113
Views
5
CrossRef citations to date
0
Altmetric
Review

Interpreting clinical assays for histone deacetylase inhibitors

&
Pages 117-141 | Published online: 30 Sep 2022

Abstract

As opposed to genetics, dealing with gene expressions by direct DNA sequence modifications, the term epigenetics applies to all the external influences that target the chromatin structure of cells with impact on gene expression unrelated to the sequence coding of DNA itself. In normal cells, epigenetics modulates gene expression through all development steps. When “imprinted” early by the environment, epigenetic changes influence the organism at an early stage and can be transmitted to the progeny. Together with DNA sequence alterations, DNA aberrant cytosine methylation and microRNA deregulation, epigenetic modifications participate in the malignant transformation of cells. Their reversible nature has led to the emergence of the promising field of epigenetic therapy. The efforts made to inhibit in particular the epigenetic enzyme family called histone deacetylases (HDACs) are described. HDAC inhibitors (HDACi) have been proposed as a viable clinical therapeutic approach for the treatment of leukemia and solid tumors, but also to a lesser degree for noncancerous diseases. Three epigenetic drugs are already arriving at the patient’s bedside, and more than 100 clinical assays for HDACi are registered on the National Cancer Institute website. They explore the eventual additive benefits of combined therapies. In the context of the pleiotropic effects of HDAC isoforms, more specific HDACi and more informative screening tests are being developed for the benefit of the patients.

Introduction

The transcriptional state of a eukaryotic gene is determined by the surrounding chromatin architecture, the state of DNA cytosine methylation in the promoter/first exons and the associated regulating microRNAs (miRNAs).

In the cell nucleus, the genome is packaged into a superstructure, the chromatin, whose elementary dynamic units are the nucleosomes. Each is made up of four associated dimers of core histones (H2A, H2B, H3, and H4) around which 147 base pairs of DNA are wrapped, the nucleosomes being finally linked together via the linker histone H1 (). Chromatin is compacted around the DNA into a so-called “closed state” when cells are resting. It is opened into an “active state” to allow for gene transcription by adenosine triphosphate (ATP)-dependent protein complexes, which remodel the chromatin architecture. Theses complexes modify the accessibility of DNA regulatory sites through both repositioning (sliding) and ejecting nucleosomes. Modeling complexes include transcription co-activators, transcription factors, and epi-enzymes.Citation1 Histone acetyltransferases (HATs), for instance, acetylate specific lysine residues of histones and convert them into an amide form, loosening histone contacts with DNA, resulting in exposed binding sites for the transcription machinery. On the other hand, other complexes function as gene silencers and deny the same machinery access to DNA. Repressive complexes include histone deacetylases (HDACs), which deacetylate specific lysine residues of the histones tails to induce tighter interactions between the now positively charged lysine (Nε protonated form) and the negatively-charged DNA phosphate groups.Citation2 Beside acetylation, several other post-translational lysine modifications in histones have been described: methylation, phosphorylation, SUMOylation, and ATP-ribosylation.Citation3,Citation4

Figure 1 The nucleosome unit and the histone tail chemical modification.

Abbreviations: HAT, histone acetyltransferase; HDM, histone demethylases; HDAC, histone deacetylase; HMT, histone methyltransferase; UBP, ubiquitin-specific protease.

Figure 1 The nucleosome unit and the histone tail chemical modification.Abbreviations: HAT, histone acetyltransferase; HDM, histone demethylases; HDAC, histone deacetylase; HMT, histone methyltransferase; UBP, ubiquitin-specific protease.

All the modifications incurred by histones form the “histone code”. For example, K9 in H3 turns the chromatin into inactivity when methylated. Phosphorylation of serine 10 in the same H3 is required for methylation of K4 and acetylation of K9 and K14. Similar enzyme crosstalk has also been described, for ubiquitination of K120 in H2B prior to methylation of K79 in H3.Citation5 Several epi-enzyme families are involved in the histones modifications. HATs and HDACsCitation2 have balancing actions for histones acetylation. Methylation of histonesCitation6 is controlled by histones methyltransferases (HMTs) and histones demethylases (HDMs), while histones arginine methylation is catalyzed by the protein arginine N-methyltransferases family of enzymes.Citation7Citation9 One, two, or three methylations are possible, with impact on gene expression/repression.Citation10 The analysis of epigenetic marks at the genome-wide scale has shown that monomethylated H3K4 is associated with transcription factors binding to enhancers, trimethylated H3K4 with transcription start sites, and dimethylated H3K4 with both transcription start sites and enhancers.Citation11

DNA methyltransferases (DNMTs) repress gene expression via DNA cytosine methylation, unfavorable to transcription factor binding. DNMTs are recruited and stabilized, on DNA, by HMTs and HDMs. Both are also able to recruit HDACs, methyl-binding proteins like methyl CpG binding protein 2 (MECP2), and several co-factors to further tune gene expression.Citation12,Citation13 Since DNMT co-factors are lacking in normal tissues, gene re-expression induced by DNMT inhibitors could be limited to tumor tissues to reduce “off-target” effects. DNA methylation in the epigenomes of human embryonic stem cells is an important field of research. The roles of DNA methylation in cancer genesis have also been extensively studied.Citation12,Citation13

Catalyzed by the ten-eleven translocation 1 (TET) family of enzymes, DNA hydroxymethylcytosine has been recently described as a step towards cytosine demethylation.Citation14 Mutations and translocations of TET are present in myeloid malignancies.Citation15 The role of hydroxymethylcytosine, if any, is not yet understood, but its existence questions all the results obtained so far when determining the cytosine methylation status.

In a further step of complexity, specialized miRNAs read the epi-code and target effectors genes to modulate their expression. MiRNAs are small non-coding RNAs of 20–22 nucleotides that inhibit gene expression when they engage either in imperfect base-pairing with their target mRNA 3′-untranslated region or affect its stability. MiRNA 29-a, -b and -c target DNMT3a and b directly and cell-specifically. HDAC4 is targeted by both miR-1 and miR-140, while miR-449-a targets HDAC1 in prostate malignant cells.Citation16 Onco-proteins like promyelocyte leukemia retinoic acid receptor-α (PML-RARα) in promyelocytic leukemia and B-cell lymphoma 6 in non-Hodgkin’s lymphoma result from translocations. It is near the premiR-223 region that the t(8;21) translocation juxtaposes the Runt-related transcription factor 1 gene on chromosome 21 with the Cytochrome B Termination 1 gene on chromosome 8, generating the acute myeloid leukemia (AML)1-Eight Twenty One fusion gene.Citation17 The recruitment, on this chimerical site, of DNMT, MeCP2, and HDAC1 repressor complexes, promotes leukemogenesis. Epi-miRNAs write their own epi-code when their cytosines are methylated. Downregulation of miR-124a induces an up regulation of its target, cyclin-dependent kinase 6 (CDK6), as well as phosphorylation of retinoblastoma, and contributes to the abnormal proliferation of acute lymphoblastic leukemia (ALL) cells both in vitro and in vivo.

Most epigenetic changes translate into either up regulation or silencing of gene expression.Citation18 When inappropriate, they predispose the organism to more mutational events via increased genomic instability and aberrant cellular signaling. The field of epigenetic being extremely prolific, we have restricted our reference list to the essentials.

HDACs

HDACs remove the acetyl group from an N-ε-acetyl lysine located near the amino terminus of a core histone, cleaving an amide bond and increasing the positive charge of the histone. The removal of acetyl groups from the histones tails stabilizes nucleosomal DNA-histones interactions by its subsequent change in electrostatic charges. It is the basis for HDAC-mediated transcriptional repression via chromatin condensation.Citation19 HDACs have been categorized into four classes. Class I HDACs (HDAC1, 2, 3, and 8) are nuclear proteins with ubiquitous expression involved in regulating cell proliferation.Citation20 HDAC2 has been shown to suppress apoptosis in tumor cells not only via both the intrinsic/mitochondrial and the extrinsic/death-receptor pathways, but also via mitotic failure and autophagic cell death, while HDAC3 is involved in bone structure and S-phase check point.Citation21,Citation22 Class II HDACs have a tissue-specific expression and can shuttle between the nucleus and the cytoplasm. They are divided into two subclasses: IIa with HDAC4, 5, 7, and 9. HDAC4 represses chondrocyte hypertrophy. HDAC7 functions in the down regulation and apoptosis of T-cells.Citation20 HDAC9 is involved in cardiomyocyte differentiation.Citation23 Class IIb includes HDAC10 and HDAC6. The latter contains two tandem catalytic domains: one is for histones deacetylation and the other for deacetylation of α-tubulin. HDAC6 has also the capacity to bind directly to ubiquitinated proteins through an ubiquitin-binding domain, to target cargo proteins for subsequent processing. HDAC6-specific effects on cell motility and the proteasome are thought to be responsible for much of the toxicity of HDACs inhibitors (HDACi). HDAC 10 and 9 are required for chromosome homologous recombination.Citation24 Class III HDACs include 7 different members of the sirtuin (SIRT) family. They are dependent on nicotinamine adenine dinucleotide (NAD+) to remove the acetyl group from lysine residues in histones and nonhistone substrates. Resveratrol from grapes and red wine is a SIRT1 activator.Citation25 HDAC11 is the only member of Class IV.

Thus, it appears that HDAC activity depends on isoform types, sub cellular localization, association into multi-protein complexes and even post-translational modifications. HDACs are also able to deacetylate nonhistone proteins such as transcription factors, chaperone proteins and effectors of DNA repair, cell-signaling and metabolism. The ongoing concept is that deacetylation stabilizes these proteins. HDACs have different developmental functions, as shown by the different phenotypes obtained in knockout mice.Citation26 Disruption of HDAC1 causes early embryonic lethality. HDAC2 knockout mice are viable but present fatal multiple cardiac defects. Germline HDAC3-deficiency causes embryonic lethality. HDAC3 conditional knockout mice gave severe deficits in membranous and endochondral bone formation. Germline deletion of HDAC4 causes premature ossification of the developing bones. HDAC6-deficiency slightly enhances trabecular bone formation. HDAC7 knockout gave vascular defects. HDAC8 is essential for neural crest progenitor cell differentiation and skull bone formation. HDAC9 knockout mice are viable at birth but have a myocardial hypertrophy.

