EP1523470A1 - Novel compounds as histone deacetylase inhibitors - Google Patents

Novel compounds as histone deacetylase inhibitors

Info

Publication number
EP1523470A1
EP1523470A1 EP03765029A EP03765029A EP1523470A1 EP 1523470 A1 EP1523470 A1 EP 1523470A1 EP 03765029 A EP03765029 A EP 03765029A EP 03765029 A EP03765029 A EP 03765029A EP 1523470 A1 EP1523470 A1 EP 1523470A1
Authority
EP
European Patent Office
Prior art keywords
compound according
cancer
medicament
compounds
manufacture
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.)
Withdrawn
Application number
EP03765029A
Other languages
German (de)
French (fr)
Inventor
Alexander B. Maurer
Sascha Hoevelmann
Elke Martin
Bernd Hentsch
Michael Gassen
Juergen Kraus
Rolf Krauss
Adam-Spencer Vincek
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.)
4SC AG
Original Assignee
4SC AG
G2M Cancer Drugs AG
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 4SC AG, G2M Cancer Drugs AG filed Critical 4SC AG
Publication of EP1523470A1 publication Critical patent/EP1523470A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C259/00Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups
    • C07C259/04Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids
    • C07C259/06Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids having carbon atoms of hydroxamic groups bound to hydrogen atoms or to acyclic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated

Definitions

  • the present invention relates to compounds as inhibitors of enzymes having histone deacetylase activity, to the processes for the preparation of those compounds, and to their use for the treatment of diseases which are associated with hypoacetylation of histones and/or other molecules, or in which induction of hyperacetylation has a beneficial effect for example by inhibition of proliferation and/or induction of differentiation and/or induction of apoptosis in transformed cells, such as cancer.
  • the compounds are useful for the treatment of other proliferative diseases, for therapy or prophylaxis of conditions associated with abnormal gene expression.
  • chromatin remodeling is a key step in the transcriptional activation of genes. Dynamic changes in the nucleosomal packaging of DNA must occur to allow transcriptional proteins to contact with the DNA template.
  • One of the most important mechanisms influencing chromatin remodeling and gene transcription are the posttranslational modification of histones and other cellular proteins by acetylation and subsequent changes in chromatin structure (Davie, 1998, Curr Opin Genet Dev 8, 173-8; Kouzarides, 1999, Curr Opin Genet Dev 9, 40-8; Strahl and Allis, 2000, Nature 403, 41-4).
  • HAT histone acetyltransferase
  • PML-RAR the fusion protein associated with acute promyelocytic leukemia (APL) inhibits p53 through mediating deacetylation and degradation of p53, thus allowing APL blasts to evade p53 dependent cancer surveillance pathways.
  • APL acute promyelocytic leukemia
  • HDAC inhibitors implicating active recruitment of HDAC to p53 by PML-RAR as the mechanism underlying p53 inhibition (Insinga et al. 2002, manuscript submitted). Therefore, factor acetylation plays a crucial role in the anti-tumor activity of HDAC inhibitors.
  • 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. 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 (Pazin and Kadonaga, 1997; Cell 89, 325-8). 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 (Pazin and Kadonaga, 1997, Cell 89, 325-8; Huynh and Bardwell, 1998, Oncogene 17, 2473-84; Wang, J. et al., 1998, Proc Natl Acad Sci U S A 95, 10860-5).
  • T3R for example was originally identified on the basis of its homology with the viral oncogene v-erbA, which in contrast to the wild type receptor does not bind ligand and functions as a constitutive repressor of transcription.
  • RARs have been associated with a number of human cancers, particularly acute promyelocytic leukemia (APL) and hepatocellular carcinoma.
  • APL acute promyelocytic leukemia
  • PML promyelocytic leukemia protein
  • PZF promyelocytic zinc finger protein
  • Another class comprises cyclic tetrapeptides, such as depsipeptide (FR901228 - Fujisawa) used successfully in a phase II trial for the treatment of T-cell lymphomas (Piekarz et al., 2001, Blood 98, 2865-8). Furthermore, MS-27-275 (Mitsui Pharmaceuticals), a compound related to the class of benzamides, is now being tested in a phase I trial patients with hematological malignancies.
  • depsipeptide FR901228 - Fujisawa
  • MS-27-275 Mitsubishi Pharmaceuticals
  • hydroxamic acids as matrix metalloproteinase inhibitors are described.
  • the compounds of the present invention are hydroxamic acids which are inhibitors of enzymes having histone deacetylase activity. Due to their HDAC-inhibitory activity they induce differentiation and/or apoptosis in a wide variety of cancer cells for three 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 and (3) clinical efficacy has been demonstrated for several other HDAC inhibitors unrelated to the presented compounds in the treatment of cancer patients.
  • 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.
  • the histone deacetylase inhibitory activity of new compounds may be determined by a number of state-of-the-art technologies such as transcriptional repression assay, Western Blot analysis which detects acetylation of histone H3 and/or histone H4, or by an enzymatic in vitro assay.
  • Histone deacetylase inhibitors can be further characterized for their cytotoxic and growth inhibitory effects on tumor cell lines and for their ability to modulate gene expression patterns in cells.
  • the present invention is directed to compounds of the general formula (I):
  • n is a non-aromatic ring system containing two to six carbon atoms, wherein the ring system can contain one ore two double bonds;
  • X is C, CH or CH 2 ;
  • Y is selected from C, CH, CH 2 , S, NR, CH 2 -CH 2 ,
  • each of the dotted lines means a single, a double or triple bond with the exclusion of a combination of a triple with triple bond and a double with a triple bond;
  • R x is independently H, -CN, alkyl, cycloalkyl, aminoalkyl, alkylamino, alkoxy, -OH, -SH, alkylthio, hydroxyalkyl, hydroxyalkylamino, halogene, haloalkyl, haloalkyloxy;
  • R is H, an alkyl or cycloalkyl group
  • Z is CH, C, or P; p is 0 or 1 ;
  • alkyl group if not stated otherwise, is preferably a linear or branched chain of 1 to 6 carbon atoms, preferably a methyl, ethyl, propyl, isopropyl, butyl, f-butyl, isobutyl, pentyl or hexyl group, a methyl, ethyl, isopropyl or ---butyl group being most preferred.
  • alkyl unless otherwise noted, is also meant to include those derivatives of alkyl defined in more detail below as "unsaturated alkyl".
  • An unsaturated alkyl group is one having one or more double bonds or triple bonds, preferably vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4- pentadienyl, 3-(1 ,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • alkyl group in the compounds of formula (I) can optionally be substituted by one or more substituents K being as defined above.
  • An cycloalkyl group denotes a non-aromatic ring system containing 3 to 8 carbon atoms, wherein one or more of the carbon atoms in the ring can be replaced by a group X, X being as defined above.
  • An alkoxy group denotes an O-alkyl group, the alkyl group being as defined above.
  • alkylthio group denotes an S-alkyl group, the alkyl group being as defined above.
  • a hydroxyalkyl group denotes an HO-alkyl group, the alkyl group being as defined above.
  • An haloalkyl group denotes an alkyl group which is substituted by one to five preferably three halogen atoms, the alkyl group being as defined above; a CF 3 being preferred.
  • haloalkyloxy group denotes an alkoxy group which is substituted by one to five preferably three halogen atoms, the alkoxy group being as defined above; a OCF 3 being preferred.
  • a hydroxyalkylamino group denotes an (HO-alkyl) 2 -N-group or HO-alkyl-NH- group, the alkyl group being as defined above.
  • An alkylamino group denotes an HN-alkyl or N-dialkyl group, the alkyl group being as defined above.
  • An aminoalkyl group denotes an H 2 N-alkyl, monoalkylaminoalkyl, or diaikylaminoalkyl group, the alkyl group being as defined above.
  • a halogen group is chlorine, bromine, fluorine or iodine, fluorine being preferred.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) in free form or in the form of pharmaceutically acceptable salts and physiologically functional derivatives or prodrugs, together with a pharmaceutically acceptable diluent or carrier therefore.
  • physiologically functional derivative refers to compounds like ethers, esters, N-alkylated or acetylated hydroxamic acids, 2,5-dihydro-[1 , 2, 4] - oxodiazolyl or 4,5-dihydro-[1 , 2, 4]-oxodiazolyl, which are not pharmaceutically active themselves but which are transformed into their pharmaceutical active form in vivo, i.e. in the subject to which the compound is administered.
  • the present invention also provides a method for the treatment or prophylaxis of a condition where there is an advantage in inhibiting histone deacetylase activity which comprises the administration of an effective amount of a compound of formula (1) and physiologically acceptable salts or physiologically functional derivatives thereof.
  • the invention is also directed to the use of compounds of the formula (I) and of their pharmacologically tolerable salts or physiologically functional derivatives for the production of a medicament for the prevention and treatment of diseases, where inhibition of histone deacetylase is of benefit.
  • the present invention provides methods for preparing the desired hydroxamic acides of the formula (I).
  • One method for the synthesis of compounds of the formula (I) comprises the conversion of an acid (formula II) to the corresponding acid chloride (formula III) and reacting the acid chloride with hydroxylamine (Watanabe et al., 1989, J. Org. Chem., 54, 17, 4088-4097; Shishido et al., 1992, J. Org. Chem., 57, 10, 2876-2883).
  • the ring n including Z can be cyclopentyl, cyclohexyl, cycloheptyl, cyclopent-1 -enyl, cyclohex-1 -enyl, cyclohept-1 -enyl, cyclopent-2-enyl, cyclohex-2-enyl, cyclohept-2-enyl, cyclohex-3- enyl, cyclohept-3-enyl.
  • the ring n including Z is cyclopentyl or cyclohexyl
  • Y is selected from CH, CH 2 , CH 2 -CH 2
  • p 0
  • Z is CH.
  • none of the carbon atoms of the alkyl groups is replaced by a group A.
  • Preferred compounds of the present invention are:
  • the compounds of the formula (I) according to the invention can form salts with inorganic or organic acids or bases.
  • examples of such salts are, for example, alkali metal salts, in particular sodium and potassium salts, or ammonium salts.
  • the compounds of formula (I) may be obtained via various methods. In preferred embodiments of the methods of the invention the two following methods of synthesis are used.
  • chlorinating agents oxalylchloride, thionylchloride, phosphorpenta chloride
  • the compounds of the present invention can be used for a variety of human and animal diseases, preferably human diseases, where inhibition of histone deacetylase activity is beneficial.
  • the compounds according to the invention and medicaments prepared therewith are generally useful to induce the differentiation and/or apoptosis of cells such as undifferentiated tumor cells.
  • the therapeutic effect of the invention may arise through one or more mechanisms, including but not limited to, the regulation of cell proliferation, cell activation, cell survival, cell differentiation, cell cycle, cell maturation and cell death or to induce systemic changes in metabolism such as changes in sugar, lipid or protein metabolism, the inhibition of new blood vessel formation (anti- angiogenesis), the inhibition of tumor spread into other organs (anti-metastatic), the inhibition of tumor spread into neighboring normal structures (anti-invasive) or the promotion of programmed cell death (apoptosis).
  • cell generation poiesis including blood cell growth and generation (prohematopoietic effect) after depletion or destruction of cells, as caused by, for example, toxic agents, radiation, immunotherapy, growth defects, malnutrition, malabsorption, immune dysregulation, anemia and the like or to provide li
  • the compounds according to the invention and medicaments prepared therewith are also useful for the treatment of a disease which is associated with gene-specific hypoacetylation of histones or other molecules, such as p53. Additionally, the compounds of the present invention may also be used in the treatment of conditions where the suppression of anti-apoptotic genes, such as BCL-XL and other BCL family members, or the induction of tumor suppressor activity of molecules such as p21 and/or p53, is required.
  • the compounds according to the invention and medicaments prepared therewith are also suitable for the treatment of diseases 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 include but are not limited to, skin cancer, melanoma, estrogen receptor-dependent and independent breast cancer, ovarian cancer, testosteron receptor-dependent and independent prostate cancer, renal cancer, colon and colorectal cancer, pancreatic cancer, bladder cancer, esophageal cancer, stomach cancer, genitourinary cancer, gastrointestinal cancer, uterine cancer, astrocytomas, gliomas, basal cancer and squameous cell carcinoma, sarcomas as Kaposi's sarcoma and osteosarcoma, head and neck cancer, small cell and non-small cell lung carcinoma, leukemia, lymphomas and other blood cell cancers, Keratoakantoma, Bowen Disease, cutaneous T-Cell Lymphoma and also for the treatment of pre-malignant lesions (such as Actinic Keratose).
  • the combinatorial treatment of the present invention is particularly useful for treating minimal residual tumor disease or tumor metastases.
  • the invention may also be beneficial by reverting inappropriate gene expression in diseases based on aberrant recruitment of histone deacetylase activity such as thyroid resistance syndrome, or in other conditions associated with abnormal gene expression, such as inflammatory disorders, diabetes, thalassemia, cirrhosis, protozoal infection, or the like and all types of autoimmune diseases, in particular rheumatoid arthritis, rheumatoid spondylitis, all forms of rheumatism, osteoarthritis, gouty arthritis, multiple sclerosis, insulin dependent diabetes mellitus and non-insulin dependent diabetes, asthma, rhinitis, uveithis, lupus erythematoidis, ulcerative colitis, Morbus Crohn, inflammatory bowel disease, as well as other chronic inflammations, chronic diarrhea and of inflammations of the skin and/or mucosa (such as Psoriasis, lchtiosis, Acne).
  • the invention also relates to the use for the protection from UV
  • proliferative diseases such as psoriasis, fibrosis, warts and other dermatological disorders.
  • proliferative disease and “cell proliferation”, are used interchangeably herein and relate to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or . hyperplastic growth, whether in vitro or in vivo.
