US20070232696A1 - Valproic acid and derivatives thereof as histone deacetylase inhibitors - Google Patents

Valproic acid and derivatives thereof as histone deacetylase inhibitors Download PDF

Info

Publication number
US20070232696A1
US20070232696A1 US11/653,855 US65385507A US2007232696A1 US 20070232696 A1 US20070232696 A1 US 20070232696A1 US 65385507 A US65385507 A US 65385507A US 2007232696 A1 US2007232696 A1 US 2007232696A1
Authority
US
United States
Prior art keywords
cells
vpa
histone deacetylase
compound
induction
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/653,855
Inventor
Martin Gottlicher
Thorsten Heinzel
Bernd Groner
Peter Herrlich
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chemotherapeutisches Forschungsinstitut Georg Speyer Haus
Original Assignee
Chemotherapeutisches Forschungsinstitut Georg Speyer Haus
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chemotherapeutisches Forschungsinstitut Georg Speyer Haus filed Critical Chemotherapeutisches Forschungsinstitut Georg Speyer Haus
Priority to US11/653,855 priority Critical patent/US20070232696A1/en
Publication of US20070232696A1 publication Critical patent/US20070232696A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/02Drugs for disorders of the endocrine system of the hypothalamic hormones, e.g. TRH, GnRH, CRH, GRH, somatostatin