HDACs and their inhibitors

In tumor cells, deletion of a single HDAC is not sufficient to induce cell death but leads to nuclear bridging and fragmentation, and ‘in fine’ to cell mitotic catastrophe. This suggests that inhibition of HDAC may be sufficient for anticancer activity and provides a rational incentive for the development of HDACi.Citation27 In the 1970s, seminal experiments showed that treatment of cells with the short-chain fatty acid NaB (sodium butyrate) caused hyperacetylation of histone octamers and led to the discovery of HDACs.Citation28 The zinc-dependent HDACs of classes I, II, and IV are now known to have a common active site made of a tubular hydrophobic channel with a zinc atom (Zn2+) at its end, forming the enzyme catalytic pocket.Citation29 The acetyl part of the lysine substrate in histones/proteins bind to the zinc atom while the protein four carbon lysine chains fits into the catalytic pocket, and deacetylation then follows. An HDACi is designed to block the HDAC catalytic activity. Several possibilities exist: irreversible or reversible binding to the enzyme catalytic site, competition with the enzymatic substrate, and deformation of the enzyme. Accordingly and as shown in , the pharmacophore model for HDACi includes a zinc-binding group, competing somehow with the natural acetyl lysine substrate, a hydrophobic cap interacting with the external surface of the active site (generally aromatic) generating specificity, and a short linker connecting these two elements, which fits in the catalytic pocket. The zinc-binding groups can be a carboxylic acid in valproic acid (VPA), a hydroxamic acid in Vorinostat, benzamide in Entinostat, sulfhydryl in Romidepsin,Citation30 and a ketone in Trapoxin. The linkers can be simple carbon chains, like in Vorinostat, or aromatic groups, like in Entinostat. HDACi in clinical trials are reported in . Trichostatine (TSA) from Streptomyces hygroscopius was isolated as an antifungal antibiotic and was incidentally shown to have an anti-proliferative activity on murine leukemia cells. Further studies demonstrated that it was a pan-HDACi. The hydroxamate portion at the end of the molecule acts as a zinc-binding group. Because of toxic side effects, it is not used clinically but participates in the rational conception of HDACi via molecular modeling, as shown in .

Figure 2 Left panel: X-ray crystallographic data for SAHA bound to HDAC8. The zinc atom in the HDAC active site is shown in grey. The hydroxamic acid group in SAHA is bound to Zn2+, the phenylamide group is outside the enzyme active site, and these two elements are linked by a short carbon chain. Right panel: Modeled tubulin bound to HDAC6. Zn2+in the active site is shown in grey. The hydroxamic acid group in tubulin is bound to the Zn2+, the phenylamide group is outside the enzyme active site and these two elements are linked by a short carbon chain. A bulk chemical entity has been grafted onto the phenylamide part of SAHA to obtain selectivity towards HDAC6 due to specific S-pi interactions from sulfur atom (in yellow).

Abbreviations: HDAC, histone deacetylase; SAHA, suberoylanilide hydroxamicacid.

Figure 2 Left panel: X-ray crystallographic data for SAHA bound to HDAC8. The zinc atom in the HDAC active site is shown in grey. The hydroxamic acid group in SAHA is bound to Zn2+, the phenylamide group is outside the enzyme active site, and these two elements are linked by a short carbon chain. Right panel: Modeled tubulin bound to HDAC6. Zn2+in the active site is shown in grey. The hydroxamic acid group in tubulin is bound to the Zn2+, the phenylamide group is outside the enzyme active site and these two elements are linked by a short carbon chain. A bulk chemical entity has been grafted onto the phenylamide part of SAHA to obtain selectivity towards HDAC6 due to specific S-pi interactions from sulfur atom (in yellow).Abbreviations: HDAC, histone deacetylase; SAHA, suberoylanilide hydroxamicacid.

Figure 3 Histone deacetylase (HDAC) inhibitors used in clinical trials arranged by chemical classes.

Figure 3 Histone deacetylase (HDAC) inhibitors used in clinical trials arranged by chemical classes.

Besides its HDAC inhibition activity, Vorinostat (, left), deriving from TSA, has a complex and not yet fully characterized activity leading to the accumulation of acetylated histones and non histone proteins. First generation HDACi are not selective except for a partial selectivity achievedCitation31,Citation32 in rare cases using bulk chemical groups to generate specific interactions with the external surface of the active site of the enzyme, like in tubacin.Citation33 Sulfur-based zinc-binding groups also showed some selectivity in compounds like Largazole, a potent and selective HDACi for HDAC1 and 2. It is a densely functionalized macrocyclic peptide isolated from the Cyanobacterium symploca sp. by Luesch and coworkers.Citation34 Entinostat and Mocetinostat have selectivity for HDAC1-3 and also against HDAC11 for the latter. Valproate and sodium butyrate (NaB) better target HDAC I and IIa. For sirtuinsCitation35 inhibition is based on NAD+competitive binding with attempts to propose a pharmacophore, according to the various inhibitor structures described.Citation36

SIRT1 activation is the novel therapeutic approach to treat chronic inflammatory diseases, and enzyme activators are therefore sought. Many screening tests to search for HDACi use short histone peptides, capturing baits and engineered cells. All have their limitations because, in vivo, HDAC are parts of mega Daltons modeling chromatin complexes that may change within each cell type.

HDAC inhibitors metabolism

The metabolism of HDACi is an important concern during clinical assays. It is studied to determine the correlation between HDACi blood concentration, effective biological effects and eventual drug interactions. The known metabolisms of some HDACi are reported in . TSA is metabolized as the inactive trichostatic acid, which is further demethylatedCitation37 for rapid clearance (). Phenyl butyrate (PB) metabolism has been described in several contexts.Citation38 PB is β-oxidized to phenylacetate, and cleared out upon glutamine addition. Vorinostat is also oxidized to 4-anilino-4-oxobutanoic acid and glucuronylated.Citation39,Citation40 Romidepsin is a disulfide prodrug. The real active form corresponds to the free thiol metabolite,Citation41 produced in vivo; the butenthiol part being thought to be the zinc-binding group. A glutathione conjugate has also been described,Citation42 which is metabolized in vivo by the cytochrome P450sCitation43 with slow and high acetylating subjects. Other HDACi stabilities have been investigated.Citation44,Citation45

Figure 4 Metabolic processes for some histone deacetylase inhibitors.

Abbreviations: PB, phenyl butyrate; TSA, trichostatin.

Figure 4 Metabolic processes for some histone deacetylase inhibitors.Abbreviations: PB, phenyl butyrate; TSA, trichostatin.

In-vitro effects of HDAC inhibitors

DNA chips studied the transcriptome of cells treated with Vorinostat and Romidepsin,Citation46 revealing that the expression of 40% of all genes was affected over a period of 16 hours. A Belinostat mRNA signature of 25 genes was sufficient to assess the overall gene modulation. Panobinostat modulated cell cycles and angiogenic genes.Citation47 Tumor antigen expression modulation and major histo-compatibility antigen (MHC) molecule inductionCitation48 have been observed with Dacinostat.Citation49 Mice bearing human tumor xenografts treated with Belinostat showed a modulation of the expression of genes active in the cellular G2/M phase. This was different from what was seen with 5-fluorouracil (5-FU), Cisplatin, Paclitaxel, or Thiotepa. Synergistic effects were obtained when combining HDACi and DNA demethylating agents,Citation50 or HDACi and all-trans retinoic acid (ATRA), a cell-differentiating agent used to treat acute promyelocytic leukemia (APL).Citation51 The influence of epigenetic modulators to modify stem cell fate and its relevance for curing diseases has been reviewed.Citation52 Successful therapeutic use of HDACi may thus depend on the cellular environment, the specific HDAC targeted, and the relative dependence of the tumor on the unique set of pathways influenced by a specific HDAC. Results are summarized in .Citation53

Table 1 In-vitro modulation of gene expression by HDACi

Clinical trials with zinc-dependent HDACi

This part of the review describes the HDACi that have been or are being investigated in clinical trials. In , all current trials are recapitulated. In , and for each molecule, some data related to epigenetic measurements are summarized.

Table 2 Clinical trials for epigenetic drugs

Table 3 In vivo HDACi effects from clinical data

PB or its sodium salt

PB or its corresponding sodium salt (NaPB) is a short chain fatty acid approved by the Food and Drug Administration (FDA) for the treatment of hyperammonemia. It stops the cell cycle in its G1–G0 phase. PB is an efficient HDACi at about 0.5 mM.Citation54,Citation55 PB induces apoptosis – probably via c-jun N-terminal kinase (JNK) – in lung carcinoma cells,Citation56 p21waf1-mediated growth arrest in MCF-7 cells,Citation57 tumor necrosis factor (TNF)-αCitation58 or peroxisome proliferator-activated receptor (PPAR)λ-mediatedCitation59 cell differentiation, and is more potent than phenylacetate in prostate cancer cells,Citation60 while increasing MHC class I expression. PB is converted in vivo into the active metabolite phenylacetate (PA) by β-oxidation in the liver and kidney mitochondria.Citation61 Most dose-limiting toxicities (DLTs) are fatigue, nausea, and somnolence. Preliminary studies have been conducted in patients with recurrent glioblastoma multiform (GBM)Citation62 (). Phase I studies have been conducted in patients with hormone refractory prostate cancers,Citation63 refractory solid tumor malignanciesCitation64 like colon carcinoma, non small cell lung cancer (NSCLC), anaplastic astrocytoma, GBM, bladder carcinoma, sarcoma, ovarian carcinoma, rectal hemangiopericytoma, and pancreatic carcinoma,Citation65 mainly as intravenous infusions but also in AML and myelodysplastic syndrome (MDS).Citation66 DLTs were neuro-cortical with milder fatigue and nausea/vomiting, light-headedness, short-term memory loss, sedation, confusion, and hypocalcemia. Although central nervous system (CNS) toxicity was observed, infusions were well tolerated (). The active metabolite PA accumulated.

In the AML/MDS study,Citation67 with sequential administration of 5-aza-cytidine (5-aza) (), partial remissions or stable diseases were obtained. Targeting different biological mechanisms is feasible with acceptable toxicity. Phase I trials in combination with several drugs have been reported. Prostate, colorectal, leiomyosarcoma, and esophageal cancers were treated in combination with 5-aza (),Citation68 metastatic colorectal cancer with fluorouracil 5-FU as a 24-hour continuous intravenous infusion (CIV).Citation69 With 5-aza, no re-expression of E-cadherin, endothelin B, and glutathione S transferase (GST) pi was observed, a result explained by the lack of dose effect or by the fact that DNA methylation is an S-phase-dependent process while in-vivo prostatic cells may be in S-phase at any given time. Stable disease was the best response. Combining 5-FU appeared also feasible.

