine acetylationHistone deacetylase 2Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIPolyamine deacetylase HDAC10Homo sapiens (human)
DNA repairPolyamine deacetylase HDAC10Homo sapiens (human)
chromatin organizationPolyamine deacetylase HDAC10Homo sapiens (human)
regulation of DNA-templated transcriptionPolyamine deacetylase HDAC10Homo sapiens (human)
macroautophagyPolyamine deacetylase HDAC10Homo sapiens (human)
positive regulation of mismatch repairPolyamine deacetylase HDAC10Homo sapiens (human)
homologous recombinationPolyamine deacetylase HDAC10Homo sapiens (human)
negative regulation of DNA-templated transcriptionPolyamine deacetylase HDAC10Homo sapiens (human)
polyamine deacetylationPolyamine deacetylase HDAC10Homo sapiens (human)
spermidine deacetylationPolyamine deacetylase HDAC10Homo sapiens (human)
epigenetic regulation of gene expressionPolyamine deacetylase HDAC10Homo sapiens (human)
chromatin organizationHistone deacetylase 11 Homo sapiens (human)
oligodendrocyte developmentHistone deacetylase 11 Homo sapiens (human)
epigenetic regulation of gene expressionHistone deacetylase 11 Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 8Homo sapiens (human)
chromatin organizationHistone deacetylase 8Homo sapiens (human)
mitotic sister chromatid cohesionHistone deacetylase 8Homo sapiens (human)
negative regulation of protein ubiquitinationHistone deacetylase 8Homo sapiens (human)
regulation of protein stabilityHistone deacetylase 8Homo sapiens (human)
regulation of telomere maintenanceHistone deacetylase 8Homo sapiens (human)
epigenetic regulation of gene expressionHistone deacetylase 8Homo sapiens (human)
polyamine deacetylationHistone deacetylase 6Homo sapiens (human)
spermidine deacetylationHistone deacetylase 6Homo sapiens (human)
positive regulation of signaling receptor activityHistone deacetylase 6Homo sapiens (human)
protein polyubiquitinationHistone deacetylase 6Homo sapiens (human)
response to amphetamineHistone deacetylase 6Homo sapiens (human)
protein deacetylationHistone deacetylase 6Homo sapiens (human)
protein quality control for misfolded or incompletely synthesized proteinsHistone deacetylase 6Homo sapiens (human)
intracellular protein transportHistone deacetylase 6Homo sapiens (human)
autophagyHistone deacetylase 6Homo sapiens (human)
actin filament organizationHistone deacetylase 6Homo sapiens (human)
negative regulation of microtubule depolymerizationHistone deacetylase 6Homo sapiens (human)
regulation of autophagyHistone deacetylase 6Homo sapiens (human)
positive regulation of epithelial cell migrationHistone deacetylase 6Homo sapiens (human)
negative regulation of hydrogen peroxide metabolic processHistone deacetylase 6Homo sapiens (human)
regulation of macroautophagyHistone deacetylase 6Homo sapiens (human)
axonal transport of mitochondrionHistone deacetylase 6Homo sapiens (human)
negative regulation of protein-containing complex assemblyHistone deacetylase 6Homo sapiens (human)
regulation of protein stabilityHistone deacetylase 6Homo sapiens (human)
protein destabilizationHistone deacetylase 6Homo sapiens (human)
lysosome localizationHistone deacetylase 6Homo sapiens (human)
protein-containing complex disassemblyHistone deacetylase 6Homo sapiens (human)
positive regulation of peptidyl-serine phosphorylationHistone deacetylase 6Homo sapiens (human)
cellular response to heatHistone deacetylase 6Homo sapiens (human)
peptidyl-lysine deacetylationHistone deacetylase 6Homo sapiens (human)
response to immobilization stressHistone deacetylase 6Homo sapiens (human)
cellular response to topologically incorrect proteinHistone deacetylase 6Homo sapiens (human)
erythrocyte enucleationHistone deacetylase 6Homo sapiens (human)
ubiquitin-dependent protein catabolic process via the multivesicular body sorting pathwayHistone deacetylase 6Homo sapiens (human)
negative regulation of protein-containing complex disassemblyHistone deacetylase 6Homo sapiens (human)
regulation of fat cell differentiationHistone deacetylase 6Homo sapiens (human)
negative regulation of gene expression, epigeneticHistone deacetylase 6Homo sapiens (human)
negative regulation of proteolysisHistone deacetylase 6Homo sapiens (human)
negative regulation of DNA-templated transcriptionHistone deacetylase 6Homo sapiens (human)
collateral sproutingHistone deacetylase 6Homo sapiens (human)
negative regulation of axon extension involved in axon guidanceHistone deacetylase 6Homo sapiens (human)
positive regulation of dendrite morphogenesisHistone deacetylase 6Homo sapiens (human)
negative regulation of oxidoreductase activityHistone deacetylase 6Homo sapiens (human)
response to corticosteroneHistone deacetylase 6Homo sapiens (human)
response to misfolded proteinHistone deacetylase 6Homo sapiens (human)
positive regulation of synaptic transmission, glutamatergicHistone deacetylase 6Homo sapiens (human)
cilium assemblyHistone deacetylase 6Homo sapiens (human)
regulation of microtubule-based movementHistone deacetylase 6Homo sapiens (human)
regulation of androgen receptor signaling pathwayHistone deacetylase 6Homo sapiens (human)
dendritic spine morphogenesisHistone deacetylase 6Homo sapiens (human)
cilium disassemblyHistone deacetylase 6Homo sapiens (human)
parkin-mediated stimulation of mitophagy in response to mitochondrial depolarizationHistone deacetylase 6Homo sapiens (human)
regulation of establishment of protein localizationHistone deacetylase 6Homo sapiens (human)
cellular response to hydrogen peroxideHistone deacetylase 6Homo sapiens (human)
aggresome assemblyHistone deacetylase 6Homo sapiens (human)
polyubiquitinated misfolded protein transportHistone deacetylase 6Homo sapiens (human)
response to growth factorHistone deacetylase 6Homo sapiens (human)
cellular response to misfolded proteinHistone deacetylase 6Homo sapiens (human)
cellular response to parathyroid hormone stimulusHistone deacetylase 6Homo sapiens (human)
response to dexamethasoneHistone deacetylase 6Homo sapiens (human)
tubulin deacetylationHistone deacetylase 6Homo sapiens (human)
positive regulation of tubulin deacetylationHistone deacetylase 6Homo sapiens (human)
positive regulation of cellular response to oxidative stressHistone deacetylase 6Homo sapiens (human)
negative regulation of protein acetylationHistone deacetylase 6Homo sapiens (human)
regulation of autophagy of mitochondrionHistone deacetylase 6Homo sapiens (human)
positive regulation of cholangiocyte proliferationHistone deacetylase 6Homo sapiens (human)
negative regulation of aggrephagyHistone deacetylase 6Homo sapiens (human)
epigenetic regulation of gene expressionHistone deacetylase 6Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 9Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 9Homo sapiens (human)
negative regulation of cytokine productionHistone deacetylase 9Homo sapiens (human)
response to amphetamineHistone deacetylase 9Homo sapiens (human)
inflammatory responseHistone deacetylase 9Homo sapiens (human)
heart developmentHistone deacetylase 9Homo sapiens (human)
neuron differentiationHistone deacetylase 9Homo sapiens (human)
B cell differentiationHistone deacetylase 9Homo sapiens (human)
cellular response to insulin stimulusHistone deacetylase 9Homo sapiens (human)
peptidyl-lysine deacetylationHistone deacetylase 9Homo sapiens (human)
B cell activationHistone deacetylase 9Homo sapiens (human)
cholesterol homeostasisHistone deacetylase 9Homo sapiens (human)
negative regulation of gene expression, epigeneticHistone deacetylase 9Homo sapiens (human)
negative regulation of DNA-templated transcriptionHistone deacetylase 9Homo sapiens (human)
regulation of skeletal muscle fiber developmentHistone deacetylase 9Homo sapiens (human)
regulation of striated muscle cell differentiationHistone deacetylase 9Homo sapiens (human)
positive regulation of cell migration involved in sprouting angiogenesisHistone deacetylase 9Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 5Homo sapiens (human)
negative regulation of transcription by RNA polymerase IIHistone deacetylase 5Homo sapiens (human)
inflammatory responseHistone deacetylase 5Homo sapiens (human)
response to xenobiotic stimulusHistone deacetylase 5Homo sapiens (human)
regulation of myotube differentiationHistone deacetylase 5Homo sapiens (human)
negative regulation of myotube differentiationHistone deacetylase 5Homo sapiens (human)
response to activityHistone deacetylase 5Homo sapiens (human)
neuron differentiationHistone deacetylase 5Homo sapiens (human)
B cell differentiationHistone deacetylase 5Homo sapiens (human)
cellular response to insulin stimulusHistone deacetylase 5Homo sapiens (human)
B cell activationHistone deacetylase 5Homo sapiens (human)
response to cocaineHistone deacetylase 5Homo sapiens (human)
regulation of protein bindingHistone deacetylase 5Homo sapiens (human)
negative regulation of gene expression, epigeneticHistone deacetylase 5Homo sapiens (human)
negative regulation of DNA-templated transcriptionHistone deacetylase 5Homo sapiens (human)
positive regulation of transcription by RNA polymerase IIHistone deacetylase 5Homo sapiens (human)
positive regulation of DNA-binding transcription factor activityHistone deacetylase 5Homo sapiens (human)
cellular response to lipopolysaccharideHistone deacetylase 5Homo sapiens (human)
negative regulation of cell migration involved in sprouting angiogenesisHistone deacetylase 5Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (122)

Processvia Protein(s)Taxonomy
transcription corepressor bindingHistone deacetylase 3Homo sapiens (human)
chromatin bindingHistone deacetylase 3Homo sapiens (human)
transcription corepressor activityHistone deacetylase 3Homo sapiens (human)
histone deacetylase activityHistone deacetylase 3Homo sapiens (human)
protein bindingHistone deacetylase 3Homo sapiens (human)
enzyme bindingHistone deacetylase 3Homo sapiens (human)
cyclin bindingHistone deacetylase 3Homo sapiens (human)
chromatin DNA bindingHistone deacetylase 3Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 3Homo sapiens (human)
histone deacetylase bindingHistone deacetylase 3Homo sapiens (human)
NF-kappaB bindingHistone deacetylase 3Homo sapiens (human)
DNA-binding transcription factor bindingHistone deacetylase 3Homo sapiens (human)
protein decrotonylase activityHistone deacetylase 3Homo sapiens (human)
histone decrotonylase activityHistone deacetylase 3Homo sapiens (human)
protein de-2-hydroxyisobutyrylase activityHistone deacetylase 3Homo sapiens (human)
transcription cis-regulatory region bindingBromodomain-containing protein 4Homo sapiens (human)
p53 bindingBromodomain-containing protein 4Homo sapiens (human)
chromatin bindingBromodomain-containing protein 4Homo sapiens (human)
transcription coregulator activityBromodomain-containing protein 4Homo sapiens (human)
transcription coactivator activityBromodomain-containing protein 4Homo sapiens (human)
protein bindingBromodomain-containing protein 4Homo sapiens (human)
RNA polymerase II CTD heptapeptide repeat kinase activityBromodomain-containing protein 4Homo sapiens (human)
enzyme bindingBromodomain-containing protein 4Homo sapiens (human)
lysine-acetylated histone bindingBromodomain-containing protein 4Homo sapiens (human)
RNA polymerase II C-terminal domain bindingBromodomain-containing protein 4Homo sapiens (human)
P-TEFb complex bindingBromodomain-containing protein 4Homo sapiens (human)
histone reader activityBromodomain-containing protein 4Homo 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)
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)
RNA bindingLeukotriene A-4 hydrolaseHomo sapiens (human)
aminopeptidase activityLeukotriene A-4 hydrolaseHomo sapiens (human)
epoxide hydrolase activityLeukotriene A-4 hydrolaseHomo sapiens (human)
leukotriene-A4 hydrolase activityLeukotriene A-4 hydrolaseHomo sapiens (human)
protein bindingLeukotriene A-4 hydrolaseHomo sapiens (human)
peptidase activityLeukotriene A-4 hydrolaseHomo sapiens (human)
zinc ion bindingLeukotriene A-4 hydrolaseHomo sapiens (human)
tripeptide aminopeptidase activityLeukotriene A-4 hydrolaseHomo sapiens (human)
metalloaminopeptidase activityLeukotriene A-4 hydrolaseHomo sapiens (human)
monooxygenase activityCytochrome P450 3A5Homo sapiens (human)
iron ion bindingCytochrome P450 3A5Homo sapiens (human)
protein bindingCytochrome P450 3A5Homo sapiens (human)
retinoic acid 4-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
oxidoreductase activityCytochrome P450 3A5Homo sapiens (human)
oxygen bindingCytochrome P450 3A5Homo sapiens (human)
heme bindingCytochrome P450 3A5Homo sapiens (human)
aromatase activityCytochrome P450 3A5Homo sapiens (human)
estrogen 16-alpha-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
testosterone 6-beta-hydroxylase activityCytochrome P450 3A5Homo sapiens (human)
transcription cis-regulatory region bindingHistone deacetylase 4Homo sapiens (human)
histone bindingHistone deacetylase 4Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingHistone deacetylase 4Homo sapiens (human)
histone deacetylase activityHistone deacetylase 4Homo sapiens (human)
protein bindingHistone deacetylase 4Homo sapiens (human)
zinc ion bindingHistone deacetylase 4Homo sapiens (human)
SUMO transferase activityHistone deacetylase 4Homo sapiens (human)
potassium ion bindingHistone deacetylase 4Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 4Homo sapiens (human)
identical protein bindingHistone deacetylase 4Homo sapiens (human)
histone deacetylase bindingHistone deacetylase 4Homo sapiens (human)
molecular adaptor activityHistone deacetylase 4Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingHistone deacetylase 4Homo sapiens (human)
DNA-binding transcription factor bindingHistone deacetylase 4Homo sapiens (human)
structural molecule activityAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
calcium ion bindingAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
protein bindingAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
structural constituent of muscleAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
electron transfer activityAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
peptidyl-aspartic acid 3-dioxygenase activityAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
transcription cis-regulatory region bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
delayed rectifier potassium channel activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
ubiquitin protein ligase bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
identical protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
protein homodimerization activityPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
C3HC4-type RING finger domain bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
scaffold protein bindingPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel activity involved in ventricular cardiac muscle cell action potential repolarizationPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
nucleosomal DNA bindingHistone deacetylase 1Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingHistone deacetylase 1Homo sapiens (human)
RNA polymerase II core promoter sequence-specific DNA bindingHistone deacetylase 1Homo sapiens (human)
core promoter sequence-specific DNA bindingHistone deacetylase 1Homo sapiens (human)
transcription corepressor bindingHistone deacetylase 1Homo sapiens (human)
p53 bindingHistone deacetylase 1Homo sapiens (human)
transcription corepressor activityHistone deacetylase 1Homo sapiens (human)
histone deacetylase activityHistone deacetylase 1Homo sapiens (human)
protein bindingHistone deacetylase 1Homo sapiens (human)
enzyme bindingHistone deacetylase 1Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 1Homo sapiens (human)
Krueppel-associated box domain bindingHistone deacetylase 1Homo sapiens (human)
histone deacetylase bindingHistone deacetylase 1Homo sapiens (human)
NF-kappaB bindingHistone deacetylase 1Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingHistone deacetylase 1Homo sapiens (human)
E-box bindingHistone deacetylase 1Homo sapiens (human)
DNA-binding transcription factor bindingHistone deacetylase 1Homo sapiens (human)
histone decrotonylase activityHistone deacetylase 1Homo sapiens (human)
promoter-specific chromatin bindingHistone deacetylase 1Homo sapiens (human)
chromatin bindingHistone deacetylase 7Homo sapiens (human)
transcription corepressor activityHistone deacetylase 7Homo sapiens (human)
histone deacetylase activityHistone deacetylase 7Homo sapiens (human)
protein kinase C bindingHistone deacetylase 7Homo sapiens (human)
protein bindingHistone deacetylase 7Homo sapiens (human)
SUMO transferase activityHistone deacetylase 7Homo sapiens (human)
protein kinase bindingHistone deacetylase 7Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 7Homo sapiens (human)
metal ion bindingHistone deacetylase 7Homo sapiens (human)
14-3-3 protein bindingHistone deacetylase 7Homo sapiens (human)
DNA-binding transcription factor bindingHistone deacetylase 7Homo 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)
nucleosomal DNA bindingHistone deacetylase 2Homo sapiens (human)
chromatin bindingHistone deacetylase 2Homo sapiens (human)
RNA bindingHistone deacetylase 2Homo sapiens (human)
histone deacetylase activityHistone deacetylase 2Homo sapiens (human)
protein bindingHistone deacetylase 2Homo sapiens (human)
enzyme bindingHistone deacetylase 2Homo sapiens (human)
heat shock protein bindingHistone deacetylase 2Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 2Homo sapiens (human)
histone bindingHistone deacetylase 2Homo sapiens (human)
histone deacetylase bindingHistone deacetylase 2Homo sapiens (human)
NF-kappaB bindingHistone deacetylase 2Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingHistone deacetylase 2Homo sapiens (human)
histone decrotonylase activityHistone deacetylase 2Homo sapiens (human)
protein de-2-hydroxyisobutyrylase activityHistone deacetylase 2Homo sapiens (human)
promoter-specific chromatin bindingHistone deacetylase 2Homo sapiens (human)
protein lysine deacetylase activityPolyamine deacetylase HDAC10Homo sapiens (human)
histone deacetylase activityPolyamine deacetylase HDAC10Homo sapiens (human)
protein bindingPolyamine deacetylase HDAC10Homo sapiens (human)
zinc ion bindingPolyamine deacetylase HDAC10Homo sapiens (human)
deacetylase activityPolyamine deacetylase HDAC10Homo sapiens (human)
enzyme bindingPolyamine deacetylase HDAC10Homo sapiens (human)
protein lysine deacetylase activityPolyamine deacetylase HDAC10Homo sapiens (human)
histone deacetylase bindingPolyamine deacetylase HDAC10Homo sapiens (human)
acetylputrescine deacetylase activityPolyamine deacetylase HDAC10Homo sapiens (human)
acetylspermidine deacetylase activityPolyamine deacetylase HDAC10Homo sapiens (human)
histone deacetylase activityHistone deacetylase 11 Homo sapiens (human)
protein bindingHistone deacetylase 11 Homo sapiens (human)
DNA-binding transcription factor bindingHistone deacetylase 11 Homo sapiens (human)
histone deacetylase activityHistone deacetylase 8Homo sapiens (human)
protein bindingHistone deacetylase 8Homo sapiens (human)
Hsp70 protein bindingHistone deacetylase 8Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 8Homo sapiens (human)
metal ion bindingHistone deacetylase 8Homo sapiens (human)
Hsp90 protein bindingHistone deacetylase 8Homo sapiens (human)
DNA-binding transcription factor bindingHistone deacetylase 8Homo sapiens (human)
histone decrotonylase activityHistone deacetylase 8Homo sapiens (human)
acetylspermidine deacetylase activityHistone deacetylase 6Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingHistone deacetylase 6Homo sapiens (human)
transcription corepressor bindingHistone deacetylase 6Homo sapiens (human)
actin bindingHistone deacetylase 6Homo sapiens (human)
histone deacetylase activityHistone deacetylase 6Homo sapiens (human)
protein bindingHistone deacetylase 6Homo sapiens (human)
beta-catenin bindingHistone deacetylase 6Homo sapiens (human)
microtubule bindingHistone deacetylase 6Homo sapiens (human)
zinc ion bindingHistone deacetylase 6Homo sapiens (human)
enzyme bindingHistone deacetylase 6Homo sapiens (human)
polyubiquitin modification-dependent protein bindingHistone deacetylase 6Homo sapiens (human)
ubiquitin protein ligase bindingHistone deacetylase 6Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 6Homo sapiens (human)
histone deacetylase bindingHistone deacetylase 6Homo sapiens (human)
tubulin deacetylase activityHistone deacetylase 6Homo sapiens (human)
alpha-tubulin bindingHistone deacetylase 6Homo sapiens (human)
ubiquitin bindingHistone deacetylase 6Homo sapiens (human)
tau protein bindingHistone deacetylase 6Homo sapiens (human)
beta-tubulin bindingHistone deacetylase 6Homo sapiens (human)
misfolded protein bindingHistone deacetylase 6Homo sapiens (human)
Hsp90 protein bindingHistone deacetylase 6Homo sapiens (human)
dynein complex bindingHistone deacetylase 6Homo sapiens (human)
transcription factor bindingHistone deacetylase 6Homo sapiens (human)
transcription corepressor activityHistone deacetylase 9Homo sapiens (human)
histone deacetylase activityHistone deacetylase 9Homo sapiens (human)
protein kinase C bindingHistone deacetylase 9Homo sapiens (human)
protein bindingHistone deacetylase 9Homo sapiens (human)
histone H3K14 deacetylase activityHistone deacetylase 9Homo sapiens (human)
histone H3K9 deacetylase activityHistone deacetylase 9Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 9Homo sapiens (human)
histone H4K16 deacetylase activityHistone deacetylase 9Homo sapiens (human)
histone deacetylase bindingHistone deacetylase 9Homo sapiens (human)
metal ion bindingHistone deacetylase 9Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingHistone deacetylase 9Homo sapiens (human)
DNA-binding transcription factor bindingHistone deacetylase 9Homo sapiens (human)
transcription cis-regulatory region bindingHistone deacetylase 5Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingHistone deacetylase 5Homo sapiens (human)
transcription corepressor bindingHistone deacetylase 5Homo sapiens (human)
chromatin bindingHistone deacetylase 5Homo sapiens (human)
histone deacetylase activityHistone deacetylase 5Homo sapiens (human)
protein kinase C bindingHistone deacetylase 5Homo sapiens (human)
protein bindingHistone deacetylase 5Homo sapiens (human)
protein lysine deacetylase activityHistone deacetylase 5Homo sapiens (human)
identical protein bindingHistone deacetylase 5Homo sapiens (human)
histone deacetylase bindingHistone deacetylase 5Homo sapiens (human)
metal ion bindingHistone deacetylase 5Homo sapiens (human)
RNA polymerase II-specific DNA-binding transcription factor bindingHistone deacetylase 5Homo sapiens (human)
DNA-binding transcription factor bindingHistone deacetylase 5Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (61)