Definitions

  • the present invention relates to the use of the drug valproic acid and derivatives thereof as inhibitors of enzymes having histone deacetylase activity.
  • the invention also relates to the use of those compounds for the manufacture of a medicament for the treatment of diseases which are associated with hypoacetylation of histones or in which induction of hyperacetylation has a beneficial effect for example by induction of differentiation and/or apoptosis in transformed cells.
  • HAT histone acetyltransferase
  • HDAC histone deacetylase
  • Nuclear hormone receptors are ligand-dependent transcription factors that control development and homeostasis through both positive and negative control of gene expression. Defects in these regulatory processes underlie the causes of many diseases and play an important role in the development of cancer.
  • TFIIB basal transcription factors
  • ER estrogen
  • RAR retinoic acid
  • T3R thyroid hormone
  • RXR retinoid X
  • Putative coactivator proteins include SRC-1/NCoA-1, GRIP1/TIF2/NCoA-2, p/CIP/ACTR/AIB1, CBP and a variety of other factors (reviewed in Xu et al., 1999, Curr Opin Genet Dev 9, 140-147).
  • SRC proteins as well as CBP have been shown to harbor intrinsic histone acetyltransferase activity and to exist in a complex with the histone acetylase P/CAF.
  • nuclear receptors including T3R, RAR and PPAR
  • N-CoR has also been reported to interact with antagonist-occupied progesterone and estrogen receptors.
  • N-CoR and SMRT have been shown to exist in large protein complexes, which also contain mSin3 proteins and histone deacetylases.
  • the ligand-induced switch of nuclear receptors from repression to activation reflects the exchange of corepressor and coactivator complexes with antagonistic enzymatic activities.
  • the N-CoR corepressor complex not only mediates repression by nuclear receptors, but also interacts with additional transcription factors including Mad-1, BCL-6 and ETO. Many of these proteins play key roles in disorders of cell proliferation and differentiation.
  • T3R for example was originally identified on the basis of its homology with the viral oncogene v-erbA, which in contrast to the wildtype receptor does not bind ligand and functions as a constitutive repressor of transcription.
  • mutations in RARs have been associated with a number of human cancers, particularly acute promyelocytic leukemia (APL) and hepatocellular carcinoma.
  • APL acute promyelocytic leukemia
  • RAR fusion proteins resulting from chromosomal translocations involve either the promyelocytic leukemia protein (PML) or the promyelocytic zinc finger protein (PLZF).
  • PML promyelocytic leukemia protein
  • PLAZF promyelocytic zinc finger protein
  • WO 99/37150 discloses a transcription therapy for cancer comprising administering a retinoid substance and an inhibitor of histone deacetylase.
  • HDAC inhibitors Several compounds are known to be HDAC inhibitors. Butyric acid, or butyrate, was the first HDAC inhibitor to be identified. In millimolar concentrations, butyrate is not specific for HDAC, it also inhibits phosphorylation and methylation of nucleoproteins as well as DNA methylation. Its analogue phenylbutyrate acts in a similar manner. More specific are trichostatin A (TSA) and trapoxin (TPX). TPX and TSA have emerged as potent inhibitors of histone deacetylases. TSA reversibly inhibits, whereas TPX irreversibly binds to and inactivates HDAC enzymes. Unlike butyrate, nonspecific inhibition of other enzyme systems has not yet been reported for TSA or TPX. TSA and TPX, however, exhibit considerable toxicity and are poorly bioavailable. Therefore they are of limited therapeutic use.
  • TSA trichostatin A
  • TPX trapoxin
  • the invention relies on the novel finding that valproic acid (VPA; 2-n-propylpentanoic acid) is capable of inhibiting histone deacetylases.
  • VPA valproic acid
  • Valproic acid is a known drug with multiple biological activities which depend on different molecular mechanisms of action.
  • VPA derivatives allowed to determine that the different activities are mediated by different molecular mechanisms of action. Teratogenicity and antiepileptic activity follow different modes of action because compounds could be isolated which are either preferentially teratogenic or preferentially antiepileptic (Nau et al., 1991, Pharmacol. Toxicol. 69, 310-321). Activation of PPAR8 was found to be strictly correlated with teratogenicity (Lampen et al., 1999, Toxicol. Appl. Pharmacol. 160, 238-249) suggesting that, both, PPAR ⁇ activation and teratogenicity require the same molecular activity of VPA.
  • PPAR ⁇ activation is caused by the HDAC inhibitory activity of VPA and its derivatives. Furthermore it is shown that the established HDAC inhibitor TSA activates PPAR ⁇ and induces the same type of F9 cell differentiation as VPA. From these results we conclude that not only activation of PPAR ⁇ but also induction of F9 cell differentiation and teratogenicity of VPA or VPA derivatives are most likely caused by HDAC inhibition.
  • the present invention is based on the finding that VPA and the derivatives described in this application are inhibitors of histone deacteylases.
  • the finding of this novel mechanism of action of VPA and compounds derived thereof, i.e. the inhibition of enzymes with histone deacetylase activity led us to the proposition that VPA due to its HDAC-inhibitory activity should be useful to induce differentiation and/or apoptosis in a wide variety of cancer cells for two reasons: (1) these enzymes are present in all cells and (2) pilot studies with model compounds such as butyrate or TSA which are different from those described in this invention had shown that HDAC inhibitors induce differentiation in a wide variety of cells.
  • the activity to induce differentiation and/or apoptosis in a wide variety of transformed cells is a much more complex biological activity than only the inhibition of proliferation. In the latter case it would not be evident, why only the proliferation of transformed (tumor) but not of normal cells should be inhibited.
  • the induction of apoptosis, differentiation or more specifically re-differentiation in dedifferentiated tumor cells provides a rationale why the compounds of this invention have beneficial effects in a wide variety of tumors by induction of differentiation and/or apoptosis. This proposition was confirmed in a wide variety of tumor cells (see examples).
  • Antiepileptic and sedating activities follow different structure activity relationships and thus obviously depend on a primary VPA activity distinct from HDAC inhibition.
  • U.S. Pat. No. 5,672,746 and WO 96/06821 disclose the use of VPA and derivatives thereof for the treatment of neuredegenerative and neuroproliferative disorders.
  • VPA ⁇ -carbon branched carboxylic acids as described by formula I wherein R 1 and R 2 independently are a linear or branched, saturated or unsaturated aliphatic C 2-25 , preferably C 3-25 hydrocarbon chain which optionally comprises one or several heteroatoms and which may be substituted, R 3 is hydroxyl, halogen, alkoxy or an optionally alkylated amino group.
  • R 1 and R 2 residues give rise to chiral compounds.
  • one of the stereoisomers has a stronger teratogenic effect than the other one (Nau et al., 1991, Pharmacol. Toxicol. 69, 310-321) and the more teratogenic isomer more efficiently activates PPAR ⁇ (Lampen et al, 1999). Therefore, this isomer can be expected to inhibit HDACs more strongly (this invention).
  • the present invention encompasses the racemic mixtures of the respective compounds, the less active isomers, and in particular the more active isomers.
  • the hydrocarbon chains R 1 and R 2 may comprise one or several heteroatoms (e.g. O, N, S) replacing carbon atoms in the hydrocarbon chain. This is due to the fact that structures very similar to that of carbon groups may be adopted by heteroatom groups when the heteroatoms have the same type of hybridization as a corresponding carbon group.
  • heteroatoms e.g. O, N, S
  • R 1 and R 2 may be substituted. Possible substituents include hydroxyl, amino, carboxylic and alkoxy groups as well as aryl and heterocyclic groups.
  • R 1 and R 2 independently comprise 2 to 10, more preferably 3 to 10 or 5 to 10 carbon atoms. It is also preferred that R 1 and R 2 independently are saturated or comprise one double bond or one triple bond.
  • one of the side chains (R 1 ) may preferably contain sp 1 hybridized carbon atoms in position 2 and 3 or heteroatoms which generate a similar structure. This side chain should comprise 3 carbon or heteroatoms but longer chains may also generate HDAC-inhibiting molecules. Also inclusion of aromatic rings or heteroatoms in R 2 is considered to generate compounds with HDAC inhibitory activity because the catalytic site of the HDAC protein apparently accommodates a wide variety of binding molecules.
  • HDAC inhibitors also compounds which have previously been disregarded as suitable antiepileptic agents are considered as HDAC inhibitors under this invention.
  • compounds having a propinyl residue as R 1 and residues of 7 or more carbons as R 2 are considered (Lampen et al, 1999).
  • the group “COR 3 ” is a carboxylic group. Also derivatization of the carboxylic group has to be considered for generating compounds with potential HDAC inhibitory activity. Such derivatives may be halides (e.g. chlorides), esters or amides.
  • R 3 is alkoxy
  • the alkoxy group comprises 1 to 25, preferably 1-10 carbon atoms.
  • R 3 is a mono- or di-alkylated amino group
  • the alkyl substituents comprise 1 to 25, preferably 1-10 carbon atoms. An unsubstituted amino group, however, is preferred.
  • compositions of formula I can be used.
  • substances can be used which are metabolized to a compound as defined in formula I in the human organism or which lead to the release of a compound as defined in formula I for example by ester hydrolysis.
  • the invention concerns the use of an ⁇ -carbon branched carboxylic acid as described by formula I or of a pharmaceutically acceptable salt thereof as an inhibitor of an enzyme having histone deacetylase activity
  • R 1 is a linear or branched, saturated or unsaturated, aliphatic C 5-25 hydrocarbon chain
  • R 2 independently is a linear or branched, saturated or unsaturated, aliphatic C 2-25 hydrocarbon chain, but not —CH 2 —CH ⁇ CH2, —CH 2 —C ⁇ CH or —CH 2 —CH 2 —CH 3
  • R 1 and R 2 are optionally substituted with hydroxyl, amino, carboxylic, alkoxy, aryl and/or heterocyclic groups
  • R 3 is hydroxyl.
  • the invention concerns the use of an ⁇ -carbon branched carboxylic acid as described by formula I or of a pharmaceutically acceptable salt thereof as an inhibitor of an enzyme having histone deacetylase activity
  • R 1 is a linear or branched, saturated or unsaturated, aliphatic C 3-25 hydrocarbon chain
  • R 2 independently is a linear or branched, saturated or unsaturated, aliphatic C 2-25 hydrocarbon chain
  • R 1 or R 2 comprise one or several heteroatoms (e.g. O, N, S). replacing carbon atoms in the hydrocarbon chain
  • R 1 and R 2 are optionally substituted with hydroxyl, amino, carboxylic, alkoxy, aryl and/or heterocyclic groups
  • R 3 is hydroxyl.
  • R 1 and R 2 do not comprise an ester group (—CO—O—).
  • the atom of R 1 which is next to the ⁇ -carbon of the carboxylic acid (derivative) of formula I and covalently linked to said ⁇ -carbon may be a carbon atom.
  • the atom of R 2 which is next to the ⁇ -carbon of the carboxylic acid (derivative) of formula I and covalently linked to said ⁇ -carbon may be a carbon atom.
  • R 1 and R 2 may be hydrocarbon chains comprising no heteroatoms O, N or S.
  • the compounds which are most preferably used according to the present invention are VPA, S-4-yn VPA, 2-EHXA (2-Ethyl-hexanoic acid).
  • the compounds are useful for inhibiting mammalian (for use of cell lines in in vitro assays and animal models systems) and in particular human (in vivo and in vitro) histone deacetylases HDAC 1-3 (class I) and HDAC 4-8 (class II).
  • the compounds may be used to induce the differentiation and/or apoptosis of cells such as undifferentiated tumour cells. Presumably, this reflects a general mechanism, as differentiation can be induced in F9 teratocarcinoma cells, MT 450 breast cancer cells, HT-29 colon carcinoma cells and several leukemia cell lines as assessed by morphological alterations and specific marker gene or protein expression. Furthermore, for example MT450 cells can be induced to undergo apoptosis (see example 6).
  • the invention also concerns the use of a compound of formula I for the induction of differentiation and/or apoptosis of transformed cells.
  • Another aspect of the present invention is the use of a compound of formula I for the manufacture of a medicament for the treatment of a disease which is associated with gene-specific hypoacetylation of histones.
  • a disease which is associated with gene-specific hypoacetylation of histones.
  • diseases which are associated with aberrant repression of specific genes which correlates with a local level of histone acetylation below the regular level.
  • Yet another aspect of the invention is the use of a compound of formula I for the manufacture of a medicament for the treatment of a disease in which the induction of hyperacetylation of histones has a beneficial effect resulting in differentiation and/or apoptosis of a patient's tumor cells and thus causing a clinical improvement of the patient's condition.
  • diseases are skin cancer, estrogen receptor-dependent and independent breast cancer, ovarian cancer, prostate cancer, renal cancer, colon and colorectal cancer, pancreatic cancer, head and neck cancer, small cell and non-small cell lung carcinoma.
  • the induction of hyperacetylation may also be beneficial by reverting inappropriate gene expression in diseases based on aberrant recruitment of histone deacetylase activity such as thyroid resistance syndrome.
  • the compounds and salts thereof can be formulated as pharmaceutical compositions (e.g. powders, granules, tablets, pills, capsules, injections, solutions, foams, enemas and the like) comprising at least one such compound alone or in admixture with pharmaceutically acceptable carriers, excipients and/or diluents.
  • the pharmaceutical compositions can be formulated in accordance with a conventional method. Specific dose levels for any particular patient will be employed depending upon a variety of factors including the activity of specific compounds employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and the severity of the particular disease undergoing therapy.
  • the active ingredient will preferably be administered in an appropriate amount, for example, selected from the range of about 10 mg/kg to 100 mg/kg body weight a day orally or intravenously.
  • the dose levels are not specifically restricted as long as serum levels of 0.05 mM to 3 mM, preferably of about 0.4 mM to 1.2 mM are achieved.
  • Another aspect of the invention is a method for the identification of substances having histone deacetylase inhibitory activity which comprises providing a derivative of valproic acid, determining its histone deacetylase inhibitory activity, and selecting the substance if the substance has histone deacetylase inhibitory activity.
  • Valproic acid can serve as a lead substance for the identification of other compounds exhibiting histone deacetylase inhibitory activity.
  • compounds may be selected which show increased HDAC inhibitory activity at lower doses and serum levels and have decreased effects on the central nervous system such as sedating activity.
  • Another parameter that may be optimised is the appearance of the hepatotoxic effect. Compounds may be selected which show a reduced liver toxicity.
  • the derivatives may be provided by synthesising compounds which comprise additional and/or modified substituents.
  • the HDAC inhibitory activity may be determined by a state-of-the-art technology such as transcription repression assay, a Western Blot which detects acetylation of histone H3 and/or histone H4, or by an enzymatic assay.
  • the transcriptional assay for repressor activity exploits activation and derepression of a Gal4-dependent reporter gene.
  • This assay can be performed either by transient transfection of mammalian cell lines (e.g. HeLa, 293T, CV-1) or with specifically constructed permanent cell lines. Transcription factors such as thyroid hormone receptor, PPAR ⁇ , retinoic acid receptor, N-CoR and AML/ETO repress transcription when they bind to a promoter containing UAS elements as fusion proteins with the heterologous DNA-binding domain of the yeast Gal4 protein.
  • the reporter gene In the absence of the Gal4-fusion protein the reporter gene has a high basal transcriptional activity due to the presence of binding sites for other transcription factors in the thymidine kinase promoter.
  • the Gal4 fusion proteins repress this activity by up to 140-fold. HDAC inhibitors induce relief of this repression which can be detected as an increase in reporter gene activity (e.g. by luciferase assay).
  • Histone deacetylase inhibitors induce the accumulation of N-terminally hyperacetylated histones H3 and H4. These acetylated histones can be detected by Western blot analysis of whole cell extracts or of histone preparations from histone deacetylase inhibitor-treated cells using antibodies specific for the acetylated N-terminal lysine residues of histones H3 and H4.
  • the enzymatic assay for HDAC activity records the release of 3 H-labeled acetic acid from hyperacetylated substrates.
  • Sources of HDAC activity can be co-immunoprecipitates with antibodies directed against N-CoR (or other repressors known to recruit HDACs) or crude cell extracts containing histone deacetylases (e.g. HeLa, 293T, F 9).
  • Substrates may be either chemically 3 H-acetylated peptides corresponding to the. N-termini of histones H3 or H4 or histone proteins isolated from metabolically labelled cells which were treated with HDAC inhibitors. After extraction with ethyl acetate the release of 3 H-labeled acetic acid is detected by liquid scintillation counting.
  • Yet another aspect of the invention is a method for profiling of the HDAC isoenzyme specificity of a compound as defined in formula I wherein the binding of the compound to HDAC and/or the competition for binding to HDAC is measured.