Pivaloyloxymethyl butyrate

Pivaloyloxymethyl butyrate (AN)-9, is an ester prodrug of butyric acid (BA)Citation70 but with a greater potency at inducing malignant cell differentiation and tumor growth inhibition. It showed more favorable toxicological, pharmacological, and pharmaceutical properties than BA in preclinical studies. BA itself induces p16 expression and growth arrest of colon cancer cells,Citation71 and modifies caspase distribution during apoptosis.Citation72 AN-9 down regulates c-jun and c-myc and induces differentiation in leukemia cells.Citation73 It is decomposed by esterases in vivo to yield butyric and pivaloyl acids and a formaldehyde molecule, responsible for toxicity resulting in visual acuity disorders. It has demonstrated a synergistic effect with other anticancer agents by reducing bcl-2 levels.Citation74 Initial studyCitation75 with I.V. of AN-9 in advanced solid malignancies () gave partial responses, and stable diseases as best responses. Later, a multicenter trial of pivaloyloxymethyl butyrateCitation76 in refractory NSCLC (), administered as a continuous I.V. infusion, gave partial responses.

VPA

VPA is a nontoxic short-chain carboxylic acid used for the treatment of epilepsy with a long clinical history and well known pharmacokinetics (PKs) and pharmacodynamics (PDs).Citation77,Citation78 VPA induces chromatin decondensation,Citation79 and differentiation in neural progenitor cells,Citation80 and inhibits HDAC activityCitation81 in the mM range (preferentially HDAC1, 2).Citation82 The antiproliferative activity was associated with aberrant cyclin D3 functionality during the C6 glioma G1 phase.Citation83 Activation of PPARδ was present in F9 cells.Citation84 VPA induces caspase-dependent and -independent apoptosis in leukemia cells,Citation85 and in AML cells expressing P-gp and multidrug resistance protein 1 (MRP1),Citation86 inhibits production of TNF-α and interleukin (IL)-6 and activates nuclear factor kappa B (NF-κB).Citation87 VPA has been evaluated in combination with other anticancer compounds. For AML, increased 5-aza cytotoxicity was associated to cyclin D1 and p27(Kip1) expression,Citation88 while sequential VPA/ATRA treatment reprograms differentiation.Citation89 VPA induces p16INK4A upregulation and apoptosis and sensitizes melanoma cells to chemotherapy.Citation90 Interestingly, most of the clinical trials reported are for combination therapies.

A Phase I was conductedCitation91 for refractory advanced cancer (colorectal, melanoma, NSCLC, and others) (). VPA/ATRA combination was evaluated for several diseases. Poor risk AMLCitation92 (), MDS and relapsed or refractory AMLCitation93 () have also been investigated. A 52% response rate was observed in MDS patients. ATRA exerted no additional effect in patients receiving the combination, but could be used to induce a second response in relapsing VPA-treated patients. In recurrent or refractory AML or MDS in a Phase II protocolCitation94 (), ATRA was administered when VPA reached the target serum concentration. The differentiation therapy with VPA was effective in 30% of patients. In 11 elderly patients, de novo AMLCitation95 () was also treated with theophyllin to increase cAMP levels and major cell differentiation.Citation96 Complete marrow response was observed in three patients, including one complete remission. Two additional patients had hematologic improvement. Patients with AML-M6 were found particularlyCitation97 responsive, probably due to T-cell acute lymphocytic leukemia 1 (TAL1)Citation98 and GATA1Citation99 interactions with HDACi, inducing differentiation in murine erythroleukemia (MEL) cells. Siitonen et alCitation100 reported a negative study trying VPA, in combination with 13-cis-retinoic acid (13-cis-RA)Citation101 and 1,25-dihydroxyvitamin D3, in 19 naive patients with MDS or chronic myelomonocytic leukemia (CMML) (). Combinations with demethylating agents have been reported. Phase I/IICitation102 study with 5-aza-2′-deoxycytidine (5-azaDc) in leukemia () included gene expression analysis (p57kip2, p15, p73, MDR1 and THBS2). Initial DNMT1 levels were too low to be informative. A Phase I studyCitation103 with 5-azaDc in AML () gave partial to complete remissions, warranting further studies of 5-azaDc alone or with alternative HDACi. A Phase ICitation104 study of epigenetic modulation with 5-aza for advanced cancers (, colon, skin melanoma, breast, other) gave stable diseases. A Phase I/II studyCitation105 with 5-aza and ATRA for AML and MDS () gave 42% positive overall responses.

Other combinations were investigated: a Phase I dose escalation combination trial with epirubicin, 5-FU, and cyclophosphamideCitation106 in breast cancer (), and a Phase I trialCitation107 with epirubicin for solid tumors (). The rationale for the combination was to facilitate epirubicin access to DNA to potentiate its strand breaks activity as a topoisomerase II inhibitor. Intrinsic epirubicin toxicity was not exacerbated. Reverse combination was found inadequate by the same group. The same group investigated combination with the topoisomerase I inhibitor karenitecin (KTN) () for treating melanoma with both Phase I/II trials.Citation108 No VPA/KTN synergistic toxicity was observed. The best response was disease stabilization. VPA plus chemoimmunotherapy was investigated in a Phase II studyCitation109 for advanced inoperable or metastatic melanoma (), HDACi having been previously found to have a tumorigenic potential in melanoma.Citation110 Some patients then received dacarbazine plus interferon-α with VPA.

The magnesium salt of VPA has been tested in phase ICitation111 for cervical cancer (squamous and in adenocarcinoma) and Phase IICitation112 clinical trials. In the Phase I study (), VPA was given per os, and the authors emphasized the requirement for new endpoint trials based on biomarker analysisCitation113,Citation114 with, in this particular case, H3 and H4 acetylation and in vivo HDAC inhibition detection. The Phase II study was conducted with hydralazine, a demethylating agent,Citation115 () to overcome chemotherapy resistance in refractory solid tumors (cervix, breast, ovarian, and others). Partial responses and disease stabilization were the best responses.

Vorinostat

Vorinostat (suberoylanilide hydroxamicacid [SAHA], Zolinza®) has probably been the most studied compound in clinical trials on several cancer types. SAHA induces differentiation,Citation116 growth arrest,Citation117 or apoptosis at micromolar concentrations. Vorinostat is an unselective zinc-bindingCitation118 hydroxamic-acid-type inhibitor of HDAC1, 2, 3, 6, and 8. In glioma cells, SAHA induced expression of DR5, TNFα, p21Waf1, and p27Kip1 and reduced expression of CDK2, CDK4, cyclin D1, and cyclin D2.Citation119 SAHA can induce thyroid cancer cell death by caspase-mediated pathways,Citation120 and induces G1 and G2-M arrest and apoptosis in several types of breast cancer cell lines,Citation121,Citation122 NSCLC,Citation123 and prostate cancer cells.Citation124 It potentiates the activity of other molecules like Paclitaxel in ovarian cancers.Citation125

Phase I trials have been described for both oral and I.V administrations. Escalating I.V. administrationCitation126 in solid tumors and hematological malignancies () gave hypotension for one schedule. In mesothelioma, with I.V. or oral formulations,Citation127 the best responses were partial (). An oral formulation for hematologic malignancies (Hodgkin’s and others) () and solid tumors (mainly mesothelioma, prostate, urothelial, thyroid, and renal)Citation128 gave one complete response while others were incomplete. Oral twice- or thrice-daily administrations in advanced leukemias and MDS (AML, CLL [chronic lymphocytic leukemia], MDS, ALL and CML [chronic myelocytic leukemia]) ()Citation129 gave two complete responses and two complete responses with incomplete blood-count recovery (all with AML treated at/below maximum tolerated dose [MTD]).

Phase II clinical trials were mainly proposed with oral formulations. A multi-institutional trialCitation130 in women with recurrent or persistent epithelial ovarian () or primary peritoneal carcinoma platinum-resistant/refractory gave one partial response. Another multicenter open-label oral trialCitation131 investigated measurable, relapsed, or refractory breast cancer, NSCLC, or colorectal cancer (). Disease stabilization was observed in eight patients. SAHA is tolerated at 200 mg only, in a daily oral schedule for 14 days–3 weeks. In recurrent and/or metastatic head and neck cancer (400 mg every day) ()Citation132 no confirmed responses have been observed. In patients with metastatic breast cancer,Citation133 there were no complete or partial responses, and the heterogeneity of the recruited patients did not allow production of significant statistical results. Eight patients were positive for estrogen and/or progesterone receptors, four had amplified CerB-2. Fatigue, nausea, diarrhea, and lymphopenia were the most frequent clinically significant adverse effects. In GBMCitation134 (), an oral dose of 200 mg followed by a 7-day rest period showed that SAHA monotherapy is well tolerated with modest single-agent activity. Although HDACi were shown to induce cell death and sensitize cells to cytotoxic chemotherapy in thyroid cancer cell lines, Woyach et alCitation135 described the lack of therapeutic effect of SAHA in patients with metastatic radioiodine-refractory thyroid carcinoma in a Phase II study (). A Phase II oral combination therapy was proposed with carboplatin (I.V.) and Paclitaxel (I.V.) for advanced solid malignanciesCitation136 (). Eleven partial responses occurred and seven disease stabilizations. The regimen requires drug–drug interaction to be determined. Encouraging results were obtained in patients with previously untreated NSCLC.

Belinostat

Belinostat (PXD101) is a recent hydroxamic acid HDACi that has growth-inhibitory and pro-apoptotic activity in several cancer types at submicromolar concentrations,Citation137,Citation138 and that has been investigated in ovarian cancers.Citation139 It down regulates thymidilate synthase, vascular endothelial growth factor (VEGF), aurora kinase, and epidermal growth factor receptor (EGFR), and up regulates cyclin A. It has been used in combination. A gene expression-signature profiling has been reported for Belinostat.Citation140 According to publicationsCitation141 PKs gave a general 1–2-hour half-life. In early trials, DNA fragmentation increased with a combination of 5-FU in HCT116 colon cancer cells in vitro and in both HT-29 and HCT116 in xenograft models,Citation142 and also a poly(ADP-ribose) polymerase (PARP) cleavage and down regulation of thymidylate synthase expression in HCT116. Improved tumor reduction was obtained in vivo with mouse HT29 and HCT116 xenograft models compared with single compounds, validating a rationale for the clinical schedule.