Processvia Protein(s)Taxonomy
nucleusHistone deacetylase 3Homo sapiens (human)
nucleoplasmHistone deacetylase 3Homo sapiens (human)
cytoplasmHistone deacetylase 3Homo sapiens (human)
Golgi apparatusHistone deacetylase 3Homo sapiens (human)
cytosolHistone deacetylase 3Homo sapiens (human)
plasma membraneHistone deacetylase 3Homo sapiens (human)
mitotic spindleHistone deacetylase 3Homo sapiens (human)
histone deacetylase complexHistone deacetylase 3Homo sapiens (human)
transcription repressor complexHistone deacetylase 3Homo sapiens (human)
nucleusHistone deacetylase 3Homo sapiens (human)
condensed nuclear chromosomeBromodomain-containing protein 4Homo sapiens (human)
nucleusBromodomain-containing protein 4Homo sapiens (human)
nucleoplasmBromodomain-containing protein 4Homo 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)
cytoplasmCytochrome P450 3A4Homo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A4Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A4Homo sapiens (human)
extracellular regionLeukotriene A-4 hydrolaseHomo sapiens (human)
nucleoplasmLeukotriene A-4 hydrolaseHomo sapiens (human)
cytosolLeukotriene A-4 hydrolaseHomo sapiens (human)
extracellular exosomeLeukotriene A-4 hydrolaseHomo sapiens (human)
tertiary granule lumenLeukotriene A-4 hydrolaseHomo sapiens (human)
ficolin-1-rich granule lumenLeukotriene A-4 hydrolaseHomo sapiens (human)
cytosolLeukotriene A-4 hydrolaseHomo sapiens (human)
nucleusLeukotriene A-4 hydrolaseHomo sapiens (human)
endoplasmic reticulum membraneCytochrome P450 3A5Homo sapiens (human)
intracellular membrane-bounded organelleCytochrome P450 3A5Homo sapiens (human)
nucleusHistone deacetylase 4Homo sapiens (human)
nucleoplasmHistone deacetylase 4Homo sapiens (human)
cytoplasmHistone deacetylase 4Homo sapiens (human)
cytosolHistone deacetylase 4Homo sapiens (human)
nuclear speckHistone deacetylase 4Homo sapiens (human)
histone deacetylase complexHistone deacetylase 4Homo sapiens (human)
chromatinHistone deacetylase 4Homo sapiens (human)
transcription repressor complexHistone deacetylase 4Homo sapiens (human)
virion membraneSpike glycoproteinSevere acute respiratory syndrome-related coronavirus
endoplasmic reticulumAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
endoplasmic reticulum membraneAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
plasma membraneAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
cortical endoplasmic reticulumAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
sarcoplasmic reticulum membraneAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
endoplasmic reticulumAspartyl/asparaginyl beta-hydroxylaseHomo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
cell surfacePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
perinuclear region of cytoplasmPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
voltage-gated potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
inward rectifier potassium channel complexPotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
plasma membranePotassium voltage-gated channel subfamily H member 2Homo sapiens (human)
nucleusHistone deacetylase 1Homo sapiens (human)
nucleoplasmHistone deacetylase 1Homo sapiens (human)
cytoplasmHistone deacetylase 1Homo sapiens (human)
cytosolHistone deacetylase 1Homo sapiens (human)
NuRD complexHistone deacetylase 1Homo sapiens (human)
neuronal cell bodyHistone deacetylase 1Homo sapiens (human)
Sin3-type complexHistone deacetylase 1Homo sapiens (human)
histone deacetylase complexHistone deacetylase 1Homo sapiens (human)
chromatinHistone deacetylase 1Homo sapiens (human)
heterochromatinHistone deacetylase 1Homo sapiens (human)
transcription repressor complexHistone deacetylase 1Homo sapiens (human)
protein-containing complexHistone deacetylase 1Homo sapiens (human)
nucleusHistone deacetylase 1Homo sapiens (human)
nucleusHistone deacetylase 7Homo sapiens (human)
nucleoplasmHistone deacetylase 7Homo sapiens (human)
cytoplasmHistone deacetylase 7Homo sapiens (human)
cytosolHistone deacetylase 7Homo 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)
chromosome, telomeric regionHistone deacetylase 2Homo sapiens (human)
nucleusHistone deacetylase 2Homo sapiens (human)
nucleoplasmHistone deacetylase 2Homo sapiens (human)
cytoplasmHistone deacetylase 2Homo sapiens (human)
NuRD complexHistone deacetylase 2Homo sapiens (human)
Sin3-type complexHistone deacetylase 2Homo sapiens (human)
histone deacetylase complexHistone deacetylase 2Homo sapiens (human)
chromatinHistone deacetylase 2Homo sapiens (human)
protein-containing complexHistone deacetylase 2Homo sapiens (human)
ESC/E(Z) complexHistone deacetylase 2Homo sapiens (human)
nucleusHistone deacetylase 2Homo sapiens (human)
nucleusPolyamine deacetylase HDAC10Homo sapiens (human)
nucleoplasmPolyamine deacetylase HDAC10Homo sapiens (human)
cytoplasmPolyamine deacetylase HDAC10Homo sapiens (human)
cytosolPolyamine deacetylase HDAC10Homo sapiens (human)
intracellular membrane-bounded organellePolyamine deacetylase HDAC10Homo sapiens (human)
histone deacetylase complexPolyamine deacetylase HDAC10Homo sapiens (human)
nucleusHistone deacetylase 11 Homo sapiens (human)
plasma membraneHistone deacetylase 11 Homo sapiens (human)
histone deacetylase complexHistone deacetylase 11 Homo sapiens (human)
nuclear chromosomeHistone deacetylase 8Homo sapiens (human)
nucleusHistone deacetylase 8Homo sapiens (human)
nucleoplasmHistone deacetylase 8Homo sapiens (human)
cytoplasmHistone deacetylase 8Homo sapiens (human)
histone deacetylase complexHistone deacetylase 8Homo sapiens (human)
nucleusHistone deacetylase 8Homo sapiens (human)
nucleusHistone deacetylase 6Homo sapiens (human)
nucleoplasmHistone deacetylase 6Homo sapiens (human)
cytoplasmHistone deacetylase 6Homo sapiens (human)
multivesicular bodyHistone deacetylase 6Homo sapiens (human)
centrosomeHistone deacetylase 6Homo sapiens (human)
cytosolHistone deacetylase 6Homo sapiens (human)
microtubuleHistone deacetylase 6Homo sapiens (human)
caveolaHistone deacetylase 6Homo sapiens (human)
inclusion bodyHistone deacetylase 6Homo sapiens (human)
aggresomeHistone deacetylase 6Homo sapiens (human)
axonHistone deacetylase 6Homo sapiens (human)
dendriteHistone deacetylase 6Homo sapiens (human)
cell leading edgeHistone deacetylase 6Homo sapiens (human)
ciliary basal bodyHistone deacetylase 6Homo sapiens (human)
perikaryonHistone deacetylase 6Homo sapiens (human)
perinuclear region of cytoplasmHistone deacetylase 6Homo sapiens (human)
axon cytoplasmHistone deacetylase 6Homo sapiens (human)
histone deacetylase complexHistone deacetylase 6Homo sapiens (human)
microtubule associated complexHistone deacetylase 6Homo sapiens (human)
nucleusHistone deacetylase 9Homo sapiens (human)
nucleoplasmHistone deacetylase 9Homo sapiens (human)
cytoplasmHistone deacetylase 9Homo sapiens (human)
histone deacetylase complexHistone deacetylase 9Homo sapiens (human)
transcription regulator complexHistone deacetylase 9Homo sapiens (human)
histone methyltransferase complexHistone deacetylase 9Homo sapiens (human)
nucleusHistone deacetylase 5Homo sapiens (human)
nucleoplasmHistone deacetylase 5Homo sapiens (human)
cytoplasmHistone deacetylase 5Homo sapiens (human)
Golgi apparatusHistone deacetylase 5Homo sapiens (human)
cytosolHistone deacetylase 5Homo sapiens (human)
nuclear speckHistone deacetylase 5Homo sapiens (human)
histone deacetylase complexHistone deacetylase 5Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (316)