  • the method may comprise the following steps: HDACs are either immune precipitated with HDAC isoform specific antibodies, with antibodies directed against corepressor complexes, or with specific antibodies against recombinant HDACs overexpressed in transgenic cells.
  • the method may further involve determination of individual HDACs present in these immune precipitates by Western blot analysis.
  • Radiolabeled VPA or compounds according to formula I are bound to the immune precipitates. The amount of bound compound is determined through measurement of bound radioactivity after appropriate washing steps.
  • a variation of this aspect involves binding of one labeled HDAC inhibitor such as VPA, TSA or trapoxin and competition of binding by a compound according to formula I.
  • Another variation of the method involves the use of alternate labeling and/or detection procedures.
  • VPA or derivatives thereof to define genes which are induced by said compounds in cells such as primary human or rodent cells, leukemic cells, other cancer cells or tumor cell lines.
  • Methods to define such genes that are induced by VPA include established technologies for screening large arrays of cDNAs, expressed sequence tags or so-called unigene collections.
  • subtractive hybridization techniques is suitable to define genes which are induced by VPA or derivatives thereof. The use of these methods to identify potential targets for drug development downstream of HDAC-inhibition, and furthermore the use of these methods to define diagnostic means in order to facilitate the therapeutic treatment of patients with suitable compounds is part of this invention.
  • VPA VPA
  • derivatives thereof for defining target genes which are selectively regulated or not regulated by VPA, particularly also in comparison to other HDAC inhibitors like trichostatin A.
  • the present invention also concerns a diagnostic method to identify tumors comprising the step of testing whether a tumor is responsive to treatment with compounds as defined by formula I.
  • the method preferably comprises the method for the identification of genes induced by VPA or a derivative thereof described supra.
  • the diagnostic method comprises the use of nucleic acid technology, preferably of hybridization or polymerase chain reaction for detection. Other types of nucleic acid technology, however, may be employed.
  • the method comprises the use of specific antibodies against differentially regulated proteins for detection. For this purpose proteins encoded by the genes showing deregulation of their expression upon VPA treatment would be expressed e.g. in recombinant expression systems and antibodies directed against these proteins would be generated. Subsequently such antibodies could be used (or patterns of antibodies) to characterize the status of a tumor or tumor cells for diagnostic and/or prognostic reasons.
  • VPA and derivatives thereof are potent HDAC inhibitors.
  • the HDAC inhibitors known so far are either nonspecific like butyrate, or toxic or poorly bioavailable in the whole organism like TSA and TPX.
  • VPA has the advantage that it is already an approved drug and has been used over decades for the treatment of epilepsy in human. Thus, a vast amount of data concerning pharmaceutical acceptability and the lack of serious side effects are available.
  • VPA should be a suitable drug for the use in humans to induce differentiation and/or apoptosis in transformed cells and by that to exert beneficial effects in a wide variety of patients suffering from cancer.
  • FIG. 1 describes the histone deacetylase inhibitor-like activation of PPAR ⁇ by VPA (example 1).
  • FIG. 2 shows that VPA activates several transcription factors in addition to PPAR ⁇ (example 2).
  • FIG. 3 shows VPA-induced accumulation of hyperacetylated histones H3 and H4 (example 3).
  • FIG. 4 shows the biochemical analysis of histone deacetylase activity in the absence or presence of VPA (example 4).
  • FIG. 5 shows indicators of VPA induced differentiation in HT-29 colonic carcinoma cells, F9-teratocarcinoma cells, and RenCa renal carcinoma cells.
  • FIG. 6 shows induction of apoptosis in MT450 breast cancer cells (example 6).
  • FIG. 7 shows the loss of viable cells upon treatment with valproic acid.
  • Renca-lacZ, Renca-lacZ/EGFR, Renca-lacZ/EGFRvIII and Renca-lacZ/ErbB2 renal carcinoma cells (A) or SKOV3 ovarian carcinoma cells, SKBR3, MCF7, MDA-MB453 and MDA-MB468 breast carcinoma cells, and A431 squamous cell carcinoma cells (B) were incubated with the indicated concentrations of valproic acid (VPA).
  • the relative number of viable cells was determined using the enzymatic MTT assay, measuring cellular metabolic activity, as described in Example 7. Each point represents the mean of a set of data determined in triplicate (example 7).
  • FIG. 8 shows the reduction in cellular biomass after treatment of cell cultures with VPA (example 8)
  • a reporter gene cell line for activation of the PPAR ⁇ ligand binding domain was constructed in CHO cells.
  • a subclone of CHO cells was used which contained a transgenic reporter gene expressing a secreted form of the human placental alkaline phosphatase under control of the glucocorticoid receptor-dependent LTR-promoter of the mouse mammary tumor virus (Göttmaschine et al. (1992) Proc. Natl. Acad. Sci. USA 89, pp. 4653-4657).
  • a hybrid receptor comprising the amino-terminus of the glucocorticoid receptor fused to the ligand binding domain of PPAR ⁇ was expressed in these cells essentially as described for the expression of the corresponding hybrid of PPAR ⁇ (Gött Anlagen et al., 1992, ibd.).
  • the ligand binding domain of PPAR ⁇ was used starting at amino acid 138 as deduced from the sequence published by Amri et al. (J. Biol. Chem. 270 (1995) pp. 2367-2371).
  • Activation of the PPAR ⁇ ligand binding domain in these cells induces expression of the alkaline phosphatase reporter gene which is detectable by an enzymatic assay from the cell culture supernatant.
  • Similar cells expressing the full length glucocorticoid receptor served as negative controls for specificity of receptor activation.
  • the PPAR ⁇ hyrid receptor expressing cells were seeded at 20% confluency into 24-well culture dishes and treated for 40 h with the PPAR ⁇ ligand carbocyclic prostaglandin I 2 (PGI, 5 ⁇ M), VPA (1 or 2 mM), or the histone deacetylase inhibitors sodium butyrate (0, 2-5 mM) and trichostatin A (TSA, 300 nM). Reporter gene activity was monitored by an enzymatic assay (alkaline phosphatase). Values except for butyrate are means ⁇ S.D.
  • FIG. 1 The highly synergistic activation of the reporter gene by VPA together with the PPAR ⁇ ligand cPGI (P+V) which is similar to the synergistic activation by Trichostatin A together with cPGI (P+T), and the lack of synergism with trichostatin (T+V) or butyrate (not shown) indicate that VPA does not act like a bona fide ligand to PPAR ⁇ . VPA rather affects PPAR ⁇ activity by a mechanism which lies in the same sequence of events by which also the inhibitors of corepressor-associated histone deacetylases induce transcriptional activity of PPAR ⁇ .
  • TR thyroid hormone receptor
  • PPAR ⁇ peroxisome proliferator activated receptor ⁇
  • RAR retinoic acid receptor
  • AML/ETO fusion protein repress transcription when they bind to a promoter containing UAS sites (Gal4 response element) as fusion proteins with the heterologous DNA binding domain of the yeast Gal4 protein.
  • a promoter containing UAS sites Gal4 response element
  • Gal4 fusion protein a luciferase reporter gene is transcribed at a high basal level due to the presence of binding sites for other transcription factors in the thymidine kinase (TK) promoter.
  • TK thymidine kinase
  • Hela cells were transfected with a UAS TK luciferase reporter plasmid (Heinzel et al., 1997, Nature 387, pp 43-48) and expression plasmids for the indicated Gal4 fusion proteins using the calcium phosphate precipitate method. After 24 h the medium was changed and cells were incubated with histone deacetylase inhibitors for a further 24 h. Transcriptional repression is measured as luciferase activity relative to the baseline of cells transfected with an expression plasmid for the Gal4 DNA binding domain alone ( FIG. 2 ). The Gal4 fusion proteins repress this baseline activity by up to 140 fold.
  • VPA at a concentration of 1 mM induces relief of this- repression which is indicated as an increase in reporter gene activity.
  • a relief of repression is also found after treatment with established histone deacetylase inhibitors (10 nM Trapoxin, 100 nM TSA) as well as after partial activation of TR and PPAR ⁇ by their respective ligands.
  • a combination of ligand and HDAC inhibitors (including VPA) results in a synergistic effect, indicating that different molecular mechanisms are involved.
  • FIG. 2 shows that VPA affects the activity of several distinct transcription factors and cofactors. This finding suggests that VPA acts on a common factor in the regulation of gene expression such as corepressor-associated histone deacetylases rather than on individual transcription factors or receptors (e.g. as a ligand).
  • FIG. 3 shows the results of such an analysis from a representative experiment. In this experiment both the time course of VPA-induced hyperacetylation (A) and the required VPA concentration (B) were determined.
  • Acetylated histones H3 and histone H4 were detected by Western blot analysis using commercially available antibodies (Upstate Biotechnology) specific for the acetylated forms of histones (Ac-H3, Cat-Nr.: 06-599; Ac-H4, Cat-Nr.: 06-598). Equal loading of the lanes was confirmed by staining a part of the polyacrylamide gel by Coomassie blue.
  • VPA For determination of the required VPA dose F9 cells were cultured in 6-well culture dishes for 8 h prior to addition of VPA at the indicated concentrations. Whole cell extracts were prepared 16 h after treatment as described above. Analysis for acetylated histones H3 and H4 was performed as described in (A). VPA concentrations in the range of blood serum levels reached during therapeutic use of VPA as antiepileptic agent in humans induce hyperactylation of histones H3 and H4. At serum levels only slightly exceeding those intended for antiepileptic therapy VPA induces histone hyperacetylation as efficiently as sodium butyrate or trichostatin A used at concentrations which are expected to have a maximum effect. This experiment indicates that VPA or a metabolite formed in F9 cells inhibits histone deacetylase activity.
  • Immune precipitates from whole cell extracts using antibodies against the corepressor N-CoR or mSin3 contain histone deacetylase activity. This enzymatic activity is measured by incubating the immune precipitates with radioactively acetylated histone substrates from cells in which histones have been hyperacetylated in the presence of 3 H-acetate. The release of 3 H-acetate is detected as a measure of enzymatic activity by extraction with ethyl acetate and subsequent liquid scintillation counting ( FIG. 4 ). Addition of the histone deacetylase inhibitor trichostatin A (TSA, 10 ⁇ 7 M) to the reaction in vitro severely inhibits the enzymatic activity.
  • TSA histone deacetylase inhibitor
  • VPA from left to right 0.2 mM, 1 mM, 5 mM
  • EHXA ethyl hexanoic acid
  • R-4-yn VPA from left to right 0.2 mM, 1 mM, 5 mM
  • S-4-yn VPA from left to right 0.2 mM, 1 mM, 5 mM
  • Histone deacetylase inhibitors and VPA in particular induce differentiation of dedifferentiated tumorigenic cells.
  • Cell differentiation is associated with cell cycle arrest, morphological alterations and the appearance of expression of markers of the differentiated phenotype. Morphological alterations where determined by microscopic evaluation of F9 and HT-29 cells.
  • F9 and HT-29 cells were cultured for 36 h in the absence or the presence of 1 mM VPA in 96-well culture dishes.
  • F9 teratocarcinoma cells were treated for 48 h with VPA (1 mM), sodium butyrate (B, 1 mM) and trichostation A (TSA, 30 nM). Differentiation was followed by morphological criteria, a reduced rate in the increase of cell number (e.g. cell cycle arrest, data not shown), the drop of 3 H-thymidine incorporation by 48% during a 12 h pulse labeling period (see above) and the appearance of nuclear AP-2 protein ( FIG. 5B ) as a specific marker of histone deacetylase inhibitor-induced differentiation of F9 cells.
  • VPA 1 mM
  • B sodium butyrate
  • TSA trichostation A
  • Nuclear AP-2 protein was detected in nuclear extract which had been prepared by mild detergent lysis (25 mM Tris, pH 7.5; 1 mM EDTA, 0.05*NP40) of treated or non-treated F9 cells, recovery of nuclei by centrifugation (3000 ⁇ g, 5 min) and lysis of nuclei in sample buffer for denaturing SDS gel electrophoresis. Nuclear extracts were separated on a 9% SDS polyacrylamide gel.
  • AP-2 protein was detected by Western blot analysis using a rabbit polyclonal antibody (Santa Cruz, Cat.-No.: SC-184) at a dilution of 1/1000 in Tris buffered saline containing 3W non-fat dry milk and 0.05% Tween 20.
  • VPA and trichostatin A induce nuclear AP-2 protein whereas the activity of butyrate at the chosen concentration is weak. Since appearance of AP-2 is a delayed effect which is only detectable after 36 to 40 h of VPA treatment the weak activity of butyrate may be caused by efficient metabolism of the compound. Nevertheless, VPA induces differentiation of the epithelial F9 cell line in a way indistinguishable from differentiation by other histone deacetylase inhibitors.
  • RenCa-LacZ cells were cultured for 36 h either in the absence or the presence of 1 mM VPA. Morphological alterations were observed by phase contrast microscopy and micrographs of representative fields were taken ( FIG. 5C )
  • MT-450 cells were cultured for 72 h in the absence or presence of 1 mM VPA. Apoptotic cells were detected by flow cytometric analysis after staining of cell surface exposed phosphatidylserine with FITC-conjugated annexin V (Becton Dickinson) according to suppliers instructions. Dead cells were excluded by propidium iodide staining. Cells positive for annexin V and negative for propidium iodide uptake (lower right quadrant in FIG. 6 ) were judged and counted as apoptotic cells.
  • Human MDA-MB468, MDA-MB453 and SKBR3 breast carcinoma cells, A431 squamous cell carcinoma cells, and SKOV3 ovarian carcinoma cells were maintained in Dulbecco's modified Eagle's medium (DMEM, BioWhittaker, Verviers, Belgium) supplemented with 10% heat inactivated fetal bovine serum (FBS), 2 mM L-glutamine, 100 units/ml penicillin, and 100 ⁇ g/ml streptomycin.
  • Human MCF7 breast carcinoma cells were grown in RPMI medium supplemented as described above.
  • Renal cell carcinoma (Renca) cells stably transfected with plasmid pZeoSV2/lacZ encoding E. coli ⁇ -galactosidase (Renca-lacZ cells) were grown in RPMI-1640 medium supplemented with 8% FBS, 2 mM L-glutamine, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin, 0.25 mg/ml Zeocin.
  • Renca-lacZ cells cotransfected with plasmids pSV2ErbB2N and pSV2neo encoding c-erbB2 and neomycin resistance (Renca-lacZ/ErbB2) (Maurer-Gebhard et al., Cancer Res.
  • Tumor cells were seeded in 96 well plates at a density of 1 ⁇ 10 4 cells/well in normal growth medium. Valproic acid was added at final concentrations of 1 or 3 mM to triplicate samples and the cells were incubated for 40 h (Renca-lacZ, Renca-lacZ/ErbB2, Renca-lacZ/EGFR, Renca-lacZ/EGFRvIII, SKBR3 and SKOV3 cells) or 70 h (A431, MCF7, MDA-MB453 and MDA-MB468 cells). Control cells were grown in the absence of valproic acid.
  • valproic acid reduces the viability of breast carcinoma cells, ovarian carcinoma cells, squamous cell carcinoma cells, renal carcinoma cells, and renal carcinoma cells expressing at high levels the ErbB2 or EGF receptor proto-oncogenes, or the oncogenically activated EGF receptor variant EGFRVIII, in a concentration dependent manner.
  • valproic acid potently reduces the number and/or viability of a wide variety of tumor cells derived from solid tumors of epithelial origin. The loss of viability could indicate a reduction in cell number upon induction of cellular differentiation and/or induction of cell death.
  • the observation of changes of cellular morphology suggest that cellular differentiation is at least responsible for a part of the effect. This induction of differentiation and/or induction of cell death suggest that valproic acid and derivatives thereof could be used for the therapy of such tumors.
  • VPA induces differentiation and/or cell death in a series of human cancer cells and reduces the total cellular biomass of human cancer cell cultures. The reduction in biomass could indicate cell loss due to cell death and/or differentiation associated cell cycle arrest. Quantitative parameters, e.g. the loss of biomass, was determined in 30 human cancer cell lines ( FIG. 8 e ) and twelve examples of dose-response curves are shown, e.g.
  • BT-549 breast cancer cells (1), estrogen dependent ZR-75 breast cancer cells (2), DMS-114 small cell lung cancer cells (3), NCI-H226 non-small cell lung cancer cells (4), SK-MEL-28 skin cancer cells (5), OVCAR-3 ovarian cancer cells (6), HUP-T3 pancreatic cancer cells (7), DU-145 prostate cancer cells (8), DETROIT-562 head and neck cancer cells, LS-174 colon cancer cells (10), A-172 brain cancer cells (11) and HL-60 leukemia cells (12) ( FIG. 8 a - d ). All cells were evaluated for morphological signs of cell death and/or differentiation.
  • FIG. 8 a - c Cells in panels 1 to 9 ( FIG. 8 a - c ) were seeded in 96 well culture dishes at densities between 3000 and 8000 per well. After recovery of 24 hours cells were cultured for 48 hours in the absence or presence of the indicated concentrations of VPA. Cultures were fixed with TCA by layering 50 ⁇ l of cold 50*TCA on top of the growth medium in each well to produce a final TCA concentration of 10%. After 1 hour of incubation at 4° C. the cells were washed five times with tap water and air dried.
  • T 0 the time at which the drug was added
  • the graphs in FIG. 8 a - d show means ⁇ S.D. from sixfold determinations.
  • FIG. 8 e summarizes the reduction of total cellular biomass of various human cancer cells by treatment with 1 mM VPA. This reduction could indicate differentiation associated cell cycle arrest and/or induction of cell death. Cells were VPA treated for 48 hours. The inhibition was calculated from six response tests performed in parallel and reductions of cellular biomass are given in percent of untreated cells with standard deviations.