In Phase I treatment of refractory solid tumors by I.V. administration ()Citation143 the caspase-dependent cleavage of cytokeratin-18 was determined to measure the level of apoptosis.Citation144 Heavily pre-treated patients with advanced hematological neoplasiaCitation145 () were also treated. In Phase II trials, investigations of I.V. administration of relapsed malignant pleural mesotheliomaCitation146 () indicated that combination strategies or alternate dosing schedules might be necessary. In resistant micro-papillary ovarian tumors (low malignant potential [LMP]) and epithelial ovarian cancers (EOC) (),Citation147 a Phase II trial with Belinostat gave partial responses or stable diseases for LMP, and stabilized diseases for EOC.

Givinostat

Givinostat (ITF2357) belongs to the hydroxamic acid family of HDACi which is very similar to SAHA. It inhibits IL-6 and VEGF production in stromal cells.Citation148

Two Phase II studies were described for relapsed/refractory Hodgkin lymphoma (HL) (). A first oral one, gave stable diseases by computed tomography (CT) scan that have been associated with a significant reduction in fluorodeoxyglucose-positron emission tomography scan uptake.Citation149 Galli et alCitation150 developed a Phase II multicenter trial in 19 heavily treated patients that were relapsing from progressive multiple myeloma (MM) (). The best responses were disease stabilization. This regimen appears as unlikely to play a significant role for advanced MM, and other combinations with currently used drugs should be investigated. A combination with the alkylating agent mechlorethamineCitation151 () was investigated in relapsed/refractory HL.

Panobinostat

Panobisnostat (LBH589) is a hydroxamic acid HDACi, which has demonstrated anti-angiogenic and anti-proliferative activities in human prostate carcinoma cell PC-3 xenografts in vivo, inducing H3 and tubulin acetylationCitation152 in human umbilical vascular endothelial cells (HUVEC), which corresponded to G2-M cell cycle arrest and inhibition of HUVEC cell proliferation and viability. Non cytotoxic concentrations of Panobinostat inhibited endothelial tube formation, matrigel invasion, AKT, extracellular signal-regulated kinase 1/2 phosphorylation, and chemokine receptor CXCR4 expression. Association with anti-VEGF therapies should be considered. Prince et al have discussed preclinical data on Panobinostat and emerging data from Phase I and II studies in cancer patients.Citation153

A Phase I study in refractory hematologic malignancies (AML, ALL, and MDS) ()Citation154 with I.V. administration appeared convenient to obtain anti-leukemic and biological effects. In cutaneous T-cell lymphoma (CTCL)Citation47 with oral formulation (), the responses ranged from disease stabilization to complete remission, showing the potential of this molecule in CTCL. In combination with DocetaxelCitation155 (), a microtubule interacting agent for castration-resistant prostate cancer, Panobinostat inhibited LnCAP androgen receptor positive prostate cancer cell proliferation, potentiated by Docetaxel. Single or combined treatments were administered with oral Panobinostat.

Dacinostat

Dacinostat (NVPLAQ824, LAQ) is a hydroxamic acid derivative similar to Panobinostat.Citation156 It showed anti-neoplastic activity and can activate genes that produce cell cycle arrest. It acetylates hsp90, inducing proteosomal degradation of Bcr-Abl and HER-2. Combination of Dacinostat with 5-azaDcCitation157 in human MDA-MB-231 and MCF-7 breast carcinoma cells showed a synergic anti-neoplastic activity for the MDA-MB-231. For the MCF-7 tumor cells, simultaneous 5-azaDc and Dacinostat administration were antagonistic, unseen when used in a sequential schedule (5-azaDc first). This is probably due to interference in the S-phase of Dacinostat since 5-azaDc is a S-phase specific interfering molecule. Dacinostat appeared to be well tolerated in clinical trials. Phase I investigationsCitation158 in advanced solid tumors included measure of HSP72 levels and was consistent with HSP90 inhibition (). Another groupCitation159 reported the same results with increased expression of Hsp70 and decreased c-Raf levels. The biological importance of these non histones mediated effects requires further study. I.V. administration for ALL, AML, CLL, CMLCitation160 () in blast crisis or advanced MDS gave some stable diseases.

PCI-24781

PCI-24781 is a broad-spectrum hydroxamic acid-based HDACi. PCI-24781 reverses drug resistance in four multidrug resistant sarcoma cell lines and synergizes with chemotherapeutic agents to enhance caspase-3/7 activity.Citation161

In refractory advanced solid tumorsCitation162 (), I.V. administration followed by dose escalation was well tolerated. Electro cardiac monitoring revealed grade ≤1 QTcF (QT interval corrected for heart rate using Fridericia’s formula) prolongation and asymptomatic nonspecific ST and T wave changes leading to discontinuation.

Entinostat

Entinostat (MS-275, SNX-275) is a benzamide HDACi, which promotes expression of genes involved in growth arrest and differentiation, like p21 and the maturation marker: gelsolin,Citation163 inducing caspase-dependant apoptosis in CLL B-cells,Citation164 p21CIP1/WAF1 differentiation or apoptosis in human leukemia cells,Citation165 and also tissue growth factor (TGF)βII receptor expression in human breast cancer.Citation166 Reported half-life in animals is about 1 hour, and species-variable protein binding was reported.Citation167 Half-life in human plasma was higher than in animals, which is supposedly to be linked to protein binding, as Entinostat was found to be 80% bound.Citation167

Phase I study in advanced solid tumors or lymphoma by oral routeCitation168 () was reasonably well tolerated. In refractory solid tumors and human lymphoid malignanciesCitation169 (), drug exposure increases linearly with dose. In AMLCitation170 (), results showed that Entinostat effectively inhibits HDAC in vivo in patients with AML and should be further tested, preferably in patients with less-advanced disease. Several protocols were designed for patients with advanced solid malignancies and lymphomasCitation171 (). PKs revealed dose-dependent and dose-proportional increases. Responses were partial remissions and prolonged disease stabilization. In a Phase II study for metastatic melanoma () with low efficacy treatment,Citation172 long-term tumor stabilizations have been observed, but no objective responses was assessed.

Mocetinostat

Mocetinostat (MGCD0103)Citation173 is a benzamide selective class I/IV HDACi (1, 2, 3, and 11). It inhibits neoplastic growth in multiple human tumor xenograft models including colon (HCT116, SW48, and Colo205), NSCLC (A549), prostate (DU145), pancreatic (PANC1), and vulval epidermal (A431) cancer models and does not interact with the potassium voltage-gated channel, subfamily H (eag-related), member 2 (HERG) channel. Gene expression induced by Mocetinostat is modest compared with other hydroxamic HDACi.Citation174

In patients with advanced solid tumorsCitation175 (), a phase I study gave disease stabilization as the best response. IL-6 induction related to HDACi activity has been postulated but not confirmed. At the tested doses, Mocetinostat appeared tolerable and exhibited favorable PK and PD profiles, as well as evidence of target inhibition in surrogate tissues. Cytogenetically analyzed patients with AML, MDS, ALL, and CMLCitation176 () were treated orally. A total of 18 of the 29 patients had abnormal cytogenetics. PK analyses indicated rapid absorption of Mocetinostat. Several administration schedules have been proposed for advanced leukemias or MDSCitation177 (). A Phase II study for HLCitation178 () demonstrated significant anti-tumor activity in relapsed/refractory post-transplant HL. For 437 patientsCitation179 (), partial responses were obtained. Extended studies are ongoing.

Tacedinaline

The long-known molecule Tacedinaline (CI-994) is a benzamide HDACi similar to MS-275Citation180 with anti-tumor activities in HCT-8 colon carcinoma.Citation181 Following Tacedinaline administration, inhibition of both histone deacetylation and cellular proliferation at the G1 to S transition phase of the cell cycle are observed.

Oral administration with food intake for solid tumorsCitation182 () did not affect the rate or extent of the drug absorption. Best responses were partial or stable diseases. Advanced solid malignancies (mainly colorectal, pancreatic and mesothelioma) were treated in combination with CapecitabineCitation183 (), an FDA-approved compound used to treat a variety of cancer. Three treatment protocols were implemented. A combination Phase II study with GemcitabineCitation184 for advanced pancreatic carcinoma () gave no improvement.

Depsipeptide

Depsipeptide (Romidepsin, FK228, FR901228) is a cyclic tetrapeptide isolated from Chromobacterium violaceum which has demonstrated anti tumor activities (A549 lung adenocarcinoma, MCF-7 and ZR-75-1 breast adenocarcinoma, and LOX IMVI melanoma cell lines) and is postulated as a Pg-p substrate.Citation185,Citation186 It is considered as a natural HDACi prodrug, as its disulfide bond is reduced in vivo to give the active speciesCitation40 and is the only reported sulfur-based HDACi used in clinical trials. It received FDA approval for cutaneous T-cell lymphoma in 2009. Romidepsin induces growth arrest and apoptosis in lung cancer cells.Citation187 Romidepsin induces p21-dependent G1 arrest and p21-independent G2 arrestCitation188 by downregulating cyclin D1 and upregulating cyclin E.Citation189 It inhibits c-Myc and Fas ligand expression,Citation190 modulates p53, ErbB1, HER2, and Raf-1 expression in lung cancer cells,Citation191 increases p21, phosphorylation of Bcl2, and apoptosis in human breast cancer cells,Citation192 increases expression of a NaI symporter in thyroid carcinoma cells for possible resensitization of radio resistant thyroid cancer,Citation193 and activates the caspase 8-mediated apoptosis and down regulates the c-FLIP protein.Citation194 Sequential treatments with 5-azaDc facilitates cancer cell recognition by T lymphocytes specific for cancer/testis antigen 1B (NY-ESO-1) as a possible option for immunotherapyCitation195 or induces tissue factor pathway inhibitor (TFPI)-2 expression in cancer cells.Citation196 Initial cardiac toxicity was resolved by convenient administration schedules but cardiac monitoring is most of the time implemented during clinical investigations. Concentrations studies in CLL and AML have correlated apoptosis induction and HDAC inhibition. Combination of Romidepsin and DNA demethylating agents is potentiated in ETO positive leukemia cells.Citation197 A gene signature specific for Romidepsin sensitivity has been reported.Citation198 Due to the recent approval of Romidepsin in CTCL, all early published clinical trials for this disease are not discussed.Citation199,Citation200 Romidepsin induced MDR-1 gene expression in several cancer cell lines.