Assay IDTitleYearJournalArticle
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
AID686947qHTS for small molecule inhibitors of Yes1 kinase: Primary Screen2013Bioorganic & medicinal chemistry letters, Aug-01, Volume: 23, Issue:15
Identification of potent Yes1 kinase inhibitors using a library screening approach.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
AID1745845Primary qHTS for Inhibitors of ATXN expression
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.
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.
AID496802Inhibition of human HDAC22010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1545757Inhibition of HDAC in human HeLa cell extracts2019European journal of medicinal chemistry, Dec-01, Volume: 183Indole: A privileged scaffold for the design of anti-cancer agents.
AID1845892Inhibition of recombinant human HDAC2 expressed in baculovirus expression system using KI-104 as substrate incubated for 3 hrs by by fluorescence-based assay2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1236462Tmax in iv dosed human measured under phase 1 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1326521Inhibition of full length recombinant human HDAC6 expressed in baculovirus expression system assessed as release of 7-amino-4-methylcoumarin by fluorogenic assay2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID446347Oral bioavailability in CD1 mouse at 15 mg/kg2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
Synthesis and biological evaluation of N-hydroxyphenylacrylamides and N-hydroxypyridin-2-ylacrylamides as novel histone deacetylase inhibitors.
AID619041Inhibition of full length recombinant HDAC1 using Fluor de Lys as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1236460AUC in iv dosed human measured under phase 1/2 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1383992Antiproliferative activity against human PC3 cells after 48 hrs by MTT assay2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1466058Growth inhibition of human A549 cells after 48 hrs by sulforhodamine B assay2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID1421922Antimalarial activity against Plasmodium falciparum 3D7 infected in human erythrocytes assessed as reduction in [3H]-hypoxanthine incorporation preincubated for 48 hrs followed by [3H]-hypoxanthine addition measured after 24 hrs by scintillation counting 2018European journal of medicinal chemistry, Oct-05, Volume: 158One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites.
AID1466057Growth inhibition of human PC3 cells after 48 hrs by sulforhodamine B assay2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID1404348Toxicity in ovariectomized nu/nu mouse implanted with human MCF7 cells assessed as effect on body weight at 10 mg/kg, sc administered once daily for 22 days2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1759720Inhibition of recombinant human FLAG-tagged HDAC3 expressed in human HEK293F cells co-expressing His6-tagged SMRT (1 to 899 residues) using Fluor-de-lys substrate as substrate incubated for 3 hrs followed by substrate addition and measured after 60 mins b2021ACS medicinal chemistry letters, Apr-08, Volume: 12, Issue:4
Redefining the Histone Deacetylase Inhibitor Pharmacophore: High Potency with No Zinc Cofactor Interaction.
AID609495Ratio of IC50 for CYP3A4 to IC50 for human HCT116 cells2011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID1676591Binding affinity to Nickel cation assessed as accounting ratio by measuring total compound detected/total compound adsorbed at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID609490Antiproliferative activity against human HCT116 cells2011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID1421924Cytotoxicity against human NFF cells after 72 hrs by sulforhodamine B assay2018European journal of medicinal chemistry, Oct-05, Volume: 158One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites.
AID618095Toxicity in athymic nu/nu Harlan mouse xenografted with human HCT116 cells assessed as mouse survival at 50 mg/kg, po QD for 21 days measured on day 222011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1236448Inhibition of human HDAC72015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID619051Competitive inhibition of HDAC6 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1676601Binding affinity to Zinc ion assessed as retention ratio by measuring compound detected in elution fraction/total compound detected at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID619259Antitumor activity against human HCT116 cells xenografted in athymic nu/nu Harlan mouse assessed as tumor growth inhibition at 100 mg/kg, po qd for 21 days measured on day 222011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID619042Antiproliferative activity against human COLO205 cells after 96 hrs by celltiter 96 assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID446345Clearance in CD1 mouse at 15 mg/kg, po2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
Synthesis and biological evaluation of N-hydroxyphenylacrylamides and N-hydroxypyridin-2-ylacrylamides as novel histone deacetylase inhibitors.
AID1441632Binding affinity to recombinant human LTA4H hydrolase assessed as change in melting temperature at 50 uM by SYPRO orange dye-based thermofluor assay2017Journal of medicinal chemistry, 03-09, Volume: 60, Issue:5
Drug Repurposing of Histone Deacetylase Inhibitors That Alleviate Neutrophilic Inflammation in Acute Lung Injury and Idiopathic Pulmonary Fibrosis via Inhibiting Leukotriene A4 Hydrolase and Blocking LTB4 Biosynthesis.
AID1441699Inhibition of recombinant human LTA4H aminopeptidase activity expressed in Escherichia coli BL21 (DE3) pLysS assessed as formation of p-NA from Ala-p-NA at 10 uM preincubated for 10 mins followed by substrate addition measured after 10 mins2017Journal of medicinal chemistry, 03-09, Volume: 60, Issue:5
Drug Repurposing of Histone Deacetylase Inhibitors That Alleviate Neutrophilic Inflammation in Acute Lung Injury and Idiopathic Pulmonary Fibrosis via Inhibiting Leukotriene A4 Hydrolase and Blocking LTB4 Biosynthesis.
AID613340Antiproliferative activity against human H1299 cells2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Optimization of the in vitro cardiac safety of hydroxamate-based histone deacetylase inhibitors.
AID1326519Inhibition of HDAC in human PC3 cells assessed as increase in amount of acetylated histone H3 at 0.3 uM after 48 hrs by Western blot analysis2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1677972Inhibition of N-His6-tagged human AspH (315-755) expressed in Escherichia coli BL21 (DE3) using 1 uM hFX-CP as substrate mixture with 3 uM 2OG, 100 uM L-ascorbic acid and 2 uM FAS incubated for 35 mins by MS analysis2020Bioorganic & medicinal chemistry, 10-15, Volume: 28, Issue:20
Small-molecule active pharmaceutical ingredients of approved cancer therapeutics inhibit human aspartate/asparagine-β-hydroxylase.
AID1187258Inhibition of HDAC in human HeLa cells nuclear extracts incubated for 30 mins by fluorescent assay2014European journal of medicinal chemistry, Oct-06, Volume: 85Indole-3-ethylsulfamoylphenylacrylamides: potent histone deacetylase inhibitors with anti-inflammatory activity.
AID619055Competitive inhibition of HDAC10 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1384000Inhibition of HDAC1 (unknown origin)2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1589107Selectivity index, ratio of IC50 for inhibition of human recombinant HDAC1 pre-incubated for 30 mins before substrate addition and measured after 30 mins by fluorescence based assay to IC50 for inhibition of human recombinant HDAC1 pre-incubated for 30 mi2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Exploring hydroxamic acid inhibitors of HDAC1 and HDAC2 using small molecule tools and molecular or homology modelling.
AID1639370Inhibition of HDAC1 (unknown origin) using Ac-Leu-GlyLys(Ac)-AMC as substrate preincubated for 5 mins followed by substrate addition and measured after 30 mins by fluorescence assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID609489Inhibition of purified recombinant HDAC12011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID1204580Induction of apoptosis in human NB4 cells assessed as cell death2015European journal of medicinal chemistry, Jun-15, Volume: 98Synthesis and antiproliferative activity of α-branched α,β-unsaturated ketones in human hematological and solid cancer cell lines.
AID1589101Inhibition Class 1 histone deacetylase in human HeLa nuclear extracts using Fluor-de- Lys-green substrate by fluorescence assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Exploring hydroxamic acid inhibitors of HDAC1 and HDAC2 using small molecule tools and molecular or homology modelling.
AID1676600Binding affinity to zinc ion assessed as accounting ratio by measuring total compound detected/total compound adsorbed at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID496804Inhibition of human HDAC42010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1371081Selectivity index, ratio of IC50 for human NFF cells to IC50 for Plasmodium falciparum2017Journal of medicinal chemistry, 06-22, Volume: 60, Issue:12
Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives.
AID1404298Antiproliferative activity against human HeLa cells after 72 hrs by resazurin dye based fluorescence assay2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1383995Antiproliferative activity against human HEL cells after 48 hrs by MTT assay2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1845898Inhibition of recombinant human HDAC8 expressed in baculovirus expression system using FDL as substrate incubated for 3 hrs by by fluorescence-based assay2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1845893Inhibition of recombinant human HDAC3 expressed in baculovirus expression system using KI-104 as substrate incubated for 3 hrs by by fluorescence-based assay2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID496803Inhibition of human HDAC32010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID619260Antitumor activity against human HCT116 cells xenografted in athymic nu/nu Harlan mouse assessed as tumor growth inhibition at 200 mg/kg, po qd for 21 days measured on day 222011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1187260Anti-inflammatory activity in LPS-stimulated mouse RAW264.7 cells assessed as suppression of TNFalpha production pre-incubated for 1 hr before LPS stimulation for 24 hrs by ELISA method2014European journal of medicinal chemistry, Oct-06, Volume: 85Indole-3-ethylsulfamoylphenylacrylamides: potent histone deacetylase inhibitors with anti-inflammatory activity.
AID324955Inhibition of wild type HDAC4 expressed in Escherichia coli at 2 uM2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Probing the elusive catalytic activity of vertebrate class IIa histone deacetylases.
AID1326509Inhibition of C-terminal His/FLAG-tagged full length recombinant human HDAC1 expressed in baculovirus expression system assessed as release of 7-amino-4-methylcoumarin by fluorogenic assay2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1677974Inhibition of N-His6-tagged human AspH (315-755) expressed in Escherichia coli BL21 (DE3) using 1 uM hFX-CP as substrate mixture with 3 uM 2OG, 100 uM L-ascorbic acid and high 20 uM FAS incubated for 35 mins by MS analysis2020Bioorganic & medicinal chemistry, 10-15, Volume: 28, Issue:20
Small-molecule active pharmaceutical ingredients of approved cancer therapeutics inhibit human aspartate/asparagine-β-hydroxylase.
AID1441700Inhibition of recombinant human LTA4H Epoxide Hydrolase expressed in Escherichia coli BL21 (DE3) pLysS at 10 uM preincubated for 10 mins followed by addition of LTA4 as substrate measured after 15 mins by reverse-phase HPLC analysis2017Journal of medicinal chemistry, 03-09, Volume: 60, Issue:5
Drug Repurposing of Histone Deacetylase Inhibitors That Alleviate Neutrophilic Inflammation in Acute Lung Injury and Idiopathic Pulmonary Fibrosis via Inhibiting Leukotriene A4 Hydrolase and Blocking LTB4 Biosynthesis.
AID1759728Inhibition of class 1 HDAC in quiescent human primary T-cell model of VSV-G pseudotyped HIV-1 latency assessed as reactivation of HIV latency incubated for 24 hrs in presence of 10% FBS by NanoGlo luciferase assay2021ACS medicinal chemistry letters, Apr-08, Volume: 12, Issue:4
Redefining the Histone Deacetylase Inhibitor Pharmacophore: High Potency with No Zinc Cofactor Interaction.
AID1236445Inhibition of human HDAC82015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1589104Potency index, ratio of IC50 for belinostat-1 to IC50 for test compound for inhibition Class 1 histone deacetylase in human HeLa nuclear extracts using Fluor-de- Lys-green substrate by fluorescence assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Exploring hydroxamic acid inhibitors of HDAC1 and HDAC2 using small molecule tools and molecular or homology modelling.
AID613295Antiproliferative activity against human HCT116 cells assessed as growth inhibition2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Optimization of the in vitro cardiac safety of hydroxamate-based histone deacetylase inhibitors.
AID609497Half life in rat liver microsomes2011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID1236471Trypanocidal activity against Trypanosoma brucei brucei assessed as killing of parasite densities of 10'4 to 10'6 cells/ml at 2 uM after 1.5 hrs by clonal dilution method2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID609498Half life in human liver microsomes2011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID1639369Antiproliferative activity against human U937 cells after 48 hrs by MTT assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID1326515Inhibition of HDAC in human PC3 cells assessed as increase in amount of acetylated histone H3 after 48 hrs by Western blot analysis2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1462230Inhibition of BRD4 (unknown origin)2017Bioorganic & medicinal chemistry letters, 09-01, Volume: 27, Issue:17
Structure-based design, synthesis and in vitro antiproliferative effects studies of novel dual BRD4/HDAC inhibitors.
AID1187259Anti-inflammatory activity in LPS-stimulated mouse RAW264.7 cells assessed as suppression of nitric oxide production pre-incubated for 1 hr before LPS stimulation for 24 hrs by Griess reagent based assay2014European journal of medicinal chemistry, Oct-06, Volume: 85Indole-3-ethylsulfamoylphenylacrylamides: potent histone deacetylase inhibitors with anti-inflammatory activity.
AID619048Competitive inhibition of HDAC3 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1466062Inhibition of recombinant human full length HDAC4 using Fluor-de-Lys as substrate after 60 mins by spectrofluorimetric analysis2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID619161Oral bioavailability in nude BALB/c mouse at 50 mg/kg2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1676599Binding affinity to cupric ion assessed as accounting ratio by measuring total compound detected/total compound adsorbed at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1326522Inhibition of His-tagged full length recombinant human HDAC8 expressed in baculovirus expression system assessed as release of 7-amino-4-methylcoumarin by fluorogenic assay2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1326510Antiproliferative activity against human HL60 cells after 48 hrs by SRB assay2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1371079Cytotoxicity against human NFF cells after 72 hrs by SRB assay2017Journal of medicinal chemistry, 06-22, Volume: 60, Issue:12
Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives.
AID1187261Anti-inflammatory activity in LPS-stimulated mouse RAW264.7 cells assessed as suppression of IL6 production pre-incubated for 1 hr before LPS stimulation for 24 hrs by ELISA method2014European journal of medicinal chemistry, Oct-06, Volume: 85Indole-3-ethylsulfamoylphenylacrylamides: potent histone deacetylase inhibitors with anti-inflammatory activity.
AID1759726Fraction unbound in rat plasma2021ACS medicinal chemistry letters, Apr-08, Volume: 12, Issue:4
Redefining the Histone Deacetylase Inhibitor Pharmacophore: High Potency with No Zinc Cofactor Interaction.
AID1676598Binding affinity to cupric ion assessed as retention ratio by measuring compound detected in elution fraction/total compound detected at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID618331Antitumor activity against human HCT116 cells xenografted in nude BALB/c mouse assessed as tumor growth inhibition at 200 mg/kg, po qd for 21 days measured on day 152011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1845897Inhibition of recombinant human HDAC9 expressed in baculovirus expression system using KI-104 as substrate incubated for 3 hrs by by fluorescence-based assay2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1404344Antitumor activity against human MCF7 cells implanted in ovariectomized nu/nu mouse assessed as tumor volume at 10 mg/kg, sc administered once daily for 22 days measured post last dose (Rvb = 869 +/- 173 mm3)2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1236449Inhibition of human HDAC92015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1639365Antiproliferative activity against human PC3 cells after 48 hrs by MTT assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID1236466Half-life in iv dosed human measured under phase 1 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1326518Inhibition of HDAC in human HCT116 cells assessed as increase in amount of acetylated histone H3 at 0.3 uM after 48 hrs by Western blot analysis2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1404347Antitumor activity against human MCF7 cells implanted in ovariectomized nu/nu mouse assessed as tumor growth inhibition at 10 mg/kg, sc administered once daily for 22 days measured post last dose relative to control2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1404343Antitumor activity against human MCF7 cells implanted in ovariectomized nu/nu mouse assessed as tumor volume at 10 mg/kg, sc administered once daily for 22 days measured on day 12 during compound dosing (Rvb = 869 +/- 173 mm3)2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1589103Potency index, ratio of IC50 for panobinostat to IC50 for test compound for inhibition Class 1 histone deacetylase in human HeLa nuclear extracts using Fluor-de- Lys-green substrate by fluorescence assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Exploring hydroxamic acid inhibitors of HDAC1 and HDAC2 using small molecule tools and molecular or homology modelling.
AID1404299Antiproliferative activity against human MCF7 cells after 72 hrs by resazurin dye based fluorescence assay2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID619053Competitive inhibition of HDAC8 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1404345Antitumor activity against human MCF7 cells implanted in ovariectomized nu/nu mouse assessed as tumor weight at 10 mg/kg, sc administered once daily for 22 days measured post last dose (Rvb = 743 +/- 142 mg)2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID609493Inhibition of human ERG by automated patch clamp assay2011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID619044Antiproliferative activity against human HCT116 cells after 96 hrs by celltiter 96 assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1676589Binding affinity to Nickel cation assessed as performance ratio ratio by measuring product of accounting ratio and retention ratio at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1589102Potency index, ratio of IC50 for BnNHC to IC50 for test compound for inhibition Class 1 histone deacetylase in human HeLa nuclear extracts using Fluor-de- Lys-green substrate by fluorescence assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Exploring hydroxamic acid inhibitors of HDAC1 and HDAC2 using small molecule tools and molecular or homology modelling.
AID1236465Half-life in po dosed human measured under phase 1 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1421923Antimalarial activity against Plasmodium falciparum Dd2 infected in human erythrocytes assessed as reduction in [3H]-hypoxanthine incorporation preincubated for 48 hrs followed by [3H]-hypoxanthine addition measured after 24 hrs by scintillation counting 2018European journal of medicinal chemistry, Oct-05, Volume: 158One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites.
AID1421926Selectivity index, ratio of IC50 for human NFF cells to IC50 for Plasmodium falciparum 3D7 infected in human erythrocytes2018European journal of medicinal chemistry, Oct-05, Volume: 158One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites.
AID1421928Selectivity index, ratio of IC50 for HEK293 cells to IC50 for Plasmodium falciparum 3D7 infected in human erythrocytes2018European journal of medicinal chemistry, Oct-05, Volume: 158One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites.
AID1383989Antiproliferative activity against human Jurkat cells after 48 hrs by MTT assay2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1441630Inhibition of recombinant human LTA4H aminopeptidase activity expressed in Escherichia coli BL21 (DE3) pLysS assessed as formation of p-NA from Ala-p-NA preincubated for 10 mins followed by substrate addition measured after 10 mins2017Journal of medicinal chemistry, 03-09, Volume: 60, Issue:5
Drug Repurposing of Histone Deacetylase Inhibitors That Alleviate Neutrophilic Inflammation in Acute Lung Injury and Idiopathic Pulmonary Fibrosis via Inhibiting Leukotriene A4 Hydrolase and Blocking LTB4 Biosynthesis.
AID1633989Inhibition of HDAC1 in human HuH7 cells assessed as increase in histone H3 K9/K14 acetylation at varying concentrations incubated for 24 hrs by Western blot analysis2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Synthesis of N'-propylhydrazide analogs of hydroxamic inhibitors of histone deacetylases (HDACs) and evaluation of their impact on activities of HDACs and replication of hepatitis C virus (HCV).
AID1236457AUC in po dosed human measured under phase 1 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1633990Inhibition of HDAC2 in human HuH7 cells assessed as increase in histone H3 K9/K14 acetylation at varying concentrations incubated for 24 hrs by Western blot analysis2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Synthesis of N'-propylhydrazide analogs of hydroxamic inhibitors of histone deacetylases (HDACs) and evaluation of their impact on activities of HDACs and replication of hepatitis C virus (HCV).
AID1901805Cytotoxicity against human HD-MB03 cells assessed as inhibition of cell growth incubated for 72 hrs measured by spectrophotometer analysis2022Journal of medicinal chemistry, 02-24, Volume: 65, Issue:4
Discovery of HDAC6-Selective Inhibitor NN-390 with
AID1466063Inhibition of recombinant human full length HDAC6 using Fluor-de-Lys as substrate after 60 mins by spectrofluorimetric analysis2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID1236446Inhibition of human HDAC42015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1404346Antitumor activity against human MCF7 cells implanted in ovariectomized nu/nu mouse assessed as tumor volume at 10 mg/kg, sc administered once daily for 22 days measured post last dose relative to control2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1466060Growth inhibition of human AsPC1 cells after 48 hrs by sulforhodamine B assay2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID609496Cardiac safety index, Ratio of IC50 for human ERG to IC50 for human HCT116 cells2011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID1466059Growth inhibition of human MDA-MB-231 cells after 48 hrs by sulforhodamine B assay2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID1187262Anti-inflammatory activity in LPS-stimulated mouse RAW264.7 cells assessed as suppression of PGE2 production pre-incubated for 1 hr before LPS stimulation for 24 hrs by enzyme immunoassay method2014European journal of medicinal chemistry, Oct-06, Volume: 85Indole-3-ethylsulfamoylphenylacrylamides: potent histone deacetylase inhibitors with anti-inflammatory activity.
AID1633992Inhibition of HDAC6 in human HuH7 cells assessed as increase in alpha-tubulin K40 acetylation at varying concentrations incubated for 24 hrs by Western blot analysis2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Synthesis of N'-propylhydrazide analogs of hydroxamic inhibitors of histone deacetylases (HDACs) and evaluation of their impact on activities of HDACs and replication of hepatitis C virus (HCV).
AID496807Inhibition of human HDAC72010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1466056Inhibition of HDAC in human HeLa cell nuclear extract using Ac-Lys(Ac)-pNA as substrate after 30 mins by fluorescence assay2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID1404297Antiproliferative activity against human A549 cells after 72 hrs by resazurin dye based fluorescence assay2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1236450Inhibition of human HDAC62015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID639364Clearance in mouse2011Journal of medicinal chemistry, Apr-28, Volume: 54, Issue:8
Discovery, synthesis, and pharmacological evaluation of spiropiperidine hydroxamic acid based derivatives as structurally novel histone deacetylase (HDAC) inhibitors.
AID1676590Binding affinity to Nickel cation assessed as retention ratio by measuring compound detected in elution fraction/total compound detected at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID618086Toxicity in athymic nu/nu Harlan mouse xenografted with human HCT116 cells assessed as body weight loss at 200 mg/kg, po QD for 7 days measured on day 52011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1676594Binding affinity to gallium ion assessed as accounting ratio by measuring total compound detected/total compound adsorbed at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID496801Inhibition of human HDAC12010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID750109Inhibition of HDAC in human HeLa cells using Fluor de Lys as substrate by fluorescence assay2013Bioorganic & medicinal chemistry letters, Jun-01, Volume: 23, Issue:11
Design, synthesis and biological evaluation of indeno[1,2-d]thiazole derivatives as potent histone deacetylase inhibitors.
AID1326512Antiproliferative activity against human PC3 cells after 48 hrs by SRB assay2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID496808Activity of human HDAC82010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID496806Inhibition of human HDAC62010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1236456Cmax in iv dosed human measured under phase 1/2 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1326516Inhibition of HDAC in human HCT116 cells assessed as increase in amount of acetylated alpha-tubulin after 48 hrs by Western blot analysis2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID618097Toxicity in athymic nu/nu Harlan mouse xenografted with human HCT116 cells assessed as mouse survival at 200 mg/kg, po QD for 21 days measured on day 222011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1189854Therapeutic index, ratio of CC50 for human HuH7 cells to EC50 for HCV genotype 1b infected in human Huh7 cells2015Journal of medicinal chemistry, Jan-22, Volume: 58, Issue:2
Hydroxamic acids block replication of hepatitis C virus.