  • proliferative conditions include, but are not limited to, pre-malignant and malignant cellular proliferation, including malignant neoplasms and tumors, cancers, leukemias, psoriasis, bone disease, fibroproliferative disorders (e.g.
  • Any type of cell may be treated, including but not limited to, lung, colon, breast, ovarian, prostate, liver, pancreas, brain, and skin and any treatment of disorders involving T-cells such as aplastic anemia and DiGeorge syndrome, Graves' disease.
  • the invention encompasses also compounds of formula (I) which are metabolized in patients to a compound of the presented formula.
  • the embodiments described in this invention apply to such compounds as well.
  • the present invention also concerns a diagnostic method to identify tumors comprising the step of testing in vitro whether a tumor is responsive to treatment either with compounds of formula (I) or in combination with established tumor therapeutics.
  • the method preferably comprises the method for the identification of genes regulated by these treatments.
  • 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 combinatorial treatment using formulations of this invention and derivatives thereof 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 human diseases.
  • Applicants found that the HDAC inhibitory and cellular differentiation- inducing activity of compounds of formula (I) can be used successfully in combination with well established and clinically used therapeutic drugs for the treatment of tumor cells of different origins.
  • Such compound based combinatorial treatment is considered to generate superior therapeutic success in human tumor patients than the corresponding therapeutic drugs used on their own.
  • Such combinatorial treatments could result in a decrease of the therapeutic doses of e.g. chemotherapeutic reagents required and could thus limit the currently observed, partly very serious side effects of frequently used therapies.
  • Chemotherapeutic or cytotoxic drugs e.g. 5-FU, taxol, cisPlatinum, camptothecin, gemcitabine, doxorubicine, irinothecan
  • differentiation inducing drugs e.g. vitamin D, retinoic acid, cytokines such as
  • Radiotherapy e.g. x-rays or gamma rays
  • immunological approaches antibody therapy, vaccination
  • immunotherapeutic/cytotoxic approaches e.g. antibodies conjugated with cytotoxic components
  • the compounds and salts thereof can be formulated, as pharmaceutical compositions (e.g. liquids, suspensions, emulsions, lozenges, cachets, ampoules, suppositories, pessaries, ointments, gels, pastes, sprays, lotions, oils, boluses, electuaries, aerosols, powders, granules, tablets, pills, capsules, injections, solutions, foams, creams, 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.
  • Administration in vivo can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosages of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
  • a suitable dose of the active compound is in the range of about 0.1 to about 500 mg per kilogram body weight preferably 0.1 to 100 mg per kilogram body weight of the subject per day.
  • the active ingredient is a salt, an ester, prodrug, or the like
  • the amount administered is calculated on the basis the parent compound and so the actual weight to be used is increased proportionately.
  • Figure 1 shows the relief of HDAC-dependent transcriptional repression in a reporter cell line, UAS TK-luc, after 24 hours treatment with Compound 1 , 2 and 3. Depicted is the fold induction of luciferase expression after treatment of cells at the indicated concentrations of Compounds 1 , 2 and 3.
  • Figure 2 shows the induction histone H4 hyperacetylation in 293T cells after treatment with Compounds 1 , 2 and 3 for six hours at the indicated concentrations.
  • FIG. 3 shows the inhibition of in-vitro HDAC activity in nuclear extracts from HeLa cells after treatment with Compounds 1, 2 and 3 at the indicated concentrations. Relative HDAC activity is shown as percent of activity of untreated nuclear extracts.
  • Figure 4 shows the inhibition of in-vitro HDAC activity of recombinant HDAC1 , HDAC6, and HDAC8 enzymes after treatment with Compound 3 at the indicated concentrations. Relative HDAC activity is shown as percent of activity of untreated HDAC enzymes.
  • Figure 5 shows on the protein level the down-regulation of the anti-apoptotic molecule BCL-XL and up-regulation of the cell cycle inhibitor p21 in K562 cells induced after treatment with Compounds 1 , 2 and 3 for 36 hours at the indicated concentrations.
  • Figure 6 shows the concentration of Compounds 1, 2 and 3 inducing a 50% reduction in cellular biomass (IC5 0 ) after treatment of various cancer cell lines for 72 hours.
  • Figure 7 shows the induction of apoptosis in BV-173 cells after treatment for 24 hours with Compounds 1, 2 and 3 at the indicated concentrations. Apoptosis is demonstrated by the increase of a sub-G1 population in Propidium Iodine treated cells, which is reduced to baseline levels after treatment with a pan-caspase inhibitor Z-VAD.
  • the resulting aldehyde was used for the next step without further purification.
  • the crude cyclohexanecarbaldehyde was dissolved in toluene (200 mL) and ethanol (150 mL). After 15 min of stirring at 70 °C carbethoxy-methylene triphenylphosphorane (33.6 g, 96.5 mmol) was added in one portion. Stirring was continued for additional 24 hours. The solvent was removed in vacuo. The product was obtained by flash chromatography in 78 % yield (12.5 g).
  • 3-Cyclohexyl-acrylic acid to a solution of 3-Cyclohexyl-acrylic acid ethyl ester (7.00 g, 38.4 mmol) in dioxane (200 mL) was added lithium hydroxide (4.03 g, 96.1 mmol) dissolved in water (70 mL). Because LiOH fell out methanol (170 mL) was added to the reaction. After 4 hours at 70 °C 3/4 of the solvent was removed in vacuo. 2 N HCI was added until the pH reached 3. The mixture was treated with ammonium chloride (150 mL) saturated solution. The product was extracted with dichloromethane (200 mL x 3).
  • 3-Cyclohexyl-N-hydroxy-acrylamide (K1 00003283): to a solution of 3-Cyclohexyl- acrylic acid (5.30 g, 31.3 mmol) in DMF (100 mL) was added EDC (1-(3- Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride) (7.80 g, 40.7 mmol) and HOBt (1 -hydroxybenzotriazole hydrate) (5.07 g, 37.6 mmol). After stirring for 30 min at room temperature hydroxylamine hydrochloride (3.26 g, 47.0 mmol) and triethylamine (6.94 mL, 50.1 mmol) were added.
  • HDAC histone deacetylase
  • HDAC enzymes and thereby repress transcription when bound to a specific promoter region.
  • This latter effect can be reenacted with a constitutive promoter containing UAS elements which recruits fusion proteins containing the heterologous DNA- binding domain of the yeast Gal4 protein fused to one of the above mentioned transcription factors.
  • 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.
  • Transcriptional reporter gene assay The transcriptional assay for repressor activity exploits activation and derepression of a Gal4-dependent reporter gene (Hildebrand et al., 2001, J Biol Chem 276, 9889-95 ; Maurer et al., 2002, Blood 99, 2647-52). This assay is performed with specifically constructed permanent cell lines. 293T cell were stably transfected with a UAS TK lucif erase reporter plasmid (Heinzel et al., 1997 Nature 387, pp43-48) and an expression plasmid for the Gal4 DNA binding domain fused to an HDAC-dependent repressor molecule. While Gal4 fusion proteins repress this activity, HDAC inhibitors induce relief of this repression which can be detected as an increase in reporter gene activity (e.g. by luciferase activity detection assay).
  • the substrate turn over may be detected and quantified by fluorometry.
  • a potential HDAC inhibitor the hydrolysis of the substrate is constrained resulting in a decreased fluorometric signal.
  • IC 5 o values may be calculated from dose-response curves.
  • the assay is separated in two steps: in the first step the substrate (Fluor de Lys / Biomol KI-104) is hydrolysed by histone deacetylases. In step two HDAC activity is terminated and the fluorophore is activated by the addition of a developer (Developer / Biomol KI-105). Nuclear extracts from HeLa cells (Biomol KI-140) or recombinant proteins and the potential HDAC inhibitor are mixed with reaction buffer (Biomol Kl- 143) to a total volume of 25 ⁇ l per well of a 96 well plate. 25 ⁇ l substrate (1 :100 dilution in reaction buffer) per well are added to start the reaction.
  • a negative control without histone deacetylase activity and a positive control without HDAC inhibitor are treated likewise.
  • the reaction is stopped after 15-60min. by adding 50 ⁇ l developer (1 :20 dilution in reaction buffer). After another 15min. incubation time at room temperature the fluorescence signal is stable for 60min and may be detected by a fluorescence reader (excitation filter: 390nm, emission filter: 460nm).
  • the Gal4 fusion protein represses the baseline activity of the TK promoter and the subsequent luciferase expression.
  • Addition of Compounds 1 , 2 and 3 relieve this repression as measured by increased expression and activity of luciferase in lysates of treated cells after 24 hours.
  • Both, Compound 1 and 2 induce luciferase reporter gene expression after stimulation of cells for 24 hours starting at a concentration of 8 ⁇ M (2-3 fold) with a maximum induction of more than 20 fold at 200 ⁇ M.
  • Compound 3 induces expression of reporter gene already at a concentration of 1.6 ⁇ M with a maximum induction of almost 40 fold at 40 ⁇ M.
  • histone deacetylase inhibitors shift the enzymatic balance between histone acetyl transferases (HAT's) and histone deacetylases (HDAC's) towards HAT's by blocking HDAC's, they induce the accumulation of N-terminally hyperacetylated histones H4.
  • HAT's histone acetyl transferases
  • HDAC's histone deacetylases
  • Hyperacetylation can be seen with Compounds 1 and 2 at minimum concentrations of 1 ,6 ⁇ M with a maximum induction. at 40 ⁇ M, whereas compound 3 induces hyperacetylation starting at 320 nM.
  • HDAC activity in nuclear extracts is dramatically reduced in the presence of Compounds 1 , 2 and 3, with a 50% reduction of HDAC activity at a concentration of 14.7 ⁇ M for Compound 1 , at 6.1 ⁇ M for Compound 2, and at 0.9 ⁇ M for Compound 3.
  • HDAC activity of recombinant HDAC1 , HDAC6, and HDAC8 is dramatically reduced in the presence of Compound 1 and 2, with a 50% reduction of HDAC activity at a concentration of 0.3 to 0.4 ⁇ M for
  • BCL-X L high expression of BCL-X L is found in many human cancers and is often a negative prognostic factor. Accordingly, downregulation of BCL-XL expression in certain cancer cells either induces apoptosis directly or sensitizes cells to apoptotic stimuli. In general, induction of apoptosis can be exploited therapeutically using HDAC inhibitors in cancer therapy
  • Protein expression profiling The expression pattern induced by histone deacetylase inhibitors can be monitored by Western Blot analysis with antibodies against p21 and BCL-X L using whole cell extracts of cells treated with the respective compounds at the indicated concentrations. Modulation of protein expression is exemplified by the induction of expression of the cell cycle inhibitor/tumor suppressor p21 and the suppression of expression of the anti-apoptotic molecule BCL-X L .
  • the reduction in cellular biomass was measured by SRB-assay.
  • SRB-assay For this assay cells were seeded in 96 well culture dishes at densities between 3000 and 8000 cells per well. After recovery of 24 hours cells were cultured for 48-72 hours in the absence or presence of the indicated concentrations of compounds. Synergistic reduction in total cellular biomass was assayed through SRB-assay by adding the compounds in the medium at the indicated concentrations and cultivate the cells for further 48 hours.
  • TCA Trichloracetat
  • test wells for each dose-response were set in parallel with 12 control wells per cell line. Measurement of the cell population density at time 0 (T 0 ; the time at which the drug was added) was also made from 12 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 from each microtiter plate the background OD measurements (i.e. OD of complete medium plus stain and OD of cells at T 0 ) were subtracted thus giving the reduction of cellular biomass of the cells.
  • the cell cycle can be divided into four different sections: During G 0/1 -phase cells are in senescence or proliferate, in S-phase cells start replicating their DNA, and during G2- and M-phase cells undergo mitosis. The cellular DNA content correlates with cell cycle progression: while cells in G0 1 phase possess one set of chromosomes, cells in G2- and M-phase possess two full sets of chromosomes. Cells in S-phase are still replicating their DNA and therefore exhibit a DNA content between one and two sets of chromosomes. Degradation of DNA is a marker for apoptosis.
  • PI propidium iodide
  • the sub-G1 area indicates the hypodiploid DNA peak corresponding to cells with fragmented DNA undergoing apoptosis Using a dye that intercalates with DNA (such as propidium iodide - PI) the cellular DNA content was determined. Between 5x10 5 - 1x10 6 cells were seeded out in cell culture dishes and treated for the desired time with the desired amount of test compound. After the incubation cells were harvested, washed in cold PBS, and the cell pellet was resuspended in 1ml 70% ethanol (-20°C). These cell pellets may be shelved for several month.
  • a dye that intercalates with DNA such as propidium iodide - PI
  • the concentrations inducing 50% growth arrest ranged from 8 to 70 ⁇ M (mean 33 ⁇ M) for Compound 1 , from 5 to 26 ⁇ M (mean 15 ⁇ M) for Compound 2, and from 0.5 to 8 ⁇ M (mean 2.6 ⁇ M) for Compound 3.

