Page last updated: 2024-12-11

panobinostat

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

Description

Panobinostat: An indole and hydroxamic acid derivative that acts as a HISTONE DEACETYLASE inhibitor. It is used as an antineoplastic agent in combination with BORTEZOMIB and DEXAMETHASONE for the treatment of MULTIPLE MYELOMA. [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

panobinostat : A hydroxamic acid obtained by formal condensation of the carboxy group of (2E)-3-[4-({[2-(2-methylindol-3-yl)ethyl]amino}methyl)phenyl]prop-2-enoic acid with the amino group of hydroxylamine. A histone deacetylase inhibitor used (as its lactate salt) in combination with bortezomib and dexamethasone for the treatment of multiple myeloma. [Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Cross-References

ID SourceID
PubMed CID6918837
CHEMBL ID483254
CHEBI ID85990
CHEBI ID93774
SCHEMBL ID183197
SCHEMBL ID164801
SCHEMBL ID22773814
MeSH IDM0540392

Synonyms (88)

Synonym
HY-10224
panobinostat (lbh589)
faridak
lbh-589
nvp-lbh-589
panobinostat
lbh-589b
farydak
nvp-lbh589
2-propenamide, n-hydroxy-3-(4-(((2-(2-methyl-1h-indol-3-yl)ethyl)amino) methyl)phenyl)-, (2e)-
lbh 589
lbh589 ,
bdbm29589
AKOS005146046
EC-000.2287
CHEMBL483254
chebi:85990 ,
BRD-K02130563-001-07-2
panobinostat;(e)-n-hydroxy-3-(4-((2-(2-methyl-1h-indol-3-yl)ethylamino)methyl)phenyl)acrylamide
A25218
(e)-n-hydroxy-3-[4-[[2-(2-methyl-1h-indol-3-yl)ethylamino]methyl]phenyl]prop-2-enamide
404950-80-7
EX-8456
panobinostat [usan:inn]
2-propenamide, n-hydroxy-3-(4-(((2-(2-methyl-1h-indol-3-yl)ethyl)amino)methyl)phenyl)-, (2e)-
9647fm7y3z ,
unii-9647fm7y3z
D10319
panobinostat (usan/inn)
(e)-n-hydroxy-3-(4-(((2-(2-methyl-1h-indol-3-yl)ethyl)amino)methyl)phenyl)acrylamide
LBH589 - PANOBINOSTAT
BCP9000844
BCPP000187
lbh58,9nvp-lbh589,panobinostat
CS-0267
n-hydroxy-3-[4-[2-(2-methyl-1h-indol-3-yl)ethylaminomethyl]phenyl]-2(e)-propenamide
S1030
panobinostat [who-dd]
panobinostat [mart.]
panobinostat [inn]
panobinostat [mi]
panobinostat [usan]
(2e)-n-hydroxy-3-[4-({[2-(2-methyl-1h-indol-3-yl)ethyl]amino}methyl)phenyl]prop-2-enamide
(e)-n-hydroxy-3-(4-{[2-(2-methyl-1h-indol-3-yl)-ethylamino]-methyl}-phenyl)-acrylamide
CCG-208762
MLS006011216
smr004702978
(e)-n-hydroxy-3-(4-((2-(2-methyl-1h-indol-3-yl)ethylamino)methyl)phenyl)acrylamide
AM808102
2-propenamide, n-hydroxy-3-[4-[[[2-(2-methyl-1h-indol-3-yl)ethyl]amino]methyl]phenyl]-, (2e)-
FPOHNWQLNRZRFC-ZHACJKMWSA-N
n-hydroxy-3 -[4-[[[2-(2-methyl-1h-indol-3-yl)ethyl]amino]methyl]phenyl]-2e-2-propenamide
n-hydroxy-3-[4-[[[2-(2-methyl-1h-indol-3-yl)ethyl]amino]methyl]phenyl]-2e-2-propenamide
SCHEMBL183197
SCHEMBL164801
gtpl7489
(e)-3-[4-[[2-(2-methyl-1h-indol-3-yl)ethylamino]methyl]phenyl]prop-2-enehydroxamic acid
nvp-lbh 589
AC-28652
mfcd09833242
(2e)-n-hydroxy-3-[4-({[2-(2-methyl-1h-indol-3-yl)ethyl]amino}methyl)phenyl]acrylamide
DB06603
J-523585
DTXSID40193506 ,
EX-A169
CHEBI:93774
bdbm198124
(2e)-n-hydroxy-3-[4-[[[2-(2-methyl-1h-indol-3-yl)ethyl]amino]methyl]phenyl]-2-propenamide
NCGC00263117-07
SW219369-1
panobinostat(lbh589)
n-hydroxy-3-[4-[[2-(2-methyl-1h-indol-3-yl)ethylamino]methyl]phenyl]-2-propenamide
404950-80-7 (free base)
AS-17046
BCP01816
Q7131441
NCGC00263117-05
nsc761190
nsc-761190
(lbh-589)
SCHEMBL22773814
dtxcid10115997
panobinostatum
l01xx42
(2e)-n-hydroxy-3-(4-(((2-(2-methyl-1h-indol-3-yl)ethyl)amino)methyl)phenyl)prop-2-enamide
(2e)-n-hydroxy-3-(4-(((2-(2-methyl-1h-indol-3-yl)ethyl)amino)methyl)phenyl)acrylamide
panobinostat (mart.)
EN300-7395075

Research Excerpts

Overview

Panobinostat is an oral pan histone-deacetylase inhibitor used in the treatment of relapsed and refractory multiple myeloma. It modulates the expression of oncogenic and immune-mediating genes involved in tumour cell growth and survival.

ExcerptReferenceRelevance
"Panobinostat (LBH589) is a novel pan-deacetylase inhibitor that is currently being evaluated in phase III clinical trials for treatment of Hodgkin's lymphoma and multiple myeloma. "( Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
Aichholz, R; Atadja, P; Délémonté, T; Fredenhagen, A; Kittelmann, M; Kuhn, A; Kühnöl, J; Oberer, L; Shultz, MD; Wang, P, 2012
)
2.04
"Panobinostat is a histone deacetylase inhibitor that may have therapeutic benefit."( Panobinostat in adults with H3 K27M-mutant diffuse midline glioma: a single-center experience.
Balakrishnan, SN; Carabenciov, ID; Daniels, DJ; Kizilbash, SH; Neth, BJ; Ruff, MW; Uhm, JH, 2022
)
2.89
"Panobinostat is an oral pan histone-deacetylase inhibitor used in the treatment of relapsed and refractory multiple myeloma. "( Outcomes with panobinostat in heavily pretreated multiple myeloma patients.
Casasanta, N; Lee, A; Moshier, E; Mouhieddine, TH; Pan, D; Richter, J; Upadhyay, R; Zubizarreta, N,
)
1.93
"Panobinostat is a nonselective pan-histone deacetylase inhibitor that is being tested in preclinical and clinical studies, including for the treatment of pediatric medulloblastoma, which has a propensity for leptomeningeal spread and diffuse midline glioma, which can infiltrate into supratentorial brain regions."( Central Nervous System Distribution of Panobinostat in Preclinical Models to Guide Dosing for Pediatric Brain Tumors.
Elmquist, WF; Larson, JD; Oh, JH; Rathi, S; Sirianni, RW; Wechsler-Reya, RJ; Zhang, W, 2023
)
1.9
"Panobinostat is a pan-deacetylase inhibitor that modulates the expression of oncogenic and immune-mediating genes involved in tumour cell growth and survival. "( Phase II trial of single-agent panobinostat consolidation improves responses after sub-optimal transplant outcomes in multiple myeloma.
Bowen, K; Cooke, RE; Gartlan, KH; Hill, GR; Kalff, A; Khong, T; Mithraprabhu, S; Ramachandran, M; Reynolds, J; Savvidou, I; Spencer, A, 2021
)
2.35
"Panobinostat is an oral histone deacetylase inhibitor (HDACi) that affects multiple cellular pathways and has demonstrated the ability to resensitize refractory-multiple myeloma cells in preclinical studies, as well as in patients with RRMM in clinical trials."( Panobinostat From Bench to Bedside: Rethinking the Treatment Paradigm for Multiple Myeloma.
Berdeja, JG; Chari, A; Laubach, JP; Richardson, PG; Richter, J; Spencer, A; Stricker, S, 2021
)
2.79
"Panobinostat (pano) is an FDA-approved histone deacetylase inhibitor. "( A Novel Indication for Panobinostat as a Senolytic Drug in NSCLC and HNSCC.
Adomako, A; McDaid, HM; Rodriguez-Gabin, A; Samaraweera, L, 2017
)
2.21
"Panobinostat is a clinically available histone deacetylase inhibitor for treating myelomas and also shows potentiality in NSCLC."( Panobinostat sensitizes KRAS-mutant non-small-cell lung cancer to gefitinib by targeting TAZ.
Chen, KC; Chen, PC; Cheng, CH; Huang, YH; Lan, CH; Lee, WY; Lin, CW; Wu, HC; Wu, WS, 2017
)
2.62
"Panobinostat (Farydak) is an orally active hydroxamic acid-derived histone deacetylase inhibitor used for the treatment of relapsed or refractory multiple myeloma. "( Physiologically Based Pharmacokinetic Model Predictions of Panobinostat (LBH589) as a Victim and Perpetrator of Drug-Drug Interactions.
Chun, DY; Einolf, HJ; Gu, H; He, H; Lin, W; Mangold, JB; Wang, L; Won, CS, 2017
)
2.14
"Panobinostat is a promising new class of anti-cancer drugs in AML."( Low expression of GFI-1 Gene is associated with Panobinostat-resistance in acute myeloid leukemia through influencing the level of HO-1.
Cheng, B; Fang, Q; Lu, T; Ma, D; Tang, S; Wang, J; Wei, D; Yu, K; Zhe, N; Zhou, Z, 2018
)
1.46
"Panobinostat is a potent pan-histone inhibitor of HDAC enzymes implicated in cancer development and progression."( Panobinostat in lymphoid and myeloid malignancies.
Dickinson, M; Khot, A; Prince, HM, 2013
)
2.55
"Panobinostat is an oral pan-deacetylase inhibitor that synergizes with bortezomib to inhibit both the aggresome and proteasome pathways in preclinical studies. "( PANORAMA 2: panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma.
Alsina, M; Coutre, SE; Gasparetto, C; Khan, M; Lonial, S; Mukhopadhyay, S; Ondovik, MS; Paley, CS; Richardson, PG; Schlossman, RL; Weber, DM, 2013
)
2.21
"Panobinostat is a potent oral pan-deacetylase inhibitor with promising clinical activity in hematologic malignancies. "( A clinical investigation of inhibitory effect of panobinostat on CYP2D6 substrate in patients with advanced cancer.
Beck, JT; Feld, R; Gazi, L; Hengelage, T; Leighl, N; Nayak, A; Porro, MG; Shepherd, FA; Woo, MM; Zhao, L, 2013
)
2.09
"Panobinostat is a potent deacetylase inhibitor that elicits synergistic effects on MM cells in combination with bortezomib."( Phase Ib study of panobinostat and bortezomib in relapsed or relapsed and refractory multiple myeloma.
Anderson, KC; Bladé, J; Bourquelot, PM; Günther, A; LeBlanc, R; Mishra, KK; Mu, S; Richardson, PG; San-Miguel, JF; Sezer, O; Siegel, D; Sopala, M; Sutherland, H; Victoria Mateos, M, 2013
)
1.45
"Panobinostat is a histone deacetylase inhibitor that has shown synergistic preclinical anti-myeloma activity when combined with other agents, recently exhibiting synergy with the alkylating agent melphalan (Sanchez et al., Leuk Res 35(3):373-379, 2011). "( A phase 1/2 study of oral panobinostat combined with melphalan for patients with relapsed or refractory multiple myeloma.
Berenson, JR; Boccia, RV; Dressler, K; Ghazal, HH; Harb, WA; Hilger, JD; Jamshed, S; Kingsley, EC; Matous, J; Nassir, Y; Noga, SJ; Swift, RA; Vescio, R; Yellin, O, 2014
)
2.15
"Panobinostat is a potent oral pan-deacetylase inhibitor that in preclinical studies has synergistic anti-myeloma activity when combined with bortezomib and dexamethasone. "( Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial.
Beksac, M; Bengoudifa, BR; Binlich, F; Bladé, J; Cavenagh, JD; Chen, W; Chuncharunee, S; Corradini, P; Corrado, C; de la Rubia, J; Dimopoulos, MA; Einsele, H; Elghandour, A; Gimsing, P; Günther, A; Hou, J; Hungria, VT; Jedrzejczak, WW; Kaufman, JL; LeBlanc, R; Lee, JH; Lee, JJ; Lonial, S; Moreau, P; Nahi, H; Nakorn, TN; Numbenjapon, T; Ocio, EM; Pulini, S; Qiu, L; Richardson, PG; Salwender, H; San-Miguel, JF; Schlossman, RL; Shelekhova, T; Siritanaratkul, N; Sohn, SK; Sopala, M; Tan, D; Veskovski, L; Wang, MC; Warzocha, K; White, D; Yong, K; Yoon, SS, 2014
)
3.29
"Panobinostat is a histone deacetylase inhibitor with antineoplastic and antiangiogenic effects in glioma that may work synergistically with bevacizumab. "( Phase II study of panobinostat in combination with bevacizumab for recurrent glioblastoma and anaplastic glioma.
Batchelor, TT; Beroukhim, R; Chi, AS; Drappatz, J; Gaffey, SC; Grimm, SA; Hempfling, K; Lee, EQ; Ligon, KL; McCluskey, C; Muzikansky, A; Nayak, L; Norden, AD; Raizer, JJ; Reardon, DA; Rinne, ML; Schiff, D; Smith, KH; Wen, PY; Wrigley, B, 2015
)
2.19
"Panobinostat is a potent, oral pan-deacetylase inhibitor that elicits anti-myeloma activity through epigenetic modulation of gene expression and disruption of protein metabolism."( Panobinostat: a novel pan-deacetylase inhibitor for the treatment of relapsed or relapsed and refractory multiple myeloma.
Alsina, M; Beksac, M; Dimopoulos, MA; Hungria, VT; Laubach, JP; Lonial, S; Moreau, P; Richardson, PG; San-Miguel, JF; Yoon, SS, 2015
)
2.58
"Panobinostat is a radiosensitizing agent and targets the epigenetics of malignancy. "( Phase I study evaluating the safety and efficacy of oral panobinostat in combination with radiotherapy or chemoradiotherapy in patients with inoperable stage III non-small-cell lung cancer.
Brown, MP; Gowda, R; Penniment, M; Singhal, N; Takhar, HS; Takhar, P, 2015
)
2.1
"Panobinostat is a potent oral deacetylase inhibitor that alters gene expression through epigenetic mechanisms and inhibits protein degradation. "( Panobinostat for the Treatment of Multiple Myeloma.
Laubach, JP; Moreau, P; Richardson, PG; San-Miguel, JF, 2015
)
3.3
"Panobinostat is a potent pan-deacetylase inhibitor that affects the growth and survival of multiple myeloma (MM) cells through alteration of epigenetic mechanisms and protein metabolism. "( Panobinostat plus bortezomib and dexamethasone in previously treated multiple myeloma: outcomes by prior treatment.
Beksac, M; Bengoudifa, BR; Binlich, F; Corrado, C; Dimopoulos, MA; Einsele, H; Elghandour, A; Guenther, A; Hou, J; Hungria, VT; Jedrzejczak, WW; Lee, JH; Lonial, S; Moreau, P; Nakorn, TN; Richardson, PG; San-Miguel, JF; Schlossman, RL; Siritanaratkul, N; Sopala, M; Yoon, SS, 2016
)
3.32
"Panobinostat is a potent oral pan-deacetylase inhibitor."( Panobinostat in combination with bortezomib in patients with relapsed or refractory peripheral T-cell lymphoma: an open-label, multicentre phase 2 trial.
Chan, YH; Fadilah, S; Goh, YT; Hwang, WY; Kim, WS; Kumar, SG; Lee, YS; Lim, ST; Ng, SC; Phipps, C; Tan, D; Tan, SY; Tay, K; Yeap, CH, 2015
)
2.58
"Panobinostat is an oral HDAC inhibitor with radiosensitizing activity. "( Phase I trial of panobinostat and fractionated stereotactic re-irradiation therapy for recurrent high grade gliomas.
Andrews, DW; Bar-Ad, V; Dicker, AP; Evans, JJ; Farrell, C; Glass, J; Judy, K; Kim, L; Lawrence, YR; Liu, H; Palmer, JD; Shi, W; Simone, N; Werner-Wasik, M, 2016
)
2.22
"Panobinostat is a promising alternative to well-studied, NCCN-recommended regimens for the treatment of RRMM. "( Panobinostat: A histone deacetylase inhibitor for the treatment of relapsed or refractory multiple myeloma.
Beggs, AE; Campbell, H; Ford, PD; Kodali, L; Wahaib, K, 2016
)
3.32
"1. Panobinostat is a recently approved histone deacetylase (HDAC) inhibitor. "( Clinical pharmacokinetics of panobinostat, a novel histone deacetylase (HDAC) inhibitor: review and perspectives.
Srinivas, NR, 2017
)
1.37
"Panobinostat is an oral pan-histone deacetylase inhibitor developed by Novartis. "( The Role of Panobinostat Plus Bortezomib and Dexamethasone in Treating Relapsed or Relapsed and Refractory Multiple Myeloma: A European Perspective.
Einsele, H; Moreau, P; San-Miguel, JF, 2016
)
2.26
"Panobinostat is a pan-deacetylase inhibitor that impedes protein destruction by disturbing the enzymatic activity of deacetylases."( Panobinostat for the management of multiple myeloma.
Gasparetto, C; Green, MM; Sivaraj, D, 2017
)
2.62
"Panobinostat is a pan-histone deacetylase inhibitor that has demonstrated clinical activity as a single agent in early phase trials of myelofibrosis."( A phase II study of panobinostat in patients with primary myelofibrosis (PMF) and post-polycythemia vera/essential thrombocythemia myelofibrosis (post-PV/ET MF).
Goldberg, JD; Hochman, T; Hoffman, R; Lu, M; Mascarenhas, J; Najfeld, V; Newsom, C; Petersen, B; Sandy, L; Ye, F; Yoon, J; Zhang, D, 2017
)
1.5
"Panobinostat is an orally available pan deacetylase inhibitor with evidence of activity in myeloid malignancies and cutaneous T cell lymphoma."( Preliminary evidence of disease response to the pan deacetylase inhibitor panobinostat (LBH589) in refractory Hodgkin Lymphoma.
Bhalla, KN; Bishton, M; DeAngelo, DJ; Dickinson, M; Fischer, T; Liu, A; Ottmann, OG; Parker, K; Prince, HM; Ritchie, D; Scott, JW; Spencer, A, 2009
)
1.31
"Panobinostat (LBH589) is a highly potent deacetylase inhibitor that has demonstrated clinical efficacy in patients with advanced cutaneous T-cell lymphoma (CTCL). "( Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T-cell lymphoma models: Defining molecular mechanisms of resistance.
Atadja, P; Fawell, S; Feng, Y; Growney, JD; Kwon, P; O'Connor, G; Pu, M; Shao, W; Yao, YM; Zhu, W, 2010
)
2.08
"Panobinostat is a well tolerated new treatment option for HCC that activates alternative pathways of apoptosis, also in p53-deficient tumors."( The pan-deacetylase inhibitor panobinostat inhibits growth of hepatocellular carcinoma models by alternative pathways of apoptosis.
Alinger, B; Di Fazio, P; Gahr, S; Hahn, EG; Hohenstein, B; Meissnitzer, M; Montalbano, R; Neureiter, D; Ocker, M; Okamoto, K; Quint, K; Sass, G; Schneider-Stock, R; Wissniowski, T, 2010
)
1.37
"Panobinostat (LBH589) is a potent histone deacetylase inhibitor (HDACi) that has shown anti-tumor activity in preclinical studies in both solid and hematological malignancies. "( The histone deacetylase inhibitor LBH589 enhances the anti-myeloma effects of chemotherapy in vitro and in vivo.
Berenson, JR; Bonavida, B; Chen, H; Li, M; Li, ZW; Sanchez, E; Shen, J; Steinberg, J; Wang, C, 2011
)
1.81
"Panobinostat (LBH589) is a potent deacetylase inhibitor acting both on histones and nonhistonic proteins, including α-tubulin."( Cytotoxic activity of the histone deacetylase inhibitor panobinostat (LBH589) in anaplastic thyroid cancer in vitro and in vivo.
Asioli, S; Bandino, A; Boccuzzi, G; Bosco, O; Bussolati, B; Catalano, MG; Compagnone, A; Fortunati, N; Gargantini, E; Grange, C; Mainini, F; Poli, R; Pugliese, M, 2012
)
1.35
"Panobinostat (LBH589) is a potent pan-histone deacetylase inhibitor. "( Phase I dose-escalating study of panobinostat (LBH589) administered intravenously to Japanese patients with advanced solid tumors.
Ando, Y; Fujiwara, Y; Inada, M; Kimura, J; Kitagawa, K; Kiyota, N; Komatsu, Y; Minami, H; Mitsuma, A; Morita, S; Oizumi, S; Sawaki, M; Tanii, H; Yuki, S, 2012
)
2.1
"Panobinostat is a histone deacetylase inhibitor with anti-neoplastic and anti-angiogenic effects and may work synergistically with VEGF inhibitors."( Phase I study of panobinostat in combination with bevacizumab for recurrent high-grade glioma.
Batchelor, TT; Beroukhim, R; Chi, AS; Ciampa, AS; Doherty, LM; Drappatz, J; Gerard, M; Grimm, SA; Hammond, S; Lafrankie, DC; Lee, EQ; Norden, AD; Raizer, JJ; Ruland, S; Schiff, D; Snodgrass, SM; Wen, PY, 2012
)
1.44
"Panobinostat is a novel oral pan-deacetylase inhibitor with promising anti-cancer activity. "( The effect of food on the bioavailability of panobinostat, an orally active pan-histone deacetylase inhibitor, in patients with advanced cancer.
Dandamudi, UB; Frank, R; Gazi, L; Hengelage, T; Lewis, LD; Porro, MG; Shapiro, GI; Woo, MM; Zhao, L, 2012
)
2.08
"Panobinostat is a potent oral pan-deacetylase inhibitor (pan-DACi)."( Phase II study of melphalan, thalidomide and prednisone combined with oral panobinostat in patients with relapsed/refractory multiple myeloma.
Alesiani, F; Ballanti, S; Boccadoro, M; Caraffa, P; Catarini, M; Cavallo, F; Corvatta, L; Gentili, S; Leoni, P; Liberati, AM; Offidani, M; Palumbo, A; Polloni, C; Pulini, S, 2012
)
1.33
"Panobinostat (LBH589) is a highly potent HDACi with demonstrated antitumor activities at low nanomolar concentration in several preclinical studies and its clinical efficacy is currently under investigation in several clinical trials."( Panobinostat for the treatment of multiple myeloma.
Bahlis, NJ; Lonial, S; Neri, P, 2012
)
2.54
"Panobinostat is a potent, oral pan-deacetylase inhibitor (pan-DACi) that increases the acetylation of proteins involved in multiple oncogenic pathways. "( Panobinostat activity in both bexarotene-exposed and -naïve patients with refractory cutaneous T-cell lymphoma: results of a phase II trial.
Assaf, C; Becker, JC; Dummer, R; Duvic, M; Grazia Bernengo, M; Lebbé, C; Marshood, M; Ortiz Romero, P; Poulalhon, N; Prince, HM; Squier, M; Tai, F; Williams, D, 2013
)
3.28
"Panobinostat is a potent oral pandeacetylase inhibitor that leads to acetylation of intracellular proteins, inhibits cellular proliferation and induces apoptosis in leukemic cell lines. "( Phase Ia/II, two-arm, open-label, dose-escalation study of oral panobinostat administered via two dosing schedules in patients with advanced hematologic malignancies.
Atadja, P; Bhalla, KN; DeAngelo, DJ; Fischer, T; Giles, FJ; Kindler, T; Liu, A; Mishra, KK; Ottmann, OG; Parker, K; Prince, HM; Scott, JW; Spencer, A; Woo, M, 2013
)
2.07

Effects

Panobinostat (LBH589) has been identified as a potential therapeutic agent for ALL with t(4;11) Studies suggest that the antineoplastic effects are associated with reduced MLL-AF4 fusion protein and reduced expression of HOX genes.

ExcerptReferenceRelevance
"Panobinostat has a clinically relevant activity profile and is a candidate for OA symptom and structure modification."( Targeting FoxO transcription factors with HDAC inhibitors for the treatment of osteoarthritis.
Akasaki, Y; Chen, E; Chu, AC; Gotoh, M; Hu, Y; Johnson, KA; Kanaya, H; Kawata, M; Kurakazu, I; Lotz, MK; Nagira, K; Ohzono, H; Okubo, N; Olmer, M, 2023
)
2.35
"Panobinostat (LBH589) has been identified as a potential therapeutic agent for ALL with t(4;11) and studies suggest that the antineoplastic effects are associated with reduced MLL-AF4 fusion protein and reduced expression of HOX genes."( Panobinostat (LBH589) increase survival in adult xenografic model of acute lymphoblastic leukemia with t(4;11) but promotes antagonistic effects in combination with MTX and 6MP.
Borges, KS; Cruzeiro, GAV; Junior, HLR; Laranjeira, ABA; Moreno, DA; Ramalho, FS; Rego, EM; Salomão, KB; Scrideli, CA; Silva, CLA; Tone, LG; Yunes, JA, 2022
)
2.89
"Panobinostat, a pan-DACi, has shown significant clinical benefit and is the first DACi approved for the treatment of MM."( Deacetylase inhibitors as a novel modality in the treatment of multiple myeloma.
Laubach, JP; Lonial, S; Maglio, ME; Moreau, P; Richardson, PG; San-Miguel, J, 2017
)
1.18
"Panobinostat has been shown to have therapeutic efficacy both in vitro and in DIPG orthotopic xenograft models; however, clinical data in patients with DIPG are lacking."( Concomitant Use of Panobinostat and Reirradiation in Progressive DIPG: Report of 2 Cases.
Altinok, D; Edwards, H; Ge, Y; Kieran, MW; Poulik, J; Sood, S; Steven, M; Taub, JW; Wang, ZJ, 2017
)
1.5

Actions

Panobinostat can cause apoptosis induction in refractory metastatic neuroblastoma in combination with MIBG therapy and chemotherapy. It was shown to inhibit CYP2D6 activity in vitro; thus understanding the magnitude of the potential clinical inhibition becomes important.

ExcerptReferenceRelevance
"Panobinostat can cause apoptosis induction in refractory metastatic neuroblastoma in combination with MIBG therapy and chemotherapy."( Successful treatment of refractory metastatic neuroblastoma with panobinostat in combination with chemotherapy agents and iodine-131-meta-iodobenzylguanidine therapy.
Bordbar, M; Shahriari, M; Shakibazad, N; Zareifar, S; Zekavat, OR, 2020
)
2.24
"Panobinostat was shown to inhibit CYP2D6 activity in vitro; thus understanding the magnitude of the potential clinical inhibition of panobinostat on co-medications that are CYP2D6 substrates becomes important."( A clinical investigation of inhibitory effect of panobinostat on CYP2D6 substrate in patients with advanced cancer.
Beck, JT; Feld, R; Gazi, L; Hengelage, T; Leighl, N; Nayak, A; Porro, MG; Shepherd, FA; Woo, MM; Zhao, L, 2013
)
1.37

Treatment

Panobinostat treatment reduced cell growth and diminished cell viability, as shown by the induced cell cycle arrest and apoptosis in vitro. Treatment also increased the proapoptotic BIK, BIM, BAX, and BAK levels, as well as increased the activity of caspase-7.

ExcerptReferenceRelevance
"(3) Panobinostat treatment induced upregulation of PD-L1 expression in both glioma and squamous cell carcinoma cells."( Histone Deacetylase Inhibitor Panobinostat Benefits the Therapeutic Efficacy of Oncolytic Herpes Simplex Virus Combined with PD-1/PD-L1 Blocking in Glioma and Squamous Cell Carcinoma Models.
Chen, X; Ji, D; Liu, Y; Ren, P; Wang, L; Wu, Y; Zhao, J; Zhou, GG; Zhou, X, 2022
)
1.49
"Panobinostat treatment led to significant reductions in multiple established plasma markers of inflammation. "( The histone deacetylase inhibitor panobinostat lowers biomarkers of cardiovascular risk and inflammation in HIV patients.
Brinkmann, CR; Dinarello, CA; Høgh Kølbæk Kjær, AS; Olesen, R; Rasmussen, TA; Søgaard, OS; Tolstrup, M; Østergaard, L, 2015
)
2.14
"Panobinostat treatment reduced cell growth and diminished cell viability, as shown by the induced cell cycle arrest and apoptosis in vitro. "( The HDACi Panobinostat Shows Growth Inhibition Both In Vitro and in a Bioluminescent Orthotopic Surgical Xenograft Model of Ovarian Cancer.
Bischof, K; Bjørge, L; Gjertsen, BT; Helland, Ø; McCormack, E; Popa, M, 2016
)
2.28
"Panobinostat treatment also increased the proapoptotic BIK, BIM, BAX, and BAK levels, as well as increased the activity of caspase-7."( Treatment with panobinostat induces glucose-regulated protein 78 acetylation and endoplasmic reticulum stress in breast cancer cells.
Atadja, P; Balusu, R; Bhalla, KN; Buckley, KM; Chen, J; Coothankandaswamy, V; Fiskus, W; Ha, K; Joshi, A; Kolhe, R; Nalluri, S; Rao, R; Yang, Y, 2010
)
1.43
"Panobinostat treatment induced downregulation of EGFR, HER2, and HER3 mRNA and protein through transcriptional and posttranslational mechanisms."( The dual EGFR/HER2 inhibitor lapatinib synergistically enhances the antitumor activity of the histone deacetylase inhibitor panobinostat in colorectal cancer models.
El-Khoueiry, A; Fazzone, W; LaBonte, MJ; Ladner, RD; Lenz, HJ; Louie, SG; Russell, J; Wilson, PM, 2011
)
1.3
"Panobinostat treatment increased histone acetylation, decreased cell proliferation and survival, and blocked cell cycle progression at G2/M with a concurrent decrease in S phase in all TNBC cell lines. "( Targeting triple-negative breast cancer cells with the histone deacetylase inhibitor panobinostat.
Burow, ME; Collins-Burow, BM; Driver, JL; Pounder, FN; Rhodes, LV; Segar, HC; Tate, CR, 2012
)
2.05
"(1) Treatment with panobinostat enhanced oHSV propagation and cytotoxicity in human glioma A172 and squamous cell carcinoma SCC9 cells. "( Histone Deacetylase Inhibitor Panobinostat Benefits the Therapeutic Efficacy of Oncolytic Herpes Simplex Virus Combined with PD-1/PD-L1 Blocking in Glioma and Squamous Cell Carcinoma Models.
Chen, X; Ji, D; Liu, Y; Ren, P; Wang, L; Wu, Y; Zhao, J; Zhou, GG; Zhou, X, 2022
)
1.34
"Cotreatment with panobinostat and BEZ235 warrants further evaluation in GBM therapy."( Enhanced efficacy of histone deacetylase inhibitor panobinostat combined with dual PI3K/mTOR inhibitor BEZ235 against glioblastoma.
Li, Q; Ma, J; Mao, W; Meng, W; Wang, B; Wang, J; Zhang, C; Zhao, Y, 2019
)
1.09
"Treatment with panobinostat (LBH589), a novel potent DACi, led to the highly aberrant modulation of several miRNAs in hepatocellular carcinoma (HCC) cell lines as shown by miRNA array analysis."( The pan-deacetylase inhibitor panobinostat suppresses the expression of oncogenic miRNAs in hepatocellular carcinoma cell lines.
Di Fazio, P; Henrici, A; Krause, M; Montalbano, R; Neureiter, D; Ocker, M; Quint, K; Slater, EP; Stiewe, T, 2015
)
1.05
"Treatment with panobinostat improved survival of athymic nude mice implanted with human CTCL cells."( Cotreatment with BCL-2 antagonist sensitizes cutaneous T-cell lymphoma to lethal action of HDAC7-Nur77-based mechanism.
Atadja, P; Balusu, R; Bhalla, KN; Chappa, P; Chen, J; Eaton, K; Fernandez, P; Fiskus, W; Jillella, A; Joshi, R; Kolhe, R; Lee, P; Natarajan, K; Rao, R; Wang, Y; Yang, Y, 2009
)
0.69
"Co-treatment with panobinostat and decitabine also caused more loss of viability of primary AML but not normal CD34(+) bone marrow progenitor cells."( Panobinostat treatment depletes EZH2 and DNMT1 levels and enhances decitabine mediated de-repression of JunB and loss of survival of human acute leukemia cells.
Atadja, P; Balusu, R; Bhalla, KN; Buckley, K; Chen, J; Fiskus, W; Joshi, A; Joshi, R; Koul, S; Mandawat, A; Rao, R; Upadhyay, S; Wang, Y; Yang, Y, 2009
)
2.12
"Treatment with panobinostat increased the levels of phosphorylated-eukaryotic translation initiation factor (p-eIF2alpha), ATF4, and CAAT/enhancer binding protein homologous protein (CHOP)."( Treatment with panobinostat induces glucose-regulated protein 78 acetylation and endoplasmic reticulum stress in breast cancer cells.
Atadja, P; Balusu, R; Bhalla, KN; Buckley, KM; Chen, J; Coothankandaswamy, V; Fiskus, W; Ha, K; Joshi, A; Kolhe, R; Nalluri, S; Rao, R; Yang, Y, 2010
)
1.05
"Treatment with panobinostat induced heat shock protein 90 acetylation; depleted the levels of heat shock protein 90 client proteins, cyclin-dependent kinase 4, c-RAF, and AKT; and abrogated bortezomib-induced aggresome formation in MCL cells. "( Role of CAAT/enhancer binding protein homologous protein in panobinostat-mediated potentiation of bortezomib-induced lethal endoplasmic reticulum stress in mantle cell lymphoma cells.
Atadja, P; Balusu, R; Bhalla, KN; Buckley, KM; Coothankandaswamy, V; Fiskus, W; Ha, K; Joshi, A; Nalluri, S; Rao, R; Savoie, A; Sotomayor, E; Tao, J, 2010
)
0.96
"Treatment with panobinostat combined with sorafenib demonstrated the highest preclinical efficacy in HCC models, providing the rationale for clinical studies with this novel combination."( Combination therapy for hepatocellular carcinoma: additive preclinical efficacy of the HDAC inhibitor panobinostat with sorafenib.
Alsinet, C; Cabellos, L; Friedman, SL; Hoshida, Y; Lachenmayer, A; Llovet, JM; Minguez, B; Thung, S; Toffanin, S; Tsai, HW; Villanueva, A; Ward, SC, 2012
)
0.93
"Treatment with panobinostat also induced the accumulation and colocalization of p62 with LC3B-II in cytosolic foci as evidenced by immunofluorescent confocal microscopy."( Combination of pan-histone deacetylase inhibitor and autophagy inhibitor exerts superior efficacy against triple-negative human breast cancer cells.
Atadja, P; Balusu, R; Bhalla, KN; Chauhan, L; Fiskus, W; Ha, K; Hembruff, SL; Mudunuru, U; Rao, R; Smith, JE; Venkannagari, S, 2012
)
0.72

Toxicity

The addition of panobinostat to bortezomib and dexamethasone has demonstrated response and progression-free survival benefits. The incidence and severity of adverse events associated with it can create a challenge for clinicians and patients.

ExcerptReferenceRelevance
" Coexposure of LBH589 and bortezomib at minimally toxic doses of either drug alone resulted in a striking induction of apoptosis in established U251, U87, and D37 GBM cell lines, as well as in GBM8, GBM10, GBM12, GBM14, and GBM56 short-term cultured cell lines."( Mitochondrial Bax translocation partially mediates synergistic cytotoxicity between histone deacetylase inhibitors and proteasome inhibitors in glioma cells.
Adjei, AA; Atadja, P; Friday, BB; Sarkaria, J; Wigle, D; Yang, L; Yu, C, 2008
)
0.35
" Two serious adverse events, rapid atrial fibrillation and tracheo-oesophageal fistula, were not attributable to study treatment."( Phase I study evaluating the safety and efficacy of oral panobinostat in combination with radiotherapy or chemoradiotherapy in patients with inoperable stage III non-small-cell lung cancer.
Brown, MP; Gowda, R; Penniment, M; Singhal, N; Takhar, HS; Takhar, P, 2015
)
0.66
" Random-effect pooled estimates were calculated for overall response rate and rates of common adverse effects."( Efficacy and Safety of Panobinostat in Relapsed or/and Refractory Multiple Myeloma: Meta Analyses of Clinical Trials and Systematic Review.
Hu, B; Hu, Y; Liu, JD; Sun, CY; Tang, L; Wang, QY; Wu, YY, 2016
)
0.74
" Despite this benefit, patients in the panobinostat arm experienced higher rates of adverse events (AEs) and higher rates of discontinuation due to AEs."( Panobinostat plus bortezomib and dexamethasone: impact of dose intensity and administration frequency on safety in the PANORAMA 1 trial.
Beksac, M; Corrado, C; Dimopoulos, MA; Einsele, H; Elghandour, A; Guenther, A; Hou, J; Hungria, VTM; Jedrzejczak, WW; Lee, JH; Lonial, S; Moreau, P; Na Nakorn, T; Paul, S; Redhu, S; Richardson, PG; Salwender, H; San-Miguel, JF; Schlossman, RL; Siritanaratkul, N; Sopala, M; Yoon, SS, 2017
)
2.17
" In this review multiple myeloma-related symptoms and adverse events resulting from treatments for multiple myeloma are discussed, with a focus on adverse events related to histone deacetylase inhibitors and histone deacetylase inhibitor combinations."( Optimal Management of Histone Deacetylase Inhibitor-Related Adverse Events in Patients With Multiple Myeloma: A Focus on Panobinostat.
Cavenagh, JD; Popat, R, 2018
)
0.69
" CONCLUSIONS CED of water-soluble panobinostat, up to a concentration of 30 μM, was not toxic and was distributed effectively in normal brain."( The distribution, clearance, and brainstem toxicity of panobinostat administered by convection-enhanced delivery.
Asby, DJ; Bienemann, AS; Boulter, LJ; Damment, SJP; Gill, SS; Johnson, D; Killick-Cole, CL; Lewis, O; Singleton, WGB; Woolley, M; Wyatt, MJ, 2018
)
1.01
" While the addition of panobinostat to bortezomib and dexamethasone has demonstrated response and progression-free survival benefits, the incidence and severity of adverse events associated with it can create a challenge for clinicians and patients."( Incidence and management of adverse events associated with panobinostat in the treatment of relapsed/refractory multiple myeloma.
Arnall, JR; Harvey, RD; Moore, DC, 2019
)
1.07
" All patients experienced adverse events (AEs), with diarrhea (80."( Phase II, Multicenter, Single-Arm, Open-Label Study to Evaluate the Efficacy and Safety of Panobinostat in Combination with Bortezomib and Dexamethasone in Japanese Patients with Relapsed or Relapsed-and-Refractory Multiple Myeloma.
Maki, A; Matsumoto, M; Shimada, F; Shimizu, K; Sunami, K; Suzuki, K, 2021
)
0.84
" Grade 3-4 adverse events occurred in 71 (91%) of 78 patients in the 20 mg three times weekly group, 69 (83%) of 83 patients in the 20 mg twice weekly group, and 60 (75%) of 80 patients in the 10 mg three times weekly group; the most common (≥20% patients in any group) grade 3-4 adverse events were thrombocytopenia (33 [42%] of 78, 26 [31%] of 83, and 19 [24%] of 83 patients) and neutropenia (18 [23%], 13 [16%], and six [8%])."( Efficacy and safety of oral panobinostat plus subcutaneous bortezomib and oral dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma (PANORAMA 3): an open-label, randomised, phase 2 study.
Abdo, A; Beksac, M; Bladé, J; Chari, A; Dimopoulos, MA; Gonçalves, IZ; Hájek, R; Hungria, VTM; Illés, Á; Jacobasch, L; Laubach, JP; Lech-Maranda, E; Lonial, S; Maison-Blanche, P; Mariz, M; Moreau, P; Polprasert, C; Rajkumar, SV; Richardson, PG; San-Miguel, JF; Schjesvold, F; Shelekhova, T; Spencer, A; Spicka, I; Sureda, A; Wróbel, T, 2021
)
0.92

Pharmacokinetics

Hepatic impairment did not alter panobinostat absorption with Tmax unchanged at 2 h. Using predictive physiologically-based pharmacokinetic (PBPK) models in the evaluation of the PK of alectinib,. ruxolitinib, and panobinstat exposures can help in using optimal doses with lower toxicity in these populations.

ExcerptReferenceRelevance
" No substantial change in T (max) or half-life was observed."( Effect of ketoconazole-mediated CYP3A4 inhibition on clinical pharmacokinetics of panobinostat (LBH589), an orally active histone deacetylase inhibitor.
Chen, LC; de Jonge, M; Hamberg, P; Hengelage, T; Li, W; Porro, MG; Sharma, S; van der Biessen, D; Verweij, J; Woo, MM; Zhao, L, 2011
)
0.59
" Correlative studies confirmed panobinostat's pharmacodynamic effect in blood, FPB, and tumor samples."( A phase I, pharmacokinetic, and pharmacodynamic study of panobinostat, an HDAC inhibitor, combined with erlotinib in patients with advanced aerodigestive tract tumors.
Akar, A; Altiok, S; Bepler, G; Chiappori, A; Gray, JE; Haura, E; Kish, JA; Kreahling, J; Lush, R; Neuger, A; Pinder-Schenck, M; Schell, MJ; Tanvetyanon, T; Tetteh, L; Williams, CC; Zhao, X, 2014
)
0.93
" Initially patients received a single oral dose of 30 mg panobinostat for a 1-week pharmacokinetic study (core phase)."( A phase I, open-label, multicenter study to evaluate the pharmacokinetics and safety of oral panobinostat in patients with advanced solid tumors and various degrees of hepatic function.
Clive, S; Gelderblom, H; Hess, D; Loman, N; Mu, S; Porro, MG; Sandstrom, P; Sharma, S; Slingerland, M; Valera, SZ; Waldron, E, 2014
)
0.87
" Pharmacokinetic parameters were derived using non-compartmental analysis."( A phase I, open-label, multicenter study to evaluate the pharmacokinetics and safety of oral panobinostat in patients with advanced solid tumors and varying degrees of renal function.
Gelderblom, H; Hess, D; Hussain, SA; Lolkema, MP; Mu, S; Porro, MG; Sharma, S; Valera, SZ; Waldron, E; Witteveen, PO, 2015
)
0.64
" Panobinostat pharmacokinetic parameters were best characterized by a three-compartment model with first-order absorption and elimination."( Population pharmacokinetics of intravenous and oral panobinostat in patients with hematologic and solid tumors.
Capdeville, R; Mu, S; Nedelman, J; Savelieva, M; Schran, H; Woo, MM, 2015
)
1.58
" The pharmacokinetic data of panobinostat in patients with hematologic malignancies and advanced solid tumors have been collated and reviewed from the various published clinical studies for over a decade."( Clinical pharmacokinetics of panobinostat, a novel histone deacetylase (HDAC) inhibitor: review and perspectives.
Srinivas, NR, 2017
)
1.04
" Pharmacokinetic studies demonstrated that exposure to panobinostat was proportional to the dose given, with no associations between pharmacokinetic parameters and age, weight, or body surface area."( Safety, pharmacokinetics, and pharmacodynamics of panobinostat in children, adolescents, and young adults with relapsed acute myeloid leukemia.
Alexander, TB; Bhojwani, D; Cooper, TM; Crews, KR; Ge, Y; Heym, KM; Inaba, H; Karol, SE; Klco, JM; Kuo, DJ; Lacayo, NJ; Mead, PE; Panetta, JC; Pui, CH; Ribeiro, RC; Rubnitz, JE; Schiff, DE; Taub, JW, 2020
)
1.06
" Using predictive physiologically-based pharmacokinetic (PBPK) models in the evaluation of the PK of alectinib, ruxolitinib, and panobinostat exposures in the presence of cancer, RIP, and HIP can help in using optimal doses with lower toxicity in these populations."( Physiologically-based pharmacokinetic model for alectinib, ruxolitinib, and panobinostat in the presence of cancer, renal impairment, and hepatic impairment.
Abu Kassab, HT; Abu Kwiak, AD; Abu Laila, SS; Abu Shameh, AJ; Al-Daoud, NM; Alhazabreh, MK; Alsmadi, MM; Alzughoul, SB; Jaber, SA; Jaradat, MM, 2021
)
1.06

Compound-Compound Interactions

Panobinostat is a histone deacetylase inhibitor that has shown synergistic preclinical anti-myeloma activity when combined with other agents.