A Phase I study in advanced, incurable cancersCitation201 () indicated that further clinical trials are warranted. Used for advanced or refractory neoplasmsCitation202 (), elimination half-life was 8.1 hours. In CCL and AMLCitation203 (), intravenous treatment gave no responses. Romidepsin effectively inhibits HDAC in vivo in patients with CLL and AML, but future studies should examine alternative administration routes. In refractory or recurrent solid tumorsCitation204 (), DLTs were not associated with changes in troponin levels or evidence of ventricular dysfunction, transient asymptomatic sick sinus syndrome and hypocalcemia. For MDS and AMLCitation205 (), intravenous administration gave no significant cardiac toxicity. Romidepsin therapy can be administered with acceptable short-term toxicity. Gastrointestinal symptoms and fatigue seemed to be treatment-limiting after multiple cycles. Phase II performed on refractory renal cell cancerCitation206 (), I.V., gave classical but serious toxicities. Two patients developed a prolonged QT interval, one patient developed grade 3 atrial fibrillation and tachycardia, and there was one sudden death. In metastatic neuroendocrine tumorsCitation207 (), adverse events were ventricular tachycardia and prolonged QT, possibly resulting in a sudden death, terminating the study prematurely. Romidepsin has serious cardiac adverse events, and risks need to be comprehensively evaluated. In lung cancers (NSCLC and SCLC) (),Citation208 Romidepsin was not appropriate. This study presented an in depth gene expression profiling.

Conclusion

A number of clinical trials have been completed and many others are ongoing using HDACi as single agents and in combination with radiotherapy and/or chemotherapy for the treatment of various hematological and solid malignancies with some promising early results. Vorinostat is the most established HDACi, and was approved in October 2006 by the FDA for the treatment of advanced forms of cutaneous T-cell lymphoma that have failed multiple other systemic treatment options. Significant single agent activity for Romidepsin has also been demonstrated in peripheral cutaneous T-cell lymphoma, and encouraging results have also been seen in HL with Mocetinostat. From the trials conducted, it is also clear that a major clinical advantage is that HDACi are well tolerated in the majority of patients. The future of HDACi lies in designing rational combination therapies. The sequence of drug administration may be of paramount importance to avoid antagonistic effects. The possibility of drug–drug interactions and enhanced toxicities is to be considered. HDACi are also evaluated in non cancerous pathologies like AIDS, graft versus host diseases, and polycythemia verae. Very soon, SIRT activators could find therapeutic applications in lung interstitial diseases. Like for the kinase inhibitors, more selective third generation HDACi are sought, yet specific tests remain to be designed to screen for bioactivity in vitro and in vivo.Citation211

Acknowledgements

This work was supported in part by the COST action TD0905 Epigenetics: bench to bedside and by the Agence Nationale de la Recherche (France).

Disclosure

The authors report no conflicts of interest in this work.