AID1676597Binding affinity to cupric ion assessed as performance ratio ratio by measuring product of accounting ratio and retention ratio at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1236472Trypanocidal activity against Trypanosoma brucei brucei assessed as killing of parasite densities of 10'4 to 10'6 cells/ml at 180 uM after 4 hrs by clonal dilution method2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1639362Inhibition of HDAC in human HeLa nuclear extract using Boc-Lys(Ac)-AMC as substrate preincubated for 5 mins followed by substrate addition and measured after 30 mins by fluorescence assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID1236443Inhibition of human HDAC22015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1236453Cmax in po dosed human measured under phase 1 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1236447Inhibition of human HDAC52015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID619047Competitive inhibition of HDAC2 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID619052Competitive inhibition of HDAC7 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1404296Antiproliferative activity against human MDA-MB-231 cells after 72 hrs by resazurin dye based fluorescence assay2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1404300Antiproliferative activity against human SK-MEL-28 cells after 72 hrs by resazurin dye based fluorescence assay2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1383993Antiproliferative activity against human HeLa cells after 48 hrs by MTT assay2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1404302Cytotoxicity against human MCF10A cells assessed as reduction in cell proliferation at 10 uM after 72 hrs by resazurin dye based fluorescence assay2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1441631Inhibition of recombinant human LTA4H Epoxide Hydrolase expressed in Escherichia coli BL21 (DE3) pLysS preincubated for 10 mins followed by addition of LTA4 as substrate measured after 15 mins by reverse-phase HPLC analysis2017Journal of medicinal chemistry, 03-09, Volume: 60, Issue:5
Drug Repurposing of Histone Deacetylase Inhibitors That Alleviate Neutrophilic Inflammation in Acute Lung Injury and Idiopathic Pulmonary Fibrosis via Inhibiting Leukotriene A4 Hydrolase and Blocking LTB4 Biosynthesis.
AID1236458AUC in iv dosed human measured under phase 1 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1236454Cmax in iv dosed human measured under phase 1 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1371034Antimalarial activity against Plasmodium falciparum infected in human erythrocytes preincubated for 48 hrs followed by [3H]-hypoxanthine addition measured after 24 hrs by scintillation counting assay2017Journal of medicinal chemistry, 06-22, Volume: 60, Issue:12
Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives.
AID619046Competitive inhibition of HDAC1 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1236441Antitrypanosomal activity against Trypanosoma brucei brucei 427 assessed as inhibition of parasite proliferation measured as ATP levels after 48 hrs by luciferase-based assay2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1702237Oral bioavailability in Balb/c mouse at 50 mg/kg measured up to 24 hrs by LC/MS/MS analysis2018Journal of medicinal chemistry, 02-22, Volume: 61, Issue:4
Design, Synthesis, and Preclinical Evaluation of Fused Pyrimidine-Based Hydroxamates for the Treatment of Hepatocellular Carcinoma.
AID496809Inhibition of human HDAC92010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1901784Therapeutic window, ratio of IC50 for human fetal neuronal stem cells to IC50 for human HD-MB03 cells2022Journal of medicinal chemistry, 02-24, Volume: 65, Issue:4
Discovery of HDAC6-Selective Inhibitor NN-390 with
AID1404339Inhibition of HDAC in human cell lysates using fluoro-substrate peptide/fluoro-deacetylated peptide as substrate incubated for 20 mins measured at 1 to 2 mins interval for 30 to 60 mins by fluorescence assay2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1326517Inhibition of HDAC in human PC3 cells assessed as increase in amount of acetylated alpha-tubulin after 48 hrs by Western blot analysis2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1371032Inhibition of HDAC1 in Plasmodium falciparum 3D7 nuclear extract using Ac-RGK(Ac)-AMC fluorogenic peptide as substrate preincubated for 1 hr followed by substrate addition measured after 10 min by fluorescence assay2017Journal of medicinal chemistry, 06-22, Volume: 60, Issue:12
Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives.
AID1525776Inhibition of HADC6 (unknown origin)2020Journal of medicinal chemistry, 01-09, Volume: 63, Issue:1
Old but Gold: Tracking the New Guise of Histone Deacetylase 6 (HDAC6) Enzyme as a Biomarker and Therapeutic Target in Rare Diseases.
AID1466064Inhibition of recombinant human full length HDAC8 using Fluor-de-Lys as substrate after 60 mins by spectrofluorimetric analysis2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID1639364Antiproliferative activity against human SK-N-BE(2) cells after 48 hrs by MTT assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID1639368Antiproliferative activity against human HEL cells after 48 hrs by MTT assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID324950Inhibition of HDAC4 H976Y mutant expressed in Escherichia coli2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Probing the elusive catalytic activity of vertebrate class IIa histone deacetylases.
AID1383994Antiproliferative activity against human K562 cells after 48 hrs by MTT assay2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1677975Inhibition of N-His6-tagged human AspH (315-755) expressed in Escherichia coli BL21 (DE3) using high 10 uM hFX-CP as substrate mixture with 10 uM 2OG, 100 uM L-ascorbic acid and 2 uM FAS incubated for 35 mins by MS analysis2020Bioorganic & medicinal chemistry, 10-15, Volume: 28, Issue:20
Small-molecule active pharmaceutical ingredients of approved cancer therapeutics inhibit human aspartate/asparagine-β-hydroxylase.
AID1845896Inhibition of recombinant human HDAC7 expressed in baculovirus expression system using KI-104 as substrate incubated for 3 hrs by by fluorescence-based assay2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1676588Binding affinity to Zinc ion assessed as performance ratio ratio by measuring product of accounting ratio and retention ratio at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1639366Antiproliferative activity against human HT-29 cells after 48 hrs by MTT assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID613341Inhibition of human ERG by automated Q-patch assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Optimization of the in vitro cardiac safety of hydroxamate-based histone deacetylase inhibitors.
AID1371053Antitrypanosomal activity against Trypanosoma brucei brucei measured after 22 hrs by fluorescence-based Alamar blue viability assay2017Journal of medicinal chemistry, 06-22, Volume: 60, Issue:12
Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives.
AID1421929Selectivity index, ratio of IC50 for HEK293 cells to IC50 for Plasmodium falciparum Dd2 infected in human erythrocytes2018European journal of medicinal chemistry, Oct-05, Volume: 158One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites.
AID1404349Drug uptake in tumor of ovariectomized nu/nu mouse implanted with human MCF7 cells at 10 mg/kg, sc administered once daily for 22 days measured post last dose by HPLC-MS/MS analysis2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID446342AUC (0 to infinity) in CD1 mouse at 5 mg/kg, iv2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
Synthesis and biological evaluation of N-hydroxyphenylacrylamides and N-hydroxypyridin-2-ylacrylamides as novel histone deacetylase inhibitors.
AID1589106Inhibition of human recombinant HDAC2 pre-incubated for 30 mins before substrate addition and measured after 30 mins by fluorescence based assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Exploring hydroxamic acid inhibitors of HDAC1 and HDAC2 using small molecule tools and molecular or homology modelling.
AID1204590Acceleration of RA-induced granulocytic differentiation in human NB4 cells in presence of all-trans retinoic acid relative to control2015European journal of medicinal chemistry, Jun-15, Volume: 98Synthesis and antiproliferative activity of α-branched α,β-unsaturated ketones in human hematological and solid cancer cell lines.
AID1759721Inhibition of class 1 HDAC in human Jurkat 2C4 model of HIV latency assessed as reactivation of HIV latency incubated for 18 to 24 hrs in presence of 0.1 % heat inactivated NHS by Steady-Glo luciferase assay2021ACS medicinal chemistry letters, Apr-08, Volume: 12, Issue:4
Redefining the Histone Deacetylase Inhibitor Pharmacophore: High Potency with No Zinc Cofactor Interaction.
AID1639363Antiproliferative activity against human HeLa cells after 48 hrs by MTT assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID1676596Binding affinity to Ferric ion assessed as retention ratio by measuring compound detected in elution fraction/total compound detected at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1404301Antiproliferative activity against human NCI-H460 cells after 72 hrs by resazurin dye based fluorescence assay2018ACS medicinal chemistry letters, Feb-08, Volume: 9, Issue:2
Biocompatible Boron-Containing Prodrugs of Belinostat for the Potential Treatment of Solid Tumors.
AID1639373Inhibition of HDAC11 (unknown origin) using Ac-Leu-GlyLys(Ac)-AMC as substrate preincubated for 5 mins followed by substrate addition and measured after 30 mins by fluorescence assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID619050Competitive inhibition of HDAC5 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID446346Volume of distribution at steady state in CD1 mouse at 15 mg/kg, po2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
Synthesis and biological evaluation of N-hydroxyphenylacrylamides and N-hydroxypyridin-2-ylacrylamides as novel histone deacetylase inhibitors.
AID619049Competitive inhibition of HDAC4 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1326513Antiproliferative activity against human A549 cells after 48 hrs by SRB assay2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID324956Inhibition of HDAC6 in HEK293 cells2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Probing the elusive catalytic activity of vertebrate class IIa histone deacetylases.
AID1383991Antiproliferative activity against human SK-N-BE(2) cells after 48 hrs by MTT assay2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1383990Antiproliferative activity against human MOLT4 cells after 48 hrs by MTT assay2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1421927Selectivity index, ratio of IC50 for human NFF cells to IC50 for Plasmodium falciparum Dd2 infected in human erythrocytes2018European journal of medicinal chemistry, Oct-05, Volume: 158One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites.
AID1676593Binding affinity to Gallium ion assessed as retention ratio by measuring compound detected in elution fraction/total compound detected at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1901780Competitive inhibition of N-terminal His-SUMO tagged zebra fish recombinant HDAC6 catalytic domain 2 transfected with pET28b(+) vector in Escherichia coli BL21 (DE3) RILP cells incubated for 10 mins followed by addition of FITC-M344 measured af ter 10 min2022Journal of medicinal chemistry, 02-24, Volume: 65, Issue:4
Discovery of HDAC6-Selective Inhibitor NN-390 with
AID1383985Inhibition of HDAC1/2 in human HeLa nuclear extract using Boc-Lys(acetyl)-AMC as substrate preincubated for 5 mins followed by substrate addition measured after 30 mins by fluorescence assay2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1236468Half-life in iv dosed human measured under phase 1/2 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1236461Tmax in po dosed human measured under phase 1 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID619056Competitive inhibition of HDAC11 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID446344Half life in CD1 mouse at 15 mg/kg, po2010Journal of medicinal chemistry, Jan-28, Volume: 53, Issue:2
Synthesis and biological evaluation of N-hydroxyphenylacrylamides and N-hydroxypyridin-2-ylacrylamides as novel histone deacetylase inhibitors.
AID1236444Inhibition of human HDAC32015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID324949Inhibition of HDAC3 in HEK293 cells2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Probing the elusive catalytic activity of vertebrate class IIa histone deacetylases.
AID619054Competitive inhibition of HDAC9 using KI-104 as substrate by fluorescence assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1676595Binding affinity to Ferric ion assessed as performance ratio ratio by measuring product of accounting ratio and retention ratio at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1371080Cytotoxicity against HEK293 cells after 48 hrs by resazurin assay2017Journal of medicinal chemistry, 06-22, Volume: 60, Issue:12
Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives.
AID1466055Inhibition of recombinant human full length HDAC2 using Fluor-de-Lys as substrate after 60 mins by spectrofluorimetric analysis2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID609494Inhibition of CYP3A42011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID1326511Antiproliferative activity against human HCT116 cells after 48 hrs by SRB assay2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1639371Inhibition of HDAC6 (unknown origin) using Ac-Leu-GlyLys(Ac)-AMC as substrate preincubated for 5 mins followed by substrate addition and measured after 30 mins by fluorescence assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID1326520Inhibition of full length recombinant human HDAC2 expressed in baculovirus expression system assessed as release of 7-amino-4-methylcoumarin by fluorogenic assay2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1639367Antiproliferative activity against human K562 cells after 48 hrs by MTT assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID1676602Binding affinity to ferric ion assessed as accounting ratio by measuring total compound detected/total compound adsorbed at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1421925Cytotoxicity against HEK293 cells after 48 hrs by resazurin dye based assay2018European journal of medicinal chemistry, Oct-05, Volume: 158One-pot, multi-component synthesis and structure-activity relationships of peptoid-based histone deacetylase (HDAC) inhibitors targeting malaria parasites.
AID1384001Inhibition of HDAC6 (unknown origin)2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1677973Inhibition of N-His6-tagged human AspH (315-755) expressed in Escherichia coli BL21 (DE3) using 1 uM hFX-CP as substrate mixture with high 200 uM 2OG, 100 uM L-ascorbic acid and 2 uM FAS incubated for 35 mins by MS analysis2020Bioorganic & medicinal chemistry, 10-15, Volume: 28, Issue:20
Small-molecule active pharmaceutical ingredients of approved cancer therapeutics inhibit human aspartate/asparagine-β-hydroxylase.
AID496805Inhibition of human HDAC52010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID613298In vitro cardiac safety index, ratio of IC50 for human ERG to IC50 for human HCT116 cells2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Optimization of the in vitro cardiac safety of hydroxamate-based histone deacetylase inhibitors.
AID619043Antiproliferative activity against human A2780 cells after 96 hrs by celltiter 96 assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1189850Cytotoxicity against human HuH7 cells assessed as inhibition of cell viability after 3 days by CellTiter 96 assay2015Journal of medicinal chemistry, Jan-22, Volume: 58, Issue:2
Hydroxamic acids block replication of hepatitis C virus.
AID618096Toxicity in athymic nu/nu Harlan mouse xenografted with human HCT116 cells assessed as mouse survival at 100 mg/kg, po QD for 21 days measured on day 222011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1236464Tmax in iv dosed human measured under phase 1/2 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1633993Antiviral activity against HCV infected in human HuH7-luc/neo cells assessed as inhibition of DNA replication incubated for 3 days by luciferase reporter gene assay2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Synthesis of N'-propylhydrazide analogs of hydroxamic inhibitors of histone deacetylases (HDACs) and evaluation of their impact on activities of HDACs and replication of hepatitis C virus (HCV).
AID609492Thermodynamic solubility of the compound at pH 6.82011Bioorganic & medicinal chemistry letters, Aug-15, Volume: 21, Issue:16
The design, synthesis and structure-activity relationships of novel isoindoline-based histone deacetylase inhibitors.
AID1676592Binding affinity to Gallium ion assessed as performance ratio ratio by measuring product of accounting ratio and retention ratio at 2.55 umol by immobilized metal-ion affinity chromatography2020Journal of medicinal chemistry, 10-22, Volume: 63, Issue:20
Immobilized Metal Affinity Chromatography as a Drug Discovery Platform for Metalloenzyme Inhibitors.
AID1633991Inhibition of HDAC3 in human HuH7 cells assessed as increase in histone H3 K9/K14acetylation at varying concentrations incubated for 24 hrs by Western blot analysis2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Synthesis of N'-propylhydrazide analogs of hydroxamic inhibitors of histone deacetylases (HDACs) and evaluation of their impact on activities of HDACs and replication of hepatitis C virus (HCV).
AID1845895Inhibition of recombinant human HDAC6 expressed in baculovirus expression system using KI-104 as substrate incubated for 3 hrs by by fluorescence-based assay2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID619258Antitumor activity against human HCT116 cells xenografted in athymic nu/nu Harlan mouse assessed as tumor growth inhibition at 50 mg/kg, po qd for 21 days measured on day 222011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID316875Inhibition of HDAC from human HeLa cells2008Journal of medicinal chemistry, Mar-27, Volume: 51, Issue:6
Histone deacetylase inhibitors: from bench to clinic.
AID1639372Inhibition of HDAC8 (unknown origin) using Ac-Leu-GlyLys(Ac)-AMC as substrate preincubated for 5 mins followed by substrate addition and measured after 30 mins by fluorescence assay2019Bioorganic & medicinal chemistry, 04-15, Volume: 27, Issue:8
Design, synthesis and activity evaluation of indole-based double - Branched HDAC1 inhibitors.
AID619045Antiproliferative activity against human PC3 cells after 96 hrs by celltiter 96 assay2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1189851Antiviral activity against HCV genotype 1b infected in human Huh7 cells after 3 days by luciferase reporter gene assay2015Journal of medicinal chemistry, Jan-22, Volume: 58, Issue:2
Hydroxamic acids block replication of hepatitis C virus.
AID324948Inhibition of HDAC1 in HEK293 cells2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Probing the elusive catalytic activity of vertebrate class IIa histone deacetylases.
AID1677971Inhibition of N-His6-tagged human AspH (315-755) expressed in Escherichia coli BL21 (DE3) at 20uM using 1 uM hFX-CP as substrate mixture with 3 uM 2OG, 100 uM L-ascorbic acid and 2 uM FAS incubated for 35 mins by MS analysis2020Bioorganic & medicinal chemistry, 10-15, Volume: 28, Issue:20
Small-molecule active pharmaceutical ingredients of approved cancer therapeutics inhibit human aspartate/asparagine-β-hydroxylase.
AID1236442Inhibition of human HDAC12015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID613343Inhibition of human ERG at 30 uM2011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Optimization of the in vitro cardiac safety of hydroxamate-based histone deacetylase inhibitors.
AID1633994Cytotoxicity against human HuH7 cells assessed as reduction in cell viability incubated for 3 days by MTT assay2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Synthesis of N'-propylhydrazide analogs of hydroxamic inhibitors of histone deacetylases (HDACs) and evaluation of their impact on activities of HDACs and replication of hepatitis C virus (HCV).
AID1759727Inhibition of class 1 HDAC in human Jurkat 2C4 model of HIV latency assessed as reactivation of HIV latency incubated for 18 to 24 hrs in presence of 5% heat inactivated NHS by Steady-Glo luciferase assay2021ACS medicinal chemistry letters, Apr-08, Volume: 12, Issue:4
Redefining the Histone Deacetylase Inhibitor Pharmacophore: High Potency with No Zinc Cofactor Interaction.
AID1649884Inhibition of HDAC8 (unknown origin)2019European journal of medicinal chemistry, Feb-15, Volume: 164Histone deacetylase 8 (HDAC8) and its inhibitors with selectivity to other isoforms: An overview.
AID639798Oral bioavailability in mouse2011Journal of medicinal chemistry, Apr-28, Volume: 54, Issue:8
Discovery, synthesis, and pharmacological evaluation of spiropiperidine hydroxamic acid based derivatives as structurally novel histone deacetylase (HDAC) inhibitors.
AID1384002Inhibition of HDAC8 (unknown origin)2018European journal of medicinal chemistry, Apr-25, Volume: 150Discovery of meta-sulfamoyl N-hydroxybenzamides as HDAC8 selective inhibitors.
AID1589105Inhibition of human recombinant HDAC1 pre-incubated for 30 mins before substrate addition and measured after 30 mins by fluorescence based assay2019Bioorganic & medicinal chemistry letters, 09-15, Volume: 29, Issue:18
Exploring hydroxamic acid inhibitors of HDAC1 and HDAC2 using small molecule tools and molecular or homology modelling.
AID1326514Inhibition of HDAC in human HCT116 cells assessed as increase in amount of acetylated histone H3 after 48 hrs by Western blot analysis2016European journal of medicinal chemistry, Oct-21, Volume: 1222-(Phenylsulfonyl)quinoline N-hydroxyacrylamides as potent anticancer agents inhibiting histone deacetylase.
AID1466061Inhibition of recombinant human full length HDAC1 using Fluor-de-Lys as substrate after 60 mins by spectrofluorimetric analysis2017European journal of medicinal chemistry, Jul-07, Volume: 1344-Indolyl-N-hydroxyphenylacrylamides as potent HDAC class I and IIB inhibitors in vitro and in vivo.
AID1371082Selectivity index, ratio of IC50 for HEK293 cells to IC50 for Plasmodium falciparum2017Journal of medicinal chemistry, 06-22, Volume: 60, Issue:12
Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives.
AID618101Antitumor activity against human HCT116 cells xenografted in athymic nude nu/nu CD1 mouse assessed as tumor growth inhibition at 40 mg/kg, ip qd for 7 days measured on day 102011Journal of medicinal chemistry, Jul-14, Volume: 54, Issue:13
Discovery of (2E)-3-{2-butyl-1-[2-(diethylamino)ethyl]-1H-benzimidazol-5-yl}-N-hydroxyacrylamide (SB939), an orally active histone deacetylase inhibitor with a superior preclinical profile.
AID1845894Inhibition of recombinant human HDAC4 expressed in baculovirus expression system using KI-104 as substrate incubated for 3 hrs by by fluorescence-based assay2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1371033Inhibition of recombinant HDAC1 in recombinant Plasmodium falciparum at 1 uM using Ac-RGK(Ac)-AMC fluorogenic peptide as substrate preincubated for 1 hr followed by substrate addition measured after 10 min by fluorescence assay2017Journal of medicinal chemistry, 06-22, Volume: 60, Issue:12
Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives.
AID1236594Inhibition of HDAC class 1 in human Huh-luc/neo7 cells assessed as histone H3 acetylation at 1 uM after 1 to 3 hrs by Western blotting analysis2015Bioorganic & medicinal chemistry letters, Jun-01, Volume: 25, Issue:11
Pyridine hydroxamic acids are specific anti-HCV agents affecting HDAC6.
AID1845891Inhibition of recombinant human HDAC1 expressed in baculovirus expression system using KI-104 as substrate incubated for 3 hrs by by fluorescence-based assay2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1633995Therapeutic index, ratio of CC50 for human HuH7 cells to EC50 for antiviral activity against HCV infected in human HuH7-luc/neo cells assessed as inhibition of DNA replication i2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Synthesis of N'-propylhydrazide analogs of hydroxamic inhibitors of histone deacetylases (HDACs) and evaluation of their impact on activities of HDACs and replication of hepatitis C virus (HCV).
AID1347160Primary screen NINDS Rhodamine qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
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.
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.
AID1347159Primary screen GU Rhodamine qHTS for Zika virus inhibitors: Unlinked NS2B-NS3 protease assay2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1347412qHTS assay to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: Counter screen cell viability and HiBit confirmation2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1745854NCATS anti-infectives library activity on HEK293 viability as a counter-qHTS vs the C. elegans viability qHTS2023Disease models & mechanisms, 03-01, Volume: 16, Issue:3
In vivo quantitative high-throughput screening for drug discovery and comparative toxicology.
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.
AID1802011HDAC6 fluorescence anisotropy assay from Article 10.1038/nchembio.2134: \\Histone deacetylase 6 structure and molecular basis of catalysis and inhibition\\2016Nature chemical biology, 09, Volume: 12, Issue:9
Histone deacetylase 6 structure and molecular basis of catalysis and inhibition.
AID1802061In vitro HDACs Inhibition Fluorescence Assay from Article 10.1111/cbdd.12787: \\PXD101 analogs with L-phenylglycine-containing branched cap as histone deacetylase inhibitors.\\2016Chemical biology & drug design, 10, Volume: 88, Issue:4
PXD101 analogs with L-phenylglycine-containing branched cap as histone deacetylase inhibitors.
AID1798526HDAC Activity Assay from Article 10.1016/j.bmcl.2008.02.025: \\Probing the elusive catalytic activity of vertebrate class IIa histone deacetylases.\\2008Bioorganic & medicinal chemistry letters, Mar-15, Volume: 18, Issue:6
Probing the elusive catalytic activity of vertebrate class IIa histone deacetylases.
AID1802240Enzymatic Inhibitory Assay from Article 10.1111/cbdd.12819: \\Design, synthesis, and preliminary bioactivity evaluation of N(1) -hydroxyterephthalamide derivatives with indole cap as novel histone deacetylase inhibitors.\\2017Chemical biology & drug design, Jan, Volume: 89, Issue:1
Design, synthesis, and preliminary bioactivity evaluation of N
AID1346068Human histone deacetylase 8 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346082Human histone deacetylase 2 (3.5.1.- Histone deacetylases (HDACs))2008The Biochemical journal, Jan-15, Volume: 409, Issue:2
Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors.
AID1346131Human histone deacetylase 7 (3.5.1.- Histone deacetylases (HDACs))2008The Biochemical journal, Jan-15, Volume: 409, Issue:2
Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors.
AID1346104Human histone deacetylase 4 (3.5.1.- Histone deacetylases (HDACs))2008The Biochemical journal, Jan-15, Volume: 409, Issue:2
Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors.
AID1346077Human histone deacetylase 3 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346066Human histone deacetylase 5 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346134Human histone deacetylase 1 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346068Human histone deacetylase 8 (3.5.1.- Histone deacetylases (HDACs))2008The Biochemical journal, Jan-15, Volume: 409, Issue:2
Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors.
AID1346032Human histone deacetylase 6 (3.5.1.- Histone deacetylases (HDACs))2008The Biochemical journal, Jan-15, Volume: 409, Issue:2
Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors.
AID1346090Human histone deacetylase 9 (3.5.1.- Histone deacetylases (HDACs))2008The Biochemical journal, Jan-15, Volume: 409, Issue:2
Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors.
AID1346032Human histone deacetylase 6 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346104Human histone deacetylase 4 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346090Human histone deacetylase 9 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346082Human histone deacetylase 2 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346077Human histone deacetylase 3 (3.5.1.- Histone deacetylases (HDACs))2008The Biochemical journal, Jan-15, Volume: 409, Issue:2
Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors.
AID1346131Human histone deacetylase 7 (3.5.1.- Histone deacetylases (HDACs))2010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1346134Human histone deacetylase 1 (3.5.1.- Histone deacetylases (HDACs))2008The Biochemical journal, Jan-15, Volume: 409, Issue:2
Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors.
AID1347414qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: Secondary screen by immunofluorescence2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (211)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's29 (13.74)29.6817
2010's146 (69.19)24.3611
2020's36 (17.06)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 47.10