ExcerptReferenceRelevance
" We aimed to define the maximum tolerated dose, toxicity, activity, and pharmacokinetics of oral panobinostat, a pan-deacetylase inhibitor, alone and in combination with docetaxel for the treatment of castration-resistant prostate cancer (CRPC)."( A phase I study of oral panobinostat alone and in combination with docetaxel in patients with castration-resistant prostate cancer.
Anand, A; Dzik-Jurasz, A; Hu, J; Rathkopf, D; Ross, RW; Scher, HI; Tanaka, E; Wong, BY; Woo, MM; Yang, W, 2010
)
0.89
" In arm II, oral panobinostat (15 mg) was administered on the same schedule in combination with docetaxel 75 mg/m(2) every 21 days."( A phase I study of oral panobinostat alone and in combination with docetaxel in patients with castration-resistant prostate cancer.
Anand, A; Dzik-Jurasz, A; Hu, J; Rathkopf, D; Ross, RW; Scher, HI; Tanaka, E; Wong, BY; Woo, MM; Yang, W, 2010
)
1.01
"Patients received oral panobinostat administered 2 or 3 times weekly (continuous or intermittent dosing in combination with intravenous gemcitabine administered on days 1, 8, and 15 every 28 days or on days 1 and 8 every 21 days)."( A phase I study of panobinostat in combination with gemcitabine in the treatment of solid tumors.
Bendell, JC; Burris, HA; Greco, FA; Infante, JR; Jones, SF; Murphy, PB; Spigel, DR; Thompson, DS; Yardley, DA, 2011
)
1.01
" The recommended doses for further study are intermittent oral panobinostat administered at a dose of 10 mg 3 times weekly for 2 weeks in combination with gemcitabine 800 mg/m2 administered intravenously on days 1 and 8 every 21 days."( A phase I study of panobinostat in combination with gemcitabine in the treatment of solid tumors.
Bendell, JC; Burris, HA; Greco, FA; Infante, JR; Jones, SF; Murphy, PB; Spigel, DR; Thompson, DS; Yardley, DA, 2011
)
0.94
" Patients received bevacizumab 10 mg/kg every other week in combination with oral panobinostat."( Phase I study of panobinostat in combination with bevacizumab for recurrent high-grade glioma.
Batchelor, TT; Beroukhim, R; Chi, AS; Ciampa, AS; Doherty, LM; Drappatz, J; Gerard, M; Grimm, SA; Hammond, S; Lafrankie, DC; Lee, EQ; Norden, AD; Raizer, JJ; Ruland, S; Schiff, D; Snodgrass, SM; Wen, PY, 2012
)
0.94
" Notably, the therapeutic efficacy of LBH589 was significantly enhanced in combination with vincristine and dexamethasone."( Preclinical activity of LBH589 alone or in combination with chemotherapy in a xenogeneic mouse model of human acute lymphoblastic leukemia.
Abizanda, G; Agirre, X; Blanco-Prieto, MJ; Calasanz, MJ; Cigudosa, JC; De Martino Rodriguez, A; Garate, L; García de Jalón, JA; José-Eneriz, ES; Martín-Subero, JI; Martinez-Climent, JA; Miranda, E; Moreno, C; Prósper, F; Ribera, JM; Rifón, J; Rio, P; Román-Gómez, J; Segura, V; Vilas-Zornoza, A, 2012
)
0.38
" In preclinical and clinical studies, panobinostat showed good anti-myeloma activity in combination with several agents."( Phase II study of melphalan, thalidomide and prednisone combined with oral panobinostat in patients with relapsed/refractory multiple myeloma.
Alesiani, F; Ballanti, S; Boccadoro, M; Caraffa, P; Catarini, M; Cavallo, F; Corvatta, L; Gentili, S; Leoni, P; Liberati, AM; Offidani, M; Palumbo, A; Polloni, C; Pulini, S, 2012
)
0.88
"To investigate reversal effect of histone deacetylase inhibitor LBH589 alone or in combination with proteasome inhibitor bortezomib on drug resistance in acute myeloid leukemia (AML) and its mechanism."( [Reversal effect of LBH589 alone or in combination with bortezomib on drug-resistance in myeloid leukemia and its mechanism].
Chen, WW; Huang, KK; Huang, M; Jiang, XJ; Meng, FY; Wang, Q; Wang, ZX; Wu, FQ; Zhou, HS, 2011
)
0.37
"To determine the maximum tolerated doses and dose-limiting toxicities of oral panobinostat in combination with paclitaxel and carboplatin when administered to patients with advanced solid tumors."( A phase I trial of oral administration of panobinostat in combination with paclitaxel and carboplatin in patients with solid tumors.
Bendell, JC; Burris, HA; Infante, JR; Jones, SF; Mohyuddin, A; Thompson, DS; Yardley, DA, 2012
)
0.87
"The recommended phase II dose is panobinostat 10 mg orally three times weekly in combination with paclitaxel 175 mg/m(2) and carboplatin AUC 5 administered intravenously on day 1 of every 21-day cycle."( A phase I trial of oral administration of panobinostat in combination with paclitaxel and carboplatin in patients with solid tumors.
Bendell, JC; Burris, HA; Infante, JR; Jones, SF; Mohyuddin, A; Thompson, DS; Yardley, DA, 2012
)
0.92
" Bortezomib, a proteasome inhibitor,markedly enhanced the cytotoxic effects of panobinostat combined with gemcitabine."( Identification of unique synergistic drug combinations associated with downexpression of survivin in a preclinical breast cancer model system.
Budman, DR; Calabro, A; Lesser, M; Rosen, L, 2012
)
0.61
" PANORAMA 2 is a phase 2 trial of panobinostat in combination with bortezomib and dexamethasone to treat patients with relapsed and bortezomib-refractory multiple myeloma (with ≥2 prior lines of therapy, including an immunomodulatory drug, and patients who had progressed on or within 60 days of the last bortezomib-based therapy)."( PANORAMA 2: panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma.
Alsina, M; Coutre, SE; Gasparetto, C; Khan, M; Lonial, S; Mukhopadhyay, S; Ondovik, MS; Paley, CS; Richardson, PG; Schlossman, RL; Weber, DM, 2013
)
1.05
" HDACi were combined with ABT-737, which targets the intrinsic apoptosis pathway, recombinant human tumour necrosis factor-related apoptosis-inducing ligand (rhTRAIL/MD5-1), that activates the extrinsic apoptosis pathway or the DNA methyl transferase inhibitor 5-azacytidine."( Preclinical screening of histone deacetylase inhibitors combined with ABT-737, rhTRAIL/MD5-1 or 5-azacytidine using syngeneic Vk*MYC multiple myeloma.
Atadja, P; Banks, KM; Bergsagel, PL; Chesi, M; Doyle, MA; Ellul, J; Faulkner, D; Johnstone, RW; Lefebure, M; Matthews, GM; Shortt, J; Vidacs, E, 2013
)
0.39
"Panobinostat is a histone deacetylase inhibitor that has shown synergistic preclinical anti-myeloma activity when combined with other agents, recently exhibiting synergy with the alkylating agent melphalan (Sanchez et al."( A phase 1/2 study of oral panobinostat combined with melphalan for patients with relapsed or refractory multiple myeloma.
Berenson, JR; Boccia, RV; Dressler, K; Ghazal, HH; Harb, WA; Hilger, JD; Jamshed, S; Kingsley, EC; Matous, J; Nassir, Y; Noga, SJ; Swift, RA; Vescio, R; Yellin, O, 2014
)
2.15
"Panobinostat, a histone deacetylase (HDAC) inhibitor, enhances antiproliferative activity in non-small cell lung cancer (NSCLC) cell lines when combined with erlotinib."( A phase I, pharmacokinetic, and pharmacodynamic study of panobinostat, an HDAC inhibitor, combined with erlotinib in patients with advanced aerodigestive tract tumors.
Akar, A; Altiok, S; Bepler, G; Chiappori, A; Gray, JE; Haura, E; Kish, JA; Kreahling, J; Lush, R; Neuger, A; Pinder-Schenck, M; Schell, MJ; Tanvetyanon, T; Tetteh, L; Williams, CC; Zhao, X, 2014
)
2.09
"2 mg/kg three times weekly for 2 weeks (starting on day 1), in combination with oral panobinostat 10, 20, or 30 mg every 96 h (starting on day 8), and oral temozolomide 150 mg/m(2)/day on days 9 through 13."( Treatment of resistant metastatic melanoma using sequential epigenetic therapy (decitabine and panobinostat) combined with chemotherapy (temozolomide).
Deutsch, J; Frees, M; Laux, D; Leon-Ferre, R; Milhem, M; Smith, BJ; Xia, C, 2014
)
0.84
"This triple agent of dual epigenetic therapy in combination with traditional chemotherapy was generally well tolerated by the cohort and appeared safe to be continued in a Phase II trial."( Treatment of resistant metastatic melanoma using sequential epigenetic therapy (decitabine and panobinostat) combined with chemotherapy (temozolomide).
Deutsch, J; Frees, M; Laux, D; Leon-Ferre, R; Milhem, M; Smith, BJ; Xia, C, 2014
)
0.62
" We conducted a multicenter phase II trial of panobinostat combined with bevacizumab in patients with recurrent high-grade glioma (HGG)."( Phase II study of panobinostat in combination with bevacizumab for recurrent glioblastoma and anaplastic glioma.
Batchelor, TT; Beroukhim, R; Chi, AS; Drappatz, J; Gaffey, SC; Grimm, SA; Hempfling, K; Lee, EQ; Ligon, KL; McCluskey, C; Muzikansky, A; Nayak, L; Norden, AD; Raizer, JJ; Reardon, DA; Rinne, ML; Schiff, D; Smith, KH; Wen, PY; Wrigley, B, 2015
)
1.01
"Patients with recurrent HGG were treated with oral panobinostat 30 mg 3 times per week, every other week, in combination with bevacizumab 10 mg/kg every other week."( Phase II study of panobinostat in combination with bevacizumab for recurrent glioblastoma and anaplastic glioma.
Batchelor, TT; Beroukhim, R; Chi, AS; Drappatz, J; Gaffey, SC; Grimm, SA; Hempfling, K; Lee, EQ; Ligon, KL; McCluskey, C; Muzikansky, A; Nayak, L; Norden, AD; Raizer, JJ; Reardon, DA; Rinne, ML; Schiff, D; Smith, KH; Wen, PY; Wrigley, B, 2015
)
1
"SAHA, Valproic Acid, Scriptaid, MS275 and LBH589 were combined with Delta24-RGD in fourteen distinct GSCs."( The HDAC Inhibitors Scriptaid and LBH589 Combined with the Oncolytic Virus Delta24-RGD Exert Enhanced Anti-Tumor Efficacy in Patient-Derived Glioblastoma Cells.
Berghauser Pont, LM; de Vrij, J; Dirven, CM; Kaufmann, JK; Kleijn, A; Kloezeman, JJ; Lamfers, ML; Leenstra, S; van den Bossche, W, 2015
)
0.42
"LBH589 and Scriptaid combined with Delta24-RGD revealed synergistic anti-tumor activity in a subset of GSCs."( The HDAC Inhibitors Scriptaid and LBH589 Combined with the Oncolytic Virus Delta24-RGD Exert Enhanced Anti-Tumor Efficacy in Patient-Derived Glioblastoma Cells.
Berghauser Pont, LM; de Vrij, J; Dirven, CM; Kaufmann, JK; Kleijn, A; Kloezeman, JJ; Lamfers, ML; Leenstra, S; van den Bossche, W, 2015
)
0.42
" Panobinostat can be combined with palliative-dose RT at doses up to 45 mg twice a week with tolerable toxicity."( Phase I study evaluating the safety and efficacy of oral panobinostat in combination with radiotherapy or chemoradiotherapy in patients with inoperable stage III non-small-cell lung cancer.
Brown, MP; Gowda, R; Penniment, M; Singhal, N; Takhar, HS; Takhar, P, 2015
)
1.57
"Panobinostat, a potent pan-deacetylase inhibitor, improved progression-free survival (PFS) in patients with relapsed and refractory multiple myeloma when combined with bortezomib and dexamethasone in a phase 3 trial, PANORAMA-1."( Panobinostat PK/PD profile in combination with bortezomib and dexamethasone in patients with relapsed and relapsed/refractory multiple myeloma.
Binlich, F; Corrado, C; Hino, M; Kuroda, Y; Lin, R; Mu, S; Shibayama, H; Suzuki, K; Tajima, T; Waldron, E, 2016
)
3.32
"Apparent panobinostat exposure-AE and exposure-ORR relationships were observed when combined with bortezomib and dexamethasone in the treatment of patients with relapsed and refractory multiple myeloma."( Panobinostat PK/PD profile in combination with bortezomib and dexamethasone in patients with relapsed and relapsed/refractory multiple myeloma.
Binlich, F; Corrado, C; Hino, M; Kuroda, Y; Lin, R; Mu, S; Shibayama, H; Suzuki, K; Tajima, T; Waldron, E, 2016
)
2.29
" Findings of many preclinical studies have shown synergistic antilymphoma activity when panobinostat is combined with the proteasome inhibitor bortezomib."( Panobinostat in combination with bortezomib in patients with relapsed or refractory peripheral T-cell lymphoma: an open-label, multicentre phase 2 trial.
Chan, YH; Fadilah, S; Goh, YT; Hwang, WY; Kim, WS; Kumar, SG; Lee, YS; Lim, ST; Ng, SC; Phipps, C; Tan, D; Tan, SY; Tay, K; Yeap, CH, 2015
)
2.08
" This phase I/II study was conducted to determine the maximum tolerated dose (MTD), safety, and efficacy of lenalidomide combined with panobinostat in relapsed/refractory HL."( A Phase I/II Trial of Panobinostat in Combination With Lenalidomide in Patients With Relapsed or Refractory Hodgkin Lymphoma.
Bartlett, NL; Blum, KA; Christian, BA; Devine, SM; Fehniger, TA; Jaglowski, SM; Maly, JJ; Phelps, MA; Sexton, JL; Wagner-Johnston, ND; Wei, L; Zhu, X, 2017
)
0.97
"This phase-I/phase-II study evaluated panobinostat in combination with ifosfamide, carboplatin, etoposide (P-ICE) in relapsed/refractory classical Hodgkin lymphoma."( Phase-I and randomized phase-II trial of panobinostat in combination with ICE (ifosfamide, carboplatin, etoposide) in relapsed or refractory classical Hodgkin lymphoma.
Claret, L; Copeland, AR; Fanale, MA; Fayad, LE; Feng, L; Fowler, N; Hagemeister, FB; Hu, B; Nastoupil, LJ; Neelapu, S; Nieto, Y; Oki, Y; Rodriguez, MA; Romaguera, J; Samaniego, F; Turturro, F; Westin, JR; Younes, A, 2018
)
1.02
" Based on a previous clinical drug-drug interaction study with ketoconazole (KTZ), the contribution of CYP3A4 in vivo was estimated to be ∼40%."( Physiologically Based Pharmacokinetic Model Predictions of Panobinostat (LBH589) as a Victim and Perpetrator of Drug-Drug Interactions.
Chun, DY; Einolf, HJ; Gu, H; He, H; Lin, W; Mangold, JB; Wang, L; Won, CS, 2017
)
0.7
"This is a phase II study of panobinostat, an oral pan-HDAC inhibitor, combined with rituximab in patients with relapsed diffuse large B cell lymphoma."( Panobinostat in combination with rituximab in heavily pretreated diffuse large B-cell lymphoma: Results of a phase II study.
Abramson, JS; Barnes, JA; Fisher, DC; Hochberg, EP; Jacobsen, E; Neuberg, D; Redd, R; Takvorian, T, 2018
)
2.22
" Both cell lines were treated with G1-S4, G2-S16 and G3-S16 either alone or in combination with bryostatin (BRY), romidepsin (RMD) or panobinostat (PNB) for 24 and 48 h."( Polyanionic carbosilane dendrimers as a new adjuvant in combination with latency reversal agents for HIV treatment.
Juárez-Sánchez, R; Muñoz, E; Muñoz-Fernández, MÁ; Pavicic, C; Relaño-Rodríguez, I, 2019
)
0.72
" Interestingly enough, G3-S16 dendrimer alone and its combination with BRY, RMD or PNB showed a significant increased expression of GFP in the THP89GFP monocyte cell line."( Polyanionic carbosilane dendrimers as a new adjuvant in combination with latency reversal agents for HIV treatment.
Juárez-Sánchez, R; Muñoz, E; Muñoz-Fernández, MÁ; Pavicic, C; Relaño-Rodríguez, I, 2019
)
0.51
"In our case, who had refractory metastatic neuroblastoma, we use histone deacetylase inhibitor (panobinostat) in combination with chemotherapy agents and iodine-131-meta-iodobenzylguanidine (MIBG) therapy."( Successful treatment of refractory metastatic neuroblastoma with panobinostat in combination with chemotherapy agents and iodine-131-meta-iodobenzylguanidine therapy.
Bordbar, M; Shahriari, M; Shakibazad, N; Zareifar, S; Zekavat, OR, 2020
)
1.01
"Panobinostat can cause apoptosis induction in refractory metastatic neuroblastoma in combination with MIBG therapy and chemotherapy."( Successful treatment of refractory metastatic neuroblastoma with panobinostat in combination with chemotherapy agents and iodine-131-meta-iodobenzylguanidine therapy.
Bordbar, M; Shahriari, M; Shakibazad, N; Zareifar, S; Zekavat, OR, 2020
)
2.24
" We completed a phase I, dose-finding trial for the mTOR inhibitor everolimus combined with the HDAC inhibitor panobinostat in advanced clear cell renal cell carcinoma (ccRCC) patients."( Phase I study of the mTOR inhibitor everolimus in combination with the histone deacetylase inhibitor panobinostat in patients with advanced clear cell renal cell carcinoma.
Adra, N; Chintala, S; Damayanti, N; George, S; Pili, R; Wood, A, 2020
)
0.99
" In addition, cellular and molecular investigations on NB4 revealed that not only panobinostat induced p21-mediated G1 arrest and ROS-mediated apoptosis, but also exerted a superior cytotoxicity when combined with c-Myc and autophagy inhibitors."( Pan-HDAC inhibitor panobinostat, as a single agent or in combination with PI3K inhibitor, induces apoptosis in APL cells: An emerging approach to overcome MSC-induced resistance.
Bashash, D; Mosleh, M; Safaroghli-Azar, A, 2020
)
1.11
" We demonstrated the synergism of [Bis + ABT199/venetoclax] in combination with panobinostat (Pano), decitabine (DAC), or olaparib (Ola), known inhibitors of BCL2, histone deacetylase, DNA methyltransferase, and poly(ADP-ribose) polymerase, respectively, in AML cells."( Enhanced cytotoxicity of bisantrene when combined with venetoclax, panobinostat, decitabine and olaparib in acute myeloid leukemia cells.
Andersson, BS; Murray, D; Nieto, Y; Popat, U; Valdez, BC; Yuan, B, 2022
)
1.18
" This study aimed to explore the in vitro and in vivo efficiency of histone deacetylase (HDAC) inhibitor panobinostat (PANO) in combination with mTOR inhibitor rapamycin (RAPA) against TNBC."( Histone deacetylase inhibitor panobinostat in combination with rapamycin confers enhanced efficacy against triple-negative breast cancer.
Chen, L; Chen, X; Kong, L; Lin, J; Mo, C; Wu, K; Xu, S; Zhang, H; Zhou, L, 2022
)
1.22
"In this study, oncolytic activity in vitro and antitumor therapeutic efficacy in vivo when combined with oHSV and panobinostat were investigated."( Histone Deacetylase Inhibitor Panobinostat Benefits the Therapeutic Efficacy of Oncolytic Herpes Simplex Virus Combined with PD-1/PD-L1 Blocking in Glioma and Squamous Cell Carcinoma Models.
Chen, X; Ji, D; Liu, Y; Ren, P; Wang, L; Wu, Y; Zhao, J; Zhou, GG; Zhou, X, 2022
)
1.22
" Plate cloning results showed that LBH589 combined with AM1241 inhibited the proliferation of cervical cancer cells compared to individual drug."( Panobinostat (LBH589) combined with AM1241 induces cervical cancer cell apoptosis through autophagy pathway.
Qu, X; Sheng, B; Wang, W; Xia, D, 2023
)
2.35
"LBH589 combined with AM1241 activated the endoplasmic reticulum emergency pathway, DNA damage repair signaling pathway, oxidative stress and autophagy pathway, ultimately promoting the apoptosis of cervical cancer cells."( Panobinostat (LBH589) combined with AM1241 induces cervical cancer cell apoptosis through autophagy pathway.
Qu, X; Sheng, B; Wang, W; Xia, D, 2023
)
2.35

Bioavailability

Panobinostat has demonstrated potent activity in multiple malignancies, including pediatric brain tumors such as DIPG. The study aimed to determine the influence of food on the oral bioavailability of panobinostats.

ExcerptReferenceRelevance
" 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
" The study aimed to determine the influence of food on the oral bioavailability of panobinostat."( The effect of food on the bioavailability of panobinostat, an orally active pan-histone deacetylase inhibitor, in patients with advanced cancer.
Dandamudi, UB; Frank, R; Gazi, L; Hengelage, T; Lewis, LD; Porro, MG; Shapiro, GI; Woo, MM; Zhao, L, 2012
)
0.86
" Bioavailability was 21."( Population pharmacokinetics of intravenous and oral panobinostat in patients with hematologic and solid tumors.
Capdeville, R; Mu, S; Nedelman, J; Savelieva, M; Schran, H; Woo, MM, 2015
)
0.67
" 23bb has a good pharmacokinetic profile with oral bioavailability of 47."( Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
Cao, D; Chen, L; Chen, X; Liu, Z; Long, C; Ma, L; Niu, T; Tang, M; Wang, F; Wang, T; Wang, X; Xiang, W; Yang, Z; Yi, Y; You, J, 2016
)
0.43
"In the present study, we assessed the therapeutic potential of LBH589, an orally bioavailable hydroxamic acid-derived nonselective HDAC inhibitor in mouse models of HD."( LBH589, A Hydroxamic Acid-Derived HDAC Inhibitor, is Neuroprotective in Mouse Models of Huntington's Disease.
Chopra, V; Hersch, S; Kazantsev, AG; Khanna, P; Kuhn, R; Paganetti, P; Quinti, L; Young, AB, 2016
)
0.43
"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
"Panobinostat, an orally bioavailable pan-HDAC inhibitor, has demonstrated potent activity in multiple malignancies, including pediatric brain tumors such as DIPG, with increased activity against H3K27M mutant cell lines."( Panobinostat penetrates the blood-brain barrier and achieves effective brain concentrations in a murine model.
Franson, A; He, M; Homan, MJ; Koschmann, C; Liu, C; Marini, BL; Matvekas, A; Pai, MP; Ravi, K; Roberts, H, 2021
)
3.51
" Here, we propose such a regimen by combining Pano with LTI6426, a first-in-class orally bioavailable protein disulfide isomerase (PDI) inhibitor."( PDI inhibitor LTI6426 enhances panobinostat efficacy in preclinical models of multiple myeloma.
Basar, AP; Dolloff, NG; Duncan, RM; Li, H; Reyes, L; Robinson, RM, 2022
)
1.01
" This orally bioavailable drug is classified as a non-selective histone deacetylase inhibitor (pan-HDACi) that inhibits class I, II, and IV HDACs at nanomolar levels due to its significant histone modifications and epigenetic mechanisms."( Molecular mechanisms underlying the clinical efficacy of panobinostat involve Stochasticity of epigenetic signaling, sensitization to anticancer drugs, and induction of cellular cell death related to cellular stresses.
Abdalla, AN; Almalki, WH; Ardianto, C; Bakrim, S; Bouyahya, A; El Omari, N; Khalid, A; Lee, LH; Ming, LC, 2023
)
1.16

Dosage Studied

The results from these studies were incorporated into the Food and Drug Administration-approved product label. Everolimus 5 mg PO daily and panobinostat 10 mgPO 3 times weekly was the recommended phase II dosing based on their maximum tolerated dose. Simulation using a compartmental BBB model suggests inadequate exposure of free panobinstat in the brain following a recommended oral dosing regimen.

ExcerptRelevanceReference
" In vivo dosing of mice with LBH589 (10 mg/kg/d) reduced angiogenesis and PC-3 tumor growth."( Targeting tumor angiogenesis with histone deacetylase inhibitors: the hydroxamic acid derivative LBH589.
Atadja, P; Kato, Y; Pili, R; Qian, DZ; Salumbides, B; Sanni, T; Shabbeer, S; Verheul, HM; Wei, Y, 2006
)
0.33
"Patients received oral panobinostat administered 2 or 3 times weekly (continuous or intermittent dosing in combination with intravenous gemcitabine administered on days 1, 8, and 15 every 28 days or on days 1 and 8 every 21 days)."( A phase I study of panobinostat in combination with gemcitabine in the treatment of solid tumors.
Bendell, JC; Burris, HA; Greco, FA; Infante, JR; Jones, SF; Murphy, PB; Spigel, DR; Thompson, DS; Yardley, DA, 2011
)
1.01
" 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
" Considering the variability in exposure following enzyme inhibition and the fact that chronic dosing of panobinostat was not studied with CYP3A inhibitors, close monitoring of panobinostat-related adverse events is necessary."( Effect of ketoconazole-mediated CYP3A4 inhibition on clinical pharmacokinetics of panobinostat (LBH589), an orally active histone deacetylase inhibitor.
Chen, LC; de Jonge, M; Hamberg, P; Hengelage, T; Li, W; Porro, MG; Sharma, S; van der Biessen, D; Verweij, J; Woo, MM; Zhao, L, 2011
)
0.81
" Due to concerns for thrombocytopenia with the weekly dosing regimen, the protocol was amended to examine an every other week regimen."( Phase I study of panobinostat in combination with bevacizumab for recurrent high-grade glioma.
Batchelor, TT; Beroukhim, R; Chi, AS; Ciampa, AS; Doherty, LM; Drappatz, J; Gerard, M; Grimm, SA; Hammond, S; Lafrankie, DC; Lee, EQ; Norden, AD; Raizer, JJ; Ruland, S; Schiff, D; Snodgrass, SM; Wen, PY, 2012
)
0.72
") panobinostat administered on different dosing schedules in patients with advanced solid tumors or lymphoma."( A phase I dose-escalation study of intravenous panobinostat in patients with lymphoma and solid tumors.
Beck, J; Gadbaw, B; Mita, M; Paul, S; Prince, HM; Sharma, S; Squier, M; Woo, MM, 2013
)
1.37
" Serial blood samples were collected following dosing for pharmacokinetic and pharmacodynamic analyses."( A phase I dose-escalation study of intravenous panobinostat in patients with lymphoma and solid tumors.
Beck, J; Gadbaw, B; Mita, M; Paul, S; Prince, HM; Sharma, S; Squier, M; Woo, MM, 2013
)
0.65
"The results of Phase II trials prove that oral panobinostat is deliverable with dosing regimens of three times per week, either weekly or biweekly."( Panobinostat in lymphoid and myeloid malignancies.
Dickinson, M; Khot, A; Prince, HM, 2013
)
2.09
" Panobinostat + melphalan appears to have tolerability issues in a dosing regimen capable of producing a response."( A phase 1/2 study of oral panobinostat combined with melphalan for patients with relapsed or refractory multiple myeloma.
Berenson, JR; Boccia, RV; Dressler, K; Ghazal, HH; Harb, WA; Hilger, JD; Jamshed, S; Kingsley, EC; Matous, J; Nassir, Y; Noga, SJ; Swift, RA; Vescio, R; Yellin, O, 2014
)
1.61
" Correlative studies in this trial may help to optimise dosing schedules in GIST."( Phase I study of panobinostat and imatinib in patients with treatment-refractory metastatic gastrointestinal stromal tumors.
Ahrens, M; Bauer, S; Bock, E; Grabellus, F; Grunewald, S; Hilger, RA; Hoiczyk, M; Mühlenberg, T; Nagarajah, J; Pink, D; Pustowka, A; Reichardt, A; Reichardt, P; Scheulen, ME; Schuler, M, 2014
)
0.74
"Although covariate analyses revealed significant effects of body size, age, and race on panobinostat pharmacokinetics, these effects were minor compared to the interindividual variability and therefore not clinically relevant when dosing panobinostat in populations similar to those studied."( Population pharmacokinetics of intravenous and oral panobinostat in patients with hematologic and solid tumors.
Capdeville, R; Mu, S; Nedelman, J; Savelieva, M; Schran, H; Woo, MM, 2015
)
0.89
" The N-hydroxyl group of this motif is highly subject to sulfation/glucoronidation-based inactivation in humans; compounds containing this motif require much higher dosing in clinic to achieve therapeutic concentrations."( Development of Allosteric Hydrazide-Containing Class I Histone Deacetylase Inhibitors for Use in Acute Myeloid Leukemia.
Chou, CJ; Inks, ES; Li, J; McClure, JJ; Peterson, YK; Zhang, C, 2016
)
0.43
" 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
" The optimal dosing of panobinostat for the treatment of MF remains somewhat ill-defined but appears to be most effective and better tolerated when administered at lower doses over a prolonged duration of therapy."( A phase II study of panobinostat in patients with primary myelofibrosis (PMF) and post-polycythemia vera/essential thrombocythemia myelofibrosis (post-PV/ET MF).
Goldberg, JD; Hochman, T; Hoffman, R; Lu, M; Mascarenhas, J; Najfeld, V; Newsom, C; Petersen, B; Sandy, L; Ye, F; Yoon, J; Zhang, D, 2017
)
1.09
" There is interest in evaluating alternate dosing schedules and novel combinations of pano for the treatment of upper aerodigestive and lung malignancies; thus we evaluated it in combination with Taxol, a chemotherapeutic with activity in both diseases."( A Novel Indication for Panobinostat as a Senolytic Drug in NSCLC and HNSCC.
Adomako, A; McDaid, HM; Rodriguez-Gabin, A; Samaraweera, L, 2017
)
0.77
" The incidences of several key AEs were lower in both arms following the planned reduction of bortezomib dosing frequency in TP2."( Panobinostat plus bortezomib and dexamethasone: impact of dose intensity and administration frequency on safety in the PANORAMA 1 trial.
Beksac, M; Corrado, C; Dimopoulos, MA; Einsele, H; Elghandour, A; Guenther, A; Hou, J; Hungria, VTM; Jedrzejczak, WW; Lee, JH; Lonial, S; Moreau, P; Na Nakorn, T; Paul, S; Redhu, S; Richardson, PG; Salwender, H; San-Miguel, JF; Schlossman, RL; Siritanaratkul, N; Sopala, M; Yoon, SS, 2017
)
1.9
" The results from these studies were incorporated into the Food and Drug Administration-approved product label, providing guidance for panobinostat dosing recommendations when it is combined with other drugs."( Physiologically Based Pharmacokinetic Model Predictions of Panobinostat (LBH589) as a Victim and Perpetrator of Drug-Drug Interactions.
Chun, DY; Einolf, HJ; Gu, H; He, H; Lin, W; Mangold, JB; Wang, L; Won, CS, 2017
)
0.9
" Lines of therapy, combination regimens, dosing and duration were measured."( Treatment patterns and medication adherence among patients diagnosed with multiple myeloma and treated with panobinostat.
Bhor, M; Bonafede, M; Chari, A; Davis, B; Eldjerou, L; Gilligan, AM; Globe, D; Stetsovsky, D; Talcott, J; Urniasz, A; Varker, H, 2018
)
0.69
" Everolimus 5 mg PO daily and panobinostat 10 mg PO 3 times weekly (weeks 1 and 2) given in 21-day cycles was the recommended phase II dosing based on their maximum tolerated dose."( Phase I study of the mTOR inhibitor everolimus in combination with the histone deacetylase inhibitor panobinostat in patients with advanced clear cell renal cell carcinoma.
Adra, N; Chintala, S; Damayanti, N; George, S; Pili, R; Wood, A, 2020
)
1.06
" We investigated the activity and safety of three different dosing regimens of oral panobinostat in combination with subcutaneous bortezomib and oral dexamethasone for this indication."( Efficacy and safety of oral panobinostat plus subcutaneous bortezomib and oral dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma (PANORAMA 3): an open-label, randomised, phase 2 study.
Abdo, A; Beksac, M; Bladé, J; Chari, A; Dimopoulos, MA; Gonçalves, IZ; Hájek, R; Hungria, VTM; Illés, Á; Jacobasch, L; Laubach, JP; Lech-Maranda, E; Lonial, S; Maison-Blanche, P; Mariz, M; Moreau, P; Polprasert, C; Rajkumar, SV; Richardson, PG; San-Miguel, JF; Schjesvold, F; Shelekhova, T; Spencer, A; Spicka, I; Sureda, A; Wróbel, T, 2021
)
1.14
" We showed that it is well-tolerated at the dosage schedule that we describe, with no serious adverse effects throughout the study period."( Panobinostat in adults with H3 K27M-mutant diffuse midline glioma: a single-center experience.
Balakrishnan, SN; Carabenciov, ID; Daniels, DJ; Kizilbash, SH; Neth, BJ; Ruff, MW; Uhm, JH, 2022
)
2.16
" In routine clinical care, PanBorDex is used primarily in later relapses and is commonly administered in attenuated dosing schedules to mitigate the treatment-related toxicity."( Panobinostat in combination with bortezomib and dexamethasone in multiply relapsed and refractory myeloma; UK routine care cohort.
Basu, S; Bhatti, Z; Collings, F; Karim, F; Kishore, B; Kothari, J; Leary, H; Maouche, N; Ramasamy, K; Reddy, U; Ryman, N; Sundararaman, S; Tseu, B; Vallance, GD, 2022
)
2.16
" Simulation using a compartmental BBB model suggests inadequate exposure of free panobinostat in the brain following a recommended oral dosing regimen in patients."( Central Nervous System Distribution of Panobinostat in Preclinical Models to Guide Dosing for Pediatric Brain Tumors.
Elmquist, WF; Larson, JD; Oh, JH; Rathi, S; Sirianni, RW; Wechsler-Reya, RJ; Zhang, W, 2023
)
1.41
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Roles (3)

RoleDescription
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).
antineoplastic agentA substance that inhibits or prevents the proliferation of neoplasms.
angiogenesis modulating agentAn agent that modulates the physiologic angiogenesis process. This is accomplished by endogenous angiogenic proteins and a variety of other chemicals and pharmaceutical agents.
[role information is derived from Chemical Entities of Biological Interest (ChEBI), Hastings J, Owen G, Dekker A, Ennis M, Kale N, Muthukrishnan V, Turner S, Swainston N, Mendes P, Steinbeck C. (2016). ChEBI in 2016: Improved services and an expanding collection of metabolites. Nucleic Acids Res]

Drug Classes (4)

ClassDescription
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.
cinnamamidesAn enamide which is cinnamamide or a derivative of cinnamamide obtained by replacement of one or more of its hydrogens.
secondary amino compoundA compound formally derived from ammonia by replacing two hydrogen atoms by organyl groups.
methylindoleAny member of the class of indoles carrying one or more methyl substituents.
[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 (29)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
PPM1D proteinHomo sapiens (human)Potency0.09300.00529.466132.9993AID1347411
Interferon betaHomo sapiens (human)Potency0.09300.00339.158239.8107AID1347411
Spike glycoproteinSevere acute respiratory syndrome-related coronavirusPotency4.46680.009610.525035.4813AID1479145
[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 8Schistosoma mansoniIC50 (µMol)0.45000.30341.33503.6200AID1446915
Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)IC50 (µMol)10.00000.00010.33717.3000AID1312866
Histone deacetylase 3Homo sapiens (human)IC50 (µMol)0.20310.00040.619610.0000AID1210336; AID1282239; AID1312853; AID1312867; AID1313942; AID1321706; AID1431805; AID1525779; AID1542181; AID1545759; AID1548728; AID1548739; AID1548756; AID1548757; AID1548758; AID1548759; AID1559803; AID1589101; AID1600732; AID1600735; AID1775551; AID1801572; AID1882466; AID748112; AID750109
Histone deacetylase 3Homo sapiens (human)Ki0.00170.00020.42378.1900AID1236444; AID1399816; AID496803; AID619048
Bromodomain-containing protein 4Homo sapiens (human)IC50 (µMol)0.00500.00040.40329.0500AID1462230
Leukotriene A-4 hydrolaseHomo sapiens (human)IC50 (µMol)10.00000.00051.28547.6500AID1441630; AID1441631
Cytochrome P450 3A5Homo sapiens (human)IC50 (µMol)0.00330.00330.70736.2000AID1371032
Phosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)IC50 (µMol)10.00000.00000.683210.0000AID1312866
Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)IC50 (µMol)10.00000.00020.595310.0000AID1312864
Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)IC50 (µMol)10.00000.00030.660710.0000AID1312865
Histone deacetylase 4Homo sapiens (human)IC50 (µMol)0.79630.00061.052610.0000AID1210337; AID1282240; AID1312855; AID1312867; AID1313945; AID1542182; AID1545759; AID1548741; AID1559809; AID1600732; AID1600735; AID1723749; AID1801572; AID1821196; AID748111; AID750109
Histone deacetylase 4Homo sapiens (human)Ki0.38210.00021.62559.1242AID1236446; AID1399818; AID1600730; AID496804; AID619049
Histone deacetylase 1Homo sapiens (human)IC50 (µMol)0.18100.00010.55439.9000AID1210334; AID1282229; AID1312847; AID1312867; AID1313935; AID1321704; AID1431806; AID1525777; AID1542179; AID1545759; AID1548726; AID1548737; AID1548752; AID1548753; AID1548754; AID1548755; AID1559802; AID1589101; AID1589105; AID1591853; AID1600732; AID1600735; AID1775549; AID1785449; AID1801572; AID1882456; AID619041; AID748114; AID750109
Histone deacetylase 1Homo sapiens (human)Ki0.00160.00000.49888.1900AID1236442; AID1399812; AID1399814; AID496801; AID619046
NAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)IC50 (µMol)20.00000.50003.848110.0000AID1559817
Histone deacetylase 7Homo sapiens (human)IC50 (µMol)1.85090.00071.02609.9000AID1282242; AID1312857; AID1312867; AID1313948; AID1542185; AID1545759; AID1548743; AID1559811; AID1600732; AID1600735; AID1801572; AID1821198; AID750109
Histone deacetylase 7Homo sapiens (human)Ki2.43310.00022.00059.5000AID1236448; AID1399820; AID496807; AID619052
Histone deacetylase 2Homo sapiens (human)IC50 (µMol)0.25270.00010.72219.9700AID1210335; AID1282238; AID1312852; AID1312867; AID1313940; AID1321705; AID1542180; AID1545759; AID1548727; AID1548738; AID1559808; AID1589101; AID1589106; AID1600732; AID1600735; AID1775550; AID1801572; AID748113; AID750109
Histone deacetylase 2Homo sapiens (human)Ki0.00770.00000.47098.1900AID1236443; AID1399815; AID1600731; AID496802; AID619047
Polyamine deacetylase HDAC10Homo sapiens (human)IC50 (µMol)0.00700.00050.72459.9000AID1282244; AID1312860; AID1312867; AID1542188; AID1545759; AID1559814; AID1600732; AID1600735; AID1821201; AID750109
Polyamine deacetylase HDAC10Homo sapiens (human)Ki0.01800.00000.76878.1900AID1399823; AID619055
Histone deacetylase 11 Homo sapiens (human)IC50 (µMol)0.83960.00030.92989.9000AID1282245; AID1312861; AID1312867; AID1542189; AID1545759; AID1559815; AID1600732; AID1600735; AID1821202; AID750109
Histone deacetylase 11 Homo sapiens (human)Ki0.60180.00011.21478.1900AID1399828; AID619056
NAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)IC50 (µMol)20.00000.00601.62509.0000AID1559816
NAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)Ki10.00000.11400.11400.1140AID1399824
Histone deacetylase 8Homo sapiens (human)IC50 (µMol)0.46000.00070.99479.9000AID1210339; AID1282231; AID1312854; AID1312867; AID1313949; AID1431803; AID1525780; AID1542186; AID1545759; AID1548740; AID1559812; AID1589101; AID1600732; AID1600735; AID1801572; AID1821199; AID1882467; AID748106; AID750109
Histone deacetylase 8Homo sapiens (human)Ki0.23040.00020.75258.1900AID1236445; AID1399817; AID1600728; AID496808; AID619053
NAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)IC50 (µMol)20.00000.85005.430010.0000AID1559818
Histone deacetylase 6Homo sapiens (human)IC50 (µMol)0.23070.00000.53769.9000AID1210338; AID1282230; AID1312859; AID1312867; AID1313947; AID1321707; AID1431804; AID1525776; AID1542184; AID1545759; AID1548729; AID1548745; AID1559804; AID1600732; AID1600735; AID1775552; AID1785450; AID1801572; AID1882260; AID1882462; AID748109; AID750109
Histone deacetylase 6Homo sapiens (human)Ki0.01490.00010.41568.1900AID1236450; AID1399822; AID1600729; AID1802011; AID496806; AID619051
Histone deacetylase 9Homo sapiens (human)IC50 (µMol)1.50710.00050.94139.9000AID1282243; AID1312858; AID1312867; AID1313950; AID1542187; AID1545759; AID1548744; AID1559813; AID1600732; AID1600735; AID1801572; AID1821200; AID750109
Histone deacetylase 9Homo sapiens (human)Ki1.80780.00021.85209.0000AID1236449; AID1399821; AID496809; AID619054
Histone deacetylase 5Homo sapiens (human)IC50 (µMol)1.19710.00070.961010.0000AID1282241; AID1312856; AID1312867; AID1313946; AID1542183; AID1545759; AID1548742; AID1559810; AID1600732; AID1600735; AID1801572; AID1816688; AID1821197; AID748110; AID750109
Histone deacetylase 5Homo sapiens (human)Ki0.05440.00021.29939.5000AID1236447; AID1399819; AID496805; AID619050
Histone deacetylase Plasmodium falciparum (malaria parasite P. falciparum)IC50 (µMol)0.00250.00060.16880.9400AID1371032; AID414980
Nuclear receptor corepressor 2Homo sapiens (human)IC50 (µMol)0.00200.00170.59528.0000AID1312853; AID1548728; AID1548756; AID1548757; AID1548758; AID1548759
[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)0.16950.03001.85126.7000AID1312869
Protein TatHIV-1 M:B_HXB2REC50 (µMol)0.13000.13003.51008.1000AID1598103
Histone deacetylase 1Homo sapiens (human)EC50 (µMol)0.16950.03001.98776.7000AID1312869
Histone deacetylase 2Homo sapiens (human)EC50 (µMol)0.16950.03001.85756.7000AID1312869
Histone deacetylase 6Homo sapiens (human)EC50 (µMol)0.15070.00521.59986.7000AID1312868
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (474)

Processvia Protein(s)Taxonomy
phosphorylationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
natural killer cell differentiationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
positive regulation of cytokine productionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
positive regulation of endothelial cell proliferationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
adaptive immune responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
mast cell chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
respiratory burst involved in defense responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
protein phosphorylationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
inflammatory responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
immune responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
positive regulation of endothelial cell migrationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
positive regulation of gene expressionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
T cell chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
natural killer cell activationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
B cell differentiationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
T cell differentiationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
positive regulation of cell migrationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
neutrophil chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
positive regulation of neutrophil apoptotic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
natural killer cell chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
B cell chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
vascular endothelial growth factor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
T cell activationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
B cell activationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
mast cell degranulationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
innate immune responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
positive regulation of angiogenesisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
T cell receptor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
B cell receptor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
mast cell differentiationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
neutrophil extravasationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
positive regulation of epithelial tube formationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
cell migrationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
phosphatidylinositol-3-phosphate biosynthetic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
phosphatidylinositol-mediated signalingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
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)
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)
intracellular glucose homeostasisPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
negative regulation of cell-matrix adhesionPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of leukocyte migrationPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
transcription by RNA polymerase IIPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
protein import into nucleusPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
immune responsePhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
extrinsic apoptotic signaling pathway via death domain receptorsPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damagePhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of lamellipodium assemblyPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
cytokine-mediated signaling pathwayPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
B cell differentiationPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
T cell differentiationPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
osteoclast differentiationPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of tumor necrosis factor productionPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
cellular response to insulin stimulusPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of RNA splicingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
regulation of toll-like receptor 4 signaling pathwayPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
substrate adhesion-dependent cell spreadingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
cellular response to UVPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
response to endoplasmic reticulum stressPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
interleukin-18-mediated signaling pathwayPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
natural killer cell mediated cytotoxicityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of protein import into nucleusPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
negative regulation of apoptotic processPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
negative regulation of osteoclast differentiationPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of glucose importPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol phosphate biosynthetic processPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
insulin-like growth factor receptor signaling pathwayPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of smooth muscle cell proliferationPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
protein stabilizationPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of filopodium assemblyPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
negative regulation of stress fiber assemblyPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
growth hormone receptor signaling pathwayPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
myeloid leukocyte migrationPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of focal adhesion disassemblyPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of endoplasmic reticulum unfolded protein responsePhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
positive regulation of protein localization to plasma membranePhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
insulin receptor signaling pathwayPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
regulation of cell-matrix adhesionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
positive regulation of gene expressionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
phosphorylationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
positive regulation of nitric oxide biosynthetic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
endothelial cell proliferationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
response to ischemiaPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
sphingosine-1-phosphate receptor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
intracellular calcium ion homeostasisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
endocytosisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
autophagyPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
homophilic cell adhesion via plasma membrane adhesion moleculesPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
cell surface receptor protein tyrosine kinase signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
G protein-coupled receptor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
positive regulation of autophagyPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
positive regulation of endothelial cell migrationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
cell migrationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
platelet activationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
positive regulation of neutrophil apoptotic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
positive regulation of Rac protein signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
embryonic cleavagePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
negative regulation of MAPK cascadePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
phosphatidylinositol phosphate biosynthetic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
negative regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
angiogenesis involved in wound healingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
platelet aggregationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
negative regulation of vascular endothelial growth factor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
negative regulation of hypoxia-induced intrinsic apoptotic signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
negative regulation of sprouting angiogenesisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
regulation of clathrin-dependent endocytosisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
phosphatidylinositol-3-phosphate biosynthetic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
phosphatidylinositol-mediated signalingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
phosphorylationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
angiogenesisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
positive regulation of cytokine productionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
adaptive immune responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
dendritic cell chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
positive regulation of acute inflammatory responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
respiratory burst involved in defense responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
sphingosine-1-phosphate receptor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
endocytosisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
inflammatory responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
immune responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
G protein-coupled receptor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
phospholipase C-activating G protein-coupled receptor signaling pathwayPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
positive regulation of cytosolic calcium ion concentrationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
positive regulation of endothelial cell migrationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
T cell chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
negative regulation of triglyceride catabolic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
neutrophil chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
secretory granule localizationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
regulation of cell adhesion mediated by integrinPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
positive regulation of Rac protein signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
natural killer cell chemotaxisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
T cell proliferationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
T cell activationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
mast cell degranulationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
positive regulation of MAP kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
phosphatidylinositol 3-kinase/protein kinase B signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
innate immune responsePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
regulation of angiogenesisPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
phosphatidylinositol phosphate biosynthetic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
negative regulation of cardiac muscle contractionPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
platelet aggregationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
cellular response to cAMPPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
neutrophil extravasationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
hepatocyte apoptotic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
regulation of calcium ion transmembrane transportPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
negative regulation of fibroblast apoptotic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
cell migrationPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
phosphatidylinositol-mediated signalingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
phosphatidylinositol-3-phosphate biosynthetic processPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo 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)
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 IINAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
rDNA heterochromatin formationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
protein deacetylationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
autophagyNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
mitotic nuclear membrane reassemblyNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
regulation of exit from mitosisNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of autophagyNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of peptidyl-threonine phosphorylationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
substantia nigra developmentNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
myelination in peripheral nervous systemNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
heterochromatin formationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
subtelomeric heterochromatin formationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
regulation of myelinationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of proteasomal ubiquitin-dependent protein catabolic processNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
cellular response to oxidative stressNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
peptidyl-lysine deacetylationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
epigenetic regulation of gene expressionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of protein catabolic processNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
regulation of phosphorylationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of DNA bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
post-translational protein modificationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
cellular lipid catabolic processNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NLRP3 inflammasome complex assemblyNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
innate immune responseNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of fat cell differentiationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of fatty acid biosynthetic processNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of meiotic nuclear divisionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of striated muscle tissue developmentNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of DNA-templated transcriptionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of transcription by RNA polymerase IINAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
cell divisionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
meiotic cell cycleNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
regulation of cell cycleNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
response to redox stateNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of cell divisionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of attachment of spindle microtubules to kinetochoreNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
cellular response to caloric restrictionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of oligodendrocyte progenitor proliferationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
cellular response to hypoxiaNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
cellular response to epinephrine stimulusNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
tubulin deacetylationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of execution phase of apoptosisNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
positive regulation of oocyte maturationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of NLRP3 inflammasome complex assemblyNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of satellite cell differentiationNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
negative regulation of reactive oxygen species metabolic processNAD-dependent protein deacetylase sirtuin-2Homo 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)
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)
single strand break repairNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of transcription by RNA polymerase IINAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
rDNA heterochromatin formationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
pyrimidine dimer repair by nucleotide-excision repairNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
DNA synthesis involved in DNA repairNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
angiogenesisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
ovulation from ovarian follicleNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
intracellular glucose homeostasisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of protein phosphorylationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of endothelial cell proliferationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of adaptive immune responseNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
chromatin organizationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
DNA methylation-dependent heterochromatin formationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
protein deacetylationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
triglyceride mobilizationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
DNA damage responseNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
response to oxidative stressNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
transforming growth factor beta receptor signaling pathwayNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
spermatogenesisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of mitotic cell cycleNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
muscle organ developmentNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of cell population proliferationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cellular response to starvationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of gene expressionNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of centrosome duplicationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of triglyceride biosynthetic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of cholesterol effluxNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of lipid storageNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of glucose metabolic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of macroautophagyNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
protein ubiquitinationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
peptidyl-lysine acetylationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
macrophage differentiationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of transforming growth factor beta receptor signaling pathwayNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of prostaglandin biosynthetic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
heterochromatin formationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
protein destabilizationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of TOR signalingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of endodeoxyribonuclease activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of NF-kappaB transcription factor activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
response to insulinNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
circadian regulation of gene expressionNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
leptin-mediated signaling pathwayNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of smooth muscle cell apoptotic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
intracellular triglyceride homeostasisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of peroxisome proliferator activated receptor signaling pathwayNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of cell population proliferationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cellular response to glucose starvationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of phosphorylationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
response to hydrogen peroxideNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
behavioral response to starvationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cholesterol homeostasisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of apoptotic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of apoptotic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of apoptotic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of canonical NF-kappaB signal transductionNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
proteasome-mediated ubiquitin-dependent protein catabolic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of cysteine-type endopeptidase activity involved in apoptotic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of DNA-binding transcription factor activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of DNA damage response, signal transduction by p53 class mediatorNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of neuron apoptotic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of blood vessel endothelial cell migrationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
response to leptinNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of MHC class II biosynthetic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of fat cell differentiationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of gluconeogenesisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of DNA repairNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of angiogenesisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of cell cycleNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of DNA-templated transcriptionNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of transcription by RNA polymerase IINAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of transcription by glucoseNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of insulin receptor signaling pathwayNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
white fat cell differentiationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of helicase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of smooth muscle cell differentiationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
maintenance of nucleus locationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
fatty acid homeostasisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of androgen receptor signaling pathwayNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of macrophage cytokine productionNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cellular response to hydrogen peroxideNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of bile acid biosynthetic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
UV-damage excision repairNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cellular response to tumor necrosis factorNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cellular response to hypoxiaNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cellular response to ionizing radiationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of protein serine/threonine kinase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of brown fat cell differentiationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
stress-induced premature senescenceNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
energy homeostasisNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
protein depropionylationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
DNA repair-dependent chromatin remodelingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
regulation of cellular response to heatNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of signal transduction by p53 class mediatorNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of protein acetylationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of intrinsic apoptotic signaling pathway in response to DNA damage by p53 class mediatorNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of oxidative stress-induced intrinsic apoptotic signaling pathwayNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of endoplasmic reticulum stress-induced intrinsic apoptotic signaling pathwayNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of adipose tissue developmentNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cellular response to leukemia inhibitory factorNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of macrophage apoptotic processNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of cAMP-dependent protein kinase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of cAMP-dependent protein kinase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of cellular response to testosterone stimulusNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of peptidyl-lysine acetylationNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
negative regulation of cellular senescenceNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of cellular senescenceNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
positive regulation of double-strand break repairNAD-dependent protein deacetylase sirtuin-1Homo 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)
negative regulation of transcription by RNA polymerase IINAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
osteoblast differentiationNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
DNA repairNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
regulation of DNA repairNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
regulation of transcription by RNA polymerase IINAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein deacetylationNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
DNA damage responseNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
homologous chromosome pairing at meiosisNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
rRNA transcriptionNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
retrotransposon silencingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
regulation of mitochondrion organizationNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
negative regulation of protein ubiquitinationNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
positive regulation of gluconeogenesisNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
negative regulation of gene expression, epigeneticNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
transcription initiation-coupled chromatin remodelingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
positive regulation of transcription by RNA polymerase INAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
regulation of protein export from nucleusNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
negative regulation of phosphatidylinositol 3-kinase/protein kinase B signal transductionNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein deglutarylationNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
R-loop processingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein depropionylationNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
DNA repair-dependent chromatin remodelingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
regulation of transcription of nucleolar large rRNA by RNA polymerase INAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
positive regulation of rRNA processingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
peptidyl-lysine desuccinylationNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
epigenetic regulation of gene expressionNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
peptidyl-lysine demalonylationNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
chromatin remodelingNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
protein deacetylationNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
aerobic respirationNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
positive regulation of insulin secretionNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
peptidyl-lysine deacetylationNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
negative regulation of ERK1 and ERK2 cascadeNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
positive regulation of superoxide dismutase activityNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
positive regulation of catalase activityNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
positive regulation of ceramide biosynthetic processNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
negative regulation of reactive oxygen species metabolic processNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo 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)
negative regulation of transcription by RNA polymerase IINuclear receptor corepressor 2Homo sapiens (human)
lactationNuclear receptor corepressor 2Homo sapiens (human)
response to organonitrogen compoundNuclear receptor corepressor 2Homo sapiens (human)
regulation of cellular ketone metabolic processNuclear receptor corepressor 2Homo sapiens (human)
cerebellum developmentNuclear receptor corepressor 2Homo sapiens (human)
response to estradiolNuclear receptor corepressor 2Homo sapiens (human)
estrous cycleNuclear receptor corepressor 2Homo sapiens (human)
negative regulation of DNA-templated transcriptionNuclear receptor corepressor 2Homo sapiens (human)
negative regulation of androgen receptor signaling pathwayNuclear receptor corepressor 2Homo sapiens (human)
negative regulation of miRNA transcriptionNuclear receptor corepressor 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (141)

Processvia Protein(s)Taxonomy
protein bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
ATP bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
1-phosphatidylinositol-3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
1-phosphatidylinositol-4,5-bisphosphate 3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
1-phosphatidylinositol-4-phosphate 3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
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)
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)
phosphotyrosine residue bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
transmembrane receptor protein tyrosine kinase adaptor activityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
insulin receptor bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
insulin-like growth factor receptor bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
neurotrophin TRKA receptor bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
protein bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
kinase activator activityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
protein phosphatase bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol 3-kinase regulator activityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol 3-kinase regulatory subunit bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
ErbB-3 class receptor bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol 3-kinase bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
insulin bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
insulin receptor substrate bindingPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
protein heterodimerization activityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol kinase activityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
enzyme-substrate adaptor activityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol 3-kinase activator activityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
1-phosphatidylinositol-3-kinase regulator activityPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
protein bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
ATP bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
1-phosphatidylinositol-3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
insulin receptor substrate bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
1-phosphatidylinositol-4,5-bisphosphate 3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
protein serine kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
1-phosphatidylinositol-4-phosphate 3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
protein kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
protein serine/threonine kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
protein bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
ATP bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
1-phosphatidylinositol-3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
1-phosphatidylinositol-4-phosphate 3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
identical protein bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
ephrin receptor bindingPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
1-phosphatidylinositol-4,5-bisphosphate 3-kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
protein serine kinase activityPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo 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)
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)
NAD+ ADP-ribosyltransferase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NAD+-protein ADP-ribosyltransferase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
chromatin bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NAD+ ADP-ribosyltransferase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
histone deacetylase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
protein bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
zinc ion bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NAD-dependent histone deacetylase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
protein lysine deacetylase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NAD-dependent protein lysine deacetylase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
histone acetyltransferase bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
histone deacetylase bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
tubulin deacetylase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
ubiquitin bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NAD-dependent histone H4K16 deacetylase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NAD+ bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
DNA-binding transcription factor bindingNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NAD-dependent protein demyristoylase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
NAD-dependent protein depalmitoylase activityNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
transcription factor bindingNAD-dependent protein deacetylase sirtuin-2Homo 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)
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)
NAD+ ADP-ribosyltransferase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
NAD+-protein ADP-ribosyltransferase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
RNA polymerase II cis-regulatory region sequence-specific DNA bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
p53 bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
transcription coactivator activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
transcription corepressor activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
histone deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
protein bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nuclear receptor bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
NAD-dependent histone deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
enzyme bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
NAD-dependent histone H3K14 deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
protein lysine deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
NAD-dependent protein lysine deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
histone bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
identical protein bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
HLH domain bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
bHLH transcription factor bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
metal ion bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
NAD-dependent histone H3K9 deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
NAD-dependent histone H4K16 deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
mitogen-activated protein kinase bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
lysine-acetylated histone bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
protein-propionyllysine depropionylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
DNA-binding transcription factor bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
histone H4K12 deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
histone H3K deacetylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
NAD-dependent histone decrotonylase activityNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
keratin filament bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
promoter-specific chromatin bindingNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
NAD+ bindingNAD-dependent protein deacetylase sirtuin-1Homo 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)
chromatin bindingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein bindingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein methyltransferase activityNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
NAD-dependent protein lysine deacetylase activityNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein-succinyllysine desuccinylase activityNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
metal ion bindingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein-glutaryllysine deglutarylase activityNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
NAD+ bindingNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
NAD-dependent histone H3K18 deacetylase activityNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein-propionyllysine depropionylase activityNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
protein-malonyllysine demalonylase activityNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
NAD+ ADP-ribosyltransferase activityNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
NAD+-protein ADP-ribosyltransferase activityNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
protein bindingNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
zinc ion bindingNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
enzyme bindingNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
NAD-dependent protein lysine deacetylase activityNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
sequence-specific DNA bindingNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
NAD+ bindingNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
NAD-dependent histone deacetylase activityNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo 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)
DNA bindingNuclear receptor corepressor 2Homo sapiens (human)
chromatin bindingNuclear receptor corepressor 2Homo sapiens (human)
transcription corepressor activityNuclear receptor corepressor 2Homo sapiens (human)
Notch bindingNuclear receptor corepressor 2Homo sapiens (human)
protein bindingNuclear receptor corepressor 2Homo sapiens (human)
nuclear glucocorticoid receptor bindingNuclear receptor corepressor 2Homo sapiens (human)
histone deacetylase bindingNuclear receptor corepressor 2Homo sapiens (human)
nuclear retinoid X receptor bindingNuclear receptor corepressor 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (79)