Abstract

The present invention is directed to compounds of the general formula (I) or pharmaceutical acceptable salts or physiologically functional derivatives thereof wherein: n is a non-aromatic ring system containing two to seven carbon atoms, wherein the ring system can contain one ore two double bonds; X is C, CH or CH2; Y is selected from C, CH, CH2, S, NR, CH2-CH2, H2C- -CH, HC- -CH2, C- -CH2, H2C- -C,or C - -C; one or more of the hydrogen atoms can opionally be substituted by one or more substituents R ; each of the dotted lines means a single, a double or triple bond with the exclusion of a combination of a triple with triple bond and a double with a triple bond; R is independently H, -CN, alkyl, cycloalkyl, aminoalkyl, alkylamino, alkoxy, -OH, -SH, alkylthio, hydroxyalkyl, hydroxyalkylamino, halogene, haloalkyl, haloalkyloxy; R is H, an alkyl or cycloalkyl group; Z is CH, C, or P; p is 0 or 1.

Description

Novel compounds as istone deacetylase inhibitors
The present invention relates to compounds as inhibitors of enzymes having histone deacetylase activity, to the processes for the preparation of those compounds, and to their use for the treatment of diseases which are associated with hypoacetylation of histones and/or other molecules, or in which induction of hyperacetylation has a beneficial effect for example by inhibition of proliferation and/or induction of differentiation and/or induction of apoptosis in transformed cells, such as cancer. Furthermore, the compounds are useful for the treatment of other proliferative diseases, for therapy or prophylaxis of conditions associated with abnormal gene expression.
BACKGROUND OF THE INVENTION
Local remodeling 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 to contact with the DNA template. One of the most important mechanisms influencing chromatin remodeling and gene transcription are the posttranslational modification of histones and other cellular proteins by acetylation and subsequent changes in chromatin structure (Davie, 1998, Curr Opin Genet Dev 8, 173-8; Kouzarides, 1999, Curr Opin Genet Dev 9, 40-8; Strahl and Allis, 2000, Nature 403, 41-4). In the case of histone hyperacetylation, changes 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 to 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 tumorigenic transformation. Additionally, other molecules such as transcription factors alter their activity and stability depending on their acetylation status. E.g. PML-RAR, the fusion protein associated with acute promyelocytic leukemia (APL) inhibits p53 through mediating deacetylation and degradation of p53, thus allowing APL blasts to evade p53 dependent cancer surveillance pathways. Expression of PML-RAR in hematopoietic precursors results in repression of p53 mediated transcriptional activation, and protection from p53-dependent apoptosis triggered by genotoxic stresses (X-rays, oxidative stress). However, the function of p53 is reinstalled in the presence of HDAC inhibitors implicating active recruitment of HDAC to p53 by PML-RAR as the mechanism underlying p53 inhibition (Insinga et al. 2002, manuscript submitted). Therefore, factor acetylation plays a crucial role in the anti-tumor activity of HDAC inhibitors.
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. 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 (Pazin and Kadonaga, 1997; Cell 89, 325-8). 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 (Pazin and Kadonaga, 1997, Cell 89, 325-8; Huynh and Bardwell, 1998, Oncogene 17, 2473-84; Wang, J. et al., 1998, Proc Natl Acad Sci U S A 95, 10860-5). T3R for example was originally identified on the basis of its homology with the viral oncogene v-erbA, which in contrast to the wild type 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 (Grignani et al., 1998, Nature 391, 815-8; Guidez et al., 1998, Blood 91 , 2634-42; He et al., 1998, Nat Genet 18, 126-35; Lin et al., 1998, Nature 391, 811-4). 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).
By now, a clinical phase II trial with the closely related butyric acid derivative Pivanex (Titan Pharmaceuticals) as a monotherapy has been completed demonstrating activity in stage lll/IV non-small cell lung cancer (Keer et al., 2002, ASCO, Abstract No. 1253). More HDAC inhibitors have been identified, with NVP-LAQ824 (Novartis) and SAHA (Aton Pharma Inc.) being members of the structural class of hydroxamic acids tested in phase I clinical trials (Marks et al., 2001, Nature Reviews Cancer 1, 194-202). Another class comprises cyclic tetrapeptides, such as depsipeptide (FR901228 - Fujisawa) used successfully in a phase II trial for the treatment of T-cell lymphomas (Piekarz et al., 2001, Blood 98, 2865-8). Furthermore, MS-27-275 (Mitsui Pharmaceuticals), a compound related to the class of benzamides, is now being tested in a phase I trial patients with hematological malignancies.
In Int. J. Chem. Kinet. 1997, 29, 729-735 3-Cyclopentyl-N-hydroxy-propionamide, 4-Cyclohexyl-N-hydroxy-butyramide and 2-Cyclohexyl-N-hydroxy-acetamide are described (see also Berndt et al., 1992, Int. J. Chem. Kinet, 24, 695-701).
The crystal structure of a histone deacetylase like protein from the hyperthermophilic bacterium aquifex aeolicus cocrystallized with the two inhibitors TSA and SAHA is described by Finnin et al., 1999, Nature, 401, 188-193. Hydroxamic acids with at least one aromatic ring or ring system as histone deacetylase inhibitors are described by Lavoie et al.,' 2001 , Bioorg. Med. Chem.
Letters 11, 2847-2850; Remiszewski et al., 2002, J. Med. Chem. 45, 4, 753-757;
Massa et al., 2001 , J. Med. Chem. 44, 2069-2072; Stemson et al., 2001, Org. Lett.
3, 26, 4239-4242; Mai et al., 2002, J. Med. Chem. 45, 1778-1784; Woo et al., 2002,
J. Med. Chem. 45, 2877-2885.
In EP1174438, WO0052033, WO0118045, WO0118171, WO0138322, WO0170675,
WO9735990, W09911659, WO0226703, WO0230879 and WO0226696 hydroxamic acids as histone deacetylase inhibitors are described.
In WO9805635 and WO9533709 hydroxamic acids as matrix metalloproteinase inhibitors are described.
The compounds of the present invention are hydroxamic acids which are inhibitors of enzymes having histone deacetylase activity. Due to their HDAC-inhibitory activity they induce differentiation and/or apoptosis in a wide variety of cancer cells for three 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 and (3) clinical efficacy has been demonstrated for several other HDAC inhibitors unrelated to the presented compounds in the treatment of cancer patients.
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.
The histone deacetylase inhibitory activity of new compounds may be determined by a number of state-of-the-art technologies such as transcriptional repression assay, Western Blot analysis which detects acetylation of histone H3 and/or histone H4, or by an enzymatic in vitro assay. Histone deacetylase inhibitors can be further characterized for their cytotoxic and growth inhibitory effects on tumor cell lines and for their ability to modulate gene expression patterns in cells.
The present invention is directed to compounds of the general formula (I):
or pharmaceutical acceptable salts or physiologically functional derivatives thereof wherein:
n is a non-aromatic ring system containing two to six carbon atoms, wherein the ring system can contain one ore two double bonds;
X is C, CH or CH2;
Y is selected from C, CH, CH2, S, NR, CH2-CH2,
H2C- -CH HC- -CH2 j C- -CH2j H2C- -C or O -C; one or more of the hydrogen atoms can optionally be substituted by one or more substituents R ;
each of the dotted lines means a single, a double or triple bond with the exclusion of a combination of a triple with triple bond and a double with a triple bond;
Rx is independently H, -CN, alkyl, cycloalkyl, aminoalkyl, alkylamino, alkoxy, -OH, -SH, alkylthio, hydroxyalkyl, hydroxyalkylamino, halogene, haloalkyl, haloalkyloxy;
R is H, an alkyl or cycloalkyl group;
Z is CH, C, or P; p is 0 or 1 ; and
with the proviso that the following compounds are excluded:
An alkyl group, if not stated otherwise, is preferably a linear or branched chain of 1 to 6 carbon atoms, preferably a methyl, ethyl, propyl, isopropyl, butyl, f-butyl, isobutyl, pentyl or hexyl group, a methyl, ethyl, isopropyl or ---butyl group being most preferred.
The term "alkyl", unless otherwise noted, is also meant to include those derivatives of alkyl defined in more detail below as "unsaturated alkyl". An unsaturated alkyl group is one having one or more double bonds or triple bonds, preferably vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4- pentadienyl, 3-(1 ,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
The alkyl group in the compounds of formula (I) can optionally be substituted by one or more substituents K being as defined above.
An cycloalkyl group denotes a non-aromatic ring system containing 3 to 8 carbon atoms, wherein one or more of the carbon atoms in the ring can be replaced by a group X, X being as defined above.
An alkoxy group denotes an O-alkyl group, the alkyl group being as defined above.
An alkylthio group denotes an S-alkyl group, the alkyl group being as defined above.
A hydroxyalkyl group denotes an HO-alkyl group, the alkyl group being as defined above. An haloalkyl group denotes an alkyl group which is substituted by one to five preferably three halogen atoms, the alkyl group being as defined above; a CF3 being preferred.
An haloalkyloxy group denotes an alkoxy group which is substituted by one to five preferably three halogen atoms, the alkoxy group being as defined above; a OCF3 being preferred.
A hydroxyalkylamino group denotes an (HO-alkyl)2-N-group or HO-alkyl-NH- group, the alkyl group being as defined above.
An alkylamino group denotes an HN-alkyl or N-dialkyl group, the alkyl group being as defined above.
An aminoalkyl group denotes an H2N-alkyl, monoalkylaminoalkyl, or diaikylaminoalkyl group, the alkyl group being as defined above.
A halogen group is chlorine, bromine, fluorine or iodine, fluorine being preferred.
The invention also provides a pharmaceutical composition comprising a compound of formula (I) in free form or in the form of pharmaceutically acceptable salts and physiologically functional derivatives or prodrugs, together with a pharmaceutically acceptable diluent or carrier therefore.
The term "physiologically functional derivative" as used herein refers to compounds like ethers, esters, N-alkylated or acetylated hydroxamic acids, 2,5-dihydro-[1 , 2, 4] - oxodiazolyl or 4,5-dihydro-[1 , 2, 4]-oxodiazolyl, which are not pharmaceutically active themselves but which are transformed into their pharmaceutical active form in vivo, i.e. in the subject to which the compound is administered.
In another aspect, the present invention also provides a method for the treatment or prophylaxis of a condition where there is an advantage in inhibiting histone deacetylase activity which comprises the administration of an effective amount of a compound of formula (1) and physiologically acceptable salts or physiologically functional derivatives thereof.
The invention is also directed to the use of compounds of the formula (I) and of their pharmacologically tolerable salts or physiologically functional derivatives for the production of a medicament for the prevention and treatment of diseases, where inhibition of histone deacetylase is of benefit.
In addition, the present invention provides methods for preparing the desired hydroxamic acides of the formula (I).
One method for the synthesis of compounds of the formula (I) comprises the conversion of an acid (formula II) to the corresponding acid chloride (formula III) and reacting the acid chloride with hydroxylamine (Watanabe et al., 1989, J. Org. Chem., 54, 17, 4088-4097; Shishido et al., 1992, J. Org. Chem., 57, 10, 2876-2883).
formula (II)
formula (III)
formula (I)
Coupling reactions of acids of the formula (II) with hydroxylamine, other methods for the preparation of compounds of the formula (I), are described by Woo et al., 2002, J. Med. Chem. 45, 2877-2885; Knorr et al., 1989, Tetrahedron Lett., 30, 1927-1930, Carpino, 1993, J. Am. Chem. Soc, 115, 4397-4398 and Albericio et.al., 1998, J. Org. Chem., 63, 9678-9683.
Another method for the preparation of compounds of the formula (I) is the reaction of the corresponding ester with hydroxylamine as described by Stowell et al., 1995, J. Med. Chem., 38, 8, 1411-1413.
In one preferred embodiments in the compounds of formula (I) the ring n including Z can be cyclopentyl, cyclohexyl, cycloheptyl, cyclopent-1 -enyl, cyclohex-1 -enyl, cyclohept-1 -enyl, cyclopent-2-enyl, cyclohex-2-enyl, cyclohept-2-enyl, cyclohex-3- enyl, cyclohept-3-enyl.
In another preferred embodiment in the compounds of the formula (I) of the present invention the ring n including Z is cyclopentyl or cyclohexyl, and Y is selected from CH, CH2, CH2-CH2, S, NR or p = 0, and Z is CH or P.
In another more preferred embodiment in the compounds of the formula (I) of the present invention the ring n including Z is cyclopentyl or cyclohexyl, Y is selected from CH, CH2, CH2-CH2, or p = 0 and Z is CH.
In another more preferred embodiment, none of the carbon atoms of the alkyl groups is replaced by a group A.
Preferred compounds of the present invention are:
3-Cyclopentyl-N-hydroxy-propionamide;
3-Cyclohexyl-N-hydroxy-propionamide;
4-Cyclohexyl-N-hydroxy-butyramide;
2-Cycloheptyl-N-hydroxy-acetamide.
The compounds of the formula (I) according to the invention can form salts with inorganic or organic acids or bases. Examples of such salts are, for example, alkali metal salts, in particular sodium and potassium salts, or ammonium salts. The compounds of formula (I) may be obtained via various methods. In preferred embodiments of the methods of the invention the two following methods of synthesis are used.
Acids or acid chlorids of α-β unsaturated compounds of the formula (I) can be obtained by hydrolysis of the corresponding ester to result the acid (Bojic et al., 1998, 354, 289-299) which can be converted with chlorinating agents (oxalylchloride, thionylchloride, phosphorpenta chloride) into the acid chloride, α-β unsaturated esters can be synthesized from the corresponding aldehyd by reacting them with Carbethoxymethylene)triphenylphosphorane (PhP=CHCOOEt) (Maryanoff et al., 1989, Chem. Rev. 89, 863-927).