Landscapes

  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present invention relates to the use of the drug valproic acid and derivatives thereof as inhibitors of enzymes having histone deacetylase activity. The invention also relates to the use of those compounds for the manufacture of a medicament for the treatment of diseases which are associated with hypoacetylation of histones or in which induction of hyperacetylation has a beneficial effect for example by induction of differentiation and/or apoptosis in transformed cells.

Description

  • The present invention relates to the use of the drug valproic acid and derivatives thereof as inhibitors of enzymes having histone deacetylase activity. The invention also relates to the use of those compounds for the manufacture of a medicament for the treatment of diseases which are associated with hypoacetylation of histones or in which induction of hyperacetylation has a beneficial effect for example by induction of differentiation and/or apoptosis in transformed cells.
  • Local remodelling of chromatin is a key step in the transcriptional activation of genes. Dynamic changes in the nucleosomal packaging of DNA must occur to allow transcriptional proteins contact with the DNA template. One of the most important mechanisms contributing to chromatin remodelling is the posttranslational modification of histones by acetylation. Change in electrostatic attraction for DNA and steric hindrance introduced by the hydrophobic acetyl group leads to destabilisation of the interaction of histones with DNA. As a result, acetylation of histones disrupts nucleosomes and allows the DNA to become accessible to the transcriptional machinery. Removal of the acetyl groups allows the histones to bind more tightly to DNA and to adjacent nucleosomes and thus maintain a transcriptionally repressed chromatin structure. Acetylation is mediated by a series of enzymes with histone acetyltransferase (HAT) activity. Conversely, acetyl groups are removed by specific histone deacetylase (HDAC) enzymes. Disruption of these mechanisms gives rise to transcriptional misregulation and may lead to leukemic transformation.
  • Nuclear hormone receptors are ligand-dependent transcription factors that control development and homeostasis through both positive and negative control of gene expression. Defects in these regulatory processes underlie the causes of many diseases and play an important role in the development of cancer.
  • Several members of the nuclear receptor superfamily have been reported to interact with basal transcription factors, including TFIIB. However, numerous lines of evidence indicate that nuclear receptors must interact with additional factors to mediate both activation and repression of target genes. A number of cofactors that associate with the ligand binding domains of estrogen (ER), retinoic acid (RAR), thyroid hormone (T3R), retinoid X (RXR), and other nuclear receptors have recently been identified. Putative coactivator proteins include SRC-1/NCoA-1, GRIP1/TIF2/NCoA-2, p/CIP/ACTR/AIB1, CBP and a variety of other factors (reviewed in Xu et al., 1999, Curr Opin Genet Dev 9, 140-147). Interestingly, SRC proteins as well as CBP have been shown to harbor intrinsic histone acetyltransferase activity and to exist in a complex with the histone acetylase P/CAF.
  • Many nuclear receptors, including T3R, RAR and PPAR, can interact with the corepressors N-CoR and SMRT in the absence of ligand and thereby inhibit transcription. Furthermore, N-CoR has also been reported to interact with antagonist-occupied progesterone and estrogen receptors. N-CoR and SMRT have been shown to exist in large protein complexes, which also contain mSin3 proteins and histone deacetylases. Thus, the ligand-induced switch of nuclear receptors from repression to activation reflects the exchange of corepressor and coactivator complexes with antagonistic enzymatic activities.
  • The N-CoR corepressor complex not only mediates repression by nuclear receptors, but also interacts with additional transcription factors including Mad-1, BCL-6 and ETO. Many of these proteins play key roles in disorders of cell proliferation and differentiation. T3R for example was originally identified on the basis of its homology with the viral oncogene v-erbA, which in contrast to the wildtype receptor does not bind ligand and functions as a constitutive repressor of transcription. Furthermore, mutations in RARs have been associated with a number of human cancers, particularly acute promyelocytic leukemia (APL) and hepatocellular carcinoma. In APL patients RAR fusion proteins resulting from chromosomal translocations involve either the promyelocytic leukemia protein (PML) or the promyelocytic zinc finger protein (PLZF). Although both fusion proteins can interact with components of the corepressor complex, the addition of retinoic acid dismisses the corepressor complex from PML-RAR, whereas PLZF-RAR interacts constitutively. These findings provide an explanation why PML-RAR APL patients achieve complete remission following retinoic acid treatment whereas PLZF-RAR APL patients respond very poorly. The hypothesis that corepressor-mediated aberrant repression may be causal for pathogenesis in APL is supported by the finding that trichostatin A, which inhibits histone deacetylase (HDAC) function is capable of overcoming the differentiation block in cells containing the PLZF-RAR fusion protein. Furthermore, a PML-RAR patient who had experienced multiple relapses after treatment with retinoic acid has recently been treated with the HDAC inhibitor phenylbutyrate, resulting in complete remission of the leukemia (Warrell et al., 1998, J. Natl. Cancer Inst. 90, 1621-1625).
  • Additional evidence that histone acetylation plays a role in cancer comes from studies on the AML1-ETO oncoprotein and on chromosomal rearrangements involving the MLL locus (Redner et al., 1999, Blood 94, 417-428).
  • WO 99/37150 discloses a transcription therapy for cancer comprising administering a retinoid substance and an inhibitor of histone deacetylase.
  • Several compounds are known to be HDAC inhibitors. Butyric acid, or butyrate, was the first HDAC inhibitor to be identified. In millimolar concentrations, butyrate is not specific for HDAC, it also inhibits phosphorylation and methylation of nucleoproteins as well as DNA methylation. Its analogue phenylbutyrate acts in a similar manner. More specific are trichostatin A (TSA) and trapoxin (TPX). TPX and TSA have emerged as potent inhibitors of histone deacetylases. TSA reversibly inhibits, whereas TPX irreversibly binds to and inactivates HDAC enzymes. Unlike butyrate, nonspecific inhibition of other enzyme systems has not yet been reported for TSA or TPX. TSA and TPX, however, exhibit considerable toxicity and are poorly bioavailable. Therefore they are of limited therapeutic use.
  • It is one object of the present invention to provide substances which can induce differentiation and/or apoptosis in a wide variety of transformed cells and therefore can be useful in the treatment of cancer.
  • The invention relies on the novel finding that valproic acid (VPA; 2-n-propylpentanoic acid) is capable of inhibiting histone deacetylases.
  • Valproic acid is a known drug with multiple biological activities which depend on different molecular mechanisms of action.
      • VPA is an antiepileptic drug.
      • VPA is teratogenic. When used as antiepileptic drug during pregnancy VPA can induce birth defects (neural tube closure defects and other malformations) in a few percent of born children. In mice, VPA is teratogenic in the majority of mouse embryos when properly dosed.
      • VPA activates a nuclear hormone receptor (PPAR8). Several additional transcription factors are also derepressed but some factors are not significantly derepressed (glucocorticoid receptor, PPARα).
      • VPA is hepatotoxic, which may depend on poorly metabolized esters with coenzyme A.
  • The use of VPA derivatives allowed to determine that the different activities are mediated by different molecular mechanisms of action. Teratogenicity and antiepileptic activity follow different modes of action because compounds could be isolated which are either preferentially teratogenic or preferentially antiepileptic (Nau et al., 1991, Pharmacol. Toxicol. 69, 310-321). Activation of PPAR8 was found to be strictly correlated with teratogenicity (Lampen et al., 1999, Toxicol. Appl. Pharmacol. 160, 238-249) suggesting that, both, PPARδ activation and teratogenicity require the same molecular activity of VPA. Also, differentiation of F9 cells strictly correlated with PPARδ activation and teratogenicity as suggested by Lampen et al., 1999, and documented by the analysis of differentiation markers (Werling et al., 2001, Mol. Pharmacol. 59, 1269-1276).
  • It is shown in the present application, that PPARδ activation is caused by the HDAC inhibitory activity of VPA and its derivatives. Furthermore it is shown that the established HDAC inhibitor TSA activates PPARδ and induces the same type of F9 cell differentiation as VPA. From these results we conclude that not only activation of PPARδ but also induction of F9 cell differentiation and teratogenicity of VPA or VPA derivatives are most likely caused by HDAC inhibition.
  • The present invention is based on the finding that VPA and the derivatives described in this application are inhibitors of histone deacteylases. The finding of this novel mechanism of action of VPA and compounds derived thereof, i.e. the inhibition of enzymes with histone deacetylase activity led us to the proposition that VPA due to its HDAC-inhibitory activity should be useful to induce differentiation and/or apoptosis in a wide variety of cancer cells for two reasons: (1) these enzymes are present in all cells and (2) pilot studies with model compounds such as butyrate or TSA which are different from those described in this invention had shown that HDAC inhibitors induce differentiation in a wide variety of cells.
  • The activity to induce differentiation and/or apoptosis in a wide variety of transformed cells is a much more complex biological activity than only the inhibition of proliferation. In the latter case it would not be evident, why only the proliferation of transformed (tumor) but not of normal cells should be inhibited. The induction of apoptosis, differentiation or more specifically re-differentiation in dedifferentiated tumor cells provides a rationale why the compounds of this invention have beneficial effects in a wide variety of tumors by induction of differentiation and/or apoptosis. This proposition was confirmed in a wide variety of tumor cells (see examples).
  • Antiepileptic and sedating activities follow different structure activity relationships and thus obviously depend on a primary VPA activity distinct from HDAC inhibition.
  • The mechanism of hepatotoxicity is poorly understood and it is unknown whether it is associated with formation of the VPA-CoA ester. The use according to the invention, e.g. HDAC inhibition, however, appears not to require CoA ester formation.
  • U.S. Pat. No. 5,672,746 and WO 96/06821 disclose the use of VPA and derivatives thereof for the treatment of neuredegenerative and neuroproliferative disorders.
  • One aspect of the present invention is the use of VPA and derivatives thereof as an inhibitor of enzymes having histone deacetylase activity. Derivatives of VPA are α-carbon branched carboxylic acids as described by formula I
    Figure US20070232696A1-20071004-C00001