References

  • ChoudhuriSCuiYKlaassenCDMolecular targets of epigenetic regulation and effectors of environmental influencesToxicol Appl Pharmacol2010245337839320381512
  • Santos-RosaHCaldasCChromatin modifier enzymes, the histone code and cancerEur J Cancer200541162381240216226460
  • OliverSSDenuJMDynamic interplay between histone H3 modifications and protein interpreters: emerging evidence for a “histone language”Chembiochem201112229930721243717
  • ZeeBMLevinRSDimaggioPAGlobal turnover of histone post-translational modifications and variants in human cellsEpigenetics Chromatin20103223321134274
  • DekkerFHaismaHJHistone acetyl transferases as emerging drug targetsDrug Discovery Today20091419/2094294819577000
  • LimSMetzgerESchüleREpigenetic regulation of cancer growth by histone demethylasesInt J Cancer201012791991199820607829
  • NicholsonTBChenTRichardSThe physiological and pathophysiological role of PRMT1-mediated protein arginine methylationPharmacol Res200960646647419643181
  • FackelmayerFOProtein arginine methyltransferases: guardians of the Arg?Trends Biochem Sci2005301266667116257219
  • TiniMNaeemHTorchiaJBiochemical analysis of arginine methylation in transcriptionMethods Mol Biol200952323524719381935
  • SpannhoffATHauserRHeinkeWThe emerging therapeutic potential of histone methyltransferase and demethylase inhibitorsChemMedChem20094101568158219739196
  • ZhangYXuKNiMNucleosome dynamics define transcriptional enhancersNature Genetics201042434334720208536
  • CopelandRAOlhavaEJScottMPTargeting epigenetic enzymes for drug discoveryCurr Opin Chem Biol201014450551020621549
  • JonesPABaylinSBThe fundamental role of epigenetic events in cancerNat Rev Genet20023641542812042769
  • KriaucionisSHeintzNThe nuclear DNA base 5-hydroxymethylcytosine is present in purkinje neurons and the brainScience2009324592992993019372393
  • Abdel-WahabOMullallyAHedvatCGenetic characterization of TET1, TET2, and TET3 alterations in myeloid malignanciesBlood2009114114414719420352
  • FabbriMCalinGAEpigenetics and miRNAs in human cancerAdv Genet201070879920920746
  • ZainJKaminetzkiDO’ConnorOAEmerging role of epigenetic therapies in cutaneous T-cell lymphomasExpert Rev Hematol20103218720321083462
  • GrahamJSKayeSBBrownRThe promises and pitfalls of epigenetic therapies in solid tumorsEur J Cancer20094571129113619211243
  • BoldenJEPeartMJJohnstoneRWAnticancer activities of histone deacetylase inhibitorsNat Rev Drug Discov20065957695784
  • HaberlandMMontgomeryRLOlsonENThe many roles of histone deacetylases in development and physiology: implications for disease and therapyNat Rev Genet2009101324219065135
  • RazidloDFWhitneyTJCasperMEHistone deacetylase 3 depletion in osteo/chondroprogenitor cells decreases bone density and increases marrow fatPLoS One201057e1149220628553
  • BhaskaraSChylaBJAmannJMDeletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage controlMol Cell2008301617218406327
  • De ZoetenEFWangLSaiHInhibition of HDAC9 increases T regulatory cell function and prevents colitis in miceGastroenterology2010138258359419879272
  • KotianSLiyanarachchiSZelentAHistone deacetylases 9 and 10 are required for homologous recombinationJ Biol Chem. Epub 2011 Jan 18.
  • KellyGA review of the sirtuin system, its clinical implications, and the potential role of dietary activators like resveratrol: part 1Altern Med Rev201015324526321155626
  • ZhangYKwonSYamaguchiTMice lacking histone deacetylase 6 have hyperacetylated tubulin but are viable and develop normallyMol Cell Biol20082851688170118180281
  • MaiAAltucciLEpi-drugs to fight cancer: from chemistry to cancer treatment, the road aheadInt J Biochem Cell Biol200941119921318790076
  • MarksPADokmanovicMHistone deacetylase inhibitors: discovery and development as anticancer agentsExpert Opin Investig Drugs2005141214971511
  • BertrandPInside HDAC with HDAC inhibitorsEur J Med Chem20104562095211620223566
  • Yurek-GeorgeACecilARMoAHThe first biologically active synthetic analogues of FK228, the depsipeptide histone deacetylase inhibitorJ Med Chem200750235720572617958342
  • KhanNJeffersMKumarSDetermination of the class and isoform selectivity of small-molecule histone deacetylase inhibitorsBiochem J2008409258158917868033
  • KrennHrubecKMarshallBLHedglinMDesign and evaluation of ‘linkerless’ hydroxamic acids as selective HDAC8 inhibitorsBioorg Med Chem Lett200717102874287817346959
  • EstiuGGreenbergEHarrisonCBStructural origin of selectivity in class II-selective histone deacetylase inhibitorsJ Med Chem200851102898290618412327
  • TaoriKPaulVJLueschHStructure and activity of largazole, a potent antiproliferative agent from the Floridian marine cyanobacterium Symploca spJ Am Chem Soc200813061806180718205365
  • HuhtiniemiTSuuronenTLahtela-KakkonenMNε-Modified lysine containing inhibitors for SIRT1 and SIRT2Bioorg Med Chem201018155616562520630764
  • HuhtiniemiTWittekindtCLaitinenTComparative and pharmacophore model for deacetylase SIRT1J Comput Aided Mol Des200620958959917103016
  • ElautGTörökGVinkenMMajor phase I biotransformation pathways of Trichostatin A in rat hepatocytes and in rat and human liver microsomesDrug Metab Dispos200230121320132812433798
  • EbbelENLeymarieNSchiavoSIdentification of phenylbutyrate-generated metabolites in Huntington disease patients using parallel liquid chromatography/electrochemical array/mass spectrometry and off-line tandem mass spectrometryAnalytical Biochem20103992152161
  • PariseRAHolleranJLBeunerJHA liquid chromatography–electrospray ionization tandem mass spectrometric assay for quantitation of the histone deacetylase inhibitor, vorinostat (suberoylanilide hydroxamicacid, SAHA), and its metabolites in human serumJ Chrom B20068402108115
  • DuLMussonDGWangAQStability studies of vorinostat and its two metabolites in human plasma, serum and urineJ Pharm Biomed Anal200642555656416824724
  • FurumaiRMatsuyamaAKobashiNFK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylasesCancer Res200262174916492112208741
  • XiaoJJByrdJMarcucciGIdentification of thiols and glutathione conjugates of depsipeptide FK228 (FR901228), a novel histone protein deacetylase inhibitor, in the bloodRapid Commun Mass Spectrom200317875776612672127
  • ShiragaTTozukaZIshimuraRIdentification of cytochrome P450 enzymes involved in the metabolism of FK228, a potent histone deacetylase inhibitor, in human liver microsomesBiol Pharm Bull200528112412915635176
  • KimHMOhSJParkSKIn vitro metabolism of KBH-A40, a novel delta-lactam-based histone deacetylase (HDAC) inhibitor, in human liver microsomes and serumXenobiotica200838328129318274957
  • FonsiMFioreFJonesPMetabolism-related liabilities of a potent histone deacetylase (HDAC) inhibitor and relevance of the route of administration on its metabolic fateXenobiotica2009391072273719569735
  • PeartMJSmythGKvan LaarRKIdentification and functional significance of genes regulated by structurally different histone deacetylase inhibitorsPNAS2005102103697370215738394
  • EllisLPanYSmythGKHistone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphomaClin Cancer Res200814144500451018628465
  • KatoNTanakaJSugitaJRegulation of the expression of MHC class I-related chain A, B (MICA, MICB) via chromatin remodeling and its impact on the susceptibility of leukemic cells to the cytotoxicity of NKG2D-expressing cellsLeukemia200721102103210817625602
  • VoDDPrinsRMBegleyJLEnhanced antitumor activity induced by adoptive T-cell transfer and adjunctive use of the histone deacetylase inhibitor LAQ824Cancer Res200969228693869919861533
  • CameronEEBachmanKEMyöhänenSSynergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancerNat Genet19992111031079916800
  • AppelbaumFRBaerMRCarabasiMHNCCN Practice guidelines for acute myelogenous leukemiaOncology20001411A536111195419
  • SnykersSVinkenMRogiersVDifferential role of epigenetic modulators in malignant and normal stem cells: a novel tool in preclinical in vitro toxicology and clinical therapyArch Toxicol200781853354417387455
  • MarksPAMillerTRichonVMHistone deacetylasesCurr Opin Pharmacol20033434435112901942
  • DiGiuseppeJAWengLJYuKHPhenylbutyrate-induced G1 arrest and apoptosis in myeloid leukemia cells: structure-function analysisLeukemia19991381243125310450753
  • CarducciMANelsonJBChan-TackKMPhenylbutyrate induces apoptosis in human prostate cancer and is more potent than phenylacetateClin Cancer Res1996223793879816181
  • ZhangXWeiLYangYYuQSodium 4-phenylbutyrate induces apoptosis of human lung carcinoma cells through activating JNK pathwayJ Cell Biochem200493481982915389886
  • GorospeMShackSGuytonKZUp-regulation and functional role of p21Waf1/Cip1 during growth arrest of human breast carcinoma MCF-7 cells by phenylacetateCell Growth Differ1996712160916158959328
  • LiuLHudginsWRMillerACTranscriptional upregulation of TGF-α by phenylacetate and phenylbuytrate is associated with differentiation of human melanoma cellsCytokine1995754494567578983
  • HanSWadaRKSidellNDifferentiation of human neuroblastoma by phenylacetate is mediated by peroxisome proliferatoractivated receptor gammaCancer Res200161103998400211358817
  • CarducciMANelsonJBChan-TackKMPhenylbutyrate induces apoptosis in human prostate cancer and is more potent than phenylacetateClin Cancer Res1996223793879816181
  • BrusilowSWPhenylacetylglutamine may replace urea as a vehicle for waste nitrogen excretionPediatr Res19912921471502014149
  • PhuphanichSBakerSDGrossmanSAOral sodium phenylbutyrate in patients with recurrent malignant gliomas: a dose escalation and pharmacologic studyNeuro Oncol20057217718215831235
  • CarducciMAGilbertJBowlingMKA phase I clinical and pharmacological evaluation of sodium phenylbutyrate on an 120-h infusion scheduleClin Cancer Res20017103047305511595694
  • GilbertJBakerSDBowlingMKA phase I dose escalation and bioavailability study of oral sodium phenylbutyrate in patients with refractory solid tumor malignanciesClin Cancer Res2001782292230011489804
  • CamachoLHOlsonJTongWPPhase I dose escalation clinical trial of phenylbutyrate sodium administered twice daily to patients with advanced solid tumorsInvest New Drugs200725213113817053987
  • GoreSDWengLJZhaiSImpact of prolonged infusions of the putative differentiating agent sodium phenylbutyrate on myelodysplastic syndromes and acute myeloid leukemiaClin Cancer Res20028496397011948101
  • MaslakPChanelSCamachoLHPilot study of combination transcriptional modulation therapy with sodium phenylbutyrate and 5-azacytidine in patients with acute myeloid leukemia or myelodysplastic syndromeLeukemia200620221221716357841
  • GilbertJBakerSDDonehowerRMethytransferase (MT) activity and gene expression in tumor biopsies from patients enrolled in a Phase I study of the MT inhibitor, 5-azacytidine (5AC), and the histone deacetylase inhibitor, phenylbutyrate (PB), in refractory solid tumorsProc Am Soc Clin Onc.200187a
  • SungMWWaxmanSCombination of cytotoxicdifferentiation therapy with 5-fluorouracil and phenylbutyrate in patients with advanced colorectal cancerAnticancer Res2007272995100117465233
  • RephaeliARabizadehEAviramADerivatives of butyric acid as potential anti-neoplastic agentsInt J Cancer199149166721874573
  • SchwartzBAvivi-GreenCPolak-CharconSSodium butyrate induces retinoblastoma protein dephosphorylation, p16 expression and growth arrest of colon cancer cellsMol Cell Biochem19981881–221309823007
  • MandalMAdamLKumarRRedistribution of activated caspase-3 to the nucleus during butyric acid-induced apoptosisBiochem Biophys Res Commun1999260377578010403841
  • RabizadehEShaklaiMNudelmanARapid alteration of c-myc and c-jun expression in leukemic cells induced to differentiate by a butyric acid prodrugFEBS Lett199332832252298348968
  • RabizadehEBaireyOAviramADoxorubicin and a butyric acid derivative effectively reduce levels of Bcl-2 protein in the cells of chronic lymphocytic leukemia patientEur J Haematol200166426327111380606
  • PatnaikARowinskyEKVillalonaMAA phase I study of pivaloyloxymethyl butyrate, a prodrug of the differentiating agent butyric acid, in patients with advanced solid malignanciesClin Cancer Res2002872142214812114414
  • ReidTValoneFLiperaWPhase II trial of the histone deacetylase inhibitor pivaloyloxymethyl butyrate (Pivanex, AN-9) in advanced non-small cell lung cancerLung Cancer200445338138615301879