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index47.10 (24.57)
Research Supply Index5.48 (2.92)
Research Growth Index5.07 (4.65)
Search Engine Demand Index72.65 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (47.10)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials0 (0.00%)5.53%
Trials30 (14.42%)5.53%
Reviews0 (0.00%)6.00%
Reviews27 (12.98%)6.00%
Case Studies0 (0.00%)4.05%
Case Studies2 (0.96%)4.05%
Observational0 (0.00%)0.25%
Observational0 (0.00%)0.25%
Other8 (100.00%)84.16%
Other149 (71.63%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (54)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
A Phase I/Ib Trial of the CDK4/6 Antagonist Ribociclib And The HDAC Inhibitor Belinostat In Patients With Metastatic Triple Negative Breast Cancer And Recurrent Ovarian Cancer With Response Prediction By Genomics (CHARGE) [NCT04315233]Phase 134 participants (Anticipated)Interventional2021-05-03Recruiting
Phase II Study of PXD101 (NSC-726630) in Relapsed and Refractory Aggressive B-Cell Lymphomas [NCT00303953]Phase 222 participants (Actual)Interventional2006-01-31Completed
A Phase 2 Study of Belinostat and SGI-110 (Guadecitabine) or ASTX727 for the Treatment of Unresectable and Metastatic Conventional Chondrosarcoma [NCT04340843]Phase 232 participants (Anticipated)Interventional2020-09-08Suspended(stopped due to Other - Completed stage 1 accrual)
Quantitative Magnetic Resonance Spectroscopic Imaging (MRSI) to Predict Early Response to Standard Radiation Therapy (RT)/Temozolomide (TMZ) ± Belinostat Therapy in Newly-Diagnosed Glioblastomas (GBM) [NCT02137759]Phase 229 participants (Actual)Interventional2014-05-07Active, not recruiting
The Maximum Tolerated Dose and to Evaluate Safety and Efficacy of Belinostat (PXD-101) in Combination With Paclitaxel Plus Carboplatin in Chemotherapy-Naive Patients With Stage IV Non-Small-Cell Lung Cancer (NSCLC) [NCT01310244]Phase 1/Phase 223 participants (Actual)Interventional2010-12-31Completed
A Phase I Study of Belinostat in Combination With Warfarin in Patients With Solid Tumors or Hematological Malignancies [NCT01317927]Phase 127 participants (Actual)Interventional2010-12-31Completed
A Phase 1 Study of AZD1775 in Combination With Belinostat in Relapsed and Refractory Myeloid Malignancies and Selected Untreated Patients With Acute Myeloid Leukemia [NCT02381548]Phase 120 participants (Actual)Interventional2015-08-18Terminated
Carfilzomib Plus Belinostat in Relapsed/Refractory Non-Hodgkin Lymphoma Subtypes: A Phase 1 Study [NCT02142530]Phase 119 participants (Actual)Interventional2014-10-31Completed
A Phase I/II Safety, Pharmacodynamic, and Pharmacokinetic Study of Intravenously Administered PXD101 Plus Carboplatin or Paclitaxel or Both in Patients With Advanced Solid Tumours [NCT00421889]Phase 1/Phase 280 participants (Actual)Interventional2005-08-31Completed
Phase Ib/II Study to Determine the Recommended Dose, Safety, and Preliminary Efficacy of Belinostat When Used in Combination With Carboplatin, Paclitaxel, and Bevacizumab in Patients With Untreated Non-small Cell Lung Cancer. [NCT01090830]Phase 1/Phase 27 participants (Actual)Interventional2010-04-30Terminated(stopped due to Principal Investigator has left institution. IND withdrawn.)
Pilot Safety and Feasibility Study of an In Vivo Sensitivity Screen Using a Direct Tumor Microinjection Technique and FDG-PET [NCT03432741]Phase 139 participants (Anticipated)Interventional2018-03-27Suspended(stopped due to funding - sponsor filing of Chapter 11 bankruptcy)
A Phase II Study of Binimetinib Plus Belinostat for Subjects With Metastatic Uveal Melanoma [NCT05170334]Phase 232 participants (Anticipated)Interventional2021-12-15Recruiting
A Phase 1 Study for the Evaluation of Excretion (Mass Balance) and Pharmacokinetics of 14C-Labeled Belinostat in Patients With Advanced Cancer [NCT01583777]Phase 16 participants (Actual)Interventional2013-09-30Completed
A Clinical Phase I / II Trial of Belinostat in Combination With Erlotinib in Patients With Non-small Cell Lung Cancer [NCT01188707]Phase 1/Phase 25 participants (Actual)Interventional2010-10-01Terminated(stopped due to Dose over MTD reached)
A Phase 1/2 Study of PXD101 (Belinostat) in Combination With Cisplatin, Doxorubicin and Cyclophosphamide in the First Line Treatment of Advanced or Recurrent Thymic Malignancies [NCT01100944]Phase 1/Phase 226 participants (Actual)Interventional2010-03-04Terminated(stopped due to Principal investigator left the National Institutes of Health (NIH).)
Phase I Pharmacokinetic Study of Belinostat for Solid Tumors and Lymphomas in Patients With Varying Degrees of Hepatic Dysfunction [NCT01273155]Phase 172 participants (Actual)Interventional2011-01-10Completed
An Open-label, Nonrandomized, Phase 1 Study to Evaluate the Safety and Pharmacokinetics of Belinostat in Patients With Relapsed/Refractory Solid Tumors or Hematological Malignancies Who Have Mild, Moderate, and Severe Renal Impairment [NCT02679131]Phase 118 participants (Actual)Interventional2016-03-31Terminated(stopped due to Close out study w/o Cohort D; Severe Impairment (eGFR=15 to <30mL/min/1.73m²))
An Open-label Randomized Phase II Trial of Belinostat (PXD101) in Combination With Carboplatin and Paclitaxel (BelCaP) Compared to Carboplatin and Paclitaxel in Patients With Previously Untreated Carcinoma of Unknown Primary [NCT00873119]Phase 289 participants (Actual)Interventional2009-02-28Completed
A Multicenter Phase II Study of Belinostat (PXD-101) in Previously Chemotherapy Treated Thymoma and Thymic Carcinoma [NCT00589290]Phase 241 participants (Actual)Interventional2007-12-31Completed
Open-label, Nonrandomized, Phase 1 Study Evaluating Safety and Pharmacokinetics of Belinostat in Patients With Relapsed/Refractory Solid Tumors or Hematological Malignancies in Wild-Type, Heterozygous, and Homozygous UGT1A1*28 Genotypes [NCT02680795]Phase 117 participants (Actual)Interventional2016-04-27Completed
Phase 1 Study of Volasertib and Belinostat in Patients With Relapsed and Refractory Aggressive B-cell and T-cell Lymphomas [NCT02875002]Phase 10 participants (Actual)Interventional2016-10-31Withdrawn
A Phase II Clinical Trial of PXD101 in Patients With Recurrent or Refractory Cutaneous and Peripheral T-Cell Lymphomas [NCT00274651]Phase 253 participants (Actual)Interventional2006-01-31Terminated(stopped due to Enrollment stopped prior to reaching expected number of patients, study had accumulated sufficient data to allow a registration study in PTCL (PXD101-CLN-19))
A Phase I Study of Belinostat in Combination With Cisplatin and Etoposide in Adults With a Focus on Small Cell Lung Cancer and Other Cancers of Neuroendocrine Origin [NCT00926640]Phase 128 participants (Actual)Interventional2009-07-01Completed
Phase I Study of Belinostat (PXD-101) and Velcade (Bortezomib) in Relapsed or Refractory Acute Leukemia/ Myelodysplastic Syndrome [NCT01075425]Phase 141 participants (Actual)Interventional2010-05-31Completed
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 Phase I Safety, Pharmacodynamic, Anti-Tumor Activity, and Pharmacokinetic Study of PXD101 Alone and in Combination With 5-Fluorouracil in Patients With Advanced Solid Tumors [NCT00413322]Phase 135 participants (Actual)Interventional2005-09-30Completed
Ph I Study to Evaluate the Safety and Efficacy of Belinostat in Combination With Nivolumab Alone and With Ipilimumab in Patients With Previously Treated Metastatic or Advanced Carcinomas Enriched for ARID1A Loss of Function (Lof) Mutation [NCT04315155]Phase 10 participants (Actual)Interventional2020-10-31Withdrawn(stopped due to study abandoned for funding issues)
RESOLVE : A Phase I Trial of Tremelimumab + Durvalumab(MEDI4736)+ Belinostat in ARID1A Mutated Cancers With Focus on Urothelial Carcinoma [NCT05154994]Phase 19 participants (Anticipated)Interventional2022-01-14Recruiting
Belinostat Combined With Azacitidine/Gemcitabine/Busulfan/Melphalan With Autologous Stem-Cell Transplantation in Refractory or Relapsed Lymphoma [NCT02701673]Phase 1/Phase 20 participants (Actual)Interventional2016-06-30Withdrawn
A Phase II Evaluation of Belinostat (NSC #726630) and Carboplatin (NSC #241240) in the Treatment of Recurrent or Persistent Platinum-Resistant Ovarian, Fallopian Tube, or Primary Peritoneal Cancer [NCT00993616]Phase 229 participants (Actual)Interventional2009-12-31Completed
A Phase I/II Clinical Trial of PXD101 in Combination With Doxorubicin in Patients With Soft Tissue Sarcomas [NCT00878800]Phase 1/Phase 241 participants (Actual)Interventional2006-12-31Completed
A Phase I/II Clinical Trial of PXD101 in Combination With Idarubicin in Patients With AML Not Suitable for Standard Intensive Therapy [NCT00878722]Phase 1/Phase 241 participants (Actual)Interventional2007-08-31Completed
A Phase II Study of Belinostat in Combination With Bortezomib in Patients With Relapsed, Refractory Multiple Myeloma [NCT00431340]Phase 24 participants (Actual)Interventional2007-03-31Terminated(stopped due to Terminated due to dose limiting toxicity)
A Phase I Study of PXD101 in Combination With 17-AAG in Advanced Malignancies [NCT00354185]Phase 136 participants (Actual)Interventional2006-05-31Terminated
A Phase II Trial of Belinostat as Consolidation Therapy With Zidovudine for Adult T-Cell Leukemia-Lymphoma [NCT02737046]Phase 210 participants (Anticipated)Interventional2016-12-12Recruiting
A Phase 1 Trial of PXD101 in Combination With 13-cis-Retinoic Acid in Advanced Solid Tumor Malignancies [NCT00334789]Phase 151 participants (Actual)Interventional2006-06-12Completed
A Phase 1 Study of PXD101 in Combination With Bortezomib (PS-341) in Patients With Advanced Solid Tumors and Lymphoma [NCT00348985]Phase 155 participants (Actual)Interventional2006-03-31Completed
A Phase II Clinical Trial of PXD101 in Patients With Advanced Multiple Myeloma [NCT00131261]Phase 225 participants (Actual)Interventional2005-01-31Completed
Phase 1/Expansion Study of Tazemetostat Plus Belinostat for the Treatment of Relapsed or Refractory Lymphoma [NCT05627245]Phase 164 participants (Anticipated)Interventional2023-03-01Recruiting
A Randomized, Phase IIB, Multicenter, Trial of Oral Azacytidine Plus Romidepsin Versus Investigator's Choice in Patients With Relapse or Refractory Peripheral T-cell Lymphoma (PTCL) [NCT04747236]Phase 250 participants (Anticipated)Interventional2021-02-19Recruiting
Open Label, Dose Escalation Trial of Oral PXD101 in Patients With Advanced Solid Tumors [NCT00413075]Phase 1121 participants (Actual)Interventional2006-06-30Completed
A Phase I/II Study of PXD101 in Patients With Unresectable Hepatocellular Carcinoma With Pharmacokinetic and Pharmacodynamic Evaluation [NCT00321594]Phase 1/Phase 254 participants (Actual)Interventional2006-05-31Completed
Phase II Study of PXD101 (NSC 726630) as Second-Line Therapy for Treatment of Patients With Malignant Pleural Mesothelioma [NCT00365053]Phase 213 participants (Actual)Interventional2006-06-30Completed
A Multicenter, Open-Label Trial of Belinostat in Patients With Relapsed or Refractory Peripheral T-Cell Lymphoma [NCT00865969]Phase 2129 participants (Actual)Interventional2008-12-15Completed
Phase 1 Dose Finding Study of Belinostat Plus Cyclophosphamide/Vincristine/Doxorubicin/Prednisone (CHOP) Regimen (BelCHOP) for Treatment of Patients With Peripheral T-cell Lymphoma(PTCL) [NCT01839097]Phase 123 participants (Actual)Interventional2013-05-31Completed
A Phase 1 Dose-Escalation Trial of Talazoparib in Combination With Belinostat for Metastatic Breast Cancer, Metastatic Castration Resistant Prostate Cancer, and Metastatic Ovarian Cancer [NCT04703920]Phase 125 participants (Anticipated)Interventional2021-03-04Recruiting
A Phase II Exploratory Study of PXD-101(Belinostat) Followed by Zevalin in Patients With Relapsed Aggressive High-Risk Lymphoma [NCT01686165]Phase 25 participants (Actual)Interventional2012-08-31Completed
A Multi-Center, Open-Label, Extension Study to Allow Continued Treatment of Patients Who Have Participated in Spectrum-Sponsored Clinical Studies With Belinostat [NCT04184869]Phase 18 participants (Actual)Interventional2019-08-05Completed
Phase II Study of the Histone Deacetylase Inhibitor PXD101 for the Treatment of Myelodysplastic Syndrome [NCT00357162]Phase 221 participants (Actual)Interventional2006-05-31Completed
A Phase I Study of PXD101 in Combination With Azacitidine (5-Aza) for Advanced Hematologic Malignancies [NCT00351975]Phase 156 participants (Actual)Interventional2006-06-30Completed
A Phase 3, Randomized, Open-Label Study Comparing the Efficacy and Safety of the Combination of Beleodaq-CHOP or Folotyn-COP to the CHOP Regimen Alone in Newly Diagnosed Patients With Peripheral T-Cell Lymphoma [NCT06072131]Phase 3504 participants (Anticipated)Interventional2023-10-04Recruiting
A Phase 2 Study of PXD101 in Patients With Relapsed or Refractory Acute Myelogenous Leukemia or Patients Over 60 With Newly-Diagnosed Acute Myelogenous Leukemia [NCT00357032]Phase 212 participants (Actual)Interventional2006-05-31Completed
A Phase 2 Study of PXD101 in Platinum Resistant Epithelial Ovarian Tumors and Micropapillary/Borderline (LMP) Ovarian Tumors [NCT00301756]Phase 232 participants (Actual)Interventional2006-09-30Completed
A Phase 1 Study of MLN4924 (Pevonedistat) and Belinostat in Relapsed/Refractory Acute Myeloid Leukemia or Myelodysplastic Syndrome [NCT03772925]Phase 130 participants (Anticipated)Interventional2019-06-20Active, not recruiting
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00274651 (5) [back to overview]Duration of Response
NCT00274651 (5) [back to overview]Time to Response
NCT00274651 (5) [back to overview]Time to Progression
NCT00274651 (5) [back to overview]Objective Response Rate in Patients With Recurrent or Refractory Peripheral T-cell Lymphoma (PTCL))
NCT00274651 (5) [back to overview]Objective Response Rate in Patients With Recurrent or Refractory Cutaneous T-cell Lymphoma (CTCL)
NCT00301756 (7) [back to overview]Time to Disease Progression (Low Malignant Potential or Micropapillary / Borderline Ovarian Tumour Group)
NCT00301756 (7) [back to overview]Time to Disease Progression (Epithelial Ovarian Cancer Group)
NCT00301756 (7) [back to overview]Stable Disease Rate, Defined as Neither Sufficient Shrinkage to Qualify for PR Nor Sufficient Increase to Qualify for PD, Taking as Reference the Smallest Sum LD Since the Treatment Started (Low Malignant Potential Group)
NCT00301756 (7) [back to overview]Stable Disease Rate, Defined as Neither Sufficient Shrinkage to Qualify for PR Nor Sufficient Increase to Qualify for PD, Taking as Reference the Smallest Sum LD Since the Treatment Started (Epithelial Ovarian Cancer Group)
NCT00301756 (7) [back to overview]Progression-free Survival
NCT00301756 (7) [back to overview]Efficacy of Belinostat in Terms of Complete or Partial Response; Disappearance of All Target Lesions or at Least a 30% Decrease in the Sum of the Longest Diameter of Target Lesions, Taking as Reference the Baseline Sum LD
NCT00301756 (7) [back to overview]Number of Grade 3 Adverse Events Using the National Cancer Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0
NCT00303953 (3) [back to overview]Overall Survival
NCT00303953 (3) [back to overview]Progression-free Survival
NCT00303953 (3) [back to overview]Assess Number of Patients Who Achieve Confirmed and Unconfirmed Complete Response (CR) or Partial Response (PR)
NCT00321594 (1) [back to overview]Dose-limiting Toxicities (DLT) and Maximum Tolerated Dose (MTD) of Belinostat in Patients With Inoperable HCC (Phase I)
NCT00357032 (3) [back to overview]Toxicity Summary
NCT00357032 (3) [back to overview]Overall Survival
NCT00357032 (3) [back to overview]Complete Response Rate
NCT00357162 (4) [back to overview]Toxicity of Belinostat in Patients With Myelodysplastic Syndrome
NCT00357162 (4) [back to overview]Number of Confirmed Responses (Complete Response, Partial Response, or Hematologic Improvement) Noted on 2 Consecutive Evaluations at Least 4 Weeks Apart
NCT00357162 (4) [back to overview]Time to Progression
NCT00357162 (4) [back to overview]Overall Survival
NCT00365053 (4) [back to overview]Objective Tumor Response Rate According to the Response Evaluation Criteria in Solid Tumors (RECIST) Committee
NCT00365053 (4) [back to overview]Toxicity Profile
NCT00365053 (4) [back to overview]Progression-free Survival
NCT00365053 (4) [back to overview]Overall Survival
NCT00421889 (9) [back to overview]Belinostat AUC (0-infinity)
NCT00421889 (9) [back to overview]Belinostat Cmax
NCT00421889 (9) [back to overview]Belinostat Mean t½
NCT00421889 (9) [back to overview]Best Overall Response (CR or PR)
NCT00421889 (9) [back to overview]Duration of Response
NCT00421889 (9) [back to overview]Time to Progression
NCT00421889 (9) [back to overview]Time to Response
NCT00421889 (9) [back to overview]Dose Limiting Toxicities (DLT), Part A
NCT00421889 (9) [back to overview]Maximum Tolerable Dose (MTD) Belinostat, Part A,
NCT00589290 (2) [back to overview]Number of Participants With Adverse Events
NCT00589290 (2) [back to overview]Number of Participants With a Partial Response
NCT00865969 (7) [back to overview]Duration of Response
NCT00865969 (7) [back to overview]Time to Response
NCT00865969 (7) [back to overview]Time to Progression
NCT00865969 (7) [back to overview]Overall Survival
NCT00865969 (7) [back to overview]Progression Free Survival
NCT00865969 (7) [back to overview]Objective Response Rate
NCT00865969 (7) [back to overview]Number of Participants With At Least One Serious Treatment-Emergent Adverse Event (TEAE)
NCT00873119 (6) [back to overview]Best Overall Response
NCT00873119 (6) [back to overview]Overall Survival (OS)
NCT00873119 (6) [back to overview]Progression Free Survival
NCT00873119 (6) [back to overview]Time to Progression (TTP)
NCT00873119 (6) [back to overview]Time to Response
NCT00873119 (6) [back to overview]Duration of Response
NCT00878722 (11) [back to overview]Duration of Response (CR and PR)
NCT00878722 (11) [back to overview]Elimination t½
NCT00878722 (11) [back to overview]Event-Free Survival
NCT00878722 (11) [back to overview]Maximum Tolerated Dose, Dose Limiting Toxicity
NCT00878722 (11) [back to overview]Overall Response
NCT00878722 (11) [back to overview]Overall Survival
NCT00878722 (11) [back to overview]Relapse-Free Survival
NCT00878722 (11) [back to overview]Remission Duration
NCT00878722 (11) [back to overview]Time to Response (CR and PR)
NCT00878722 (11) [back to overview]Belinostat Cmax
NCT00878722 (11) [back to overview]Belinostat AUC (Area Under Curve)
NCT00878800 (11) [back to overview]Dose Limiting Toxicity (DLT)
NCT00878800 (11) [back to overview]Duration of Response
NCT00878800 (11) [back to overview]Maximum Tolerated Dose (MTD) of Doxorubicin
NCT00878800 (11) [back to overview]Maximum Tolerated Dose (MTD) PXD101
NCT00878800 (11) [back to overview]Objective Response (CR and PR)
NCT00878800 (11) [back to overview]Time to Response
NCT00878800 (11) [back to overview]Time to Progression
NCT00878800 (11) [back to overview]Belinostat AUC (Time 0 to Last Measurement)
NCT00878800 (11) [back to overview]Belinostat Cmax
NCT00878800 (11) [back to overview]Belinostat t½
NCT00878800 (11) [back to overview]Disease Control Rate (CR or PR or SD)
NCT00993616 (3) [back to overview]Frequency and Severity of Observed Adverse Effects Graded According to NCI CTCAE Version 3.0
NCT00993616 (3) [back to overview]Objective Response by Response Evaluation Criteria in Solid Tumors (RECIST) Criteria (Version 1.1)
NCT00993616 (3) [back to overview]Progression Free Survival at 6 Months
NCT01100944 (21) [back to overview]Time to Maximum Plasma Concentration (Tmax)
NCT01100944 (21) [back to overview]Time to Response
NCT01100944 (21) [back to overview]Treatment-related Grade 3 and 4 Adverse Events (Highest Grade Per Event Per Patient)
NCT01100944 (21) [back to overview]Relative Changes in the Number of Tregs With Treatment
NCT01100944 (21) [back to overview]Relative Changes in T Cell Immunoglobulin Domain and Mucin Domain-3 (TIM3)-Expressing Cluster of Differentiation 8 (CD8)+Tcells
NCT01100944 (21) [back to overview]Relative Change Observed in Total Protein Hyperacetylation of Cluster of Differentiation 3 (CD3)+T Cells With Belinostat
NCT01100944 (21) [back to overview]Number of Participants With Serious and Non-serious Adverse Events
NCT01100944 (21) [back to overview]Number of Participants With Grade 3 and 4 Dose Limiting Toxicity (DLT) at 2000mg/m(2) Belinostat
NCT01100944 (21) [back to overview]Clinical Response
NCT01100944 (21) [back to overview]Total Clearance (CL) of Belinostat
NCT01100944 (21) [back to overview]Time to Half Life (t1/2) of Belinostat
NCT01100944 (21) [back to overview]Progression Free Survival (PFS)
NCT01100944 (21) [back to overview]Disease Control Rate (DCR)
NCT01100944 (21) [back to overview]Overall Survival (OS)
NCT01100944 (21) [back to overview]Objective Response Rate (Partial Response (PR) + Complete Response (CR) of Belinostat in Combination With Cisplatin, Doxorubicin and Cyclophosphamide in the First Line Treatment of Patients With Advanced Thymic Malignancies
NCT01100944 (21) [back to overview]Maximum Tolerated Dose (MTD) of Belinostat
NCT01100944 (21) [back to overview]Maximum Plasma Concentration (Cmax)/Dose
NCT01100944 (21) [back to overview]Maximum Observed Plasma Concentration (Cmax) of Belinostat
NCT01100944 (21) [back to overview]Duration of Response
NCT01100944 (21) [back to overview]Area Under the Plasma Concentration vs. Time Curve Extrapolated to Infinity (AUC(INF)/Dose
NCT01100944 (21) [back to overview]Area Under the Plasma Concentration vs. Time Curve Extrapolated to Infinity (AUC(INF))
NCT01273155 (13) [back to overview]Best Response
NCT01273155 (13) [back to overview]Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Volume of Distribution at Steady State (Vss)
NCT01273155 (13) [back to overview]Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Clearance (CL)
NCT01273155 (13) [back to overview]Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Area Under the Plasma Concentration Time Curve Extrapolated to Infinity(AUC0-inf) of Belinostat
NCT01273155 (13) [back to overview]Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0).
NCT01273155 (13) [back to overview]Maximum Tolerated Dose (MTD) of Belinostat According to Degree of Liver Dysfunction
NCT01273155 (13) [back to overview]Dose Limiting Toxicity (DLTs)
NCT01273155 (13) [back to overview]Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Metabolic Ratios of Area Under the Plasma Concentration Time Curve Extrapolated to Infinity (AUC0-inf) for the Belinostat Metabolic Pathway
NCT01273155 (13) [back to overview]Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Area Under the Plasma Concentration Time Curve Extrapolated to Infinity(AUC0-inf)
NCT01273155 (13) [back to overview]Pharmacokinetics (PK) of a Single-dose Belinostat (400 mg/m^2)) According to Degree of Liver Dysfunction: Maximum Plasma Concentrations (Cmax)
NCT01273155 (13) [back to overview]Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Metabolic Ratios of Maximum Plasma Concentrations (Cmax) for the Belinostat Metabolic Pathway
NCT01273155 (13) [back to overview]Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Half-Life (t1/2)
NCT01273155 (13) [back to overview]Number of Participants Experiencing Adverse Events Considered to be at Least Possibly Related to the Study Drug
NCT01686165 (4) [back to overview]Overall Response
NCT01686165 (4) [back to overview]Complete Response Rate
NCT01686165 (4) [back to overview]Occurrence of Adverse Events and Serious Adverse Events
NCT01686165 (4) [back to overview]Progression-free Survival