    wherein R1 and R2 independently are a linear or branched, saturated or unsaturated aliphatic C2-25, preferably C3-25 hydrocarbon chain which optionally comprises one or several heteroatoms and which may be substituted, R3 is hydroxyl, halogen, alkoxy or an optionally alkylated amino group.
  • Different R1 and R2 residues give rise to chiral compounds. Usually one of the stereoisomers has a stronger teratogenic effect than the other one (Nau et al., 1991, Pharmacol. Toxicol. 69, 310-321) and the more teratogenic isomer more efficiently activates PPARδ (Lampen et al, 1999). Therefore, this isomer can be expected to inhibit HDACs more strongly (this invention). The present invention encompasses the racemic mixtures of the respective compounds, the less active isomers, and in particular the more active isomers.
  • The hydrocarbon chains R1 and R2 may comprise one or several heteroatoms (e.g. O, N, S) replacing carbon atoms in the hydrocarbon chain. This is due to the fact that structures very similar to that of carbon groups may be adopted by heteroatom groups when the heteroatoms have the same type of hybridization as a corresponding carbon group.
  • R1 and R2 may be substituted. Possible substituents include hydroxyl, amino, carboxylic and alkoxy groups as well as aryl and heterocyclic groups.
  • Preferably, R1 and R2 independently comprise 2 to 10, more preferably 3 to 10 or 5 to 10 carbon atoms. It is also preferred that R1 and R2 independently are saturated or comprise one double bond or one triple bond. In particular, one of the side chains (R1) may preferably contain sp1 hybridized carbon atoms in position 2 and 3 or heteroatoms which generate a similar structure. This side chain should comprise 3 carbon or heteroatoms but longer chains may also generate HDAC-inhibiting molecules. Also inclusion of aromatic rings or heteroatoms in R2 is considered to generate compounds with HDAC inhibitory activity because the catalytic site of the HDAC protein apparently accommodates a wide variety of binding molecules. With the novel observation that teratogenic VPA derivatives are HDAC inhibitors, also compounds which have previously been disregarded as suitable antiepileptic agents are considered as HDAC inhibitors under this invention. In particular, but not exclusively, compounds having a propinyl residue as R1 and residues of 7 or more carbons as R2, are considered (Lampen et al, 1999).
  • Preferably, the group “COR3” is a carboxylic group. Also derivatization of the carboxylic group has to be considered for generating compounds with potential HDAC inhibitory activity. Such derivatives may be halides (e.g. chlorides), esters or amides. When R3 is alkoxy, the alkoxy group comprises 1 to 25, preferably 1-10 carbon atoms. When R3 is a mono- or di-alkylated amino group, the alkyl substituents comprise 1 to 25, preferably 1-10 carbon atoms. An unsubstituted amino group, however, is preferred.
  • According to the present invention also pharmaceutically acceptable salts of a compound of formula I can be used. According to the present invention also substances can be used which are metabolized to a compound as defined in formula I in the human organism or which lead to the release of a compound as defined in formula I for example by ester hydrolysis.
  • In a particular embodiment, the invention concerns the use of an α-carbon branched carboxylic acid as described by formula I or of a pharmaceutically acceptable salt thereof as an inhibitor of an enzyme having histone deacetylase activity wherein R1 is a linear or branched, saturated or unsaturated, aliphatic C5-25 hydrocarbon chain, R2 independently is a linear or branched, saturated or unsaturated, aliphatic C2-25 hydrocarbon chain, but not —CH2—CH═CH2, —CH2—C≡CH or —CH2—CH2—CH3, R1 and R2 are optionally substituted with hydroxyl, amino, carboxylic, alkoxy, aryl and/or heterocyclic groups, and R3 is hydroxyl.
  • In yet another embodiment the invention concerns the use of an α-carbon branched carboxylic acid as described by formula I or of a pharmaceutically acceptable salt thereof as an inhibitor of an enzyme having histone deacetylase activity wherein R1 is a linear or branched, saturated or unsaturated, aliphatic C3-25 hydrocarbon chain, and R2 independently is a linear or branched, saturated or unsaturated, aliphatic C2-25 hydrocarbon chain, R1 or R2 comprise one or several heteroatoms (e.g. O, N, S). replacing carbon atoms in the hydrocarbon chain, R1 and R2 are optionally substituted with hydroxyl, amino, carboxylic, alkoxy, aryl and/or heterocyclic groups, and R3 is hydroxyl.
  • In yet another embodiment of the invention R1 and R2 do not comprise an ester group (—CO—O—). The atom of R1 which is next to the α-carbon of the carboxylic acid (derivative) of formula I and covalently linked to said α-carbon may be a carbon atom. The atom of R2 which is next to the α-carbon of the carboxylic acid (derivative) of formula I and covalently linked to said α-carbon may be a carbon atom. R1 and R2 may be hydrocarbon chains comprising no heteroatoms O, N or S.
  • The compounds which are most preferably used according to the present invention are VPA, S-4-yn VPA, 2-EHXA (2-Ethyl-hexanoic acid).
  • The compounds are useful for inhibiting mammalian (for use of cell lines in in vitro assays and animal models systems) and in particular human (in vivo and in vitro) histone deacetylases HDAC 1-3 (class I) and HDAC 4-8 (class II).
  • The compounds may be used to induce the differentiation and/or apoptosis of cells such as undifferentiated tumour cells. Presumably, this reflects a general mechanism, as differentiation can be induced in F9 teratocarcinoma cells, MT 450 breast cancer cells, HT-29 colon carcinoma cells and several leukemia cell lines as assessed by morphological alterations and specific marker gene or protein expression. Furthermore, for example MT450 cells can be induced to undergo apoptosis (see example 6).
  • The invention also concerns the use of a compound of formula I for the induction of differentiation and/or apoptosis of transformed cells.
  • Another aspect of the present invention is the use of a compound of formula I for the manufacture of a medicament for the treatment of a disease which is associated with gene-specific hypoacetylation of histones. There are a number of diseases which are associated with aberrant repression of specific genes which correlates with a local level of histone acetylation below the regular level.
  • Yet another aspect of the invention is the use of a compound of formula I for the manufacture of a medicament for the treatment of a disease in which the induction of hyperacetylation of histones has a beneficial effect resulting in differentiation and/or apoptosis of a patient's tumor cells and thus causing a clinical improvement of the patient's condition. Examples of such diseases are skin cancer, estrogen receptor-dependent and independent breast cancer, ovarian cancer, prostate cancer, renal cancer, colon and colorectal cancer, pancreatic cancer, head and neck cancer, small cell and non-small cell lung carcinoma. The induction of hyperacetylation may also be beneficial by reverting inappropriate gene expression in diseases based on aberrant recruitment of histone deacetylase activity such as thyroid resistance syndrome.
  • The compounds and salts thereof can be formulated as pharmaceutical compositions (e.g. powders, granules, tablets, pills, capsules, injections, solutions, foams, enemas and the like) comprising at least one such compound alone or in admixture with pharmaceutically acceptable carriers, excipients and/or diluents. The pharmaceutical compositions can be formulated in accordance with a conventional method. Specific dose levels for any particular patient will be employed depending upon a variety of factors including the activity of specific compounds employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and the severity of the particular disease undergoing therapy. The active ingredient will preferably be administered in an appropriate amount, for example, selected from the range of about 10 mg/kg to 100 mg/kg body weight a day orally or intravenously. The dose levels are not specifically restricted as long as serum levels of 0.05 mM to 3 mM, preferably of about 0.4 mM to 1.2 mM are achieved.
  • Another aspect of the invention is a method for the identification of substances having histone deacetylase inhibitory activity which comprises providing a derivative of valproic acid, determining its histone deacetylase inhibitory activity, and selecting the substance if the substance has histone deacetylase inhibitory activity. Valproic acid can serve as a lead substance for the identification of other compounds exhibiting histone deacetylase inhibitory activity. Thereby compounds may be selected which show increased HDAC inhibitory activity at lower doses and serum levels and have decreased effects on the central nervous system such as sedating activity. Another parameter that may be optimised is the appearance of the hepatotoxic effect. Compounds may be selected which show a reduced liver toxicity. The derivatives may be provided by synthesising compounds which comprise additional and/or modified substituents. The HDAC inhibitory activity may be determined by a state-of-the-art technology such as transcription repression assay, a Western Blot which detects acetylation of histone H3 and/or histone H4, or by an enzymatic assay.
  • The transcriptional assay for repressor activity exploits activation and derepression of a Gal4-dependent reporter gene. This assay can be performed either by transient transfection of mammalian cell lines (e.g. HeLa, 293T, CV-1) or with specifically constructed permanent cell lines. Transcription factors such as thyroid hormone receptor, PPARδ, retinoic acid receptor, N-CoR and AML/ETO repress transcription when they bind to a promoter containing UAS elements as fusion proteins with the heterologous DNA-binding domain of the yeast Gal4 protein. In the absence of the Gal4-fusion protein the reporter gene has a high basal transcriptional activity due to the presence of binding sites for other transcription factors in the thymidine kinase promoter. The Gal4 fusion proteins repress this activity by up to 140-fold. HDAC inhibitors induce relief of this repression which can be detected as an increase in reporter gene activity (e.g. by luciferase assay).
  • Histone deacetylase inhibitors induce the accumulation of N-terminally hyperacetylated histones H3 and H4. These acetylated histones can be detected by Western blot analysis of whole cell extracts or of histone preparations from histone deacetylase inhibitor-treated cells using antibodies specific for the acetylated N-terminal lysine residues of histones H3 and H4.
  • The enzymatic assay for HDAC activity records the release of 3H-labeled acetic acid from hyperacetylated substrates. Sources of HDAC activity can be co-immunoprecipitates with antibodies directed against N-CoR (or other repressors known to recruit HDACs) or crude cell extracts containing histone deacetylases (e.g. HeLa, 293T, F 9). Substrates may be either chemically 3H-acetylated peptides corresponding to the. N-termini of histones H3 or H4 or histone proteins isolated from metabolically labelled cells which were treated with HDAC inhibitors. After extraction with ethyl acetate the release of 3H-labeled acetic acid is detected by liquid scintillation counting.
  • Yet another aspect of the invention is a method for profiling of the HDAC isoenzyme specificity of a compound as defined in formula I wherein the binding of the compound to HDAC and/or the competition for binding to HDAC is measured.
  • The method may comprise the following steps: HDACs are either immune precipitated with HDAC isoform specific antibodies, with antibodies directed against corepressor complexes, or with specific antibodies against recombinant HDACs overexpressed in transgenic cells. The method may further involve determination of individual HDACs present in these immune precipitates by Western blot analysis. Radiolabeled VPA or compounds according to formula I are bound to the immune precipitates. The amount of bound compound is determined through measurement of bound radioactivity after appropriate washing steps. A variation of this aspect involves binding of one labeled HDAC inhibitor such as VPA, TSA or trapoxin and competition of binding by a compound according to formula I. Another variation of the method involves the use of alternate labeling and/or detection procedures.
  • It is preferred that compounds are selected which specifically inhibit only a subset of HDACs.
  • Another aspect of the invention is the use of VPA or derivatives thereof to define genes which are induced by said compounds in cells such as primary human or rodent cells, leukemic cells, other cancer cells or tumor cell lines. Methods to define such genes that are induced by VPA include established technologies for screening large arrays of cDNAs, expressed sequence tags or so-called unigene collections. Also the use of subtractive hybridization techniques is suitable to define genes which are induced by VPA or derivatives thereof. The use of these methods to identify potential targets for drug development downstream of HDAC-inhibition, and furthermore the use of these methods to define diagnostic means in order to facilitate the therapeutic treatment of patients with suitable compounds is part of this invention. Considering the low general toxicity of VPA in the organism compared to other HDAC-inhibitors it is a specific aspect of this invention to use VPA or derivatives thereof for defining target genes which are selectively regulated or not regulated by VPA, particularly also in comparison to other HDAC inhibitors like trichostatin A.
  • The present invention also concerns a diagnostic method to identify tumors comprising the step of testing whether a tumor is responsive to treatment with compounds as defined by formula I. The method preferably comprises the method for the identification of genes induced by VPA or a derivative thereof described supra. In a particular embodiment, the diagnostic method comprises the use of nucleic acid technology, preferably of hybridization or polymerase chain reaction for detection. Other types of nucleic acid technology, however, may be employed. In another embodiment the method comprises the use of specific antibodies against differentially regulated proteins for detection. For this purpose proteins encoded by the genes showing deregulation of their expression upon VPA treatment would be expressed e.g. in recombinant expression systems and antibodies directed against these proteins would be generated. Subsequently such antibodies could be used (or patterns of antibodies) to characterize the status of a tumor or tumor cells for diagnostic and/or prognostic reasons.
  • The present invention provides novel possibilities to treat various cancer diseases. Applicant found that VPA and derivatives thereof are potent HDAC inhibitors. The HDAC inhibitors known so far are either nonspecific like butyrate, or toxic or poorly bioavailable in the whole organism like TSA and TPX. VPA has the advantage that it is already an approved drug and has been used over decades for the treatment of epilepsy in human. Thus, a vast amount of data concerning pharmaceutical acceptability and the lack of serious side effects are available. Thus VPA should be a suitable drug for the use in humans to induce differentiation and/or apoptosis in transformed cells and by that to exert beneficial effects in a wide variety of patients suffering from cancer.
  • FIG. 1 describes the histone deacetylase inhibitor-like activation of PPARδ by VPA (example 1).
  • FIG. 2 shows that VPA activates several transcription factors in addition to PPARδ (example 2).
  • FIG. 3 shows VPA-induced accumulation of hyperacetylated histones H3 and H4 (example 3).
  • FIG. 4 shows the biochemical analysis of histone deacetylase activity in the absence or presence of VPA (example 4).
  • FIG. 5 shows indicators of VPA induced differentiation in HT-29 colonic carcinoma cells, F9-teratocarcinoma cells, and RenCa renal carcinoma cells. The phenotypes of F9-teratocarcinoma cells differentiated by VPA or the histone deacetylase inhibitor trichostatin A appear identical (example 5).
  • FIG. 6 shows induction of apoptosis in MT450 breast cancer cells (example 6).
  • FIG. 7 shows the the loss of viable cells upon treatment with valproic acid. Renca-lacZ, Renca-lacZ/EGFR, Renca-lacZ/EGFRvIII and Renca-lacZ/ErbB2 renal carcinoma cells (A) or SKOV3 ovarian carcinoma cells, SKBR3, MCF7, MDA-MB453 and MDA-MB468 breast carcinoma cells, and A431 squamous cell carcinoma cells (B) were incubated with the indicated concentrations of valproic acid (VPA). The relative number of viable cells was determined using the enzymatic MTT assay, measuring cellular metabolic activity, as described in Example 7. Each point represents the mean of a set of data determined in triplicate (example 7).
  • FIG. 8 shows the reduction in cellular biomass after treatment of cell cultures with VPA (example 8)
  • The following examples further illustrate the invention.
  • EXAMPLE 1
  • Activation of a PPARδ-Glucocorticoid Receptor Hybrid Protein by VPA
  • A reporter gene cell line for activation of the PPARδ ligand binding domain was constructed in CHO cells. A subclone of CHO cells was used which contained a transgenic reporter gene expressing a secreted form of the human placental alkaline phosphatase under control of the glucocorticoid receptor-dependent LTR-promoter of the mouse mammary tumor virus (Göttlicher et al. (1992) Proc. Natl. Acad. Sci. USA 89, pp. 4653-4657). A hybrid receptor comprising the amino-terminus of the glucocorticoid receptor fused to the ligand binding domain of PPARδ was expressed in these cells essentially as described for the expression of the corresponding hybrid of PPARα (Göttlicher et al., 1992, ibd.). The ligand binding domain of PPARδ was used starting at amino acid 138 as deduced from the sequence published by Amri et al. (J. Biol. Chem. 270 (1995) pp. 2367-2371). Activation of the PPARδ ligand binding domain in these cells induces expression of the alkaline phosphatase reporter gene which is detectable by an enzymatic assay from the cell culture supernatant. Similar cells expressing the full length glucocorticoid receptor served as negative controls for specificity of receptor activation. For the experiment shown in FIG. 1 the PPARδ hyrid receptor expressing cells were seeded at 20% confluency into 24-well culture dishes and treated for 40 h with the PPARδ ligand carbocyclic prostaglandin I2 (PGI, 5 μM), VPA (1 or 2 mM), or the histone deacetylase inhibitors sodium butyrate (0, 2-5 mM) and trichostatin A (TSA, 300 nM). Reporter gene activity was monitored by an enzymatic assay (alkaline phosphatase). Values except for butyrate are means±S.D. from triplicate determinations in 2 independent experiments which were normalized according to cPGI-induced activity (FIG. 1). The highly synergistic activation of the reporter gene by VPA together with the PPARδ ligand cPGI (P+V) which is similar to the synergistic activation by Trichostatin A together with cPGI (P+T), and the lack of synergism with trichostatin (T+V) or butyrate (not shown) indicate that VPA does not act like a bona fide ligand to PPARδ. VPA rather affects PPARδ activity by a mechanism which lies in the same sequence of events by which also the inhibitors of corepressor-associated histone deacetylases induce transcriptional activity of PPARδ.