Other methods for preparing different acids are described by Mancuso et al., 1981, Synthesis, 165-185; or Bal et al., 1981 , Tetrahedron, 37, 2091-2096.
The compounds of the present invention can be used for a variety of human and animal diseases, preferably human diseases, where inhibition of histone deacetylase activity is beneficial.
Therefore the compounds according to the invention and medicaments prepared therewith are generally useful to induce the differentiation and/or apoptosis of cells such as undifferentiated tumor cells. The therapeutic effect of the invention may arise through one or more mechanisms, including but not limited to, the regulation of cell proliferation, cell activation, cell survival, cell differentiation, cell cycle, cell maturation and cell death or to induce systemic changes in metabolism such as changes in sugar, lipid or protein metabolism, the inhibition of new blood vessel formation (anti- angiogenesis), the inhibition of tumor spread into other organs (anti-metastatic), the inhibition of tumor spread into neighboring normal structures (anti-invasive) or the promotion of programmed cell death (apoptosis).
They can also be used to support cell generation poiesis, including blood cell growth and generation (prohematopoietic effect) after depletion or destruction of cells, as caused by, for example, toxic agents, radiation, immunotherapy, growth defects, malnutrition, malabsorption, immune dysregulation, anemia and the like or to provide li
a therapeutic control of tissue generation and degradation, and therapeutic modification of cell and tissue maintenance and blood cell homeostasis.
The compounds according to the invention and medicaments prepared therewith are also useful for the treatment of a disease which is associated with gene-specific hypoacetylation of histones or other molecules, such as p53. Additionally, the compounds of the present invention may also be used in the treatment of conditions where the suppression of anti-apoptotic genes, such as BCL-XL and other BCL family members, or the induction of tumor suppressor activity of molecules such as p21 and/or p53, is required.
The compounds according to the invention and medicaments prepared therewith are also suitable for the treatment of diseases 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 include but are not limited to, skin cancer, melanoma, estrogen receptor-dependent and independent breast cancer, ovarian cancer, testosteron receptor-dependent and independent prostate cancer, renal cancer, colon and colorectal cancer, pancreatic cancer, bladder cancer, esophageal cancer, stomach cancer, genitourinary cancer, gastrointestinal cancer, uterine cancer, astrocytomas, gliomas, basal cancer and squameous cell carcinoma, sarcomas as Kaposi's sarcoma and osteosarcoma, head and neck cancer, small cell and non-small cell lung carcinoma, leukemia, lymphomas and other blood cell cancers, Keratoakantoma, Bowen Disease, cutaneous T-Cell Lymphoma and also for the treatment of pre-malignant lesions (such as Actinic Keratose). The combinatorial treatment of the present invention is particularly useful for treating minimal residual tumor disease or tumor metastases.
Additionally, the invention may also be beneficial by reverting inappropriate gene expression in diseases based on aberrant recruitment of histone deacetylase activity such as thyroid resistance syndrome, or in other conditions associated with abnormal gene expression, such as inflammatory disorders, diabetes, thalassemia, cirrhosis, protozoal infection, or the like and all types of autoimmune diseases, in particular rheumatoid arthritis, rheumatoid spondylitis, all forms of rheumatism, osteoarthritis, gouty arthritis, multiple sclerosis, insulin dependent diabetes mellitus and non-insulin dependent diabetes, asthma, rhinitis, uveithis, lupus erythematoidis, ulcerative colitis, Morbus Crohn, inflammatory bowel disease, as well as other chronic inflammations, chronic diarrhea and of inflammations of the skin and/or mucosa (such as Psoriasis, lchtiosis, Acne). The invention also relates to the use for the protection from UV light and for the treatment of sun burn.
Furthermore, the compounds according to the invention and medicaments prepared therewith are useful for the treatment of other proliferative diseases such as psoriasis, fibrosis, warts and other dermatological disorders. The terms "proliferative disease", and "cell proliferation", are used interchangeably herein and relate to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or . hyperplastic growth, whether in vitro or in vivo. Examples of proliferative conditions include, but are not limited to, pre-malignant and malignant cellular proliferation, including malignant neoplasms and tumors, cancers, leukemias, psoriasis, bone disease, fibroproliferative disorders (e.g. of connective tissues), and atherosclerosis. Any type of cell may be treated, including but not limited to, lung, colon, breast, ovarian, prostate, liver, pancreas, brain, and skin and any treatment of disorders involving T-cells such as aplastic anemia and DiGeorge syndrome, Graves' disease.
The invention encompasses also compounds of formula (I) which are metabolized in patients to a compound of the presented formula. The embodiments described in this invention apply to such compounds as well.
The present invention also concerns a diagnostic method to identify tumors comprising the step of testing in vitro whether a tumor is responsive to treatment either with compounds of formula (I) or in combination with established tumor therapeutics. The method preferably comprises the method for the identification of genes regulated by these treatments. 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 combinatorial treatment using formulations of this invention and derivatives thereof 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.
In general the present invention provides novel possibilities to treat various human diseases. Applicants found that the HDAC inhibitory and cellular differentiation- inducing activity of compounds of formula (I) can be used successfully in combination with well established and clinically used therapeutic drugs for the treatment of tumor cells of different origins. Such compound based combinatorial treatment is considered to generate superior therapeutic success in human tumor patients than the corresponding therapeutic drugs used on their own. It is an object of the present invention to provide combinatorial therapeutic approaches using the presented compounds for the treatment of cancer. Such combinatorial treatments could result in a decrease of the therapeutic doses of e.g. chemotherapeutic reagents required and could thus limit the currently observed, partly very serious side effects of frequently used therapies.
Aspects of the present invention are the combination of compounds of formula (I) with, but not restricted to, therapeutic principles currently in clinical use or in clinical development, such as
Chemotherapeutic or cytotoxic drugs (e.g. 5-FU, taxol, cisPlatinum, camptothecin, gemcitabine, doxorubicine, irinothecan) - , differentiation inducing drugs (e.g. vitamin D, retinoic acid, cytokines such as
11-3, 11-6, SCF, G-CSF, GM-CSF, TNF)
Radiation therapy (e.g. x-rays or gamma rays) immunological approaches (antibody therapy, vaccination) combined immunotherapeutic/cytotoxic approaches (e.g. antibodies conjugated with cytotoxic components) anti-angiogenesis approaches.
The compounds and salts thereof can be formulated, as pharmaceutical compositions (e.g. liquids, suspensions, emulsions, lozenges, cachets, ampoules, suppositories, pessaries, ointments, gels, pastes, sprays, lotions, oils, boluses, electuaries, aerosols, powders, granules, tablets, pills, capsules, injections, solutions, foams, creams, 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 age, body weight, general health, sex, diet, and prior medication, and the severity of the particular disease of the patient, and the activity of specific compounds employed, time of administration, route of administration, rate of excretion, the duration of the treatment, other drugs, compounds, and/or materials used in combination. It will be appreciated that the appropriate dosage of the active compounds, and compositions comprising the active compounds, can vary from patient to patient. Determining the optimal dosage will generally involve balancing of the level of therapeutic benefit against any risk or deleterious side effects of the treatments of the present invention.
Administration in vivo can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosages of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
In general, a suitable dose of the active compound is in the range of about 0.1 to about 500 mg per kilogram body weight preferably 0.1 to 100 mg per kilogram body weight of the subject per day. Where the active ingredient is a salt, an ester, prodrug, or the like, the amount administered is calculated on the basis the parent compound and so the actual weight to be used is increased proportionately.
Figures
Figure 1 shows the relief of HDAC-dependent transcriptional repression in a reporter cell line, UAS TK-luc, after 24 hours treatment with Compound 1 , 2 and 3. Depicted is the fold induction of luciferase expression after treatment of cells at the indicated concentrations of Compounds 1 , 2 and 3.
Figure 2 shows the induction histone H4 hyperacetylation in 293T cells after treatment with Compounds 1 , 2 and 3 for six hours at the indicated concentrations.
Figure 3 shows the inhibition of in-vitro HDAC activity in nuclear extracts from HeLa cells after treatment with Compounds 1, 2 and 3 at the indicated concentrations. Relative HDAC activity is shown as percent of activity of untreated nuclear extracts.
Figure 4 shows the inhibition of in-vitro HDAC activity of recombinant HDAC1 , HDAC6, and HDAC8 enzymes after treatment with Compound 3 at the indicated concentrations. Relative HDAC activity is shown as percent of activity of untreated HDAC enzymes.
Figure 5 shows on the protein level the down-regulation of the anti-apoptotic molecule BCL-XL and up-regulation of the cell cycle inhibitor p21 in K562 cells induced after treatment with Compounds 1 , 2 and 3 for 36 hours at the indicated concentrations.
Figure 6 shows the concentration of Compounds 1, 2 and 3 inducing a 50% reduction in cellular biomass (IC50) after treatment of various cancer cell lines for 72 hours.
Figure 7 shows the induction of apoptosis in BV-173 cells after treatment for 24 hours with Compounds 1, 2 and 3 at the indicated concentrations. Apoptosis is demonstrated by the increase of a sub-G1 population in Propidium Iodine treated cells, which is reduced to baseline levels after treatment with a pan-caspase inhibitor Z-VAD. EXAMPLES Example 1
Synthesis of Compound 1 (3-Cyclopentyl-N-hvdroxy-propionamide)
To a solution of 3-Cyclopentyl-propionyl chloride (11.0 mL, 11.6 g, 72.0 mmol) in dichloromethane (50 mL) hydroxylamine hydrochloride (5.00 g, 72.0 mmol) and sodium bicarbonate (12.0 g, 144 mmol) were added. After stirring for 24 h at room temp, the reaction was quenched by addition of saturated ammonium chloride solution (50 mL). The layers were separated. The water layer was extracted with ethyl acetate (4 x 50 mL). The solvent of the combined organic layers was removed in vacuo. Precipitation of the crude product out of ethyl acetate by adding petroleum ether yielded the hydroxamate (5.25 g, 33.4 mmol, 46%) as a white solid. 1H NMR (300 MHz, [Dβ-DMSO]: δ = 0.74-0.91 (m, 2 H), 1.05-1.25 (m, 4 H), 1.36 (q, J = 7.4
Hz, 4 H), 1.54-1.71 (m, 5 H), 1.94 (t, J = 7.6 Hz, 2 H), 8.59 (s, 1 H) and 10.28 (s, 1 H) (NH and OH). 13C NMR (75 MHz, [Dβ-DMSO]: δ = 25.6, 26.0, 29.7, 32.4, 36.5, 169.2
(C(=0)NHOH). MS: m/z calcd for (C8H14N02) [M+H]+ 158; found 158.
Example2:
Synthesis of Compound 2 (3-Cvclohexyl-N-hvdroxy-propionamide): To a solution of 3-Cyclohexyl-propionyl chloride (12.1 mL, 12.6 g, 72.0 mmol) in dichloromethane (50 mL) hydroxylamine hydrochloride (5.00 g, 72.0 mmol) and sodium bicarbonate (12.0 g, 144 mmol) were added. After stirring for 24 h at room temperature the reaction was quenched by addition of saturated ammonium chloride solution (50 mL). The layers were separated. The water layer was extracted with ethyl acetate (4 x 50 mL). The solvent of the combined organic layers was removed in vacuo. Precipitation of the crude product out of ethyl acetate by adding petroleum ether yielded the hydroxamate (7.21 g, 42 mmol, 58%) as a white solid. 1H-NMR (300 MHz, [D6-DMSO]: δ = 0.95-1.12 (m, 2 H), 1.38-1.69 (m, 6 H), 1.62-1.77 (m, 3 H), 1.94 (t, J = 7.6 Hz, 2 H), 8.58 (s, 1 H) and 10.28 (s, 1 H) (NH and OH); 13C-NMR (75 MHz, [D6-DMSO]: δ = 25.0, 31.8, 32.0, 32.3, 39.5, 169.5 (C(=)NHOH); MS: m/z calc, for (C9H17N02) [M+H]+ 172; found 172. -e 3:
Synthesis of Compound 3 (3-Cvclohexyl-N-hvdroxy-acrylamide)
3-Cyclohexyl-acrylic acid ethyl ester: to a cooled solution of oxalyl chloride (11.1 mL, 105 mmol) in dichloromethane (250 mL) was added at -78 °C a solution of dimethylsulfoxide (14.6 mL, 206 mmol) in dichloromethane (250 mL). After 5 min at the same temperature cyclohexylmethanol (10.8 mL, 87.5 mmol) and after additional 5 min triethylamine (60.7 mL, 438 mmol) were added. The reaction remained at -78 °C for two hours and then let warm to room temperature. The solvent was removed in vacuo. The resulting aldehyde was used for the next step without further purification. The crude cyclohexanecarbaldehyde was dissolved in toluene (200 mL) and ethanol (150 mL). After 15 min of stirring at 70 °C carbethoxy-methylene triphenylphosphorane (33.6 g, 96.5 mmol) was added in one portion. Stirring was continued for additional 24 hours. The solvent was removed in vacuo. The product was obtained by flash chromatography in 78 % yield (12.5 g). 1H NMR (300 MHz, [Dβ-DMSO]: δ = 1.07-1.34 (m, 8 H, CH3 and cyclohexyl-CH ), 1.60-1.80 (m, 5 H, cyclohexyl-CH2), 2.04-2.18 (m, 1 H, cyclohexyl-CH); 4.16 (q, J = 7.1 Hz, 2 H, Et- CH2), 5.74 (dd, J = 15.8 Hz and J = 1.5 Hz, 1 H, C=C-H), 6.89 (dd, J = 15.8 Hz and J = 6.8 Hz, 1 H, C=C-H).