Processvia Protein(s)Taxonomy
cytosolPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
plasma membranePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
phosphatidylinositol 3-kinase complex, class IAPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
phosphatidylinositol 3-kinase complexPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
cytoplasmPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
plasma membranePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit delta isoformHomo sapiens (human)
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)
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)
nucleusPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
cytoplasmPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
cis-Golgi networkPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
cytosolPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
plasma membranePhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
cell-cell junctionPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol 3-kinase complex, class IAPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
membranePhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
perinuclear region of cytoplasmPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
perinuclear endoplasmic reticulum membranePhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
phosphatidylinositol 3-kinase complexPhosphatidylinositol 3-kinase regulatory subunit alphaHomo sapiens (human)
nucleusPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
nucleoplasmPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
nucleolusPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
cytosolPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
phosphatidylinositol 3-kinase complex, class IAPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
midbodyPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
intracellular membrane-bounded organellePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
plasma membranePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
cytoplasmPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
phosphatidylinositol 3-kinase complexPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoformHomo sapiens (human)
cytoplasmPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
cytosolPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
plasma membranePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
phosphatidylinositol 3-kinase complex, class IAPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
membranePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
phosphatidylinositol 3-kinase complex, class IBPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
plasma membranePhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo sapiens (human)
cytoplasmPhosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform Homo 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
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)
chromosome, telomeric regionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
nucleusNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
chromosomeNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
nucleolusNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
cytoplasmNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
mitochondrionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
centrosomeNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
centrioleNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
spindleNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
cytosolNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
microtubuleNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
plasma membraneNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
growth coneNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
midbodyNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
paranodal junctionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
paranode region of axonNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
perikaryonNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
myelin sheathNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
lateral loopNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
Schmidt-Lanterman incisureNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
juxtaparanode region of axonNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
perinuclear region of cytoplasmNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
mitotic spindleNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
meiotic spindleNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
glial cell projectionNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
heterochromatinNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
chromatin silencing complexNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
nucleusNAD-dependent protein deacetylase sirtuin-2Homo sapiens (human)
nucleusHistone deacetylase 7Homo sapiens (human)
nucleoplasmHistone deacetylase 7Homo sapiens (human)
cytoplasmHistone deacetylase 7Homo sapiens (human)
cytosolHistone deacetylase 7Homo 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)
nucleolusNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cytoplasmNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
ESC/E(Z) complexNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cytosolNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
fibrillar centerNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nucleusNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nuclear envelopeNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nuclear inner membraneNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nucleoplasmNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nucleolusNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cytoplasmNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
mitochondrionNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
cytosolNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
PML bodyNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
eNoSc complexNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
chromatinNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
euchromatinNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
heterochromatinNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
chromatin silencing complexNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
rDNA heterochromatinNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nucleusNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nuclear inner membraneNAD-dependent protein deacetylase sirtuin-1Homo sapiens (human)
nucleoplasmNAD-dependent protein deacetylase sirtuin-1Homo 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)
nucleusNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
cytoplasmNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
nucleusNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
nucleoplasmNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
nucleolusNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
nucleolus organizer regionNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
nuclear speckNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
site of double-strand breakNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
chromatinNAD-dependent protein deacetylase sirtuin-7Homo sapiens (human)
nucleoplasmNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
mitochondrionNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
mitochondrial matrixNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
protein-containing complexNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo sapiens (human)
nucleusNAD-dependent protein deacetylase sirtuin-3, mitochondrialHomo 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)
nucleusNuclear receptor corepressor 2Homo sapiens (human)
nucleoplasmNuclear receptor corepressor 2Homo sapiens (human)
membraneNuclear receptor corepressor 2Homo sapiens (human)
nuclear matrixNuclear receptor corepressor 2Homo sapiens (human)
nuclear bodyNuclear receptor corepressor 2Homo sapiens (human)
chromatinNuclear receptor corepressor 2Homo sapiens (human)
transcription repressor complexNuclear receptor corepressor 2Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (464)

Assay IDTitleYearJournalArticle
AID1801572In-vitro HDAC Enzymatic Endpoint Assay from Article 10.1021/acschembio.5b00640: \\An Isochemogenic Set of Inhibitors To Define the Therapeutic Potential of Histone Deacetylases in u00DF-Cell Protection.\\2016ACS chemical biology, Feb-19, Volume: 11, Issue:2
An Isochemogenic Set of Inhibitors To Define the Therapeutic Potential of Histone Deacetylases in β-Cell Protection.
AID1799021pfHDAC-1 Enzyme Assay from Article 10.1021/jm801654y: \\Identification and characterization of small molecule inhibitors of a class I histone deacetylase from Plasmodium falciparum.\\2009Journal of medicinal chemistry, Apr-23, Volume: 52, Issue:8
Identification and characterization of small molecule inhibitors of a class I histone deacetylase from Plasmodium falciparum.
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.
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.
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.
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.
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.
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.
AID1313935Inhibition of C-terminal His-tagged and C-terminal FLAG-tagged full length human recombinant HDAC1 expressed in baculovirus coexpressed in fall armyworm Sf9 cells using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as substrate 2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
AID1313947Inhibition of human recombinant HDAC6 expressed in baculovirus coexpressed in fall armyworm Sf9 cells using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as substrate after 60 mins by fluorescence assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
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.
AID1312927Inhibition of HDAC6 in human MV4-11 cells assessed as upregulation of acetylated alpha-tubulin level after 6 hrs by Western blot method2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1431805Inhibition of human KDAC3 using FITC-labeled p53 acetylated peptide as substrate after 60 mins by fluorescence assay2017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1210340Activity of recombinant human CYP2B6 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
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.
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.
AID1548745Inhibition of recombinant HDAC6 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1548756Inhibition of recombinant human C-terminal GST/His-tagged HDAC3 (1 to 428 residues) co-expressed with human N-terminal GST-tagged NCOR2 (395 to 489 residues) in baculovirus infected Sf9 cells using Boc-Lys(acetyl)-AMC as substrate preincubated for 90 mins2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1591852Inhibition of recombinant human full-length C-terminal FLAG-tagged HDAC1 expressed in baculovirus expression system at 1000 nM using Ac-peptide-AMC as substrate preincubated for 15 mins followed by substrate addition by fluorescence assay relative to cont2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
AID1236478Antitrypanosomal activity against Trypanosoma brucei brucei 427 assessed as inhibition of parasite proliferation measured as ATP levels after 48 hrs by luciferase-based assay in presence of melarsoprol2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
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.
AID1545759Inhibition of HDAC (unknown origin)2019European journal of medicinal chemistry, Dec-01, Volume: 183Indole: A privileged scaffold for the design of anti-cancer agents.
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.
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.
AID1548939Cmax in BALB/c mouse at 50 mg/kg, po via gavage after 24 hrs by Lc-MS/MS analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1399811Solubility in pH 7.4 phosphate buffer solution at 100 uM after 24 hrs by HPLC method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1548741Inhibition of recombinant HDAC4 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1600733Antiproliferative activity against human CAL27 cells after 72 hrs by microplate reader based MTT assay2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1821217Antitumor activity against human MV4-11 cells xenografted in BALB/c mouse assessed as tumor growth inhibition at 4 mg/kg, ip administered for 24 days
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.
AID1399813Pro-apoptotic activity in human HUT78 cells after 18 hrs by caspase-Glo 3/7 assay2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1313942Inhibition full length human recombinant HDAC3 expressed in baculovirus using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as substrate after 60 mins by fluorescence assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
AID1686358Cytotoxicity against human MV4-11 cells assessed as reduction in cell viability incubated for 48 hrs by Cell-titer-blue cell viability assay2016Journal of medicinal chemistry, 11-10, Volume: 59, Issue:21
Development of Allosteric Hydrazide-Containing Class I Histone Deacetylase Inhibitors for Use in Acute Myeloid Leukemia.
AID1548942Oral bioavailability in BALB/c mouse at 50 mg/kg dosed via gavage after 24 hrs by Lc-MS/MS analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1282299Inhibition of HDAC1/2/3 in human HCT116 cells assessed as upregulation of histone H3 acetylation at 10 to 1000 nM after 6 hrs by Western blot analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1775572Inhibition of HDAC in human MM1.S cells assessed as increase in H3K9Ac at > 500 nM after 48 hrs by Western blot analysis2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Design and Synthesis of Novel Epigenetic Inhibitors Targeting Histone Deacetylases, DNA Methyltransferase 1, and Lysine Methyltransferase G9a with
AID1821220Drug uptake in BALB/c mouse xenografted with human AML MV4-11 cells assessed as tumor tissue at 4mg/kg, po measured after 0.5 to 6 hrs by LC-MS/MS
AID1399834Selectivity ratio of Ki for human recombinant SIRT3 to Ki for human recombinant HDAC12018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
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).
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).
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.
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.
AID496806Inhibition of human HDAC62010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1542187Inhibition of HDAC9 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
AID1282244Inhibition of recombinant HDAC10 (unknown origin) using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1559865Antitumor activity against human HT-29 cells xenografted in nude mouse assessed as relative tumor volume at 50 mg/kg, po qd for 21 days relative to control (Rvb = 16.68 +/- 5.10 No_unit)2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
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.
AID1431806Inhibition of human KDAC1 using substrate A after 60 mins by fluorescence assay2017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1210348Activity of recombinant human CYP2A6 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1591856Inhibition of wild-type human N-terminal GST-tagged CDK2/cycA2 expressed in Sf21 insect cells using FAM-labelled substrate preincubated for 10 mins followed by substrate addition and measured after 30 mins by fluorescence assay2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
AID1559803Inhibition of recombinant His6/GST-tagged human HDAC3 expressed in baculovirus infected High5 insect cells using Ac-Lys-Tyr-Lys(epsilon-acetyl)-AMC as substrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
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.
AID1559812Inhibition of recombinant human His6/GST-tagged HDAC8 expressed in baculovirus infected High5 insect cells using Ac-Lys-Tyr-Lys(epsilon-acetyl)-AMC as susbtrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1548728Inhibition of recombinant human C-terminal GST/His-tagged HDAC3 (1 to 428 residues) co-expressed with human N-terminal GST-tagged NCOR2 (395 to 489 residues) in baculovirus infected Sf9 cells using Boc-Lys(acetyl)-AMC as substrate preincubated for 1 hr fo2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1431818Selectivity ratio of IC50 for human KDAC8 to IC50 for human KDAC32017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1399826Inhibition of human recombinant SIRT3 using fluoro-lysine sirtuin 2 deacetylase substrate after 45 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1821190Half life in CD-1 mouse at 50 mg/kg, po measured upto 24 hrs
AID1548764Inhibition of HDAC6 in human MV4-11 cells assessed as tubulin acetylation at 500 nM incubated for 3 hrs followed by compound washout and measured after 30 mins by Western blot analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1559817Inhibition of recombinant human N-terminal GST-tagged SIRT2 expressed in Escherichia coli using Ac-Arg-His-Lys-Lys(Ac)-AMC as susbtrate after 2 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1591857Antiproliferative activity against human A549 cells measured after 72 hrs by CCK8 assay2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
AID1600732Inhibition of HDAC in human A2780 cells using Boc-Lys(epsilon-Ac)-AMC as substrate preincubated for 18 hrs followed by substrate addition and further incubation for 3 hrs by microplate reader based fluorescence assay2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
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.
AID1821246Drug distribution in BALB/c mouse liver measured at 4 mg/kg, po or ip after 2 to 8 hrs after 0.5 hrs
AID1431812Selectivity ratio of IC50 for human KDAC1 to IC50 for human KDAC32017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1399824Inhibition of human recombinant SIRT1 using fluorogenic HDAC substrate after 20 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1598093Cytotoxicity against human HepG2 cells assessed as reduction in cell growth incubated for 48 hrs by CellTiter-Glo assay2019Journal of medicinal chemistry, 05-23, Volume: 62, Issue:10
Identification of 5-Substituted 2-Acylaminothiazoles That Activate Tat-Mediated Transcription in HIV-1 Latency Models.
AID1282300Inhibition of HDAC6 in human HCT116 cells assessed as upregulation of alpha tubulin acetylation at 10 to 1000 nM after 6 hrs by Western blot analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1312916Cytotoxicity against human HBL1 cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1312866Inhibition of N-terminal His6-tagged recombinant full-length human p110delta/untagged recombinant full length human p85alpha expressed in baculovirus infected insect Sf9 cells incubated for 2 hrs by kinase-glo assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1210324Activity of recombinant human CYP3A5 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1845902Reversal of HIV-1 latency infected in human U1 cells assessed as fold increase in p24 expression level at 31.1 nM incubated for 48 hrs by ELISA2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1845914Inhibition of HIV-1 latency in human CD4+ve Th cells infected NL4.3-Luc virus assessed as p24 expression level incubated for 48 hrs by v450 dye based flow cytometry2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID414980Inhibition of Plasmodium falciparum HDAC1 expressed in Drosophila melanogaster S2 cells2009Journal of medicinal chemistry, Apr-23, Volume: 52, Issue:8
Identification and characterization of small molecule inhibitors of a class I histone deacetylase from Plasmodium falciparum.
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).
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.
AID1525777Inhibition of HADC1 (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.
AID1821197Inhibition of human recombinant HDAC5 using Boc-Lys(triflouroacetyI)-AMC substrate incubated for 2 hrs by fluorescence based assay
AID1821202Inhibition of human recombinant HDAC11 using Boc-Lys(triflouroacetyI)-AMC substrate incubated for 2 hrs by fluorescence based assay
AID1282243Inhibition of recombinant HDAC9 (unknown origin) using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1399809Clearance in human hepatocytes assessed per million cells at 1 uM after 5 to 90 mins by LC-MS/MS analysis2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1236470Trypanocidal activity against Trypanosoma brucei brucei assessed as killing of parasite densities of 10'4 to 10'6 cells/ml at 2.8 uM after 16 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).
AID1210350Activity of recombinant human CYP2C8 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1312895Toxicity in NOD/SCID mouse xenografted with human MM1S cells assessed as mortality at 10 mg/kg, ip administered every 2 days for 6 days2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
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.
AID1600735Inhibition of HDAC in human CAL27 cells using Boc-Lys(epsilon-Ac)-AMC as substrate preincubated for 18 hrs followed by substrate addition and further incubation for 3 hrs by microplate reader based fluorescence assay2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1821234Antitumor activity against human MV4-11 cells xenografted in balb/c mouse assessed as increase accumulation of AcHH4 protein level at 4 mg/kg, ip measured after 8 hrs relative to control
AID1775552Inhibition of recombinant HDAC6 (unknown origin)2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Design and Synthesis of Novel Epigenetic Inhibitors Targeting Histone Deacetylases, DNA Methyltransferase 1, and Lysine Methyltransferase G9a with
AID1127840Cytotoxicity against human M14 cells assessed as cell viability after 24 hrs using GF-AFC as substrate by ApoTox-Glo triplex assay2014Journal of medicinal chemistry, Feb-27, Volume: 57, Issue:4
Novel pyrrolidine diketopiperazines selectively inhibit melanoma cells via induction of late-onset apoptosis.
AID1210354Activity of recombinant human CYP2C18 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1399835Selectivity ratio of Ki for human recombinant SIRT7 to Ki for human recombinant HDAC12018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
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.
AID1591858Antiproliferative activity against human HepG2 cells measured after 72 hrs by CCK8 assay2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
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).
AID1312849Cytotoxicity against human MV4-11 cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1775569Antiproliferative activity against human MM1.S cells assessed as inhibition of cell proliferation measured up to 72 hrs by CellTiter96 Aqueous one reagent based assay2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Design and Synthesis of Novel Epigenetic Inhibitors Targeting Histone Deacetylases, DNA Methyltransferase 1, and Lysine Methyltransferase G9a with
AID1313950Inhibition of C-terminal His-tagged human recombinant HDAC9 (604 to 1066 residues) expressed in baculovirus coexpressed in fall armyworm Sf9 cells using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as substrate after 60 mins by2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
AID496801Inhibition of human HDAC12010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
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.
AID1559804Inhibition of recombinant His6/GST-tagged human HDAC6 expressed in baculovirus infected High5 insect cells using Boc-Lys(epsion-acetyl)-AMC as substrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1845899Reversal of HIV-1 latency infected in human U1 cells assessed as fold increase in p24 expression level at 15.6 nM incubated for 48 hrs by ELISA2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1282282Antitumor activity against human Ramos cells xenografted in NOD/SCID mouse assessed as tumor mass change at 10 mg/kg, ip q2d for 6 days2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1821249Drug distribution in BALB/c mouse heart measured at 4 mg/kg, po or ip after 2 to 8 hrs after 0.5 hrs
AID1312919Cytotoxicity against human SUDHL4 cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID618092Toxicity in nude BALB/c mouse xenografted with human HCT116 cells assessed as body weight loss at 50 mg/kg, po QD for 21 days measured on day 122011Journal 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.
AID1821193AUC (0 to infinity) in CD-1 mouse at 50 mg/kg, po measured upto 24 hrs
AID1821221Drug uptake in BALB/c mouse xenografted with human AML MV4-11 cells assessed as tumor tissue at 4 mg/kg ip measured after 0.5 to 6 hrs by LC-MS/MS
AID1313940Inhibition full length human recombinant HDAC2 expressed in baculovirus coexpressed in fall armyworm Sf9 cells using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as substrate after 60 mins by fluorescence assay2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
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.
AID1282230Inhibition of human recombinant HDAC6 using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
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.
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).
AID748113Inhibition of HDAC2 (unknown origin) after 60 mins by fluorescence assay2013Journal of medicinal chemistry, Jun-13, Volume: 56, Issue:11
Discovery of the first histone deacetylase 6/8 dual inhibitors.
AID1846017Anticancer activity against human UW426 cells assessed as reduction in cell viability incubated for 72 hrs by CCK assay2021European journal of medicinal chemistry, Apr-05, Volume: 215Medulloblastoma drugs in development: Current leads, trials and drawbacks.
AID1312868Inhibition of HDAC6 in human A2780S cells assessed as tubulin acetylation incubated for 6 hrs by cytoblot assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1600726Selectivity index, ratio of IC50 for C-terminal His-fusion tagged/N-terminal Strep-2 tagged recombinant human HDAC8 (1 to 377 residues) expressed in insect cells to IC50 for C-terminal GST-tagged recombinant human HDAC2 (1 to 488 residues) expressed in Ba2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1548755Inhibition of recombinant human HDAC1 using Boc-Lys(acetyl)-AMC as substrate preincubated for 10 mins followed by substrate addition and measured after 2 hrs by fluorescence based microplate reader analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
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).
AID1542189Inhibition of HDAC11 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
AID1210356Activity of recombinant human CYP2C19 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
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.
AID1236467Half-life in po dosed human measured under phase 2 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
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.
AID748109Inhibition of HDAC6 (unknown origin) after 60 mins by fluorescence assay2013Journal of medicinal chemistry, Jun-13, Volume: 56, Issue:11
Discovery of the first histone deacetylase 6/8 dual inhibitors.
AID1821238Drug distribution in BALB/c mouse spleen measured at 4 mg/kg, po or ip after 0.5 hrs
AID1882467Inhibition of HDAC8 (unknown origin)2022Journal of medicinal chemistry, 02-24, Volume: 65, Issue:4
Chasing a Breath of Fresh Air in Cystic Fibrosis (CF): Therapeutic Potential of Selective HDAC6 Inhibitors to Tackle Multiple Pathways in CF Pathophysiology.
AID1846016Anticancer activity against human UW228 cells assessed as reduction in cell viability incubated for 72 hrs by CCK assay2021European journal of medicinal chemistry, Apr-05, Volume: 215Medulloblastoma drugs in development: Current leads, trials and drawbacks.
AID1431817Selectivity ratio of IC50 for human KDAC3 to IC50 for human KDAC62017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1559810Inhibition of recombinant N-terminal GST-tagged full length human HDAC5 expressed in baculovirus infected Sf9 cells using Ac-Leu-Gly-Lys(Tfa)-AMC as substrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1312861Inhibition of full length human recombinant HDAC11 expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition measured after 1 hr by fluorescence 2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1210352Activity of recombinant human CYP2C9 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1548759Inhibition of recombinant human C-terminal GST/His-tagged HDAC3 (1 to 428 residues) co-expressed with human N-terminal GST-tagged NCOR2 (395 to 489 residues) in baculovirus infected Sf9 cells using Boc-Lys(acetyl)-AMC as substrate preincubated for 10 mins2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1399821Inhibition of human recombinant HDAC9 using fluorogenic HDAC substrate class 2a after 30 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
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.
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.
AID1559814Inhibition of recombinant human His6/GST-tagged HDAC10 expressed in baculovirus infected High5 insect cells using Ac-Lys-Tyr-Lys(epsilon-acetyl)-AMC as susbtrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1282288Toxicity in Balb/c nude mouse xenografted with human HCT116 cells assessed as change in body weight at 10 mg/kg, ip q2d for 6 days2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1542180Inhibition of HDAC2 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
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.
AID1559809Inhibition of recombinant human His6/GST-tagged HDAC4 expressed in baculovirus infected High5 insect cells using Ac-Leu-Gly-Lys(Tfa)-AMC as substrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
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).
AID1548737Inhibition of recombinant HDAC1 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1591855Inhibition of wild-type human N-terminal GST-tagged CDK2/cycA2 expressed in Sf21 insect cells at 100 nM using FAM-labelled substrate preincubated for 10 mins followed by substrate addition and measured after 30 mins by fluorescence assay relative to contr2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
AID1312914Cytotoxicity against human MM1S cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1559854Antiproliferative activity against human MCF7 cells assessed as reduction in cell growth after 72 hrs2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1399832Selectivity ratio of Ki for human recombinant SIRT1 to Ki for human recombinant HDAC12018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
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.
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).
AID1548274Antiproliferative activity against bortezomib resistant human KM3/BTZ cells incubated for 48 hrs by MTT assay
AID1548757Inhibition of recombinant human C-terminal GST/His-tagged HDAC3 (1 to 428 residues) co-expressed with human N-terminal GST-tagged NCOR2 (395 to 489 residues) in baculovirus infected Sf9 cells using Boc-Lys(acetyl)-AMC as substrate preincubated for 60 mins2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1236459AUC in po dosed human measured under phase 2 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1548744Inhibition of recombinant HDAC9 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1821241Drug distribution in BALB/c mouse gut measured at 4 mg/kg, po or ip after 0.5 hrs after 0.5 hrs
AID1785449Inhibition of HDAC1 (unknown origin) using Ac-peptide-AMC as substrate incubated for 1 hr by fluorescence method
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.
AID1591853Inhibition of recombinant human full-length C-terminal FLAG-tagged HDAC1 expressed in baculovirus expression system using Ac-peptide-AMC as substrate preincubated for 15 mins followed by substrate addition by fluorescence assay2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
AID1542183Inhibition of HDAC5 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
AID1126987Inhibition of HDAC2 in human U937 cells assessed as increase of intracellular acetylated histone H3 level at 1 uM after 24 hrs by Western blot analysis2014Journal of medicinal chemistry, Apr-24, Volume: 57, Issue:8
Discovery of the first N-hydroxycinnamamide-based histone deacetylase 1/3 dual inhibitors with potent oral antitumor activity.
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.
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).
AID1399816Inhibition of human recombinant HDAC3 using fluorogenic HDAC substrate after 10 mins by spectrophotometric method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1312853Inhibition of full length C-terminal His-tagged human recombinant HDAC3/NCOR2 (395 to 489 residues) expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substr2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1821195Oral bioavailability in CD-1 mouse at 50 mg/kg measured upto 24 hrs
AID1312852Inhibition of full length human recombinant HDAC2 expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition measured after 1 hr by fluorescence a2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1431811Selectivity ratio of IC50 for human KDAC3 to IC50 for human KDAC12017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1210332Activity of recombinant human CYP2D6 expressed in supersomes assessed as enzyme-mediated drug metabolism measured as compound remaining at 10 uM after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1542182Inhibition of HDAC4 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
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.
AID1399810Solubility in PG/EtOH (96%) at 30 mg/mL after 24 hrs by UV-spectrometric method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1548933Mutagenecity in Salmonella typhimurium TA100 at 10 uM in presence of S9 liver homogenate after 2 to 3 days by mini-Ames test2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
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.
AID1431804Inhibition of human KDAC6 using FITC-labeled histone H4 acetylated peptide as substrate after 60 mins by fluorescence assay2017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
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.
AID1542179Inhibition of HDAC1 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
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.
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).
AID1821250Drug distribution in BALB/c mouse brain measured at 4 mg/kg, po or ip after 2 to 8 hrs after 0.5 hrs
AID1821247Drug distribution in BALB/c mouse kidney measured at 4 mg/kg, po or ip after 2 to 8 hrs after 0.5 hrs
AID1821235Antitumor activity against human MV4-11 cells xenografted in balb/c mouse assessed as increase in accumulation of AcHH3 protein level at 4 mg/kg, ip measured after 8 hrs relative to control
AID1548738Inhibition of recombinant HDAC2 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1598139Activation of Tat-mediated HIV1 transcription in J-Lat 10.6 cells harboring LTR driven GFP reporter co-expressing CMV driven RFP reporter assessed as maximum LTR activity at 0.1 uM incubated for 48 hr by FACSCalibur flow cytometry relative to control2019Journal of medicinal chemistry, 05-23, Volume: 62, Issue:10
Identification of 5-Substituted 2-Acylaminothiazoles That Activate Tat-Mediated Transcription in HIV-1 Latency Models.
AID1321704Inhibition of recombinant HDAC1 (unknown origin)2016Journal of medicinal chemistry, 10-13, Volume: 59, Issue:19
Design, Synthesis, and Biological Evaluation of First-in-Class Dual Acting Histone Deacetylases (HDACs) and Phosphodiesterase 5 (PDE5) Inhibitors for the Treatment of Alzheimer's Disease.
AID1312888Antitumor activity against human MM1S cells xenografted in NOD/SCID mouse assessed as change in tumor mass at 10 mg/kg, ip administered every 2 days for 6 days measured every 2 days of compound dosing relative to control2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
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.
AID1399827Inhibition of SIRT7 (unknown origin)2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1600734Antiproliferative activity against human A2780 cells after 72 hrs by microplate reader based MTT assay2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1282302Inhibition of HDAC1/2/3 in human MV4-11 cells assessed as upregulation of histone H3 acetylation at 10 to 1000 nM after 6 hrs by Western blot analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1558024Induction of apoptosis in mouse SM1 cells at 100 nM by caspase3/7 reagent based luminescence assay2019Journal of medicinal chemistry, 09-26, Volume: 62, Issue:18
Discovery of a New Isoxazole-3-hydroxamate-Based Histone Deacetylase 6 Inhibitor SS-208 with Antitumor Activity in Syngeneic Melanoma Mouse Models.
AID1282242Inhibition of recombinant HDAC7 (unknown origin) using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID748111Inhibition of HDAC4 (unknown origin) after 60 mins by fluorescence assay2013Journal of medicinal chemistry, Jun-13, Volume: 56, Issue:11
Discovery of the first histone deacetylase 6/8 dual inhibitors.
AID748112Inhibition of HDAC3 (unknown origin) after 60 mins by fluorescence assay2013Journal of medicinal chemistry, Jun-13, Volume: 56, Issue:11
Discovery of the first histone deacetylase 6/8 dual inhibitors.
AID1312859Inhibition of full length human recombinant HDAC6 expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition measured after 1 hr by fluorescence a2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1282238Inhibition of recombinant HDAC2 (unknown origin) using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
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).
AID1542184Inhibition of HDAC6 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
AID1602601Cytotoxicity against mouse KPC monolayer cells assessed as reduction in tumor spheroid intensity after 72 hrs by Alamar Blue assay2019Journal of medicinal chemistry, 03-14, Volume: 62, Issue:5
Identification and Characterization of AES-135, a Hydroxamic Acid-Based HDAC Inhibitor That Prolongs Survival in an Orthotopic Mouse Model of Pancreatic Cancer.
AID748110Inhibition of HDAC5 (unknown origin) after 60 mins by fluorescence assay2013Journal of medicinal chemistry, Jun-13, Volume: 56, Issue:11
Discovery of the first histone deacetylase 6/8 dual inhibitors.
AID1312918Cytotoxicity against human Raji cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1312915Cytotoxicity against human OCI-LY1 cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1312909Toxicity in NOD/SCID mouse xenografted with human MM1S cells assessed as body weight loss at 10 mg/kg, ip administered every 2 days for 6 days measured on day 12 post last dose2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1210326Activity of recombinant human CYP4A11 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1399815Inhibition of human recombinant HDAC2 using fluorogenic HDAC substrate after 15 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1431814Selectivity ratio of IC50 for human KDAC8 to IC50 for human KDAC12017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1127852Induction of apoptosis human M14 cells assessed as caspase activity at 100 uM after 4 hrs using DEVD peptide as substrate by ApoTox-Glo triplex assay2014Journal of medicinal chemistry, Feb-27, Volume: 57, Issue:4
Novel pyrrolidine diketopiperazines selectively inhibit melanoma cells via induction of late-onset apoptosis.
AID1600729Inhibition of N-terminal GST-tagged recombinant human HDAC6 (1 to 1215 residues) expressed in Baculovirus infected insect cells using Boc-Lys(Ac)-AMC as substrate preincubated for 5 mins followed by substrate addition and further incubation for 90 mins me2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1282245Inhibition of recombinant HDAC11 (unknown origin) using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
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).
AID748106Inhibition of HDAC8 (unknown origin) after 60 mins by fluorescence assay2013Journal of medicinal chemistry, Jun-13, Volume: 56, Issue:11
Discovery of the first histone deacetylase 6/8 dual inhibitors.
AID1312869Inhibition of HDAC1/2/3 in human A2780S cells assessed as histone H3 acetylation incubated for 6 hrs by cytoblot assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1210338Inhibition of HDAC6 (unknown origin) expressed in HEK293 cells using [3H]acetylated human histone H4 peptide as substrate by scintillation counting2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
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.
AID1282231Inhibition of human recombinant HDAC8 using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1821216Antitumor activity against human MV4-11 cells xenografted in BALB/c mouse assessed as tumor growth inhibition at 2 to 4 mg/kg, po administered for 24 days
AID1598133Selectivity ratio of EC50 for activation of non-specific gene expression in HEK293- FlpIn-FM cells harboring LTR driven CBR reporter co-expressing CMV driven CBG reporter assessed as CMV driven gene expression to EC50 for activation of Tat-mediated HIV1 t2019Journal of medicinal chemistry, 05-23, Volume: 62, Issue:10
Identification of 5-Substituted 2-Acylaminothiazoles That Activate Tat-Mediated Transcription in HIV-1 Latency Models.
AID1559808Inhibition of recombinant human His6/GST-tagged HDAC2 expressed in baculovirus infected High5 insect cells using Ac-Lys-Tyr-Lys(epsilon-acetyl)-AMC as susbtrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1210346Activity of recombinant human CYP1B1 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1559868Antitumor activity against human HT-29 cells xenografted in nude mouse assessed as reduction in tumor growth at 50 mg/kg, po qd for 21 days relative to control2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1785450Inhibition of HDAC6 (unknown origin) using Ac-peptide-AMC as substrate incubated for 1 hr by fluorescence method
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.
AID1845900Reversal of HIV-1 latency infected in human ACH-2 cells assessed as fold increase in p24 expression level at 15.6 nM incubated for 48 hrs by ELISA2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1559852Antiproliferative activity against human NCI-H1975 cells assessed as reduction in cell growth after 72 hrs2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1882462Inhibition of HDAC6 (unknown origin)2022Journal of medicinal chemistry, 02-24, Volume: 65, Issue:4
Chasing a Breath of Fresh Air in Cystic Fibrosis (CF): Therapeutic Potential of Selective HDAC6 Inhibitors to Tackle Multiple Pathways in CF Pathophysiology.
AID1127841Induction of apoptosis human M14 cells assessed as caspase activity after 24 to 48 hrs using DEVD peptide as substrate by ApoTox-Glo triplex assay2014Journal of medicinal chemistry, Feb-27, Volume: 57, Issue:4
Novel pyrrolidine diketopiperazines selectively inhibit melanoma cells via induction of late-onset apoptosis.
AID1210339Inhibition of his-strep-tagged HDAC8 (unknown origin) expressed in SF9 cells using [3H]acetylated human histone H4 peptide as substrate by scintillation counting2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1210318Activity of recombinant human CYP2E1 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1312906Toxicity in NOD/SCID mouse xenografted with human Ramos cells assessed as body weight loss at 10 mg/kg, ip administered every 2 days for 6 days measured on day 7 post last dose2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
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.
AID708192Growth inhibition of mouse B16 cells incubated for 48 hrs by MTT assay2012Journal of medicinal chemistry, Nov-26, Volume: 55, Issue:22
Selective histone deacetylase 6 inhibitors bearing substituted urea linkers inhibit melanoma cell growth.
AID1821239Drug distribution in BALB/c mouse liver measured at 4 mg/kg, po or ip after 0.5 hrs after 0.5 hrs
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.
AID1210316Activity of recombinant human CYP2D6 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1312912Cytotoxicity against human U266 cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID496809Inhibition of human HDAC92010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1210328Drug metabolism in Escherichia coli JM109 assessed as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
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.
AID1821198Inhibition of human recombinant HDAC7 using Boc-Lys(triflouroacetyI)-AMC substrate incubated for 2 hrs by fluorescence based assay
AID496805Inhibition of human HDAC52010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1882260Inhibition of HDAC6 (unknown origin)2022European journal of medicinal chemistry, Feb-05, Volume: 229A review on the treatment of multiple myeloma with small molecular agents in the past five years.
AID1882456Inhibition of HDAC1 (unknown origin)2022Journal of medicinal chemistry, 02-24, Volume: 65, Issue:4
Chasing a Breath of Fresh Air in Cystic Fibrosis (CF): Therapeutic Potential of Selective HDAC6 Inhibitors to Tackle Multiple Pathways in CF Pathophysiology.
AID1321705Inhibition of recombinant HDAC2 (unknown origin)2016Journal of medicinal chemistry, 10-13, Volume: 59, Issue:19
Design, Synthesis, and Biological Evaluation of First-in-Class Dual Acting Histone Deacetylases (HDACs) and Phosphodiesterase 5 (PDE5) Inhibitors for the Treatment of Alzheimer's Disease.
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).
AID1559862Antitumor activity against human HT-29 cells xenografted in nude mouse assessed as tumor volume at 50 mg/kg, po qd for 21 days measured on day 21 (Rvb = 2854 +/- 940 mm3)2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1312865Inhibition of His-tagged full length recombinant human p110gamma expressed in baculovirus expression system incubated for 1 hr by ADP-gloreagen assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID620532Reactivation of MeCp2 mutant expression in human GM11272 cells at 1 to 100 nM by PCR method2011Bioorganic & medicinal chemistry letters, Sep-15, Volume: 21, Issue:18
Clonal Rett Syndrome cell lines to test compounds for activation of wild-type MeCP2 expression.
AID1282239Inhibition of recombinant HDAC3 (unknown origin) using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1548727Inhibition of recombinant human HDAC2 expressed in baculovirus expression system using Boc-Lys(acetyl)-AMC as substrate preincubated for 1 hr followed by substrate addition and measured after 2 hrs by fluorescence based micro plate reader analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1559859Antitumor activity against azoxymethane-induced colitis-associated cancer C57BL/6 mouse model assessed as tumor incidence at 50 mg/kg, po via gavage administered with AOM followed by 3 cycles of 2.5% DSS given in water for 5 days and compound administered2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1821248Drug distribution in BALB/c mouse gut measured at 4 mg/kg, po or ip after 2 to 8 hrs after 0.5 hrs
AID1548276Antiproliferative activity against bortezomib resistant human KM3/BTZ cells assessed combination index in presence of bortezomib at 1:1 compound to bortezomib ratio incubated for 48 hrs by MTT assay
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.
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.
AID1845901Reversal of HIV-1 latency infected in human ACH-2 cells assessed as fold increase in p24 expression level at 31.1 nM incubated for 48 hrs by ELISA2021European journal of medicinal chemistry, Mar-05, Volume: 213HIV latency reversal agents: A potential path for functional cure?
AID1431822Selectivity ratio of IC50 for human KDAC1 to IC50 for human KDAC62017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1210336Inhibition of HDAC3 (unknown origin) expressed in HEK293 cells using [3H]acetylated human histone H4 peptide as substrate by scintillation counting2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1431815Selectivity ratio of IC50 for human KDAC1 to IC50 for human KDAC82017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1600730Inhibition of C-terminal His-tagged/N-terminal GST-tagged recombinant human HDAC4 (627 to 1084 residues) expressed in Baculovirus infected insect cells using Boc-Lys(TFa)-AMC as substrate preincubated for 5 mins followed by substrate addition and further 2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1559811Inhibition of human N-terminal GST-tagged HDAC7 (518 to end residues) expressed in baculovirus infected Sf9 cells using Ac-Leu-Gly-Lys(Tfa)-AMC as susbtrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1313945Inhibition of N-terminal GST-tagged and C-terminal His-tagged human recombinant HDAC4 (627 to 1084 residues ) expressed in baculovirus coexpressed in fall armyworm Sf9 cells using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
AID1723749Inhibition of human recombinant HDAC4 using Boc-Lys-(epsilon-Tfa)-AMC fluorogenic substrate incubated for 90 mins by fluorescence based assay2020Journal of medicinal chemistry, 09-24, Volume: 63, Issue:18
Multicomponent Synthesis, Binding Mode, and Structure-Activity Relationship of Selective Histone Deacetylase 6 (HDAC6) Inhibitors with Bifurcated Capping Groups.
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).
AID1210322Activity of recombinant human CYP3A4 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis in presence of cytochrome b52012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1424834Induction of apoptosis in human MV4-11 cells at 30 nM after 24 to 48 hrs by Annexin V-PI staining based flow cytometry2018European journal of medicinal chemistry, May-25, Volume: 152Design and biological evaluation of tetrahydro-β-carboline derivatives as highly potent histone deacetylase 6 (HDAC6) inhibitors.
AID414981Antimalarial activity against Plasmodium falciparum 3D7 by [3H]hypoxanthine uptake2009Journal of medicinal chemistry, Apr-23, Volume: 52, Issue:8
Identification and characterization of small molecule inhibitors of a class I histone deacetylase from Plasmodium falciparum.
AID1548762Inhibition of class I HDAC in human MV4-11 cells assessed as reduction in histone H3 levels at 500 nM incubated for 3 hrs followed by compound washout and measured after 1 hr by Western blot analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
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.
AID1559813Inhibition of recombinant human C-terminal His-tagged HDAC9 (604 to 1066 residues) expressed in baculovirus infected Sf9 cells using Ac-Leu-Gly-Lys(Tfa)-AMC as substrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) 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.
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.
AID1598102Activation of Tat-mediated HIV1 transcription in HEK293- FlpIn-FM cells harboring LTR driven CBR reporter co-expressing CMV driven CBG reporter assessed as maximum LTR activity at 0.2 uM incubated for 48 hr using Chroma-Glo substrate by luciferase dual re2019Journal of medicinal chemistry, 05-23, Volume: 62, Issue:10
Identification of 5-Substituted 2-Acylaminothiazoles That Activate Tat-Mediated Transcription in HIV-1 Latency Models.
AID1312854Inhibition of full length C-terminal His-tagged human recombinant HDAC8 expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition measured after 2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
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).
AID1559856Antiproliferative activity against human T47D cells assessed as reduction in cell growth after 72 hrs2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1399829Selectivity ratio of Ki for human recombinant HDAC4 to Ki for human recombinant HDAC12018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1821222Plasma concentration in BALB/c mouse xenografted with human AML MV4-11 cells at 4 mg/kg, po
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.
AID1600727Selectivity index, ratio of IC50 for C-terminal His-tagged/N-terminal GST-tagged recombinant human HDAC4 (627 to 1084 residues) expressed in Baculovirus infected insect cells to IC50 for C-terminal GST-tagged recombinant human HDAC2 (1 to 488 residues) ex2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1321707Inhibition of recombinant HDAC6 (unknown origin)2016Journal of medicinal chemistry, 10-13, Volume: 59, Issue:19
Design, Synthesis, and Biological Evaluation of First-in-Class Dual Acting Histone Deacetylases (HDACs) and Phosphodiesterase 5 (PDE5) Inhibitors for the Treatment of Alzheimer's Disease.
AID1399818Inhibition of human recombinant HDAC4 using fluorogenic HDAC substrate class 2a after 30 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1313946Inhibition of N-terminal GST-tagged full length human recombinant HDAC5 expressed in baculovirus coexpressed in fall armyworm Sf9 cells using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as substrate after 60 mins by fluorescen2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
AID1775549Inhibition of recombinant HDAC1 (unknown origin)2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Design and Synthesis of Novel Epigenetic Inhibitors Targeting Histone Deacetylases, DNA Methyltransferase 1, and Lysine Methyltransferase G9a with
AID1542181Inhibition of HDAC3 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
AID1548726Inhibition of recombinant HDAC1 (unknown origin) using Boc-Lys(acetyl)-AMC as substrate preincubated for 1 hr followed by substrate addition and measured after 2 hrs by fluorescence based micro plate reader analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1548890Induction of apoptosis in human MV4-11 cells harboring wild type p53/FLT3-ITD mutant assessed as cleavage of pro-caspase 3 at 100 nM after 24 hrs by Western blot analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1600728Inhibition of C-terminal His-fusion tagged/N-terminal Strep-2 tagged recombinant human HDAC8 (1 to 377 residues) expressed in insect cells using Boc-Lys(TFa)-AMC as substrate preincubated for 5 mins followed by substrate addition and further incubation fo2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1210330Activity of recombinant human CYP3A4 expressed in supersomes assessed as enzyme-mediated drug metabolism measured as compound remaining at 10 uM after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1816688Inhibition of HDAC5 (unknown origin) using Ac-peptide as substrate preincubated for 15 mins followed by substrate addition by fluorescence assay2021European journal of medicinal chemistry, Aug-05, Volume: 220Design, synthesis, and biological evaluation of novel dual inhibitors targeting lysine specific demethylase 1 (LSD1) and histone deacetylases (HDAC) for treatment of gastric cancer.
AID1821187Cmax in CD-1 mouse at 50 mg/kg, po measured upto 24 hrs
AID1821243Drug distribution in BALB/c mouse brain measured at 4 mg/kg, po or ip after 0.5 hrs after 0.5 hrs
AID1821196Inhibition of human recombinant HDAC4 using Boc-Lys(triflouroacetyI)-AMC substrate incubated for 2 hrs by fluorescence based assay
AID1548941AUC (0 to infinity) in BALB/c mouse at 50 mg/kg, po via gavage after 24 hrs by Lc-MS/MS analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
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.
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.
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.
AID1821240Drug distribution in BALB/c mouse kidney measured at 4 mg/kg, po or ip after 0.5 hrs after 0.5 hrs
AID1559855Antiproliferative activity against human MDA-MB-231 cells assessed as reduction in cell growth after 72 hrs2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
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.
AID1312881Antitumor activity against human Ramos cells xenografted in NOD/SCID mouse assessed as change in tumor mass at 10 mg/kg, ip administered every 2 days for 6 days measured every 2 days of compound dosing relative to control2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1446915Inhibition of Schistosoma mansoni KDAC8 using (FAM)-labeled peptide as substrate after 60 mins by microfluidic assay2017Bioorganic & medicinal chemistry, 04-01, Volume: 25, Issue:7
Structural insights of SmKDAC8 inhibitors: Targeting Schistosoma epigenetics through a combined structure-based 3D QSAR, in vitro and synthesis strategy.
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.
AID1821201Inhibition of human recombinant HDAC10 using Boc-Lys(triflouroacetyI)-AMC substrate incubated for 2 hrs by fluorescence based assay
AID1821218Antitumor activity against human MV4-11 cells xenografted in BALB/c mouse assessed as tumor growth inhibition at 4 mg/kg, po administered for 24 days
AID1312850Cytotoxicity against human A2780S cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1431803Inhibition of human KDAC8 after 60 mins by fluorescence assay2017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1548940Half life in BALB/c mouse at 50 mg/kg, po via gavage after 24 hrs by Lc-MS/MS analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1126989Inhibition of HDAC6 in human U937 cells assessed as increase of intracellular acetylated alpha-tubulin level at 1 uM after 24 hrs by Western blot analysis2014Journal of medicinal chemistry, Apr-24, Volume: 57, Issue:8
Discovery of the first N-hydroxycinnamamide-based histone deacetylase 1/3 dual inhibitors with potent oral antitumor activity.
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.
AID1399817Inhibition of human recombinant HDAC8 using fluorogenic HDAC substrate after 15 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1548758Inhibition of recombinant human C-terminal GST/His-tagged HDAC3 (1 to 428 residues) co-expressed with human N-terminal GST-tagged NCOR2 (395 to 489 residues) in baculovirus infected Sf9 cells using Boc-Lys(acetyl)-AMC as substrate preincubated for 30 mins2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1548915Induction of apoptosis in human RS4:11 cells harboring wild type p53/FLT3 assessed as cleavage of pro-caspase 3 at 100 nM after 24 hrs by Western blot analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
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).
AID1312856Inhibition of human recombinant HDAC5 expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition measured after 1 hr by fluorescence assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1548742Inhibition of recombinant HDAC5 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1313948Inhibition of N-terminal GST-tagged human recombinant HDAC7 (518 to end residues) expressed in baculovirus coexpressed in fall armyworm Sf9 cells using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as substrate after 60 mins by 2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
AID1312860Inhibition of human recombinant HDAC10 expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition measured after 1 hr by fluorescence assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1548753Inhibition of recombinant human HDAC1 using Boc-Lys(acetyl)-AMC as substrate preincubated for 60 mins followed by substrate addition and measured after 2 hrs by fluorescence based microplate reader analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
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.
AID1399812Inhibition of human recombinant HDAC1 using Fluor de Lys as substrate after 2 hrs by fluorescence method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1548740Inhibition of recombinant HDAC8 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1821200Inhibition of human recombinant HDAC9 using Boc-Lys(triflouroacetyI)-AMC substrate incubated for 2 hrs by fluorescence based assay
AID1821260Drug distribution in tumor of BALB/c mouse xenografted with human MV4-11 cells assessed as measured after 8 hrs
AID1210342Activity of recombinant human CYP1A1 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
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.
AID1399833Selectivity ratio of Ki for human recombinant SIRT2 to Ki for human recombinant HDAC12018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID618329Antitumor activity against human HCT116 cells xenografted in nude BALB/c mouse assessed as tumor growth inhibition at 50 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.
AID1312847Inhibition of full length C-terminal His/FLAG-tagged human recombinant HDAC1 expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition measured a2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1312858Inhibition of C-terminal His-tagged human recombinant HDAC9 (604 to 1066 residues) expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition meas2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1312923Inhibition of HDAC1/2/3 in human MV4-11 cells assessed as upregulation of acetylated histone H3 level after 6 hrs by Western blot method2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1559857Antiproliferative activity against human NCI-N87 cells assessed as reduction in cell growth after 72 hrs2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1548729Inhibition of recombinant HDAC6 (unknown origin) using Boc-Lys(acetyl)-AMC as substrate preincubated for 1 hr followed by substrate addition and measured after 2 hrs by fluorescence based micro plate reader analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1548743Inhibition of recombinant HDAC7 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1775571Permeability of the compound in PBS/EtOH buffer (70:30) incubated for 18 hrs by UV plate reader based PAMPA assay2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Design and Synthesis of Novel Epigenetic Inhibitors Targeting Histone Deacetylases, DNA Methyltransferase 1, and Lysine Methyltransferase G9a with
AID1431816Selectivity ratio of IC50 for human KDAC6 to IC50 for human KDAC32017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1126986Inhibition of HDAC1 in human U937 cells assessed as increase of intracellular acetylated histone H3 level at 1 uM after 24 hrs by Western blot analysis2014Journal of medicinal chemistry, Apr-24, Volume: 57, Issue:8
Discovery of the first N-hydroxycinnamamide-based histone deacetylase 1/3 dual inhibitors with potent oral antitumor activity.
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.
AID1542188Inhibition of HDAC10 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
AID1399819Inhibition of human recombinant HDAC5 using fluorogenic HDAC substrate class 2a after 30 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
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.
AID1282240Inhibition of recombinant HDAC4 (unknown origin) using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
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.
AID1312928Inhibition of HDAC6 in human HCT116 cells assessed as upregulation of acetylated alpha-tubulin level after 6 hrs by Western blot method2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1559816Inhibition of recombinant human N-terminal GST-tagged SIRT1 expressed in Escherichia coli using Ac-Arg-His-Lys-Lys(Ac)-AMC as susbtrate after 1 hr by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1431821Selectivity ratio of IC50 for human KDAC6 to IC50 for human KDAC82017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase 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.
AID1548763Inhibition of class I HDAC in human MV4-11 cells assessed as reduction in histone H3 levels at 500 nM incubated for 3 hrs followed by compound washout and measured after 6 hr by Western blot analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
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.
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.
AID1399814Inhibition of human recombinant HDAC1 using fluorogenic HDAC substrate after 15 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1591854Inhibition of wild-type human N-terminal GST-tagged CDK2/cycA2 expressed in Sf21 insect cells at 10 nM using FAM-labelled substrate preincubated for 10 mins followed by substrate addition and measured after 30 mins by fluorescence assay relative to contro2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
AID1775551Inhibition of recombinant HDAC3 (unknown origin)2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Design and Synthesis of Novel Epigenetic Inhibitors Targeting Histone Deacetylases, DNA Methyltransferase 1, and Lysine Methyltransferase G9a with
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.
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.
AID1431819Selectivity ratio of IC50 for human KDAC3 to IC50 for human KDAC82017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1775570Cytotoxicity against human THLE-2 cells assessed as decrease in cell viability measured after 72 hrs by Vialight-plus cell proliferation assay2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Design and Synthesis of Novel Epigenetic Inhibitors Targeting Histone Deacetylases, DNA Methyltransferase 1, and Lysine Methyltransferase G9a with
AID1399830Selectivity ratio of Ki for human recombinant HDAC7 to Ki for human recombinant HDAC12018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1821223Plasma concentration in BALB/c mouse xenografted with human AML MV4-11 cells at 4 mg/kg, ip
AID1127853Cytotoxicity against human M14 cells assessed as cell viability at 100 uM after 4 hrs using GF-AFC as substrate by ApoTox-Glo triplex assay2014Journal of medicinal chemistry, Feb-27, Volume: 57, Issue:4
Novel pyrrolidine diketopiperazines selectively inhibit melanoma cells via induction of late-onset apoptosis.
AID496803Inhibition of human HDAC32010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1821244Drug distribution in BALB/c mouse lung measured at 4 mg/kg, po or ip after 0.5 hrs after 0.5 hrs
AID1600731Inhibition of C-terminal GST-tagged recombinant human HDAC2 (1 to 488 residues) expressed in Baculovirus infected insect cells using Boc-Lys(Ac)-AMC as substrate preincubated for 5 mins followed by substrate addition and further incubation for 90 mins mea2019Bioorganic & medicinal chemistry, 10-01, Volume: 27, Issue:19
Novel α,β-unsaturated hydroxamic acid derivatives overcome cisplatin resistance.
AID1558023Cytotoxicity against mouse SM1 cells at 100 nM by ApoTox-Glo triplex assay2019Journal of medicinal chemistry, 09-26, Volume: 62, Issue:18
Discovery of a New Isoxazole-3-hydroxamate-Based Histone Deacetylase 6 Inhibitor SS-208 with Antitumor Activity in Syngeneic Melanoma Mouse Models.
AID1591851Inhibition of recombinant human full-length C-terminal FLAG-tagged HDAC1 expressed in baculovirus expression system at 10 nM using Ac-peptide-AMC as substrate preincubated for 15 mins followed by substrate addition by fluorescence assay relative to contro2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
AID1525779Inhibition of HADC3 (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.
AID1821259Drug distribution in tumor of BALB/c mouse xenografted with human MV4-11 cells assessed as measured after 2 hrs
AID1312917Cytotoxicity against human Ramos cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1559815Inhibition of recombinant human His6/GST-tagged HDAC11 expressed in baculovirus infected High5 insect cells using Ac-Lys-Tyr-Lys(epsilon-acetyl)-AMC as susbtrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1559905Toxicity in nude mouse xenografted with human HT-29 cells assessed as body weight at 50 mg/kg, po qd for 21 days measured on day 21 (Rvb = 19.1 gms)2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1282229Inhibition of human recombinant HDAC1 using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1127851Cytotoxicity against human M14 cells assessed as cell viability at 100 uM after 4 hrs using bis-AAF-R110 as substrate by ApoTox-Glo triplex assay2014Journal of medicinal chemistry, Feb-27, Volume: 57, Issue:4
Novel pyrrolidine diketopiperazines selectively inhibit melanoma cells via induction of late-onset apoptosis.
AID1236455Cmax in po dosed human measured under phase 2 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1598094Cytotoxicity against HEK293 cells assessed as reduction in cell growth incubated for 48 hrs by MTS assay2019Journal of medicinal chemistry, 05-23, Volume: 62, Issue:10
Identification of 5-Substituted 2-Acylaminothiazoles That Activate Tat-Mediated Transcription in HIV-1 Latency Models.
AID748114Inhibition of HDAC1 (unknown origin) after 60 mins by fluorescence assay2013Journal of medicinal chemistry, Jun-13, Volume: 56, Issue:11
Discovery of the first histone deacetylase 6/8 dual inhibitors.
AID1399823Inhibition of human recombinant HDAC10 using fluorogenic HDAC substrate after 45 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
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.
AID1431820Selectivity ratio of IC50 for human KDAC8 to IC50 for human KDAC62017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
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.
AID1479839Cell cycle arrest in human MV4-11 cells assessed as accumulation at sub-G1 phase at 30 to 50 nM after 24 hrs by propidium iodide staining-based flow cytometric method2018Journal of medicinal chemistry, 04-26, Volume: 61, Issue:8
Marbostat-100 Defines a New Class of Potent and Selective Antiinflammatory and Antirheumatic Histone Deacetylase 6 Inhibitors.
AID1321706Inhibition of recombinant HDAC3 (unknown origin)2016Journal of medicinal chemistry, 10-13, Volume: 59, Issue:19
Design, Synthesis, and Biological Evaluation of First-in-Class Dual Acting Histone Deacetylases (HDACs) and Phosphodiesterase 5 (PDE5) Inhibitors for the Treatment of Alzheimer's Disease.
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.
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.
AID1282241Inhibition of recombinant HDAC5 (unknown origin) using AMC labeled AC-peptide as substrate incubated for 1 hr by fluorescence analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1846018Anticancer activity against human MED-MEB-8A cells assessed as reduction in cell viability incubated for 72 hrs by CCK assay2021European journal of medicinal chemistry, Apr-05, Volume: 215Medulloblastoma drugs in development: Current leads, trials and drawbacks.
AID1821258Drug distribution in tumor of BALB/c mouse xenografted with human MV4-11 cells assessed as measured after 0.5 hrs
AID1559853Antiproliferative activity against human EBC1 cells assessed as reduction in cell growth after 72 hrs2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1598104Activation of non-specific gene expression in human J-Lat 10.6 cells harboring LTR driven GFP reporter co-expressing CMV driven RFP reporter assessed as CMV driven gene expression incubated for 48 hr by FACSCalibur flow cytometry2019Journal of medicinal chemistry, 05-23, Volume: 62, Issue:10
Identification of 5-Substituted 2-Acylaminothiazoles That Activate Tat-Mediated Transcription in HIV-1 Latency Models.
AID1559851Antiproliferative activity against human Bel7402 cells assessed as reduction in cell growth after 72 hrs2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1548275Antiproliferative activity against bortezomib resistant human KM3/BTZ cells in presence of bortezomib incubated for 48 hrs by MTT assay
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.
AID1399822Inhibition of human recombinant HDAC6 using fluorogenic HDAC substrate after 30 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1559858Antiproliferative activity against human HT-29 cells assessed as reduction in cell growth after 72 hrs2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1686371Cytotoxicity against human PBMC assessed as reduction in cell viability at 10 nM incubated for 48 hrs by Cell-titer-blue cell viability assay2016Journal of medicinal chemistry, 11-10, Volume: 59, Issue:21
Development of Allosteric Hydrazide-Containing Class I Histone Deacetylase Inhibitors for Use in Acute Myeloid Leukemia.
AID1312913Cytotoxicity against human RPMI8226 cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1126988Inhibition of HDAC3 in human U937 cells assessed as increase of intracellular acetylated histone H4 level at 1 uM after 24 hrs by Western blot analysis2014Journal of medicinal chemistry, Apr-24, Volume: 57, Issue:8
Discovery of the first N-hydroxycinnamamide-based histone deacetylase 1/3 dual inhibitors with potent oral antitumor activity.
AID496808Activity of human HDAC82010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID618098Toxicity in nude BALB/c 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.
AID1542185Inhibition of HDAC7 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
AID1210335Inhibition of flag-tagged HDAC2 (unknown origin) expressed in SF21 cells using [3H]acetylated human histone H4 peptide as substrate by scintillation counting2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1821199Inhibition of human recombinant HDAC8 using Boc-Lys(triflouroacetyI)-AMC substrate incubated for 2 hrs by fluorescence based assay
AID1559802Inhibition of recombinant His6/GST-tagged human HDAC1 expressed in baculovirus infected High5 insect cells using Ac-Lys-Tyr-Lys(epsilon-acetyl)-AMC as substrate after 24 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1548739Inhibition of recombinant HDAC3 (unknown origin)2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1548752Inhibition of recombinant human HDAC1 using Boc-Lys(acetyl)-AMC as substrate preincubated for 90 mins followed by substrate addition and measured after 2 hrs by fluorescence based microplate reader analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID496807Inhibition of human HDAC72010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1479834Inhibition of HDAC6 in human MV4-11 cells assessed as induction of alpha-tubulin hyperacetylation at 50 nM after 2 to 24 hrs by Western blot method2018Journal of medicinal chemistry, 04-26, Volume: 61, Issue:8
Marbostat-100 Defines a New Class of Potent and Selective Antiinflammatory and Antirheumatic Histone Deacetylase 6 Inhibitors.
AID1399831Selectivity ratio of Ki for human recombinant HDAC9 to Ki for human recombinant HDAC12018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1882466Inhibition of HDAC3 (unknown origin)2022Journal of medicinal chemistry, 02-24, Volume: 65, Issue:4
Chasing a Breath of Fresh Air in Cystic Fibrosis (CF): Therapeutic Potential of Selective HDAC6 Inhibitors to Tackle Multiple Pathways in CF Pathophysiology.
AID1210334Inhibition of HDAC1 (unknown origin) expressed in HEK293 cells using [3H]acetylated human histone H4 peptide as substrate by scintillation counting2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1548754Inhibition of recombinant human HDAC1 using Boc-Lys(acetyl)-AMC as substrate preincubated for 30 mins followed by substrate addition and measured after 2 hrs by fluorescence based microplate reader analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1763569Cytotoxicity against human PANC1 cells assessed as reduction in cell viability incubated upto 72 hrs by MTT assay2021Bioorganic & medicinal chemistry letters, 07-01, Volume: 43Design and evaluation of 1,2,3-dithiazoles and fused 1,2,4-dithiazines as anti-cancer agents.
AID1399820Inhibition of human recombinant HDAC7 using fluorogenic HDAC substrate class 2a after 45 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1548730Antiproliferative activity against human MV4-11 cells assessed as reduction in cell viability after 48 hrs by CellTiter-Blue dye based spectrophotometric analysis2020Journal of medicinal chemistry, 05-28, Volume: 63, Issue:10
Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity.
AID1868471Inhibition of HIV-1 latency reversal in GFP-fused human J-lat 9.2 cells at 0.1 uM incubated for 24 hrs in presence of pan-PKC inhibitor Go 6983 by flow cytometry analysis2022Journal of natural products, 05-27, Volume: 85, Issue:5
Ansellone J, a Potent
AID1236477Antitrypanosomal activity against Trypanosoma brucei brucei 427 assessed as inhibition of parasite proliferation measured as ATP levels after 48 hrs by luciferase-based assay in presence of nifurtimox2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1559818Inhibition of recombinant human N-terminal His-tagged SIRT3 expressed in Escherichia coli using Ac-Arg-His-Lys-Lys(Ac)-AMC as susbtrate after 2 hrs by fluorescence based assay2020Journal of medicinal chemistry, 01-23, Volume: 63, Issue:2
Synthesis and in Vitro and in Vivo Biological Evaluation of Tissue-Specific Bisthiazole Histone Deacetylase (HDAC) Inhibitors.
AID1598103Activation of Tat-mediated HIV1 transcription in J-Lat 10.6 cells harboring LTR driven GFP reporter co-expressing CMV driven RFP reporter assessed as LTR-driven gene expression incubated for 48 hr by FACSCalibur flow cytometry2019Journal of medicinal chemistry, 05-23, Volume: 62, Issue:10
Identification of 5-Substituted 2-Acylaminothiazoles That Activate Tat-Mediated Transcription in HIV-1 Latency Models.
AID1525780Inhibition of HADC8 (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.
AID1312864Inhibition of recombinant human p110beta expressed in baculovirus infected insect Sf9 cells incubated for 1 hr by ADP-gloreagen assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1210320Activity of recombinant human CYP3A4 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
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.
AID1821242Drug distribution in BALB/c mouse heart measured at 4 mg/kg, po or ip after 0.5 hrs after 0.5 hrs
AID1210337Inhibition of HDAC4 (unknown origin) expressed in HEK293 cells using [3H]acetylated human histone H4 peptide as substrate by scintillation counting2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
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.
AID1821245Drug distribution in BALB/c mouse spleen measured at 4 mg/kg, po or ip after 2 to 8 hrs after 0.5 hrs
AID1431813Selectivity ratio of IC50 for human KDAC6 to IC50 for human KDAC12017European journal of medicinal chemistry, Feb-15, Volume: 127Design and synthesis of benzodiazepine analogs as isoform-selective human lysine deacetylase inhibitors.
AID1479836Induction of HDAC6 degradation in human MV4-11 cells at 50 nM after 24 hrs by Western blot method2018Journal of medicinal chemistry, 04-26, Volume: 61, Issue:8
Marbostat-100 Defines a New Class of Potent and Selective Antiinflammatory and Antirheumatic Histone Deacetylase 6 Inhibitors.
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.
AID1312857Inhibition of N-terminal GST-tagged human recombinant HDAC7 (518 to end residues) expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrate addition measu2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
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.
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).
AID1312851Cytotoxicity against human HCT116 cells assessed as growth inhibition after 24 hrs by MTT assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1591859Antiproliferative activity against human CAL148 cells measured after 72 hrs by CCK8 assay2019Bioorganic & medicinal chemistry letters, 08-15, Volume: 29, Issue:16
Discovery of novel 9H-purin derivatives as dual inhibitors of HDAC1 and CDK2.
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.
AID1775550Inhibition of recombinant HDAC2 (unknown origin)2021Journal of medicinal chemistry, 03-25, Volume: 64, Issue:6
Design and Synthesis of Novel Epigenetic Inhibitors Targeting Histone Deacetylases, DNA Methyltransferase 1, and Lysine Methyltransferase G9a with
AID496804Inhibition of human HDAC42010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1312924Inhibition of HDAC1/2/3 in human HCT116 cells assessed as upregulation of acetylated histone H3 level after 6 hrs by Western blot method2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
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.
AID1236476Antitrypanosomal activity against Trypanosoma brucei brucei 427 assessed as inhibition of parasite proliferation measured as ATP levels after 48 hrs by luciferase-based assay in presence of suramin2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1312867Inhibition of HDAC in human HeLa cell nuclear extract using Ac-Leu-Gly-Lys (Ac)-AMC as substrate after 30 mins by fluorescence assay2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1399828Inhibition of human recombinant HDAC11 using fluorogenic HDAC substrate class 2a after 30 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1236475Antitrypanosomal activity against Trypanosoma brucei brucei 427 assessed as inhibition of parasite proliferation measured as ATP levels after 48 hrs by luciferase-based assay in presence of pentamidine2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
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.
AID1542186Inhibition of HDAC8 (unknown origin)2019Journal of medicinal chemistry, 04-11, Volume: 62, Issue:7
Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy.
AID1313949Inhibition of C-terminal His-tagged full length human recombinant HDAC8 expressed in baculovirus coexpressed in fall armyworm Sf9 cells using carboxyfluorescein (FAM)-labeled acetylated/ trifluoroacetylated peptide as substrate after 60 mins by fluorescen2016Bioorganic & medicinal chemistry, 09-15, Volume: 24, Issue:18
Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.
AID496802Inhibition of human HDAC22010Nature chemical biology, Mar, Volume: 6, Issue:3
Chemical phylogenetics of histone deacetylases.
AID1312855Inhibition of N-terminal GST/C-terminal His-tagged human recombinant HDAC4 (627 to 1084 residues) expressed in baculovirus infected insect Sf9 cells using Ac-peptide-AMC as substrate assessed as release of AMC preincubated for 15 mins followed by substrat2016Journal of medicinal chemistry, 06-09, Volume: 59, Issue:11
Development of Purine-Based Hydroxamic Acid Derivatives: Potent Histone Deacetylase Inhibitors with Marked in Vitro and in Vivo Antitumor Activities.
AID1282298Inhibition of HDAC6 in human MV4-11 cells assessed as upregulation of alpha tubulin acetylation at 10 to 1000 nM after 6 hrs by Western blot analysis2016Journal of medicinal chemistry, Feb-25, Volume: 59, Issue:4
Discovery of Selective Histone Deacetylase 6 Inhibitors Using the Quinazoline as the Cap for the Treatment of Cancer.
AID1210344Activity of recombinant human CYP1A2 expressed in Escherichia coli JM109 co-expressing P450 reductase assessed as enzyme-mediated drug metabolism measured as compound remaining after 1 hr by LC-MS analysis2012Drug metabolism and disposition: the biological fate of chemicals, May, Volume: 40, Issue:5
Biocatalytic synthesis and structure elucidation of cyclized metabolites of the deacetylase inhibitor panobinostat (LBH589).
AID1236463Tmax in po dosed human measured under phase 2 trial2015Bioorganic & medicinal chemistry, Aug-15, Volume: 23, Issue:16
Evaluation of histone deacetylase inhibitors (HDACi) as therapeutic leads for human African trypanosomiasis (HAT).
AID1399825Inhibition of human recombinant SIRT2 using fluoro-lysine sirtuin 2 deacetylase substrate after 60 mins by fluorimetrc method2018Bioorganic & medicinal chemistry letters, 09-15, Volume: 28, Issue:17
Squaramides as novel class I and IIB histone deacetylase inhibitors for topical treatment of cutaneous t-cell lymphoma.
AID1821251Drug distribution in BALB/c mouse lung measured at 4 mg/kg, po or ip after 2 to 8 hrs after 0.5 hrs
AID618261Antitumor activity against human HCT116 cells xenografted in nude BALB/c mouse assessed as tumor growth inhibition at 50 mg/kg, po qd for 21 days measured on day 182011Journal 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.
AID1347415qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: tertiary screen by RT-qPCR2020ACS 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.
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.
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.
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.
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))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.
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.
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.
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.
AID1347412qHTS assay to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: Counter screen cell viability and HiBit confirmation2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
AID1347411qHTS to identify inhibitors of the type 1 interferon - major histocompatibility complex class I in skeletal muscle: primary screen against the NCATS Mechanism Interrogation Plate v5.0 (MIPE) Libary2020ACS chemical biology, 07-17, Volume: 15, Issue:7
High-Throughput Screening to Identify Inhibitors of the Type I Interferon-Major Histocompatibility Complex Class I Pathway in Skeletal Muscle.
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.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (674)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's58 (8.61)29.6817
2010's477 (70.77)24.3611
2020's139 (20.62)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 48.38