  • LoscherWBasic pharmacology of valproate: a review after 35 years of clinical use for the treatment of epilepsyCNS Drugs2002161066969412269861
  • PeruccaEPharmacological and therapeutic properties of valproate: a summary after 35 years of clinical experienceCNS Drugs2002161069571412269862
  • MarchionDCBicakuEDaudAIValproic acid alters chromatin structure by regulation of chromatin modulation proteinsCancer Res20056593815382215867379
  • JungGAYoonJYMoonBSValproic acid induces differentiation and inhibition of proliferation in neural progenitor cells via the beta-catenin-Ras-ERK-p21Cip/WAF1 pathwayBMC Cell Biol20089667819068119
  • GottlicherMMinucciSZhuJValproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cellsEMBO J200120246969697811742974
  • GurvichNTsygankovaOMMeinkothJLHistone deacetylase is a target of valproic acid-mediated cellular differentiationCancer Res20046431079108614871841
  • BaconCLGallagherHCHaugheyJCAntiproliferative action of valproate is associated with aberrant expression and nuclear translocation of cyclin D3 during the C6 glioma G1 phaseJ Neurochem2002831121912358724
  • WerlingUSiehlerSLitfinMInduction of differentiation in F9 cells and activation of peroxisome proliferator-activated receptor delta by valproic acid and its teratogenic derivativesMol Pharmacol20015951269127611306712
  • KawagoeRKawagoeHSanoKValproic acid induces apoptosis in human leukemia cells by stimulating both caspase dependent and -independent apoptotic signaling pathwaysLeuk Res200226549550211916526
  • TangRFaussatAMMajdakPValproic acid inhibits proliferation and induces apoptosis in acute myeloid leukemia cells expressing P-gp and MRP1Leukemia20041871246125115116123
  • IchiyamaTOkadaKLiptonJMSodium valproate inhibits production of TNF-α and IL-6 and activation of NF-κBBrain Res20008571–224625110700573
  • SiitonenTKoistinenPSavolainenERIncrease in Ara-C cytotoxicity in the presence of valproate, a histone deacetylase inhibitor, is associated with the concurrent expression of cyclin D1 and p27(Kip 1) in acute myeloblastic leukemia cellsLeuk Res200529111335134215936818
  • CiminoGLo-CocoFFenuSSequential valproic acid/Alltrans retinoic acid treatment reprograms differentiation in refractory and highrisk acute myeloid leukemiaCancer Res200666178903891116951208
  • ValentiniAGravinaPFedericiGValproic acid induces apoptosis, p16INK4A upregulation and sensitization to chemotherapy in human melanoma cellsCancer Biol Ther20076218519117218782
  • AtmacaAAl-BatranSEMaurerAValproic acid (VPA) in patients with refractory advanced cancer: a dose escalating Phase I clinical trialBritish J Cancer2007972177182
  • BugGRitterMWassmannBClinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemiaCancer2005104122717272516294345
  • KuendgenAKnippSFoxFResults of a Phase 2 study of valproic acid alone or in combination with alltrans retinoic acid in 75 patients with myelodysplastic syndrome and relapsed or refractory acute myeloid leukemiaAnn Hematol200584Suppl 1616616270213
  • PilatrinoCCilloniDMessaEIncrease in platelet count in older, poor-risk patients with acute myeloid leukemia or myelodysplastic syndrome treated with valproic acid and all-trans retinoic acidCancer2005104110110915895376
  • RaffouxEChaibiPDombretHValproic acid and all-trans retinoic acid for the treatment of elderly patients with acute myeloid leukemiaHaematologica200590798698815996941
  • DuprezELillehaugJRNaoeTcAMP signalling is decisive for recovery of nuclear bodies (PODs) during maturation of RA-resistant t(15;17) promyelocytic leukemia NB4 cells expressing PML-RAR alphaOncogene19961211245124598649787
  • KuendgenASchmidMSchlenkRThe histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemiaCancer2006106111211916323176
  • HuangSBrandtSJmSin3A regulates murine erythroleukemia cell differentiation through association with the TAL1 (or SCL) transcription factorMol Cell Biol20002062248225910688671
  • WatamotoKTowatariMOzawaYAltered interaction of HDAC5 with GATA-1 during MEL cell differentiationOncogene200322579176918414668799
  • SiitonenTTimonenTJuvonenEValproic acid combined with 13-cis retinoic acid and 1,25-dihydroxyvitamin D3 in the treatment of patients with myelodysplastic syndromesHaematologica20079281119112217650442
  • SantiniVFerriniPRDifferentiation therapy of myelodysplastic syndromes: fact or fiction?Br J Heamatol1998102511241138
  • Garcia-ManeroGKantarjianHMSanchez-GonzalezBPhase 1/2 study of the combination of 5-aza-2′-deoxycytidine with valproic acid in patients with leukemiaBlood2006108103271327916882711
  • BlumWKlisovicRBHackansonBPhase I study of decitabine alone or in combination with valproic acid in acute myeloid leukemiaJ Clin Oncol200725253884389117679729
  • BraitehFSorianoAOGarcia-ManeroGPhase I study of epigenetic modulation with 5-azacytidine and valproic acid in patients with advanced cancersClin Cancer Res200814196296630118829512
  • SorianoAOYangHFaderlSSafety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndromeBlood200711072302230817596541
  • MünsterPMarchionDBicakuEClinical and biological effects of valproic acid as a histone deacetylase inhibitor on tumor and surrogate tissues: Phase I/II trial of valproic acid and epirubicin/FECClin Cancer Res20091572488249619318486
  • MünsterPMarchionDBicakuEPhase I trial of histone deacetylase inhibition by valproic acid followed by the topoisomerase II inhibitor epirubicin in advanced solid tumors: a clinical and translational studyJ Clin Oncology2007251519791985
  • DaudAIDawsonJDeContiRCPotentiation of a topoisomerase I inhibitor, karenitecin, by the histone deacetylase inhibitor valproic acid in melanoma: translational and Phase I/II clinical trialClin Cancer Res20091572479248919318485
  • RoccaAMinucciSTostiGA Phase I–II study of the histone deacetylase inhibitor valproic acid plus chemoimmunotherapy in patients with advanced melanomaBritish J Cancer200910012836
  • FachettiFPrevidiSBallariniMModulation of pro- and anti-apoptotic factors in human melanoma cells exposed to histone deacetylase inhibitorsApoptosis20049557358215314285
  • Chavez-BlancoASegura-PachecoBPerez-CardenasEHistone acetylation and histone deacetylase activity of magnesium valproate in tumor and peripheral blood of patients with cervical cancer. A Phase I studyMol Cancer2005412216001982
  • CandelariaMGallardo-RinconDArceCA Phase II study of epigenetic therapy with hydralazine and magnesium valproate to overcome chemotherapy resistance in refractory solid tumorsAnn Oncol20071891529153817761710
  • ParulekarWREinsenhauerEANovel endpoints and design of early clinical trialsAnnal Oncol200213Suppl 4139143
  • HunsbergerSRubinsteinLVDanceyJDose escalation trial designs based on a molecularly targeted endpointStat Med200524142171218115909289
  • Segura-PachecoBPerez-CardenasETaja-ChayebLGlobal DNA hypermethylation-associated cancer chemotherapy resistance and its reversion with the demethylating agent hydralazineJ Transl Med200643216893460
  • RichonVMWebbYMergerRSecond generation hybrid polar compounds are potent inducers of transformed cell differentiationProc Natl Acad Sci U S A19969312570557088650156
  • ButlerLMAgusDBScherHLSuberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivoCancer Res200060785165517011016644
  • FinninMSDonigianJRCohenAStructures of a histone deacetylase homologue bound to the TSA and SAHA inhibitorsNature1999401674918819310490031
  • YinDOngJMHuJSuberoylanilide hydroxamic acid, a histone deacetylase inhibitor: effects on gene expression and growth of glioma cells in vitro and in vivoClin Cancer Res20071331045105217289901
  • MitsiadesCSPoulakiVMcMullanCNovel histone deacetylase inhibitors in the treatment of thyroid cancerClin Cancer Res200511103958396515897598
  • MunsterPNTroso-SandovalTRosenNThe histone deacetylase inhibitor suberoylanilide hydroxamic acid induces differentiation of human breast cancer cellsCancer Res200161238492849711731433
  • BaliPPranpatMSwabyRActivity of suberoylanilide hydroxamic acid against human breast cancer cells with amplification of her-2Clin Cancer Res200511176382638916144943
  • KomatsuNKawamataNTakeuchiSSAHA, a HDAC inhibitor, has profound anti-growth activity against non-small cell lung cancer cellsOncol Rep200615118719116328054
  • ButlerLMAgusDBScherHISuberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivoCancer Res200060185165517011016644
  • DietrichCGreenbergVLDesimoneCPSuberoylanilide hydroxamic acid potentiates paclitaxel-induced apoptosis in ovarian cancer cell linesGynecol Oncol2006116112613019875160
  • KellyWKRichonVMO’ConnorOPhase I clinical trial of histone deacetylase inhibitor: suberoylanilide hydroxamic acid administered intravenouslyClin Cancer Res.2003910 Pt 13578358814506144
  • KrugLMCurleyTSchwartzLPotential role of histone deacetylase inhibitors in mesothelioma: clinical experience with suberoylanilide hydroxamic acidClin Lung Cancer20067425726116512979
  • KellyWKO’ConnorOAKrugLMPhase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancerJ Clin Oncol200523173923393115897550
  • Garcia-ManeroGYangnHBueso-RamosCPhase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias andmyelodysplastic syndromesBlood200811131060106617962510
  • ModesittSCSillMHoffmanJSA Phase II study of vorinostat in the treatment of persistent or recurrent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group studyGynecol Oncol2008109218218618295319
  • VansteenkisteJVan CutsemEDumezHEarly Phase II trial of oral vorinostat in relapsed or refractory breast, colorectal, or nonsmall cell lung cancerInvest New Drugs200826548348818425418
  • BlumenscheinGRJrKiesMSPapadimitrakopoulouVAPhase II trial of the histone deacetylase inhibitor vorinostat (Zolinza, suberoylanilide hydroxamic acid, SAHA) in patients with recurrent and/or metastatic head and neck cancerInvest New Drugs2008261818717960324
  • LuuTHMorganRJLeongLA phase II trial of vorinostat (suberoylanilide hydroxamic acid) in metastatic breast cancer: a California Cancer Consortium studyClin Cancer Res200814217138714218981013
  • GalanisEJaeckleKAMaurerMJPhase II trial of vorinostat in recurrent glioblastoma multiforme: a north central cancer treatment group studyJ Clin Oncol200927122052205819307505
  • WoyachJAKloosRTRingelMDLack of therapeutic effect of the histone deacetylase inhibitor vorinostat in patients with metastatic radioiodine-refractory thyroid carcinomaJ Clin Endocrinol Metab200994116417018854394
  • RamalingamSSPariseRARamanathanRKPhase I and pharmacokinetic study of vorinostat, a histone deacetylase inhibitor, in combination with carboplatin and paclitaxel for advanced solid malignanciesClin Cancer Res200713123605361017510206
  • PlumbJAFinnPWWilliamsRJPharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101Mol Cancer Ther20032872172812939461
  • GimsingPBelinostat: a new broad acting antineoplastic histone deacetylase inhibitorExpert Opin Investig Drugs2009184501508
  • GianXLaRochelleWJAraGActivity of PXD101, a histone deacetylase inhibitor, in preclinical ovarian cancer studiesMol Cancer Ther2006582086209516928830
  • MonksAHoseCDPezzoliPGene expression-signature of belinostat in cell lines is specific for histone deacetylase inhibitor treatment, with a corresponding signature in xenograftsAnticancer Drugs200920868269219606018
  • WarrenKEMcCullyCDvingeHPlasma and cerebrospinal fluid pharmacokinetics of the histone deacetylase inhibitor, belinostat (PXD101), in non-human primatesCancer Chemother Pharmacol200862343343717960383
  • TumberACollinsLSPetersenKThe histone deacetylase inhibitor PXD101 synergises with 5-Xuorouracil to inhibit colon cancer cell growth in vitro and in vivoCancer Chemother Pharmacol20076027528317124594
  • SteeleNLPlumbJAVidalLA Phase 1 pharmacokinetic and pharmacodynamic study of the histone deacetylase inhibitor belinostat in patients with advanced solid tumorsClin Cancer Res200814380481018245542
  • TakadaMKataokaAToiMA close association between alteration in growth kinetics by neoadjuvant chemotherapy and survival outcome in primary breast cancerInt J Oncol200425239740515254737
  • GimsingPHansenMKnudsenLMA phase I clinical trial of the histone deacetylase inhibitor belinostat in patients with advanced hematological neoplasiaEur J Hematol2008813170176