3-Cyclohexyl-acrylic acid: to a solution of 3-Cyclohexyl-acrylic acid ethyl ester (7.00 g, 38.4 mmol) in dioxane (200 mL) was added lithium hydroxide (4.03 g, 96.1 mmol) dissolved in water (70 mL). Because LiOH fell out methanol (170 mL) was added to the reaction. After 4 hours at 70 °C 3/4 of the solvent was removed in vacuo. 2 N HCI was added until the pH reached 3. The mixture was treated with ammonium chloride (150 mL) saturated solution. The product was extracted with dichloromethane (200 mL x 3). The combined organic phases were washed with saturated sodium solution (50 mL) and dried with magnesium sulfate. After filtration the solvent was removed in vacuo. The liquid was left at room temperature for 48 hours for crystallization to occur. The colourless crystalline product (2.01 g, 13.1 mmol) was obtained in 34% yield. 1H NMR (300 MHz, [D6-DMSO]: δ = 1.01 -1.37 (m, 5 H, cyclohexyl-CH2), 1.55- 1.79 (m, 5 H, cyclohexyl-CH2), 2.06-2.21 (m, 1 H, cyclohexyl-CH); 5.69 (dd, J = 15.7 Hz and J = 1.5 Hz, 1 H, C=C-H), 6.76 (dd, J = 15.7 Hz and J = 6.8 Hz, 1 H, C=C- H).12.01 (broad s, 1 H, OH). 3C NMR (75 MHz, [D6-DMSO]: δ = 25.1 , 25.4, 31.1 (cyclohexyl-CH2), 39.4 (cyclohexyl-CH), 119.5 and 153.4 (C=C) 167.3 (COOH). MS: m/z calcd for (C9402) [M+H]+ 155; found 155.
3-Cyclohexyl-N-hydroxy-acrylamide (K1 00003283): to a solution of 3-Cyclohexyl- acrylic acid (5.30 g, 31.3 mmol) in DMF (100 mL) was added EDC (1-(3- Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride) (7.80 g, 40.7 mmol) and HOBt (1 -hydroxybenzotriazole hydrate) (5.07 g, 37.6 mmol). After stirring for 30 min at room temperature hydroxylamine hydrochloride (3.26 g, 47.0 mmol) and triethylamine (6.94 mL, 50.1 mmol) were added. After additional 18 hours stirring at room temperature the precipitate was filtered and washed with DMF. The DMF phases were combined. The solvent was concentrated in vacuo. 1 N HCI (50 mL) was added. After extraction with ethyl acetate (3 x 100 mL) and dichloromethane (2 x 100 mL) the organic layers were combined, washed with 1 N HCI (10 mL) and sodium chloride saturated solution (10 mL) and dried with magnesium sulfate. The solvent was removed in vacuo. The product was crystallized from ethyl acetate by addition of petroleum ether. This step removed large amounts of HOBt, the substance was then further purified by preparative HPLC using an acetonitrile / water gradient. This yielded (1.54 g, 9.08 mmol) of 3-Cyclohexyl-N-hydroxy-acrylamide. 1H NMR (300 MHz, [D6-DMSO]: δ = 0.99-1.35 (m, 5 H) and 1.56-1.73 (m, 5 H)
(cyclohexyl-CH2), 2.00-2.13 (m, 1 H, cyclohexyl CH), 5.67 (d, J = 15.7 Hz, 1 H, C=C- H), 6.58 (dd, J = 15.7 and J = 6.3 Hz, 1 H, C=C-H), 8.77 (s, 1 H) and 10.49 (s, 1 H) (NH and OH). 13C NMR (75 MHz, [D6-DMSO]: δ = 25.2, 25.5, 31.5 (cyclohexyl-CH2), 41.1 (cyclohexyl-CH), 118.8 and 147.1 (C=C), 162.8 (C(=0)NHOH). MS: m/z calcd for (C95N02) [M+H]+ 170; found 170.
Example 4
Compounds 1, 2 and 3 inhibit HDAC activity, induce histone H4 hyperacetylation, and relieve transcriptional repression in a reporter cell line (Fig. 1 to 4). Histone hyperacetylation correlates with an open, de-condensed chromatin structure and gene activation, while hypoacetylation correlates with chromatin condensation and transcriptional repression. 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. Transcription factors such as thyroid hormone receptor, PPARδ, retinoic acid receptor, N-CoR and AML/ETO recruit
HDAC enzymes and thereby repress transcription when bound to a specific promoter region. This latter effect can be reenacted with a constitutive promoter containing UAS elements which recruits fusion proteins containing the heterologous DNA- binding domain of the yeast Gal4 protein fused to one of the above mentioned transcription factors. 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.
Methods
Transcriptional reporter gene assay. The transcriptional assay for repressor activity exploits activation and derepression of a Gal4-dependent reporter gene (Hildebrand et al., 2001, J Biol Chem 276, 9889-95 ; Maurer et al., 2002, Blood 99, 2647-52). This assay is performed with specifically constructed permanent cell lines. 293T cell were stably transfected with a UAS TK lucif erase reporter plasmid (Heinzel et al., 1997 Nature 387, pp43-48) and an expression plasmid for the Gal4 DNA binding domain fused to an HDAC-dependent repressor molecule. While Gal4 fusion proteins repress this activity, HDAC inhibitors induce relief of this repression which can be detected as an increase in reporter gene activity (e.g. by luciferase activity detection assay).
Induction of histone hyperacetylation. These acetylated histones can be detected by Western Blot analysis of whole cell extracts from histone deacetylase inhibitor- treated 293T cells using antibodies specific for the acetylated N-terminal lysine residues of histones H4 (Gόttlicher et al., 2001 , EMBO J 20, 6969-78).
In vitro inhibition of recombinant HDAC's. The determination of histone deacetylase activity in HeLa nuclear extracts or recombinant HDAC proteins from Highδ insect cells is based on the specific deacetylation of an artificial substrate (Fluor de Lys, Biomol). The substrate turn over may be detected and quantified by fluorometry. By addition of a potential HDAC inhibitor the hydrolysis of the substrate is constrained resulting in a decreased fluorometric signal. IC5o values may be calculated from dose-response curves.
The assay is separated in two steps: in the first step the substrate (Fluor de Lys / Biomol KI-104) is hydrolysed by histone deacetylases. In step two HDAC activity is terminated and the fluorophore is activated by the addition of a developer (Developer / Biomol KI-105). Nuclear extracts from HeLa cells (Biomol KI-140) or recombinant proteins and the potential HDAC inhibitor are mixed with reaction buffer (Biomol Kl- 143) to a total volume of 25μl per well of a 96 well plate. 25μl substrate (1 :100 dilution in reaction buffer) per well are added to start the reaction. A negative control without histone deacetylase activity and a positive control without HDAC inhibitor are treated likewise. The reaction is stopped after 15-60min. by adding 50μl developer (1 :20 dilution in reaction buffer). After another 15min. incubation time at room temperature the fluorescence signal is stable for 60min and may be detected by a fluorescence reader (excitation filter: 390nm, emission filter: 460nm).
Results
As can be seen in Figure 1 , the Gal4 fusion protein represses the baseline activity of the TK promoter and the subsequent luciferase expression. Addition of Compounds 1 , 2 and 3 relieve this repression as measured by increased expression and activity of luciferase in lysates of treated cells after 24 hours. Both, Compound 1 and 2 induce luciferase reporter gene expression after stimulation of cells for 24 hours starting at a concentration of 8 μM (2-3 fold) with a maximum induction of more than 20 fold at 200 μM. Compound 3, however, induces expression of reporter gene already at a concentration of 1.6 μM with a maximum induction of almost 40 fold at 40 μM.
Since histone deacetylase inhibitors shift the enzymatic balance between histone acetyl transferases (HAT's) and histone deacetylases (HDAC's) towards HAT's by blocking HDAC's, they induce the accumulation of N-terminally hyperacetylated histones H4. This can be seen in Figure 2 by the induction of histone hyperacetylation after treatment of 293T cells for 6 hours with Compounds 1 , 2 and 3. Hyperacetylation can be seen with Compounds 1 and 2 at minimum concentrations of 1 ,6 μM with a maximum induction. at 40 μM, whereas compound 3 induces hyperacetylation starting at 320 nM. Since most HDAC's reside in the nucleus, highest HDAC activity can be measured in nuclear extracts. As demonstrated in Figure 3, HDAC activity in nuclear extracts is dramatically reduced in the presence of Compounds 1 , 2 and 3, with a 50% reduction of HDAC activity at a concentration of 14.7 μM for Compound 1 , at 6.1 μM for Compound 2, and at 0.9 μM for Compound 3.
Results obtained with Compound 3 in nuclear extracts could be confirmed with recombinant HDAC's. As demonstrated in Figure 4, HDAC activity of recombinant HDAC1 , HDAC6, and HDAC8 is dramatically reduced in the presence of Compound 1 and 2, with a 50% reduction of HDAC activity at a concentration of 0.3 to 0.4 μM for
Compound 3.
Example 5
Induction of growth arrest, apoptosis and down-regulation of BCL-X after treatment of cancer cells with Compounds 1 , 2 and 3. The treatment of tumor cell lines with histone deacetylase inhibitors leads to histone- hyperacetylation and the transcriptional regulation of target genes. Although the discrete mechanism of action varies, cancer therapy still depends on an ability to engender apoptosis in cancer cells as a final common pathway. HDAC inhibitors have already been shown to induce apoptosis in certain cancer cells through down-regulation of the anti-apoptotic molecules, such as BCL-XL and BCL-2. Anti-apoptotic BCL2 family members seem to be involved in resistance of tumors to apoptosis. For example, high expression of BCL-XL is found in many human cancers and is often a negative prognostic factor. Accordingly, downregulation of BCL-XL expression in certain cancer cells either induces apoptosis directly or sensitizes cells to apoptotic stimuli. In general, induction of apoptosis can be exploited therapeutically using HDAC inhibitors in cancer therapy
Methods
Protein expression profiling. The expression pattern induced by histone deacetylase inhibitors can be monitored by Western Blot analysis with antibodies against p21 and BCL-XL using whole cell extracts of cells treated with the respective compounds at the indicated concentrations. Modulation of protein expression is exemplified by the induction of expression of the cell cycle inhibitor/tumor suppressor p21 and the suppression of expression of the anti-apoptotic molecule BCL-XL.
Growth inhibiton of tumor cell lines. The reduction in cellular biomass was measured by SRB-assay. For this assay cells were seeded in 96 well culture dishes at densities between 3000 and 8000 cells per well. After recovery of 24 hours cells were cultured for 48-72 hours in the absence or presence of the indicated concentrations of compounds. Synergistic reduction in total cellular biomass was assayed through SRB-assay by adding the compounds in the medium at the indicated concentrations and cultivate the cells for further 48 hours.
Cells were fixed with cold Trichloracetat (TCA) producing a final TCA concentration of 10%. After 1 hour of incubation at 4°C the cells were washed five times with water and air dried. Fixed cells were stained for 30 minutes with 0,4% (wt/vol) Sulforhodamine B (SRB) 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. Optical densities (OD) were read on a Molecular Devices Versa Max tunable microplate reader at 520-550 nm. Four test wells for each dose-response were set in parallel with 12 control wells per cell line. Measurement of the cell population density at time 0 (T0; the time at which the drug was added) was also made from 12 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 from each microtiter plate 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 cellular biomass of the cells.
Measurement of Apoptosis. FACS analysis of the cell cycle by propidium iodide (PI) staining. The cell cycle can be divided into four different sections: During G0/1 -phase cells are in senescence or proliferate, in S-phase cells start replicating their DNA, and during G2- and M-phase cells undergo mitosis. The cellular DNA content correlates with cell cycle progression: while cells in G01 phase possess one set of chromosomes, cells in G2- and M-phase possess two full sets of chromosomes. Cells in S-phase are still replicating their DNA and therefore exhibit a DNA content between one and two sets of chromosomes. Degradation of DNA is a marker for apoptosis. The sub-G1 area indicates the hypodiploid DNA peak corresponding to cells with fragmented DNA undergoing apoptosis Using a dye that intercalates with DNA (such as propidium iodide - PI) the cellular DNA content was determined. Between 5x105 - 1x106 cells were seeded out in cell culture dishes and treated for the desired time with the desired amount of test compound. After the incubation cells were harvested, washed in cold PBS, and the cell pellet was resuspended in 1ml 70% ethanol (-20°C). These cell pellets may be shelved for several month. For PI staining 3ml cold sodium citrate solution (38mM, pH 7,4) was added, cells were pelleted and stained with 500μl sodium citrate solution (38mM, pH 7,4) containing 50μg/ml PI and 5μg/ml RNase. After a 30 minute incubation time at 37°C in darkness, cells were analyzed via FACS analysis. The sub-Grpeak constitutes the amount of apoptotic cells.
Results
The expression of the growth arrest signaling tumor suppressor protein p21 (p21/waf) and the anti-apoptotic protein BCL-XL was analyzed uppn treatment with Compounds 1, 2 and 3. A down-regulation of the latter is regarded as one important pre-requisite for the induction of apoptosis. Figure 5 shows that Compounds 1 , 2 and 3 in fact induces increased protein expression levels of p21 and at the same time down- regulates BCL-XL protein levels. Both, Compound 2 and 3 induce p21 expression and BCL-XL downregulation at 8 μM, whereas Compound 1 induces p21 expression at 40 μM and BCL-XL downregulation at 200 μM.
Growth of various human tumor cell lines of breast, colon, pancreas and prostate origin were inhibited by Compounds 1, 2, and 3 (Figure 6). The concentrations inducing 50% growth arrest ranged from 8 to 70 μM (mean 33 μM) for Compound 1 , from 5 to 26 μM (mean 15 μM) for Compound 2, and from 0.5 to 8 μM (mean 2.6 μM) for Compound 3.
Studies using the BV-173 cell line consolidated the activity of Compound 1 and 2 as inducers of programmed cell death (apoptosis). Examples of this type of analysis are presented in Figure 7. The sub-G1 area (M1) indicates the hypodiploid DNA peak corresponding- to cells with fragmented DNA undergoing apoptosis, which is dramatically increased to 50 - 60 % in cells treated with Compound 1 at 100 μM, Compound 2 at 50 μM and Compound 3 at 5 μM. This apoptotic process is however completely blocked in the presence of a pan-caspase inhibitor, Z-VAD, which again confirms the induction of apoptosis by Compounds 1 , 2, and 3.