Duration of Response

Duration of response was defined as the time from first notation of response until the time of first notation of disease progression. (NCT00274651)
Timeframe: throughout the study, or for a maximum of 2 years

InterventionDays (Median)
Arm A (CTCL, ITT Population)83
Arm B (PTCL, ITT Population)109

[back to top]

Time to Response

Time to response was defined as the interval between the first date of treatment and the first notation of response. (NCT00274651)
Timeframe: throughout the study, or for a maximum of 2 years

InterventionDays (Median)
Arm A (CTCL, ITT Population)40
Arm B (PTCL, ITT Population)100

[back to top]

Time to Progression

Time to progression was defined as the interval between the first date of treatment and the first notation of disease progression. (NCT00274651)
Timeframe: throughout the study, or for a maximum of 2 years

InterventionDays (Median)
Arm A (CTCL, ITT Population)43
Arm B (PTCL, ITT Population)82

[back to top]

Objective Response Rate in Patients With Recurrent or Refractory Peripheral T-cell Lymphoma (PTCL))

Tumor response was assessed using the revised criteria of Cheson (Cheson 2007).Tumor assessments were done using conventional radiographic methods, e.g. CT or CT/PET. (NCT00274651)
Timeframe: throughout the study, or for a maximum of 2 years

Interventionpercentage of patients with OR (Number)
PTCL (ITT Population)25

[back to top]

Objective Response Rate in Patients With Recurrent or Refractory Cutaneous T-cell Lymphoma (CTCL)

Tumor response was assessed using Cheson (Cheson 2007) and SWAT criteria. The SWAT score represents the product of the percentage total body surface area (TBSA) involvement of each lesion type (patch, plaque, and tumor or ulceration), multiplied by a weighting factor. (NCT00274651)
Timeframe: throughout the study, or for a maximum of 2 years

Interventionparticipants (Number)
Arm A (CTCL, ITT Population)4

[back to top]

Time to Disease Progression (Low Malignant Potential or Micropapillary / Borderline Ovarian Tumour Group)

Assessed by RECIST criteria. Summarized using summary statistics, such as the mean, median, counts and proportion. (NCT00301756)
Timeframe: Up to 5 years

Interventionmonths (Median)
Treatment (Enzyme Inhibitor Therapy)13.4

[back to top]

Time to Disease Progression (Epithelial Ovarian Cancer Group)

Assessed by RECIST criteria. Summarized using summary statistics, such as the mean, median, counts and proportion. (NCT00301756)
Timeframe: Up to 5 years

Interventionmonths (Median)
Treatment (Enzyme Inhibitor Therapy)2.3

[back to top]

Stable Disease Rate, Defined as Neither Sufficient Shrinkage to Qualify for PR Nor Sufficient Increase to Qualify for PD, Taking as Reference the Smallest Sum LD Since the Treatment Started (Low Malignant Potential Group)

Stable disease rate, defined as neither sufficient shrinkage to qualify for Partial Response nor sufficient increase to qualify for Progressive disease, taking as reference the smallest sum LD since the treatment started. Summarized using summary statistics, such as the mean, median, counts and proportion. Assessed by RECIST criteria. (NCT00301756)
Timeframe: Up to 5 years

Interventionparticipants (Number)
Treatment (Enzyme Inhibitor Therapy)10

[back to top]

Stable Disease Rate, Defined as Neither Sufficient Shrinkage to Qualify for PR Nor Sufficient Increase to Qualify for PD, Taking as Reference the Smallest Sum LD Since the Treatment Started (Epithelial Ovarian Cancer Group)

Stable disease rate, defined as neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum LD since the treatment started. Summarized using summary statistics, such as the mean, median, counts and proportion. Assessed by RECIST criteria. (NCT00301756)
Timeframe: Up to 5 years

Interventionparticipants (Number)
Treatment (Enzyme Inhibitor Therapy)9

[back to top]

Progression-free Survival

Computed using the Kaplan-Meier method. Ninety-five percent confidence intervals will be constructed and selected results will be illustrated using figures and plots. (NCT00301756)
Timeframe: Duration of time from start of treatment to time of progression, assessed up to 5 years

Interventionmonths (Median)
Treatment (Enzyme Inhibitor Therapy)13.4

[back to top]

Efficacy of Belinostat in Terms of Complete or Partial Response; Disappearance of All Target Lesions or at Least a 30% Decrease in the Sum of the Longest Diameter of Target Lesions, Taking as Reference the Baseline Sum LD

Efficacy of belinostat in terms of complete or partial response; disappearance of all target lesions or at least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD. Assessed by Response Evaluation Criteria in Solid Tumors (RECIST) or Rustin criteria. (NCT00301756)
Timeframe: Up to 5 years

Interventionparticipants (Number)
Treatment (Enzyme Inhibitor Therapy)0

[back to top]

Number of Grade 3 Adverse Events Using the National Cancer Common Terminology Criteria for Adverse Events (CTCAE) Version 3.0

Events of Thrombosis, Hypersensitivity and ALP will be tabulated. (NCT00301756)
Timeframe: Up to 5 years

Interventionevents (Number)
Treatment (Belinostat / Enzyme Inhibitor Therapy)5

[back to top]

Overall Survival

Measured from time of registration to death, or last contact date (NCT00303953)
Timeframe: assessed every 3 months for 3 years

Interventionyears (Median)
PXD1010.9

[back to top]

Progression-free Survival

Measured from date of registration to time of first documentation of progression or death, or last contact date. Progression is defined as a 50% increase in sum of products of greatest diameters (SPD) of target measurable lesions over the smallest sum observed (over baseline if no decrease during therapy) using the same techniques as baseline; appearance of a new lesion/site; unequivocal progression of non-measurable disease in the opinion of the treating physician; death due to disease without prior documentation of progression. (NCT00303953)
Timeframe: assessed at week 8, then every 3 months for 3 years

Interventionyears (Median)
PXD1010.2

[back to top]

Assess Number of Patients Who Achieve Confirmed and Unconfirmed Complete Response (CR) or Partial Response (PR)

Complete Response(CR) is a complete disappearance of all disease with the exception of nodes. 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. CR Unconfirmed (CRU) does not qualify for CR above, due to a residual nodal mass or an indeterminate BM. Partial Response(PR) is a 50% decrease in the 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. (NCT00303953)
Timeframe: assessed at week 8, and every 3 months for 3 years

Interventionparticipants (Number)
Complete Response (CR)Complete Response Unconfirmed (CRU)Partial Response (PR)No response
PXD10100020

[back to top]

Dose-limiting Toxicities (DLT) and Maximum Tolerated Dose (MTD) of Belinostat in Patients With Inoperable HCC (Phase I)

DLT is defined as any grade 4 hematological toxicity and any grade 3 or 4 non hematological toxicity during cycle 1, excluding alopecia. Specifically, grade 3 nausea, vomiting, or diarrhea that does not respond to therapy is considered dose-limiting. Also, delays in treatment greater than 2 weeks are also dose-limiting. MTD is defined as the dose below which >= 2 of 3 or >= 2 of 6 patients experience DLT. Graded using the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00321594)
Timeframe: Course 1

InterventionParticipants (Count of Participants)
Phase I, Level 10
Phase I, Level 20
Phase I, Level 30
Phase I, Level 41

[back to top]

Toxicity Summary

Assessed using Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. Grade 3 and above toxicities possibly, probably or definitely related to treatment. (NCT00357032)
Timeframe: Up to 1 year

Interventionparticipants (Number)
Grade 3 : DehydrationGrade 3 : Febrile neutropeniaGrade 3 : HematomaGrade 3 : Mucositis/stomatitisGrade 3 : NauseaGrade 3 : Prolonged QTc intervalGrade 3 : Sodium, serum-lowGrade 4 : DehydrationGrade 4 : Febrile neutropeniaGrade 4 : HematomaGrade 4 : Mucositis/stomatitisGrade 4 : NauseaGrade 4 : Prolonged QTc intervalGrade 4 : Sodium, serum-low
Treatment (Belinostat)11011110010000

[back to top]

Overall Survival

Survival endpoints will be summarized by the method of Kaplan-Meier (NCT00357032)
Timeframe: Up to 1 year

InterventionMonths (Median)
Treatment (Belinostat)9.1

[back to top]

Complete Response Rate

Clinical responses were measured according to International Working Group criteria. Bone marrow studies were repeated at a minimum of every three cycles. Per International Working Group criteria: Complete Response (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 (NCT00357032)
Timeframe: Up to 1 year

Interventionpercentage of subjects (Number)
Treatment (Belinostat)0

[back to top]

Toxicity of Belinostat in Patients With Myelodysplastic Syndrome

Graded using the Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. Reporting events deemed at least possibly related to study treatment. (NCT00357162)
Timeframe: Prior to each course (every 21 days), and every 3 months for up to 3 years after completion of study treatment

Interventionparticipants (Number)
Grade 3 Hematologic Adverse EventGrade 4 Hematologic Adverse EventsGrade 3 Non-Hemeatologic Adverse EventsGrade 4 Non-hematologic Adverse Events
Treatment (Enzyme Inhibitor Therapy)131151

[back to top]

Number of Confirmed Responses (Complete Response, Partial Response, or Hematologic Improvement) Noted on 2 Consecutive Evaluations at Least 4 Weeks Apart

"Complete Response (CR)~A CR is defined as a participant with bone marrow showing less than 5% myeloblasts with no evidence of dysplasia and with adequate peripheral blood counts for at least 2 months (hemoglobin > 11 g/dl, neutrophils ≥ 1500/mm3, platelets ≥ 100,000/mm3) and with no blasts in the peripheral.~Partial Response (PR)~All the CR criteria except bone marrow blasts decreased by ≥ 50% over pretreatment, or a less advanced WHO classification than pretreatment.~Hematologic Improvement (HI)~A 2g/dl increase in hemoglobin for participants with <11g/dl hemoglobin at pretreatment, or an increase of >30,000/mm^3 platelets for participants with <100,000/mm^3 at pretreatment, or a 100% increase in neutrophil counts for participants with <1500/mm^3 at pretreatment" (NCT00357162)
Timeframe: 12 weeks

Interventionparticipants (Number)
Confirmed Hematologic Improvement (HI)Confirmed Partial Response (PR)COnfirmed Complete Response (CR)
Treatment (Enzyme Inhibitor Therapy)100

[back to top]

Time to Progression

Estimated using the method of Kaplan-Meier. (NCT00357162)
Timeframe: Time from registration to the date of progression or last follow-up, assessed up to 3 years

Interventionmonths (Median)
Treatment (Enzyme Inhibitor Therapy)14.9

[back to top]

Overall Survival

Estimated using the method of Kaplan-Meier. (NCT00357162)
Timeframe: From date of registration to the date of last follow-up or death due to any cause, assessed up to 3 years

Interventionmonths (Median)
Treatment (Enzyme Inhibitor Therapy)17.9

[back to top]

Objective Tumor Response Rate According to the Response Evaluation Criteria in Solid Tumors (RECIST) Committee

Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by CT, MRI or X-ray: 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 (NCT00365053)
Timeframe: Up to 3 years

Interventionpercentage of participants (Number)
Treatment (Belinostat)0

[back to top]

Toxicity Profile

Toxicity information recorded will include the type, severity, time of onset, time of resolution, and the probable association with the study regimen. Toxicities table summarizes the observed incidence by severity and type of toxicity for toxicities that are related to treatment and greater than grade 1. Adverse events assessed by the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. (NCT00365053)
Timeframe: Up to 3 years

InterventionParticipants (Count of Participants)
Grade 2 : HemoglobinGrade 2 : LymphopeniaGrade 2 : AlbuminGrade 2 : AnorexiaGrade 2 : ASTGrade 2 : ConstipationGrade 2 : CreatinineGrade 2 : SweatingGrade 2 : DyspneaGrade 2 : FatigueGrade 2 : Glucose (hyperglycemia)Grade 2 : HypoxiaGrade 2 : HypotensionGrade 2 : InfectionGrade 2 : NauseaGrade 2 : PainGrade 2 : Sodium (Hyponatremia)Grade 2 : Supraventricular arrhythmia NOSGrade 3 : HemoglobinGrade 3 : LymphopeniaGrade 3 : AlbuminGrade 3 : AnorexiaGrade 3 : ASTGrade 3 : ConstipationGrade 3 : CreatinineGrade 3 : SweatingGrade 3 : DyspneaGrade 3 : FatigueGrade 3 : Glucose (hyperglycemia)Grade 3 : HypoxiaGrade 3 : HypotensionGrade 3 : InfectionGrade 3 : NauseaGrade 3 : PainGrade 3 : Sodium (Hyponatremia)Grade 3 : Supraventricular arrhythmia NOSGrade 4 : HemoglobinGrade 4 : LymphopeniaGrade 4 : AlbuminGrade 4 : AnorexiaGrade 4 : ASTGrade 4 : ConstipationGrade 4 : CreatinineGrade 4 : SweatingGrade 4 : DyspneaGrade 4 : FatigueGrade 4 : Glucose (hyperglycemia)Grade 4 : HypoxiaGrade 4 : HypotensionGrade 4 : InfectionGrade 4 : NauseaGrade 4 : PainGrade 4 : Sodium (Hyponatremia)Grade 4 : Supraventricular arrhythmia NOS
Treatment (Belinostat)611202113060122102000010001200000031000000001001000000

[back to top]

Progression-free Survival

Estimated using the product-limit method of Kaplan and Meier. Progression is defined using Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0), as a 20% increase in the sum of the longest diameter of target lesions, or a measurable increase in a non-target lesion, or the appearance of new lesions. (NCT00365053)
Timeframe: Up to 3 years

InterventionMonths (Mean)
Treatment (Belinostat)1.2

[back to top]

Overall Survival

Estimated using the product-limit method of Kaplan and Meier. (NCT00365053)
Timeframe: Up to 3 years

InterventionMonths (Median)
Treatment (Belinostat)4.5

[back to top]

Belinostat AUC (0-infinity)

(NCT00421889)
Timeframe: Cycle 1 Day 1: Cycle 1 day 1: Pre-Infusion, 0 min, 5 min, 15 min, 30 min, 1 h, 2 h, 3 h, 3 h 15 min, 3 h 30 min, 4 h, 5 h, 6 h, 6 h 15 min, 6 h 30 min, 7h, 8h, 9h, 24h

Interventionng*h/mL (Mean)
Belinostat 600 mg/30 Minutes9116
Belinostat 800 mg/30 Min11785
Belinostat 1000 mg/30 Min21057
Belinostat 1000 mg/3-hours31515
Belinostat 1000 mg/6-hours13107

[back to top]

Belinostat Cmax

(NCT00421889)
Timeframe: Cycle 1 day 1: Pre-Infusion, 0 min, 5 min, 15 min, 30 min, 1 h, 2 h, 3 h, 3 h 15 min, 3 h 30 min, 4 h, 5 h, 6 h, 6 h 15 min, 6 h 30 min, 7h, 8h, 9h, 24h

Interventionng/mL (Mean)
Belinostat 600 mg/30 Minutes18592
Belinostat 800 mg/30 Min22525
Belinostat 1000 mg/30 Min31517
Belinostat 1000 mg/3-hours8340
Belinostat 1000 mg/6-hours2873

[back to top]

Belinostat Mean t½

(NCT00421889)
Timeframe: Cycle 1 Day 1: Cycle 1 day 1: Pre-Infusion, 0 min, 5 min, 15 min, 30 min, 1 h, 2 h, 3 h, 3 h 15 min, 3 h 30 min, 4 h, 5 h, 6 h, 6 h 15 min, 6 h 30 min, 7h, 8h, 9h, 24h

Interventionhours (Mean)
Belinostat 600 mg/30 Minutes1.41
Belinostat 800 mg/30 Min1.16
Belinostat 1000 mg/30 Min0.898
Belinostat 1000 mg/3-hours3.76
Belinostat 1000 mg/6-hours1.9

[back to top]

Best Overall Response (CR or PR)

Best overall responses were assessed by RECIST (Response Evaluation Criteria in Solid Tumors) criteria. Clinical tumor evaluation will take place after each cycle. Formal radiological evaluation after every 2 cycles. If a response is noted, a follow-up radiographic assessment must be performed 4 weeks (+ 1 week) after the response is noted (NCT00421889)
Timeframe: Throughout study until PD (progressive disease) or lost to follow up

Interventionparticipants (Number)
Part A: Dose Escalation 600/5/NA1
Part A: Dose Escalation 600/NA/1750
Part A: Dose Escalation 600/5/1750
Part A: Dose Escalation 800/5/1750
Part A: Dose Escalation 1000/5/1751
Part B: Ovarian Cancer MTD15
Part C: 3 Hours Infusion, Solid Tumors Except Ovarian Cancer0
Part C: 6 Hours Infusion Solid Tumors, Except Ovarian Cancer0
Part D: Bladder Cancer MTD4

[back to top]

Duration of Response

Defined as interval from the time criteria for CR or PR are met, until the first date that recurrent or progressive disease is objectively documented. (NCT00421889)
Timeframe: Throughout study

Interventiondays (Median)
Part B: Ovarian Cancer MTDNA
Part D: Bladder Cancer MTDNA

[back to top]

Time to Progression

Time to progression, defined as the interval between the first dates of treatment until the first notation of disease progression. RECIST criteria (NCT00421889)
Timeframe: Throughout study

Interventiondays (Median)
Part B: Ovarian Cancer MTD195
Part C: 3-6 Hours Infusion, Solid Tumors Except Ovarian Cancer150
Part D: Bladder Cancer MTD136

[back to top]

Time to Response

Time to response (RECIST) was assessed as the interval between the first dates of treatment until the first notation of response. (NCT00421889)
Timeframe: Throughout study

Interventiondays (Median)
Part B: Ovarian Cancer MTDNA
Part D: Bladder Cancer MTDNA

[back to top]

Dose Limiting Toxicities (DLT), Part A

To determine the number of participants experiencing dose limiting toxicities of belinostat (PXD101) in doses up to 1000 mg/m²/day administered in combination with standard doses of carboplatin and paclitaxel or both. (NCT00421889)
Timeframe: Cycle 1

Interventionparticipants (Number)
Part A0

[back to top]

Maximum Tolerable Dose (MTD) Belinostat, Part A,

To determine the maximum tolerated dose of belinostat (PXD101) in doses up to 1000 mg/m²/day administered in combination with standard doses of carboplatin (AUC of 5) and paclitaxel (175 mg/m2). (NCT00421889)
Timeframe: Cycle 1

Interventionmg/m2 (Number)
Part A1000

[back to top]

Number of Participants With Adverse Events

Here are the number of participants with adverse events. For a detailed list of adverse events see the adverse event module. (NCT00589290)
Timeframe: 26 months

InterventionParticipants (Number)
Belinostat41

[back to top]

Number of Participants With a Partial Response

Response is defined by the Response Evaluation Criteria in Solid Tumor (RECIST). Partial response (PR) is at least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD. For additional details about the RECIST criteria see the protocol Link module. (NCT00589290)
Timeframe: 25.5 months

InterventionParticipants (Number)
Thymoma Patients2
Thymic Patients0

[back to top]

Duration of Response

The Duration of Response was assessed by IWG criteria per the IRC from the date the measurement criteria were first met for CR or PR (whichever status was recorded first) until the first subsequent date that relapse or progression was documented. It was estimated by the Kaplan-Meier method. Response is defined as complete response (CR) or partial response (PR). CR is defined as the disappearance of all evidence of disease. PR is defined as a regression of measurable disease and no new sites. (NCT00865969)
Timeframe: 24 months

Interventionmonths (Median)
Belinostat13.6

[back to top]