  • EXAMPLE 2
  • Activation of Transcriptional Repressors by VPA
  • The transcription factors thyroid hormone receptor (TR), peroxisome proliferator activated receptor δ (PPARδ), retinoic acid receptor (RAR), the corepressor N-CoR and the AML/ETO fusion protein repress transcription when they bind to a promoter containing UAS sites (Gal4 response element) as fusion proteins with the heterologous DNA binding domain of the yeast Gal4 protein. In the absence of the Gal4 fusion protein a luciferase reporter gene is transcribed at a high basal level due to the presence of binding sites for other transcription factors in the thymidine kinase (TK) promoter. Hela cells were transfected with a UAS TK luciferase reporter plasmid (Heinzel et al., 1997, Nature 387, pp 43-48) and expression plasmids for the indicated Gal4 fusion proteins using the calcium phosphate precipitate method. After 24 h the medium was changed and cells were incubated with histone deacetylase inhibitors for a further 24 h. Transcriptional repression is measured as luciferase activity relative to the baseline of cells transfected with an expression plasmid for the Gal4 DNA binding domain alone (FIG. 2). The Gal4 fusion proteins repress this baseline activity by up to 140 fold. VPA at a concentration of 1 mM (close to the serum levels which are reached during therapeutic use) induces relief of this- repression which is indicated as an increase in reporter gene activity. A relief of repression is also found after treatment with established histone deacetylase inhibitors (10 nM Trapoxin, 100 nM TSA) as well as after partial activation of TR and PPARδ by their respective ligands. A combination of ligand and HDAC inhibitors (including VPA) results in a synergistic effect, indicating that different molecular mechanisms are involved. FIG. 2 shows that VPA affects the activity of several distinct transcription factors and cofactors. This finding suggests that VPA acts on a common factor in the regulation of gene expression such as corepressor-associated histone deacetylases rather than on individual transcription factors or receptors (e.g. as a ligand).
  • EXAMPLE 3
  • Accumulation of hyperacetylated histones in VPA-treated cells VPA and established histone deacetylase inhibitors like sodium butyrate (NaBu) or trichostatin A (TSA) induce the accumulation of hyperacetylated histones H3 and H4. These acetylated histones can be detected by Western blot analysis in cell extracts of appropriately treated cells. FIG. 3 shows the results of such an analysis from a representative experiment. In this experiment both the time course of VPA-induced hyperacetylation (A) and the required VPA concentration (B) were determined.
  • (A) For the time course analysis F9 cells were seeded into 6-well culture dishes 30 h before the intended time point of analysis. Individual cultures were treated at the indicated time points before analysis by addition of 10-fold concentrated stock solutions in culture medium of VPA or trichostatin A. Whole cell extracts were prepared by rinsing the cell cultures twice in ice-cold phosphate buffered saline and lysis of cells in 250 μl of sample buffer for denaturing SDS gel electrophoresis. DNA of collected samples was sheared by sonication and samples were separated on a 15% denaturing polyacrylamide gel. Acetylated histones H3 and histone H4 were detected by Western blot analysis using commercially available antibodies (Upstate Biotechnology) specific for the acetylated forms of histones (Ac-H3, Cat-Nr.: 06-599; Ac-H4, Cat-Nr.: 06-598). Equal loading of the lanes was confirmed by staining a part of the polyacrylamide gel by Coomassie blue.
  • (B) For determination of the required VPA dose F9 cells were cultured in 6-well culture dishes for 8 h prior to addition of VPA at the indicated concentrations. Whole cell extracts were prepared 16 h after treatment as described above. Analysis for acetylated histones H3 and H4 was performed as described in (A). VPA concentrations in the range of blood serum levels reached during therapeutic use of VPA as antiepileptic agent in humans induce hyperactylation of histones H3 and H4. At serum levels only slightly exceeding those intended for antiepileptic therapy VPA induces histone hyperacetylation as efficiently as sodium butyrate or trichostatin A used at concentrations which are expected to have a maximum effect. This experiment indicates that VPA or a metabolite formed in F9 cells inhibits histone deacetylase activity.
  • EXAMPLE 4
  • VPA and Derivatives Inhibit Histone Deacetylase Activity in vitro
  • Immune precipitates from whole cell extracts using antibodies against the corepressor N-CoR or mSin3 contain histone deacetylase activity. This enzymatic activity is measured by incubating the immune precipitates with radioactively acetylated histone substrates from cells in which histones have been hyperacetylated in the presence of 3H-acetate. The release of 3H-acetate is detected as a measure of enzymatic activity by extraction with ethyl acetate and subsequent liquid scintillation counting (FIG. 4). Addition of the histone deacetylase inhibitor trichostatin A (TSA, 10−7 M) to the reaction in vitro severely inhibits the enzymatic activity. VPA (from left to right 0.2 mM, 1 mM, 5 mM) and the related compounds ethyl hexanoic acid (EHXA, from left to right 0.008 mM, 0.04 mM, 0.2 mM, 1 mM, 5 mM), R-4-yn VPA (from left to right 0.2 mM, 1 mM, 5 mM) and S-4-yn VPA (from left to right 0.2 mM, 1 mM, 5 mM) were tested for HDAC inhibitory activity. The assays were performed with N-CoR immunoprecipitates from 293T cells in duplicate. Immunoprecipitates were pretreated with HDAC inhibitors for 15 min prior to the addition of substrate and subsequent incubation for 2.5 h at 37° C. (untreated enzyme activity 2,205 cpm=100%). Precipitates of a preimmune serum served as a negative control. EC50 values are 0.6 mM for VPA, 0.2 mM for EHXA and 0.3 mM for S-4-yn VPA, whereas the stereoisomer R-4-yn VPA is inactive. These data show that VPA by itself rather than a cellular metabolite inhibits histone deacetylase activity.
  • EXAMPLE 5
  • Induction of Cell Differentiation in F9 Teratocarcinoma, HT-29 Colonic Cancer, and RenCa Renal Carcinoma Cells.
  • Histone deacetylase inhibitors and VPA in particular induce differentiation of dedifferentiated tumorigenic cells. Cell differentiation is associated with cell cycle arrest, morphological alterations and the appearance of expression of markers of the differentiated phenotype. Morphological alterations where determined by microscopic evaluation of F9 and HT-29 cells. One parameter of differentiation, the cell cycle arrest, was shown in F9 teratocacrcinoma, estrogen independent MT-450 breast cancer and HT-29 colonic carcinoma cells by means of the reduced incorporation of 3H-thymidine into cultured cells. F9 and HT-29 cells were cultured for 36 h in the absence or the presence of 1 mM VPA in 96-well culture dishes. 37 kBq of 3H-thymidine were added for additional 12 h of culture. MT-450 cells were cultured for 72 h prior to a 1 h 3H-thymidine labelling period. Incorporation of 3H-thymidine into DNA was determined by automatic cell harvesting and liquid scintillation counting. VPA pretreatment reduced the rate of thymidine incorporation by 48±5%, 63±8%, and 52±8% in F9, MT-450, and HT-29 cells, respectively. The dose-response for the reduction of thymidine incorporation into HT-29 cells (FIG. 5A) was determined by the same experimental procedure. In addition, the induction of a cell differentiation marker was shown in F9 teratocarcinoma cells (FIG. 5B).
  • F9 teratocarcinoma cells were treated for 48 h with VPA (1 mM), sodium butyrate (B, 1 mM) and trichostation A (TSA, 30 nM). Differentiation was followed by morphological criteria, a reduced rate in the increase of cell number (e.g. cell cycle arrest, data not shown), the drop of 3H-thymidine incorporation by 48% during a 12 h pulse labeling period (see above) and the appearance of nuclear AP-2 protein (FIG. 5B) as a specific marker of histone deacetylase inhibitor-induced differentiation of F9 cells. Nuclear AP-2 protein was detected in nuclear extract which had been prepared by mild detergent lysis (25 mM Tris, pH 7.5; 1 mM EDTA, 0.05*NP40) of treated or non-treated F9 cells, recovery of nuclei by centrifugation (3000×g, 5 min) and lysis of nuclei in sample buffer for denaturing SDS gel electrophoresis. Nuclear extracts were separated on a 9% SDS polyacrylamide gel. AP-2 protein was detected by Western blot analysis using a rabbit polyclonal antibody (Santa Cruz, Cat.-No.: SC-184) at a dilution of 1/1000 in Tris buffered saline containing 3W non-fat dry milk and 0.05% Tween 20. Both VPA and trichostatin A induce nuclear AP-2 protein whereas the activity of butyrate at the chosen concentration is weak. Since appearance of AP-2 is a delayed effect which is only detectable after 36 to 40 h of VPA treatment the weak activity of butyrate may be caused by efficient metabolism of the compound. Nevertheless, VPA induces differentiation of the epithelial F9 cell line in a way indistinguishable from differentiation by other histone deacetylase inhibitors.
  • Induction of differentiation in RenCa-LacZ cells by VPA was determined by alterations in cell morphology. RenCa-LacZ cells were cultured for 36 h either in the absence or the presence of 1 mM VPA. Morphological alterations were observed by phase contrast microscopy and micrographs of representative fields were taken (FIG. 5C)
  • EXAMPLE 6
  • Induction of Apoptosis in MT-450 Breast Cancer Cells
  • MT-450 cells were cultured for 72 h in the absence or presence of 1 mM VPA. Apoptotic cells were detected by flow cytometric analysis after staining of cell surface exposed phosphatidylserine with FITC-conjugated annexin V (Becton Dickinson) according to suppliers instructions. Dead cells were excluded by propidium iodide staining. Cells positive for annexin V and negative for propidium iodide uptake (lower right quadrant in FIG. 6) were judged and counted as apoptotic cells.
  • EXAMPLE 7
  • Loss of Viable Tumor Cells Upon Treatment with Valproic Acid (MTT Tests)
  • Cell Lines and Cell Culture
  • Human MDA-MB468, MDA-MB453 and SKBR3 breast carcinoma cells, A431 squamous cell carcinoma cells, and SKOV3 ovarian carcinoma cells were maintained in Dulbecco's modified Eagle's medium (DMEM, BioWhittaker, Verviers, Belgium) supplemented with 10% heat inactivated fetal bovine serum (FBS), 2 mM L-glutamine, 100 units/ml penicillin, and 100 μg/ml streptomycin. Human MCF7 breast carcinoma cells were grown in RPMI medium supplemented as described above.
  • Renal cell carcinoma (Renca) cells stably transfected with plasmid pZeoSV2/lacZ encoding E. coli β-galactosidase (Renca-lacZ cells) (Maurer-Gebhard et al., Cancer Res. 58: 2661-2666, 1998) were grown in RPMI-1640 medium supplemented with 8% FBS, 2 mM L-glutamine, 100 U/ml penicillin, 100 μg/ml streptomycin, 0.25 mg/ml Zeocin. Renca-lacZ cells cotransfected with plasmids pSV2ErbB2N and pSV2neo encoding c-erbB2 and neomycin resistance (Renca-lacZ/ErbB2) (Maurer-Gebhard et al., Cancer Res. 58: 2661-2666, 1998), or plasmids pLTR-EGFR or pLTR-EGFRvIII and pSV2neo encoding epidermal growth factor (EGF) receptor, the oncogenically activated EGF receptor variant EGFRvIII, and neomycin resistance (Renca-lacZ/EGFR and Renca-lacZ/EGFRvIII) (Schmidt et al., Oncogene 18: 1711-1721, 1999) were grown in the same medium further containing 0.48 mg/ml G418.
  • Cell Viability Assays
  • Tumor cells were seeded in 96 well plates at a density of 1×104 cells/well in normal growth medium. Valproic acid was added at final concentrations of 1 or 3 mM to triplicate samples and the cells were incubated for 40 h (Renca-lacZ, Renca-lacZ/ErbB2, Renca-lacZ/EGFR, Renca-lacZ/EGFRvIII, SKBR3 and SKOV3 cells) or 70 h (A431, MCF7, MDA-MB453 and MDA-MB468 cells). Control cells were grown in the absence of valproic acid. Ten μl of 10 mg/ml 3-(4,5-dimethylthiazole-2-yl)-2,5 diphenyltetrazolium bromide (MTT) (Sigma, Deisenhofen, Germany) in PBS were added to each well and the cells were incubated for another 3 h. Cells were lysed by the addition of 90 μl of lysis buffer (20% SDS in 50% dimethyl formamide, pH 4.7). After solubilization of the formazan product, the absorption at 590 nm was determined in a microplate reader (Dynatech, Denkendorf, Germany) and the relative amount of viable cells in comparison to cells grown without the addition of valproic acid was calculated.
  • Results
  • The results presented in FIG. 7 show that valproic acid reduces the viability of breast carcinoma cells, ovarian carcinoma cells, squamous cell carcinoma cells, renal carcinoma cells, and renal carcinoma cells expressing at high levels the ErbB2 or EGF receptor proto-oncogenes, or the oncogenically activated EGF receptor variant EGFRVIII, in a concentration dependent manner. These results demonstrate that valproic acid potently reduces the number and/or viability of a wide variety of tumor cells derived from solid tumors of epithelial origin. The loss of viability could indicate a reduction in cell number upon induction of cellular differentiation and/or induction of cell death. The observation of changes of cellular morphology suggest that cellular differentiation is at least responsible for a part of the effect. This induction of differentiation and/or induction of cell death suggest that valproic acid and derivatives thereof could be used for the therapy of such tumors.
  • EXAMPLE 8
  • Reduction in Cellular Biomass After Treatment of Human Cancer Cell Cultures with Valproic Acid (see FIG. 8).
  • VPA induces differentiation and/or cell death in a series of human cancer cells and reduces the total cellular biomass of human cancer cell cultures. The reduction in biomass could indicate cell loss due to cell death and/or differentiation associated cell cycle arrest. Quantitative parameters, e.g. the loss of biomass, was determined in 30 human cancer cell lines (FIG. 8 e) and twelve examples of dose-response curves are shown, e.g. BT-549 breast cancer cells (1), estrogen dependent ZR-75 breast cancer cells (2), DMS-114 small cell lung cancer cells (3), NCI-H226 non-small cell lung cancer cells (4), SK-MEL-28 skin cancer cells (5), OVCAR-3 ovarian cancer cells (6), HUP-T3 pancreatic cancer cells (7), DU-145 prostate cancer cells (8), DETROIT-562 head and neck cancer cells, LS-174 colon cancer cells (10), A-172 brain cancer cells (11) and HL-60 leukemia cells (12) (FIG. 8 a-d). All cells were evaluated for morphological signs of cell death and/or differentiation. All cultures contained an increased number of dying cells at the highest tested VPA concentration and in some cultures such as SW-1116 colon cancer cells (FIG. 8 e) most cells were dying already at 1 mM VPA during the experiment. PC-3 (FIG. 8 e) and DU-145 (FIG. 8 c) cells change their normal round morphology to a long fibroblast-like shape. Also U87MG (FIG. 8 e) cells increase in length and develop spider-like filamentous extensions.
  • Cells in panels 1 to 9 (FIG. 8 a-c) were seeded in 96 well culture dishes at densities between 3000 and 8000 per well. After recovery of 24 hours cells were cultured for 48 hours in the absence or presence of the indicated concentrations of VPA. Cultures were fixed with TCA by layering 50 μl of cold 50*TCA on top of the growth medium in each well to produce a final TCA concentration of 10%. After 1 hour of incubation at 4° C. the cells were washed five times with tap water and air dried. Fixed cells were stained for 30 minutes with 0.4% (wt/vol) Sulforhodamine B dissolved in 1% acetic acid and washed four times with 1% acetic acid to remove unbound dye.After air drying bound dye was solubilized with 10 mM unbuffered Tris base (pH 10.5) for 5 minutes on a gyratory shaker. Optical densities were read on a Titertek Multiskan Plus plate reader at a single wavelength of 550 nm. Six test wells for each dose-response were set in parallel with 12 control wells per cell line. A measure of the cell population density at time 0 (T0; the time at which the drug was added) was also made from 12 extra reference wells of cells fixed with TCA just prior to drug addition to the test plates. Background OD of complete medium with 5% FBS fixed and stained as described above was also determined in 12 separate wells.
  • From the unprocessed OD data the background OD measurements (i.e. OD of complete medium plus stain and OD of cells at T0) were subtracted thus giving the reduction of total cellular biomass of the cells.
  • Cells in panels 10 to 12 (FIG. 8 d) were cultured 36 to 50 hours as indicated in the absence or presence of the indicated concentrations of VPA in 96 well dishes. 37 kBq of 3H-thymidine were added for additional 12 hours of culture. Incorporation of 3H-thymidine into DNA was determined by automatic cell harvesting and liquid scintillation counting.
  • The graphs in FIG. 8 a-d show means±S.D. from sixfold determinations.
  • In addition cancer cells of further organ origins have been treated with valproic acid in the same way as described for experiments presented in FIG. 8 a-c. FIG. 8 e summarizes the reduction of total cellular biomass of various human cancer cells by treatment with 1 mM VPA. This reduction could indicate differentiation associated cell cycle arrest and/or induction of cell death. Cells were VPA treated for 48 hours. The inhibition was calculated from six response tests performed in parallel and reductions of cellular biomass are given in percent of untreated cells with standard deviations.