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 Index48.38 (24.57)
Research Supply Index6.65 (2.92)
Research Growth Index5.39 (4.65)
Search Engine Demand Index77.88 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (48.38)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials95 (14.01%)5.53%
Reviews72 (10.62%)6.00%
Case Studies10 (1.47%)4.05%
Observational0 (0.00%)0.25%
Other501 (73.89%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (147)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
Phase I Study of the Combination of Bevacizumab, Everolimus and LBH589 (BEL) for the Treatment of Advanced Solid Tumors [NCT01055795]Phase 114 participants (Actual)Interventional2010-03-31Completed
A Phase Ib, Open-label, Multi-center Dose-finding Study of Oral Panobinostat (LBH589) in Combination With Ara-C and Mitoxantrone as Salvage Therapy for Refractory or Relapsed Acute Myeloid Leukemia [NCT01055483]Phase 159 participants (Actual)Interventional2009-09-30Completed
A Phase IB Study of the Histone Deacetylase Inhibitor Panobinostat (LBH589) Given Orally in Combination With Carboplatin and Paclitaxel in Patients With Advanced Solid Tumors [NCT01159418]Phase 136 participants (Anticipated)Interventional2008-06-30Active, not recruiting
A Pilot/Phase I Study of Panobinostat (LBH589) in Patients With Metastatic Melanoma [NCT01065467]Phase 116 participants (Actual)Interventional2010-02-28Completed
A Phase I Dose Finding Study Of Panobinostat In Children With Refractory Hematologic Malignancies [NCT01321346]Phase 130 participants (Actual)Interventional2011-03-31Completed
A Phase Ib, Dose-finding Study of Oral Panobinostat (LBH589) in Combination With Idarubicin and Cytarabine Induction and High-dose Cytarabine-based Consolidation Therapy in Adult Patients Less Than or Equal to 65 Years Old With Acute Myeloid Leukemia (AML [NCT01242774]Phase 146 participants (Actual)Interventional2010-10-31Completed
A Phase I/II Open Label Study of LBH589, a Novel Histone Deacetylase Inhibitor (HDACi), in Patients With Primary Myelofibrosis (PMF) and Post-polycythemia/Essential Thrombocythemia Myelofibrosis (Post-PV/ET MF) [NCT01298934]Phase 1/Phase 222 participants (Actual)Interventional2009-09-30Active, not recruiting
A Phase 1 Study of HDAC Inhibitor Panobinostat (LBH 589) Administered in Combination With Ipilimumab in Subjects With Unresectable Stage III or Stage IV Melanoma [NCT02032810]Phase 117 participants (Actual)Interventional2014-01-07Completed
Efficacy and Safety Assessment of Oral LBH589 in Adult Patients With Advanced Soft Tissue Sarcoma After Pre-treatment Failure: an Open-label, Multicenter Phase II Study [NCT01136499]Phase 253 participants (Actual)Interventional2010-01-31Completed
Phase Ib Study to Assess Safety and Tolerability of PDR001 in Combination With Panobinostat in Metastatic Melanoma and Non-small Cell Lung Cancer After Failure of Prior Anti PD1 or PD-L1 Therapy [NCT03982134]Phase 10 participants (Actual)Interventional2019-09-30Withdrawn(stopped due to Sponsor decision to withdraw)
Multicenter Phase I Study of Imatinib, a Platelet-derived Growth Factor Receptor Inhibitor, and LBH589, a Histone Deacetylase Inhibitor, in the Treatment of Newly Diagnosed and Recurrent Chordoma [NCT01175109]Phase 136 participants (Anticipated)Interventional2011-10-31Active, not recruiting
Evaluation of Panobinostat (LBH589) as Maintenance Therapy in Multiple Myeloma Following Autologous Hematopoietic Cell Transplantation [NCT02722941]Phase 230 participants (Actual)Interventional2016-06-10Completed
Phase I/II Study of the Combination of Panobinostat and Carfilzomib in Patients With Relapsed/Refractory Multiple Myeloma [NCT01496118]Phase 1/Phase 280 participants (Actual)Interventional2011-12-31Completed
A Phase II, Multicentre Study of Oral LBH589 in Patients With Chronic Phase Chronic Myeloid Leukemia With Resistant Disease Following Treatment With at Least Two Fusion Gene of the BCR and ABL Genes (BCR-ABL) Tyrosine Kinase Inhibitors [NCT00451035]Phase 229 participants (Actual)Interventional2007-02-19Terminated
CLBH589DUS108T: Panobinostat With Carfilzomib and Dexamethasone for Relapsed/Refractory Multiple Myeloma: Correlation With In Vitro Chemosensitivity Testing [NCT03256045]Phase 29 participants (Actual)Interventional2018-02-08Terminated(stopped due to Terminated due to loss of funding and former PI left the institution)
A Phase 2 Study of Panobinostat in Combination With Everolimus for Children and Young Adults With Gliomas Harboring H3.3 or H3.1 K27M Mutation [NCT03632317]Phase 20 participants (Actual)Interventional2019-10-31Withdrawn(stopped due to low accrual)
A Phase I/II Multicenter, National, Open-Label Study of Panobinostat in Combination With Idarubicin and Cytarabine in Patients Aged 65 Years or Older With Newly Diagnosed Acute Myeloblastic Leukaemia (AML) [NCT00840346]Phase 1/Phase 246 participants (Anticipated)Interventional2009-09-30Completed
An Expanded Treatment Protocol of Panobinostat (LBH589) in Combination With Bortezomib and Dexamethasone in Patients With Multiple Myeloma Who Have Had at Least One Prior Line of Therapy [NCT02204553]0 participants Expanded AccessNo longer available
A Randomized Phase II Study of Oral Panobinostat (LBH589) With or Without Rituximab to Treat Relapsed or Refractory Diffuse Large B Cell Lymphoma [NCT01238692]Phase 242 participants (Actual)Interventional2010-11-30Completed
A Randomized, Phase II Trial Evaluating the Efficacy and Safety of Lenalidomide, Bortezomib and Dexamethasone (RVD) With or Without Panobinostat in Transplant Eligible, Newly Diagnosed Multiple Myeloma [NCT02720510]Phase 26 participants (Actual)Interventional2016-06-14Terminated(stopped due to A study was terminated due to low enrollment.)
A Phase II, Multi-center, Single Arm, Open Label Study to Evaluate the Efficacy and Safety of Panobinostat in Combination With Bortezomib and Dexamethasone in Japanese Patients With Relapsed/Refractory Multiple Myeloma [NCT02290431]Phase 231 participants (Actual)Interventional2014-12-16Completed
Phase II Study of Panobinostat (LBH589) for Recurrent Glioblastoma (GBM) Undergoing Planned Surgical Resection [NCT01115036]Phase 20 participants (Actual)Interventional2010-04-30Withdrawn(stopped due to No subjects were enrolled on this study, so study was closed and IND withdrawn.)
Serial Measurements of Molecular and Architectural Responses to Therapy (SMMART) Trial: PRIME [NCT03878524]Phase 12 participants (Actual)Interventional2020-04-01Active, not recruiting
A Phase II Study of the Histone Deacetylase Inhibitor Panobinostat (LBH589) in Patients With Advanced Small Cell Lung Cancer (SCLC) [NCT01222936]Phase 221 participants (Actual)Interventional2008-05-31Completed
A Phase II Study of Oral LBH589 in Adult Patients With Refractory Cutaneous T-Cell Lymphoma [NCT00425555]Phase 2139 participants (Actual)Interventional2007-01-31Completed
A Disease Registry Encompassing the Care of Patients With Multiple Myeloma on Panobinostat (RECOMM) [NCT04150289]248 participants (Actual)Observational [Patient Registry]2019-11-04Completed
Phase I Clinical Trial With LBH589 and Infusional 5-FU/LV in Patients With Metastatic Colorectal Cancer Who Failed 5-FU Based Chemotherapy [NCT01238965]Phase 17 participants (Actual)Interventional2010-10-31Terminated(stopped due to Adverse Events)
A Phase I Dose Finding Study of the Pan-DAC Inhibitor Panobinostat (LBH589) in Combination With Etoposide and Cisplatin in the First Line Treatment of Extensive-Stage Small Cell Lung Cancer - An ICORG In-House Study [NCT01160731]Phase 10 participants (Actual)Interventional2009-11-30Withdrawn
A Phase II Study of Panobinostat in Pediatric, Adolescent and Young Adult Patients With Solid Tumors Including Osteosarcoma, Malignant Rhabdoid Tumor/Atypical Teratoid Rhabdoid Tumors and Neuroblastoma [NCT04897880]Phase 225 participants (Actual)Interventional2019-01-09Terminated(stopped due to Drug supply)
Phase I Trial of Cisplatin and Pemetrexed in Combination With Panobinostat in Advanced Solid Tumors, With Emphasis on Non-Small Cell Lung Cancer [NCT01336842]Phase 123 participants (Actual)Interventional2011-04-30Completed
A Safety and Tolerability Study of RAD001 (mTOR Inhibitor) in Combination With Two Dosing Schedules of LBH589B (Histone Deacetylase Inhibitor) in Solid Tumors/ Lymphomas With Enrichment for EBV-Driven Tumors [NCT01341834]Phase 149 participants (Actual)Interventional2010-03-31Active, not recruiting
Panobinostat in Combination With Rituximab For Relapsed/Refractory Diffuse Large B Cell Lymphoma [NCT01282476]Phase 218 participants (Actual)Interventional2011-06-30Terminated(stopped due to slow accrual)
A One Year, Open Label, Multicenter Trial of LBH589 Alone or in Combination With ESA in Red Blood Cell Transfusion-dependent LOW and INT-1 MDS Patients Being Either Refractory to ESA or With a Low Probability of Response - the GErman PAnobinostat Low Risk [NCT01034657]Phase 234 participants (Actual)Interventional2009-11-30Terminated(stopped due to The study was terminated due to lack of efficacy of single agent LBH589 in the 4 month open label core phase and due to enrollment difficulties.)
A Phase II Trial of LBH589 in Patients With Metastatic Medullary Thyroid Cancer and Radioactive Iodine Resistant Differentiated Thyroid Cancer [NCT01013597]Phase 213 participants (Actual)Interventional2010-01-31Completed
Phase 1 Trial of Panobinostat in Children With Diffuse Intrinsic Pontine Glioma [NCT02717455]Phase 153 participants (Actual)Interventional2016-06-28Active, not recruiting
Phase I Study of LBH589 in Combination With Erlotinib for Advanced Aerodigestive Tract Cancers (CLBH5889CUS11T) [NCT00738751]Phase 144 participants (Actual)Interventional2008-11-30Completed
An Open-label, Multi-center, Expanded Treatment Protocol of Oral Panobinostat in Combination With Bortezomib and Dexamethasone in Patients With Relapsed, and Relapsed and Refractory Multiple Myeloma [NCT02568943]0 participants Expanded AccessNo longer available
A Phase I/IIa Trial of VTD-panobinostat Treatment and Panobinostat Maintenance in Relapsed and Relapsed/Refractory Multiple Myeloma Patients [NCT02145715]Phase 1/Phase 254 participants (Anticipated)Interventional2013-01-31Active, not recruiting
Panobinostat and Ruxolitinib In MyElofibrosis (PRIME STUDY) - Phase I/II Study of Combination Oral JAK2 Tyrosine Kinase Inhibitor (JAK2-TKI) and Histone Deacetylase Inhibitor (HDACI) Therapy in Patients With Myelofibrosis [NCT01693601]Phase 1/Phase 215 participants (Actual)Interventional2013-01-31Completed
Phase I Study of LBH589, A Novel Oral Deacetylase Inhibitor in Patients With Recurrent or Refractory Hodgkin or Non-Hodgkin's Lymphoma [NCT01032148]Phase 18 participants (Actual)Interventional2009-12-31Terminated(stopped due to lack of funding)
Phase I/ II Trial of Carboplatin and Etoposide Plus LBH589 for Previously Untreated Extensive Stage Small Cell Lung Cancer [NCT00958022]Phase 17 participants (Actual)Interventional2009-09-30Terminated(stopped due to Based on the tolerabilty challenges of the combination)
A Phase II Study of Panobinostat (LBH589) as Second-Line Therapy in Patients With Chronic Graft-Versus-Host Disease [NCT01028313]Phase 20 participants (Actual)InterventionalWithdrawn(stopped due to A decision was made to not move forward with the study. No participants were enrolled or treated.)
Pilot Study Evaluating the Expression of ERB-B4 After Treatment With HDAC Inhibitor in ER+ Tamoxifen Refractory Breast Cancer [NCT00993642]Early Phase 10 participants (Actual)Interventional2009-09-30Withdrawn(stopped due to Lack of accrual)
An Open-label Phase 2 Study of Intravenous Bortezomib and Oral Panobinostat (LBH589) in Adult Patients With Relapsed/Refractory Peripheral T-cell Lymphoma or NK/T-cell Lymphoma After Failure of Conventional Chemotherapy [NCT00901147]Phase 225 participants (Actual)Interventional2009-11-30Completed
Phase II Trial of LBH589 (Panobinostat) in Relapsed or Relapsed and Refractory Waldenstrom's Macroglobulinemia [NCT00936611]Phase 239 participants (Actual)Interventional2009-07-31Completed
A Phase II Study of Oral Single Agent Panobinostat in Patients With Refractory de Novo or Secondary Acute Myelogenous Leukemia (AML) [NCT00880269]Phase 259 participants (Actual)Interventional2009-08-31Completed
A Randomized, Triple-arm, Controlled, Open-label, Multicenter Phase II Study Assessing Two Different Doses of Panobinostat in Combination With Carfilzomib and Dexamethasone in Relapsed or Relapsed and Refractory Multiple Myeloma [NCT02756663]Phase 20 participants (Actual)Interventional2016-12-31Withdrawn
A Multicenter, Randomized, Open-label Phase 2 Study Evaluating the Safety and Efficacy of Three Different Regimens of Oral Panobinostat in Combination With Subcutaneous Bortezomib and Oral Dexamethasone in Patients With Relapsed or Relapsed/Refractory Mul [NCT02654990]Phase 2249 participants (Actual)Interventional2016-04-27Completed
Open Label, Multi-center, Phase IV Study of Ruxolitinib or Ruxolitinib and Panobinostat Combination, for Patients Who Have Completed Prior Global Novartis or Incyte Sponsored Studies [NCT02386800]Phase 4356 participants (Anticipated)Interventional2015-03-05Recruiting
A Phase II Study of Oral LBH589 in Adult Patients With Cutaneous T-Cell Lymphoma Who Are Intolerant to or Have Progressed on or After Prior HDAC Inhibitor [NCT00490776]Phase 29 participants (Actual)Interventional2007-07-05Terminated(stopped due to Recruitment was stopped on 15 June 2009 (early termination date) due to low prevalence of study population and slow accrual.)
A Phase I Study Evaluating the Combination of the Deacetylase Inhibitor, LBH589 Plus the mTOR Inhibitor RAD001, in Relapsed and Refractory Adult Patients With Lymphoma [NCT00962507]Phase 111 participants (Actual)Interventional2009-07-31Completed
Phase I, Dose Finding Trial of the Combination of Panobinostat and Stereotactic Radiation in the Treatment of Brain Tumors [NCT01324635]Phase 117 participants (Actual)Interventional2012-05-31Terminated(stopped due to Arm A - reached goal; Arm B - poor accrual)
A Phase II Study Of Oral LBH589 In Adult Patients With Multiple Myeloma Who Have Received At Least Two Prior Lines Of Therapy And Whose Disease Is Refractory To The Most Recent Line Of Therapy [NCT00445068]Phase 238 participants (Actual)Interventional2007-04-16Terminated
A Phase II Study of Oral LBH589 in Patients With Cutaneous T-cell Lymphoma and Adult T-cell Leukemia/Lymphoma [NCT00699296]Phase 24 participants (Actual)Interventional2008-05-31Terminated
A Phase I and Dose Expansion Cohort Study of Panobinostat in Combination With Fludarabine and Cytarabine in Pediatric Patients With Refractory or Relapsed Acute Myeloid Leukemia or Myelodysplastic Syndrome [NCT02676323]Phase 119 participants (Actual)Interventional2016-05-03Terminated(stopped due to Slow accrual)
A Phase I Study of LBH589 (Panobinostat) in Combination With External Beam Radiotherapy for the Treatment of Prostate Cancer, Esophageal Cancer and Head and Neck Cancer [NCT00670553]Phase 17 participants (Actual)Interventional2008-09-30Completed
Phase II Trial of LBH589 in Refractory Colorectal Cancer [NCT00690677]Phase 229 participants (Actual)Interventional2008-06-30Completed
A Phase I/II Study of Oral Melphalan Combined With LBH589 for Patients With Relapsed or Refractory Multiple Myeloma (MM) [NCT00743288]Phase 1/Phase 240 participants (Actual)Interventional2008-07-31Completed
A Phase II, Multicentre Study of Oral LBH589 in Patients With Accelerated Phase or Blast Phase (Blast Crisis) Chronic Myeloid Leukemia With Resistant Disease Following Treatment With at Least Two BCR-ABL Tyrosine Kinase Inhibitors [NCT00449761]Phase 227 participants (Actual)Interventional2007-02-23Terminated
A Phase Ib, Open-label, Two Arm Study of i.v. and Oral Panobinostat (LBH589) in Combination With i.v. Trastuzumab (Herceptin®) and i.v. Paclitaxel as Treatment for Adult Female Patients With HER2 Overexpressing Metastatic Breast Cancer (MBC) [NCT00788931]Phase 115 participants (Actual)Interventional2008-12-31Completed
Phase Ib/II: Epigenetic Modification of Chemosensitivity and Apoptosis in Metastatic Melanoma: Treatment of a Resistant Disease Using Decitabine, Temozolomide and Panobinostat [NCT00925132]Phase 1/Phase 239 participants (Actual)Interventional2009-12-31Terminated(stopped due to Change in the number of approved drugs for metastatic melanoma)
A Phase IA/II, Two-arm, Multi-center, Open-label, Dose-escalation Study of LBH589 Administered Orally Via Different Dosing Schedules in Adult Patients With Advanced Hematological Malignancies [NCT00621244]Phase 1/Phase 2175 participants (Actual)Interventional2003-03-01Completed
A Phase Ib/IIb, Open-label, Multi-center, Study of Oral Panobinostat (LBH589) Administered With 5-Azacitidine (in Adult Patients With Myelodysplastic Syndromes (MDS), Chronic Myelomonocytic Leukemia (CMML), or Acute Myeloid Leukemia (AML). [NCT00946647]Phase 1/Phase 2113 participants (Actual)Interventional2009-12-02Completed
A Phase I/II Study of LBH589 Plus Decitabine for Patients Age ≥ 60 Years With High Risk MDS or AML [NCT00691938]Phase 1/Phase 252 participants (Actual)Interventional2008-06-30Completed
Phase II Trial of LBH589 (Panobinostat) in Adult Patients With Recurrent Malignant Gliomas [NCT00848523]Phase 26 participants (Actual)Interventional2008-11-30Terminated(stopped due to insufficient accrual)
A Phase I/II Study of the Histone Deacetylase (HDAC) Inhibitor LBH589 (Panobinostat) in Combination With mTOR Inhibitor RAD001 (Everolimus) in Patients With Relapsed Multiple Myeloma or Lymphoma [NCT00918333]Phase 1/Phase 2124 participants (Actual)Interventional2009-06-30Completed
Phase II Trial of Oral LBH 589, a Novel Histone Deacetylase (HDAC) Inhibitor, in Relapsed or Refractory Chronic Lymphocytic Leukemia and Mantle Cell Lymphoma [NCT01090973]Phase 21 participants (Actual)Interventional2010-03-31Terminated(stopped due to Pharmaceutical company request.)
Phase I Exploratory Study of Panobinostat IV in Combination With Bortezomib in Relapsed/Refractory Multiple Myeloma Patients [NCT00891033]Phase 112 participants (Actual)Interventional2009-04-30Terminated(stopped due to This study was terminated because the drug company stopped making the study drug)
Pemetrexed and LBH589 in Previously-Treated Patients With Advanced Non-Small Cell Lung Cancer [NCT00907179]Phase 112 participants (Actual)Interventional2009-07-31Terminated(stopped due to Widespread use firstline Pemetrexed; slow recruitment; funding withdrawn..)
A Phase I Dose Escalation Study of LBH589 in Combination With Imatinib Mesylate for Patients With Chronic Myeloid Leukemia in Cytogenetic Remission With Residual Disease Detectable by Q-PCR [NCT00686218]Phase 19 participants (Actual)Interventional2008-05-31Completed
Phase I/II Study of LBH589 and Bevacizumab in Patients With Recurrent High Grade Glioma [NCT00859222]Phase 1/Phase 251 participants (Actual)Interventional2009-03-31Completed
A Phase Ib/IIa Trial of Panobinostat in Combination With Trastuzumab in Adult Female Patients With HER2 Positive Metastatic Breast Cancer Whose Disease Has Progressed During or Following Therapy With Trastuzumab [NCT00567879]Phase 1/Phase 256 participants (Actual)Interventional2008-04-30Terminated(stopped due to The study was terminated early due to insufficient evidence of clinical benefit.)
A Phase II, Open-label Multicenter Trial of Panobinostat Monotherapy in Women With HER2-negative Locally Recurrent or Metastatic Breast Cancer [NCT00777049]Phase 254 participants (Actual)Interventional2009-02-28Completed
A Phase II, Open Label, Single Arm Study of i.v. Panobinostat (LBH589) in Patients With Metastatic Hormone Refractory Prostate Cancer [NCT00667862]Phase 235 participants (Actual)Interventional2008-03-18Completed
A Phase II Study of Oral Panobinostat in Adult Patients With Relapsed/Refractory Classical Hodgkins Lymphoma After High-dose Chemotherapy With Autologous Stem Cell Transplant [NCT00742027]Phase 2129 participants (Actual)Interventional2008-09-16Completed
A Phase I, Open-label, Multicenter Study to Evaluate the Pharmacokinetics and Safety of Oral Panobinostat in Patients With Advanced Solid Tumors and Various Degrees of Hepatic Function [NCT01007968]Phase 125 participants (Actual)Interventional2010-03-31Completed
A Phase I Trial of Sorafenib and LBH589 in the Treatment of Advanced HCC [NCT00823290]Phase 12 participants (Actual)Interventional2009-01-31Terminated(stopped due to Investigator left site)
Phase I/II Randomized Trial of LBH589 (Panobinostat) at Two Dose Levels Combined With Bicalutamide (Casodex) in Men With Castration-resistant Prostate Cancer [NCT00878436]Phase 1/Phase 252 participants (Actual)Interventional2009-06-30Completed
A Phase I Trial of Panobinostat (LBH589) and Epirubicin in Patients With Solid Tumor Malignancies [NCT00878904]Phase 140 participants (Actual)Interventional2009-09-13Completed
A Phase Ib, Multi-center, Open-label, Dose-escalation Study of Oral LBH589 When Administered in Combination With Oral Lenalidomide & Dexamethasone in Adult Patients With Multiple Myeloma [NCT00532675]Phase 146 participants (Actual)Interventional2008-04-22Completed
A Phase II Study of LBH589, a Novel Histone Deacetylase Inhibitor, in Relapsed and Refractory Adult Patients With Acute Leukemia (AL) or in Newly Diagnosed Patients Over the Age of 60 [NCT00723203]Phase 216 participants (Actual)Interventional2008-04-30Terminated(stopped due to Terminated early due to a lack of efficacy)
A Phase IB, Open-Label, Multicenter Study to Investigate the Effect of Oral LBH589 on Dextromethorphan, a CYP2D6 Substrate, and to Assess the Efficacy and Safety of Oral LBH589 in Patients With Advanced Solid Tumors [NCT00535951]Phase 118 participants (Actual)Interventional2007-11-30Completed
A Phase I Safety and Tolerability Study of LBH589 in Combination With Sorafenib in Patients With Advanced Renal Cell Carcinoma, Soft Tissue Sarcoma, and Non-small Cell Lung Carcinoma (NSCLC) of Non-squamous Histologies. [NCT01005797]Phase 138 participants (Actual)Interventional2009-11-30Completed
Panobinostat Combined With High-Dose Gemcitabine/Busulfan/Melphalan With Autologous Stem Cell Transplant for Patients With Refractory/Relapsed Myeloma [NCT02506959]Phase 283 participants (Actual)Interventional2015-09-14Active, not recruiting
Phase I, Prospective, Open-label, Multi-centric, Dose Finding Trial of Combination of IGEV and Panobinostat Before Autologous Stem Cell Transplant in Patients With Hodgkin's Lymphoma [NCT01884428]Phase 124 participants (Anticipated)Interventional2011-07-31Recruiting
Panobinostat Combined With High-Dose Gemcitabine/Busulfan/Melphalan With Autologous Stem Cell Transplant for Patients With Refractory/Relapsed Lymphoma [NCT02961816]Phase 20 participants (Actual)Interventional2017-06-30Withdrawn(stopped due to Lack of funding)
A Phase I Open Label Study Of Panobinostat In Combination With Lenalidomide, Bortezomib, And Dexamethasone In Patients With Relapsed And Relapsed/Refractory Multiple Myeloma [NCT01965353]Phase 121 participants (Actual)Interventional2013-10-31Completed
A Phase I Dose-escalation Study of LBH589 Administered Orally in Adult Patients With Advanced Solid Tumors or Cutaneous T-cell Lymphoma [NCT00412997]Phase 114 participants (Actual)Interventional2006-11-30Completed
Phase I/II Trial of Tamoxifen Following Epigenetic Regeneration of Estrogen Receptor Using Decitabine and LBH 589 in Patients With Triple Negative Metastatic Breast Cancer [NCT01194908]Phase 1/Phase 25 participants (Actual)Interventional2010-07-31Terminated(stopped due to Slow accrual)
Phase Ib, Open-label, Multi-center Study to Characterize the Safety, Tolerability and Pharmacodynamics (PD) of PDR001 in Combination With LCL161, Everolimus (RAD001) or Panobinostat (LBH589) [NCT02890069]Phase 1298 participants (Actual)Interventional2016-10-14Completed
A Phase Ib Open-label, Multicenter Cross-over Study to Investigate the Effect of Food on the Rate and Extent of Oral LBH589 Absorption in Patients With Advanced Solid Tumors [NCT00570284]Phase 136 participants (Actual)Interventional2007-11-30Completed
A Phase II, Multi-center, Single Arm, Open Label Study of Panobinostat in Combination With Bortezomib and Dexamethasone in Patients With Relapsed and Bortezomib-refractory Multiple Myeloma [NCT01083602]Phase 255 participants (Actual)Interventional2010-06-30Completed
A Phase I Study of LBH589 in Combination With Paclitaxel and Carboplatin +/- Bevacizumab the Treatment of Solid Tumors [NCT00556088]Phase 140 participants (Anticipated)Interventional2007-12-31Completed
A Phase I, Open-label, Multi-center Study to Evaluate the Pharmacokinetics and Safety of Oral Panobinostat in Patients With Advanced Solid Tumors and Varying Degrees of Renal Function [NCT00997399]Phase 137 participants (Actual)Interventional2010-03-31Completed
A Phase IA, Dose-escalating Study of LBH589 Administered Intravenously in Adult Patients With Advanced Solid Tumors [NCT00739414]Phase 114 participants (Actual)Interventional2008-07-31Completed
A Phase III Randomized, Double Blind, Placebo Controlled Multi-center Study of Panobinostat for Maintenance of Response in Patients With Hodgkin's Lymphoma Who Are at Risk for Relapse After High Dose Chemotherapy and Autologous Stem Cell Transplant (ASCT) [NCT01034163]Phase 341 participants (Actual)Interventional2010-06-30Completed
Phase II Trial of Oral Panobinostat (LBH589), a Novel Deacetylase Inhibitor (DACi) in Patients With Primary Myelofibrosis (PMF), Post Essential Thrombocythemia (ET) Myelofibrosis and Post- Polycythemia Vera (PV) Myelofibrosis [NCT00931762]Phase 235 participants (Actual)Interventional2009-08-31Terminated
Phase I Study of Panobinostat Plus ICE Chemotherapy Followed by a Randomized Phase-II Study of ICE Compared With Panobinostat Plus ICE for Patients With Relapsed and Refractory Classical Hodgkin Lymphoma [NCT01169636]Phase 1/Phase 262 participants (Actual)Interventional2011-01-31Completed
A Phase 2 Study to Evaluate the Efficacy of Epigenetic Modulation in Relapsed/Refractory Diffuse Large B-Cell Lymphoma (DLBCL) [NCT00978432]Phase 250 participants (Actual)Interventional2012-02-29Terminated(stopped due to The toxicity seemed to outweigh the benefit.)
An Open Label, Single Arm, Phase Ib Dose Finding Study of i.v. Panobinostat (LBH589) With Docetaxel and Prednisone in Patients With Hormone Refractory Prostate Cancer [NCT00663832]Phase 144 participants (Actual)Interventional2008-02-29Completed
A Phase I/II Trial Evaluating the Use of Histone Deacetylase Inhibitor LBH589 in Addition to Corticosteroids in Patients With Acute Graft Versus Host Disease [NCT01111526]Phase 1/Phase 222 participants (Actual)Interventional2010-04-30Completed
Phase I-II Study of Carfilzomib, Lenalidomide, Dexamethasone, and Panobinostat, Ca-R-Pa-Diem, as Induction Therapy for Newly Diagnosed, Untreated, Transplant-Eligible, Multiple Myeloma Patients [NCT02802163]Phase 1/Phase 20 participants (Actual)Interventional2017-06-30Withdrawn(stopped due to study drug unavailable)
A Phase I Study of LBH589 in Combination With Gemcitabine in the Treatment of Solid Tumors [NCT00550199]Phase 117 participants (Actual)Interventional2007-11-30Terminated(stopped due to Study terminated due to LBH589 toxicity.)
Phase I Study of Combination of Sorafenib and LBH589 in Hepatocellular Carcinoma [NCT00873002]Phase 13 participants (Actual)Interventional2009-03-31Terminated(stopped due to Dose Limiting Toxicity)
A Phase I Study of Panobinostat in Combination With Daratumumab, Bortezomib, and Dexamethasone in Patients With Relapsed/Refractory Multiple Myeloma [NCT04956302]Phase 11 participants (Actual)Interventional2021-09-27Terminated(stopped due to PI Decision)
A Multicenter, Randomized, Double Blind, Placebo Controlled Phase III Study of Panobinostat in Combination With Bortezomib and Dexamethasone in Patients With Relapsed Multiple Myeloma [NCT01023308]Phase 3767 participants (Actual)Interventional2009-12-21Completed
Phase 1 Trial of Marizomib Alone and in Combination With Panobinostat for Children With Diffuse Intrinsic Pontine Glioma [NCT04341311]Phase 145 participants (Anticipated)Interventional2020-08-10Active, not recruiting
An Open-label, Single Center, Study to Determine the Absorption, Distribution, Metabolism, and Excretion (ADME) of LBH589 After a Single Oral Administration of 20 mg (14C) LBH589 in Advanced Cancer Patients [NCT00472368]Phase 14 participants (Actual)Interventional2007-05-31Completed
A Phase IB, Study to Investigate the Effect of Ketoconazole, a CYP3A4 Inhibitor, on Oral LBH589 and to Assess the Efficacy and Safety of Oral LBH589 in Patients With Advanced Solid Tumors. [NCT00503451]Phase 114 participants (Actual)Interventional2007-09-30Completed
A Phase IA/IB, Two Arm, Multi-center, Open-label, Dose Escalation Study of Oral LBH589 Alone and in Combination With IV Docetaxel and Oral Prednisone in Hormone Refractory Prostate Cancer (HRPC) [NCT00493766]Phase 116 participants (Actual)Interventional2006-05-31Terminated(stopped due to Because of a strategic decision by Novartis)
A Phase Ib, Multi-center, Open-label, Dose-escalation Study of Oral LBH589 When Administered in Combination With Bortezomib in Adult Patients With Multiple Myeloma [NCT00532389]Phase 162 participants (Actual)Interventional2007-10-31Completed
Phase II Study of LBH589 for Patients With Low or Intermediate-1 Risk Myelodysplastic Syndrome [NCT00939159]Phase 217 participants (Actual)Interventional2009-08-31Terminated(stopped due to Low Accrual)
A Phase IA/IB, Two Arm, Multi-center, Open-label, Dose Escalation Study of Oral LBH589 Alone and in Combination With IV Docetaxel and Oral Prednisone in Hormone Refractory Prostate Cancer (HRPC) [NCT00419536]Phase 1108 participants Interventional2006-05-31Terminated
Phase I/II of Panobinostat (LBH589) Plus Everolimus (RAD001) in Patients With Relapsed and Refractory Lymphoma [NCT00967044]Phase 1/Phase 231 participants (Actual)Interventional2009-11-30Completed
A Phase II Trial of Panobinostat and Lenalidomide in Patients With Relapsed or Refractory Hodgkin's Lymphoma [NCT01460940]Phase 224 participants (Actual)Interventional2011-10-13Completed
An Open-label, Uncontrolled Phase II Trial of HDAC-Inhibitor LBH589 in Patients With Chemo-refractory Metastatic Gastric Cancer Overexpressing Histone Deacetylases (HDACs) - CLBH589BDE03T [NCT01528501]Phase 228 participants (Anticipated)Interventional2009-06-30Terminated(stopped due to medical/ethical reasons)
A Phase I Study of the Combination of Panobinostat and Carfilzomib in Patients With Relapsed and/or Refractory Multiple Myeloma [NCT01549431]Phase 132 participants (Actual)Interventional2012-01-31Completed
A Phase I-II Pilot Study to Assess the Safety and Efficacy of Combined Administration With Pegylated Interferon-alpha2a and the Histone Deacetylase Inhibitor (HDACi) Panobinostat for Reducing the Residual Reservoir of HIV-1 Infected Cells in cART-Treated [NCT02471430]Phase 1/Phase 234 participants (Anticipated)Interventional2016-05-31Active, not recruiting
A Phase II Study of LBH589 in the Treatment of Patients With Refractory Clear Cell Renal Carcinoma [NCT00550277]Phase 220 participants (Actual)Interventional2008-01-31Completed
A Phase I Study of LBH589 in Combination With Capecitabine ± Lapatinib [NCT00632489]Phase 120 participants (Actual)Interventional2008-05-31Completed
A Randomized Phase II, Open-label Multicenter Trial of Panobinostat Monotherapy in Women With HER2 Positive Locally Recurrent or Metastatic Breast Cancer [NCT00777335]Phase 24 participants (Actual)Interventional2009-02-28Terminated(stopped due to Very low recruitement rate.)
Study to Investigate Outcome of Individualized Treatment Based on Pharmacogenomic Profiling & Ex Vivo Drug Sensitivity Testing of Patient-derived Organoids in Patients With Metastatic Colorectal Cancer [NCT05725200]Phase 240 participants (Anticipated)Interventional2022-09-27Recruiting
A Phase I/II Study of MLN9708 (Ixazomib) in Combination With Panobinostat and Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma [NCT02057640]Phase 1/Phase 216 participants (Actual)Interventional2014-05-22Completed
A Phase I Study of Panobinostat in Combination With Bortezomib in the Treatment of Relapsed and/or Refractory Mantle Cell Lymphoma [NCT01504776]Phase 13 participants (Actual)Interventional2011-04-30Completed
Phase I Study to Determine the Safety and Tolerability of Escalating Doses of Panobinostat (LBH589) in Patients With Sickle Cell Disease [NCT01245179]Phase 118 participants (Anticipated)Interventional2010-11-30Recruiting
The Safety and Efficacy of The Histone Deacetylase Inhibitor Panobinostat for Purging HIV-1 From The Latent Reservoir (CLEAR) Study [NCT01680094]Phase 1/Phase 215 participants (Actual)Interventional2012-09-30Completed
A Phase II Trial of LBH589 in Refractory Myelodysplastic Syndromes (MDS) Patients [NCT00594230]Phase 226 participants (Actual)Interventional2008-01-31Terminated(stopped due to Per protocol, the results of a planned interim analysis demonstrated insufficient efficacy and led to early termination of the study.)
A Phase II Study of Oral Panobinostat in Adult Patients With Diffuse Large B-cell Lymphoma Relapsed/Refractory After High-dose Chemotherapy With Autologous Stem Cell Transfusion (ASCT) or Not Eligible for ASCT [NCT01523834]Phase 235 participants (Actual)Interventional2011-02-28Completed
A Phase 1b, Open-label, Multi-center, Single Arm, Dose Finding Study to Assess Safety and Pharmacokinetics of the Oral Combination of Panobinostat and Ruxolitinib in Patients With Primary Myelofibrosis (PMF), Post-polycythemia Vera-myelofibrosis (PPV-MF) [NCT01433445]Phase 161 participants (Actual)Interventional2011-11-01Completed
Phase I/Ib Trial of the Efficacy and Safety of Combination Therapy of Lenalidomide/Bortezomib/Dexamethasone and Panobinostat in Transplant Eligible Patients With Newly Diagnosed Multiple Myeloma (MM) [NCT01440582]Phase 177 participants (Actual)Interventional2013-02-18Completed
A Phase I Dose Finding and Proof-of-concept Study of the Histone Deacetylase Inhibitor Panobinostat (LBH589) in Combination With Standard Dose Cytarabine and Daunorubicin for Older Patients With Untreated Acute Myeloid Leukemia or Advanced Myelodysplastic [NCT01463046]Phase 129 participants (Actual)Interventional2012-01-31Completed
Phase II Study of Panobinostat (LBH589) Given in Combination With Bortezomib (Velcade) in Patients With Pancreatic Cancer Progressing on Gemcitabine Therapy Alone or Gemcitabine in Combination [NCT01056601]Phase 27 participants (Actual)Interventional2010-09-30Terminated(stopped due to Funding not available)
A Phase II Trial of Panobinostat in Patients With Neuroendocrine Tumors [NCT00985946]Phase 215 participants (Actual)Interventional2010-05-31Terminated(stopped due to Study did not meet statistical requirements to continue.)
An Open Label Single Arm Phase I/II Study of MTX110 Delivered by Convection-enhanced Delivery (CED) in Patients With Diffuse Intrinsic Pontine Glioma (DIPG) Previously Treated With External Beam Radiation Therapy [NCT03566199]Phase 1/Phase 27 participants (Actual)Interventional2018-05-22Completed
A Phase II Study Incorporating Panobinostat, Bortezomib and Liposomal Vincristine Into Re-Induction Therapy for Relapsed Pediatric T-Cell Acute Lymphoblastic Leukemia or Lymphoma [NCT02518750]Phase 23 participants (Actual)Interventional2016-11-23Terminated(stopped due to Due to slow accrual)
A Phase II Trial Evaluating the Use of a Histone Deacetylase Inhibitor Panobinostat for Graft Versus Host Disease (GVHD) Prevention [NCT02588339]Phase 242 participants (Actual)Interventional2016-03-04Completed
A Phase Ib, Open-label, Multi-center, Dose-escalation Study of Oral Panobinostat (LBH589) Administered With 5-Azacitidine (Vidaza®) in Adult Japanese Patients With Myelodysplastic Syndromes (MDS), Chronic Myelomonocytic Leukemia (CMML) or Acute Myeloid Le [NCT01613976]Phase 110 participants (Actual)Interventional2012-08-31Completed
Phase I/II Study With Oral Panobinostat Maintenance Therapy Following Allogeneic Stem Cell Transplantation in Patients With High Risk MDS or AML (PANOBEST) [NCT01451268]Phase 1/Phase 262 participants (Anticipated)Interventional2011-01-31Active, not recruiting
Phase I/II Study of Panobinostat (LBH589) and Letrozole in Patients With Triple Negative Metastatic Breast Cancer [NCT01105312]Phase 1/Phase 228 participants (Actual)Interventional2010-09-30Completed
A Phase II Study to Investigate Biological Correlates of Clinical Response to Panobinostat in Haematological Malignancy [NCT01658241]Phase 230 participants (Actual)Interventional2012-07-16Completed
A Phase II, Single-Center, Open-Label Study Of Oral Panobinostat (LBH589) When Administered In Combination With Lenalidomide And Weekly Dexamethasone In Patients With Multiple Myeloma [NCT01651039]Phase 232 participants (Actual)Interventional2012-07-31Completed
A Randomized, Multicenter Phase III Study to Assess the Efficacy of Panobinostat Maintenance Therapy vs. Standard of Care Following Allogeneic Stem Cell Transplantation in Patients With High-risk AML or MDS (ETAL-4 / HOVON-145) [NCT04326764]Phase 352 participants (Actual)Interventional2018-07-24Terminated(stopped due to Change of marketing authorisation holder)
A Phase I/II Study of the HDAC Inhibitor LBH-589 in Combination With the mTOR Inhibitor Everolimus (RAD001) in Metastatic Renal Cell Carcinoma [NCT01582009]Phase 1/Phase 226 participants (Actual)Interventional2010-03-31Terminated(stopped due to pts off study, PI left institute)
A Phase II Study of the Histone Deacetylase (HDAC) Inhibitor LBH589 (Panobinostat) in Patients With Relapsed or Refractory Non-Hodgkin Lymphoma [NCT01261247]Phase 241 participants (Actual)Interventional2011-01-17Completed
An Open-label Multi-center Single Agent Panobinostat Roll-over Protocol for Patients Who Have Completed a Previous Novartis-sponsored Panobinostat Study and Are Judged by the Investigator to Benefit From Continued Single Agent Panobinostat Treatment [NCT01802879]Phase 29 participants (Actual)Interventional2013-06-24Completed
A Feasibility Study Examining the Use of Non-Invasive Focused Ultrasound (FUS) With Oral Panobinostat Administration in Children With Progressive Diffuse Midline Glioma (DMG) [NCT04804709]Phase 13 participants (Actual)Interventional2021-07-28Active, not recruiting
Phase 1/1b Study of the Efficacy and Safety of the Combination of Panobinostat + Carfilzomib in Patients With Relapsed/Refractory Myeloma [NCT01301807]Phase 163 participants (Actual)Interventional2011-07-28Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT00425555 (13) [back to overview]Progression-free Survival (PFS)
NCT00425555 (13) [back to overview]Maximum Plasma Concentration (Cmax) of Panobinostat
NCT00425555 (13) [back to overview]Duration of Response (DOS)
NCT00425555 (13) [back to overview]Overall Response Rate of Participants Using the Modified Severity-Weighted Assessment Tool (mSWAT)
NCT00425555 (13) [back to overview]Skindex-29 Measurements of Average Sub-scores for Emotions From Baseline up to Cycle 12
NCT00425555 (13) [back to overview]Time to Response for Responders
NCT00425555 (13) [back to overview]Area Under the Plasma Concentration AUC0-24, AUC0-48 and AUC 0-infinity of Panobinostat
NCT00425555 (13) [back to overview]The Overall Response Rate Using mSWAT Skin Score
NCT00425555 (13) [back to overview]Time to Peak Concentration (Tmax) of Panobinostat
NCT00425555 (13) [back to overview]Time of Clast (Tlast) of Panobinostat
NCT00425555 (13) [back to overview]Last Observed Plasma Concentration (Clast) of Panobinostat
NCT00425555 (13) [back to overview]Skindex-29 Measurements of Average Sub-scores for Physical Symptoms From Baseline up to Cycle 12
NCT00425555 (13) [back to overview]Skindex-29 Measurements of Average Sub-scores for Functioning From Baseline up to Cycle 12
NCT00445068 (6) [back to overview]Safety and Tolerability
NCT00445068 (6) [back to overview]Maximum Plasma Concentration (Cmax) of Panobinostat
NCT00445068 (6) [back to overview]Time of Clast (Tlast) of Panobinostat
NCT00445068 (6) [back to overview]Area Under the Plasma Concentration (AUC0-24) of Panobinostat
NCT00445068 (6) [back to overview]Last Observed Plasma Concentration (Clast) of Panobinostat
NCT00445068 (6) [back to overview]Time to Peak Concentration (Tmax) of Panobinostat
NCT00449761 (9) [back to overview]QT Interval (QTc) in Participants Receiving Oral Panobinostat at Baseline and Change From Baseline to Extreme Value
NCT00449761 (9) [back to overview]Safety and Tolerability of Panobinostat
NCT00449761 (9) [back to overview]Time of Clast (Tlast) of Panobinostat
NCT00449761 (9) [back to overview]Time to Peak Concentration (Tmax) of Panobinostat
NCT00449761 (9) [back to overview]Maximum Plasma Concentration (Cmax) of Panobinostat
NCT00449761 (9) [back to overview]Last Observed Plasma Concentration (Clast) of Panobinostat
NCT00449761 (9) [back to overview]Area Under the Plasma Concentration (AUC0-24) of Panobinostat
NCT00449761 (9) [back to overview]Participants With Hematologic Response
NCT00449761 (9) [back to overview]BCR-ABL Mutations of Participants at Study Entry and, in Responding Participants and at the Time of Disease Progression
NCT00451035 (9) [back to overview]QT Interval (QTc) in Participants Receiving Oral Panobinostat at Baseline and Change From Baseline to Extreme Value
NCT00451035 (9) [back to overview]Time of Clast (Tlast) of Panobinostat
NCT00451035 (9) [back to overview]Area Under the Plasma Concentration (AUC0-24) of Panobinostat
NCT00451035 (9) [back to overview]Safety and Tolerability Profile of Oral Panobinostat
NCT00451035 (9) [back to overview]BCR-ABL Mutations of Participants at Study Entry and, in Responding Participants and at the Time of Disease Progression
NCT00451035 (9) [back to overview]Last Observed Plasma Concentration (Clast) of Panobinostat
NCT00451035 (9) [back to overview]Major (Complete/Partial) Cytogenetic Response (MCyR) Rate
NCT00451035 (9) [back to overview]Maximum Plasma Concentration (Cmax) of Panobinostat
NCT00451035 (9) [back to overview]Time to Peak Concentration (Tmax) of Panobinostat
NCT00550277 (3) [back to overview]Number of Participants Experiencing ≥Grade 2 Adverse Events
NCT00550277 (3) [back to overview]Number of Participants With Overall Response
NCT00550277 (3) [back to overview]Progression-free Survival
NCT00567879 (2) [back to overview]Number of Participants With Best Overall Response
NCT00567879 (2) [back to overview]Number of Participants With Dose Limiting Toxicities (DLTs)
NCT00594230 (2) [back to overview]Overall Response Rate (CR, Marrow CR + PR) of LBH in Patients With Relapsed or Refractory MDS.
NCT00594230 (2) [back to overview]Safety and Tolerability of LBH589 in Patients With Relapsed/Refractory MDS by Measuring the Number of Participants With Adverse Events
NCT00621244 (17) [back to overview]Number of Participants DLT in Arm 2 in Dose Escalation Phase
NCT00621244 (17) [back to overview]Number of Participants DLT in Arm 1 in Dose Escalation Phase
NCT00621244 (17) [back to overview]Maximum Plasma Concentration of Panobinostat After the First Dose in Arms 1 and 2
NCT00621244 (17) [back to overview]Half Life of Panobinostat After Multiple Doses in Arm 1 on Day 15
NCT00621244 (17) [back to overview]Percentages of Participants With Histone Acetylation Induction in Peripheral Blood in Arm 1 (MWF Every Week), Group X
NCT00621244 (17) [back to overview]Maximum Plasma Concentration of Panobinostat After Multiple Doses in Arm 1 on Day 15
NCT00621244 (17) [back to overview]Highest Percent Change of Fetal Hemoglobin From Baseline in Arm 2 (MWF Every Other Week)
NCT00621244 (17) [back to overview]Percentage of Participants With Histone Acetylation Induction in Peripheral Blood in Arm 2 (MWF Every Other Week), Group Y
NCT00621244 (17) [back to overview]Highest Percent Change in Fetal Hemoglobin From Baseline in Arm 1 (MWF Every Week)
NCT00621244 (17) [back to overview]Half Life of Panobinostat After the First Dose in Arms 1 and 2
NCT00621244 (17) [back to overview]Response as Per Investigator Assessment for Patients With Myelodysplastic Syndromes (MDS)
NCT00621244 (17) [back to overview]Percentages of Participants With Histone Acetylation Induction in Peripheral Blood in Arm 2 (MWF Every Other Week), Group X
NCT00621244 (17) [back to overview]Response as Per Investigator Assessment for Patients With Hodgkin's Lymphoma (HD)
NCT00621244 (17) [back to overview]Response as Per Investigator Assessment for Patients With Acute Myelogenous Leukemia (AML) in Expansion Phase
NCT00621244 (17) [back to overview]Geometric Mean Ratio (GMR) Comparing Treatment Days in Arm 1
NCT00621244 (17) [back to overview]Percentages of Participants With Histone Acetylation Induction in Peripheral Blood in Arm 1 (MWF Every Week), Group Y
NCT00621244 (17) [back to overview]Response as Per Investigator Assessment for Patients With Acute Myelogenous Leukemia (AML)
NCT00632489 (2) [back to overview]To Determine the Maximum Tolerated Doses (MTD) and Dose-limiting Toxicities (DLT) of LBH589 in Combination With Capecitabine When Administered to Patients With Refractory and Advanced Tumor Types That Are Sensitive to 5-fluorouracil
NCT00632489 (2) [back to overview]To Determine the Maximum Tolerated Doses (MTD) and Dose-limiting Toxicities (DLT) of LBH589 in Combination With Capecitabine When Administered to Patients With Refractory and Advanced Tumor Types That Are Sensitive to 5-fluorouracil
NCT00667862 (5) [back to overview]Percentage of Participants With PSA Progression Rate at 24 Weeks
NCT00667862 (5) [back to overview]Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)
NCT00667862 (5) [back to overview]Percentage of Participants With Tumor Response Rate
NCT00667862 (5) [back to overview]Percentage of Participants With Progression-Free Survival (PFS) Rate at 24 Weeks
NCT00667862 (5) [back to overview]Percentage of Participants With Prostate Specific Antigen (PSA) Response Rate at 24 Weeks
NCT00691938 (10) [back to overview]Overall Survival
NCT00691938 (10) [back to overview]Event-free Survival
NCT00691938 (10) [back to overview]Rate of Cytogenetic Complete Remission (CRc)
NCT00691938 (10) [back to overview]Time to Response
NCT00691938 (10) [back to overview]Rates of Morphologic Complete Remission With Incomplete Count Recovery (CRi)
NCT00691938 (10) [back to overview]Remission Duration
NCT00691938 (10) [back to overview]Rate of Hematologic Improvement.
NCT00691938 (10) [back to overview]Phase II: Overall Rate of Morphologic Complete Remission (CR) + Cytogenetic Complete Remission (CRc) + Morphologic Complete Remission With Incomplete Blood Count Recovery (CRi)
NCT00691938 (10) [back to overview]Phase I: Maximum Tolerated Dose (MTD) of LBH589 When Given in Combination With Decitabine
NCT00691938 (10) [back to overview]Safety and Tolerability of Regimen as Measured by the Rate of the Most Common Adverse Events Experienced
NCT00723203 (1) [back to overview]Hematological Response Rate
NCT00742027 (11) [back to overview]Maximum Observed Concentration (Cmax) of Panobinostat
NCT00742027 (11) [back to overview]Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), And Deaths as a Measure of Safety and Tolerability of Panobinostat
NCT00742027 (11) [back to overview]Area Under the Plasma Concentration-Time Curve From Time Zero to 28 Hours (AUC0-28) for Panobinostat
NCT00742027 (11) [back to overview]Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC0-∞) for Panobinostat
NCT00742027 (11) [back to overview]Duration of Overall Disease Response
NCT00742027 (11) [back to overview]Objective Response Rate (ORR) as Assessed by the Investigator Based on Cheson Response Criteria
NCT00742027 (11) [back to overview]Response Rate Based on Central Review of Computed Tomography (CT) Scan/Magnetic Resonance Imaging (MRI)
NCT00742027 (11) [back to overview]Progression Free Survival (PFS)
NCT00742027 (11) [back to overview]The Overall Survival (OS)
NCT00742027 (11) [back to overview]The Time to Reach Maximum Plasma Concentration (Tmax) of Panobinostat
NCT00742027 (11) [back to overview]Time To Overall Disease Response in Responders
NCT00743288 (5) [back to overview]Maximum Tolerated Dose (MTD)
NCT00743288 (5) [back to overview]Duration of Response
NCT00743288 (5) [back to overview]MTD
NCT00743288 (5) [back to overview]Overall Response Rate (ORR) and Clinical Benefit Rate (CBR) [ORR= Complete Response (CR) + Very Good Partial Response (VGPR) + Partial Response (PR)]; CBR=ORR + Minimal Response (MR)] Following Treatment With Panobinostat and Melphalan
NCT00743288 (5) [back to overview]Time to Progression
NCT00777049 (1) [back to overview]Objective Response Rate (as Determined by Investigator): the Percentage of Patients Assigned to a Treatment Arm With a Confirmed Best Response of CR or PR.
NCT00777335 (2) [back to overview]Corrected QT Interval Fridericia's Formula (QTcF)
NCT00777335 (2) [back to overview]Overall Response (OR) Rate (as Determined by the Investigator): the Number of Patients Assigned to a Treatment Arm With a Confirmed Best Response of Complete Response(CR) or Partial Response (PR).
NCT00859222 (9) [back to overview]6-Month Progression-Free Survival (PFS6) [Phase I]
NCT00859222 (9) [back to overview]Overall Survival [Phase II]
NCT00859222 (9) [back to overview]Best Radiographic Response
NCT00859222 (9) [back to overview]Progression-Free Survival (PFS) [Phase II]
NCT00859222 (9) [back to overview]Progression-Free Survival (PFS) [Phase I]
NCT00859222 (9) [back to overview]Overall Survival (OS) [Phase I]
NCT00859222 (9) [back to overview]LBH589 Maximum Tolerated Dose (MTD) [Phase I]
NCT00859222 (9) [back to overview]Dose Limiting Toxicity (DLT) [Phase I]
NCT00859222 (9) [back to overview]6-Month Progression-Free Survival (PFS6) [Phase II]
NCT00878436 (2) [back to overview]Percentage of Patients Free of Progression and Without Symptomatic Deterioration
NCT00878436 (2) [back to overview]Percentage of Patients Free of Progression and Without Symptomatic Deterioration
NCT00880269 (1) [back to overview]Best Response as Per Investigator Assessment by Stratum (FAS)
NCT00918333 (5) [back to overview]Duration of Response (Phase II)
NCT00918333 (5) [back to overview]Number of Phase I Participants With Dose-Limiting Toxicity Events (Phase I)
NCT00918333 (5) [back to overview]Overall Response Rate (Phase II)
NCT00918333 (5) [back to overview]Overall Survival Time (Phase II)
NCT00918333 (5) [back to overview]Progression-free Survival (Phase II)
NCT00925132 (2) [back to overview]Phase I - Number of Participants With Dose Limiting Toxicities (DLTs) at a Given Dose Level
NCT00925132 (2) [back to overview]Phase 2 -Number of Patients With a Decrease in Tumor Size Using RECIST and CHOI's Criteria
NCT00931762 (4) [back to overview]Change From Baseline in Myelofibrosis Symptom Assessment Form (MF-SAF) Scale Scores At Cycle 6
NCT00931762 (4) [back to overview]Number of Participants With Adverse Events (AEs), and Serious Adverse Events (SAEs)
NCT00931762 (4) [back to overview]Overall Response (OR) Rate Per International Working Group for Myelofibrosis Research and Treatment (IWG-MRT) Criteria
NCT00931762 (4) [back to overview]Change From Baseline in Myelofibrosis Symptom Assessment Form (MF-SAF) Scale Scores
NCT00936611 (5) [back to overview]Median Duration of Response
NCT00936611 (5) [back to overview]Median Progression Free Survival
NCT00936611 (5) [back to overview]Median Time to Progression
NCT00936611 (5) [back to overview]Number of Participants With Grade 3 and 4 Treatment-Related Thrombocytopenia
NCT00936611 (5) [back to overview]Overall Response Rate
NCT00939159 (1) [back to overview]Overall Response Rate Based on the Hematologic Improvement
NCT00946647 (10) [back to overview]Hematologic Improvement (HI) for Myeloid Dysplastic Syndromes(MDS)/Chronic Myelomonocytic Leukemia (CMML) Patients Per Investigator (Phase Llb)
NCT00946647 (10) [back to overview]Overall Response Rate (ORR) Assessed by Best Overall Response: Participants With AML Per Investigator (Phase Llb)
NCT00946647 (10) [back to overview]Overall Response Rate (ORR) Assessed by Best Overall Response: Participants With MDS/CMML Per Investigator (Phase Llb)
NCT00946647 (10) [back to overview]Clinical Response Other Than Composite Clinical Response for Myeloid Dysplastic Syndromes(MDS)/Chronic Myelomonocytic Leukemia (CMML) Patients Per Investigator (Phase Llb)
NCT00946647 (10) [back to overview]Clinical Response Other Than Composite Clinical Response for Acute Myelogenous Leukemia (AML) Patients Per Investigator (Phase Llb)
NCT00946647 (10) [back to overview]1-year Survival Rate (Phase Llb)
NCT00946647 (10) [back to overview]Number of Dose Limiting Toxicity (DLT) (Phase lb)
NCT00946647 (10) [back to overview]Number of Participants With Dose Limiting Toxicity (DLT) (Phase lb)
NCT00946647 (10) [back to overview]Time to Progression (TTP) (Phase Llb)
NCT00946647 (10) [back to overview]Composite Complete Response (Phase Llb)
NCT00967044 (1) [back to overview]Maximum Tolerated Dose (MTD) of Everolimus With Panobinostat
NCT00978432 (2) [back to overview]Overall Response Rate
NCT00978432 (2) [back to overview]Summary of Adverse Events (AEs)
NCT00985946 (4) [back to overview]Evaluate the Overall Survival of Patients With Gastrointestinal Neuroendocrine Tumors Treated With Panobinostat
NCT00985946 (4) [back to overview]Evaluate the Time to Progression for Patients With Gastrointestinal Neuroendocrine Tumors Treated With Panobinostat
NCT00985946 (4) [back to overview]Number of Participants With Toxicities
NCT00985946 (4) [back to overview]Tumor Response Rate of Patients With Gastrointestinal Neuroendocrine Tumors Using Response Evaluation Criteria in Solid Tumors (RECIST) Criteria.
NCT01013597 (5) [back to overview]Tumor Response Rate to LBH589.
NCT01013597 (5) [back to overview]Overall Survival
NCT01013597 (5) [back to overview]Impact of LBH589 on Tumor Markers for Thyroid Cancer
NCT01013597 (5) [back to overview]Time to Progression of Thyroid Cancer
NCT01013597 (5) [back to overview]Toxicity of LBH589
NCT01023308 (11) [back to overview]Overall Response Rate in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.
NCT01023308 (11) [back to overview]European Organization for Research and Treatment of Cancer Multiple Myeloma Module (EORTC) QLQ-MY20-Change From Baseline by Treatment Group
NCT01023308 (11) [back to overview]European Organization for Research and Treatment of Cancer Multiple Myeloma Module (EORTC ) QLQ-C30 - Summary Statistics by Treatment Group
NCT01023308 (11) [back to overview]Time to Response Per Investigator Assessment (mEBMT Criteria) of Response Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.
NCT01023308 (11) [back to overview]Time to Progression/Relapse Per Investigator Assessment (mEBMT Criteria) Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.
NCT01023308 (11) [back to overview]Progression-free Survival Events in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.
NCT01023308 (11) [back to overview]Progression Free Survival in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.
NCT01023308 (11) [back to overview]Overall Survival in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone
NCT01023308 (11) [back to overview]Duration of Response Per Investigator Assessment (mEBMT Criteria) Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.
NCT01023308 (11) [back to overview]Overall Survival in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone
NCT01023308 (11) [back to overview]Functional Assessment of Chronic Illness Therapy (FACIT) Measurement System : FACT/GOG-NTX-Change From Baseline by Treatment Group
NCT01034163 (1) [back to overview]Number of Participants With Adverse Events
NCT01034657 (9) [back to overview]Frequency Distribution of IPSS Score Status - Randomized Phase
NCT01034657 (9) [back to overview]Percentage of Participants With Objective Response During the Randomized Phase
NCT01034657 (9) [back to overview]Frequency Distribution of IPSS Score Status - Core Phase
NCT01034657 (9) [back to overview]Percentage of Participants With HI-E - Randomized Phase
NCT01034657 (9) [back to overview]Percentage of Participants With Objective Response During Core Phase
NCT01034657 (9) [back to overview]Percentage of Participants With Hematological Response of the Erythropoetic System (HI-E) - Core Phase
NCT01034657 (9) [back to overview]Overall Survival (OS) - Overall Period
NCT01034657 (9) [back to overview]Mean Single Scoring Values of the IPSS - Randomized Phase
NCT01034657 (9) [back to overview]Mean Single Scoring Values of the IPSS - Core Phase
NCT01056601 (3) [back to overview]Number of Participants by Tumor Response
NCT01056601 (3) [back to overview]Progression-Free Survival
NCT01056601 (3) [back to overview]Duration of Response
NCT01083602 (6) [back to overview]Over All Survival
NCT01083602 (6) [back to overview]Overall Response Rate (PR+nCR+CR)
NCT01083602 (6) [back to overview]Progression-free Survival
NCT01083602 (6) [back to overview]Time to Response (Greater Than or Equal to PR) Based on Investigator Assessment
NCT01083602 (6) [back to overview]Responders to Treatment
NCT01083602 (6) [back to overview]Time to Progression
NCT01090973 (1) [back to overview]Number of Participants With Adverse Events (AEs)
NCT01105312 (9) [back to overview]Clinical Benefit Rate
NCT01105312 (9) [back to overview]Duration of Response (Phase II)
NCT01105312 (9) [back to overview]Confirmed Response Rate (Phase I)
NCT01105312 (9) [back to overview]Maximum-tolerated Dose (Phase I)
NCT01105312 (9) [back to overview]Progression-free Survival (Phase II)
NCT01105312 (9) [back to overview]Response Rate (Phase II)
NCT01105312 (9) [back to overview]Survival Time (Phase II)
NCT01105312 (9) [back to overview]Time to Treatment Failure
NCT01105312 (9) [back to overview]Time-to-disease Progression (Phase II)
NCT01111526 (9) [back to overview]Occurrence of Possibly Related Adverse Events
NCT01111526 (9) [back to overview]Chronic GVHD Severity at MTD
NCT01111526 (9) [back to overview]Occurrence of Discontinuation of All Immune Suppression
NCT01111526 (9) [back to overview]Phase II: Overall Rate of Response (ORR)
NCT01111526 (9) [back to overview]Chronic GVHD Onset
NCT01111526 (9) [back to overview]Incidence of GVHD Flares Requiring Increasing Immune Suppressive Therapy
NCT01111526 (9) [back to overview]Phase I: Maximum Tolerated Dose (MTD) in Milligrams
NCT01111526 (9) [back to overview]Stable or Improved Chronic GVHD Severity Score
NCT01111526 (9) [back to overview]Overall Survival (OS)
NCT01169636 (3) [back to overview]Number of Participants With Complete Remission (CR)
NCT01169636 (3) [back to overview]Percentage of Participants With Failure Free Survival (FFS)
NCT01169636 (3) [back to overview]Maximum Tolerated Dose (MTD) of Panobinostat + ICE
NCT01261247 (4) [back to overview]Proportion of Confirmed Responses Defined to be a CR or PR Noted as the Objective Status
NCT01261247 (4) [back to overview]Duration of Response
NCT01261247 (4) [back to overview]Median Overall Survival Time
NCT01261247 (4) [back to overview]Median Progression-free Survival Time
NCT01282476 (3) [back to overview]Overall Response Rate
NCT01282476 (3) [back to overview]Toxicities
NCT01282476 (3) [back to overview]Progression-free Survival Rate
NCT01460940 (3) [back to overview]Assess the Safety and Tolerability of Combined Lenalidomide and Panobinostat in Patients With Previously Treated Hodgkin's Lymphoma.
NCT01460940 (3) [back to overview]Progression-free Survival in Patients With Previously Treated Hodgkin's Lymphoma Receiving Combined Lenalidomide and Panobinostat
NCT01460940 (3) [back to overview]Determine the Overall Response Rate (ORR), Including Complete Responses (CR) and Partial Responses (PR)
NCT01496118 (5) [back to overview]Number of Phase I Patients (Dose Level 1-6) Experiencing a Dose-Limiting Toxicity (DLT) to Determine the Optimal Dosage
NCT01496118 (5) [back to overview]Time-to-progression (TTP)
NCT01496118 (5) [back to overview]Progression-free-survival (PFS)
NCT01496118 (5) [back to overview]Phase II: Overall Response Rate
NCT01496118 (5) [back to overview]Overall-survival (OS)
NCT01523834 (5) [back to overview]Time to Response (TTR)
NCT01523834 (5) [back to overview]Overall Survival (OS)
NCT01523834 (5) [back to overview]Progression Free Survival (PFS)
NCT01523834 (5) [back to overview]Complete Response (CR) Rate
NCT01523834 (5) [back to overview]Overall Response Rate (ORR) at the End of the Induction Phase
NCT01582009 (5) [back to overview]6-month Overall Survival Rate
NCT01582009 (5) [back to overview]Median Progression Free Survival
NCT01582009 (5) [back to overview]Number of Participants With an Adverse Event.
NCT01582009 (5) [back to overview]Number of Participants With Clinical Response
NCT01582009 (5) [back to overview]Progression-free Survival (PFS)
NCT01651039 (8) [back to overview]Disease Control Rate for Lens Refractory Rate
NCT01651039 (8) [back to overview]Overall Response Rate for Len Refractory Patients
NCT01651039 (8) [back to overview]Response Rates
NCT01651039 (8) [back to overview]Response Rates for Len Refractory Patients
NCT01651039 (8) [back to overview]Clinical Benefit Rate
NCT01651039 (8) [back to overview]Clinical Benefit Rate for Len Refractory Patients
NCT01651039 (8) [back to overview]Disease Control Rate
NCT01651039 (8) [back to overview]The Best Overall Response Rate (ORR)
NCT01693601 (6) [back to overview]Number of Patients That Achieve Stable Disease or Clinical Improvement
NCT01693601 (6) [back to overview]Number of Participants Who Experienced Dose-Limiting (DLTs)
NCT01693601 (6) [back to overview]Percent Change in Spleen Volume
NCT01693601 (6) [back to overview]Percent Change in Spleen Size for Responders and Non-responders
NCT01693601 (6) [back to overview]Percent Change in Spleen Length
NCT01693601 (6) [back to overview]Number of Participants With Percent Change on Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF)
NCT01802879 (1) [back to overview]Overview of Adverse Events (Safety Set)
NCT02032810 (4) [back to overview]Progression Free Survival (PFS)
NCT02032810 (4) [back to overview]Immune Related Overall Response Rate (ORR)
NCT02032810 (4) [back to overview]Overall Survival (OS)
NCT02032810 (4) [back to overview]Maximum Tolerated Dose (MTD)
NCT02057640 (4) [back to overview]Number of Participants With Dose Limiting Toxicity According to CTCAE Version 4.03
NCT02057640 (4) [back to overview]Response to Combination Therapy (Panobinostat, Dexamethasone, MLN9708)
NCT02057640 (4) [back to overview]Progression-free Survival
NCT02057640 (4) [back to overview]Overall Survival
NCT02290431 (16) [back to overview]Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: CL/F
NCT02290431 (16) [back to overview]Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: AUClast, AUC0-24h, AUC0-48h, AUCinf
NCT02290431 (16) [back to overview]Time to Progression/Relapse (TTP) Per Investigator
NCT02290431 (16) [back to overview]Progression Free Survival (PFS)
NCT02290431 (16) [back to overview]Percentage of Participants With Near Complete Response (nCR)/ Complete Response (CR) Rate
NCT02290431 (16) [back to overview]Overall Survival (OS)
NCT02290431 (16) [back to overview]Overall Response Rate (ORR)
NCT02290431 (16) [back to overview]Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: Tmax
NCT02290431 (16) [back to overview]Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: T1/2
NCT02290431 (16) [back to overview]Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: Lambda_z
NCT02290431 (16) [back to overview]Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: Cmax
NCT02290431 (16) [back to overview]Minimal Response Rate (MRR) Per Investigator
NCT02290431 (16) [back to overview]Duration of Response (DOR) Per Investigator
NCT02290431 (16) [back to overview]Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: Vz/F
NCT02290431 (16) [back to overview]Time to Response (TTR) Per Investigator
NCT02290431 (16) [back to overview]Quality of Life (QoL) as Measured by FACT/GOG-Ntx Total Score
NCT02588339 (7) [back to overview]Number of Participants With Non-relapse Mortality
NCT02588339 (7) [back to overview]Number of Participants With Primary Disease Relapse
NCT02588339 (7) [back to overview]Percentage of Participants With Overall Survival (OS)
NCT02588339 (7) [back to overview]Time to Stable Engraftment
NCT02588339 (7) [back to overview]Number of Participants Stratified by Chronic Graft Versus Host Disease (GVHD) Stage
NCT02588339 (7) [back to overview]Number of Participants Stratified by Acute Graft Versus Host Disease GVHD Stage
NCT02588339 (7) [back to overview]Percentage of Participants With Relapse-free Survival (RFS)
NCT02722941 (4) [back to overview]Overall Survival (OS)
NCT02722941 (4) [back to overview]Relative Dose Intensity (RDI) Per Cohort
NCT02722941 (4) [back to overview]Progression Free Survival (PFS)
NCT02722941 (4) [back to overview]Complete Response Rate
NCT03256045 (6) [back to overview]Multiple Myeloma Cells In-vitro Drug Sensitivity to Panobinostat, by Subject
NCT03256045 (6) [back to overview]Synergy of Panobinostat and Carfilzomib in Combination, Per Subject.
NCT03256045 (6) [back to overview]Synergy of Panobinostat and Dexamethasone in Combination, Per Subject
NCT03256045 (6) [back to overview]Multiple Myeloma Cells In-vitro Drug Sensitivity to Carfilzomib, by Subject
NCT03256045 (6) [back to overview]Best Overall Response, by Subject
NCT03256045 (6) [back to overview]Multiple Myeloma Cells In-vitro Drug Sensitivity to Dexamethasone, by Subject
NCT03566199 (2) [back to overview]Proportion of Participants With Grade 3 or Higher, Treatment-related, Adverse Events
NCT03566199 (2) [back to overview]Overall Survival Rate (OS) at 12 Months