Time to Response

Time to response was defined as the time (in weeks) from first administration of treatment until first response. Response is defined as complete response (CR) or partial response (PR). CR is defined as the disappearance of all evidence of disease. PR is defined as a regression of measurable disease and no new sites. (NCT00865969)
Timeframe: 24 months

Interventionweeks (Median)
Belinostat5.6

[back to top]

Time to Progression

Time to progression was defined as the time (in months) from first administration of treatment to the date of disease progression based on tumor assessments made according to the IWG criteria as assessed by the IRC. The progression is defined as any new lesion or increase by ≥ 50% of previously involved sites from nadir. (NCT00865969)
Timeframe: 24 months

Interventionmonths (Median)
Belinostat2

[back to top]

Overall Survival

Overall Survival was the time from first administration of study treatment until the date of death. (NCT00865969)
Timeframe: 24 months

Interventionmonths (Median)
Belinostat7.9

[back to top]

Progression Free Survival

Progression-free survival (PFS) was the duration of time from first administration of study treatment to date of first documented progression or death from any cause. It was based on tumor assessments made according to the IWG criteria as assessed by the IRC. The progression is defined as any new lesion or increase by ≥ 50% of previously involved sites from nadir. (NCT00865969)
Timeframe: 24 months

Interventionmonths (Median)
Belinostat1.6

[back to top]

Objective Response Rate

Objective response rate was defined as the percentage of participants with a complete response (CR) or a partial response (PR) according to International Working Group (IWG) criteria. The response was assessed based on clinical and radiological criteria. CR is defined as the disappearance of all evidence of disease. PR is defined as a regression of measurable disease and no new sites. As pre-defined, the primary endpoint analysis for this study was based on the Independent Review Committee (IRC) assessment of response. (NCT00865969)
Timeframe: 24 months

Interventionpercentage of participants (Number)
Belinostat25.8

[back to top]

Number of Participants With At Least One Serious Treatment-Emergent Adverse Event (TEAE)

A TEAE was defined as an event with an onset date and time on or after the first dosing start date and time, or on or after the first dosing start date if the onset time was missing. A serious TEAE was any untoward medical occurrence that at any dose results in death, prolonged hospitalization, persistent or significant disability or congenital abnormalities. (NCT00865969)
Timeframe: 24 months

InterventionParticipants (Count of Participants)
Belinostat61

[back to top]

Best Overall Response

The best overall response in an individual patient according to the RECIST criteria (Eisenhauer 2009 ) is the best response recorded from the start of the treatment until disease progression/recurrence. Objective response is defined as best overall response of complete response (CR) or partial response (PR) (NCT00873119)
Timeframe: Tumor assessment every 6 weeks for the treatment period. Subsequent assessments every 6 weeks for the initial 6 months, then every 9 weeks for 6 months, then every 12 weeks for 12 months and then every 6 months until 5 years from the start of study

Interventionpercentage of participants (Number)
Arm A - BelCaP43.2
Arm B - CaP22.2

[back to top]

Overall Survival (OS)

Time from the date of randomization to the date of death (NCT00873119)
Timeframe: Tumor assessment every 6 weeks for the treatment period. Subsequent assessments every 6 weeks for the initial 6 months, then every 9 weeks for 6 months, then every 12 weeks for 12 months and then every 6 months until 5 years from the start of study

Interventionmonths (Median)
Arm A - BelCaP11.5
Arm B - CaP9.1

[back to top]

Progression Free Survival

Time from the date of randomization to the time of disease progression or death due to any cause, measured by RECIST criteria (Response Evaluation Criteria In Solid Tumors). (NCT00873119)
Timeframe: Tumor assessment every 6 weeks for the treatment period. Subsequent assessments every 6 weeks for the initial 6 months, then every 9 weeks for 6 months, then every 12 weeks for 12 months and then every 6 months until 5 years from the start of study

Interventionmonths (Median)
Arm A - BelCaP5.4
Arm B - CaP5.3

[back to top]

Time to Progression (TTP)

Time from the date of randomization to the time of disease progression (NCT00873119)
Timeframe: Tumor assessment every 6 weeks for the treatment period. Subsequent assessments every 6 weeks for the initial 6 months, then every 9 weeks for 6 months, then every 12 weeks for 12 months and then every 6 months until 5 years from the start of study

Interventionmonths (Median)
Arm A - BelCaP5.7
Arm B - CaP5.6

[back to top]

Time to Response

For patients with overall best response being CR or PR, time to response was measured as the time from randomization to the first time when the measurement criteria for CR or PR (whichever status is recorded first) were met (NCT00873119)
Timeframe: Tumor assessment every 6 weeks for the treatment period. Subsequent assessments every 6 weeks for the initial 6 months, then every 9 weeks for 6 months, then every 12 weeks for 12 months and then every 6 months until 5 years from the start of study

Interventionmonths (Median)
Arm A - BelCaP3.2
Arm B - CaPNA

[back to top]

Duration of Response

Duration of overall response was measured from the time that measurement criteria were first met for CR or PR (whichever status was recorded first) until the first date that PD ([Progressive Disease]) or death was documented (NCT00873119)
Timeframe: Tumor assessment every 6 weeks for the treatment period. Subsequent assessments every 6 weeks for the initial 6 months, then every 9 weeks for 6 months, then every 12 weeks for 12 months and then every 6 months until 5 years from the start of study

Interventionmonths (Median)
Arm A - BelCaP4.6
Arm B - CaP6.5

[back to top]

Duration of Response (CR and PR)

Duration of Response (CR and PR) in Weeks (NCT00878722)
Timeframe: Throughout study, after each cycle for the first two cycles, then after every second cycle

Interventionweeks (Mean)
Arm A, Step 226.3
Arm A, Step 320.6
Arm A, Step 428.1
Arm B, Steps 1-65.4
Arm B, Step 730.3
Arm B, Step 819.6
Arm B, Step 94.3

[back to top]

Elimination t½

(NCT00878722)
Timeframe: Cycle 1, Samples taken in Cycle 1 only, prior to initial dose on days 4 and 5 and at end of infusion, 5, 15, and 30 min, and 1, 2, 3, 4, and 6 hours post infusion

InterventionHours (Mean)
Arm A, Cycle 1 Day 412.7
Arm A, Cycle 1 Day 513.1
Arm B, Cycle 1 Day 1 and Day 24.21

[back to top]

Event-Free Survival

Event-free survival: time (weeks) from entry into study until treatment failure, disease relapse or death from any cause. (NCT00878722)
Timeframe: Throughout study, after each cycle for the first two cycles, then after every second cycle

InterventionWeeks (Median)
Arm A, Step 15.1
Arm A, Step 23.1
Arm A, Step 38.0
Arm A, Step 46.5
Arm B, Steps 1-62.0
Arm B, Step 75.4
Arm B, Step 85.4
Arm B, Step 96.4

[back to top]

Maximum Tolerated Dose, Dose Limiting Toxicity

DLT (dose limiting toxicities): patients with any of the toxicities: 1.Haematological toxicity is not included in the definition due to bone marrow involvement by the disease except for following grade 4 ANC (absolute neutrophil count) and PLT (platelet count) for 6 weeks with less than 5% blasts in bone marrow. 2.Drug related non hematological Grade 3 or 4 toxicity except alopecia, brief nausea and vomiting, diarrhea, rash, arthralgias and myalgias. Treatment interventions should palliate toxicity symptoms prior to concluding a DLT has occurred (e.g if nausea and vomiting to Grade 3 have been associated with the drug). If despite standard treatment Grade 3 nausea and or vomiting persisted then a DLT was considered to have occurred. Grade 4 diarrhea in spite of standard therapeutic measures was included in DLT definition. 3.Inability to tolerate full dosing cycle due to toxicity or any drug-related adverse event resulting in more than 14 day treatment delay in the next treatment cycle (NCT00878722)
Timeframe: First Cycle

Interventionparticipants (Number)
Arm A, Step 10
Arm A, Step 20
Arm A, Step 30
Arm A, Step 40
Arm B, Steps 1-60
Arm B, Step 70
Arm B, Step 80
Arm B, Step 90

[back to top]

Overall Response

Efficacy measured as Response rate (complete response ([CR] and Complete remission with incomplete recovery of platelets [CRi]) and partial response ([PR])) using the response criteria of the International Working Group (Cheson et al 2003). CR includes CRi, CRc (Cytogenetic complete remission), and CRm (Molecular complete remission). (NCT00878722)
Timeframe: Throughout study, after each cycle for the first two cycles, then after every second cycle

Interventionparticipants (Number)
Arm A, Step 10
Arm A, Step 21
Arm A, Step 31
Arm A, Step 41
Arm B, Steps 1-61
Arm B, Step 71
Arm B, Step 81
Arm B, Step 93

[back to top]

Overall Survival

Overall survival: time in weeks from entry into study until death from any cause. All patients without this endpoint at the time of discontinuation or the end of trial have been censored. (NCT00878722)
Timeframe: Throughout study, after each cycle for the first two cycles, then after every second cycle

InterventionWeeks (Median)
Arm A, Step 410
Arm B, Steps 1-64.9
Arm B, Step 73.7
Arm B, Step 93.9

[back to top]

Relapse-Free Survival

Relapse-free survival: time (weeks) from leukemia-free state to relapse or death from any cause. (NCT00878722)
Timeframe: Throughout study, after each cycle for the first two cycles, then after every second cycle

InterventionWeeks (Number)
Arm A, Step 226.3
Arm A, Step 320.6
Arm A, Step 428.1
Arm B, Step 730.3
Arm B, Step 96.1

[back to top]

Remission Duration

Remission duration: time (weeks) from date of remission status to disease relapse. (NCT00878722)
Timeframe: Throughout study, after each cycle for the first two cycles, then after every second cycle

InterventionWeeks (Mean)
Arm A, Step 226.3
Arm A, Step 320.6
Arm B, Steps 1-65.4
Arm B, Step 730.3
Arm B, Step 819.6
Arm B, Step 93.0

[back to top]

Time to Response (CR and PR)

Time to response: time in weeks from first treatment to obtainment of the particular response status (CR and PR) (NCT00878722)
Timeframe: Throughout study, after each cycle for the first two cycles, then after every second cycle

InterventionWeeks (Median)
Arm A, Step 22.6
Arm A, Step 37.9
Arm A, Step 46.0
Arm B, Steps 1-64.0
Arm B, Step 71.9
Arm B, Step 84.0
Arm B, Step 91.0

[back to top]

Belinostat Cmax

Cmax: Arm A: at Cycle 1 Day 4, Cycle 1 Day 5 Arm B: Cycle 1 Day 1 and Cycle 1 Day 2 (NCT00878722)
Timeframe: Samples taken in Cycle 1 only, prior to initial dose on days 4 and 5 and at end of infusion, 5, 15, and 30 min, and 1, 2, 3, 4, and 6 hours post infusion

Interventionng/mL (Mean)
Arm A, Cycle 1 Day 438744
Arm A, Cycle 1 Day 531952
Arm B, Cycle 1 Day 1 and Day 29657

[back to top]

Belinostat AUC (Area Under Curve)

(NCT00878722)
Timeframe: Cycle 1, prior to initial dose on days 4 and 5 and at end of infusion, 5, 15, and 30 min, and 1, 2, 3, 4, and 6 hours post infusion

Interventionng*hrs/mL (Mean)
Arm A, Cycle 1 Day 421850
Arm A, Cycle 1 Day 521322
Arm B, Cycle 1 Day 1 and Day 2172823

[back to top]

Dose Limiting Toxicity (DLT)

Dose Limiting Toxicity (DLT) of PXD101 and doxorubicin combination treatment (NCT00878800)
Timeframe: Throughout study

InterventionDose limiting toxicity (Number)
Dose Escalation0
MTD Expansion0

[back to top]

Duration of Response

(NCT00878800)
Timeframe: Throughout study, after every 2 cycles

InterventionMonths (Median)
Dose Escalation3.9
MTD Expansion7.9

[back to top]

Maximum Tolerated Dose (MTD) of Doxorubicin

Maximum Tolerated Dose (MTD) of doxorubicin (NCT00878800)
Timeframe: During Cohort 1 to 4, Cycle 1 only, up to 3 weeks

Interventionmg/m2 (Number)
Dose Escalation75

[back to top]

Maximum Tolerated Dose (MTD) PXD101

Maximum Tolerated Dose (MTD) of PXD101treatment (NCT00878800)
Timeframe: During Cohort 1 to 4, Cycle 1 only, up to 3 weeks

Interventionmg/m² (Number)
Dose Escalation1000

[back to top]

Objective Response (CR and PR)

Measured by response rate using the RECIST (Response Evaluation Criteria in Solid Tumors) response criteria (response rate: Complete Response (CR) and Partial Response (PR)) following up to 6 cycles of treatment. (NCT00878800)
Timeframe: Throughout study, after every 2 cycles

Interventionpercentage of participants (Number)
Dose Escalation8
MTD Expansion12.5

[back to top]

Time to Response

(NCT00878800)
Timeframe: Throughout study, after every 2 cycles

Interventionmonths (Median)
Dose EscalationNA
MTD ExpansionNA

[back to top]

Time to Progression

(NCT00878800)
Timeframe: Throughout study, after every 2 cycles

Interventionmonths (Median)
Dose Escalation3.7
MTD Expansion6.0

[back to top]

Belinostat AUC (Time 0 to Last Measurement)

Measure the AUC of belinostat alone (Day 4 values) and in the presence of doxorubicin (Day 5 values) at the Maximum Tolerated Dose level: belinostat 1000 mg/m2 and doxorubicin 75 mg/m2 (NCT00878800)
Timeframe: Cycle 1, Day 4 and Day 5, pre-infusion, at end of infusion and at 5 min, 15 min, 30 min, 1 h, 2 h, 2 h and 15 min, 2 h and 30 min, 3 h, 4 h, 6 h, 8 h and 24 h post infusion

Interventionng*h/mL (Geometric Mean)
Day 4 - Belinostat 1000 mg/m²23100
Day 5 - Belinostat 1000 mg/m² and Doxorubicin 75 mg/m²22100

[back to top]

Belinostat Cmax

Measure the Cmax of belinostat alone (Day 4 values) and in the presence of doxorubicin (Day 5 values) at the Maximum Tolerated Dose level: belinostat 1000 mg/m2 and doxorubicin 75 mg/m2 (NCT00878800)
Timeframe: Cycle 1, Day 4 and Day 5, pre-infusion, at end of infusion and at 5 min, 15 min, 30 min, 1 h, 2 h, 2 h and 15 min, 2 h and 30 min, 3 h, 4 h, 6 h, 8 h and 24 h post infusion

Interventionng/mL (Geometric Mean)
Day 4 - Belinostat 1000 mg/m²41100
Day 5 - Belinostat 1000 mg/m² and Doxorubicin 75 mg/m²37000

[back to top]

Belinostat t½

Measure the t½ of belinostat alone (Day 4 values) and in the presence of doxorubicin (Day 5 values) at the Maximum Tolerated Dose level: belinostat 1000 mg/m2 and doxorubicin 75 mg/m2 (NCT00878800)
Timeframe: Cycle 1, Day 4 and Day 5, pre-infusion, at end of infusion and at 5 min, 15 min, 30 min, 1 h, 2 h, 2 h and 15 min, 2 h and 30 min, 3 h, 4 h, 6 h, 8 h and 24 h post infusion

Interventionhours (Geometric Mean)
Day 4 - Belinostat 1000 mg/m²1.48
Day 5 - Belinostat 1000 mg/m² and Doxorubicin 75 mg/m²2.13

[back to top]

Disease Control Rate (CR or PR or SD)

The disease control rate, defined as best overall response of either objective response or stable disease (CR or PR or SD) following up to 6 cycles of treatment with confirmation according to the RECIST criteria (NCT00878800)
Timeframe: Throughout study, after every 2 cycles

Interventionpercentage of participants (Number)
Dose Escalation72.0
MTD Expansion68.8

[back to top]

Frequency and Severity of Observed Adverse Effects Graded According to NCI CTCAE Version 3.0

(NCT00993616)
Timeframe: Every cycle during treatment and 30 days after the end of treatment

,,,,,
InterventionParticipants (Count of Participants)
LeukopeniaThrombocytopeniaNeutropeniaAnemiaOther hematologicAllergy/immunologyAuditory/earCardiacCoagulationConstitutionalDermatologicEndocrineNauseaVomitingGastrointestinalGenitourinary/renalHemorrhageLymphaticsMetabolicMusculoskeletalNeurosensoryOther neurologicalOcular/visualPainPulmonaryVascular
Grade 012141362623262625102226614102625261725212226152125
Grade 155210100111031149811172531860
Grade 294690210162051700010120402
Grade 312420100010023200020000000
Grade 402200100000000000000000000
Grade 500000000000000001000000000

[back to top]

Objective Response by Response Evaluation Criteria in Solid Tumors (RECIST) Criteria (Version 1.1)

Per Response Evaluation Criteria in Solid Tumors (RECIST) Criteria (version 1.1): Complete Response (CR) is disappearance of all target and non-target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm; Partial Response (PR) is at least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum diameters; Increasing Disease is at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progressions); Stable Disease (SD): Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest (NCT00993616)
Timeframe: From study entry, up to 5 years

Interventionparticipants (Number)
Complete ResponseIncrease DiseasePartial ResponseStable DiseaseIndeterminate
Treatment181125

[back to top]

Progression Free Survival at 6 Months

Progression is defined according to RECIST v1.0 as at least a 20% increase in the sum of LD target lesions taking as reference the smallest sum LD recorded since study entry, the appearance of one or more new lesions, death due to disease without prior objective documentation of progression, global deterioration in health status attributable to the disease requiring a change in therapy without objective evidence of progression, or unequivocal progression of existing non-target lesions. (NCT00993616)
Timeframe: Every other cycle for 6 months

Interventionpercentage of participants (Number)
Treatment29.6

[back to top]

Time to Maximum Plasma Concentration (Tmax)

Time to reach peak concentration after drug administration. (NCT01100944)
Timeframe: on day 1: pre-belinostat and 0, 0.5, 1 and 2 hours after belinostat infusion, on day 3: 0, 0.8, 0.25, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post-last belinostat dose

InterventionHour (Mean)
Phase I Dose Level 1 + Phase 225.28
Phase I Dose Level 250.0

[back to top]

Time to Response

Time to response is the time between the first day of treatment until first date of response (complete response (CR) + partial response (PR)) (whichever is first recorded). (NCT01100944)
Timeframe: From the first day of treatment until the date of first documented response, assessed up to 43 months

Interventiondays (Median)
Phase I Dose Level 1 & Phase I Dose Level 244.5

[back to top] [back to top]

Relative Changes in the Number of Tregs With Treatment

Changes in marker levels from baseline. All pre values were set to 1 so that we could measure changes relative to the baseline. (NCT01100944)
Timeframe: Baseline Cycle 1 Day 1 (C1D1), Cycle 1 Day 2 (C1D2), Cycle 1 Day 3 (C1D3), and Cycle 2 Day 1 (C2D1)

Interventionrelative fold change (Median)
C1D2C1D3C2D1
All Participants0.580.471.12

[back to top]

Relative Changes in T Cell Immunoglobulin Domain and Mucin Domain-3 (TIM3)-Expressing Cluster of Differentiation 8 (CD8)+Tcells

Changes in marker levels from baseline. All pre values were set to 1 so that we could measure changes relative to the baseline. (NCT01100944)
Timeframe: Baseline Cycle 1 Day 1 (C1D1), Cycle 1 Day 2 (C1D2), Cycle 1 Day 3 (C1D3), and Cycle 2 Day 1 (C2D1)

InterventionRelative fold change (Median)
C1D2C1D3C2D1
All Participants0.810.661.01

[back to top]

Relative Change Observed in Total Protein Hyperacetylation of Cluster of Differentiation 3 (CD3)+T Cells With Belinostat

Changes in marker levels from baseline. All pre values were set to 1 so that we could measure changes relative to the baseline. (NCT01100944)
Timeframe: Baseline Cycle 1 Day 1 (C1D1), Cycle 1 Day 2 (C1D2), Cycle 1 Day 3 (C1D3), and Cycle 2 Day 1 (C2D1)

InterventionRelative fold change (Median)
C1D2C1D3C2D1
All Participants3.452.491.15

[back to top]

Number of Participants With Serious and Non-serious Adverse Events

Here is the number of participants with serious and non-serious adverse events. For a detailed list of events, see the adverse event module. (NCT01100944)
Timeframe: up to 122 months

InterventionParticipants (Count of Participants)
SeriousNon-serious
All Participants2026

[back to top]

Number of Participants With Grade 3 and 4 Dose Limiting Toxicity (DLT) at 2000mg/m(2) Belinostat

A DLT is defined as grade 4 neutropenia lasting more than 7 days despite prophylactic or therapeutic use of granulocyte colony-stimulating factor (G-CSF), febrile neutropenia defined as absolute neutrophil count (ANC) less than 1000/mm(3) and temperature more than 38.5 degrees Celsius or 100.4 degrees Fahrenheit or life threatening sepsis, grade 4 thrombocytopenia or grade 3 thrombocytopenia associated with bleeding during the first cycle of therapy. Any grade 3 or 4 non-hematologic toxicity was considered dose limiting with the following exceptions: grade 3 diarrhea lasting less than 48 hours, grade 3 nausea and/or vomiting lasting less than 48 hours, grade 3 electrolyte abnormalities lasting less than 48 hours, grade 3 creatinine elevation lasting less than 48 hours. (NCT01100944)
Timeframe: up to 122 months

InterventionParticipants (Count of Participants)
Grade 3 NauseaGrade 3 DiarrheaGrade 4 NeutropeniaGrade 4 Thrombocytopenia
Phase I Dose Level 22222

[back to top]

Clinical Response

Response is defined by the Response Evaluation Criteria in Solid Tumors (RECIST). Complete response (CR) is disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. Partial response (PR) is at least a 30% decrease in the sum of the diameters of target lesions, taking as reference the baseline sum diameters. Progressive disease (PD) is at least a 20% increase in the sum of the diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of at least 5 mm. (Note: the appearance of one or more new lesions is also considered progressions). Stable disease (SD) is neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study. (NCT01100944)
Timeframe: 43 months

,,
InterventionParticipants (Count of Participants)
Complete Response (CR)Partial Response (PR)Stable Disease (SD)Progressive Disease (PD)
Thymic and Thymoma Participants19141
Thymic Particpants03101
Thymoma Participants1640

[back to top]

Total Clearance (CL) of Belinostat

Clearance is the amount of time for the drug to be eliminated from the body. (NCT01100944)
Timeframe: on day 1: pre-belinostat and 0, 0.5, 1 and 2 hours after belinostat infusion, on day 3: 0, 0.8, 0.25, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post-last belinostat dose

InterventionL/hr (Mean)
Phase 1 Dose Level 1 + Phase 2133.5
Phase 1 Dose Level 2132.1

[back to top]

Time to Half Life (t1/2) of Belinostat

Half life is the duration of time for the drug to be reduced to half the original amount. (NCT01100944)
Timeframe: on day 1: pre-belinostat and 0, 0.5, 1 and 2 hours after belinostat infusion, on day 3: 0, 0.8, 0.25, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post-last belinostat dose.