Claims (12)

1-23. (canceled)
24. The use of a compound of Formula I as an inhibitor of an enzyme having histone deacetylase activity,
Figure US20070232696A1-20071004-C00002
wherein R1 and R2 independently are a linear or branched, saturated or unsaturated, aliphatic C2-25 hydrocarbon chain which may be substituted with a hydroxyl, amino, alkoxy, aryl or heterocyclic group, R3 is hydroxyl, halogen, alkoxy or an optionally alkylated amino group; or a pharmaceutically acceptable salt of a compound of the Formula I; or a prodrug or pharmaceutically active metabolite of a compound of the Formula I, or a pharmaceutically acceptable salt of a prodrug or metabolite thereof.
25. The use according to claim 24, wherein R1 and R2 independently are a linear or branched, saturated or unsaturated, C2-10 hydrocarbon chain.
26. The use according to claim 24, wherein the compound is selected from the group consisting of VPA, S-4-yn VPA, 2-ethyl hexanoic acid and pharmaceutically acceptable salts, prodrugs or pharmaceutically active metabolites thereof.
27. The use according to anyone of claims 24 to 26, wherein the enzyme having histone deacetylase activity is mammalian, preferably human histone deacetylase.
28. The use according to claim 27, wherein the human histone deacetylase is selected from the group consisting of histone deacetylases HDAC 1-3 (class I) and HDAC 4-8 (class II).
29. The use according to claim 24, wherein the compound is used for the induction of differentiation of cells.
30. The use according to claim 29, wherein the compound is used for the induction of differentiation of transformed cells.
31. The use according to claim 24, wherein the compound is used for the induction of apoptosis of transformed cells.
32-49. (canceled)
50. A method of treating a disease or disorder associated with the hyperacetylation of histones, comprising administering to a subject in need of such treatment, an agent capable of inhibiting histone deacetylase, selected from the group consisting of valproic acid, S-4-yn valproic acid, and 2-ethyl-hexanoic acid or a pharmaceutically acceptable salt; or a prodrug or pharmaceutically active metabolite, or a pharmaceutically acceptable salt of a prodrug or metabolite thereof, wherein the disease is an endocrine disorder associated with induction of histone deacetylase.
51. The method of claim 50, wherein the disease is thyroid resistance syndrome.
US11/653,855 2000-07-07 2007-01-17 Valproic acid and derivatives thereof as histone deacetylase inhibitors Abandoned US20070232696A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/653,855 US20070232696A1 (en) 2000-07-07 2007-01-17 Valproic acid and derivatives thereof as histone deacetylase inhibitors

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP00.114088.8 2000-07-07
EP00114088A EP1170008A1 (en) 2000-07-07 2000-07-07 Valproic acid and derivatives thereof as histone deacetylase inhibitors
US10/332,353 US7265154B2 (en) 2000-07-07 2001-07-05 Valproic acid and derivatives thereof as histone deacetylase inhibitors
PCT/EP2001/007704 WO2002007722A2 (en) 2000-07-07 2001-07-05 Valproic acid and derivatives thereof as histone deacetylase inhibitors
US11/653,855 US20070232696A1 (en) 2000-07-07 2007-01-17 Valproic acid and derivatives thereof as histone deacetylase inhibitors

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US10/332,353 Division US7265154B2 (en) 2000-07-07 2001-07-05 Valproic acid and derivatives thereof as histone deacetylase inhibitors
PCT/EP2001/007704 Division WO2002007722A2 (en) 2000-07-07 2001-07-05 Valproic acid and derivatives thereof as histone deacetylase inhibitors

Publications (1)

Publication Number Publication Date
US20070232696A1 true US20070232696A1 (en) 2007-10-04

Family

ID=8169134

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/332,353 Expired - Fee Related US7265154B2 (en) 2000-07-07 2001-07-05 Valproic acid and derivatives thereof as histone deacetylase inhibitors
US11/653,855 Abandoned US20070232696A1 (en) 2000-07-07 2007-01-17 Valproic acid and derivatives thereof as histone deacetylase inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/332,353 Expired - Fee Related US7265154B2 (en) 2000-07-07 2001-07-05 Valproic acid and derivatives thereof as histone deacetylase inhibitors

Country Status (10)

Country Link
US (2) US7265154B2 (en)
EP (2) EP1170008A1 (en)
JP (1) JP2004504347A (en)
AT (1) ATE295725T1 (en)
AU (2) AU2001283925B2 (en)
CA (1) CA2414967C (en)
DE (1) DE60110912T2 (en)
ES (1) ES2239677T3 (en)
PT (1) PT1301184E (en)
WO (1) WO2002007722A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100317739A1 (en) * 2007-12-14 2010-12-16 Brown Milton L Histone deacetylase inhibitors