Progression-free Survival (PFS)

PFS is defined as time from first dose of study treatment to progression or death due to any cause, based on modified European Society for Bone and Marrow Transplantation (EBMT) criteria per Investigator's assessment (NCT00425555)
Timeframe: Baseline up to Cycle 12, an average of 12 months

Interventionmonths (Median)
Bexarotene Exposed127.0
Bexarotene Naive113.0
All Participants114.0

[back to top]

Maximum Plasma Concentration (Cmax) of Panobinostat

Cmax is defined as the maximum observed drug concentration observed in plasma over all PK sample concentrations. It will be obtained from the Cmax parameter calculated by WinNonlin®. If there is no measurable concentration in the subject's PK profile, then Cmax will be missing for that subject. Cmax will be reported in units of ng/mL. (NCT00425555)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng/mL (Mean)
Day 1Day 8
All Participants11.37913.490

[back to top]

Duration of Response (DOS)

Duration of response and time to response were summarized. A Kaplan-Meier analysis of time to progression was performed, including estimation and 95% confidence interval of the median time to progression and progression-free survival. (NCT00425555)
Timeframe: Baseline up to Cycle 12, an average of 12 months

InterventionDays (Median)
Bexarotene Exposed170.0
Bexarotene NaiveNA
All Participants280.0

[back to top]

Overall Response Rate of Participants Using the Modified Severity-Weighted Assessment Tool (mSWAT)

"Skin response was primarily classified based on an assessment using mSWAT, provided there was documented evidence of stable disease or better in lymph node/viscera.The mSWAT is a tool specifically developed to evaluate the extent of skin disease in CTCL (Olsen et al 2007).~Responses in the skin based on SWAT are defined as:~Complete Response (CR): no evidence of skin disease~Partial Response (PR): ≥ 50% decrease of the modified SWAT score compared with baseline~Stable Disease (SD): Neither CR, PR, or PD as compared with baseline, i.e. change from baseline is less than a 50% decrease but also less than a 25 % increase in the modified SWAT score~Progressive Disease (PD): ≥ 25% increase in the modified SWAT score compared with baseline." (NCT00425555)
Timeframe: Baseline up to 6 Months of Follow up

Interventionpercentage (Number)
Bexarotene Exposed16.7
Bexarotene Naive20.3
All Participants18.5

[back to top]

Skindex-29 Measurements of Average Sub-scores for Emotions From Baseline up to Cycle 12

"Skin index measurement (Skindex-29) was a self-administered 29-item dermatology patient-reported outcome measure that explores the domains of Functioning, Emotions and Symptoms. Data from the Skindex-29 came from the Dermatology Survey page. Scoring of the Skindex-29 was performed as per the developers' scoring algorithms. The score for each dimension was found by adding responses of 1 to 5 for each question, with higher scores indicating worse quality of life. The Skindex-29 yields three scale scores that assess emotions scores range from 29 to 116, with higher scores indicating worse health-related quality of life. Average sub-scores for emotions, physical symptoms, and functioning was displayed." (NCT00425555)
Timeframe: Baseline up to Cycle 12, an average of 12 months

,
Interventionscore on a scale (Mean)
BaselineCycle 2Cycle 3Cycle 4Cycle 5Cycle 6Cycle 7Cycle 8Cycle 9Cycle 10Cycle 11Cycle 12
Bexarotene Exposed52.248.645.940.835.834.539.339.444.643.648.342.0
Bexarotene Naive50.646.644.134.734.435.436.835.832.843.543.543.1

[back to top]

Time to Response for Responders

Time to response were summarized. A Kaplan-Meier analysis of time to progression was performed, including estimation and 95% confidence interval of the median time to progression and progression-free survival. (NCT00425555)
Timeframe: Baseline up to Cycle 12, an average of 12 months

InterventionDays (Median)
Bexarotene Exposed69.5
Bexarotene Naive85.5
All Participants82.0

[back to top]

Area Under the Plasma Concentration AUC0-24, AUC0-48 and AUC 0-infinity of Panobinostat

AUC is defined as the area under concentration-time curve as a measure of drug exposure. The area under the plasma concentration-time curve from time zero to 24 hours. The area under the plasma concentration-time curve from time zero to 48 hours. AUC (0-inf) is defined as the area under the concentration-time curve from time zero (pre-dose) extrapolated to infinite time. (NCT00425555)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng*hr/ml (Mean)
Day 1 - AUC0-24Day 8 - AUC0-24Day 1 - AUC0-48Day 8 - AUC0-48Day 1- AUC (0-inf)Day 8 - AUC (0-inf)
All Participants110.138134.033132.019159.750134.828162.673

[back to top]

The Overall Response Rate Using mSWAT Skin Score

Estimate of the response rate of participants with resistant cutaneous T-cell lymphoma (CTCL) treated with Panobinostat using the mSWAT skin scores and 95% CI will be analyzed. (NCT00425555)
Timeframe: Baseline up to Cycle 12, an average of 12 months

,,
InterventionParticipants (Count of Participants)
mSWAT skin response (CR/PR)flare considered as disease progression ( f.i.p ) - Number of respondersPhysician's Global Assessment- Responder (CR/PR)
All Participants2633
Bexarotene Exposed1215
Bexarotene Naive1418

[back to top]

Time to Peak Concentration (Tmax) of Panobinostat

Tmax is defined as the time at which the Cmax occurs. It will be obtained from the Tmax parameter calculated by WinNonlin®. If there is no measurable Cmax in the subject's PK profile, then Tmax will be missing for that subject. Tmax will be reported in units of h. (NCT00425555)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionh (hours) (Median)
Day 1Day 8
All Participants1.51.5

[back to top]

Time of Clast (Tlast) of Panobinostat

Time of Clast (Tlast) will be obtained from the Tlast parameter calculated by WinNonlin®. If there is no measurable concentration in the subject's PK profile, then Tlast will be missing for that participants. Tlast will be reported in units of h. (NCT00425555)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

InterventionHours (Median)
Day 1Day 8
All Participants47.9NA

[back to top]

Last Observed Plasma Concentration (Clast) of Panobinostat

Clast is defined as the Last observed (quantifiable) plasma concentration (Clast), in units of ng/mL. Blood samples were collected to assess Clast. (NCT00425555)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng/mL (Mean)
Day 1Day 8
All Participants0.6321.524

[back to top]

Skindex-29 Measurements of Average Sub-scores for Physical Symptoms From Baseline up to Cycle 12

"Skindex-29 was a self-administered 29-item dermatology patient-reported outcome measure that explores the domains of Functioning, Emotions and Symptoms. Data from the Skindex-29 came from the Dermatology Survey page. Scoring of the Skindex-29 was performed as per the developers' scoring algorithms. The score for each dimension was found by adding responses of 1 to 5 for each question, with higher scores indicating worse quality of life. The Skindex-29 yields three scale scores that assess physical symptoms scores range from 29 to 116, with higher scores indicating worse health-related quality of life. Average sub-scores for emotions, physical symptoms, and functioning was displayed." (NCT00425555)
Timeframe: Baseline up to Cycle 12, an average of 12 months

,
Interventionscore on a scale (Mean)
BaselineCycle 2Cycle 3Cycle 4Cycle 5Cycle 6Cycle 7Cycle 8Cycle 9Cycle 10Cycle 11Cycle 12
Bexarotene Exposed60.151.451.147.343.243.545.542.747.146.846.737.7
Bexarotene Naive55.347.742.935.235.636.237.736.535.947.048.345.1

[back to top]

Skindex-29 Measurements of Average Sub-scores for Functioning From Baseline up to Cycle 12

"Skindex-29 was a self-administered 29-item dermatology patient-reported outcome measure that explores the domains of Functioning, Emotions and Symptoms. Data from the Skindex-29 came from the Dermatology Survey page. Scoring of the Skindex-29 was performed as per the developers' scoring algorithms. The score for each dimension was found by adding responses of 1 to 5 for each question, with higher scores indicating worse quality of life. The Skindex-29 yields three scale scores that assess functioning scores range from 29 to 116, with higher scores indicating worse health-related quality of life. Average sub-scores for emotions, physical symptoms, and functioning was displayed." (NCT00425555)
Timeframe: Baseline up to Cycle 12, an average of 12 months

,
Interventionscore on a scale (Mean)
BaselineCycle 2Cycle 3Cycle 4Cycle 5Cycle 6Cycle 7Cycle 8Cycle 9Cycle 10Cycle 11Cycle 12
Bexarotene Exposed46.743.542.638.832.833.033.134.437.235.240.026.3
Bexarotene Naive44.743.339.230.731.429.528.330.528.741.538.136.2

[back to top]

Safety and Tolerability

Adverse Event (AE) are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. Serious adverse events are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards. (NCT00445068)
Timeframe: From Start of the Study up to 57 Weeks approximately.

InterventionParticipants (Count of Participants)
Participants with Adverse EventsDeathsOn treatment deathsSerious Adverse EventsStudy-drug-related Serious Adverse EventsAdverse Events Leading to discontinuation
Panobinostat38731738

[back to top]

Maximum Plasma Concentration (Cmax) of Panobinostat

Cmax is defined as the maximum observed drug concentration observed in plasma over all PK sample concentrations. It will be obtained from the Cmax parameter calculated by WinNonlin®. If there is no measurable concentration in the subject's PK profile, then Cmax will be missing for that subject. Cmax will be reported in units of ng/mL. (NCT00445068)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng/mL (Mean)
Day 1Day 8
Panobinostat7.69.7

[back to top]

Time of Clast (Tlast) of Panobinostat

Time of Clast (Tlast) will be obtained from the Tlast parameter calculated by WinNonlin®. If there is no measurable concentration in the subject's PK profile, then Tlast will be missing for that participants. Tlast will be reported in units of h. (NCT00445068)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

InterventionHours (Median)
Day 1Day 8
Panobinostat47.824.3

[back to top]

Area Under the Plasma Concentration (AUC0-24) of Panobinostat

Area under the curve (AUC) is defined as the area under concentration-time curve as a measure of drug exposure. The area under the plasma concentration-time curve from time zero to 24 hours. (NCT00445068)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng*hr/ml (Mean)
Day 1Day 8
Panobinostat72.081.6

[back to top]

Last Observed Plasma Concentration (Clast) of Panobinostat

Clast is defined as the Last observed (quantifiable) plasma concentration (Clast), in units of ng/mL. Blood samples were collected to assess Clast. (NCT00445068)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng/mL (Mean)
Day 1Day 8
Panobinostat0.31.1

[back to top]

Time to Peak Concentration (Tmax) of Panobinostat

Tmax is defined as the time at which the Cmax occurs. It will be obtained from the Tmax parameter calculated by WinNonlin®. If there is no measurable Cmax in the subject's pharmacokinetic (PK) profile, then Tmax will be missing for that subject. Tmax will be reported in units of h. (NCT00445068)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

InterventionHours (Median)
Day 1Day 8
Panobinostat1.71.2

[back to top]

QT Interval (QTc) in Participants Receiving Oral Panobinostat at Baseline and Change From Baseline to Extreme Value

QTc monitoring was performed on specified days (Cycle1: Day1, 5 and 26), as well as a single pre-dose ECG once weekly during Cycle1: Week2 and Week3, Cycle2, and all subsequent cycles. Patient eligibility was ensured by a screening QTcF interval calculated by eResearchTechnology(eRT) prior to the baseline assessments. Treatment decisions were based on QTc determined by the automated reading at the investigational site (commonly used the Bazett's correction,QTcB) or measured and calculated by trained personnel at the site. Dosing relied on the investigator's assessment of the 6 baseline ECGs (the average of the 6pre-dose QTc intervals) performed prior to Cycle1/Day1 dosing, of the 3pre-dose ECGs during Cycle1:Day5 and 26, and of the single pre-dose ECGs performed once weekly for the remaining weeks of Cycle1 and subsequent cycles. The Baseline and Change From Baseline to Extreme Value QTcF interval for analysis was calculated by eRT based on the 6 baseline ECGs obtained on Cycle1/Day1. (NCT00449761)
Timeframe: From Start of the Study up to Study Termination (approximately up to 18 Months).

Interventionms (Mean)
BaselineChange from Baseline
Panobinostat396.624.5

[back to top]

Safety and Tolerability of Panobinostat

Adverse Events (AE) are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. Serious adverse events are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards (NCT00449761)
Timeframe: From Start of the Study up to Study Termination (approximately up to 18 Months).

InterventionParticipants (Count of Participants)
Participants with Adverse EventsDeathsSerious Adverse Events
Panobinostat271613

[back to top]

Time of Clast (Tlast) of Panobinostat

Time of Clast (Tlast) will be obtained from the Tlast parameter calculated by WinNonlin®. If there is no measurable concentration in the subject's PK profile, then Tlast will be missing for that participants. Tlast will be reported in units of h. (NCT00449761)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

InterventionHours (Median)
Day 1Day 8
Panobinostat23.924.1

[back to top]

Time to Peak Concentration (Tmax) of Panobinostat

Tmax is defined as the time at which the Cmax occurs. It will be obtained from the Tmax parameter calculated by WinNonlin®. If there is no measurable Cmax in the subject's Pharmacokinetic (PK) profile, then Tmax will be missing for that subject. Tmax will be reported in units of h. (NCT00449761)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

InterventionHours (Median)
Day 1Day 8
Panobinostat1.51.5

[back to top]

Maximum Plasma Concentration (Cmax) of Panobinostat

Cmax is defined as the maximum observed drug concentration observed in plasma over all PK sample concentrations. It will be obtained from the Cmax parameter calculated by WinNonlin®. If there is no measurable concentration in the subject's PK profile, then Cmax will be missing for that subject. Cmax will be reported in units of ng/mL. (NCT00449761)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng/mL (Mean)
Day 1Day 8
Panobinostat13.520.9

[back to top]

Last Observed Plasma Concentration (Clast) of Panobinostat

Clast is defined as the Last observed (quantifiable) plasma concentration (Clast), in units of ng/mL. Blood samples were collected to assess Clast. (NCT00449761)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng/mL (Mean)
Day 1Day 8
Panobinostat1.94.7

[back to top]

Area Under the Plasma Concentration (AUC0-24) of Panobinostat

Area under the curve (AUC) is defined as the area under concentration-time curve as a measure of drug exposure. The area under the plasma concentration-time curve from time zero to 24 hours. (NCT00449761)
Timeframe: Pre-dose, 0.25, 1-2, and 3-4 hours post dose on Day 1 and Day 8

Interventionng.hr/mL (Mean)
Day 1Day 8
Panobinostat139148

[back to top]

Participants With Hematologic Response

The primary efficacy variable was hematologic response, a composite endpoint defined as the overall of complete hematologic response (CHR), and of no evidence of leukemia (NEL) and of the return to chronic phase (RTC). (NCT00449761)
Timeframe: From Start of the Study up to Study Termination (approximately up to 18 Months).

InterventionParticipants (Count of Participants)
Panobinostat0

[back to top]

BCR-ABL Mutations of Participants at Study Entry and, in Responding Participants and at the Time of Disease Progression

A fusion gene of the BCR and ABL genes (BCR-ABL) messenger ribose nucleic acid (mRNA) expression (molecular response) was performed by quantitative polymerase chain reaction (qPCR) and mutational analysis was performed by direct sequencing technology, and both analyses were performed by Genzyme. (NCT00449761)
Timeframe: From Start of the Study up to Study Termination (approximately up to 18 Months).