Claims

Claims
1. Compounds of the general formula (I)
or pharmaceutical acceptable salts or physiologically functional derivatives thereof wherein: n is a non-aromatic ring system containing two to seven carbon atoms, wherein the ring system can contain one ore two double bonds; X is C, CH or CH2; Y is selected from C, CH, CH2, S, NR, CH2-CH2,
H C- -CH> HC- -CH2j C- -CH2j H2C- -Cj0r O -C; one or more of the hydrogen atoms can optionally be substituted by one or more substituents R^; each of the dotted lines means a single, a double or triple bond with the exclusion of a combination of a triple with triple bond and a double with a triple bond;
R" is independently H, -CN, alkyl, cycloalkyl, aminoalkyl, alkylamino, alkoxy, -OH, -SH, alkylthio, hydroxyalkyl, hydroxyalkylamino, halogene, haloalkyl, haloalkyloxy;
R is H, an alkyl or cycloalkyl group;
Z is CH, C, or P;
p is 0 or 1 ; and
with the proviso that the following compounds are excluded:
2. The compound of claim 1 , wherein n = cyclopentyl or cyclohexyl.
3. The compound of claim 1 , wherein n = cyclopentyl or cyclohexyl and Z is CH.
4. A pharmaceutical composition comprising a compound as defined in any of claims 1 to 3 in free form or in the form of pharmaceutically acceptable salts or physiologically functional derivatives.
5. A compound according to any of claims 1 to 3, including the compounds excluded in claim 1 for the use as a medicament.
6. The use of a compound according to claim 5 in the manufacture of a medicament for use in treatment of a disease or a therapeutic indication as inhibitors of enzyms having histone deacetylase activity.
7. The use of a compound according to claim 5 in the manufacture of a medicament for use in treatment of a disease or a therapeutic indication in which inhibition of histone deacetylase activity is beneficial.
8. The use according to claim 6 where the human histone deacetylase is selected from the group consisting of HDACs 1-10 or a member of the SIR2 protein family.
9. The use of a compound according to claim 5 in the manufacture of a medicament for the induction of differentiation of cells.
10. The use of a compound according to claim 5 in the manufacture of a medicament for the induction of differentiation of transformed cells.
11. The use of a compound according to claim 5 in the manufacture of a medicament for the induction of apoptosis of transformed cells.
12. The use of a compound according to claim 5 in the manufacture of a medicament for the inhibition of proliferation of transformed cells.
13. The use of a compound according to claim 1 for the inhibition of histone deacetylase activity.
14. The use of a compound according to claim 5 in the manufacture of a medicament, in which the induction of hyperacetylation of histones has a beneficial effect.
15. The use of a compound according to claim 5 in the manufacture of a medicament for use in treatment of a disease or a therapeutic indication selected from the group consisting of skin cancer, melanoma, 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, leukemias and other types of blood cell cancer and endocrine disease based on aberrant
recruitment of histone deacetylase such as thyroid resistance syndrome. i
16. The use of a compound according to claim 5 in the manufacture of a medicament for use in the inhibition of abnormal gene expression such as inflammatory disorders, diabetes, thalassemia, cirrhosis or protozoal infection.
17. A process for the preparation of a compound according to claim 1 which comprises the step of reacting an acid of formula (II)
formula (ll)
(wherein n, X, Y, Z, and p are as defined in ciaim 1) or an acid chloride of formula (III)
formula (III)
(wherein n, X, Y, Z, and p are as defined in claim 1) with hydroxylamine.
EP03765029A 2002-07-23 2003-07-17 Novel compounds as histone deacetylase inhibitors Withdrawn EP1523470A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
DE10233412A DE10233412A1 (en) 2002-07-23 2002-07-23 New compounds as histone deacetylase inhibitors
DE10233412 2002-07-23
PCT/EP2003/007794 WO2004009536A1 (en) 2002-07-23 2003-07-17 Novel compounds as histone deacetylase inhibitors

Publications (1)

Publication Number Publication Date
EP1523470A1 true EP1523470A1 (en) 2005-04-20

Family

ID=30128280

Family Applications (1)

Application Number Title Priority Date Filing Date
EP03765029A Withdrawn EP1523470A1 (en) 2002-07-23 2003-07-17 Novel compounds as histone deacetylase inhibitors

Country Status (6)

Country Link
EP (1) EP1523470A1 (en)
JP (1) JP4644829B2 (en)
AU (1) AU2003254371A1 (en)
CA (1) CA2491131C (en)
DE (1) DE10233412A1 (en)
WO (1) WO2004009536A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7884105B2 (en) 2005-10-27 2011-02-08 Janssen Pharmaceutica, N.V. Squaric acid derivatives as inhibitors of histone deacetylase
US8101616B2 (en) 2006-01-19 2012-01-24 Janssen Pharmaceutica N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
US8138198B2 (en) 2005-05-18 2012-03-20 Angibaud Patrick Rene Substituted aminopropenyl piperidine or morpholine derivatives as novel inhibitors of histone deacetylase
US8193205B2 (en) 2004-07-28 2012-06-05 Janssen Pharmaceutica N.V. Substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylase
US8268833B2 (en) 2002-03-13 2012-09-18 Janssen Pharmaceutica, N.V. Inhibitors of histone deacetylase