  • RamalingamSSBelaniCPRuelCPhase II study of belinostat (PXD101), a histone deacetylase inhibitor, for second line therapy of advanced malignant pleural mesotheliomaJ Thorac Oncol2009419710119096314
  • MackayHJHirteHColganTPhase II trial of the histone deacetylase inhibitor belinostat in women with platinum resistant epithelial ovarian cancer and micropapillary (LMP) ovarian tumoursEur J Cancer20104691573157920304628
  • GolayJCuppiniLLeoniFThe histone deacetylase inhibitor ITF2357 has anti-leukemic activity in vitro and in vivo and inhibits IL-6 and VEGF production by stromal cellsLeukemia20072191892190017637810
  • VivianiSBonfanteVFasolaCPhase II study of the histonedeacetylase inhibitor ITF2357 in relapsed/refractory Hodgkin’s lymphoma patientsJ Clin Oncol.200826 abstract 8532.
  • GalliMSalmoiraghiSGolayJA Phase II multiple dose clinical trial of histone deacetylase inhibitor ITF2357 in patients with relapsed or progressive multiple myelomaAnn Hematol201089218519019633847
  • Carlo-StellaCGuidettiAVivianiSSafety and clinical activity of the histone deacetylase inhibitor givinostat in combination with meclorethamine in relapsed/refractory Hodgkin lymphoma (HL)J Clin Oncol.201028 abstract 3068.
  • QianDZKatoYShabbeerSTargeting tumor angiogenesis with histone deacetylase inhibitors: the hydroxamic acid derivative LBH589Clin Cancer Res200612263464216428510
  • PrinceHMBishtonMJJohnstoneRWPanobinostat (LBH589): a potent pan-deacetylase inhibitor with promising activity against hematologic and solid tumorsFuture Oncol20095560161219519200
  • GilesFFischerTCortesJPhase I Study of Intravenous LBH589, a novel cinnamic hydroxamic acid analogue histone deacetylase inhibitor, in patients with refractory hematologic malignanciesClin Cancer Res200612154628463516899611
  • RathkopfDWongBYRossRWA Phase I study of oral panobinostat alone and in combination with docetaxel in patients with castration-resistant prostate cancerCancer Chemother Pharmacol201066118118920217089
  • RemiszewskiSWThe discovery of NVPLAQ824: from concept to clinicCurr Med Chem200310222393240214529481
  • HurtubiseAMomparlerRLEffect of histone deacetylase inhibitor LAQ824 on antineoplastic action of 5-Aza-2-deoxycytidine (decitabine) on human breast carcinoma cellsCancer Chemother Pharmacol200658561862516783580
  • De BonoJSKristeleitRTolcherAPhase I pharmacokinetic and pharmacodynamic study of LAQ824, a hydroxamate histone deacetylase inhibitor with a heat shock protein-90 inhibitory profile, in patients with advanced solid tumorsClin Cancer Res200814206663667318927309
  • KristeleitRSTandyDAtadjaPEffects of the histone deacetylase inhibitor (HDACI) LAQ824 on histone acetylation, Hsp70 and c-Raf in peripheral blood lymphocytes from patients with advanced solid tumours enrolled in a phase I clinical trialJ Clin Oncol.200422 abstract 3023.
  • OttmannOGDeangeloDJStoneRMA Phase I, pharmacokinetic (PK) and pharmacodynamic (PD) study of a novel histone deacetylase inhibitor LAQ824 in patients with hematologic malignanciesJ Clin Oncol.200422 abstract 3024.
  • ChoyECaoYHornicekFEffect of histone deacetylase inhibitor (HDACI) PCI-24781 on chemotherapy-induced apoptosis in multidrug-resistant sarcoma cell linesJ Clin Oncol.201028 abstract 10089.
  • UndeviaSDJanischLSchilskyRLPhase I study of the safety, pharmacokinetics (PK) and pharmacodynamics (PD) of the histone deacetylase inhibitor (HDACi) PCI-24781J Clin Oncol.200826 abstract 14514.
  • JaboinJWildJHamidiHMS-27-275, an inhibitor of histone deacetylase, has marked in vitro and in vivo antitumor activity against pediatric solid tumorsCancer Res200262216108611512414635
  • LucasDMDavisMEParthunMRThe histone deacetylase inhibitor MS-275 induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia cellsLeukemia20041871207121415116122
  • RosatoRRAlmenaraJAGrantSThe histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1Cancer Res200363133637364512839953
  • LeeBIParkSHKimJWMS-275, a histone deacetylase inhibitor, selectively induces transforming growth factor beta type II receptor expression in human breast cancer cellsCancer Res200161393193411221885
  • AcharyaMRSparreboomASausvilleEAInterspecies differences in plasma protein binding of MS-275, a novel histone deacetylase inhibitorCancer Chemother Pharmacol200657327528116028097
  • RyanQCHeadleeDAcharyaMPhase I and pharmacokinetic study of MS-275, a histone deacetylase inhibitor, in patients with advanced and refractory solid tumors or lymphomaJ Clin Oncol200523173912392215851766
  • KummarSGutierrezMGardnerERPhase I trial of MS-275, a histone deacetylase inhibitor, administered weekly in refractory solid tumors and lymphoid malignanciesClin Cancer Res20071318 Pt 15411541717875771
  • GojoIJiemjitATrepelJBPhase 1 and pharmacologic study of MS-275, a histone deacetylase inhibitor, in adults with refractory and relapsed acute leukemiasBlood200710972781279017179232
  • GoreLRothenbergMLO’BryantCLA phase I and pharmacokinetic study of the oral histone deacetylase inhibitor, MS-275, in patients with refractory solid tumors and lymphomasClin Cancer Res200814144517452518579665
  • HauschildATrefzerUGarbeCMulticenter phase II trial of the histone deacetylase inhibitor pyridylmethyl-N-{4-[(2-aminophenyl)-carbamoyl]-benzyl}-carbamate in pretreated metastatic melanomaMelanoma Res200818427427818626312
  • ZhouNMoradeiORaeppelSDiscovery of N-(2-aminophenyl)-4-[(4-pyridin-3-ylpyrimidin-2-ylamino)methyl]benzamide (MGCD0103), an orally active histone deacethylase inhibitorJ Med Chem200851144072407518570366
  • FournelMBonfilsCHouYMGCD0103, a novel isotype-selective histone deacetylase inhibitor, has broad spectrum antitumor activity in vitro and in vivoMol Cancer Ther20087475976818413790
  • SiuLLPiliRDuranIPhase I study of MGCD0103 given as a three-times-per-week oral dose in patients with advanced solid tumorsJ Clin Oncol200826121940194718421048
  • Garcia-ManeroGAssoulineSCortesJPhase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemiaBlood2008112498198918495956
  • LancetJENicholsGAssoulineSA Phase I study of MGCD0103 given as a twice weekly oral dose in patients with advanced leukemias or myelodysplastic syndromes (MDS)J Clin Oncol.200725 abstract 2516.
  • BociekRGKuruvillaGJProBIsotype-selective histone deacetylase (HDAC) inhibitor MGCD0103 demonstrates clinical activity and safety in patients with relapsed/refractory classical Hodgkin Lymphoma (HL)J Clin Oncol.200826 abstract 8507.
  • MartellREYounesAAssoulineSEPhase II study of MGCD0103 in patients with relapsed follicular lymphoma (FL): Study reinitiation and update of clinical efficacy and safetyJ Clin Oncol.201028 abstract 8086.
  • BergerMRBischoffHFritschiESynthesis, toxicity, and therapeutic efficacy of 4-amino-N-(2-aminophenyl) benzamide: a new compound preferentially active in slowly growing tumorsCancer Treat Rep19856912141514243841025
  • KrakerAJMizzenCAHartlBGModulation of histone acetylation by [4-(acetylamino)-N-(2-amino-phenyl) benzamide] in HCT-8 colon carcinomaMol Cancer Ther20032440140812700284
  • PrakashSFosterBJMeyerMChronic oral administration of CI-994: a Phase 1 studyInvest New Drugs200119111111291827
  • UndeviaSDKindlerHLJanischLA Phase I study of the oral combination of CI-994, a putative histone deacetylase inhibitor, and capecitabineAnn Oncol200415111705171115520075
  • RichardsDABoehmKAWaterhouseDMGemcitabine plus CI-994 offers no advantage over gemcitabine alone in the treatment of patients with advanced pancreatic cancer: results of a phase II randomized, double-blind, placebo-controlled, multicenter studyAnn Oncol20061771096110216641168
  • UedaHMandaTMatsumotoSFR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968, III: antitumor activities on experimental tumors in miceJ Antibiot19944733153238175484
  • UedaHNakajimaHHoriYFR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. I. Taxonomy, fermentation, isolation, physicochemical and biological properties, and antitumor activityJ Antibiot19944733013107513682
  • YuXDWangSYChenGAApoptosis induced by depsipeptide FK228 coincides with inhibition of survival signaling in lung cancer cellsCancer J200713210511317476138
  • SandorVRobbinsARRobeyRFR901228 causes mitotic arrest but does not alter microtubule polymerizationAnticancer Drugs2000111144545411001385
  • SandorVSenderowiczAMertinsSP21-dependent G(1) arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228Br J Cancer200083681782510952788
  • WangRBrunnerTZhangLFungal metabolite FR901228 inhibits c-Myc and Fas ligand expressionOncogene19981712150315089794227
  • YuXGuoZSMarcuMGModulation of p53, ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228J Natl Cancer Inst200294750451311929951
  • RajgolikarGChanKKWangHCEffects of a novel antitumor depsipeptide, FR901228, on human breast cancer cellsBreast Cancer Res Treat199851129389877027
  • KitazonoMRobeyRZhanZLow concentrations of the histone deacetylase inhibitor, depsipeptide (FR901228), increase expression of the Na/I symporter and iodine accumulation in poorly differentiated thyroid carcinoma cellsJ Clin Endocrinol Metab20018673430343511443220
  • AronJLParthunMRMarcucciGDepsipeptide (FR901228) induces histone acetylation and inhibition of histone deacetylase in chronic lymphocytic leukemia cells concurrent with activation of caspase 8-mediated apoptosis and down-regulation of c-FLIP proteinBlood2003102265265812649137
  • WeiserTSGuoZSOhnmachtGASequential 5-aza-2 deoxycytidine-depsipeptide FR901228 treatment induces apoptosis preferentially in cancer cells and facilitates their recognition by cytolytic T lymphocytes specific for NY-ESO-1J Immunother200124215116111265773
  • SteinerFAHongJAFischetteMRSequential 5-Aza 2-deoxycytidine/depsipeptide FK228 treatment induces tissue factor pathway inhibitor 2 (TFPI-2) expression in cancer cellsOncogene200524142386239715735751
  • KlisovicMIMaghrabyEAParthunMRDepsipeptide (FR 901228) promotes histone acetylation, gene transcription, apoptosis and its activity is enhanced by DNA methyltransferase inhibitors in AML1/ETO-positive leukemic cellsLeukemia200317235035812592335
  • SasakawaYNaoeYSogoNMarker genes to predict sensitivityto FK228, a histone deacetylase inhibitorBiochem Pharmacol200569460361615670579
  • PiekarzRLFryeRTurnerMPhase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphomaJ Clin Oncol200927325410541719826128
  • WooSGardnerERChenXPopulation pharmacokinetics of romidepsin in patients with cutaneous T-cell lymphoma and relapsed peripheral T-cell lymphomaClin Cancer Res20091541496150319228751
  • MarshallJLRizviNKauhJA Phase I trial of depsipeptide (FR901228) in patients with advanced cancerJ Exp Ther Oncol20022632533212440223
  • SandorVBakkesSRobeyRWPhase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasmsClin Cancer Res20028371872811895901
  • ByrdJCMarcucciGParthunMRA phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemiaBlood2005105395996715466934
  • FouladiMFurmanWLChinTPhase I study of depsipeptide in pediatric patients with refractory solid tumors: a Children’s Oncology Group reportJ Clin Oncol200624223678368516877737
  • KlimekVMFircanisSMaslakPTolerability, pharmacodynamics, and pharmacokinetics studies of depsipeptide (romidepsin) in patients with acute myelogenous leukemia or advanced myelodysplastic syndromesClin Cancer Res200814382683218245545
  • StadlerWMMargolinKFerberSA Phase II study of depsipeptide in refractory metastatic renal cell cancerClin Genitourin Cancer200651576016859580
  • ShahMHBinkleyPChanKCardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumorsClin Cancer Res200612133997400316818698
  • SchrumpDSFischetteMRNguyenDMClinical and molecular responses in lung cancer patients receiving RomidepsinClin Cancer Res200814118819818172270
  • SosmanJAAronsonFRSznolMConcurrent phase I trials of intravenous interleukin 6 in solid tumor patients: reversible dose-limiting neurological toxicityClin Cancer Res19973139469815535
  • MaloneyAWorkmanPHSP90 as a new therapeutic target for cancer therapy: the story unfoldsExpert Opin Biol Ther20022132411772336
  • BantscheffMHopfCSavitskiMMChemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes.Nat Biotechnol.2011 In press.