InterventionParticipants (Count of Participants)
Panobinostat0

[back to top]

QT Interval (QTc) in Participants Receiving Oral Panobinostat at Baseline and Change From Baseline to Extreme Value

QTc monitoring was performed on specified days (Cycle1: Day1, 5 and 26), as well as a single pre-dose ECG once weekly during Cycle1: Week2 and Week3, Cycle2, and all subsequent cycles. Patient eligibility was ensured by a screening QTcF interval calculated by eResearchTechnology(eRT) prior to the baseline assessments. Treatment decisions were based on QTc determined by the automated reading at the investigational site (commonly used the Bazett's correction,QTcB) or measured and calculated by trained personnel at the site. Dosing relied on the investigator's assessment of the 6 baseline ECGs (the average of the 6pre-dose QTc intervals) performed prior to Cycle1/Day1 dosing, of the 3pre-dose ECGs during Cycle1:Day5 and 26, and of the single pre-dose ECGs performed once weekly for the remaining weeks of Cycle1 and subsequent cycles. The Baseline and Change From Baseline to Extreme Value QTcF interval for analysis was calculated by eRT based on the 6 baseline ECGs obtained on Cycle1/Day1. (NCT00451035)
Timeframe: From Start of the Study up to End of Study (approximately up to 19 Months)

Interventionms (Mean)
BaselineChange from Baseline
Panobinostat (LBH589)403.129.1

[back to top]

Time of Clast (Tlast) of Panobinostat

Time of last sampling point. Tlast will be reported in units of hr (NCT00451035)
Timeframe: Pre-dose, and 0.25, 1-2, 3-4, 24, and 48 hours post-dose on Day 1

Interventionhr (Mean)
Panobinostat (LBH589)24.7

[back to top]

Area Under the Plasma Concentration (AUC0-24) of Panobinostat

Area under the curve (AUC) is defined as the area under concentration-time curve as a measure of drug exposure. The area under the plasma concentration-time curve from time zero to 24 hours. (NCT00451035)
Timeframe: Pre-dose, and 0.25, 1-2, 3-4, 24, and 48 hours post-dose on Day 1

Interventionng*hr/mL (Mean)
Panobinostat (LBH589)96.0

[back to top]

Safety and Tolerability Profile of Oral Panobinostat

Adverse events (AEs) are defined as any unfavorable and unintended diagnosis, symptom, sign (including an abnormal laboratory finding), syndrome or disease which either occurs during study, having been absent at baseline, or, if present at baseline, appears to worsen. Serious adverse events (SAEs) are any untoward medical occurrences that result in death, are life threatening, require (or prolong) hospitalization, cause persistent or significant disability/incapacity, result in congenital anomalies or birth defects, or are other conditions which in judgment of investigators represent significant hazards. (NCT00451035)
Timeframe: From Start of the Study up to 28 Days After the last dose of Study Drug (approximately up to 19 Months)

InterventionParticipants (Count of Participants)
AEsSAEsDeathsAEs leading to discontinuation
Panobinostat (LBH589)28436

[back to top]

BCR-ABL Mutations of Participants at Study Entry and, in Responding Participants and at the Time of Disease Progression

BCR-ABL messenger ribose nucleic acid (mRNA) expression (molecular response) was performed by quantitative polymerase chain reaction (qPCR) and mutational analysis was performed by direct sequencing technology, and both analyses were performed by Genzyme. (NCT00451035)
Timeframe: From Start of the Study up to End of Study (approximately up to 19 Months)

InterventionParticipants (Count of Participants)
Panobinostat (LBH589)0

[back to top]

Last Observed Plasma Concentration (Clast) of Panobinostat

Clast is defined as the Last observed (quantifiable) plasma concentration at last sampling time. (NCT00451035)
Timeframe: Pre-dose, and 0.25, 1-2, 3-4, 24, and 48 hours post-dose on Day 1

Interventionng/mL (Mean)
Panobinostat (LBH589)1.8

[back to top]

Major (Complete/Partial) Cytogenetic Response (MCyR) Rate

The CyR, based on the percentage of Ph+ metaphases by karyotype analysis on a bone marrow aspirate, was ideally assessed from a minimum of 20 metaphases in each bone marrow sample. The CyR was defined as: major response including complete (CCyR; 0% Ph+ metaphases) or partial (PCyR; 1 to 35% Ph+ metaphases), minor (36 to 65% Ph+ metaphases), minimal (66 to 95% Ph+ metaphases) or none (96 to 100% Ph+ metaphases). (NCT00451035)
Timeframe: From Start of the Study up to End of Study (approximately up to 19 Months)

InterventionParticipants (Count of Participants)
Panobinostat (LBH589)0

[back to top]

Maximum Plasma Concentration (Cmax) of Panobinostat

Cmax is defined as the Maximum (peak) plasma drug concentration after single dose administration. Cmax will be reported in units of nanogram/milliliter (ng/ML). (NCT00451035)
Timeframe: Pre-dose, and 0.25, 1-2, 3-4, 24, and 48 hours post-dose on Day 1

Interventionng/mL (Mean)
Panobinostat (LBH589)9.1

[back to top]

Time to Peak Concentration (Tmax) of Panobinostat

Time to reach peak or maximum plasma drug concentration following drug administration. Tmax will be reported in units of hour (hr). (NCT00451035)
Timeframe: Pre-dose, and 0.25, 1-2, 3-4, 24, and 48 hours post-dose on Day 1

Interventionhr (Median)
Panobinostat (LBH589)1.5

[back to top]

Number of Participants Experiencing ≥Grade 2 Adverse Events

An adverse event (AE) is the development of an undesirable medical condition, or the deterioration of a preexisting medical condition (other than the condition that is being treated by the trial) following or during exposure to a pharmaceutical product, whether or not considered causally related to the product. The number of participants experiencing such adverse events that are related to the study drug are reported here. (NCT00550277)
Timeframe: 18 months

InterventionParticipants (Count of Participants)
Treatment15

[back to top]

Number of Participants With Overall Response

"Response was evaluated in this study using the new international criteria proposed by the Response Evaluation Criteria in Solid Tumors (RECIST) Committee. Overall response is defined as the proportion of participants whose disease either decreased (partial response- PR) or disappeared (Complete response - CR).~Complete Response (CR): Disappearance of all target lesions. Partial Response (PR): At least a 30% decrease in the sum of the longest diameter (LD) of target lesions, taking as reference the baseline sum LD" (NCT00550277)
Timeframe: 18 months

InterventionParticipants (Count of Participants)
Treatment0

[back to top]

Progression-free Survival

Progression-free survival was defined as the interval from the date of first treatment with panobinostat until the date that disease progression or death occurred. Progressive disease (PD): 20% increase in the sum of diameters of target measurable lesions above the smallest sum observed (over baseline if no decrease in the sum is observed during therapy), with a minimum absolute increase of 5 mm. (NCT00550277)
Timeframe: 18 months

Interventionmonths (Median)
Treatment1.7

[back to top]

Number of Participants With Best Overall Response

Tumors were assessed according to Response Evaluation Criteria in Solid tumors (RECIST). Complete response (CR): disappearance of all lesions (i.e. all evidence of disease, not just the target lesions) determined by 2 observations not less than 4 weeks apart; Partial response (PR): > 30% decrease in the sum of longest diameters of target lesions compared to baseline, with response or stable disease observed in non-target lesions, and no new lesions; Stable disease (SD): neither sufficient shrinkage to qualify for response or sufficient increase to qualify for progressive disease in target lesions, with response or stable disease observed in non-target lesions, and no new lesions; Progressive disease (PD): > 20% increase in the sum of longest diameters of target lesions compared to smallest sum longest diameter recorded. In addition, the sum must also demonstrate an absolute increase of at least 5mm. (NCT00567879)
Timeframe: day 21

,,,,,
InterventionParticipants (Number)
Complete responsePartial responseStable diseaseProgressive diseaseUnknown
Escalation: i.v. Arm - 10mg/m^200430
Escalation: i.v. Arm - 15mg/m^200160
Escalation/Expansion: i.v. Arm -20mg/m^201893
Oral Arm - Schedule A 20mg00141
Oral Arm - Schedule B 15 mg00372
Oral Arm - Schedule B 20mg00030

[back to top]

Number of Participants With Dose Limiting Toxicities (DLTs)

Safety data was reviewed to determine the DLTs. DLTs comprised adverse events (AEs) or abnormal laboratory values that occurred at any time and were assessed as clinically relevant and meeting any of the following criteria: considered to be related to the study treatment and unrelated to disease, disease progression, inter-current illness, or concomitant medications. Toxicities were assessed using the National Cancer Institute common terminology criteria for adverse events (NCI CTCAE), version 3.0. Disease related symptoms were not considered a DLT. (NCT00567879)
Timeframe: day 21

InterventionParticipants (Number)
Escalation: i.v. Arm - 10mg/m^20
Escalation: i.v. Arm - 15mg/m^20
Escalation/Expansion: i.v. Arm -20mg/m^21
Expansion: i.v. Arm -20mg/m^21
Oral Arm - Schedule A 20mg1
Oral Arm - Schedule B 15 mg3
Oral Arm - Schedule B 20mg1

[back to top]

Overall Response Rate (CR, Marrow CR + PR) of LBH in Patients With Relapsed or Refractory MDS.

Overall response rate (ORR) is defined by the modified International Working Group (IWG) Response Criteria for MDS. In the marrow, Complete Response (CR) is <= 5% blasts present with normal maturation of all cell lines. In peripheral blood, CR is defined as hemoglobin >= 11 g/dL, ANC >= 1000/mL, and platelets >= 100,000 with 0% blasts present. Partial Response (PR) is defined the same as CR with blasts decreased by >= 50% and >= 5% blasts in the marrow. (NCT00594230)
Timeframe: Every 8 weeks up to 24 months on-study.

Interventionparticipants (Number)
Panobinostat 30 mg0
Panobinostat 20 mg0

[back to top]

Safety and Tolerability of LBH589 in Patients With Relapsed/Refractory MDS by Measuring the Number of Participants With Adverse Events

The reported incidence of AEs and SAEs with an onset on or after the initiation of therapy was graded according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) version 4.0 (NCT00594230)
Timeframe: 24 months

InterventionParticipants (Count of Participants)
Panobinostat 30 mg15
Panobinostat 20 mg9

[back to top]

Number of Participants DLT in Arm 2 in Dose Escalation Phase

"Maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) for intermittent dosing schedule (MWF weekly).~A 3-parameter version of a Bayesian logistic regression model with overdose control (Babb, Rogatko, and Zacks 1998) was used during the dose escalation phase for dose level selection and determination of the MTD." (NCT00621244)
Timeframe: Cycle 1 (28-day treamtent cycle)

InterventionParticipants (Number)
Arm 2, Group X (30 mg)0
Arm 2, Group X (45 mg)0
Arm 2, Group X (60 mg)0
Arm 2, Group X (80 mg)4
Arm 2, Group Y (30 mg)0
Arm 2, Group Y (45 mg)0
Arm 2, Group Y (60 mg) - MTD3

[back to top]

Number of Participants DLT in Arm 1 in Dose Escalation Phase

Maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) for consecutive dosing schedule (MWF weekly). A 3-parameter version of a Bayesian logistic regression model with overdose control (Babb, Rogatko, and Zacks 1998) was used during the dose escalation phase for dose level selection and determination of the MTD. (NCT00621244)
Timeframe: Cycle 1 (28-day treatment cycle)

InterventionParticipants (Number)
Arm 1, Group X (20 mg)0
Arm 1, Group X (30 mg)0
Arm 1, Group X (40 mg)2
Arm 1, Group X (60 mg) - MTD1
Arm 1, Group X (80 mg)4
Arm 1, Group Y (20 mg)0
Arm 1, Group Y (30 mg)0
Arm 1, Group Y (40 mg) - MTD5
Arm 1, Group Y (60 mg)4

[back to top]

Maximum Plasma Concentration of Panobinostat After the First Dose in Arms 1 and 2

(NCT00621244)
Timeframe: Day 1

Interventionng/mL (Mean)
20 mg19.5
30 mg39.8
40 mg58
45 mg54
60 mg66.9
80 mg63.5

[back to top]

Half Life of Panobinostat After Multiple Doses in Arm 1 on Day 15

(NCT00621244)
Timeframe: Day 15

Interventionhour (Mean)
20 mg20.1
30 mg19.7
40 mg21.4
60 mg17.9
80 mg17.7

[back to top]

Percentages of Participants With Histone Acetylation Induction in Peripheral Blood in Arm 1 (MWF Every Week), Group X

Reporting the number of patients with a reading at the timepoint in the dose group. (NCT00621244)
Timeframe: Days 1, 5, 8, 10, 15

,,,,
InterventionPercentages of participants (Number)
Day 1 (total=0, 1, 0, 0, 0)Day 5 (total = 6, 4, 8, 17, 8)Day 8 (total = 5, 4, 8, 17, 7)Day 10 (total=0, 0, 0, 0, 0, 1)Day 15 (total=3, 3, 4, 2, 2)
Arm 1, Group X (20 mg)NA100.0100.0NA66.7
Arm 1, Group X (30 mg)100.075.0100.0NA33.3
Arm 1, Group X (40 mg)NA87.575.0NA50.0
Arm 1, Group X (60 mg)NA82.482.4NA50.0
Arm 1, Group X (80 mg)NA37.557.1100.050.0

[back to top]

Maximum Plasma Concentration of Panobinostat After Multiple Doses in Arm 1 on Day 15

From day 15 by dose with schedule: MWF every week (NCT00621244)
Timeframe: Day 15

Interventionng/mL (Mean)
20 mg33.6
30 mg38.4
40 mg41.6
60 mg51.8
80 mg69.6

[back to top]

Highest Percent Change of Fetal Hemoglobin From Baseline in Arm 2 (MWF Every Other Week)

All blood samples were drawn immediately prior to each administration of LBH589 dose and at the end of treatment (≤ 7 days post last dose (preferably ≥ 4 days [96 hours])) (NCT00621244)
Timeframe: Post dose to pre-dose (up to 3.5 years)

InterventionPercent Change (Mean)
Arm 2, Group X (30 mg)96.2
Arm 2, Group X (45 mg)67.7
Arm 2, Group X (60 mg)59.3
Arm 2, Group X (80 mg)34.2
Arm 2, Group Y (30 mg)0.0
Arm 2, Group Y (45 mg)200.7
Arm 2, Group Y (60 mg)376.0

[back to top]

Percentage of Participants With Histone Acetylation Induction in Peripheral Blood in Arm 2 (MWF Every Other Week), Group Y

(NCT00621244)
Timeframe: Days 5, 8, 10, 12, 15, End of study (up to 3.5 years)

,,
InterventionPercentages of participants (Number)
Day 5 (total=1, 4, 6)Day 8 (total = 1, 4, 6)Day 10 (total=1, 4, 5)Day 12 (Total=0, 3, 5)Day 15 (Total=1, 3, 5)End of Study (Total=0, 2, 0)
Arm 2, Group Y (30 mg)100.0100.0NANA100.0NA
Arm 2, Group Y (45 mg)100.0100.050.033.333.350.0
Arm 2, Group Y (60 mg)100.083.360.040.00NA

[back to top]

Highest Percent Change in Fetal Hemoglobin From Baseline in Arm 1 (MWF Every Week)

All blood samples were drawn immediately prior to each administration of LBH589 dose and at the end of treatment (≤ 7 days post last dose (preferably ≥ 4 days [96 hours])) (NCT00621244)
Timeframe: Post dose to pre-dose (up to 3.5 years)

InterventionPercent Change (Mean)
Arm 1, Group X (20 mg)56.6
Arm 1, Group x (30 mg)22.5
Arm 1, Group X (40 mg)63.2
Arm 1, Group X (60 mg)591.4
Arm 1, Group X (80 mg)31
Arm 1, Group Y (20 mg)66.7
Arm 1, Group Y (30 mg)150.0
Arm 1, Group Y (40mg)196.3
Arm 1, Group Y (60 mg)1998.5

[back to top]

Half Life of Panobinostat After the First Dose in Arms 1 and 2

(NCT00621244)
Timeframe: Day 1

Interventionhour (Mean)
20 mg13.8
30 mg18.2
40 mg13.6
45 mg19.7
60 mg15.4
80 mg14.6

[back to top]

Response as Per Investigator Assessment for Patients With Myelodysplastic Syndromes (MDS)

Response as per investigator assessment for patients include complete response, stable disease, progressive disease/failure, partial remission. (NCT00621244)
Timeframe: 3.5 years

,
InterventionParticipants (Number)
Complete response (CR)Stable disease (SD)Progressive disease (PD)/failureMissingPartial remission (PR)
Arm 1, Group X04410
Arm 2, Group X01001

[back to top]

Percentages of Participants With Histone Acetylation Induction in Peripheral Blood in Arm 2 (MWF Every Other Week), Group X

(NCT00621244)
Timeframe: Days 5, 8, 10, 12, 15, End of study, Unscheduled (up to 3.5 years)

,,,
InterventionPercentages of participants (Number)
Day 5 (total=4, 9, 6, 8)Day 8 (total = 4, 7, 5, 7)Day 10 (Total=3, 8, 5, 7)Day 12 (Total=4, 6, 4, 8)Day 15 (Total=4, 6, 2, 5)End of Study (Total=1, 4, 2, 0)Unscheduled (Total=2, 0, 0, 0)
Arm 2, Group X (30 mg)50.050.033.325.075.0100.050.0
Arm 2, Group X (45 mg)88.9100.037.5NA50.075.0NA
Arm 2, Group X (60 mg)83.380.080.050.0100.050.0NA
Arm 2, Group X (80 mg)100.0100.0100.050.060.00.0NA

[back to top]

Response as Per Investigator Assessment for Patients With Hodgkin's Lymphoma (HD)

Response as per investigator assessment for patients include complete response, partial remission, stable disease, progressive disease (PD)/failure. (NCT00621244)
Timeframe: 3.5 years

,
InterventionParticipants (Number)
Complete response (CR)Partial remission (PR)Stable disease (SD)Progressive disease (PD)/failureMissing
Arm 1, Group Y141115
Arm 2, Group Y13420

[back to top]

Response as Per Investigator Assessment for Patients With Acute Myelogenous Leukemia (AML) in Expansion Phase

Stage 2 did not open for enrollment. (NCT00621244)
Timeframe: 1.2 years

InterventionParticipants (Number)
Complete response (CR)Progressive disease (PD)/failureStable disease (SD)Not evaluableMissing
Arm 1, Group X15625

[back to top]

Geometric Mean Ratio (GMR) Comparing Treatment Days in Arm 1

MWF Every week schedule n = number of subjects with non-missing values. (NCT00621244)
Timeframe: Day 15/day 1

,,,,
InterventionRatio (Geometric Mean)
AUCC (max) N=8,12,18,17,4
20 mg2.161.86
30 mg1.071.02
40 mg0.980.63
60 mg1.170.74
80 mg1.121.41

[back to top]

Percentages of Participants With Histone Acetylation Induction in Peripheral Blood in Arm 1 (MWF Every Week), Group Y

(NCT00621244)
Timeframe: Days 5, 8, end of study (up to 3.5 years)

,,,
InterventionPercentages of participants (Number)
Day 5 (total= 1, 2, 15, 4)Day 8 (total = 1, 3, 11, 4)End of Study (total=1, 0, 0, 0)
Arm 1, Group Y (20 mg)100.0100.00.0
Arm 1, Group Y (30 mg)100.0100.0NA
Arm 1, Group Y (40 mg)66.772.7NA
Arm 1, Group Y (60 mg)75.075.0NA

[back to top]

Response as Per Investigator Assessment for Patients With Acute Myelogenous Leukemia (AML)

Response as per investigator assessment for patients include complete response, progressive disease/failure, stable disease. (NCT00621244)
Timeframe: 3.5 years

,
InterventionParticipants (Number)
Complete response (CR)Partial remission (PR)Stable disease (SD)Progressive disease (PD)/failureNot evaluableMissing
Arm 1, Group X212517311
Arm 2, Group X0110418

[back to top]

To Determine the Maximum Tolerated Doses (MTD) and Dose-limiting Toxicities (DLT) of LBH589 in Combination With Capecitabine When Administered to Patients With Refractory and Advanced Tumor Types That Are Sensitive to 5-fluorouracil

MTD for Capecitabine, BID (NCT00632489)
Timeframe: 18 months

Interventionmg/m2 (Number)
LBH589 With Capecitabine100

[back to top]

To Determine the Maximum Tolerated Doses (MTD) and Dose-limiting Toxicities (DLT) of LBH589 in Combination With Capecitabine When Administered to Patients With Refractory and Advanced Tumor Types That Are Sensitive to 5-fluorouracil

MTD for Panobinostat, twice weekly (NCT00632489)
Timeframe: 18 months

Interventionmg (Number)
LBH589 With Capecitabine30

[back to top]

Percentage of Participants With PSA Progression Rate at 24 Weeks

The PSA progression was defined as a 50% rise from nadir and a minimum rise of 2 nanogram per milligram (ng/mL). Disease Progression as per RECIST criteria: Measurable lesions: If target lesion was lymph node then it had to be at least 2 cm at baseline to assess change in size. Bone lesions: (non-target lesions) appearance of >=2 unequivocal new lesions confirmed on a second scan at least 6 weeks later. (NCT00667862)
Timeframe: 24 weeks

Interventionpercentage of participants (Number)
Panobinostat48.6

[back to top]

Number of Participants With Adverse Events (AEs) and Serious Adverse Events (SAEs)

An AE was an adverse medical event which occurs in a participant of the study and which is not necessarily in a causal relationship with the treatment the participant receives. SAEs were AEs leading to death, are life-threatening, require hospitalizations or prolongation of hospitalizations, represent an innate malformation or a congenital abnormality. (NCT00667862)
Timeframe: From start of the study up to at least 4 weeks following the last dose of study treatment (Up to approximately 2.7 years)

InterventionParticipants (Count of Participants)
AEsSAEs
Panobinostat3514

[back to top]

Percentage of Participants With Tumor Response Rate

The tumor response rate was defined as a percentage of participants with confirmed Complete Response (CR) or Partial Response (PR) per RECIST criteria. CR: Disappearance of all target lesions and all nontarget lesions. PR: At least a 30% decrease in the sum of the longest diameter of all target lesions, taking as reference the baseline sum of the longest diameters. (NCT00667862)
Timeframe: Every 12 weeks up to approximately 2.7 years

Interventionpercentage of participants (Number)
Panobinostat0

[back to top]

Percentage of Participants With Progression-Free Survival (PFS) Rate at 24 Weeks

The PFS rate was defined as the percentage of participants that were alive without documented disease progression at the end of 24 weeks from first study treatment. Disease Progression as per response evaluation criteria in solid tumors (RECIST) criteria: Measurable lesions: If target lesion was lymph node then it had to be at least 2 centimeter (cm) at baseline to assess change in size. Bone lesions: (non-target lesions) appearance of greater than or equal to (>=) 2 unequivocal new lesions confirmed on a second scan at least 6 weeks later. (NCT00667862)
Timeframe: 24 weeks

Interventionpercentage of participants (Number)
Panobinostat11.4

[back to top]

Percentage of Participants With Prostate Specific Antigen (PSA) Response Rate at 24 Weeks

The PSA response was defined as a 50% decrease in PSA from baseline maintained for >= 4 weeks, and without clinical or radiographic evidence of disease progression during this time period. Disease Progression as per RECIST criteria: Measurable lesions: If target lesion was lymph node then it had to be at least 2 cm at baseline to assess change in size. Bone lesions: (non-target lesions) appearance of >= 2 unequivocal new lesions confirmed on a second scan at least 6 weeks later. (NCT00667862)
Timeframe: 24 weeks

Interventionpercentage of participants (Number)
Panobinostat0

[back to top]

Overall Survival

Overall survival is defined as the date of first dose of study drug to the date of death from any cause. (NCT00691938)
Timeframe: Completion of follow-up (median follow-up was 58 months)

Interventionmonths (Median)
Level 1-Phase II (All Patients Enrolled)6.44

[back to top]

Event-free Survival

Event-free survival is defined as the interval from the date of first dose of study drug to date of treatment failure, relapse from CR, or death due to any cause. (NCT00691938)
Timeframe: Completion of follow-up (median follow-up was 58 months)

Interventiondays (Median)
Level 1-Phase II (All Patients Enrolled)104

[back to top]

Rate of Cytogenetic Complete Remission (CRc)

Cytogenetic complete remission (CRc). A CRc will be defined by the achievement of a CR with reversion to a normal karyotype in a minimum of 20 metaphases analyzed by cytogenetics. (NCT00691938)
Timeframe: Up to 12 months

Interventionpercentage of participants (Number)
Level 1-Phase II (All Patients Enrolled)3.9

[back to top]

Time to Response

Time to response is defined as the date of the first dose of study drug to the date that all criteria for CR or CRi are fulfilled. (NCT00691938)
Timeframe: Up to 12 months

Interventiondays (Median)
Level 1-Phase II (All Patients Enrolled)91.5

[back to top]

Rates of Morphologic Complete Remission With Incomplete Count Recovery (CRi)

Morphologic complete remission with incomplete blood count recovery (CRi): Achievement of all of the criteria for CR except for residual neutropenia (< 1,000/μL) or thrombocytopenia (< 100,000/μL). (NCT00691938)
Timeframe: Up to 12 months

Interventionpercentage of participants (Number)
Level 1-Phase II (All Patients Enrolled)3.9

[back to top]

Remission Duration

Defined as the first date that all criteria for CR, CRi or HI are fulfilled to the date of treatment failure, relapse from CR, or death due to any cause. (NCT00691938)
Timeframe: Completion of follow-up (median follow-up was 58 months)

Interventiondays (Median)
Level 1-Phase II (All Patients Enrolled)361

[back to top]

Rate of Hematologic Improvement.

"-Hematologic improvement (HI). Includes the following categories:~Erythroid response: Hemoglobin increase by ≥ 1.5 g/dL over baseline in which the pretreatment hemoglobin is < 11 g/dL or reduction of RBC transfusions of at least 4 RBC transfusions / 8 wk compared with the pretreatment transfusion number in the previous 8 weeks.~Platelet response. Absolute increase of > 30 x 10^9/L for patients starting with 20 x 10^9/L or increase from < 20 x 10^9/L to > 20 x 109/L and by at least 100%~Neutrophil response. At least 100% increase and an absolute increase > 0.5 x 109/L for patients with pretreatment neutrophils < 1.0 x 109/L)" (NCT00691938)
Timeframe: Up to 12 months

Interventionpercentage of participants (Number)
Level 1-Phase II (All Patients Enrolled)2.0

[back to top]

Phase II: Overall Rate of Morphologic Complete Remission (CR) + Cytogenetic Complete Remission (CRc) + Morphologic Complete Remission With Incomplete Blood Count Recovery (CRi)

"Morphologic complete remission (CR). A CR designation requires that the patient achieve the morphologic leukemia-free state with less than 5% blasts in an aspirate sample with marrow spicules and with a count of at least 200 nucleated cells. There should be no blasts with Auer rods or persistence of extramedullary disease. Patients must also have an absolute neutrophil count of more than 1,000/μLand platelets of 100,000/μL.~Cytogenetic complete remission (CRc). A CRc will be defined by the achievement of a CR with reversion to a normal karyotype in a minimum of 20 metaphases analyzed by cytogenetics.~Morphologic complete remission with incomplete blood count recovery (CRi): Achievement of all of the criteria for CR except for residual neutropenia (< 1,000/μL) or thrombocytopenia (< 100,000/μL)." (NCT00691938)
Timeframe: Up to 12 months

Interventionpercentage of participants (Number)
Level 1-Phase II (All Patients Enrolled)11.8

[back to top]

Phase I: Maximum Tolerated Dose (MTD) of LBH589 When Given in Combination With Decitabine

(NCT00691938)
Timeframe: Completion of Phase I enrollment for MTD (approximately 26 months)

Interventionmg (level 5b dosing schedule) (Number)
Phase I (Includes Levels 1-5)40

[back to top]

Safety and Tolerability of Regimen as Measured by the Rate of the Most Common Adverse Events Experienced

Adverse events will be assessed according to the Common Toxicity Criteria for Adverse Events (CTCAE) version 3.0. (NCT00691938)
Timeframe: Up to 13 months after start of treatment

Interventionpercentage of participants (Number)
FatigueFebrile neutropeniaDiarrheaNausea
Level 1-Phase II (All Patients Enrolled)88767569

[back to top]

Hematological Response Rate

Morphologic CR: morphologic leukemia-free state with absolute neutrophil count > 1000/uL and platelet count ≥ 100,000/uL and independent of blood transfusions. Cytogenic CR: morphologic CR along with reversion to a normal karyotype by cytogenetic analysis. Molecular CR: morphologic CR with no residual disease by molecular or flow cytometric detection methods. Morphologic CR with incomplete blood recovery (CRi): morphologic CR except for residual neutropenia (<1000/uL) and/or thrombocytopenia (<1000,000/uL). PR: same hematologic values for a CR but with a decrease of at least 50% in percentage of blasts to a post-treatment value of 5% to 25% in bone marrow aspirate. (If the pre-treatment blast percentage was 50-100% this must decrease to a value between 5-25%. If the pre-treatment blast percentage was 20-49% this must decrease by at least half to a value > 5%.) A value ≤ 5% is also considered a PR if Auer rods are present. Hematological response = morphologic CR+PR. (NCT00723203)
Timeframe: Up to 6 cycles of treatment, up to 24 weeks.

Interventionpercentage of responding participants (Number)
Treatment (Panobinostat)0

[back to top]

Maximum Observed Concentration (Cmax) of Panobinostat

(NCT00742027)
Timeframe: Cycle 1, Day 1: Pre-dose, 0.25, 1, 3, 5, 7, 24, and 28 hours post-dose

Interventionnanograms per milliliter (ng/mL) (Mean)
Panobinostat41.88

[back to top]

Percentage of Participants With Adverse Events (AEs), Serious Adverse Events (SAEs), And Deaths as a Measure of Safety and Tolerability of Panobinostat

An AE is the appearance of (or worsening of any pre-existing) undesirable sign(s), symptom(s), or medical condition(s) occurring after signing the informed consent even if the event is not considered to be related to the study drug(s). SAEs are AEs leading to death, are life-threatening, require hospitalizations or prolongation of hospitalizations, represent an innate malformation or a congenital abnormality. (NCT00742027)
Timeframe: Up to approximately 5 years

Interventionpercentage of participants (Number)
AEsSAEsDeaths
Panobinostat10039.545.0

[back to top]

Area Under the Plasma Concentration-Time Curve From Time Zero to 28 Hours (AUC0-28) for Panobinostat

(NCT00742027)
Timeframe: Cycle 1, Day 1: Pre-dose, 0.25, 1, 3, 5, 7, 24, and 28 hours post-dose

Interventionhour*nanograms per milliliter (h.ng/mL) (Mean)
Panobinostat233.38

[back to top]

Area Under the Plasma Concentration-Time Curve From Time Zero to Infinity (AUC0-∞) for Panobinostat

(NCT00742027)
Timeframe: Cycle 1, Day 1: Pre-dose, 0.25, 1, 3, 5, 7, 24, and 28 hours post-dose

Interventionh.ng/mL (Mean)
Panobinostat239.36

[back to top]

Duration of Overall Disease Response

Duration of overall response (CR or PR) was defined as the time from the date of first documented disease response (CR or PR) to the date of first documented progression or death due to lymphoma. Per Cheson response criteria, CR= is a complete normalization of all index nodal and extranodal lesions and complete disappearance of all extranodal lesions. PR= is a 50% decrease in the SPD for up to 6 identified dominant lesions, including splenic and hepatic nodules from baseline. Participants were considered responders if they had a partial or complete response to a treatment while in the study. (NCT00742027)
Timeframe: From the start of treatment up to approximately 5 years

Interventionweeks (Median)
Panobinostat30.1

[back to top]

Objective Response Rate (ORR) as Assessed by the Investigator Based on Cheson Response Criteria

ORR was number of participants with best overall disease response of complete response(CR)/partial response(PR).Best overall disease response was best disease response recorded from start of treatment until disease progression/recurrence.CR=complete normalization of all index nodal,extranodal lesions,complete disappearance of all extranodal lesions.PR=50% decrease in SPD for up to 6 identified dominant lesions (splenic,hepatic nodules) from baseline.Stable=neither sufficient shrinkage to qualify for PR nor sufficient increase for progressive disease (PD), reference smallest sum diameters while on study.Progression=50% increase in sum of longest diameter of all target lesions,from smallest sum of longest diameter of all target lesions recorded at or after baseline/a new lesion/progression of non-target lesions.Unknown is progression not documented,one/more of index lesions not assessed or have been assessed using a different method than baseline at the time of radiologic evaluation. (NCT00742027)
Timeframe: From the start of the treatment of last participant up to 32 weeks

InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
Panobinostat53071149

[back to top]

Response Rate Based on Central Review of Computed Tomography (CT) Scan/Magnetic Resonance Imaging (MRI)

Best overall radiological response (CT/MRI) was recorded from start of treatment until progression/ recurrence. CR=complete normalization of all index nodal,extranodal lesions,complete disappearance of all extranodal lesions.PR=50% decrease in SPD for up to 6 identified dominant lesions (splenic,hepatic nodules) from baseline.Stable=neither sufficient shrinkage to qualify for PR nor sufficient increase for progressive disease (PD), reference smallest sum diameters while on study.Progression=50% increase in sum of longest diameter of all target lesions,from smallest sum of longest diameter of all target lesions recorded at or after baseline/a new lesion/progression of non-target lesions.Unknown is progression not documented,one/more of index lesions not assessed or have been assessed using a different method than baseline at the time of radiologic evaluation. (NCT00742027)
Timeframe: From start of treatment until progression/recurrence or start of a new cancer therapy (up to approximately 5 years)

Interventionpercentage of participants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive DiseaseUnknown
Panobinostat0.820.956.615.56.2

[back to top]

Progression Free Survival (PFS)

Progression-free survival (PFS) was defined as the time from the date of randomization/start of treatment to the date of event defined as the first documented progression or death due to any cause. If a participant has not had an event, progression-free survival was censored at the date of the last adequate assessment. (NCT00742027)
Timeframe: From the start of treatment up to approximately 5 years

Interventionmonths (Median)
Panobinostat6.1

[back to top]

The Overall Survival (OS)

OS was the duration from date of randomization to date of death from any cause. If a participant has not had an event, overall survival was censored at the date of the last adequate assessment. (NCT00742027)
Timeframe: Baseline to date of death from any cause (up to approximately 5 years)

Interventionmonths (Median)
Panobinostat34.9

[back to top]

The Time to Reach Maximum Plasma Concentration (Tmax) of Panobinostat

(NCT00742027)
Timeframe: Cycle 1, Day 1: Pre-dose, 0.25, 1, 3, 5, 7, 24, and 28 hours post-dose

Interventionhours (Median)
Panobinostat1.1

[back to top]

Time To Overall Disease Response in Responders

Time to overall disease response (CR or PR) was defined as the time from the date of randomization/start of treatment to the date of first documented disease response (PR or CR). Per Cheson response criteria, CR= is a complete normalization of all index nodal and extranodal lesions and complete disappearance of all extranodal lesions. PR= is a 50% decrease in the SPD for up to 6 identified dominant lesions, including splenic and hepatic nodules from baseline. Participants were considered responders if they had a partial or complete response to a treatment while in the study as per Investigator's assessment. (NCT00742027)
Timeframe: From the start of treatment up to approximately 5 years

Interventionweeks (Median)
Panobinostat9.9

[back to top]

Maximum Tolerated Dose (MTD)

Phase 1: to determine the MTD of panobinostat (LBH589) in combination with melphalan to be used in the Phase 2 portion of the study (NCT00743288)
Timeframe: 12 months

Interventionmg LBH589 (Number)
Melphalan and Panobinostat Schedule D320

[back to top]

Duration of Response

(NCT00743288)
Timeframe: First evidence of PR or better (for overall response) and MR or better (for clinical benefit response) to start of disease progression or death

Interventionmonths (Median)
Melphalan and Panobinostat Schedule B8.1

[back to top]

MTD

Phase 1: to determine MTD of melphalan in combination with panobinostat to be used in the Phase 2 portion of the study (NCT00743288)
Timeframe: 12 months

Interventionmg/kg melphalan (Number)
Melphalan and Panobinostat Schedule D30.05

[back to top]

Overall Response Rate (ORR) and Clinical Benefit Rate (CBR) [ORR= Complete Response (CR) + Very Good Partial Response (VGPR) + Partial Response (PR)]; CBR=ORR + Minimal Response (MR)] Following Treatment With Panobinostat and Melphalan

Responses were evaluated according to criteria modified from those developed by Blade et al., 1998 The reference point for evaluating response improvement is the baseline. This baseline reference point is also valid when a patient has already achieved a response and transitions through into a better response grade. (NCT00743288)
Timeframe: 24 months

,,,,
Interventionparticipants (Number)
CRVGPRPRMRSD (stable disease)Progressive disease (PD)ORR (CR+VGPR+ PR)CBR (ORR+MR)
Melphalan and Panobinostat All Patients0210231433
Melphalan and Panobinostat Schedule A00001200
Melphalan and Panobinostat Schedule B02105133
Melphalan and Panobinostat Schedule C00005200
Melphalan and Panobinostat Schedule D000012900

[back to top]

Time to Progression

(NCT00743288)
Timeframe: Time from the start of treatment to progressive disease

Interventionmonths (Median)
Melphalan and Panobinostat1.6

[back to top]

Objective Response Rate (as Determined by Investigator): the Percentage of Patients Assigned to a Treatment Arm With a Confirmed Best Response of CR or PR.

The assessment of overall response (OR) is based on the response of target lesion, of non-target lesion, and on presence of new lesions (RECIST criteria version 1.0 using imaging techniques; as per investigator assessment). (NCT00777049)
Timeframe: 6 years and 2 months

,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable Disease / Incompete ResponseProgressive DiseaseMissing
ER- and PgR- (Arm II)104142
ER+ and/or PgR+ (Arm I)0113145

[back to top]

Corrected QT Interval Fridericia's Formula (QTcF)

Prolonged QTcF: QTcF >450 msec and increase of baseline on greater than or equal to 60 msec. (NCT00777335)
Timeframe: Panobinostat intra-venous (i.v.): All cycles pre-dose measurements. For cycles 1 and 2, post-dose measurements as well. / Panobinostat oral: Pre-dose and post-dose measurements for all cycles. Note: each cycle = 3 weeks

,
Interventionparticipants (Number)
Electrocardiogram QT normalElectrocardiogram QT prolonged
Panobinostat Intra-venous (i.v.)11
Panobinostat Oral20

[back to top]

Overall Response (OR) Rate (as Determined by the Investigator): the Number of Patients Assigned to a Treatment Arm With a Confirmed Best Response of Complete Response(CR) or Partial Response (PR).

"The assessment of OR is based on the response of target lesion, of non-target lesion and on presence of new lesions (RECIST Criteria (V1.0)-assessed by CT scan spiral and bone scan)~CR:Disappearance of all target lesions~PR:>=30% increase in the sum of the longest diameter (SLD),taking as reference the nadir SLD~Progressive Disease (PD):>=20% increase in the SLD, taking as reference the nadir SLD, or the appearance of one or more new lesions~Stable Disease(SD):Neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the nadir SLD" (NCT00777335)
Timeframe: At screening, every 2 cycles (i.e. 6 weeks) during the first 6 cycles, every 3 cycles (i.e. 9 weeks) during the subsequent cycles and at the End of Treatment (EOT) visit. After the EOT, the tumor assessments should be performed every 9 weeks.

,
Interventionparticipants (Number)
Progressive Disease (PD)Stable Disease (SD)Complete Response (CR)Partial Reponse (PR)
Panobinostat Intra-venous (i.v.)2000
Panobinostat Oral1100

[back to top]

6-Month Progression-Free Survival (PFS6) [Phase I]

PFS6 is the proportion of patients remaining alive and progression-free at 6-months from study entry. Progressive disease was established based on Response Assessment in Neuro-Oncology (RANO) criteria (Wen et al JCO 2010). (NCT00859222)
Timeframe: Disease was assessed radiographically to document clinical progression every cycle on treatment and post-treatment every 8 weeks up to 12 months. Participants were followed for PFS6 up to 6 months since study entry.

Interventionproportion of participants (Number)
All Phase I Participants0.25

[back to top]

Overall Survival [Phase II]

OS is defined as the time from study entry to death or date last known alive. (NCT00859222)
Timeframe: Participants were followed long-term for survival every 4 months from the end of treatment until death or lost to follow-up. Phase II participants were followed for OS up to 27 months on this study.

Interventionmonths (Median)
Phase II GBM: Bevacizumab + LBH589 30 mg Every Other Week9
Phase II AG: Bevacizumab + LBH589 30 mg Every Other Week17

[back to top]

Best Radiographic Response

Radiographic response was established based on Response Assessment in Neuro-Oncology (RANO) criteria (Wen et al JCO 2010) with 5 potential categories: Complete Response (CR), Partial Response (PR), Stable Disease (SD), Progressive disease (PD) and Unknown status. (NCT00859222)
Timeframe: Disease was assessed radiographically for response every cycle on treatment. Treatment duration in cycles was a median (range) of 2 (1-6) PI Cohort 1, 4.5 (2-6) PI Cohort 2, 6 (2-10) PI Cohort 3, 5 PII GBM and 7 PII AG.

,,,,
Interventionparticipants (Number)
Complete ResponsePartial ResponseStable DiseaseProgressive Disease
Phase I Cohort 1: Bevacizumab +LBH589 20 mg Every Week0021
Phase I Cohort 2: Bevacizumab + LBH589 20 mg Every Other Week0120
Phase I Cohort 3: Bevacizumab + LBH589 30 mg Every Other Week0231
Phase II AG: Bevacizumab + LBH589 30 mg Every Other Week0492
Phase II GBM: Bevacizumab + LBH589 30 mg Every Other Week07143

[back to top]

Progression-Free Survival (PFS) [Phase II]

PFS is defined as the time from study entry to the earliest documentation of disease progression or death. Patients alive without evidence of PD were censored at the date of last disease assessment. (NCT00859222)
Timeframe: Disease was assessed radiographically to document clinical progression every cycle on treatment and post-treatment every 8 weeks up to 12 months.

Interventionmonths (Median)
Phase II GBM: Bevacizumab + LBH589 30 mg Every Other Week5
Phase II AG: Bevacizumab + LBH589 30 mg Every Other Week7

[back to top]

Progression-Free Survival (PFS) [Phase I]

PFS is defined as the time from study entry to the earliest documentation of disease progression or death. Patients alive without evidence of PD were censored at the date of last disease assessment. Progressive disease was established based on Response Assessment in Neuro-Oncology (RANO) criteria (Wen et al JCO 2010). (NCT00859222)
Timeframe: Disease was assessed radiographically to document clinical progression every cycle on treatment and post-treatment every 8 weeks up to 12 months.

Interventionmonths (Median)
All Phase I Participants4.3

[back to top]

Overall Survival (OS) [Phase I]

OS is defined as the time from study entry to death or date last known alive. (NCT00859222)
Timeframe: Participants were followed long-term for survival every 4 months from the end of treatment until death or lost to follow-up. Phase I participants were followed for OS up to 12.1 months on this study.

Interventionmonths (Median)
All Phase I Participants8.2

[back to top]

LBH589 Maximum Tolerated Dose (MTD) [Phase I]

The MTD LBH589 in combination with bevacizumab 10 mg/kg intravenously (IV) on days 1 and 15 of each 28 day cycle is determined by the number of patients who experience a dose limiting toxicity (DLT). See subsequent primary outcome measure for the DLT definition. The MTD is defined as the highest dose at which fewer than one-third of patients experience a DLT. If no DLTs are observed, the MTD is not reached but the highest dose received may be the Recommended Phase II Dose (RP2D). The MTD was not reached with 0 of 6 DLTs observed in the highest dose cohort but due to safety concerns higher doses of LBH589 with bevacizumab were neither planned nor tested. The RP2D was 30 mg/day orally, 3x per week, every other week. (NCT00859222)
Timeframe: Participants were assessed every 2 weeks while on study; The observation period for MTD evaluation was the first 30 days of treatment.

Interventionmg/day orally, 3x per wk, every other wk (Number)
All Phase I Participants30

[back to top]

Dose Limiting Toxicity (DLT) [Phase I]

A DLT was defined as an adverse event that (a) is related to the LBH589 and/or bevacizumab with an attribution of possible, probable, or definite, and (b) occurs during and/or begins during the first 30 days of the study treatment, and (c) meets any of the following criteria: grade 3 thrombocytopenia; grade 4 neutropenia lasting 7 days; grade 4 anemia lasting 7 days despite transfusion or growth factors; febrile neutropenia if ANC<0.5 x10^9/L; a QT interval corrected for heart rate (QTc) of 500-515 msec that did not stabilize to <480 msec after one week; a second occurrence of QTc 500-515 msec; any QTc >515 msec; any deep vein thrombosis (DVT) or pulmonary embolism (PE) while on fully therapeutic anticoagulation therapy; Grade 3 proteinuria lasting 14 days; or any other clinically significant Grade 3 toxicity despite maximal medical therapy lasting 7 days, any Grade 4 toxicity despite maximal medical therapy; or any Grade 3 or 4 toxicity resulting in study drug discontinuation. (NCT00859222)
Timeframe: Participants were assessed every 2 weeks while on study; The observation period for DLT evaluation was the first 30 days of treatment.

Interventionparticipants with DLT (Number)
Phase I Cohort 1: Bevacizumab +LBH589 20 mg Every Week1
Phase I Cohort 2: Bevacizumab + LBH589 20 mg Every Other Week0
Phase I Cohort 3: Bevacizumab + LBH589 30 mg Every Other Week0

[back to top]

6-Month Progression-Free Survival (PFS6) [Phase II]

PFS6 is the proportion of patients remaining alive and progression-free at 6-months from study entry. Progressive disease was established based on RANO criteria (Wen et al JCO 2010). (NCT00859222)
Timeframe: Disease was assessed radiographically to document clinical progression every cycle on treatment and post-treatment every 8 weeks up to 12 months. Participants were followed for PFS6 up to 6 months since study entry.