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ534831A (en) 2002-03-13 2007-01-26 Janssen Pharmaceutica Nv Carbonylamino-derivatives having histone deacetylase (HDAC) inhibiting enzymatic activity
DE60321667D1 (en) 2002-03-13 2008-07-31 Janssen Pharmaceutica Nv Piperazinyl-, piperidinyl- und morpholinylderivate als neue inhibitoren von histon-deacetylase
NZ534771A (en) 2002-03-13 2006-04-28 Janssen Pharmaceutica Nv Sulfonylamino-derivatives as novel inhibitors of histone deacetylase
RS51189B (en) 2004-07-28 2010-10-31 Janssen Pharmaceutica N.V. Substituted propenyl piperazine derivatives as novel inhibitors of histone deacetylase
NZ599464A (en) 2005-02-03 2014-03-28 Topotarget Uk Ltd Combination therapies using hdac inhibitors
US8835501B2 (en) 2005-05-13 2014-09-16 Topotarget Uk Limited Pharmaceutical formulations of HDAC inhibitors
EP1904452A2 (en) 2005-07-14 2008-04-02 Takeda San Diego, Inc. Histone deacetylase inhibitors
AU2006313517B2 (en) 2005-11-10 2013-06-27 Topotarget Uk Limited Histone deacetylase (HDAC) inhibitors (PXD101) for the treatment of cancer alone or in combination with chemotherapeutic agent
AU2007206950B2 (en) 2006-01-19 2012-02-02 Janssen Pharmaceutica N.V. Substituted indolyl-alkyl-amino-derivatives as inhibitors of histone deacetylase
DK1979328T3 (en) 2006-01-19 2013-03-25 Janssen Pharmaceutica Nv Pyridine and pyrimidine derivatives as histone deacetylase inhibitors
EP1979326B1 (en) 2006-01-19 2012-10-03 Janssen Pharmaceutica N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase
JP5225104B2 (en) 2006-01-19 2013-07-03 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Aminophenyl derivatives as new inhibitors of histone deacetylase
CN101374828B (en) 2006-01-19 2012-09-19 詹森药业有限公司 Heterocyclylalkyl derivatives as novel inhibitors of histone deacetylase
CN101868446A (en) 2007-09-25 2010-10-20 托波塔吉特英国有限公司 The synthetic method of some hydroxamic acid compound
WO2009118370A1 (en) 2008-03-27 2009-10-01 Janssen Pharmaceutica Nv Aza-bicyclohexyl substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylase
CN102659630B (en) * 2011-05-04 2016-01-06 成都地奥九泓制药厂 A kind of hydroxamic acid compound and its production and use
TWI650139B (en) * 2017-10-11 2019-02-11 行政院原子能委員會核能硏究所 Hydroxamic acid-containing contrast agent containing radioactive isotope fluorine, preparation method thereof and use thereof
KR102236356B1 (en) * 2017-11-24 2021-04-05 주식회사 종근당 Compositions for Preventing or Treating Lupus
KR20190118251A (en) * 2018-04-10 2019-10-18 주식회사 종근당 Compositions for Preventing or Treating Dry Eye

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS50121218A (en) * 1974-03-01 1975-09-23
US3994997A (en) * 1975-05-09 1976-11-30 Gulf Oil Corporation O,O-diethyl-O-carboxamidophosphate esters
US4604407A (en) * 1985-04-04 1986-08-05 E. R. Squibb & Sons, Inc. Hydroxamates
JPH01221371A (en) * 1988-02-29 1989-09-04 Hokko Chem Ind Co Ltd Production of cyclic oxyamine derivative
US6043389A (en) * 1997-03-11 2000-03-28 Mor Research Applications, Ltd. Hydroxy and ether-containing oxyalkylene esters and uses thereof
JP4269041B2 (en) * 1999-03-02 2009-05-27 国立大学法人九州工業大学 Novel cyclic tetrapeptide derivatives and their pharmaceutical uses
JP2001081031A (en) * 1999-08-30 2001-03-27 Schering Ag Benzamide derivative-containing preparation having improved solubility and oral adsorption
ATE489360T1 (en) * 2000-03-24 2010-12-15 Methylgene Inc HISTONE DEACETYLASE INHIBITORS

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2004009536A1 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8524711B2 (en) 2002-03-13 2013-09-03 Janssen Pharmaceutica N.V. Amino-derivatives as novel inhibitors of histone deacetylase
US8697717B2 (en) 2002-03-13 2014-04-15 Janssen Pharmaceutica N.V. Inhibitors of histone deacetylase
US9556161B2 (en) 2002-03-13 2017-01-31 Janssen Pharmaceutica Nv Inhibitors of histone deacetylase
US9533979B2 (en) 2002-03-13 2017-01-03 Janssen Pharmaceutica Nv Amino-derivatives as novel inhibitors of histone deacetylase
US8268833B2 (en) 2002-03-13 2012-09-18 Janssen Pharmaceutica, N.V. Inhibitors of histone deacetylase
US8343988B2 (en) 2002-03-13 2013-01-01 Janssen Pharmaceutica, N.V Inhibitors of histone deacetylase
US9150560B2 (en) 2002-03-13 2015-10-06 Janssen Pharmaceutica Nv Inhibitors of histone deacetylase
US8455498B2 (en) 2002-03-13 2013-06-04 Janssen Pharmaceutica N.V. Inhibitors of histone deacetylase
US8916554B2 (en) 2002-03-13 2014-12-23 Janssen Pharmaceutica, N.V. Amino-derivatives as novel inhibitors of histone deacetylase
US9150543B2 (en) 2004-07-28 2015-10-06 Janssen Pharmaceutica N. V. Substituted indolyl alkyl amino derivatives as inhibitors of histone deacetylase
US8193205B2 (en) 2004-07-28 2012-06-05 Janssen Pharmaceutica N.V. Substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylase
US9636341B2 (en) 2004-07-28 2017-05-02 Janssen Pharmaceutica N.V. Substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylase
US8592441B2 (en) 2004-07-28 2013-11-26 Janssen Pharmaceutica N.V. Substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylase
US8524728B2 (en) 2004-07-28 2013-09-03 Janssen Pharmaceutica N.V. Substituted indolyl alkyl amino derivatives as novel inhibitors of histone deacetylase
US8138198B2 (en) 2005-05-18 2012-03-20 Angibaud Patrick Rene Substituted aminopropenyl piperidine or morpholine derivatives as novel inhibitors of histone deacetylase
US8377935B2 (en) 2005-05-18 2013-02-19 Janssen Pharmaceutica N.V. Substituted aminopropenyl piperidine or morpholine derivatives as novel inhibitors of histone deacetylase
US7884105B2 (en) 2005-10-27 2011-02-08 Janssen Pharmaceutica, N.V. Squaric acid derivatives as inhibitors of histone deacetylase
US8101616B2 (en) 2006-01-19 2012-01-24 Janssen Pharmaceutica N.V. Pyridine and pyrimidine derivatives as inhibitors of histone deacetylase

Also Published As

Publication number Publication date
JP2005533840A (en) 2005-11-10
AU2003254371A1 (en) 2004-02-09
CA2491131A1 (en) 2004-01-29
JP4644829B2 (en) 2011-03-09
DE10233412A1 (en) 2004-02-12
CA2491131C (en) 2013-05-07
WO2004009536A1 (en) 2004-01-29

Similar Documents

Publication Publication Date Title
CA2491131C (en) Novel compounds as histone deacetylase inhibitors
CN110088105B (en) Small molecule inhibitors of JAK family kinases
US9115090B2 (en) Zn2+-chelating motif-tethered short-chain fatty acids as a novel class of histone deacetylase inhibitors
CN101528677B (en) Hdac inhibitors
US8227516B2 (en) Compounds as histone deacetylase inhibitors
JPWO2003070691A1 (en) N-hydroxycarboxamide derivatives
US20090176837A1 (en) Compounds with activity at retinoic acid receptors
KR20030077551A (en) Tetralone derivatives as antitumor agents
US11578058B2 (en) Heterocyclic compounds for inhibiting TYK2 activities
HUE033281T2 (en) p38 MAP KINASE INHIBITORS
WO2000015603A1 (en) Benzene derivatives and medicinal use thereof
CA3119912A1 (en) A crystalline spirocyclic compound inhibitor of tryptophan hydroxylase 1 (tph1) for treating diseases or disorders associated with peripheral serotonin
CN104010501A (en) Agpnistis of SRC homology-2 containing protein tyrosine phosphatase-1 and treatment methods using the same
CN107162921B (en) Phenoxyacetic acid derivatives, preparation method thereof and application thereof as medicines
EP3301085A1 (en) Retinoid derivatives with antitumor activity
CN101883487A (en) Acid mimic compounds for the inhibition of isoprenyl-S-cysteinyl methyltransferase
JP7254703B2 (en) ROR gamma modulators and uses thereof
EA022196B1 (en) 4-SUBSTITUTED-3-BENZYLOXY-BICYCLO[3.1.0]HEXANE COMPOUNDS AS mGluR 2/3 ANTAGONISTS
JP2020505412A (en) ROR gamma modulator and use thereof
CN109602734A (en) For treating the Compounds and methods for of leukaemia
EA023171B1 (en) Thio derivatives bearing lactams as potent hdac inhibitors and their uses as medicaments
CN102477001A (en) Benzamide histone deacetylase inhibitor
CN107556315B (en) Imidazole derivatives containing four-membered rings
CN113149867B (en) Curcumin histone deacetylase inhibitor, and preparation method and medical application thereof
EP1140058A1 (en) N-formyl hydroxylamine derivatives as antibacterial agents

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20041228

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

DAX Request for extension of the european patent (deleted)
RIN1 Information on inventor provided before grant (corrected)

Inventor name: VINCEK, ADAM-SPENCER

Inventor name: KRAUSS, ROLF

Inventor name: KRAUS, JUERGEN

Inventor name: GASSEN, MICHAEL

Inventor name: HENTSCH, BERND

Inventor name: MARTIN, ELKE

Inventor name: HOEVELMANN, SASCHA

Inventor name: MAURER, ALEXANDER, B.

17Q First examination report despatched

Effective date: 20080620

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: 4SC DISCOVERY GMBH

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20150203