InterventionHour (Mean)
Phase 1 Dose Level 1 + Phase 20.47
Phase 1 Dose Level 20.40

[back to top]

Progression Free Survival (PFS)

Duration of time from start of treatment to time of progression or death whichever occurs first. (NCT01100944)
Timeframe: Start of treatment to time of disease progression or death whichever occurs first, assessed up to 43 months

Interventionmonths (Median)
Thymic Participants7.2
Thymoma ParticipantsNA
Thymic and Thymoma Participants9

[back to top]

Disease Control Rate (DCR)

DCR is defined as stable disease (SD) + partial response (PR) + complete response (CR) per the Response Criteria in Solid Tumors (RECIST). (NCT01100944)
Timeframe: 43 months

Interventionpercentage of participants (Number)
Thymic Participants93
Thymoma Participants100
Thymic and Thymoma Participants96

[back to top]

Overall Survival (OS)

Overall survival is defined as the on-study date until the date of death or progression as appropriate. (NCT01100944)
Timeframe: Start of treatment to time of death, assessed up to 43 months

Interventionmonths (Median)
Thymic Participants21.4
Thymoma ParticipantsNA
Thymic and Thymoma Participants28.5

[back to top]

Objective Response Rate (Partial Response (PR) + Complete Response (CR) of Belinostat in Combination With Cisplatin, Doxorubicin and Cyclophosphamide in the First Line Treatment of Patients With Advanced Thymic Malignancies

Objective response rate is the number of participants with a best objective response of partial response (PR) + complete response (CR) per the Response Criteria in Solid Tumors (RECIST) divided by the number of participants who had treatment. (NCT01100944)
Timeframe: 43 months

Interventionpercentage of participants (Number)
Thymic Participants21
Thymoma Participants64
Thymic and Thymoma Participants40

[back to top]

Maximum Tolerated Dose (MTD) of Belinostat

The MTD is defined as the highest dose at which less than 2 out of 6 patients experienced a dose limiting toxicity (DLT). A DLT is defined as grade 4 neutropenia lasting more than 7 days despite prophylactic or therapeutic use of granulocyte colony-stimulating factor (G-CSF), febrile neutropenia defined as absolute neutrophil count (ANC) less than 1000/mm(3) and temperature more than 38.5 degrees Celsius or 100.4 degrees Fahrenheit or life threatening sepsis, grade 4 thrombocytopenia or grade 3 thrombocytopenia associated with bleeding during the first cycle of therapy. Any grade 3 or 4 non-hematologic toxicity was considered dose limiting with the following exceptions: grade 3 diarrhea lasting less than 48 hours, grade 3 nausea and/or vomiting lasting less than 48 hours, grade 3 electrolyte abnormalities lasting less than 48 hours, grade 3 creatinine elevation lasting less than 48 hours. (NCT01100944)
Timeframe: 2 years

Interventionmg/m(2) (Number)
Phase I Dose Level 1 & Phase I Dose Level 21000

[back to top]

Maximum Plasma Concentration (Cmax)/Dose

Plasma concentrations of Belinostat were measured using a newly designed and validated ultra high-performance liquid chromatography (HPLC) with tandem mass spectrometric (MS/MS) assay, with a lower limit of quantification of 5ng/mL. (NCT01100944)
Timeframe: on day 1: pre-belinostat and 0, 0.5, 1 and 2 hours after belinostat infusion, on day 3: 0, 0.8, 0.25, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post-last belinostat dose

Interventionng/ml/mg (Mean)
Phase I Dose Level 1 + Phase 20.36
Phase I Dose Level 20.31

[back to top]

Maximum Observed Plasma Concentration (Cmax) of Belinostat

Plasma concentrations of Belinostat were measured using a newly designed and validated ultra high-performance liquid chromatography (HPLC) with tandem mass spectrometric (MS/MS) assay, with a lower limit of quantification of 5ng/mL. (NCT01100944)
Timeframe: on day 1: pre-belinostat and 0, 0.5, 1 and 2 hours after belinostat infusion, on day 3: 0, 0.8, 0.25, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post-last belinostat dose

Interventionng/ml (Mean)
Phase I Dose Level 1 + Phase 2650.6
Phase I Dose Level 21202

[back to top]

Duration of Response

Duration of response is measured from the time measurement criteria (e.g. Response Evaluation Criteria in Solid Tumors (RECIST)) are met for complete response (CR) or partial response (PR) (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). (NCT01100944)
Timeframe: From the time of first response until date of progression, assessed up to 43 months

Interventionmonths (Median)
Thymic Participants7.4
Thymoma ParticipantsNA
Thymic and Thymoma Participants7.4

[back to top]

Area Under the Plasma Concentration vs. Time Curve Extrapolated to Infinity (AUC(INF)/Dose

AUC is a measure of the serum concentration of Belinostat over time. It is used to characterize drug absorption. (NCT01100944)
Timeframe: on day 1: pre-belinostat and 0, 0.5, 1 and 2 hours after belinostat infusion, on day 3: 0, 0.8, 0.25, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post-last belinostat dose

Interventionhr*ng/ml/mg (Mean)
Phase I Dose Level 1 + Phase 210.78
Phase I Dose Level 27.57

[back to top]

Area Under the Plasma Concentration vs. Time Curve Extrapolated to Infinity (AUC(INF))

AUC is a measure of the serum concentration of Belinostat over time. It is used to characterize drug absorption. (NCT01100944)
Timeframe: on day 1: pre-belinostat and 0, 0.5, 1 and 2 hours after belinostat infusion, on day 3: 0, 0.8, 0.25, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours post-last belinostat dose

Interventionhr*ng/ml (Mean)
Phase I Dose Level 1 + Phase 219756
Phase I Dose Level 229686

[back to top]

Best Response

Radiologic response assessments by computed tomography (CT) scans were performed at baseline and every two cycles of treatment based on the Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1. Per RECIST v1.1 Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions, taking as reference the baseline sum diameters; Stable Disease (SD), neither sufficient shrinkage to qualify for a PR nor sufficient increase to qualify for Progression of Disease (PD), taking as reference the smallest sum diameters while on study; PD, >=20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study) and an absolute increase of at least 5mm or the appearance of new lesions; Complete Response (CR), Disappearance of all target lesions. Any pathological lymph nodes (whether target or non-target) must have reduction in short axis to <10 mm. (NCT01273155)
Timeframe: at baseline and every two 21-day cycles of treatment, up to 12 cycles

,,,,,,
InterventionParticipants (Count of Participants)
Partial ResponseStable DiseaseProgressive Disease
Mild Dysfunction-Belinostat 1000 mg/m(2)017
Mild Dysfunction-Belinostat 750 mg/m(2)036
Moderate Dysfunction-Belinostat 500 mg/m(2)022
Moderate Dysfunction-Belinostat 750 mg/m(2)012
Normal Function-Belinostat 1000 mg/m(2)068
Severe Dysfunction-Belinostat 250 mg/m(2)006
Severe Dysfunction-Belinostat 350 mg/m(2)003

[back to top]

Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Volume of Distribution at Steady State (Vss)

Belinostat Apparent volume of distribution at steady state (Vss) on Cycle 1 Day-7 as a function of degree of liver dysfunction. (NCT01273155)
Timeframe: Cycle 1 Day-7 prior to infusion; 15 and 25 minutes after the start of infusion; and 5, 10, 15, 30, 60, and 90 minutes, and 2, 4, 6, 8, and 24 hours after the end of infusion.

InterventionL/m^2 (Mean)
Normal Function30.1
Mild Dysfunction25.7
Moderate Dysfunction38.1
Severe Dysfunction29.6

[back to top]

Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Clearance (CL)

Clearance (CL) of Belinostat on Cycle 1 Day-7 as a function of degree of liver dysfunction (NCT01273155)
Timeframe: Cycle 1 Day-7 prior to infusion; 15 and 25 minutes after the start of infusion; and 5, 10, 15, 30, 60, and 90 minutes, and 2, 4, 6, 8, and 24 hours after the end of infusion.

InterventionmL/min/m^2 (Mean)
Normal Function661
Mild Dysfunction542
Moderate Dysfunction505
Severe Dysfunction444

[back to top]

Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Area Under the Plasma Concentration Time Curve Extrapolated to Infinity(AUC0-inf) of Belinostat

Area Under the Plasma Concentration Time Curve Extrapolated to Infinity(AUC0-inf) for Belinostat glucuronide, a Belinostat Metabolite on Cycle 1 Day-7 as a function of degree of liver dysfunction. (NCT01273155)
Timeframe: Cycle 1 Day -7 prior to infusion; 15 and 25 minutes after the start of infusion; and 5, 10, 15, 30, 60, and 90 minutes, and 2, 4, 6, 8, and 24 hours after the end of infusion.

Interventionmg*min/mL (Mean)
Normal Function15.1
Mild Dysfunction18.4
Moderate Dysfunction18.4
Severe Dysfunction23.4

[back to top]

Number of Participants With Serious and Non-serious Adverse Events Assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0).

Here is the count of participants with serious and non-serious adverse events assessed by the Common Terminology Criteria in Adverse Events (CTCAE v4.0). A non-serious adverse event is any untoward medical occurrence. A serious adverse event is an adverse event or suspected adverse reaction that results in death, a life threatening adverse drug experience, hospitalization, disruption of the ability to conduct normal life functions, congenital anomaly/birth defect or important medical events that jeopardize the patient or subject and may require medical or surgical intervention to prevent one of the previous outcomes mentioned. (NCT01273155)
Timeframe: From Cycle 1 Day -7 up to 12 (21-day) cycles.

InterventionParticipants (Count of Participants)
Normal Function-Belinostat 1000 mg/m(2)14
Mild Dysfunction-Belinostat 750 mg/m(2)11
Mild Dysfunction-Belinostat 1000 mg/m(2)14
Moderate Dysfunction-Belinostat 500 mg/m(2)5
Moderate Dysfunction-Belinostat 750 mg/m(2)4
Severe Dysfunction-Belinostat 250 mg/m(2)9
Severe Dysfunction-Belinostat 350 mg/m(2)6

[back to top]

Maximum Tolerated Dose (MTD) of Belinostat According to Degree of Liver Dysfunction

In order to maintain consistent dosing across the hepatic dysfunction groups, the dose recommended for cohorts with greater liver dysfunction could be no greater than the dose for cohorts of lesser dysfunction. In other words, it was assumed that a particular group would not tolerate a dose not tolerated by a group with lesser dysfunction and conversely, will tolerate a dose tolerated by a group with greater dysfunction. If a higher dose was tolerated in a group of greater dysfunction, but not in the group of lesser dysfunction, the lower dose would be recommended for both groups. The highest dose to be explored was no greater than the recommended dose for patients with normal liver function. (NCT01273155)
Timeframe: First cycle of therapy, 28 days

Interventionmg/m(2) (Number)
Mild Dysfunction1000
Moderate DysfunctionNA
Severe DysfunctionNA

[back to top]

Dose Limiting Toxicity (DLTs)

A DLT was defined as an adverse event deemed possibly, probably, or definitely related to administration of study drugs and met the following criteria: grade≥3 non-hematological toxicity (except grade ≥3 diarrhea, nausea, vomiting responsive to supportive therapy);grade≥3 rise in creatinine (except grade 3 able to be corrected to grade 1 or baseline with intravenous fluids within 24hrs); grade≥3 electrolyte toxicities (except those able to be corrected to grade 1 or baseline within 48hrs); grade 4 thrombocytopenia; grade 4 neutropenia for >5 days or febrile neutropenia; any neurotoxicity grade≥2 not reversible to grade 1 or baseline within 2wks; or any delay in treatment by ≥2wks due to treatment-related toxicity. Worsening liver function, as defined by a rise in serum bilirubin not related to tumor progression, was considered a DLT if a patient with mild dysfunction became severe for 1wk, or if a patient in either the moderate/severe groups had a >1.5x increase in bilirubin for 1 wk. (NCT01273155)
Timeframe: First cycle of therapy, 28 days.

InterventionToxicities (Number)
Normal Function-Belinostat 1000 mg/m(2)0
Mild Dysfunction-Belinostat 750 mg/m(2)0
Mild Dysfunction-Belinostat 1000 mg/m(2)0
Moderate Dysfunction-Belinostat 500 mg/m(2)0
Moderate Dysfunction-Belinostat 750 mg/m(2)0
Severe Dysfunction-Belinostat 250 mg/m(2)0
Severe Dysfunction-Belinostat 350 mg/m(2)0

[back to top]

Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Metabolic Ratios of Area Under the Plasma Concentration Time Curve Extrapolated to Infinity (AUC0-inf) for the Belinostat Metabolic Pathway

Metabolic ratios of Area Under the Plasma Concentration Time Curve Extrapolated to Infinity (AUC0-inf), reported as geometric mean (geometric standard deviation), for the belinostat metabolic pathway on Cycle 1 Day-7 as a function of degree of liver dysfunction. (NCT01273155)
Timeframe: Cycle 1 Day-7 prior to infusion; 15 and 25 minutes after the start of infusion; and 5, 10, 15, 30, 60, and 90 minutes, and 2, 4, 6, 8, and 24 hours after the end of infusion.

,,,
Interventionratio (Geometric Mean)
Belinostat glucuronide/belinostatMethyl belinostat/belinostatM21/belinostatM24/belinostatM26/belinostatM24/M26M26/M21
Mild Dysfunction21.80.5740.7831.010.4232.350.531
Moderate Dysfunction18.10.8921.240.9160.6661.380.539
Normal Function18.70.4220.9011.240.4522.741.15
Severe Dysfunction17.70.7591.130.8250.6111.350.541

[back to top]

Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Area Under the Plasma Concentration Time Curve Extrapolated to Infinity(AUC0-inf)

Area Under the Plasma Concentration Time Curve Extrapolated to Infinity (AUC0-inf) for belinostat and four metabolites on Cycle 1 Day -7 as a function of degree of liver dysfunction. (NCT01273155)
Timeframe: Cycle 1 Day-7 prior to infusion; 15 and 25 minutes after start of infusion; and 5, 10, 15, 60, 60, and 90 minutes, and 2, 4, 6, 8, and 24 hours after the end of infusion.

,,,
Interventionµg*min/mL (Mean)
BelinostatMethyl belinostatM21M24M26
Mild Dysfunction859483840916396
Moderate Dysfunction8348291058885600
Normal Function7193546471009361
Severe Dysfunction10129301295970740

[back to top]

Pharmacokinetics (PK) of a Single-dose Belinostat (400 mg/m^2)) According to Degree of Liver Dysfunction: Maximum Plasma Concentrations (Cmax)

Maximum plasma concentrations (Cmax) for belinostat and five metabolites on Cycle 1 Day -7 as a function of degree of liver dysfunction. (NCT01273155)
Timeframe: Cycle 1 Day -7 prior to infusion; 15 and 25 minutes after the start of infusion; and 5, 10, 15, 30, 60, and 90 minutes, and 2, 4, 6, 8, and 24 hours after the end of infusion.

,,,
Interventionµg/mL (Mean)
BelinostatBelinostat glucuronideMethyl belinostatM21M24M26
Mild Dysfunction26.690.82.991.792.351.80
Moderate Dysfunction22.167.23.261.641.821.83
Normal Function21.980.82.101.262.561.68
Severe Dysfunction25.072.83.351.931.882.24

[back to top]

Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Metabolic Ratios of Maximum Plasma Concentrations (Cmax) for the Belinostat Metabolic Pathway

Metabolic ratios of Maximum Plasma Concentrations (Cmax), reported as a geometric mean (geometric standard deviation), for the belinostat metabolic pathway on Cycle 1 Day-7 as a function of degree of liver dysfunction. (NCT01273155)
Timeframe: Cycle 1 Day-7 prior to infusion; 15 and 25 minutes after the start of infusion; and 5, 10, 15, 30, 60, and 90 minutes, and 2, 4, 6, 8, and 24 hours after the end of infusion.

,,,
Interventionratio (Geometric Mean)
Belinostat glucuronide/belinostatMethyl belinostat/belinostatM21/belinostatM24/belinostatM26/belinostatM24/M26M26/M21
Mild Dysfunction3.570.1120.06420.08700.06661.320.96
Moderate Dysfunction2.880.1490.07680.07380.08240.8951.07
Normal Function3.780.09480.05220.1170.07521.561.44
Severe Dysfunction2.850.1340.07590.07100.08060.6111.06

[back to top]

Pharmacokinetics (PK) of Single-dose Belinostat (400 mg/m^2) According to Degree of Liver Dysfunction: Half-Life (t1/2)

Half-life Period (t1/2) of belinostat and five metabolites on Cycle 1 Day -7 as a function of degree of liver function. (NCT01273155)
Timeframe: Cycle 1 Day -7 prior to infusion; 15 and 25 minutes after the start of infusion; and 5, 10, 15, 30, 60, and 90 minutes, and 2, 4, 6, 8, and 24 hours after the end of infusion.

,,,
Interventionmin (Mean)
BelinostatBelinostat glucuronideMethyl belinostatM21M24M26
Mild Dysfunction12724680.3460253151
Moderate Dysfunction144238136.1486223177
Normal Function11828079.6305264131
Severe Dysfunction157267133485260153

[back to top] [back to top]

Overall Response

To document the overall response for patients with relapsed aggressive high-risk non-Hodgkin's lymphoma (NHL) treated with two cycles PXD-101 followed by one cycle of the Zevalin regimen. (NCT01686165)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
PXD-101 (d1-d5) for 2 Cycles > Zevalin 1 Cycle0

[back to top]

Complete Response Rate

To document the complete response rate for patients with relapsed aggressive high-risk non-Hodgkin's lymphoma (NHL) treated with two cycles PXD-101 followed by one cycle of the Zevalin regimen. (NCT01686165)
Timeframe: Up to 5 years

InterventionParticipants (Count of Participants)
PXD-101 (d1-d5) for 2 Cycles > Zevalin 1 Cycle0

[back to top]

Occurrence of Adverse Events and Serious Adverse Events

The proportion of patients with a given adverse event will be tabulated and the 95% confidence interval computed. (NCT01686165)
Timeframe: Up to 30 days after patient receives last dose of study drug

InterventionParticipants (Count of Participants)
PXD-101 (d1-d5) for 2 Cycles > Zevalin 1 Cycle5

[back to top]

Progression-free Survival

Will be estimated using a Kaplan-Meier estimate. The observed 2-year progression-free survival rate will be estimated (with a 95% confidence interval) from the Kaplan-Meier curve. (NCT01686165)
Timeframe: 2 years

InterventionParticipants (Count of Participants)
PXD-101 (d1-d5) for 2 Cycles > Zevalin 1 Cycle0

[back to top]