Interventionproportion of participants (Number)
Phase II GBM: Bevacizumab + LBH589 30 mg Every Other Week0.304
Phase II AG: Bevacizumab + LBH589 30 mg Every Other Week0.467

[back to top]

Percentage of Patients Free of Progression and Without Symptomatic Deterioration

"measured by PSA and /or metastases progression criteria by body CT following RECIST criteria 1.1 and/or bones scan following the appearance of at least 2 new bone metastases and confirmation of 2 additional bone metastasis on a subsequent bone scan 6-8 weeks later and/or clinical progression.~Only participants who completed two or more treatment cycles were assessed for this outcome measure." (NCT00878436)
Timeframe: 9 months

Interventionpercentage of Participants (Number)
LBH 40mg24
LBH 20mg9

[back to top]

Percentage of Patients Free of Progression and Without Symptomatic Deterioration

"measured by PSA and /or metastases progression criteria by body CT following RECIST criteria 1.1 and/or bones scan following the appearance of at least 2 new bone metastases and confirmation of 2 additional bone metastasis on a subsequent bone scan 6-8 weeks later and/or clinical progression.~Only participants who completed two or more treatment cycles were assessed for this outcome measure." (NCT00878436)
Timeframe: 6 months

Interventionpercentage of participants (Number)
LBH 40mg42
LBH 20mg19

[back to top]

Best Response as Per Investigator Assessment by Stratum (FAS)

Response to treatment was defined as complete remission rate (CRR). CRR is complete remission (CR) and morphologic CR with incomplete blood count recovery (residual neutropenia or thrombocytopenia) (CRi). To stop or to proceed with Stage 2 of a given stratum of the Simon's optimal 2-stage design was based on the number of patients with CR/CRi and a safety evaluation of the patients from Stage 1 in that stratum. If early results clearly indicated that the drug was not active or worthy of further investigation, enrollment of that particular stratum would be terminated. CR and CRi were assessed by the Investigator according to IWG response Criteria for AML (Cheson et al 2003). As per protocol we would continue to stage II if ≥ 4 patients out of 26 patients enrolled to stage I had a CR or a CRi. As per response observed there was only 1 patient with CR/CRi in stratum A and 2 patients with CR/CRi in Stratum B. (NCT00880269)
Timeframe: 6 cycles of treatment with a 28-day treatment cycle (Day 168)

,
InterventionPercentage of Participants (Number)
Complete remission rate (CR/CRi)Complete remission (CR)CR with incomplete blood count recovery (CRi)Partial remission (PR)Treatment failureUnknown
Stratum A3.103.1040.656.3
Stratum B7.43.73.7040.751.9

[back to top]

Duration of Response (Phase II)

Duration of response is defined as the time from the date at which the objective status is first noted to be (for myeloma) a complete response (CR, defined as Negative immunofixation(IFE) of the serum and urine, < 5% plasma cells in bone marrow(BM), Disappearance of plasmacytomas), stringent CR(sCR, defined as CR plus Normal serum FLC ratio, Absence of clonal cells in BM), very good partial response(VGPR, defined as PR plus Serum and urine M-component detectable by IFE but not on electrophoresis), partial response(PR, defined as a ≥ 50% reduction of serum M-protein and/or reduction in 24-h urinary M-protein by ≥ 90% or to <200 mg per 24 h) or minor response(MR, defined as ≥25% but < 49% reduction of serum M-protein and reduction in 24h urine M-protein by 50-89%); (for lymphoma) a CR(defined as no evidence of measurable disease), or PR(defined as regression of measurable disease and no new sites of disease) noted as the objective status. Estimated using the method of Kaplan-Meier. (NCT00918333)
Timeframe: The time from the date at which the patient's objective status is first noted to be a CR, sCR, VGPR, PR, or minor response to the earliest date progression is documented, assessed up to 2 years post-treatment

Interventionmonths (Median)
Phase II (Lymphoma Patients Receiving 20 mg LBL589)9.1
Phase II (Myeloma Patients Receiving 20 mg LBH589)NA
Phase II (Lymphoma Patients Receiving 30/40 mg LBH589)12.9

[back to top]

Number of Phase I Participants With Dose-Limiting Toxicity Events (Phase I)

The Maximum Tolerated Dose (MTD) is defined as the dose level below the lowest dose that induces dose-limiting toxicity (DLT) in at least one-third of patients graded according to NCI Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. Dose-limiting toxicities include non-hematologic events graded 3 or higher and deemed at least possibly related to treatment. A total of 6 patients treated at the MTD will be sufficient to identify common toxicities at the MTD. The number of patients reporting a dose-limiting event are reported. (NCT00918333)
Timeframe: 4 weeks

InterventionParticipants (Count of Participants)
Phase I (Panobinostat + Everolimus)0

[back to top]

Overall Response Rate (Phase II)

For myeloma, a complete response(CR, defined as Negative immunofixation(IFE) of the serum and urine, < 5% plasma cells in bone marrow(BM), Disappearance of plasmacytomas), stringent CR(sCR, defined as CR plus Normal serum FLC ratio, Absence of clonal cells in BM), very good partial response(VGPR, defined as PR plus Serum and urine M-component detectable by IFE but not on electrophoresis), partial response(PR, defined as a ≥ 50% reduction of serum M-protein and/or reduction in 24-h urinary M-protein by ≥ 90% or to <200 mg per 24 h), or minor response(MR, defined as ≥25% but < 49% reduction of serum M-protein and reduction in 24h urine M-protein by 50-89%) noted as the objective status. For lymphoma, a CR(defined as no evidence of measurable disease), or PR(defined as regression of measurable disease and no new sites of disease) noted as the objective status. Percentage of successes will be estimated by 100 times the number of successes divided by the total number of evaluable patients. (NCT00918333)
Timeframe: Up to 12 courses

Interventionpercentage of patients (Number)
Phase II (Lymphoma Patients Receiving 20 mg LBL589)39
Phase II (Myeloma Patients Receiving 20 mg LBH589)7
Phase II (Lymphoma Patients Receiving 30/40 mg LBH589)20
Phase II (Myeloma Patients Receiving 30/40 mg LBH589)0

[back to top]

Overall Survival Time (Phase II)

Overall survival time is defined as the time from registration to death due to any cause. The median and 95% confidence intervals will be estimated using the method of Kaplan-Meier. (NCT00918333)
Timeframe: Time from registration to death due to any cause, assessed up to 2 years post-treatment

Interventionmonths (Median)
Phase II (Lymphoma Patients Receiving 20 mg LBL589)17.1
Phase II (Myeloma Patients Receiving 20 mg LBH589)16.6
Phase II (Lymphoma Patients Receiving 30/40 mg LBH589)35.4
Phase II (Myeloma Patients Receiving 30/40 mg LBH589)21.7

[back to top]

Progression-free Survival (Phase II)

Progression-free survival time is defined as the time from registration to progression or death due to any cause. Progression is defined for myeloma as Any one or more of the following: Increase of 25% from lowest value in, Serum M-component (absolute increase must be ≥ 0.5 g/dl), Serum M-component increase ≥ 1 g/dl, if lowest M component was ≥ 5 g/dl,Urine M-component (absolute increase must be ≥ 200 mg/24 h), Bone marrow plasma cell percentage (absolute % must be ≥10%) Or any one or more of the following felt related to the underlying clonal plasma cell proliferative disorder: Development of new soft tissue plasmacytomas or bone lesions, Hypercalcemia (≥11.5 mg/dl) Decrease in hemoglobin of ≥2 g/dl, Serum creatinine level ≥2 mg/dl. Progression is defined for lymphoma as any new lesion or increase by ≥50% of previously involved sites from nadir. The median and 95% confidence intervals are estimated using the method of Kaplan-Meier. (NCT00918333)
Timeframe: Time from registration to progression or death due to any cause, assessed up to 2 years post-treatment

Interventionmonths (Median)
Phase II (Lymphoma Patients Receiving 20 mg LBL589)3.7
Phase II (Myeloma Patients Receiving 20 mg LBH589)2.3
Phase II (Lymphoma Patients Receiving 30/40 mg LBH589)4.2
Phase II (Myeloma Patients Receiving 30/40 mg LBH589)4.3

[back to top]

Phase I - Number of Participants With Dose Limiting Toxicities (DLTs) at a Given Dose Level

"A DLT was any Grade 4 toxicity for neutrophils or platelets for ≥ 7 days, Grade 3 toxicity for neutrophils for ≥ 21 days, any Grade 3 solid organ toxicity not explainable by another cause (e.g. neurotoxicity, GI toxicity), metabolic/laboratory toxicity (≥10 x ULN AST) for ≥ 14 days, any Grade 4 infection or QTcF> 500msec EKG. DLTs were assessed according to the Common Toxicity Criteria for Adverse Events (CTCAE) version 3.0.~All adverse events were collected and graded to determine DLTs as assessed according to the Common Toxicity Criteria for Adverse Events (CTCAE) version 3.0. DLTs were assessed to determine the recommended Decitabine and Panobinostat dose for the Phase II portion of the trial." (NCT00925132)
Timeframe: 6 weeks (one full cycle)

InterventionParticipants (Count of Participants)
Cohort 1: Experienced DLTCohort 2: Experienced DLTCohort 3: Experienced DLTCohort 4: Experienced DLT
Phase I Dose Escalation0000

[back to top]

Phase 2 -Number of Patients With a Decrease in Tumor Size Using RECIST and CHOI's Criteria

"Tumor response rate was assessed using RECIST criteria in which a complete response was the disappearance of all target lesions; Partial response was a 30% decrease in the sum of the longest dimension (LD) of target lesions, relative to baseline measurement; Progressive disease was an increase of 20% or more in the sum of the LD of target lesions; and Stable disease was a decrease in tumor size of less than 30% or increase of less than 20%.~Tumor response rate was also assessed using CHOI's criteria in which a response was a 10% decrease in tumor size or a 15% decrease in tumor density on contrast-enhanced computed tomography scan.~Tumor response rates were assessed in order to determine effectiveness of the treatment regimen which was defined by the response rate of at least 30% as measured by either the RECIST or Choi's criteria, and ineffective if the rate is less than 15% on both." (NCT00925132)
Timeframe: 12 weeks (2 cycles)

InterventionParticipants (Count of Participants)
Progressive Disease (PD)Stable Disease (SD)
Phase II:Decitabine 0.2 mg/kg +Panobinostat 30mg +Temozolomide125

[back to top]

Change From Baseline in Myelofibrosis Symptom Assessment Form (MF-SAF) Scale Scores At Cycle 6

The MF-SAF consists of seven questions about key symptoms and impact of MF. Questions were scored on a scale of 0-10, with higher scores indicating more severe symptoms and greater inactivity. Questions 1-6 investigate the following symptoms: night sweats, pruritus/itching, abdominal discomfort, pain under the ribs, early satiety and bone/muscle pain. Question 7 asked participants to report levels of inactivity. Negative change from baseline indicates improvement. (NCT00931762)
Timeframe: Baseline, Cycle 6 (each cycle was of 28 days)

Interventionscore on a scale (Mean)
Cycle 6, Day 1: Fatigue Right nowCycle 6, Day 1: Fatigue Usual Past 24 HoursCycle 6, Day 1: Fatigue Worst Past 24 HoursCycle 6, Day 1: General ActivityCycle 6, Day 1: MoodCycle 6, Day 1: Walking AbilityCycle 6, Day 1: Normal WorkCycle 6, Day 1: Relations with Other PeopleCycle 6, Day 1: Enjoyment of LifeCycle 6, Day 1: Filling up quickly when you eatCycle 6, Day 1: Abdominal pain or DiscomfortCycle 6, Day 1: InactivityCycle 6, Day 1: CoughCycle 6, Day 1: Night SweatsCycle 6, Day 1: ItchingCycle 6, Day 1: Bone PainCycle 6, Day 1: What is Overall Quality of Life?
Panobinostat-1.5-1.0-3.0-3.5-3.5-3.0-4.0-4.5-5.01.00.5-0.5-1.5-0.51.5-2.5-2.5

[back to top]

Number of Participants With Adverse Events (AEs), and Serious Adverse Events (SAEs)

An adverse event is the appearance or worsening of any undesirable sign, symptom, or medical condition occurring after starting the study drug even if the event is not considered to be related to study drug. SAEs was an undesirable sign, symptom or medical condition which is: life-threatening, persistent or significant disability/incapacity, congenital anomaly/birth defect, requires hospitalizations or prolongation of hospitalizations, or is medically significant (NCT00931762)
Timeframe: AE: From the start of treatment up to 28 days after last study dose (up to approximately 2 years); SAE: From time of informed consent up to 4 weeks after last study dose (approximately 2 years)

InterventionParticipants (Count of Participants)
AEsSAEs
Panobinostat3519

[back to top]

Overall Response (OR) Rate Per International Working Group for Myelofibrosis Research and Treatment (IWG-MRT) Criteria

Overall response (OR) = Complete remission (CR) + Partial remission (PR) + Clinical improvement (CI). CR: complete resolution of disease-related symptoms and signs with hemoglobin level 110 grams per deciliter (g/L), platelet count 100 x 10^9/liter (L), and absolute neutrophil count (ANC) 1.0 x 10^9/L, all 3 blood counts < upper normal limit (UNL), normal leukocyte differential, and bone marrow histologic remission. PR= all of the criteria for CR except the requirement for bone marrow histologic remission. CI= minimum 20 g/L increase in hemoglobin for transfusion or becoming transfusion independent or reduction in splenomegaly ≥ 50% or 100% increase in platelet count and absolute platelet count of 50 x 10^9/L and 100% increase in ANC of at least 0.5 x 10^9/L. (NCT00931762)
Timeframe: Up to approximately 2 years

Interventionpercentage of participants (Number)
Panobinostat2.9

[back to top]

Change From Baseline in Myelofibrosis Symptom Assessment Form (MF-SAF) Scale Scores

The MF-SAF consists of seven questions about key symptoms and impact of MF. Questions were scored on a scale of 0-10, with higher scores indicating more severe symptoms and greater inactivity. Questions 1-6 investigate the following symptoms: night sweats, pruritus/itching, abdominal discomfort, pain under the ribs, early satiety and bone/muscle pain. Question 7 asked participants to report levels of inactivity. Negative change from baseline indicates improvement. (NCT00931762)
Timeframe: Baseline, Cycles 2, and 4 (each cycle was of 28-days)

Interventionscore on a scale (Mean)
Cycle 2, Day 1: Fatigue Right nowCycle 2, Day 1: Fatigue Usual Past 24 HoursCycle 2, Day 1: Fatigue Worst Past 24 HoursCycle 2, Day 1: General ActivityCycle 2, Day 1: MoodCycle 2, Day 1: Walking AbilityCycle 2, Day 1: Normal WorkCycle 2, Day 1: Relations with Other PeopleCycle 2, Day 1: Enjoyment of LifeCycle 2, Day 1: Filling up quickly when you eatCycle 2, Day 1: Abdominal Pain or DiscomfortCycle 2, Day 1: InactivityCycle 2, Day 1: CoughCycle 2, Day 1: Night SweatsCycle 2, Day 1: ItchingCycle 2, Day 1: Bone PainCycle 2, Day 1: What is Overall Quality of Life?Cycle 4, Day 1: Fatigue Right nowCycle 4, Day 1: Fatigue Usual Past 24 HoursCycle 4, Day 1: Fatigue Worst Past 24 HoursCycle 4, Day 1: General ActivityCycle 4, Day 1: MoodCycle 4, Day 1: Walking AbilityCycle 4, Day 1: Normal WorkCycle 4, Day 1: Relations with Other PeopleCycle 4, Day 1: Enjoyment of LifeCycle 4, Day 1: Filling up quickly when you eatCycle 4, Day 1: Abdominal Pain or DiscomfortCycle 4, Day 1: InactivityCycle 4, Day 1: CoughCycle 4, Day 1: Night SweatsCycle 4, Day 1: ItchingCycle 4, Day 1: Bone PainCycle 4, Day 1: What is Overall Quality of Life?
Panobinostat0.00.50.70.00.5-0.60.2-0.10.30.7-0.00.8-0.1-1.0-1.5-1.0-0.40.00.81.0-0.50.3-0.60.9-0.60.4-0.11.00.6-0.9-1.1-0.1-1.8-0.3

[back to top]

Median Duration of Response

"Duration of response DR is defined as the time from the date of first response after treatment to the date of disease progression Patients who have died for any cause or are alive without progression are censored at the date the patient is last know to be progression-free.~Duration of response DR is defined as the time from the date of first response after treatment to the date of disease progression or death for any cause. Patients who are alive without progression are censored at the date the patient is last know to be progression-free." (NCT00936611)
Timeframe: Assessed after 2nd cycle and then every subsequent cycle for 6 cycles and then every 3 months. If taken off treatment at cycle 2 for disease progression, assessed every 3 months. The median (range) follow up from treatment start is 7.7 month (0.9 - 29.7).

Interventionmonths (Median)
LBH5896.9

[back to top]

Median Progression Free Survival

Progression free survival (PFS) is defined as the time from start of treatment to disease progression or death from any cause as estimated by Kaplan Meier methods. Patients who have not progressed and are alive are censored at the date the patient is known to be progression-free. (NCT00936611)
Timeframe: Assessed after 2nd cycle and then every subsequent cycle for 6 cycles and then every 3 months. If taken off treatment at cycle 2 for disease progression, assessed every 3 months. The median (range) follow up from treatment start is 7.7 month (0.9 - 29.7).

Interventionmonths (Median)
LBH5896.6

[back to top]

Median Time to Progression

Time to progression (TTP) is defined as the time from start of treatment to progression. Patients who have not progressed are censored at the date the patient is last known to be progression free. (NCT00936611)
Timeframe: Assessed after 2nd cycle and then every subsequent cycle for 6 cycles and then every 3 months. If taken off treatment at cycle 2 for disease progression, assessed every 3 months. The median (range) follow up from treatment start is 7.7 month (0.9 - 29.7).

Interventionmonths (Median)
LBH5896.6

[back to top] [back to top]

Overall Response Rate

"Overall response rate is percentage of participants with complete (CR), very good partial (VGPR), partial (PR), or minimal response (MR) as best response during treatment.~CR~Disappearance of monoclonal protein by immunofixation~No histologic evidence of bone marrow involvement~Resolution of any adenopathy/organomegaly (confirmed by CT scan), or signs or symptoms attributable to WM.~Second immunofixation required for confirmation.~VGPR~-At least 90% reduction of serum monoclonal IgM concentration on protein electrophoresis.~PR~At least 50% reduction of serum monoclonal IgM concentration on protein electrophoresis and at least 50% decrease in adenopathy/organomegaly on physical examination or on CT scan.~No new symptoms or signs of active disease.~MR~At least 25% but less than 50% reduction of serum monoclonal IgM by protein electrophoresis.~No new symptoms or signs of active disease." (NCT00936611)
Timeframe: Assessed after 2nd cycle and then every subsequent cycle for 6 cycles. The median number of completed cycles of therapy was 5 (0- 32). As such observed up to ~32 months.

Interventionpercentage of participants (Number)
LBH58947

[back to top]

Overall Response Rate Based on the Hematologic Improvement

"Overall response rate defined by International Working Group (IWG) response criteria in myelodysplasia. Hematologic Improvement (HI) responses, at least 9 weeks: Erythroid response (pretreatment, <11 g/dL): Hgb increase by 1.5 g/dL; Relevant reduction units of Red blood cell (RBC) transfusions by absolute number at least 4 RBC transfusions/8 week compared with pretreatment transfusion number in previous 8 weeks; Only RBC transfusions for Hgb of 9.0 g/dL pretreatment count in RBC transfusion response evaluation; Platelet response (pretreatment,<100x10^9/L): If starting with >20x10^9/L platelets:~absolute increase 30x10^9/L, Increase from baseline <20 x10^9/L to >20x10^9/L and by =/> 100%; Neutrophil response (pretreatment, <1.0x10^9/L): =/> 100% increase & absolute increase >0.5x10^9/L; Progression or relapse after HI: At least 1 of the following: =/>50% decrement from max response levels in granulocytes or platelets; Reduction in Hgb by 1.5 g/dL; or Transfusion dependence ." (NCT00939159)
Timeframe: Assessment with 28-day cycle until response, then every 3 cycles as needed, for up to 24 months

InterventionPercentage of Participants (Number)
LBH5898

[back to top]

Hematologic Improvement (HI) for Myeloid Dysplastic Syndromes(MDS)/Chronic Myelomonocytic Leukemia (CMML) Patients Per Investigator (Phase Llb)

"Hematologic response consists of Erythroid response (HI-E), Platelet response (HI-P) and Neutrophil response (HI-N).~HI-E: Hgb increase by ≥ 1.5 g/dL over pretreatment & relevant reduction of units of RBC transfusions by an absolute number of at least 4 units of PRBCs/8 weeks compared with the pretreatment transfusion number in the previous 8 weeks. Only RBC transfusions given for a Hgb of ≤ 9.0 g/dL pretreatment will count in the RBC transfusion response evaluation.~HI-P: Absolute increase of ≥ 30 x 109/L over pretreatment or patients starting with ≥ 20 x 109/L platelets OR increase from <20 x 109/L at pretreatment to > 20 x 109/L and by at least 100%.~HI-N: At least 100% increase and an absolute increase > 0.5 x 109/L over pretreatment value." (NCT00946647)
Timeframe: 48 months

,
InterventionPercentage of participants (Number)
Erythroid response (HI-E)Platelet response (HI-P)Neutrophil response (HI-N)
5-Azacytidine31.024.113.8
Panobinostat + 5-Azacytidine25.835.519.4

[back to top]

Overall Response Rate (ORR) Assessed by Best Overall Response: Participants With AML Per Investigator (Phase Llb)

"Best overall response as measured by complete remission (CR) or complete response with incomplete blood count recovery (CRi) or partial remission (PR).~Overall response patients achieved other than the composite CR by individual response category: CR, CRi or PR." (NCT00946647)
Timeframe: 48 months

,
InterventionPercentage of participants (Number)
Clinical response (CR, CRi, PR)Complete remission (CR)Compl remiss. with incompl blood cnt recovery(CRi)Partial remission (PR)
5-Azacytidine30.815.47.77.7
Panobinostat + 5-Azacytidine22.211.111.10.0

[back to top]

Overall Response Rate (ORR) Assessed by Best Overall Response: Participants With MDS/CMML Per Investigator (Phase Llb)

"Best overall response as measured by complete remission (CR) or bone marrow CR (BM-CR) or partial remission (PR) or hematologic improvement (HI).~Overall response patients achieved other than the composite CR by individual response category: CR, CRi, mCR or PR as defined by the International Working Group (IWG) response criteria." (NCT00946647)
Timeframe: 48 months

,
InterventionPercentage of participants (Number)
Clinical response (CR, BM-CR, PR, HI)Complete remission (CR)Bone marrow CR (BM-CR)Partial remission (PR)
5-Azacytidine41.46.93.46.9
Panobinostat + 5-Azacytidine41.916.112.90.0

[back to top]

Clinical Response Other Than Composite Clinical Response for Myeloid Dysplastic Syndromes(MDS)/Chronic Myelomonocytic Leukemia (CMML) Patients Per Investigator (Phase Llb)

This is the best overall response as measured by Clinical response. Clinical response is defined as having complete remission (CR), bone marrow complete remission (BM-CR), partial remission or hematologic improvement (HI) as defined by the International Working Group (IWG) response criteria. (NCT00946647)
Timeframe: 48 months

InterventionPercentage of participants (Number)
Panobinostat + 5-Azacytidine41.9
5-Azacytidine41.4

[back to top]

Clinical Response Other Than Composite Clinical Response for Acute Myelogenous Leukemia (AML) Patients Per Investigator (Phase Llb)

This is the best overall response as measured by Clinical response. Clinical response is defined as having complete remission (CR), complete remission with incomplete blood count recovery (CRi) or partial remission as defined by the International Working Group (IWG) response criteria. (NCT00946647)
Timeframe: 48 months

InterventionPercentage of participants (Number)
Panobinostat + 5-Azacytidine22.2
5-Azacytidine30.8

[back to top]

1-year Survival Rate (Phase Llb)

Overall survival was defined as the time from date of randomization to date of death due to any cause. If a patient was not known to have died, survival was censored at the date of last contact. Patients not known to have died were censored for 'Lost to follow-up' if the time between their last contact date and the analysis cut-off date was longer than 3 months and 2 weeks (104 days) during the first year after study evaluation completion, and longer than 6 months and 2 weeks (194 days), thereafter. The 1-year survival rate was obtained from the Kaplan-Meier analysis of overall survival, and its variance was estimated by Greenwood's formula. (NCT00946647)
Timeframe: 12 months

Interventionmonths (Median)
Panobinostat + 5-Azacytidine14.9
5-Azacytidine15.6

[back to top]

Number of Dose Limiting Toxicity (DLT) (Phase lb)

Dose limiting toxicity (DLT) was defined as a toxicity requiring treatment withdrawal and included the following: Non-hematologic toxicity qualifying for DLT and Hematologic toxicity qualifying for DLT (NCT00946647)
Timeframe: within the first 28 days (cycle 1)

InterventionDLTs (Number)
PAN + 5-Aza 20 mg2
PAN + 5-Aza 30 mg6
PAN + 5-Aza 40 mg4

[back to top]

Number of Participants With Dose Limiting Toxicity (DLT) (Phase lb)

Dose limiting toxicity (DLT) was defined as a toxicity requiring treatment withdrawal and included the following: Non-hematologic toxicity qualifying for DLT and Hematologic toxicity qualifying for DLT (NCT00946647)
Timeframe: within the first 28 days (cycle 1)

InterventionParticipants (Number)
PAN + 5-Aza 20 mg1
PAN + 5-Aza 30 mg3
PAN + 5-Aza 40 mg2

[back to top]

Time to Progression (TTP) (Phase Llb)

"Time to progression (TTP) was defined as the time from the date of randomization to the date of the first documented PD per investigator's assessment or death due to study indication.~Time to progression was analyzed by the Kaplan Meier method. Based on the Guidelines for Implementation of international working group (IWG) response criteria in AML, MDS and CMML according to Cheson 2003 and 2006." (NCT00946647)
Timeframe: 48 months

Interventionmonths (Median)
Panobinostat + 5-AzacytidineNA
5-Azacytidine15.2

[back to top]

Composite Complete Response (Phase Llb)

Composite complete response is defined as complete response (CR), Complete response with incomplete blood count recovery (CRi) or bone marrow complete response (BM-CR) as defined by the International Working Group (IWG) response criteria. (NCT00946647)
Timeframe: 48 months

InterventionPercentage of participants (Number)
Panobinostat + 5-Azacytidine27.5
5-Azacytidine14.3

[back to top]

Maximum Tolerated Dose (MTD) of Everolimus With Panobinostat

MTD of the novel combination of Everolimus + Panobinostat (LBH589) in a phase-I study in participants with relapsed lymphoma (Hodgkin and non-Hodgkin) where MTD is defined as the highest dose at which no more than 1 in 6 of the participants in the cohort experiences one or more dose limiting toxicities (DLTs) in the first 28 day treatment cycle. Thirty patients were enrolled onto four dose levels: Everolimus (mg, orally) 5, 5, 10, 10 daily or Panobinostat (mg, orally) 10, 20, 20, 30 three times per week. The MTD was established without the use of colony stimulating factor in cycle 1. (NCT00967044)
Timeframe: 28 day treatment cycle

Interventionmg, orally (Number)
Everolimus (daily)Panobinostat (three times weekly)
Panobinostat + Everolimus1020

[back to top]

Overall Response Rate

Overall Response Rate is the number of participants with a partial and complete response assessed by the Updated Cheson criteria for lymphoma. A complete response is complete disappearance of all detectable clinical and radiographic evidence of disease and disappearance of all disease-related symptoms if present before therapy and normalization of those biochemical abnormalities. If a subject has residual lesions on CT scan and the disease was fluorodeoxyglucose (FDG) avid pre-treatment, then that subject will be considered to be in a complete response. Partial response is a greater than or equal to 50% decrease in the sum of the products of the greatest diameters of 6 largest dominant nodes or nodal masses. No increase in size of nodes, liver or spleen and no new sites of disease (NCT00978432)
Timeframe: after 2 cycles of each study drug or after 2 cycles of doublet, up to 24 weeks

Interventionparticipants (Number)
Arm 1a (RAD001 Followed by LBH589)1
Arm 1b (LBH589 Followed by RAD001)0
Doublet (Combination RAD001 and LBH589)4

[back to top]

Summary of Adverse Events (AEs)

Counts of adverse events or treatment-emergent adverse events (TEAE, defined as newly occurring or worsening after first dose) experienced by patients on study drug(s). National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE version 3.0) is used to assess severity. Relatedness to study drug is assessed by the investigator. (NCT00978432)
Timeframe: From the time of first dose of study drug until 4 weeks after participant has stopped study drug; up to 1 year

,,
Interventionevents (Number)
Blood and lymphatic AEsGastrointestinal AEsGeneral Disorder AEsInvestigation (Lab) AEsMetabolism and Nutrition AEsMusculoskeletal and Connective Tissue AEsNervous System AEsRenal and Urinary AEsReproductive System AEsRespiratory, thoracic and mediastinal AEsSkin and Subcutaneous Tissue AEsVascular AEs
Arm 1a (RAD001 Followed by LBH589)1830665413001082
Arm 1b (LBH589 Followed by RAD001)6105321311421
Doublet (Combination RAD001 and LBH589)423919234525411531

[back to top]

Evaluate the Overall Survival of Patients With Gastrointestinal Neuroendocrine Tumors Treated With Panobinostat

(NCT00985946)
Timeframe: Up to 5 years

Interventionmonths (Median)
Panobinostat47.27

[back to top]

Evaluate the Time to Progression for Patients With Gastrointestinal Neuroendocrine Tumors Treated With Panobinostat

(NCT00985946)
Timeframe: Up to 5 years

Interventionmonths (Median)
Panobinostat9.9

[back to top]

Number of Participants With Toxicities

Evaluate the toxicity and tolerability of panobinostat in the patient population (NCT00985946)
Timeframe: up to 5 years

Interventionparticipants (Number)
Panobinostat5

[back to top]

Tumor Response Rate of Patients With Gastrointestinal Neuroendocrine Tumors Using Response Evaluation Criteria in Solid Tumors (RECIST) Criteria.

Confirmed anti-tumor response rate will be validated by the Response Evaluation Criteria in Solid Tumors (RECIST). All participants included in the study will be assessed for response to the proposed panobinostat treatment, even if there are protocol treatment deviations. Each participant will be assigned one of the following categories: complete response, partial response, stable disease, progressive disease, early death from malignant disease, early death from toxicity, early death because of other cause, or unknown. (NCT00985946)
Timeframe: every 8 weeks, up to 5 years

Interventionparticipants (Number)
Stable DiseaseComplete ResponsePartial ResponseProgressive DiseaseEarly death from malignant diseaseEarly death from toxicityEarly death because of other causeUnknown
Panobinostat150000000

[back to top]

Tumor Response Rate to LBH589.

"per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v.1.0) for target lesions and assessed by CT/MRI: Response includes Complete Response (CR, disappearance of all target lesions), or Partial Response (PR, >=30% decrease in the sum of the longest diameter of target lesions). No Response includes Stable Disease (SD, neither sufficient shrinkage to qualify for partial response nor sufficient increase to qualify for progressive disease), and Progressive Disease (PD, at least a 20% increase in the sum of the longest diameters of target lesions, taking as reference the smallest sum longest diameter recorded since the baseline measurements, or the appearance of one or more new lesion(s).)" (NCT01013597)
Timeframe: Every 8 weeks.

Interventionparticipants (Number)
No responseResponse
LBH589130

[back to top]

Overall Survival

For a given patient, overall survival (OS) is defined as the number of days from the day of first LBH589 administration until the patient's death. If a patient was alive at the time of analysis, then the patient's data is censored at the date of the last available evaluation.Survival was assessed every three months until death or final data analysis, whichever occurred first. (NCT01013597)
Timeframe: Every 3 months up to 5 years

Interventionmonths (Median)
LBH58918.4

[back to top]

Impact of LBH589 on Tumor Markers for Thyroid Cancer

Change in serum Thyroglobulin level from baseline to end of treatment. Treatment continued until either extraordinary medical circumstances, disease progression, toxicity, subject withdrawal, or death. At the time subjects came off of study treatment for one of the reasons already listed, a sample was collected for tumor markers. (NCT01013597)
Timeframe: Baseline and end of treatment, up to 1 year

Interventionng/mL (Mean)
LBH5894.58

[back to top]

Time to Progression of Thyroid Cancer

Progression is defined using the 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. Time to progression is defined as the number of days from the day of first LBH589 administration to the day the patient experienced an event of disease progression or death, whichever came first. Progression was assessed every 3 months until death or up to 5 years, whichever occurred first. (NCT01013597)
Timeframe: Every 3 months until progression up to 5 years

Interventionmonths (Median)
LBH5893.6

[back to top]

Toxicity of LBH589

Most frequent toxicities at least possibly related to panobinostat, grades 2-4 (grading based on NCI common terminology criteria for adverse events CTCAE version 3). Toxicities were collected from the time the patient provided informed consent until 4 weeks after the patient stopped LBH589. (NCT01013597)
Timeframe: Every 4 weeks, up to 5 years

Interventionparticipants (Number)
AnemiaLeukopeniaNeutropeniaLymphopeniaThrombocytopeniaHypoalbuminemiaFatigueDiarrheaHeadacheBone PainHyperglycemiaHypocalcemiaAstheniaRashAnorexiaAnxietyElevated GGTPulmonary embolism
LBH589377883841112114111

[back to top]

Overall Response Rate in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.

Best overall response based on mEBMT criteria per investigator assessment (NCT01023308)
Timeframe: 45 months

Intervention% participants with response (Number)
Panobinostat + Bortezomib60.7
Placebo + Bortezomib54.6

[back to top]

European Organization for Research and Treatment of Cancer Multiple Myeloma Module (EORTC) QLQ-MY20-Change From Baseline by Treatment Group

"Higher values in the disease symptoms and side effects of treatment scores indicate worsening. Higher scores in the future perspective and body image scores indicate improvement. LS Means and SEM are estimated from the repeated measures model. Following factors and covariates are included in the repeated measurement model: time, treatment, treatment by time interaction, number of prior lines of anti-MM therapy (1/ 2 and 3), prior use of BTZ (Yes/ No), baseline score.Disease Symptom is the sum of 20 questions, total score ranges from 0 (best possible outcome) to 100 (worst possible outcome), All subscales of EORTC QLQ-MY20 have the same score range of 0 -100. Decrease in symptom scores from baseline indicate improvement in symptoms." (NCT01023308)
Timeframe: 12, 24 and 48 weeks

,
Interventionscore on a scale (Least Squares Mean)
Disease Symptom wk 12 change baseline (n=215,243)Disease Symptom wk 24 change baseline (n=148,177)Disease Symptom wk 48 change baseline (n=37,26)Side effects of treatment wk 12 chge (n=213,242)Side effects of treatment wk 24 chge (n=148,175)Side effects of treatment wk 48 chge (n=37,26)Future perspective wk 12 chge (n=214,242)Future perspective wk 24 chge (n=148,176)Future perspective wk 48 chge (n=37,26)Body image wk 12 chge (n=213,240)Body image wk 24 chge (n=147,175)Body image wk 48 chge (n=37,26)
Panobinostat + Bortezomib-4.795-4.401-2.8368.1629.0163.3575.3193.8774.331-7.178-11.463-2.161
Placebo + Bortezomib-4.865-6.797-6.6265.5247.7313.6546.1945.8396.951-6.22-7.358-4.666

[back to top]

European Organization for Research and Treatment of Cancer Multiple Myeloma Module (EORTC ) QLQ-C30 - Summary Statistics by Treatment Group

"The EORTC QLQ-C30 measures functional dimensions (physical, role, emotional, cognitive, and social), three multi-item symptom scales (fatigue, nausea/vomiting, and pain), six single-item symptom scales (dyspnea, sleep disturbance, appetite loss, constipation, diarrhea and financial impact) and a global health status/QoL scale. Disease Symptom is the sum of 30 questions, total score ranges from 0 (best possible outcome) to 100 (worst possible outcome), All subscales of EORTC QLQ-C30 have the same score range of 0 -100. For global health status and other functional scales,an increase from baseline indicates improvement of QoL. Whereas for symptoms scales, fatigue, dyspnea, insomnia, appetite loss, constipation and diarrhea, decrease in scores from baseline indicate improvement in symptoms." (NCT01023308)
Timeframe: 12, 24 and 48 weeks

,
Interventionscore on a scale (Least Squares Mean)
Global health wk 12 change baseline (n=216,239)Global health wk 24 change baseline (n=150,176)Global health wk 48 change baseline (n=38,26)Physical functioning wk 12 chge (n=217,242)Physical functioning wk 24 chge (n=151,177)Physical functioning wk 48 chge (n=38,26)Role functioning wk 12 chge (n=215,237)Role functioning wk 24 chge (n=150,176)Role functioning wk 48 chge (n=38,26)Cognitive functioning wk 12 chge (n=216,240)Cognitive functioning wk 24 chge (n=149,176)Cognitive functioning wk 48 chge (n=38,26)Social functioning wk 12 chge (n=216,240)Social functioning wk 24 chge (n=148,171)Social functioning wk 48 chge (n=37,26)Fatigue wk 12 chge (n=217,241)Fatigue wk 24 chge (n=151,176)Fatigue wk 48 chge(n=38,26)Dyspnea wk 12 chge (n=217,240)Dyspnea wk 24 chge (n=151,177)Dyspnea wk 48 chge (n=38,26)Insomnia wk 12 chge (n=216,239)Insomnia wk 24 chge (n=149,176)Insomnia wk 48 chge (n=38,26)Appetite loss wk 12 chge (n=217,239)Appetite loss wk 24 chge (n=151,176)Appetite loss wk 48 chge (n=38,26)Constipation wk 12 chge (n=215,240)Constipation wk 24 chge (n=151,177)Constipation wk 48 chge (n=38,25)Diarrhea wk 12 chge (n=217,241)Diarrhea wk 24 chge (n=150,177)Diarrhea wk 48 chge (n=38,26)
Panobinostat + Bortezomib-9.853-7.867-2.986-9.67-9.516-2.88-11.159-11.875-5.927-4.464-6.053-5.568-8.502-8.925-6.10415.12212.6774.64613.9647.9394.1186.28310.023-2.46415.16716.5743.9994.135-0.153-0.35818.88823.16320.48
Placebo + Bortezomib-4.044-1.5184.345-5.393-6.4562.037-6.762-11.263-0.401-1.023-3.542-4.042-3.991-6.3384.6177.9399.203-2.6256.2665.3082.827.6256.104-3.4425.3835.861-2.9636.420.524-0.94610.20616.40610.996

[back to top]

Time to Response Per Investigator Assessment (mEBMT Criteria) of Response Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.

(NCT01023308)
Timeframe: 45 months

Interventiontime to response in months (Median)
Panobinostat + Bortezomib1.51
Placebo + Bortezomib2.00

[back to top]

Time to Progression/Relapse Per Investigator Assessment (mEBMT Criteria) Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.

(NCT01023308)
Timeframe: 45 months

Interventionmonths (Median)
Panobinostat + Bortezomib12.71
Placebo + Bortezomib8.54

[back to top]

Progression-free Survival Events in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.

(NCT01023308)
Timeframe: 45 months

Interventionnumber of events (Number)
Panobinostat + Bortezomib207
Placebo + Bortezomib260

[back to top]

Progression Free Survival in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.

(NCT01023308)
Timeframe: 45 months

Interventionmonths (Median)
Panobinostat + Bortezomib11.99
Placebo + Bortezomib8.80

[back to top]

Overall Survival in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone

survival time in months (NCT01023308)
Timeframe: 45 months

Interventionmonths (Median)
Panobinostat + Bortezomib40.28
Placebo + Bortezomib35.78

[back to top]

Duration of Response Per Investigator Assessment (mEBMT Criteria) Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone.

(NCT01023308)
Timeframe: 45 months

Interventionduration of response in months (Median)
Panobinostat + Bortezomib13.14
Placebo + Bortezomib10.87

[back to top]

Overall Survival in Patients Treated With Panobinostat in Combination With Bortezomib and Dexamethasone vs. Patients Treated by Placebo in Combination With Bortezomib and Dexamethasone

Number of OS events (NCT01023308)
Timeframe: 45 months

InterventionNumber of OS events (Number)
Panobinostat + Bortezomib134
Placebo + Bortezomib152

[back to top]

Functional Assessment of Chronic Illness Therapy (FACIT) Measurement System : FACT/GOG-NTX-Change From Baseline by Treatment Group

Chronic Illness Therapy (FACIT) Measurement System and focuses on four general quality of life domains for physical well being, functional well-being, social/family well-being, and emotional well-being, and includes additional items to characterize treatment-related neurotoxicity. Higher subscales/total scores represent higher QOL. In the case of the neurotoxicity subscale, lower scores correspond to higher neurotoxicity. The recall period referenced in the questionnaire is the past 7 days.Ranges for FACT-G subscales are as follows:.PWB, scale 0 -28, , NtxS scale 0-44, FACT/GOG-Ntx trial outcome index scale is 0-100 and FACT-G scale is also scaled 0-100. An increase from baseline in these scores indicate improvement. (NCT01023308)
Timeframe: 12, 24 and 48 weeks

,
Interventionscore on a scale (Least Squares Mean)
Neurotoxicity wk 12 change baseline (n=212,240)Neurotoxicity wk 24 change baseline (n=148,174)Neurotoxicity wk 48 change baseline (n=35,26)Physical wellbeing wk 12 chge (n=215,240)Physical wellbeingwk 24 chge (n=150,176)Physical wellbeing wk 48 chge (n=38,26)Trial Outcomes wk 12 chge (n=209,236)Trial Outcomes wk 24 chge (n=148,173)Trial Outcomes wk 48 chge (n=35,26)FACT-G Total wk 12 chge (n=213,240)FACT-G Totalwk 24 chge (n=147,175)FACT-G Total wk 48 chge (n=37,26)FACT/GOGNTX Total wk 12 chge (n=206,230)FACT/GOGNTX Total wk 24 chge (n=146,172)FACT/GOGNTX Total wk 48 chge (n=35,26)
Panobinostat + Bortezomib-4.481-4.564-3.158-3.29-3.044-2.037-10.573-9.84-6.633-6.658-6.076-2.704-11.176-10.581-5.871
Placebo + Bortezomib-3.337-4.739-2.133-1.952-2.2590.203-6.874-8.894-2.821-4.106-4.609-1.435-7.524-9.179-3.151

[back to top]

Number of Participants With Adverse Events

Safety monitoring was conducted throughout the study. (NCT01034163)
Timeframe: 23 months

InterventionParticipants (Number)
Panobinostat (PAN)26
Placebo11

[back to top]

Frequency Distribution of IPSS Score Status - Randomized Phase

The IPSS score values were calculated based on the results of bone marrow analysis. A score value of 0 has bone marrow blast <5%, karyotype of normal, sole: -Y, del 5Q, del 20q and cytopenias (lineages affected) of 0 to 1. Score value of 0.5 has 5-10 bone marrow blasts, karyotype of Others and cytopenias of 2 to 3. A score value of 1.0 has complex >= 3 chromosomal abnormalities and/or chromosome 7 anomalies. A score of 1.5 has 11-20 bone marrow blasts and a score of 2.0 has 21-30 bone marrow blasts. The prognostic score is determined by the sum of the single scoring values. The risk groups are determined as follows: Low = 0 points (5.7 years of median survival); intermediate -1 (INT-1) = 0.5-1.0 points (3.5 years of median survival); INT-2 = 1.5-2.0 points (1.2 years of median survival); and high >=2.5 points (6 months of median survival). (NCT01034657)
Timeframe: 52 weeks

,,
InterventionPercentage of participants (Number)
Week 52 (randomized phase), LowWeek 52 (randomized phase), , INT-1Week 52 (randomized phase), INT-2Week 52 (randomized phase), HIgh
LBH5890.016.70.00.0
LBH589 + Epoetin Alfa0.016.70.00.0
Not Randomized100.00.00.00.0

[back to top]

Percentage of Participants With Objective Response During the Randomized Phase

Objective response (complete remission (CR) + partial remission (PR) and HI-platelet (HI-P) response + HI-neutrophil (HI-N) response) was assessed according to the modified IWG criteria: CR bone marrow with 5% myeloblasts with normal maturation of al cell lines (persistent dysplasia is noted) and peripheral blood with Hgb >= 11 g/dL platelets >=100 X 10^9/L, neutrophils >= 1.0 x 10^9/L and blasts 0%. PR = All CR if abnormal before treatment except bone marrow blasts decreased by>=50% over pretreatment but still >5% (ellularity and morphology not relevant). HI-P (pretreatment, < 100 x 109/L) = absolute increase of ≥ 30 x 109/L for participants starting with > 20 x 109/L and platelets Increase from < 20 x 109/L to > 20 x 109/L and by at least 100%; HI-N (pretreatment, < 1.0 x 109/L) = at least 100% increase and an absolute increase > 0.5 x 10^9/L. (NCT01034657)
Timeframe: 32 weeks, 48 weeks

,,
InterventionPercentage of participants (Number)
Week 32 ( n=5,1,1)Week 48 (n=6,5,1)
LBH5890.00.0
LBH589 + Epoetin Alfa0.00.0
Not Randomized0.00.0

[back to top]

Frequency Distribution of IPSS Score Status - Core Phase

The IPSS score values were calculated based on the results of bone marrow analysis. A score value of 0 has bone marrow blast <5%, karyotype of normal, sole: -Y, del 5Q, del 20q and cytopenias (lineages affected) of 0 to 1. Score value of 0.5 has 5-10 bone marrow blasts, karyotype of Others and cytopenias of 2 to 3. A score value of 1.0 has complex >= 3 chromosomal abnormalities and/or chromosome 7 anomalies. A score of 1.5 has 11-20 bone marrow blasts and a score of 2.0 has 21-30 bone marrow blasts. The prognostic score is determined by the sum of the single scoring values. The risk groups are determined as follows: Low = 0 points (5.7 years of median survival); intermediate -1 (INT-1) = 0.5-1.0 points (3.5 years of median survival); INT-2 = 1.5-2.0 points (1.2 years of median survival); and high >=2.5 points (6 months of median survival). (NCT01034657)
Timeframe: baseline

InterventionPercentage of participants (Number)
Baseline (core phase), LowBaseline (core phase),, INT-1Baseline (core phase),, INT-2Baseline (core phase), High
LBH58932.467.60.00.0

[back to top]

Percentage of Participants With HI-E - Randomized Phase

HI-E was assessed according to the modified international working group (IWG) criteria for HI. Erythroid response (pretreatment, <11 g/dL): Hgb increase by ≥ 1.5 g/dL, relevant reduction of units of RBC transfusions by an absolute number of at least 4 RBC transfusions/8 wk compared with the pretreatment transfusion number in the previous 8 wk, and only RBC transfusions given for a Hgb of ≤ 9.0 g/dL pretreatment were counted in the RBC transfusion response evaluation; Platelet response (pretreatment, < 100 x 109/L): absolute increase of ≥ 30 x 109/L for participants starting with > 20 x 109/L and platelets Increase from < 20 x 109/L to > 20 x 109/L and by at least 100%; Neutrophil response (pretreatment, < 1.0 x 109/L): at least 100% increase and an absolute increase > 0.5 x 109/L; Progression or relapse after HI: At least 1 of the following: At least 50% decrement from maximum response levels in granulocytes or platelets, reduction in Hgb by ≥1.5 g/dL, or transfusion dependence. (NCT01034657)
Timeframe: 32 weeks, 52 weeks

,,
InterventionPercentage of participants (Number)
32 weeks (n=5,1,1)52 weeks (n=4,3,1)
LBH5890.00.0
LBH589 + Epoetin Alfa0.00.0
Not Randomized0.00.0

[back to top]

Percentage of Participants With Objective Response During Core Phase

Objective response (complete remission (CR) + partial remission (PR) and HI-platelet (HI-P) response + HI-neutrophil (HI-N) response) was assessed according to the modified IWG criteria: CR bone marrow with 5% myeloblasts with normal maturation of al cell lines (persistent dysplasia is noted) and peripheral blood with Hgb >= 11 g/dL platelets >=100 X 10^9/L, neutrophils >= 1.0 x 10^9/L and blasts 0%. PR = All CR if abnormal before treatment except bone marrow blasts decreased by>=50% over pretreatment but still >5% (ellularity and morphology not relevant). HI-P (pretreatment, < 100 x 109/L) = absolute increase of ≥ 30 x 109/L for participants starting with > 20 x 109/L and platelets Increase from < 20 x 109/L to > 20 x 109/L and by at least 100%; HI-N (pretreatment, < 1.0 x 109/L) = at least 100% increase and an absolute increase > 0.5 x 10^9/L. (NCT01034657)
Timeframe: 16 weeks

InterventionPercentage of participants (Number)
LBH5890.0

[back to top]

Percentage of Participants With Hematological Response of the Erythropoetic System (HI-E) - Core Phase

HI-E was assessed according to the modified international working group (IWG) criteria for HI. Erythroid response (pretreatment, <11 g/dL): Hgb increase by ≥ 1.5 g/dL, relevant reduction of units of RBC transfusions by an absolute number of at least 4 RBC transfusions/8 wk compared with the pretreatment transfusion number in the previous 8 wk, and only RBC transfusions given for a Hgb of ≤ 9.0 g/dL pretreatment were counted in the RBC transfusion response evaluation; Platelet response (pretreatment, < 100 x 109/L): absolute increase of ≥ 30 x 109/L for participants starting with > 20 x 109/L and platelets Increase from < 20 x 109/L to > 20 x 109/L and by at least 100%; Neutrophil response (pretreatment, < 1.0 x 109/L): at least 100% increase and an absolute increase > 0.5 x 109/L; Progression or relapse after HI: At least 1 of the following: At least 50% decrement from maximum response levels in granulocytes or platelets, reduction in Hgb by ≥1.5 g/dL, or transfusion dependence. (NCT01034657)
Timeframe: 16 weeks

InterventionPercentage of participants (Number)
LBH5890.0

[back to top]

Overall Survival (OS) - Overall Period

OS was defined as the time from start of treatment to death from any cause. (NCT01034657)
Timeframe: 48 weeks

Interventionmonths (Median)
LBH589NA

[back to top]

Mean Single Scoring Values of the IPSS - Randomized Phase

The IPSS score values were calculated based on the results of bone marrow analysis. A score value of 0 has bone marrow blast <5%, karyotype of normal, sole: -Y, del 5Q, del 20q and cytopenias (lineages affected) of 0 to 1. Score value of 0.5 has 5-10 bone marrow blasts, karyotype of Others and cytopenias of 2 to 3. A score value of 1.0 has complex >= 3 chromosomal abnormalities and/or chromosome 7 anomalies. A score of 1.5 has 11-20 bone marrow blasts and a score of 2.0 has 21-30 bone marrow blasts. The prognostic score is determined by the sum of the single scoring values. The risk groups are determined as follows: Low = 0 points (5.7 years of median survival); intermediate -1 (INT-1) = 0.5-1.0 points (3.5 years of median survival); INT-2 = 1.5-2.0 points (1.2 years of median survival); and high >=2.5 points (6 months of median survival). (NCT01034657)
Timeframe: 52 weeks

Interventionunits on a scale (Mean)
LBH5891.0
LBH589 + Epoetin Alfa1.0
Not Randomized0.0

[back to top]

Mean Single Scoring Values of the IPSS - Core Phase

The IPSS score values were calculated based on the results of bone marrow analysis. A score value of 0 has bone marrow blast <5%, karyotype of normal, sole: -Y, del 5Q, del 20q and cytopenias (lineages affected) of 0 to 1. Score value of 0.5 has 5-10 bone marrow blasts, karyotype of Others and cytopenias of 2 to 3. A score value of 1.0 has complex >= 3 chromosomal abnormalities and/or chromosome 7 anomalies. A score of 1.5 has 11-20 bone marrow blasts and a score of 2.0 has 21-30 bone marrow blasts. The prognostic score is determined by the sum of the single scoring values. The risk groups are determined as follows: Low = 0 points (5.7 years of median survival); intermediate -1 (INT-1) = 0.5-1.0 points (3.5 years of median survival); INT-2 = 1.5-2.0 points (1.2 years of median survival); and high >=2.5 points (6 months of median survival). (NCT01034657)
Timeframe: baseline

Interventionunits on a scale (Mean)
LBH5890.43

[back to top]

Number of Participants by Tumor Response

Number of patients whose tumor has responded to study therapy is determined using Response Evaluation Criteria In Solid Tumors. Progressive Disease (PD) is assessed if the sum of the diameters has increased by ≥ 20% and ≥ 5 mm from nadir (including baseline if it is the smallest sum). Objective response is measured by tumor reduction as defined in the RECIST criteria. Tumor shrinkage must be at least 30% to qualify as an objective response. (NCT01056601)
Timeframe: Up to 1 Year

InterventionParticipants (Number)
Progressive DiseaseNo data availableObjective Response
Pancreatic Cancer Patients520

[back to top]

Progression-Free Survival

Median number of months before disease progressed in patient on gemcitabine when treated with the combination of panobinostat and bortezomib. Progression free survival is measured from randomization until the subject has documented disease progression by an objective measure. Subjects must be alive with no more than 20% increase in tumor size to qualify for progression free survival. Changes in tumor size are defined by RECIST criteria. (NCT01056601)
Timeframe: Up to 1 Year

InterventionMonths (Median)
Pancreatic Cancer Patients2.1

[back to top]

Duration of Response

Duration of response is calculated as (Date of First Disease Progression or Death as a Result of any Cause whichever Comes First - Date of First Objective Status Assessment of Confirmed Complete or Partial Response as defined by RECIST criteria). (NCT01056601)
Timeframe: Up to 1 Year

InterventionWeeks (Number)
Pancreatic Cancer Patients0

[back to top]

Over All Survival

Kaplan Meier estimates- median time to event (NCT01083602)
Timeframe: 24 weeks

InterventionDays (Median)
Panobinostat + Bortezomib & Dexamethasone559.0

[back to top]

Overall Response Rate (PR+nCR+CR)

Overall response rate=(PR+nCR+CR) CR= < 5% plasma cells in bone marrow. No confirmation on bone marrow plasma cell (additional assessment) is needed to document CR except patients with non-secretory myeloma where the bone marrow examination must be repeated after an interval of at least 6 weeks, Absence of M-protein in serum and urine by immunofixation,nCR same as CR without out Absence of M-protein in serum and urine by immunofixation,PR+ 50% reduction of serum M-protein and sofft tissue Plasmacytomas all for more than 6 weeks. (NCT01083602)
Timeframe: after eight cycyles of treatment (24 weeks)

Interventionpercentage of participants (Number)
Panobinostat + Bortezomib & Dexamethasone34.5

[back to top]

Progression-free Survival

Progression-free survival (PFS) was defined as the time from the date of first study treatment to first occurrence of documented progressive disease /relapse or death. Time from randomization until disease progression or death by Kaplan-Meier estimates (NCT01083602)
Timeframe: 24 weeks

Interventiondays (Median)
Panobinostat + Bortezomib & Dexamethasone164.0

[back to top]

Time to Response (Greater Than or Equal to PR) Based on Investigator Assessment

Time to response is defined as the time from the date of first administration of study treatment to the date of first documented evidence of CR or nCR or PR (whichever status is recorded first). Patients who do not have a response of PR or better by the data cut-off date are censored. (NCT01083602)
Timeframe: after eight cycyles of treatment (24 weeks)

InterventionDays (Mean)
Panobinostat + Bortezomib & Dexamethasone51.8

[back to top]

Responders to Treatment

The primary endpoint for this phase II study of patients with bortezomib-refractory MM is response after a maximum of 8 cycles of therapy as defined by the modified EBMT criteria. (NCT01083602)
Timeframe: after eight cycyles of treatment (24 weeks)

Interventionparticipants (Number)
Complete Response (CR)near Complete Response(nCR)Partial Response (PR)Minimal Response (MR)No ChangePregressive Disease (PD)Unknown
Panobinostat + Bortezomib & Dexamethasone0118102033

[back to top]

Time to Progression

Time from randomization until objective tumor progression; does not include deaths-- Kaplan-Meier estimates (NCT01083602)
Timeframe: 24 weeks

InterventionDays (Median)
Panobinostat + Bortezomib & Dexamethasone164.0

[back to top]

Number of Participants With Adverse Events (AEs)

"Investigators intended to evaluate the safety and tolerability profile of LBH589. Assessments would consist of monitoring and recording all adverse events and serious adverse events, the regular monitoring of hematology, blood chemistry and urine values, vital signs, ECOG performance status, and the regular physical examinations and ECG assessments.~Adverse events will be assessed according to the Common Toxicity Criteria for Adverse Events (CTCAE) version 3.0. CTCAE v3.0 can be accessed on the National Institute of Health (NIH)/NCI website at http://ctep.cancer.gov/forms/CTCAEv3.pdf." (NCT01090973)
Timeframe: 8 weeks (2 cycles) unless treatment continues due to partial or complete response

Interventionparticipants (Number)
Oral Drug Treatment1

[back to top]

Clinical Benefit Rate

Clinical benefit rate will be estimated by the total number of patients with an objective status of CR, PR, or SD for duration of at least 6 months divided by the total number of evaluable patients. All evaluable patients will be used for this analysis. Exact binomial 95% confidence intervals for the true clinical benefit rate will be calculated. (NCT01105312)
Timeframe: from baseline up to 6 months

Interventionpercentage of participants (Number)
Phase II0

[back to top]

Duration of Response (Phase II)

Duration of response is defined for all evaluable patients who have achieved a confirmed response as the date at which the patient's objective status is first noted to be a CR or PR to the earliest date progression is documented. The distribution of duration of response will be estimated using the method of Kaplan-Meier. (NCT01105312)
Timeframe: from baseline up to 5 years post-registration

Interventionmonths (Median)
Phase IINA

[back to top]

Confirmed Response Rate (Phase I)

A confirmed response is defined to be a CR or PR (as determined by RECIST criteria) noted as the objective status on 2 consecutive evaluations at least 4 weeks apart. Response will be evaluated using all cycles of treatment. All patients meeting the eligibility criteria who have signed a consent form and have begun treatment will be evaluable for response. The number of confirmed responses will be reported here. (NCT01105312)
Timeframe: from baseline up to 5 years

InterventionParticipants (Count of Participants)
Phase I: Dose Level One0
Phase I: Dose Level Two2

[back to top]

Maximum-tolerated Dose (Phase I)

MTD is defined as the dose level below the lowest dose that induces dose limiting toxicity in at least one-third of patients (at least 2 of a maximum of 6> new patients). If dose-limiting toxicity (DLT) is not seen in any of the 3 patients, 3 new patients will be accrued and treated at the next higher dose level. If DLT are seen in 2 or 3 of 3 patients treated at a given dose level, then the next 3 patients will be treated at the next lower dose level, if only 3 patients were enrolled and treated at this lower dose level. The number of DLT's will be reported here. (NCT01105312)
Timeframe: Up to 2.5 months

InterventionParticipants (Count of Participants)
Phase I: Dose Level One1
Phase I: Dose Level Two3

[back to top]

Progression-free Survival (Phase II)

Progression-free survival (PFS) is defined as the time from registration to progression or death due to any cause. PFS at 6 months will be estimated. The distribution of PFS will be estimated using the method of Kaplan-Meier. (NCT01105312)
Timeframe: from baseline up to 6 months

Interventionmonths (Median)
Phase II2.1

[back to top]

Response Rate (Phase II)

"A confirmed response is defined to be a CR or PR (as determined by RECIST (version 1.1 criteria) noted as the objective status on 2 consecutive evaluations at least 4 weeks apart. Response will be evaluated using all cycles of treatment. All patients meeting the eligibility criteria who have signed a consent form and have begun treatment will be evaluable for response.~A CR is defined as:~All of the following must be true:~Disappearance of all non-nodal target lesions~Each target lymph node must have reduction in short axis to <1.0 cm~A PR is defined as:~At least a 30% decrease in the sum of the longest diameters of the non-nodal target lesions and the short axes of the target lymph nodes taking as reference the BSD (Section 11.41)" (NCT01105312)
Timeframe: from baseline up to 5 years post-registration

Interventionpercentage of participants (Number)
Phase II0

[back to top]

Survival Time (Phase II)

Survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier (NCT01105312)
Timeframe: from baseline up to 5 years post-registration

Interventionmonths (Median)
Phase II16.1

[back to top]

Time to Treatment Failure

Time to treatment failure (TTF) is defined as the time from the date of registration to the date at which the patient is removed from treatment due to progression, unacceptable adverse events, or refusal. The distribution of TTF will be estimated using the method of Kaplan-Meier (NCT01105312)
Timeframe: from baseline up to 5 years post-registration

Interventionmonths (Median)
Phase II2

[back to top]

Time-to-disease Progression (Phase II)

"Time-to-disease progression (TTP) is defined as the time from registration to documentation of disease progression. If a patient dies without a documentation of disease progression, the patient will be considered to have had tumor progression at the time of their death unless there is sufficient documented evidence to conclude no progression occurred prior to death. The distribution of TTP will be estimated using the method of Kaplan-Meier. Progression is defined as at least one of the following:~At least one new malignant lesion or a lymph node whose short axis has increased to >1.5 cm~At least a 20% increase in the sum of diameters of target lesions taking as reference the MSD. In addition, the sum must also demonstrate an absolute increase of at least 0.5 cm" (NCT01105312)
Timeframe: from baseline up to 6 months

Interventionmonths (Median)
Phase II2.1

[back to top] [back to top]

Chronic GVHD Severity at MTD

Chronic GVHD maximum severity grade at MTD, in participants without overlap syndrome at initiation of study therapy. Maximum c-GVHD severity: Mild, Moderate, Severe. (NCT01111526)
Timeframe: Up to 1 year

Interventionparticipants (Number)
MildModerateSevere
LBH589, in Addition to Glucocorticoids211

[back to top]

Occurrence of Discontinuation of All Immune Suppression

Number of participants discontinuing all immune suppression without subsequent flare by 1 year post initiation of therapy. (NCT01111526)
Timeframe: 1 year

Interventionparticipants (Number)
LBH589, in Addition to Glucocorticoids0

[back to top]

Phase II: Overall Rate of Response (ORR)

Rate of Complete Response (CR), Partial Response (PR), Progressive Disease (PD) and Stable Disease (SD). Assessment of GVHD will include the skin, liver and gut. Other possible etiologies of organ disease such as C difficile enterocolitis, viral infection, drug reaction, veno-occlusive disease of the liver, etc., will be excluded by appropriate tests. CR is defined as resolution of GVHD in all evaluable organs with no subsequent additional treatment given for acute GVHD. PR is defined as improvement in ≥ one evaluable organ without deterioration in at least one other. PD is defined as deterioration in at least on evaluable organ. SD is defined as the absence of any difference sufficient to meet minimal criteria for improvement or deterioration in any evaluable organs. (NCT01111526)
Timeframe: 1 year, 2 months

Interventionparticipants (Number)
Complete ResponsePartial ResponseProgressive DiseaseStable Disease
LBH589, in Addition to Glucocorticoids13210

[back to top]

Chronic GVHD Onset

Chronic GVHD onset in participants without overlap syndrome at initiation of study therapy. Number of participants with overlap syndrome at MTD. (NCT01111526)
Timeframe: Up to 1 year

Interventionparticipants (Number)
LBH589, in Addition to Glucocorticoids6

[back to top]

Incidence of GVHD Flares Requiring Increasing Immune Suppressive Therapy

Number of participants with GVHD flares requiring increasing immune suppressive therapy within 36 days of study initiation. Cumulative incidence of GVHD flares requiring increasing immune suppressive therapy will be analyzed using the competing risk method by Gray (1988). GVHD flares (progressive disease (PD)) may result in discontinuation from Panobinostat. (NCT01111526)
Timeframe: Up to 36 days per participant

Interventionparticipants (Number)
LBH589, in Addition to Glucocorticoids1

[back to top]

Phase I: Maximum Tolerated Dose (MTD) in Milligrams

MTD of LBH589 in addition to glucocorticoids as treatment for Graft Versus Host Disease (GVHD) manifestations. MTD in Milligrams (mg), taken by mouth (PO), 3 times per week, for 4 weeks. The oral formulation replaced the IV formulation (which became unavailable) after the first 4 participants were treated. Dose limiting toxicity (DLT) is defined by the occurrence of Common Toxicity Criteria (CTC) grade 3 or greater toxicity that is unexpected with transplantation, except for hematological toxicity, where DLT is defined as absolute neutrophil count (ANC) <750, and for those participants who were platelet transfusion independent is defined as platelets <10 K. (NCT01111526)
Timeframe: 2 years, 8 months

InterventionMTD of oral LBH589 in milligrams (Number)
LBH589, in Addition to Glucocorticoids5

[back to top]

Stable or Improved Chronic GVHD Severity Score

Stable or improved Chronic GVHD score: Improved Mild to None; Improved Severe to Moderate; Improved Moderate to None, Remained Stable at Mild. (NCT01111526)
Timeframe: 1 year

Interventionparticipants (Number)
Improved: Mild to NoneImproved: Severe to ModerateImproved: Moderate to NoneStable: Remained Stable at Mild
LBH589, in Addition to Glucocorticoids1111

[back to top]

Overall Survival (OS)

Overall Survival (OS) at one year post initiation of therapy. (NCT01111526)
Timeframe: 1 year

Interventionparticipants (Number)
OS at 1 yearDeath before 1 year: sepsisDeath before 1 year: relapseDeath before 1 year: GVHDDeath before 1 year: Cardiogenic shock
LBH589, in Addition to Glucocorticoids111211

[back to top]

Number of Participants With Complete Remission (CR)

Will be assessed by Kaplan-Meier methods. (NCT01169636)
Timeframe: Assessed after 3 cycles of ICE (2 months)

,,
InterventionParticipants (Count of Participants)
Complete ResponsePartial ResponseStable DiseaseProgressive Disease
Phase I: Panobinostat + ICE (Maximal Tolerated Dose (MTD)21422
Phase II Panobinostat + ICE9020
Phase II: Standard of Care (ICE)8121

[back to top]

Percentage of Participants With Failure Free Survival (FFS)

FFS duration was calculated from date of study entry to date of progression or death or change of therapy, whichever came first. (NCT01169636)
Timeframe: 16 months

Interventionpercentage of participants (Number)
Phase I: Panobinostat + ICE (Maximal Tolerated Dose (MTD)61
Phase II: Standard of Care (ICE)82
Phase II Panobinostat + ICE75

[back to top]

Maximum Tolerated Dose (MTD) of Panobinostat + ICE

Maximum Tolerated Dose (MTD) of Panobinostat + ICE (NCT01169636)
Timeframe: From first dose of panobinostat (or chemotherapy, in the arm of ICE alone) until 30 days after last dose, up to 6 years

Interventionmg (Number)
Phase I: Panobinostat + ICE (Maximal Tolerated Dose (MTD)30

[back to top]

Proportion of Confirmed Responses Defined to be a CR or PR Noted as the Objective Status

"The primary endpoint of this phase II trial is the proportion of confirmed responses (complete response (CR) or partial response (PR)) noted as the objective status and will be considered synonymous with success for this study.Response will be evaluated using all cycles of treatment. A CR is defined using the Cheson et al. Revised Response Criteria for Malignant Lymphoma as Disappearance of all evidence of disease. A PR is defined as Regression of measurable disease and no new sites with ≥50% decrease in SPD of up to 6 largest dominant masses; no increase in size of other nodes. The proportion of successes will be estimated by the number of successes divided by the total number of evaluable patients." (NCT01261247)
Timeframe: Every 28 days for up to 2 years

Interventionproportion of CR or PR patients (Number)
Arm I (LBH589)0.21

[back to top]

Duration of Response

Duration of response is defined for all evaluable patients who have achieved a confirmed response as the date at which the patient's objective status is first noted to be a CR or PR to the earliest date progression (PD) is documented. The distribution of duration of response will be estimated using the method of Kaplan-Meier. (CR: Disappearance of all evidence of disease, PR: Regression of measurable disease and no new sites, PD: Any new lesion or increase by ≥50% of previously involved sites from nadir). (NCT01261247)
Timeframe: Every 6 months for up to 2 years

Interventionmonths (Median)
Arm I (LBH589)19.7

[back to top]

Median Overall Survival Time

The median overall survival time is defined as the time from registration to death due to any cause. The distribution of survival time will be estimated using the method of Kaplan-Meier. (NCT01261247)
Timeframe: Every 6 months for up to 2 years

Interventionmonths (Median)
Arm I (LBH589)14.9

[back to top]

Median Progression-free Survival Time

The median progression-free survival time is defined as the time from registration to progression or death due to any cause. The distribution of progression-free survival will be estimated using the method of Kaplan-Meier. Progression is defined using the Cheson et al. Revised Response Criteria for Malignant Lymphoma as: Any new lesion or increase by ≥50% of previously involved sites from nadir, Appearance of a new lesion(s) > 1.5 cm in any axis, ≥50% increase from nadir in SPD of more than one node, or ≥50% increase in longest diameter of a previously identified node > 1 cm in short axis, Lesions PET positive if FDG-avid lymphoma or PET positive prior to therapy, > 50% increase from nadir in the SPD of any previous lesions, New or recurrent involvement. (NCT01261247)
Timeframe: Every 6 months for up to 2 years

Interventionmonths (Median)
Arm I (LBH589)3.1

[back to top]

Overall Response Rate

"Overall response rate is defined by the Revised International Workshop Response Criteria (2007) (see reference in protocol section).~Overall response (OR) = Complete response (CR) + Partial response (PR)" (NCT01282476)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Panobinostat/Rituximab11

[back to top]

Toxicities

Evaluate safety of this combination in relapsed/refractory DLBCL patients Grade 1 = mild, Grade 2 = moderate, Grade 3 = severe, Grade 4 = life-threatening (NCT01282476)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Platelet count decreased72314638Fatigue72314638Anemia72314638Diarrhea72314638Nausea72314638Lymphocyte count decreased72314638Anorexia72314638Hypophosphatemia72314638Neutrophil count decreased72314638Hypoalbuminemia72314638Dyspnea72314638Constipation72314638Alkaline phosphatase increased72314638Creatinine increased72314638White blood cell decreased72314638Hypocalcemia72314638Hypokalemia72314638Hypomagnesemia72314638Dry mouth72314638Flatulence72314638Gatstroesophageal reflux disease72314638Mucositis oral72314638Vomiting72314638Edema face72314638Edema limbs72314638Fever72314638Hepatobiliary disorders - other72314638Aspartate aminotransferase increased72314638Blood bilirubin increased72314638Metabolism and nutrition disorders - other72314638Bone pain72314638Dysgeusia72314638Headache72314638Insomnia72314638Epistaxis72314638Skin and subcutaneous tissue disorders - other72314638
Grade 3Grade 4Did not have anyGrade 1Grade 2
Panobinostat/Rituximab7
Panobinostat/Rituximab8
Panobinostat/Rituximab5
Panobinostat/Rituximab2
Panobinostat/Rituximab10
Panobinostat/Rituximab13
Panobinostat/Rituximab4
Panobinostat/Rituximab14
Panobinostat/Rituximab3
Panobinostat/Rituximab15
Panobinostat/Rituximab16
Panobinostat/Rituximab17
Panobinostat/Rituximab1
Panobinostat/Rituximab0

[back to top]

Progression-free Survival Rate

Progression is defined by the Revised International Workshop Response Criteria (2007) (see reference in protocol section). (NCT01282476)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Panobinostat/Rituximab6

[back to top]

Assess the Safety and Tolerability of Combined Lenalidomide and Panobinostat in Patients With Previously Treated Hodgkin's Lymphoma.

Safety and tolerability will be assessed for patients using the NIH-NCI Common Terminology Criteria (CTCAE) version 4.0 (NCT01460940)
Timeframe: up to 24 months

Interventionpercentage of patients (Number)
neutropeniathrombocytopeniafebrile neutropeniahypophosphatemia
Lenalidomide and Panobinostat58.341.725.025.0

[back to top]

Progression-free Survival in Patients With Previously Treated Hodgkin's Lymphoma Receiving Combined Lenalidomide and Panobinostat

Determined from the date of start of therapy to death from any cause or censored at the last date the patient is known to be alive (NCT01460940)
Timeframe: 3-5 years

Interventionmonths (Median)
Lenalidomide and Panobinostat3.8

[back to top]

Determine the Overall Response Rate (ORR), Including Complete Responses (CR) and Partial Responses (PR)

Overall response rate (CR + PR) will be determined using the International response criteria with combined panobinostat and lenalidomide in patients with relapsed or refractory Hodgkin's lymphoma. (NCT01460940)
Timeframe: up to 24 months

Interventionpercentage of patients (Number)
Lenalidomide and Panobinostat16.7

[back to top]

Number of Phase I Patients (Dose Level 1-6) Experiencing a Dose-Limiting Toxicity (DLT) to Determine the Optimal Dosage

"Using a traditional 3+3 dose escalation design, successive cohorts of subjects will receive a fixed dose level of the drug combination. The MTD is defined as the highest dose level at which ≤1 of 6 subjects experiences a dose-limiting toxicity (DLT) assessed by NCI CTCAE v4.0. DLT is defined as any of the following that are determined to be related to study treatment during Cycle 1:~Grade 4 neutropenia for >7 days, Febrile neutropenia, Grade 3 thrombocytopenia with ≥ Grade 2 bleeding, Grade 4 thrombocytopenia > 7 days, ≥ Grade 2 neuropathy with uncontrolled pain, ≥ Grade 3 non-hematologic drug-related toxicity (excluding alopecia), despite optimal supportive care lasting >72 hours or requiring a dose reduction in the first cycle and Patients who are unable to receive 75% of the required doses of both agents secondary to toxicity. Number of Participants With such Dose Limiting Toxicities (DLT) at each level are reported here." (NCT01496118)
Timeframe: 28 days from the start of study treatment

InterventionParticipants (Count of Participants)
Dose Level 10
Dose Level 20
Dose Level 30
Dose Level 40
Dose Level 50
Dose Level 60

[back to top]

Time-to-progression (TTP)

Measured from date of first treatment until date of first documented progression as per International Myeloma Working Group Uniform Response Criteria. Progressive disease id defined as an increase of ≥ 25% from the nadir in at least one of the following criteria: serum M-protein (absolute increase must be ≥0.5 g/dL); urine M-protein (absolute increase must be ≥200 mg/24h) ; only in patients with non-measurable serum and urine M-protein levels: difference in involved and uninvolved FLC levels (absolute increase must be >10 mg/dL); only in patients with non-measurable serum and urine M-protein levels and non-measurable disease by FLC levels, bone marrow plasma cell percentage (absolute % must be ≥10%) OR Definite development of new bone lesions or soft tissue plasmacytomas OR definite increase in the size of existing bone lesions or soft tissue plasmacytomas OR Development of hypercalcemia (corrected serum Ca >11.5mg/dL) for patients without hypercalcemia at baseline (NCT01496118)
Timeframe: up to 6 years

Interventionmonths (Median)
Dose Level 4 - Escalation and Expansion11.6
Dose Level 6 - Escalation and Expansion11.7

[back to top]

Progression-free-survival (PFS)

Measured from date of first protocol treatment until date of tumor progression or death as per International Myeloma Working Group Uniform Response Criteria. Progressive disease id defined as an increase of ≥ 25% from the nadir in at least one of the following criteria: serum M-protein (absolute increase must be ≥0.5 g/dL); urine M-protein (absolute increase must be ≥200 mg/24h) ; only in patients with non-measurable serum and urine M-protein levels: difference in involved and uninvolved FLC levels (absolute increase must be >10 mg/dL); only in patients with non-measurable serum and urine M-protein levels and non-measurable disease by FLC levels, bone marrow plasma cell percentage (absolute % must be ≥10%) OR Definite development of new bone lesions or soft tissue plasmacytomas OR definite increase in the size of existing bone lesions or soft tissue plasmacytomas OR Development of hypercalcemia (corrected serum Ca >11.5mg/dL) for patients without hypercalcemia at baseline (NCT01496118)
Timeframe: up to 6 years

Interventionmonths (Median)
Dose Level 4 - Escalation and Expansion12.1
Dose Level 6 - Escalation and Expansion10.3

[back to top]

Phase II: Overall Response Rate

Defined as the percentage of patients with confirmed complete response, very good partial response, or partial response (CR, VGPR, or PR) according to International Myeloma Working Group Uniform Response Criteria. CR=bone marrow contains ≤5% plasma cells; negative fixation on serum and urine; disappearance of soft tissue plasmacytomas. VGPR=≥90% reduction from baseline serum; urine M-protein level 100mg for 24h. PR=≥50% reduction from baseline in serum M-protein; disappearance of any soft tissue plasmacytomas. (NCT01496118)
Timeframe: up to 6 years

Interventionpercentage of participants (Number)
Dose Level 4 - Escalation and Expansion74
Dose Level 6 - Escalation and Expansion84

[back to top]

Overall-survival (OS)

Measured from time of first study treatment until date of death or date last known alive. (NCT01496118)
Timeframe: up to 6 years

Interventionmonths (Median)
Dose Level 4 - Escalation and Expansion29.2
Dose Level 6 - Escalation and Expansion44.6

[back to top]

Time to Response (TTR)

TTR is defined as the time from enrolment to Overall Response (NCT01523834)
Timeframe: 36 months

Interventionmonths (Median)
Panobinosat2.6

[back to top]

Overall Survival (OS)

OS is defined as the time from enrolment to death from any case (NCT01523834)
Timeframe: 36 months

Interventionmonths (Median)
Panobinosat7.6

[back to top]

Progression Free Survival (PFS)

PFS is defined as the time from enrolment to disease progression or relapse or death from any cause (NCT01523834)
Timeframe: 36 months

Interventionmonths (Median)
Panobinosat2.4

[back to top]

Complete Response (CR) Rate

Proportion of CR at the end of the induction phase according to the Cheson 1999 response criteria (NCT01523834)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Panobinosat11.4

[back to top]

Overall Response Rate (ORR) at the End of the Induction Phase

ORR is defined as the proportion of patients achieving a Complete Response (CR) or Partial Response (PR) according to the Cheson 1999 response criteria (NCT01523834)
Timeframe: 6 months

Interventionpercentage of participants (Number)
Panobinosat17.1

[back to top]

6-month Overall Survival Rate

6-month overall survival rate (NCT01582009)
Timeframe: The time from registration up to 3 years

Interventionpercentage of participants (Number)
Arm I: Oral Panobinostat and Oral Everolimus96

[back to top]

Median Progression Free Survival

Median progression free survival. Assessed using Kaplan Meier and Proportional Hazards. (NCT01582009)
Timeframe: The time from registration up to 3 years

Interventionmonths (Median)
Arm I: Oral Panobinostat and Oral Everolimus5.3

[back to top]

Number of Participants With an Adverse Event.

Number of participants with an adverse event. Please refer to the adverse event reporting for more detail. (NCT01582009)
Timeframe: The time from registration up to 3 years

InterventionParticipants (Count of Participants)
Arm I: Oral Panobinostat and Oral Everolimus26

[back to top]

Number of Participants With Clinical Response

Number of participants with clinical response. Response will be evaluated in this study using the new international criteria proposed by the Response Evaluation Criteria in Solid Tumors ver 1.0 Committee [JNCI 92(3):205-216, 2000]. Changes in only the largest diameter (unidimensional measurement) of the tumor lesions are used in the RECIST ver. 1.0 criteria. (NCT01582009)
Timeframe: The time from registration up to 3 years

InterventionParticipants (Count of Participants)
Arm I: Oral Panobinostat and Oral Everolimus0

[back to top]

Progression-free Survival (PFS)

6 month PFS survival rate. Calculated as the total number of failures (deaths or progression) divided by the total follow-up or exposure time of patients on study. Assessed using Kaplan Meier and Proportional Hazards. (NCT01582009)
Timeframe: The time from registration to documentation of disease progression up to 3 years

Interventionpercentage of participants (Number)
Arm I: Oral Panobinostat and Oral Everolimus28

[back to top]

Disease Control Rate for Lens Refractory Rate

The number of response rates in Len refractory participants with SD, MR, PR, VGPR, or CR (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone21

[back to top]

Overall Response Rate for Len Refractory Patients

The primary endpoint will be the best overall response rate (ORR) (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone8

[back to top]

Response Rates

"Response Rates evaluated using the International Uniform Response Criteria the International Myeloma Working Group (2003).~CR-Negative immunofixation on the serum and urine and Disappearance of any soft tissue plasmacytomas and 5% plasma cells in bone marrow VGPR-Serum and urine M-component detectable by immunofixation but not on electrophoresis or 90 or greater reduction in serum M-component plus urine M-component <100 mg per 24 h PR-50% reduction of serum M-protein and reduction in 24-h urinary M-protein by 90% or to <200 mg per 24 h MR-≥ 25% but < 49% reduction of serum M protein and reduction in 24 hour urine M protein by 50 - 89%, which still exceeds 200 mg/24hrs. In addition; if present at baseline, 25-49% reduction in the size of soft tissue plasmacytomas also required No increase in size or number of lytic bone lesions.~SD-Not meeting criteria for CR, VGPR, PR or progressive disease PD-Laboratory or Biochemical Relapse increase of 25% from baseline" (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Complete Response (CR)Very Good Partial Response (VGPR)Partial Response (PR)Minimal Response (MR)Stable Disease (SD)Progressive Disease (PD)
Panobinostat, Lenalidomide and Dexamethasone245961

[back to top]

Response Rates for Len Refractory Patients

"Response Rates evaluated using the International Uniform Response Criteria the International Myeloma Working Group (2003).~CR-Negative immunofixation on the serum and urine and Disappearance of any soft tissue plasmacytomas and 5% plasma cells in bone marrow VGPR-Serum and urine M-component detectable by immunofixation but not on electrophoresis or 90 or greater reduction in serum M-component plus urine M-component <100 mg per 24 h PR-50% reduction of serum M-protein and reduction in 24-h urinary M-protein by 90% or to <200 mg per 24 h MR-≥ 25% but < 49% reduction of serum M protein and reduction in 24 hour urine M protein by 50 - 89%, which still exceeds 200 mg/24hrs. In addition; if present at baseline, 25-49% reduction in the size of soft tissue plasmacytomas also required No increase in size or number of lytic bone lesions.~SD-Not meeting criteria for CR, VGPR, PR or progressive disease PD-Laboratory or Biochemical Relapse increase of 25% from baseline" (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Complete ResponseVery Good Partial ResponsePartial ResponseMinimal ResponseStable DiseaseProgressive Disease
Panobinostat, Lenalidomide and Dexamethasone143761

[back to top]

Clinical Benefit Rate

The number of response rates in participants that have achieved MR, PR, VGPR, CR (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone20

[back to top]

Clinical Benefit Rate for Len Refractory Patients

The number of response rates in Lens Refractory participants that have achieved MR, PR, VGPR, CR (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone15

[back to top]

Disease Control Rate

The number of response rates participants with SD, MR, PR, VGPR, or CR (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone26

[back to top]

The Best Overall Response Rate (ORR)

The primary endpoint will be the best overall response rate (ORR). Per Response Evaluation Criteria In Solid Tumors Criteria (RECIST v1.0) for target lesions and assessed by MRI: Complete Response (CR), Disappearance of all target lesions; Partial Response (PR), >=30% decrease in the sum of the longest diameter of target lesions; Overall Response (OR) = CR + PR. (NCT01651039)
Timeframe: up to 4 years

InterventionParticipants (Count of Participants)
Panobinostat, Lenalidomide and Dexamethasone11

[back to top]

Number of Patients That Achieve Stable Disease or Clinical Improvement

"Number of patients that have either stable disease or clinical improvement treatment response as defined by the International Working Group for Myelofibrosis Research and Treatment (IWG-MRT).~Stable disease (SD) - response that is not complete remission, partial remission, clinical improvement, anemia response, spleen response, symptoms response, or progressive disease.~Clinical improvement (CI) - a response in anemia, splenomegaly, or MF-SB that is not associated with progressive splenomegaly or increase in severity of anemia, thrombocytopenia, or neutropenia." (NCT01693601)
Timeframe: at least 6 months

,,,
InterventionParticipants (Count of Participants)
Stable diseaseClinical ImprovementNo response evaluated
Panobinostat and Ruxolitinib Cohort 1300
Panobinostat and Ruxolitinib Cohort 2411
Panobinostat and Ruxolitinib Cohort 3120
Panobinostat and Ruxolitinib Cohort 4300

[back to top]

Number of Participants Who Experienced Dose-Limiting (DLTs)

Panobinostat related adverse events requiring dose reduction or discontinuing prior to Cycle 6, Day 29 (C6D29) (NCT01693601)
Timeframe: up to cycle 6, day 29

,,,
InterventionParticipants (Count of Participants)
requiring Panaobinostat dose reductionDiscontinuing Panobinostat
Panobinostat and Ruxolitinib Cohort 110
Panobinostat and Ruxolitinib Cohort 202
Panobinostat and Ruxolitinib Cohort 330
Panobinostat and Ruxolitinib Cohort 410

[back to top]

Percent Change in Spleen Volume

Percent change in spleen volume at C6D29 as compared to baseline (NCT01693601)
Timeframe: Baseline and Cycle 6, Day 29

Interventionpercent change (Median)
Panobinostat and Ruxolitinib Cohort 1-41
Panobinostat and Ruxolitinib Cohort 2-8
Panobinostat and Ruxolitinib Cohort 3-40
Panobinostat and Ruxolitinib Cohort 4-17

[back to top]

Percent Change in Spleen Size for Responders and Non-responders

Percent change in spleen length size by palpation at cycle 6, day 29 (C6D29) from baseline (NCT01693601)
Timeframe: Cycle 6, Day 29

Interventionpercent change (Median)
Responders-100
Non-Responders-29

[back to top]

Percent Change in Spleen Length

Percent change in spleen length at C6D29 (NCT01693601)
Timeframe: Cycle 6, Day 29

Interventionpercent change (Median)
Panobinostat and Ruxolitinib Cohort 1-83
Panobinostat and Ruxolitinib Cohort 2-17
Panobinostat and Ruxolitinib Cohort 3-14
Panobinostat and Ruxolitinib Cohort 4-31

[back to top]

Number of Participants With Percent Change on Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF)

Symptom responders defined as having a percent change in MPN-SAF score from Screening/C1D1 to C6D29 of more than 50%.MPN-SAF is an 18-item instrument. Each item score 0-10 averaged, total scale from0-10, with higher score indicating more symptoms. (NCT01693601)
Timeframe: baseline, C1D1 and Cycle 6 Day 29

InterventionParticipants (Count of Participants)
Panobinostat and Ruxolitinib Cohort 11
Panobinostat and Ruxolitinib Cohort 22
Panobinostat and Ruxolitinib Cohort 30
Panobinostat and Ruxolitinib Cohort 40

[back to top]

Overview of Adverse Events (Safety Set)

Adverse events were collected from baseline up to 30 days post treatment at scheduled visits. Severity of adverse events was assessed according to the current version of Common Terminology Criteria for Adverse Events (CTCAE). If CTCAE grading did not exist for an adverse event, the severity of mild, moderate, severe, and life-threatening, corresponding to Grades 1 - 4, was used (NCT01802879)
Timeframe: Baseline up to approximately 60 months

Interventionparticipants (Number)
Any adverse event (AE)Any treatment related AEAny serious adverse event (SAE)Grade 3 or 4 AEGrade 3 or 4 AE - suspected to be relatedAEs leading discontinuationAEs leading to dose adjust/ temp dose interruptionOn-treatment death
Panobinostat - 10 to 40 mg/Day TIW QoW62231020

[back to top]

Progression Free Survival (PFS)

Progression Free Survival (PFS): is defined for each participant as the time from first dosing to the first observation of disease progression or death due to any cause. If a subject has not progressed or died at the time of analysis, PFS will be censored on the date of the last disease assessment. PFS will be calculated by Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. Progressive Disease (PD) = 20% increase in the sum of the longest diameter of target lesions. (NCT02032810)
Timeframe: Up to 6 months

InterventionMonths (Median)
Panobinostat 5 mg4.19
Panobinostat 10 mg1.81

[back to top] [back to top]

Overall Survival (OS)

Overall survival: The time from randomization until death from any cause. (NCT02032810)
Timeframe: 36 months

InterventionMonths (Median)
Panobinostat 5 mg25.91
Panobinostat 10 mg12

[back to top]

Maximum Tolerated Dose (MTD)

"MTD and recommended Phase 2 dose (RP2D) of panobinostat (PAN), administered in combination with ipilimumab (IPI) in participants with unresectable Stage III or Stage IV melanoma.~The goal is to enroll up to 36 patients for determination of the MTD, but will be successfully concluded earlier if 12 patients are accrued at the dose determined to be the MTD, with 3 dose limiting toxicities (DLTs).~All participants who receive study any drug therapy will be evaluated for safety. Safety assessments will be based on medical review of adverse event reports and the results of vital sign measurements, physical examinations, and clinical laboratory tests. Triplicate 12-lead electrocardiograms (ECGs) will be collected prior to dosing on Day 1 of induction cycle 1. MTD of PAN in milligrams (mg), with IPI at 3 mg/kg." (NCT02032810)
Timeframe: Up to 36 months

Interventionmg (Number)
Dose Escalation of Panobinostat, Plus Ipilimumab5

[back to top]

Number of Participants With Dose Limiting Toxicity According to CTCAE Version 4.03

Number of Participants with Dose Limiting Toxicity of MLN9708 (lxazomib) according to CTCAE version 4.03 (NCT02057640)
Timeframe: at 28 days from start of treatment

InterventionParticipants (Count of Participants)
Dose Levels 14

[back to top]

Response to Combination Therapy (Panobinostat, Dexamethasone, MLN9708)

Response to intervention as measured by international uniform response criteria and clinical benefit response according to modified EBMT response criteria, comparing myeloma panels obtained at the beginning of each cycle that include SPEP, 24 h UPEP, serum and urine IFEs, and serum free light chains to results at screening. In addition a baseline bone marrow exam and skeletal survey will be obtained and repeated as clinically indicated and for assessment of complete remission (bone marrow) (NCT02057640)
Timeframe: 4 months (102 days)

Interventionparticipants (Number)
DL1 - 3mg Lxazomib0
DL2 - 4mg Lxazomib0

[back to top]

Progression-free Survival

"Progression-free survival will be the number of days from study entry to progression or death of any cause, whichever comes first.~Progression-free survival is survival with absence of progressive disease, defined by~An increase of 25% from lowest response value in any one or more of the following:~Serum M-component (absolute increase must be >0.5 g/100 ml) *~Urine M-component (absolute increase must be >200mg per 24 h)~Only in patients without measurable serum and urine M-protein levels: the difference between involved and uninvolved FLC levels (absolute increase must be >100 mg/l)~Bone marrow plasma cell percentage (absolute % must be >10%)~And / or:~Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas~Development of hypercalcemia (corrected serum calcium >11.5 mg/100 ml) that can be attributed solely to the plasma cell proliferative disorder" (NCT02057640)
Timeframe: 1 year from start of treatment

Interventionmonths (Median)
DL1 - 3mg Lxazomib1.2
DL2 - 4mg Lxazomib3.5

[back to top]

Overall Survival

Overall survival for all will be the number of months from study entry to death from any cause. (NCT02057640)
Timeframe: up to 3 years from start of treatment

Interventionmonths (Median)
DL1 - 3mg Lxazomib12.8
DL2 - 4mg Lxazomib17.6

[back to top]

Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: CL/F

CL/F: The apparent total body clearance of drug from the plasma (NCT02290431)
Timeframe: Predose, 0.5h, 1h, 2h, 3h, 4h 8h, 24h 48h post dose

InterventionLitre/hour (L/h) (Geometric Mean)
PAN: C1D1PAN: C1D8
LBH589 + Bortezomib + Dexamethasone172114

[back to top]

Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: AUClast, AUC0-24h, AUC0-48h, AUCinf

PK sample collection was performed in subjects who agreed to blood samplings for the PK assessments of PAN and BTZ. The order of administration of the 3 study treatment components was 1) PAN, 2) Dex, and 3) BTZ. (NCT02290431)
Timeframe: Predose, 0.5h, 1h, 2h, 3h, 4h 8h, 24h, 48h post dose

Interventionh.ng/mL (Geometric Mean)
PAN: AUClast C1D1PAN: AUClast C1D8BJB432: AUClast C1D1BJB432: AUClast C1D8PAN: AUC0-24h C1D1PAN: AUC0-24h C1D8BJB432: AUC0-24h C1D1BJB432: AUC0-24h C1D8PAN: AUC0-48h C1D1PAN: AUC0-48h C1D8BJB432: AUC0-48h C1D1BJB432: AUC0-48h C1D8PAN: AUCinf C1D1PAN: AUCinf C1D8
LBH589 + Bortezomib + Dexamethasone10615637.816687.112518.583.810615636.8169116175

[back to top]

Time to Progression/Relapse (TTP) Per Investigator

TTP is defined as the time from the date of the first dose of study treatment to the date of the first documented disease progression or relapse (NCT02290431)
Timeframe: duration of study up to approx. 4 years

Interventionmonths (Median)
LBH589 + Bortezomib + Dexamethasone15.3

[back to top]

Progression Free Survival (PFS)

PFS is defined as time from first dose of study treatment to progression or death due to any cause, based on modified European Society for Bone and Marrow Transplantation (EBMT) criteria per Investigator's assessment (NCT02290431)
Timeframe: duration of study up to approx. 4 years

Interventionmonths (Median)
LBH589 + Bortezomib + Dexamethasone15.3

[back to top]

Percentage of Participants With Near Complete Response (nCR)/ Complete Response (CR) Rate

nCR plus CR rate after 8 cycles of therapy as defined by the modified European Society for Bone and Marrow Transplantation (EBMT) criteria per investigator assessment as the proportion of participants with nCR or CR as their best overall response. (NCT02290431)
Timeframe: after 24 weeks (8 cycles; cycle = 21 days)

InterventionPercentage of participants (Number)
LBH589 + Bortezomib + Dexamethasone48.4

[back to top]

Overall Survival (OS)

OS is defined as time from first dose of study treatment to death (NCT02290431)
Timeframe: up to 30 days after end of study, approx. 4 years

Interventionmonths (Median)
LBH589 + Bortezomib + DexamethasoneNA

[back to top]

Overall Response Rate (ORR)

ORR is defined as the proportion of participants with CR, nCR or partial response (PR) based on modified EBMT criteria per investigator assessment (NCT02290431)
Timeframe: 24 weeks (8 cycles; cycle = 21 days)

InterventionPercentage of participants (Number)
LBH589 + Bortezomib + Dexamethasone80.6

[back to top]

Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: Tmax

Tmax: The time to reach maximum (peak) plasma concentration. PK sample collection was performed in subjects who agreed to blood samplings for the PK assessments of PAN and BTZ. The order of administration of the 3 study treatment components was 1) PAN, 2) Dex, and 3) BTZ. (NCT02290431)
Timeframe: Predose, 0.5h, 1h, 2h, 3h, 4h 8h, 24h, 48h post dose

Interventionhour (h) (Median)
PAN: C1D1PAN: C1D8BJB432: C1D1BJB432: C1D8
LBH589 + Bortezomib + Dexamethasone2.002.0024.024.0

[back to top]

Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: T1/2

T1/2: The elimination half-life associated with the terminal slope (Lambda_z) of a semi logarithmic concentration-time curve (NCT02290431)
Timeframe: Predose, 0.5h, 1h, 2h, 3h, 4h 8h, 24h 48h post dose

Interventionhour (h) (Geometric Mean)
PAN: C1D1PAN: C1D8
LBH589 + Bortezomib + Dexamethasone13.716.5

[back to top]

Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: Lambda_z

Lambda_z: The terminal elimination rate constant (h-1) (NCT02290431)
Timeframe: Predose, 0.5h, 1h, 2h, 3h, 4h 8h, 24h 48h post dose

Intervention1/hour (1/h) (Geometric Mean)
PAN: C1D1PAN: C1D8
LBH589 + Bortezomib + Dexamethasone0.05060.0421

[back to top]

Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: Cmax

Cmax: The maximum (peak) observed plasma concentration. PK sample collection was performed in subjects who agreed to blood samplings for the PK assessments of PAN and BTZ. The order of administration of the 3 study treatment components was 1) PAN, 2) Dex, and 3) BTZ. (NCT02290431)
Timeframe: Predose, 0.5h, 1h, 2h, 3h, 4h 8h, 24h, 48h post dose

Interventionng/mL (Geometric Mean)
PAN: Cycle 1 Day 1 (C1D1)PAN: Cycle 1 Day 8 (C1D8)BJB432: C1D1BJB432: C1D8
LBH589 + Bortezomib + Dexamethasone11.518.21.064.38

[back to top]

Minimal Response Rate (MRR) Per Investigator

MRR is based on modified EBMT criteria per investigator assessment (NCT02290431)
Timeframe: after 24 weeks (8 cycles; cycle = 21 days)

InterventionPercentage of participants (Number)
LBH589 + Bortezomib + Dexamethasone9.7

[back to top]

Duration of Response (DOR) Per Investigator

DOR is defined as the time from date of the first documented CR/nCR or PR to the date of the first documented progression or relapse or death due to MM (NCT02290431)
Timeframe: duration of study up to approx. 4 years

Interventionmonths (Median)
LBH589 + Bortezomib + Dexamethasone22.7

[back to top]

Composite PharmacoKinetics (PK) of Panobinostat and Bortezomib: Vz/F

Vz/F: The apparent volume of distribution during terminal phase (associated with Lambda_z) (NCT02290431)
Timeframe: Predose, 0.5h, 1h, 2h, 3h, 4h 8h, 24h 48h post dose

InterventionLitre (L) (Geometric Mean)
PAN: C1D1PAN: C1D8
LBH589 + Bortezomib + Dexamethasone33902720

[back to top]

Time to Response (TTR) Per Investigator

TTR is defined as the time from the date of first dose of study treatment to first documented response (PR or nCR or CR) per modified EBMT criteria as assessed by investigator (NCT02290431)
Timeframe: duration of study up to approx. 4 years

Interventionmonths (Median)
LBH589 + Bortezomib + Dexamethasone1.4

[back to top]

Quality of Life (QoL) as Measured by FACT/GOG-Ntx Total Score

QoL as measured by Functional Assessment of Cancer Therapy/ Gynecology Oncology Group Neurotoxicity (FACT/GOG-NTX) scale calculated scores and changes from baseline were summarized by visit. The FACT/GOG-Ntx is a measure to assess neurotoxicity from systemic chemotherapy. The recall period for this measure is the past 7 days. FACT/GOG-Ntx Total Score: 0 - 152 (28 + 28 + 24 + 28 + 44 = 152). (FACT-G Physical Well-Being Score: 0 - 28, FACT-G Social/Family Well-Being Score: 0 - 28, FACT-G Emotional Well-Being Score: 0 - 24, FACT-G Functional Well-Being Score: 0 - 28, FACT/GOG-Ntx Neurotoxicity Subscale Score: 0 - 44). 4. The scales are combined. The higher the score, the better the QOL. (NCT02290431)
Timeframe: Baseline, Weeks 12, 24, 36, 48, 60, 72, 84, 96, 108, 120, 132, and 156

Interventionscores on a scale (Median)
BaselineWeek 12Week 24Week 36Week 48Week 60Week 72Week 84Week 96Week 108Week 120Week 132Week 156
LBH589 + Bortezomib + Dexamethasone112.3391.75100.33125.83114.25109.92108.00101.00121.67120.17125.25114.00119.8

[back to top]

Number of Participants With Non-relapse Mortality

Incidence of primary disease relapse and non-relapse related death will be reported per standard definitions. These will be treated as competing risk events. Non-relapse death is defined as death in continuous remission from primary disease requiring transplantation. (NCT02588339)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Panobinostat (PANO) Therapy1

[back to top]

Number of Participants With Primary Disease Relapse

Incidence of primary disease relapse and non-relapse related death will be reported per standard definitions. These will be treated as competing risk events. (NCT02588339)
Timeframe: 1 year

InterventionParticipants (Count of Participants)
Panobinostat (PANO) Therapy7

[back to top]

Percentage of Participants With Overall Survival (OS)

Overall survival: Time from transplant date to death from any cause. Time-to-event data such as overall survival is measured from the date of transplantation. OS will be analyzed using the Kaplan-Meier method. (NCT02588339)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Panobinostat (PANO) Therapy87

[back to top]

Time to Stable Engraftment

Stable engraftment for white blood count (WBC) is defined as a sustained absolute neutrophil count > 500 over 3 days without cytokine support. Stable platelet engraftments is defined as count of > 20,000 over 7 days without transfusion support. Time to engraftment is defined as time from day 0 to day of sustained engraftment per above criteria for both platelets and WBC. (NCT02588339)
Timeframe: 100 days post transplant

Interventiondays (Median)
ANC engraftmentPlatelet engraftment
Panobinostat (PANO) Therapy1516

[back to top]

Number of Participants Stratified by Chronic Graft Versus Host Disease (GVHD) Stage

GVHD with onset after 100 days post-HCT with presence of at least one diagnostic manifestation of chronic c-GVHD or distinct manifestation confirmed by biopsy or other relevant tests (e.g., PFT). Classified as: 1- Classic chronic GVHD - meets criteria for chronic GVHD and has no features consistent with aGVHD or 2-Overlap syndrome - features of acute and chronic GVHD exist together. C-GVHD will be measured prospectively in all participants on days 90+/-14 , 120 +/- 14, 150 +/- 14, 180+/- 14, 270+/- 30, and 365 +/- 30 as per standardized scoring system. (NCT02588339)
Timeframe: 100 days post transplant

InterventionParticipants (Count of Participants)
Mild GVHDModerate GVHD
Panobinostat (PANO) Therapy102

[back to top]

Number of Participants Stratified by Acute Graft Versus Host Disease GVHD Stage

Cumulative incidence of acute GVHD grades II-IV by day 100. Investigators will consider ≥43% incidence of grade II-IV aGVHD not acceptable. Investigators will use 23% incidence rate of GVHD as target. GVHD severity stage and grading and distribution will be measured weekly from day of transplant to day 90 +/- 14 using standard scoring system. Stage of GVHD will be given for each site of involvement (e.g. skin, liver, and gut), as well as a composite score for overall acute GVHD grade. Pathologic confirmation of aGVHD will be dictated by usual clinical practice, and not mandated by this protocol. (NCT02588339)
Timeframe: 100 days post transplant

InterventionParticipants (Count of Participants)
Grade II GVHDGrade III GVHD
Panobinostat (PANO) Therapy61

[back to top]

Percentage of Participants With Relapse-free Survival (RFS)

Relapse-free survival: Time from transplant date to death or primary disease relapse. Time-to-event data such as relapse-free survival is measured from the date of transplantation. RFS will be analyzed using the Kaplan-Meier method. (NCT02588339)
Timeframe: 1 year

Interventionpercentage of participants (Number)
Panobinostat (PANO) Therapy77

[back to top]

Overall Survival (OS)

OS: The length of time from either the date of diagnosis or the start of treatment for a disease, such as cancer, that patients diagnosed with the disease are still alive. (NCT02722941)
Timeframe: at 2 years

Interventionpercentage of participants (Median)
Cohort A: Maintenance Therapy100
Cohort B: Maintenance Therapy100

[back to top]

Relative Dose Intensity (RDI) Per Cohort

Investigators will calculate RDI for each cohort. Relative dose intensity (RDI) represents the ratio of the amount of a drug actually delivered [actual dose intensity (DI)] to the amount planned (planned DI). The purpose of calculating RDI is to evaluate whether the planned DI of a chemotherapy treatment was actually achieved which may suggest the feasibility of planned treatment regimen. There are multitude of reports demonstrating a correlation between RDI and survival in cancer treatment. RDI = (total dose received by the patient = mg)/(planned full dose of drug = mg). (NCT02722941)
Timeframe: Up to 2 years

Interventionpercentage of dose (Median)
Cohort A: Maintenance Therapy97.9
Cohort B: Maintenance Therapy89.6

[back to top]

Progression Free Survival (PFS)

"Progressive Disease (PD) according to Uniform Response Reporting Criteria for Multiple Myeloma by the International Myeloma Working Group (IMWG). Increase of 25% from lowest response value in any of the following:~Serum M- component (absolute increase must be ≥ 0.5 g/dL)~Urine M-component (absolute increase must be ≥ 200 mg/24 h)~Only in patients without measurable serum and urine M protein levels and without measurable disease by free light chain (FLC) levels, bone marrow plasma cell percentage (absolute percentage must be ≥ 10% )~Definite development of new bone lesions or soft tissue plasmacytomas or definite increase in the size of existing bone lesions or soft tissue plasmacytomas~Development of hypercalcemia (corrected serum calcium > 11.5 mg/dL) that can be attributed solely to the plasma cell proliferative disorder" (NCT02722941)
Timeframe: at 2 years

Interventionpercentage of participants (Median)
Cohort A: Maintenance Therapy71.8
Cohort B: Maintenance Therapy53.3

[back to top]

Complete Response Rate

Complete Response (CR) rate to panobinostat maintenance therapy after autologous HCT. CR: Negative immunofixation of serum and urine, disappearance of any soft tissue plasmacytomas, and < 5% plasma cells in bone marrow. (NCT02722941)
Timeframe: Up to 5 years

Interventionpercentage (Number)
Cohort A: Maintenance Therapy73.33
Cohort B: Maintenance Therapy66.6

[back to top]

Multiple Myeloma Cells In-vitro Drug Sensitivity to Panobinostat, by Subject

Multiple myeloma cells are added to assay with panobinostat. Cell viability and IC50 is determined. IC50 is the inhibitory concentration that kills 50% of cells. Results are reported in concentrations E-9. (NCT03256045)
Timeframe: At baseline

InterventionnM (Number)
Subject MM-82Subject MM-91Subject MM-92Subject MM-95Subject MM-96Subject MM-98Subject MM-99Subject MM-109Subject MM-111
Chemo Assay and Treatment With Panobinostat, Carfilzomib, and Dexamethasone3.85.92.619.81.34.48.513.62.0

[back to top]

Synergy of Panobinostat and Carfilzomib in Combination, Per Subject.

Concentration of carfilzomib and panobinostat in combination leading to 90% growth inhibition of multiple myeloma cells in-vitro. The combination index (CI) is calculated using formula: CI = ([D1] in the combination / [D1] alone) + ([D2] in the combination / [D2] alone). Key: CI = 1 no synergy; CI<1 synergy; CI>1 antagonism. (NCT03256045)
Timeframe: At baseline

InterventionCombination index (Number)
Subject MM-91Subject MM-92Subject MM-95Subject MM-96Subject MM-98Subject MM-99Subject MM-109Subject MM-111
Chemo Assay and Treatment With Panobinostat, Carfilzomib, and Dexamethasone2.52.40.32.22.61.81.762.2

[back to top]

Synergy of Panobinostat and Dexamethasone in Combination, Per Subject

Concentration of panobinostat and dexamethasone in combination leading to 90% growth inhibition of multiple myeloma cells in-vitro. The combination index (CI) is calculated using formula: CI = ([D1] in the combination / [D1] alone) + ([D2] in the combination / [D2] alone). Key: CI = 1 no synergy; CI<1 synergy; CI>1 antagonism. (NCT03256045)
Timeframe: At baseline

InterventionCI score (Number)
Subject MM-91Subject MM-92Subject MM-95Subject MM-96Subject MM-98Subject MM-99Subject MM-109Subject MM-111
Chemo Assay and Treatment With Panobinostat, Carfilzomib, and Dexamethasone1.32.61.40.61.20.94.61.2

[back to top]

Multiple Myeloma Cells In-vitro Drug Sensitivity to Carfilzomib, by Subject

Multiple myeloma cells are added to assay with carfilzomib. Cell viability and IC50 is determined. IC50 is the inhibitory concentration that kills 50% of cells. (NCT03256045)
Timeframe: At baseline

InterventionnM (Number)
Subject MM-82Subject MM-91Subject MM-92Subject MM-95Subject MM-96Subject MM-98Subject MM-99Subject MM-109Subject MM-111
Chemo Assay and Treatment With Panobinostat, Carfilzomib, and Dexamethasone17.50.4790.030.2690.00480.1590.1590.1080.0152

[back to top]

Best Overall Response, by Subject

Assessed using the International Myeloma Working Group Uniform Response Criteria for Multiple Myeloma. Key: 1 = very good partial response (VGPR); 2 = partial response (PR); 3 = minimal response (MR); 4 = stable disease (SD); 5 = progressive disease (PD). Please see IC50 and CI results in the other primary outcome measures. (NCT03256045)
Timeframe: 14 months

InterventionIMWG Uniform Response Grade (Number)
Subject MM-82Subject MM-91Subject MM-92Subject MM-95Subject MM-96Subject MM-98Subject MM-99Subject MM-109Subject M-111
Chemo Assay and Treatment With Panobinostat, Carfilzomib, and Dexamethasone422111341

[back to top]

Multiple Myeloma Cells In-vitro Drug Sensitivity to Dexamethasone, by Subject

Multiple myeloma cells are added to assay with dexamethasone. Cell viability and IC50 is determined. IC50 is the inhibitory concentration that kills 50% of cells. Results are reported in concentrations E-6. (NCT03256045)
Timeframe: At baseline

InterventionuM (Number)
Subject MM-82Subject MM-91Subject MM-95Subject MM-96Subject MM-98Subject MM-99Subject MM-109Subject MM-111
Chemo Assay and Treatment With Panobinostat, Carfilzomib, and Dexamethasone3.710.971.114.88.6801.1

[back to top]

Proportion of Participants With Grade 3 or Higher, Treatment-related, Adverse Events

Adverse events and clinically significant laboratory abnormalities which meet Grade 3, 4, or 5 criteria according to Common Terminology Criteria for Adverse Events (CTCAE) classified by investigators and treating physicians as related to study treatment (probable, possible, and definite) will be summarized by maximum intensity/grade. Adverse events will be graded according to CTCAE version 4.0. (NCT03566199)
Timeframe: Up to 12 Months

Interventionproportion of participants (Number)
Muscle weakness right-sided (Grade 3)Stridor (Grade 3)Vagus nerve disorder (Grade 3)Gait disturbance (Grade 3)Neutrophil count decreased (Grade 3)
Phase 1: Treatment (MTX110)0.1420.1420.1420.1420.142

[back to top]

Overall Survival Rate (OS) at 12 Months

Overall survival is defined as the percentage of participants alive from time of diagnosis up to 12 months. (NCT03566199)
Timeframe: Up to 12 Months

Interventionpercentage of participants (Number)
Phase 1: Treatment (MTX110)85.7

[back to top]