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7621606B2 (en) * 2001-08-27 2009-11-24 Advanced Cell Technology, Inc. Trans-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
EP1170008A1 (en) 2000-07-07 2002-01-09 Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus Valproic acid and derivatives thereof as histone deacetylase inhibitors
GB0024362D0 (en) * 2000-10-05 2000-11-22 Glaxo Group Ltd Medicaments
EP1293205A1 (en) * 2001-09-18 2003-03-19 G2M Cancer Drugs AG Valproic acid and derivatives thereof for the combination therapy of human cancers, for the treatment of tumour metastasis and minimal residual disease
US6706686B2 (en) 2001-09-27 2004-03-16 The Regents Of The University Of Colorado Inhibition of histone deacetylase as a treatment for cardiac hypertrophy
EP1443928B1 (en) 2001-10-16 2011-07-27 Sloan-Kettering Institute For Cancer Research Treatment of neurodegenerative diseases and cancer of the brain
EP1490013B1 (en) 2002-03-07 2010-02-17 University Of Delaware Methods for enhancing oligonucleotide-mediated nucleic acid sequence alteration using compositions comprising hydroxyurea
IL149404A0 (en) * 2002-04-29 2002-11-10 Yissum Res Dev Co METHODS AND COMPOSITIONS FOR MODULATING β-CATENIN PHOSPHORYLATION
GB0226855D0 (en) 2002-11-18 2002-12-24 Queen Mary & Westfield College Histone deacetylase inhibitors
WO2004050885A2 (en) * 2002-12-05 2004-06-17 Imperial College Innovations Limited Control of apoptosis using a complex of an oligonucleotide and a regulatory peptide
US7244751B2 (en) 2003-02-14 2007-07-17 Shenzhen Chipscreen Biosciences Ltd. Histone deacetylase inhibitors of novel benzamide derivatives with potent differentiation and anti-proliferation activity
CA2518318A1 (en) * 2003-03-17 2004-09-30 Takeda San Diego, Inc. Histone deacetylase inhibitors
EP1491188A1 (en) * 2003-06-25 2004-12-29 G2M Cancer Drugs AG Topical use of valproic acid for the prevention or treatment of skin disorders
WO2005065681A1 (en) * 2003-12-19 2005-07-21 Takeda San Diego, Inc. N- hydroxy-3-(3-(1h-imidazol-2-yl)-phenyl)-acrylamide derivatives and related compounds as histone deacetylase (hdac) inhibitors for the treatment of cancer
US20050137234A1 (en) * 2003-12-19 2005-06-23 Syrrx, Inc. Histone deacetylase inhibitors
PL1591109T3 (en) * 2004-04-30 2008-11-28 Topotarget Germany Ag Formulation comprising histone deacetylase inhibitor exhibiting biphasic release
KR100659779B1 (en) 2004-09-04 2006-12-19 주식회사 바이넥스 Osteogenic differentiation of human mesenchymal stem cells using histone deacetylase inhibitors
JP2008524246A (en) * 2004-12-16 2008-07-10 タケダ サン ディエゴ インコーポレイテッド Histone deacetylase inhibitor
US7642253B2 (en) * 2005-05-11 2010-01-05 Takeda San Diego, Inc. Histone deacetylase inhibitors
WO2007011626A2 (en) * 2005-07-14 2007-01-25 Takeda San Diego, Inc. Histone deacetylase inhibitors
EP1976835A2 (en) * 2006-01-13 2008-10-08 Takeda San Diego, Inc. Histone deacetylase inhibitors
WO2008011603A2 (en) * 2006-07-20 2008-01-24 Wisconsin Alumni Research Foundation Modulating notch1 signaling pathway for treating neuroendocrine tumors
US20080085874A1 (en) * 2006-08-28 2008-04-10 The Regents Of The University Of California Small molecule potentiator of hormonal therapy for breast cancer
GB0620823D0 (en) 2006-10-19 2006-11-29 Univ London Histone deacetylase inhibitors
US20100093862A1 (en) * 2006-12-06 2010-04-15 Sapporo Medical Univeristy And Japan Science And Technology Agency Potentiation of cellular immunity using histone deacetylase (hdac) inhibitors.
US20090023186A1 (en) * 2007-07-22 2009-01-22 Excellgene Sa Use of valproic acid for enhancing production of recombinant proteins in mammalian cells
US8623853B2 (en) 2008-07-23 2014-01-07 The Brigham And Women's Hospital, Inc. Treatment of cancers characterized by chromosomal rearrangement of the NUT gene
ES2473792T3 (en) 2009-04-03 2014-07-07 Naturewise Biotech & Medicals Corporation Kinematic compounds and derivatives thereof for histone deacetylase inhibition
US7994357B2 (en) 2009-04-03 2011-08-09 Naturewise Biotech & Medicals Corporation Cinamic compounds and derivatives therefrom for the inhibition of histone deacetylase
EP2277387B1 (en) 2009-07-22 2016-10-19 NatureWise Biotech & Medicals Corporation New use of histone deacetylase inhibitors in changing mrjp3 protein in royal jelly
EP2475769B1 (en) * 2009-09-07 2016-11-09 Jawaharlal Nehru Centre for Advanced Scientific Research Inhibition of histone acetyltransferases by ctk7a and methods thereof
US8921533B2 (en) 2011-07-25 2014-12-30 Chromatin Technologies Glycosylated valproic acid analogs and uses thereof
EP3052662B1 (en) 2013-10-01 2019-05-15 The J. David Gladstone Institutes Compositions, systems and methods for gene expression noise drug screening and uses thereof
WO2015168621A1 (en) * 2014-05-02 2015-11-05 Oncoethix Sa Method of treating acute myeloid leukemia and/or acute lymphoblastic leukemia using thienotriazolodiazepine compounds
CN105622712B (en) * 2014-10-27 2019-08-27 天津金耀集团有限公司 A kind of steroidal 3- ketone -4- alkene object synthesis technology
TWI794171B (en) 2016-05-11 2023-03-01 美商滬亞生物國際有限公司 Combination therapies of hdac inhibitors and pd-l1 inhibitors
US20210386787A1 (en) * 2018-10-12 2021-12-16 Life Technologies Corporation Hematopoietic Stem and Progenitor Cell Expansion System
CN111358777A (en) * 2020-02-24 2020-07-03 军事科学院军事医学研究院环境医学与作业医学研究所 Method for improving mitochondrial function of peripheral blood mononuclear cells and application

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5672746A (en) * 1994-08-30 1997-09-30 American Biogenetic Sciences, Inc. Antiproliferative and neurotrophic molecules

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6300373B1 (en) * 1993-09-10 2001-10-09 American Biogenetic Sciences, Inc. Antiproliferative and neurotrophic molecules
CA2174737C (en) * 1993-10-29 2008-04-22 Douglas V. Faller Physiologically stable compositions of butyric acid, and butyric acid salts and derivatives as anti-neoplastic agents
JPH11514857A (en) * 1995-09-20 1999-12-21 メルク エンド カンパニー インコーポレーテッド Histone deacetylase as a target for antiprotozoal agents
US6777217B1 (en) * 1996-03-26 2004-08-17 President And Fellows Of Harvard College Histone deacetylases, and uses related thereto
WO1997047307A1 (en) * 1996-06-14 1997-12-18 The Uab Research Foundation Use of histone deacetylase inhibitors to activate transgene expression
WO1998029114A1 (en) * 1996-12-30 1998-07-09 Bar-Ilan University Tricarboxylic acid-containing oxyalkyl esters and uses thereof
US6387673B1 (en) * 1997-05-01 2002-05-14 The Salk Institute For Biological Studies Compounds useful for the modulation of processes mediated by nuclear hormone receptors, methods for the identification and use of such compounds
EP1037533A1 (en) * 1997-11-10 2000-09-27 The Salk Institute for Biological Studies Methods for the use of inhibitors of co-repressors for the treatment of neoplastic diseases
US6262116B1 (en) * 1998-01-23 2001-07-17 Sloan-Kettering Institute For Cancer Research Transcription therapy for cancers
EP1170008A1 (en) 2000-07-07 2002-01-09 Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus Valproic acid and derivatives thereof as histone deacetylase inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5672746A (en) * 1994-08-30 1997-09-30 American Biogenetic Sciences, Inc. Antiproliferative and neurotrophic molecules

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100317739A1 (en) * 2007-12-14 2010-12-16 Brown Milton L Histone deacetylase inhibitors
US8293513B2 (en) 2007-12-14 2012-10-23 Georgetown University Histone deacetylase inhibitors

Also Published As

Publication number Publication date
EP1301184A2 (en) 2003-04-16
EP1170008A1 (en) 2002-01-09
US7265154B2 (en) 2007-09-04
WO2002007722A3 (en) 2002-07-18
AU2001283925B2 (en) 2005-07-14
ES2239677T3 (en) 2005-10-01
ATE295725T1 (en) 2005-06-15
DE60110912D1 (en) 2005-06-23
AU8392501A (en) 2002-02-05
JP2004504347A (en) 2004-02-12
CA2414967A1 (en) 2002-01-31
PT1301184E (en) 2005-07-29
EP1301184B1 (en) 2005-05-18
DE60110912T2 (en) 2005-10-20
CA2414967C (en) 2009-08-25
US20040087652A1 (en) 2004-05-06
WO2002007722A2 (en) 2002-01-31

Similar Documents

Publication Publication Date Title
US7265154B2 (en) Valproic acid and derivatives thereof as histone deacetylase inhibitors
AU2001283925A1 (en) Valproic acid and derivatives thereof as histone deacetylase inhibitors
Nelson et al. 13-cis Retinoic acid induces apoptosis and cell cycle arrest in human SEB-1 sebocytes
Ye et al. Small molecule inhibitors targeting activator protein 1 (AP-1) miniperspective
Ross et al. Retinoids in embryonal development
Clay et al. Influence of J series prostaglandins on apoptosis and tumorigenesis of breast cancer cells
Hazra et al. Peroxisome proliferator-activated receptor γ induces a phenotypic switch from activated to quiescent hepatic stellate cells
Nagai et al. Significance of the transcription factor KLF5 in cardiovascular remodeling
Niles Signaling pathways in retinoid chemoprevention and treatment of cancer
Kim et al. Small heterodimer partner and fibroblast growth factor 19 inhibit expression of NPC1L1 in mouse intestine and cholesterol absorption
Liu et al. Growth inhibition and apoptosis in human Philadelphia chromosome-positive lymphoblastic leukemia cell lines by treatment with the dual PPARα/γ ligand TZD18
Matsushima-Nishiwaki et al. Molecular mechanism for growth suppression of human hepatocellular carcinoma cells by acyclic retinoid
Shang et al. Protein acylation: mechanisms, biological functions and therapeutic targets
Nagpal et al. Retinoid antagonism of NF-IL6: insight into the mechanism of antiproliferative effects of retinoids in Kaposi’s sarcoma
JP4896743B2 (en) Combination of PRAME inhibitor and HDAC inhibitor
Scott et al. Glutamate-stimulated peroxynitrite production in a brain-derived endothelial cell line is dependent on N-methyl-D-aspartate (NMDA) receptor activation
Hall-Glenn et al. CCN2/CTGF is required for matrix organization and to protect growth plate chondrocytes from cellular stress
Lucarelli et al. Troglitazione affects survival of human osteosarcoma cells
Estévez-Silva et al. Pridopidine Promotes Synaptogenesis and Reduces Spatial Memory Deficits in the Alzheimer's Disease APP/PS1 Mouse Model
Vourtsis et al. Effect of an all-trans-retinoic acid conjugate with spermine on viability of human prostate cancer and endothelial cells in vitro and angiogenesis in vivo
Wright et al. Functional activation of PPARγ in human upper aerodigestive cancer cell lines
Lee et al. Interferon regulatory factor-1 (IRF-1) regulates VEGF-induced angiogenesis in HUVECs
Hayashi et al. Effect of 9-cis-retinoic acid on oral squamous cell carcinoma cell lines
Bozzo et al. Antiproliferative effect of conjugated linoleic acid in caco-2 cells: involvement of PPARγ and APC/β-catenin pathways
JP2013177329A (en) Rxr partial agonist

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION