WO2008019825A1 - Use of tricyclic indole derivatives for the treatment of muscular diseases - Google Patents

Use of tricyclic indole derivatives for the treatment of muscular diseases Download PDF

Info

Publication number
WO2008019825A1
WO2008019825A1 PCT/EP2007/007183 EP2007007183W WO2008019825A1 WO 2008019825 A1 WO2008019825 A1 WO 2008019825A1 EP 2007007183 W EP2007007183 W EP 2007007183W WO 2008019825 A1 WO2008019825 A1 WO 2008019825A1
Authority
WO
WIPO (PCT)
Prior art keywords
carboxylic acid
tetrahydro
carbazole
alkyl
fluoro
Prior art date
Application number
PCT/EP2007/007183
Other languages
French (fr)
Inventor
Holger Deppe
Alexandre Briguet
Günther Metz
Michael Erb
Original Assignee
Santhera Pharmaceuticals (Schweiz) 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
Priority claimed from EP06016934A external-priority patent/EP1891949A1/en
Application filed by Santhera Pharmaceuticals (Schweiz) Ag filed Critical Santhera Pharmaceuticals (Schweiz) Ag
Publication of WO2008019825A1 publication Critical patent/WO2008019825A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system

Definitions

  • the present invention relates to the use of a compound for the preparation of a medicament for the prophylaxis and/or treatment of muscular atrophy.
  • a general loss of skeletal muscle mass is a characteristic, debilitating response to fasting, as well as many severe diseases, including advanced cancer, renal failure, sepsis, and diabetes.
  • atrophy of specific muscles results from their disuse or denervation.
  • overall rates of protein synthesis are suppressed and rates of protein degradation are consistently elevated; this response accounts for the majority of the rapid loss of muscle protein.
  • animal models of human diseases e.g., fasting, diabetes, cancer cachexia, acidosis, sepsis, disuse atrophy, denervation, and glucocorticoid treatment, most of the accelerated proteolysis in muscle appears to be due to an activation of the Ub-proteasome pathway. This activation is associated with the upregulation of muscle specific ligases.
  • Muscle specific ubiquitin ligases are an essential part of the ubiquitin-proteasome pathway for protein degradation. Indeed, increased expression of the ubiquitin ligases MAFbx and MURF1 is a critical component in muscle atrophy. These so called atrogins are upregulated in a rat atrophy model (Gomes et al., PNAS 98, 2001 , 14440-14445). Moreover, mice deficient of MAFbx, which is sometimes also referred to as atrogini, or MURF1 are resistent to atrophy (Bodine et al., Science 294, 2001 , 1704-1708).
  • FOXO transcription factors induce are essential to upregulate the atrophy-related ubiquitin ligases MAFbx and MURF and thus contribute to developing skeletal muscle atrophy (Goldberg et al., Cell 117, 2004, 399-412).
  • FOXO transcription factors belong to the large Forkhead family of proteins, a family of transcriptional regulators characterized by a conserved DNA-binding domain termed the 'forkhead box'.
  • the Forkhead family is present in all eukaryotes.
  • the FOXO Forkhead subgroup contains four members (FOXO1, FOXO3, FOXO4, and FOXO6).
  • the transcriptional activity of FOXO transcription factors is regulated via its acetylation level. Whereas the majority of data describe that acetylation of FOXO factors represses target gene transcription, some suggest that FOXO acetylation increases target gene transcription. This discrepancy has been attributed to cell type and the target-gene-specific effects of FOXO. (Greer et al., Oncogene 24, 2005, 7410- 7425).
  • HATs histone acetyltransferases
  • HDACs histone deacetylases
  • the class III HDACs are homologous to the yeast transcriptional repressor Sir2p and have no sequence similarity to class I and Il HDACs; these Sir2 proteins are also called sirtuins (Denu, Curr. Opin. Chem. Biol. 9, 2005, 431-440).
  • Sirtuins have been found in bacteria to eukaryotes. The hallmark of the family is a domain of approximately 260 amino acids that has a high degree of sequence similarity in all sirtuins.
  • the family is divided into five classes (I-IV and U) on the basis of a phylogenetic analysis of 60 sirtuins from a wide array of organisms. Class I and class IV are further divided into three and two subgroups, respectively.
  • the U-class sirtuins are found only in Gram-positive bacteria.
  • the human genome encodes seven sirtuins, with representatives from classes I-IV.
  • Non-histone substrates of the most studied human Sir2 homolog, SIRT1 were shown to include p53, FOXO proteins, p300, NFkB and PGC-Ia, implicating sirtuins in apoptosis, cell survival, transcription and metabolism.
  • SIRT1 is one of multiple deacetylases involved in the regulation of FOXO dependent transcription in mammalian cells (Brunet et al., Science 303, 2004, 2011-2015).
  • Atrophy is a common form of muscle tissue loss which can be long-lasting but, nevertheless, is frequently reversible.
  • the classic example would be the muscle deterioration seen in bed ridden patients but genetically determined diseases such as for spinal muscular atrophy can cause life-threatening muscle atrophy as well.
  • Loss of muscle tissue is also observed as a consequence of cancer, cardiac disease, infectious disease, chronic obstructive pulmonary disease (COPD), rheumatic disease, inflammatory bowel disease, end-stage renal disease, liver cirrhosis, severe injury, where it is referred to as cachexia.
  • COPD chronic obstructive pulmonary disease
  • Therapeutic options for muscle atrophy include physiotherapy and similar interventions such as electro-stimulation of affected muscle groups, while pharmacological interventions available to prevent muscle loss or accelerate recovery from muscle weakness are not at all satisfying.
  • Those agents include appetite stimulants such as megestrol, fish oil and certain unsatured fatty acids and canabinoid-like substances.
  • Figure 1 shows a histogram representing the mRNA levels of atrogin-1 measured in cultured C2C12 myotubes treated for 24 h with solvent only (1% DMSO), 10 ⁇ M Dex, 10 ⁇ M Dex and 10 ng/ml IGF-1 or 10 ⁇ M Dex and 10 ⁇ M Compound 16.
  • the invention relates to the use of a compound according to formula (I):
  • each of R 1 and R 2 is, independently from each other, halo, hydroxy, C1-C10 alkyl, Ci-C 6 haloalkyl, C1-C10 alkoxy, Ci-C 6 haloalkoxy, Z-C 6 -Ci 0 aryl, Z-C 5 -Ci 0 heteroaryl, Z-C 3 -C 8 heterocyclyl, Z-C 3 -C 8 cycloalkyl, C 2 -Ci 2 alkenyl, C 2 -Ci 2 alkynyl, Z-C 5 -Ci 0 cycloalkenyl,
  • R 3 is independently from each other fluoro, methyl, methoxy, hydroxy or amino
  • R 4 is independently from each other fluoro, chloro, methyl, methoxy, cyano, hydroxy, amino, CF 3 , CHF 2 , CH 2 F, OCF 3 , OCHF 2 or OCH 2 F;
  • Z is a bond or d-C 6 alkylene, wherein the Ci-C 6 alkylene group is optionally substituted with one or more fluorine atoms; m is 1, 2, 3 or 4; n is 1 , 2, 3 or 4; and p is 0, 1 , 2 or 3 or its tautomeric, enantiomeric, diastereomeric or pharmaceutically acceptable salts thereof for the preparation of a medicament for the prophylaxis and/or treatment of muscular atrophy.
  • Compounds of formula (I) preferably used in the present invention are those compounds in which one or more of the residues contained therein have the meanings given below. It is understood, that the preferably used compounds cover any compound obtained by combining any of the definitions disclosed within this description for the various substituents. With respect to all compounds of the formulas (I) the present invention also includes all tautomeric and stereoisomers forms and mixtures thereof in all ratios, and their pharmaceutically acceptable salts.
  • the substituents R 1 and R 2 as well as the indices m, n and p of the formula (I) independently from each other have the following meaning.
  • one or more of the substituents R 1 and R 2 as well as the indices m, n and p can have the preferred or more preferred meanings given below.
  • R 1 is halo, C1-C6 alkyl, Z-Ce-Ci 0 aryl, Z-C 5 -Ci 0 heteroaryl or Z-C 3 -C 8 cycloalkyl, wherein alkyl, aryl, heteroaryl and cycloalkyl are optionally substituted with one or more substituents R 3 . More preferably, R 1 is halo, preferably chloro or fluoro, more preferably chloro, or Z-cyclopropyl which is unsubstituted or substituted with one or more substituents R 3 .
  • the index m is 1 or 2. More preferably, the index m is 2.
  • R 2 is as defined above. Preferably, R 2 is aminocarbonyl.
  • the index n is 1.
  • each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocycloalkenyl or cycloalkenyl group in the definition of R 1 and R 2 is unsubstituted or substituted by 1 to 4 substituents R 3 , more preferably 1 to 3 substituents R 3 , most preferably 1 to 2 substituents R 3 .
  • two substituents R 3 are simultaneously attached to the same carbon atom of the alkyl, aryl, heteroaryl, cycloalkyl, heterocyclyl or cycloalkenyl group.
  • each aryl or heteroaryl group in the definition of R 1 and R 2 is unsubstituted or substituted by 1 to 4 substituents R 4 , more preferably 1 to 3 substituents R 4 , most preferably 1 to 2 substituents R 4 .
  • R 3 is as defined as above.
  • R 3 is fluoro, methyl or methoxy.
  • R 3 is fluoro.
  • R 3 is preferably attached to the carbon atom connected to the tricyclic ring system if Z is a bond or to the carbon atom connected to the alkylene chain if Z is a d-C 6 alkylene group.
  • R 4 is as defined above.
  • R 4 is fluoro, methyl, methoxy, hydroxy, amino or CF 3 .
  • Z is as defined above.
  • Z is a bond or a Ci-C 2 alkylene optionally substituted with 1 to 4 fluoro.
  • Z is -CF 2 -, -CH 2 -CF 2 -, -CH 2 -CH 2 -CF 2 - or -CH 2 -CH 2 -CH 2 - CF 2 -, wherein the substituted alkyl chain is connected with the tricyclic moiety via the CF 2 group.
  • the index m is as defined above.
  • n is as defined above.
  • n is 1 , 2 or 3, more preferably, n is 1 or 2.
  • n is 1 and R 2 is attached to position 1 of the ring.
  • the index p is as defined above. Preferably, p is 1 or 2.
  • tautomerism like e.g. keto-enol tautomerism, of compounds of general formula (I)
  • the individual forms like e.g. the keto and enol form, and together as mixtures in any ratio are also within the scope of the invention.
  • stereoisomers like e.g. enantiomers, cis/trans isomers, conformers and the like.
  • the compounds of general formula (I) are separated into their enantiomers and used in accordance with the present invention as such.
  • the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically utilizable salts.
  • the compounds of the formula (I) which contain acidic groups can be present on these groups and can be used according to the invention, for example, as alkali metal salts, alkaline earth metal salts or as ammonium salts. More precise examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanolamine or amino acids.
  • Compounds of the formula (I) which contain one or more basic groups i.e.
  • acids which can be protonated, can be present and can be used according to the invention in the form of their addition salts with inorganic or organic acids.
  • suitable acids include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfo ⁇ ic acids, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to the person skilled in the art. If the compounds of the formula (I) simultaneously contain acidic and basic groups in
  • C 1 -C 1 0 Alkyl means a straight-chain or branched carbon chain having 1 - 10 carbon atoms, e.g. methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n- pentane, n-hexane, n-heptane, n-octane, n-nonane or n-decane.
  • Each hydrogen of a Ci- C 1 0 alkyl carbon may be replaced by a substituent.
  • C 2 -C1 2 Alkenyl means a straight-chain or branched carbon chain of 2- 12 carbon atoms having one or more double bonds in the chain.
  • C 2 -Ci 2 Alkynyl means a straight-chain or branched carbon chain of 2- 12 carbon atoms having one or more triple bonds in the chain.
  • C 3 -C 8 Cycloalkyl as employed herein includes saturated monocyclic hydrocarbon group having 3 to 8 carbons., e.g. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl. Each hydrogen of a C 3 -C 8 cycloalkyl carbon may be replaced by a substituent.
  • C 5 -Ci 0 Cycloalkenyl means a cyclic alkyl chain having 5 - 11 carbon atoms, which is partially unsaturated e.g.
  • cyclopentenyl cyclopentadienyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, cyclononenyl or cyclodecenyl.
  • Each hydrogen of a C 5 -Ci 0 cycloalkenyl carbon may be replaced by a substituent.
  • Halo means fluoro, chloro, bromo or iodo. It is generally preferred that halogen is fluoro or chloro.
  • C 3 -C 8 Heterocyclyl refers to a nonaromatic 3-8 membered monocyclic ring system having 1-3 heteroatoms selected from O, N, or S. Any ring atom can be substituted.
  • Examples of C 3 -C 8 heterocyclyl include tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino, imidazolidinyl and pyrrolidinyl.
  • C 5 -C 10 Heterocycloalkenyl means a partially saturated, nonaromatic 5-10 membered monocyclic ring system having 1-3 heteroatoms selected from O, N, or S. Any ring atom can be substituted.
  • Examples of C 5 -Ci 0 heterocycloalkenyl include tetrahydropyridyl and dihydropyranyl.
  • C 6 -Ci 0 Aryl refers to an aromatic monocyclic or bicyclic hydrocarbon ring system, wherein any ring atom capable of substitution can be substituted.
  • Examples of C 6 -Ci 0 aryl moieties include phenyl and naphthyl.
  • C 5 -Ci 0 Heteroaryl refers to an aromatic 5-10 membered monocyclic or bicyclic ring system having 1-3 heteroatoms selected from O, N, N(O) or S. Any ring atom can be substituted. Examples of C 5 -Ci 0 Heteroaryl include
  • Acyl means an alkylcarbonyl, cycloalkylcarbonyl, arylcarbonyl, heterocyclylcarbonyl, or heteroarylcarbonyl substituent, any of which may be further substituted.
  • aminocarbonyl C1-C10 alkoxycarbonyl
  • hydrocarbonyl hydrazinocarbonyl
  • hydroxyaminocarbonvl refer to the radicals -C(O)NH, -C(O)O-Ci-Ci 0 alkyl, -C(O)NHNH 2 and -C(O)NH 2 NH 2 , respectively.
  • Ci-C 6 alkylamino and " Ci-C 6 dialkylamino” refer to -NH-Ci-C 6 alkyl and - N(CrC 6 alkyl) 2 radicals, respectively.
  • alkoxy refers to an -O- C 1 -C 10 alkyl radical.
  • mercapto refers to an SH radical.
  • Pharmacological agents presently available to prevent muscle loss or accelerate recovery from muscle weakness are not at all satisfying. Those agents include appetite stimulants such as megestrol, fish oil and certain unsatured fatty acids and canabinoid- like substances.
  • An alternative approach to reduce atrophy, cachexia or dystrophy or to improve recovery from atrophy is offered by interventions that slow or stop the degradation of muscle proteins which frequently is mediated by the ubiquitin- proteasome system. Activation of this proteolytic enzyme complex as seen in clinical conditions of atrophy, cachexia and dystrophy critically depends on the ligation of peptides and proteins to ubiquitin moieties. Atrogins are a class of muscle-specific ligases that couple target proteins and peptides with ubiquitin moieties thereby mediating proteolysis of muscle proteins.
  • a method for treatment of muscular atrophies is offered by identifying pharmacological agents that would appropriately modulate any of the molecules involved in the signaling cascade that leads to pathologically increased muscle protein degradation.
  • a solution to this problem would be offered by identifying pharmacological modulators of either of the known atrogins or, alternatively, pharmacological modulators of a suitable member of upstream activators of atrogin expression.
  • the identification of such pharmacological modulators offers a broad commercial application since atrophy, cachexia and dystrophy is common to a number of chronic disease states characterized by long periods of immobility. There is also a potential to apply this treatment approach in a subset of the geriatric population.
  • inhibitors of sirtuins downregulate atrogin levels in a model for muscle atrophy in cells. Without being bound to a theory it is contemplated that the treatment of differentiated myotubes with an inhibitor of SIRT1 results in the suppression of atrogin-1 mRNA levels.
  • muscular atrophy is associated with the upregulation of muscle specific ligases, e.g. atrogins, therefore, inhibitors of sirtuins can be used for the treatment of muscular atrophies.
  • the compounds of formula (I) have been previously described as inhibitors of sirtuins and as such have been proposed for the treatment of various diseases such as cancer, diabetes, Parkinson's or Alzheimer's disease or obesity (e.g. US-A-2005/0209300). It has now been shown for the first time that tricyclic indol derivatives exhibit an unexpectedly high activity in the modulation of atrogin levels. Said compounds therefore are potential drugs for treating muscular atrophy.
  • the compounds of formula (I) can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets.
  • oral liquid preparations such as, for example, suspensions, elixirs and solutions
  • carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets.
  • tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques. Such compositions and preparations should contain at least 0.1 percent of active compound. The percentage of active compound in these compositions may, of course, be varied and may conveniently be between about 2 percent to about 60 percent of the weight of the unit. The amount of active compound in such therapeutically useful compositions is such that an effective dosage will be obtained.
  • the active compounds can also be administered intranasally as, for example, liquid drops or spray.
  • the tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacia, com starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin.
  • a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
  • Various other materials may be present as coatings or to modify the physical form of the dosage unit. For instance, tablets may be coated with shellac, sugar or both.
  • a syrup or elixir may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor.
  • Compounds of formula (I) may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxy-propylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • Any suitable route of administration may be employed for providing a mammal, especially a human, with an effective dose of a compound of the present invention.
  • oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed.
  • Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like.
  • compounds of formula (I) are administered orally.
  • the effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art.
  • the effective dosage is chosen in a range between 0.01 milligram to about 100 milligram per kilogram of mammal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form. More preferably, the dosage is between 0.01 milligram to about 50 milligram per kilogram of animal body weight. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • THF tetrahydrofurane t R (min) HPLC retention time
  • the compounds of the present invention can be prepared by the synthesis routes shown below or by methods described previously (DE 2226703, DE 2431292, V.A. Parshin, H.V. Alekseeva, A.I. Bokanov, LM. Alekseeva, V.G. Granik, "Synthesis and pharmacological activity of 1 ,2,3,4-Tetrahydrocarbazole-i-carboxamides", Voprosy Biologicheskoi, Meditsinskoi i Farmatsevticheskoi Khimii 2001, (4), 40-45).
  • keto-esters are reacted in an organic solvent such as Et 2 O with a brominating agent such as bromine at a suitable temperature for a given time to yield the corresponding 3-Bromo-2-oxo-cycloalkane- carboxylic acid esters.
  • a brominating agent such as bromine
  • optionally substituted ⁇ -bromo-ketoesters are reacted with optionally substituted anilines in a given solvent or without a solvent at an appropriate temperature for a given time to yield of 2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid esters.
  • the reaction can be carried out either in a flask without a solvent by carefully mixing the two starting materials or in a microwave reaction system.
  • the reaction can be carried out at elevated temperatures, for example at an oil bath temperature higher than 130° C.
  • the products can be purified by standard procedures or may precipitate directly from a solution of the reaction mixture dissolved in an appropriate solvent upon cooling and may then be used in subsequent reactions without further purification.
  • 2,3,4,9-tetrahydro-IH-carbazole-i-carboxylic acid- amides can be obtained from the corresponding esters by treatment with a inorganic base such as LiOH in a mixture of solvents such as water, THF and EtOH followed by the conversion to the carboxylic acid amide with a coupling reagent and NH 3 in water, dioxane or EtOH.
  • a inorganic base such as LiOH in a mixture of solvents such as water, THF and EtOH
  • Such compounds can then be purified by standard purification procedures such as flash chromatography or preparative HPLC.
  • the solvent is evaporated and the reaction mixture can be diluted with an appropriate organic solvent, such as EtOAc or DCM, which is then washed with aqueous solutions, such as water, HCI, NaHSO 4 , bicarbonate, NaH 2 PO 4 , phosphate buffer (pH 7), brine, Na 2 CO 3 or any combination thereof.
  • aqueous solutions such as water, HCI, NaHSO 4 , bicarbonate, NaH 2 PO 4 , phosphate buffer (pH 7), brine, Na 2 CO 3 or any combination thereof.
  • the combined organic solvents can then be concentrated and the reaction mixture purified by chromatography.
  • Example 3 Synthesis of ⁇ -chloro-S-fluoro ⁇ AS-tetrahydro-IH-carbazole-i- carboxylic acid ethylester (3) and 6-chloro-7-fluoro-2,3,4,9-tetrahydro-1H- carbazole-1-carboxylic acid ethylester (4)
  • Example 4 Synthesis of S-chloro-e-fluoro ⁇ A ⁇ -tetrahydro-IH-carbazole-i- carboxylic acid ethylester (5) and 7-chloro-6-fluoro-2,3,4,9-tetrahydro-1H- carbazole-1-carboxylic acid ethylester (6)
  • Example 5 Synthesis of 5,6-dichloro-2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid ethylester (7) and 6,7-dichloro -2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid ethylester (8)
  • Example 16 6-chloro-7-fluoro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid amides (22a) and (22b)
  • the SIRT1 deacetylase activity assay is an assay system designed to measure the lysine deacetylase activity of recombinant human SIRT1.
  • the assay procedure has two main steps. First, the Fluor-de Lys-SIRT1 substrate, which comprises the p53 sequence Arg-His-Lys-Lys( ⁇ -acetyl), is incubated with human recombinant SIRT1 together with the cosubstrate NAD+. Deacetylation of Fluor de Lys-SIRT1 sensitizes it so that, in the second step, treatment with the Fluor de LysTM developer (trypsin) produces a fluorophore.
  • the Fluor de LysTM developer trypsin
  • the fluorophore is excited with 360 nm light and the emitted light (460 nm) is detected on a fluorometric plate reader.
  • NAD+ is consumed in the reaction to produce nicotinamide (NAM) and O-acetyl-ADP-ribose.
  • IC 50 values of the SIRT 1 enzymatic assay were grouped in 2 classes: a ⁇ 0.1 ⁇ M and 0.1 ⁇ b ⁇ 1 ⁇ M.
  • C2C12 myoblasts were seeded at a density of 40O00 cells per well in 6-well plates previously coated with 0.1% gelatin. The cells were then grown for 2 days in growth medium containing 20% fetal calf serum. To initiate myotube differentiation, the growth medium was replaced by fusion medium containing 5% horse serum and the cells were incubated for another 4 days until myotubes formed.
  • myotubes were then pre-treated with 10 ng/ml IGF-1 , 10 ⁇ M Compound 16 (6-Chloro-2,3,4,9-tetrahydro-1 H-carbazole-1-carboxylic acid amide) or solvent only (1% DMSO) for 1 hour. After this pre-incubation, myotube atrophy was induced by adding 10 ⁇ M of the glucocorticoid dexamethasone (Dex) and the cultures were incubated for another 24 hours.
  • Dex glucocorticoid dexamethasone
  • mice Four groups of mice can be included in this analysis: untreated non-starved mice, compound treated non-starved mice, untreated starved mice and compound treated starved mice.
  • the bi-daily treatment typically starts on the day before the food deprivation and the last application takes place one hour before the mice are sacrificed.
  • vehicle 10% Cyclodextrin 4% DMSO in PBS can be used.
  • a volume of 10 micro liters per gram bodyweight can be applied.
  • the final incubation volume is 25 ⁇ l.
  • a control incubation is included for each compound tested where 0.1 M phosphate buffer pH7.4 is added instead of NADPH (minus NADPH). Two control compounds are included with each species. All incubations are performed singularly for each test compound.
  • Each compound is incubated for O 1 5, 15, 30 and 45 min.
  • the control (minus NADPH) is incubated for 45 min only.
  • the reactions are stopped by the addition of 50 ⁇ l methanol containing internal standard at the appropriate time points.
  • the incubation plates are centrifuged at 2,500 rpm for 20 min at 4°C to precipitate the protein.
  • cryopreserved hepatocytes Suspensions of cryopreserved hepatocytes are used for human hepatocyte stability assay (pooled from 3 individuals). All cryopreserved hepatocytes are purchased from In Vitro Technologies, Xenotech or TCS.
  • Incubations are performed at a test or control compound concentration of 3 ⁇ M at a cell density of 0.5x10 6 viable cells/mlL The final DMSO concentration in the incubation is 0.25%. Control incubations are also performed in the absence of cells to reveal any non-enzymatic degradation.
  • Duplicate samples (50 ⁇ l) are removed from the incubation mixture at 0, 5, 10, 20, 40 and 60 min (control sample at 60 min only) and added to methanol, containing internal standard (100 ⁇ l), to stop the reaction. Tolbutamide, 7-hydroxycoumarin, and testosterone are used as control compounds. The samples are centrifuged (2500 rpm at 4 0 C for 20 min) and the supernatants at each time point are pooled for cassette analysis by LC-MS/MS using generic methods. Data Analysis
  • Caco-2 cells obtained from the ATCC at passage number 27 are used. Cells (passage number 40-60) are seeded on to Millipore Multiscreen Caco-2 plates at 1 x 105 cells/cm 2 . They are cultured for 20 days in DMEM and media is changed every two or three days. On day 20 the permeability study is performed.
  • HBSS Hanks Balanced Salt Solution pH7.4 buffer with 25 mM HEPES and 10 mM glucose at 37°C is used as the medium in permeability studies. Incubations are carried out in an atmosphere of 5% CO 2 with a relative humidity of 95%. On day 20, the monolayers are prepared by rinsing both basolateral and apical surfaces twice with HBSS at 37 0 C. Cells are then incubated with HBSS in both apical and basolateral compartments for 40 min to stabilize physiological parameters. HBSS is then removed from the apical compartment and replaced with test compound dosing solutions.
  • the solutions are made by diluting 10 mM test compound in DMSO with HBSS to give a final test compound concentration of 10 ⁇ M (final DMSO concentration 1 %).
  • the fluorescent integrity marker lucifer yellow is also included in the dosing solution.
  • Analytical standards are made from dosing solutions. Test compound permeability is assessed in duplicate. On each plate compounds of known permeability characteristics are run as controls. The apical compartment inserts are then placed into 'companion' plates containing fresh HBSS. For basolateral to apical (B-A) permeability determination the experiment is initiated by replacing buffer in the inserts then placing them in companion plates containing dosing solutions. At 120 min the companion plate is removed and apical and basolateral samples diluted for analysis by LC-MS/MS. The starting concentration (C 0 ) and experimental recovery is calculated from both apical and basolateral compartment concentrations.
  • Lucifer yellow permeation is low if monolayers have not been damaged.
  • Test and control compounds are quantified by LC-MS/MS cassette analysis using a 5-point calibration with appropriate dilution of the samples. Generic analytical conditions are used.
  • the permeability coefficient for each compound (P app ) is calculated from the following equation: C 0 X A
  • C 0 is the donor compartment concentration at time zero and A is the area of the cell monolayer.
  • C 0 is obtained from analysis of donor and receiver compartments at the end of the incubation period. It is assumed that all of the test compound measured after 120 min incubation was initially present in the donor compartment at 0 min.
  • An asymmetry index (Al) is derived as follows:
  • the apparent permeability (P app (A-B)) values of test compounds are compared to those of control compounds, atenolol and propranolol, that have human absorption of approximately 50 and 90% respectively (Zhao, Y.H., et a/., (2001). Evaluation of Human Intestinal Absorption Data and Subsequent Derivation of a Quantitative Structure-Activity Relationship (QSAR) with the Abraham Descriptors. Journal of Pharmaceutical Sciences. 90 (6), 749-784).
  • Talinolol a known P-gp substrate (Deferme, S., MoIs, R., Van Driessche, W., Augustijns, P. (2002).
  • the selective CYP1A inhibitor, alpha-naphthoflavone is screened alongside the test compounds as a positive control.
  • the selective CYP2C9 inhibitor, sulphaphenazole, is screened alongside the test compounds as a positive control.
  • the selective CYP2C19 inhibitor, tranylcypromine, is screened alongside the test compounds as a positive control.
  • CYP3A4 Inhibition Six test compound concentrations (0.05, 0.25, 0.5, 2.5, 5, 25 ⁇ M in DMSO; final DMSO concentration 0.26%) are incubated with human liver microsomes (0.25 mg/ml) and
  • the selective CYP3A4 inhibitor, ketoconazole is screened alongside the test compounds as a positive control.
  • the reactions are terminated by the addition of methanol containing internal standard. The samples are then centrifuged, and the supematants are combined, for the simultaneous analysis of 4-hydroxytolbutamide, 4- hydroxymephenytoin, dextrorphan, and 1 -hydroxymidazolam plus internal standard by LC-MS/MS.
  • test compound 5 ⁇ M, 0.5% final DMSO concentration
  • buffer pH 7.4
  • 10% plasma v/v in buffer
  • the experiment is performed using equilibrium dialysis with the two compartments separated by a semi-permeable membrane.
  • the buffer solution is added to one side of the membrane and the plasma solution to the other side.
  • Standards are prepared in plasma and buffer and are incubated at 37°C.
  • Corresponding solutions for each compound are analyzed in cassettes by LC-MS/MS.

Abstract

The present invention relates to the use of a compound for the preparation of a medicament for the treatment of muscular atrophy.

Description

Use of tricyclic indole derivatives for the treatment of muscular diseases
The present invention relates to the use of a compound for the preparation of a medicament for the prophylaxis and/or treatment of muscular atrophy.
A general loss of skeletal muscle mass is a characteristic, debilitating response to fasting, as well as many severe diseases, including advanced cancer, renal failure, sepsis, and diabetes. In addition, atrophy of specific muscles results from their disuse or denervation. In most types of muscle atrophy overall rates of protein synthesis are suppressed and rates of protein degradation are consistently elevated; this response accounts for the majority of the rapid loss of muscle protein. In a variety of animal models of human diseases e.g., fasting, diabetes, cancer cachexia, acidosis, sepsis, disuse atrophy, denervation, and glucocorticoid treatment, most of the accelerated proteolysis in muscle appears to be due to an activation of the Ub-proteasome pathway. This activation is associated with the upregulation of muscle specific ligases.
Muscle specific ubiquitin ligases are an essential part of the ubiquitin-proteasome pathway for protein degradation. Indeed, increased expression of the ubiquitin ligases MAFbx and MURF1 is a critical component in muscle atrophy. These so called atrogins are upregulated in a rat atrophy model (Gomes et al., PNAS 98, 2001 , 14440-14445). Moreover, mice deficient of MAFbx, which is sometimes also referred to as atrogini, or MURF1 are resistent to atrophy (Bodine et al., Science 294, 2001 , 1704-1708). FOXO transcription factors induce are essential to upregulate the atrophy-related ubiquitin ligases MAFbx and MURF and thus contribute to developing skeletal muscle atrophy (Goldberg et al., Cell 117, 2004, 399-412). Skeletal muscle FOXO1 transgenic mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control. (Kamei et al., J. Biol. Chem. 279, 2004, 41114- 411123).
FOXO transcription factors belong to the large Forkhead family of proteins, a family of transcriptional regulators characterized by a conserved DNA-binding domain termed the 'forkhead box'. The Forkhead family is present in all eukaryotes. In humans, the FOXO Forkhead subgroup contains four members (FOXO1, FOXO3, FOXO4, and FOXO6). The transcriptional activity of FOXO transcription factors is regulated via its acetylation level. Whereas the majority of data describe that acetylation of FOXO factors represses target gene transcription, some suggest that FOXO acetylation increases target gene transcription. This discrepancy has been attributed to cell type and the target-gene-specific effects of FOXO. (Greer et al., Oncogene 24, 2005, 7410- 7425).
Protein acetylation regulates a wide variety of cellular functions, including the recognition of DNA by proteins, protein-protein interactions, and protein stability. Post- translational modification of proteins at lysine residues by reversible acetylation is catalyzed by the opposing activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs), which act on both histone and non-histone substrates despite their names. HDACs are grouped into three classes on the basis of their homology to yeast transcriptional repressors. The class III HDACs are homologous to the yeast transcriptional repressor Sir2p and have no sequence similarity to class I and Il HDACs; these Sir2 proteins are also called sirtuins (Denu, Curr. Opin. Chem. Biol. 9, 2005, 431-440).
Sirtuins have been found in bacteria to eukaryotes. The hallmark of the family is a domain of approximately 260 amino acids that has a high degree of sequence similarity in all sirtuins. The family is divided into five classes (I-IV and U) on the basis of a phylogenetic analysis of 60 sirtuins from a wide array of organisms. Class I and class IV are further divided into three and two subgroups, respectively. The U-class sirtuins are found only in Gram-positive bacteria. The human genome encodes seven sirtuins, with representatives from classes I-IV.
Non-histone substrates of the most studied human Sir2 homolog, SIRT1, were shown to include p53, FOXO proteins, p300, NFkB and PGC-Ia, implicating sirtuins in apoptosis, cell survival, transcription and metabolism. Data indicate that SIRT1 is one of multiple deacetylases involved in the regulation of FOXO dependent transcription in mammalian cells (Brunet et al., Science 303, 2004, 2011-2015).
Atrophy is a common form of muscle tissue loss which can be long-lasting but, nevertheless, is frequently reversible. The classic example would be the muscle deterioration seen in bed ridden patients but genetically determined diseases such as for spinal muscular atrophy can cause life-threatening muscle atrophy as well. Loss of muscle tissue is also observed as a consequence of cancer, cardiac disease, infectious disease, chronic obstructive pulmonary disease (COPD), rheumatic disease, inflammatory bowel disease, end-stage renal disease, liver cirrhosis, severe injury, where it is referred to as cachexia.
Finally, severe and life threatening loss of muscle tissue is observed in genetic diseases causing muscle dystrophies exemplified in Duchenne Muscular Dystrophy, limb girdle muscular dystrophies and congenital muscle dystrophies.
Preventing loss of muscle tissue or accelerating the recovery from muscle weakness and/or loss due to atrophy, cachexia or dystrophy would be of great clinical benefit and highly desirable since the current treatment options are very limited. Therapeutic options for muscle atrophy include physiotherapy and similar interventions such as electro-stimulation of affected muscle groups, while pharmacological interventions available to prevent muscle loss or accelerate recovery from muscle weakness are not at all satisfying. Those agents include appetite stimulants such as megestrol, fish oil and certain unsatured fatty acids and canabinoid-like substances.
It is the object of the present invention to provide a compound for the treatment of muscular atrophy which has superior properties compared to known compounds conventionally used in the therapy of muscular atrophy, such as a better efficacy and improved patient compliance.
Said object is achieved by the use of tricyclic indole derivatives according to formula (I) for the preparation of a medicament for the prophylaxis and/or treatment of muscular atrophy.
Brief description of the drawings
Figure 1 shows a histogram representing the mRNA levels of atrogin-1 measured in cultured C2C12 myotubes treated for 24 h with solvent only (1% DMSO), 10 μM Dex, 10 μM Dex and 10 ng/ml IGF-1 or 10 μM Dex and 10 μM Compound 16. The invention relates to the use of a compound according to formula (I):
Figure imgf000005_0001
wherein each of R1 and R2 is, independently from each other, halo, hydroxy, C1-C10 alkyl, Ci-C6 haloalkyl, C1-C10 alkoxy, Ci-C6 haloalkoxy, Z-C6-Ci0 aryl, Z-C5-Ci0 heteroaryl, Z-C3-C8 heterocyclyl, Z-C3-C8 cycloalkyl, C2-Ci2 alkenyl, C2-Ci2 alkynyl, Z-C5-Ci0 cycloalkenyl,
Z-C5-Ci0 heterocycloalkenyl, carboxy, carboxylate, cyano, nitro, amino, CrC6 alkyl amino, Ci-C6 dialkyl amino, mercapto, S(O)NH2, S(O)2NH2, CrC4 alkylenedioxy, oxo, acyl, aminocarbonyl, Ci-Ci0 alkoxycarbonyl, hydrazinocarbonyl, Ci-C6alkyl hydrazinocarbonyl, d-C6 dialkyl hydrazinocarbonyl, hydroxyamino carbonyl, or alkoxyamino carbonyl, wherein each alkyl, heterocyclyl, cycloalkyl and cycloalkenyl is optionally substituted by one or more R3; and wherein each aryl and heteroaryl is optionally substituted by one or more R4;
R3 is independently from each other fluoro, methyl, methoxy, hydroxy or amino; R4 is independently from each other fluoro, chloro, methyl, methoxy, cyano, hydroxy, amino, CF3, CHF2, CH2F, OCF3, OCHF2 or OCH2F;
Z is a bond or d-C6 alkylene, wherein the Ci-C6 alkylene group is optionally substituted with one or more fluorine atoms; m is 1, 2, 3 or 4; n is 1 , 2, 3 or 4; and p is 0, 1 , 2 or 3 or its tautomeric, enantiomeric, diastereomeric or pharmaceutically acceptable salts thereof for the preparation of a medicament for the prophylaxis and/or treatment of muscular atrophy.
Compounds of formula (I) preferably used in the present invention are those compounds in which one or more of the residues contained therein have the meanings given below. It is understood, that the preferably used compounds cover any compound obtained by combining any of the definitions disclosed within this description for the various substituents. With respect to all compounds of the formulas (I) the present invention also includes all tautomeric and stereoisomers forms and mixtures thereof in all ratios, and their pharmaceutically acceptable salts.
In preferred embodiments of the present invention, the substituents R1 and R2 as well as the indices m, n and p of the formula (I) independently from each other have the following meaning. Hence, one or more of the substituents R1 and R2 as well as the indices m, n and p can have the preferred or more preferred meanings given below.
In a preferred embodiment of the present invention, R1 is halo, C1-C6 alkyl, Z-Ce-Ci0 aryl, Z-C5-Ci0 heteroaryl or Z-C3-C8 cycloalkyl, wherein alkyl, aryl, heteroaryl and cycloalkyl are optionally substituted with one or more substituents R3. More preferably, R1 is halo, preferably chloro or fluoro, more preferably chloro, or Z-cyclopropyl which is unsubstituted or substituted with one or more substituents R3.
In a preferred embodiment, the index m is 1 or 2. More preferably, the index m is 2.
R2 is as defined above. Preferably, R2 is aminocarbonyl.
It is further preferred in the present invention that the index n is 1.
It is further preferred that each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocycloalkenyl or cycloalkenyl group in the definition of R1 and R2 is unsubstituted or substituted by 1 to 4 substituents R3, more preferably 1 to 3 substituents R3, most preferably 1 to 2 substituents R3. In one preferred embodiment, two substituents R3 are simultaneously attached to the same carbon atom of the alkyl, aryl, heteroaryl, cycloalkyl, heterocyclyl or cycloalkenyl group.
It is further preferred that each aryl or heteroaryl group in the definition of R1 and R2 is unsubstituted or substituted by 1 to 4 substituents R4, more preferably 1 to 3 substituents R4, most preferably 1 to 2 substituents R4.
R3 is as defined as above. Preferably, R3 is fluoro, methyl or methoxy. Most preferably, R3 is fluoro. In the case of a Z-C3-C8 cycloalkyl group, R3 is preferably attached to the carbon atom connected to the tricyclic ring system if Z is a bond or to the carbon atom connected to the alkylene chain if Z is a d-C6 alkylene group. R4 is as defined above. Preferably, R4 is fluoro, methyl, methoxy, hydroxy, amino or CF3.
Z is as defined above. Preferably, Z is a bond or a Ci-C2 alkylene optionally substituted with 1 to 4 fluoro.
In a preferred embodiment, Z is -CF2-, -CH2-CF2-, -CH2-CH2-CF2- or -CH2-CH2-CH2- CF2-, wherein the substituted alkyl chain is connected with the tricyclic moiety via the CF2 group.
The index m is as defined above. Preferably, m is 1 , 2 or 3, more preferably, m is 1 or 2. If m = 1 then the substituent R1 is preferably located on position 6 of the tricyclic indole ring system.
The index n is as defined above. Preferably, n is 1 , 2 or 3, more preferably, n is 1 or 2. In a preferred embodiment, n is 1 and R2 is attached to position 1 of the ring.
The index p is as defined above. Preferably, p is 1 or 2.
Where tautomerism, like e.g. keto-enol tautomerism, of compounds of general formula (I), may occur, the individual forms, like e.g. the keto and enol form, and together as mixtures in any ratio are also within the scope of the invention. Same applies for stereoisomers, like e.g. enantiomers, cis/trans isomers, conformers and the like.
Preferably, the compounds of general formula (I) are separated into their enantiomers and used in accordance with the present invention as such.
Preferred embodiments of the compounds according to present invention are shown below.
Figure imgf000007_0001
Figure imgf000008_0001
In case the compounds according to formula (I) contain one or more acidic or basic groups, the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically utilizable salts. Thus, the compounds of the formula (I) which contain acidic groups can be present on these groups and can be used according to the invention, for example, as alkali metal salts, alkaline earth metal salts or as ammonium salts. More precise examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanolamine or amino acids. Compounds of the formula (I) which contain one or more basic groups, i.e. groups which can be protonated, can be present and can be used according to the invention in the form of their addition salts with inorganic or organic acids. Examples for suitable acids include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfoπic acids, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to the person skilled in the art. If the compounds of the formula (I) simultaneously contain acidic and basic groups in the molecule, the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions).
Within the meaning of the present invention the terms are used as follows:
"C1-C10 Alkyl" means a straight-chain or branched carbon chain having 1 - 10 carbon atoms, e.g. methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n- pentane, n-hexane, n-heptane, n-octane, n-nonane or n-decane. Each hydrogen of a Ci- C10 alkyl carbon may be replaced by a substituent.
"C2-C12 Alkenyl" means a straight-chain or branched carbon chain of 2- 12 carbon atoms having one or more double bonds in the chain. Examples of C2-Ci2 alkenyl include -CH=CH2, -CH=CH-CH3, -CH2-CH=CH2, -CH=CH-CH2-CH3, -CH=CH-CH=CH2, -CH2-CH2- CH2-CH=CH2, -CH2-CH2-CH2-CH2-CH=CH2, -CH2-(CH2J4-CH=CH2, -CH2-(CH2)S-CH=CH2, -CH2-(CHz)6-CH=CH2, -CH2-(CH2JrCH=CH2, -CH2-(CH2)8-CH=CH2 and -CH2-(CHz)9- CH=CH2. Each hydrogen of a C2-Ci2 alkenyl carbon may be replaced by a substituent.
"C2-Ci2 Alkynyl" means a straight-chain or branched carbon chain of 2- 12 carbon atoms having one or more triple bonds in the chain. Examples of C2-Ci2 alkynyl include -C≡CH, -C=C-CH3, -CH2-C=CH, -C=C-CH2-CH3, -C≡C-C≡CH, -CH2-CH2-CH2-C=CH, -CH2- CH2-CH2-CH2-C=CH, -CH2-(CHz)4-C=CH, -CH2-(CHz)5-C=CH, -CH2-(CHz)6-C=CH, -CH2- (CH2)7-C≡CH, -CH2-(CHZ)8-C=CH and -CH2-(CH2)9-C≡CH. Each hydrogen of a C2-Ci2 alkynyl carbon may be replaced by a substituent.
"C3-C8 Cycloalkyl" as employed herein includes saturated monocyclic hydrocarbon group having 3 to 8 carbons., e.g. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl. Each hydrogen of a C3-C8 cycloalkyl carbon may be replaced by a substituent. "C5-Ci0 Cycloalkenyl" means a cyclic alkyl chain having 5 - 11 carbon atoms, which is partially unsaturated e.g. cyclopentenyl, cyclopentadienyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, cyclononenyl or cyclodecenyl. Each hydrogen of a C5-Ci0 cycloalkenyl carbon may be replaced by a substituent.
"Halo" means fluoro, chloro, bromo or iodo. It is generally preferred that halogen is fluoro or chloro.
"C3-C8 Heterocyclyl" refers to a nonaromatic 3-8 membered monocyclic ring system having 1-3 heteroatoms selected from O, N, or S. Any ring atom can be substituted. Examples of C3-C8 heterocyclyl include tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino, imidazolidinyl and pyrrolidinyl.
"C5-C10 Heterocycloalkenyl" means a partially saturated, nonaromatic 5-10 membered monocyclic ring system having 1-3 heteroatoms selected from O, N, or S. Any ring atom can be substituted. Examples of C5-Ci0 heterocycloalkenyl include tetrahydropyridyl and dihydropyranyl.
"C6-Ci0 Aryl" refers to an aromatic monocyclic or bicyclic hydrocarbon ring system, wherein any ring atom capable of substitution can be substituted. Examples of C6-Ci0 aryl moieties include phenyl and naphthyl.
"C5-Ci0 Heteroaryl" refers to an aromatic 5-10 membered monocyclic or bicyclic ring system having 1-3 heteroatoms selected from O, N, N(O) or S. Any ring atom can be substituted. Examples of C5-Ci0 Heteroaryl include
"Acyl" means an alkylcarbonyl, cycloalkylcarbonyl, arylcarbonyl, heterocyclylcarbonyl, or heteroarylcarbonyl substituent, any of which may be further substituted.
The terms "aminocarbonyl," "C1-C10 alkoxycarbonyl", "hydrazinocarbonyl", and "hydroxyaminocarbonvl" refer to the radicals -C(O)NH, -C(O)O-Ci-Ci0 alkyl, -C(O)NHNH2 and -C(O)NH2NH2, respectively. The terms " Ci-C6 alkylamino" and " Ci-C6 dialkylamino" refer to -NH-Ci-C6 alkyl and - N(CrC6 alkyl)2 radicals, respectively.
The term "alkoxy" refers to an -O- C1-C10 alkyl radical. The term "mercapto" refers to an SH radical.
Pharmacological agents presently available to prevent muscle loss or accelerate recovery from muscle weakness are not at all satisfying. Those agents include appetite stimulants such as megestrol, fish oil and certain unsatured fatty acids and canabinoid- like substances. An alternative approach to reduce atrophy, cachexia or dystrophy or to improve recovery from atrophy is offered by interventions that slow or stop the degradation of muscle proteins which frequently is mediated by the ubiquitin- proteasome system. Activation of this proteolytic enzyme complex as seen in clinical conditions of atrophy, cachexia and dystrophy critically depends on the ligation of peptides and proteins to ubiquitin moieties. Atrogins are a class of muscle-specific ligases that couple target proteins and peptides with ubiquitin moieties thereby mediating proteolysis of muscle proteins.
A method for treatment of muscular atrophies is offered by identifying pharmacological agents that would appropriately modulate any of the molecules involved in the signaling cascade that leads to pathologically increased muscle protein degradation. Specifically, a solution to this problem would be offered by identifying pharmacological modulators of either of the known atrogins or, alternatively, pharmacological modulators of a suitable member of upstream activators of atrogin expression. The identification of such pharmacological modulators offers a broad commercial application since atrophy, cachexia and dystrophy is common to a number of chronic disease states characterized by long periods of immobility. There is also a potential to apply this treatment approach in a subset of the geriatric population.
Surprisingly, it has been found, that inhibitors of sirtuins downregulate atrogin levels in a model for muscle atrophy in cells. Without being bound to a theory it is contemplated that the treatment of differentiated myotubes with an inhibitor of SIRT1 results in the suppression of atrogin-1 mRNA levels. As discussed above, muscular atrophy is associated with the upregulation of muscle specific ligases, e.g. atrogins, therefore, inhibitors of sirtuins can be used for the treatment of muscular atrophies. The compounds of formula (I) have been previously described as inhibitors of sirtuins and as such have been proposed for the treatment of various diseases such as cancer, diabetes, Parkinson's or Alzheimer's disease or obesity (e.g. US-A-2005/0209300). It has now been shown for the first time that tricyclic indol derivatives exhibit an unexpectedly high activity in the modulation of atrogin levels. Said compounds therefore are potential drugs for treating muscular atrophy.
In practical use, the compounds of formula (I) can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets.
Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques. Such compositions and preparations should contain at least 0.1 percent of active compound. The percentage of active compound in these compositions may, of course, be varied and may conveniently be between about 2 percent to about 60 percent of the weight of the unit. The amount of active compound in such therapeutically useful compositions is such that an effective dosage will be obtained. The active compounds can also be administered intranasally as, for example, liquid drops or spray.
The tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacia, com starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin. When a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil. Various other materials may be present as coatings or to modify the physical form of the dosage unit. For instance, tablets may be coated with shellac, sugar or both. A syrup or elixir may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor.
Compounds of formula (I) may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxy-propylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
Any suitable route of administration may be employed for providing a mammal, especially a human, with an effective dose of a compound of the present invention. For example, oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like. Preferably compounds of formula (I) are administered orally.
The effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art.
Preferably, the effective dosage is chosen in a range between 0.01 milligram to about 100 milligram per kilogram of mammal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form. More preferably, the dosage is between 0.01 milligram to about 50 milligram per kilogram of animal body weight. This dosage regimen may be adjusted to provide the optimal therapeutic response.
In the schemes and examples below, the abbreviations used therein have the following meanings:
AcOH acetic acid
Boc tert-butoxycarbonyl
CDI 1 , 1 '-carbonyldiimidazole
CHx cyclohexane
DCM dichloromethane
DIPEA ethyl-diisopropylamine
DMA N,N-dimethylacetamide
DMAP 4-dimethylaminopyridine
DMF N,N-dimethylformamide
DMSO dimethylsulfoxide
EDC 1 -(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
Et2O diethyl ether
EtOAc ethyl acetate
EtOH ethanol
HOBt 1 -hydroxybenzotriazole h hour(s)
LiOH lithiumhydroxide
MeCN acetonitrile
MeOH methanol
Mes-CI mesylchloride
MW molecular weight
NHS N-hydroxysuccinimid
TEA triethylamine
TFA trifluoroacetic acid
TFAA trifluoroacetic acid anhydride
THF tetrahydrofurane tR (min) HPLC retention time The compounds of the present invention can be prepared by the synthesis routes shown below or by methods described previously (DE 2226703, DE 2431292, V.A. Parshin, H.V. Alekseeva, A.I. Bokanov, LM. Alekseeva, V.G. Granik, "Synthesis and pharmacological activity of 1 ,2,3,4-Tetrahydrocarbazole-i-carboxamides", Voprosy Biologicheskoi, Meditsinskoi i Farmatsevticheskoi Khimii 2001, (4), 40-45).
Reaction Scheme 1:
Synthesis of 3-Bromo-2-oxo-cycloalkane-carboxylic acid esters:
Figure imgf000015_0001
As shown in reaction Scheme 1 , optionally substituted keto-esters are reacted in an organic solvent such as Et2O with a brominating agent such as bromine at a suitable temperature for a given time to yield the corresponding 3-Bromo-2-oxo-cycloalkane- carboxylic acid esters.
Reaction Scheme 2:
Synthesis of 2,3,4,9-Tetrahydro-1H-carbazole-1-carboxylic acid esters
Figure imgf000016_0001
Figure imgf000016_0002
As shown in Reaction Scheme 2, optionally substituted α-bromo-ketoesters are reacted with optionally substituted anilines in a given solvent or without a solvent at an appropriate temperature for a given time to yield of 2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid esters. The reaction can be carried out either in a flask without a solvent by carefully mixing the two starting materials or in a microwave reaction system. The reaction can be carried out at elevated temperatures, for example at an oil bath temperature higher than 130° C. The products can be purified by standard procedures or may precipitate directly from a solution of the reaction mixture dissolved in an appropriate solvent upon cooling and may then be used in subsequent reactions without further purification.
Reaction Scheme 3:
Synthesis of 2,3,4,9-Tetrahydro-IH-carbazole-i-carboxylic acid amides:
Figure imgf000017_0001
As shown in Reaction Scheme 3, 2,3,4,9-tetrahydro-IH-carbazole-i-carboxylic acid- amides can be obtained from the corresponding esters by treatment with a inorganic base such as LiOH in a mixture of solvents such as water, THF and EtOH followed by the conversion to the carboxylic acid amide with a coupling reagent and NH3 in water, dioxane or EtOH. Such compounds can then be purified by standard purification procedures such as flash chromatography or preparative HPLC.
Generally, after the reaction is complete, the solvent is evaporated and the reaction mixture can be diluted with an appropriate organic solvent, such as EtOAc or DCM, which is then washed with aqueous solutions, such as water, HCI, NaHSO4, bicarbonate, NaH2PO4, phosphate buffer (pH 7), brine, Na2CO3 or any combination thereof. The combined organic solvents can then be concentrated and the reaction mixture purified by chromatography. The following examples are provided to illustrate the invention and are not limiting the scope of the invention in any manner.
Example 1
Synthesis of the intermediate 1,3-bromo-2-oxo-cyclohexane-carboxylic acid ethylester (i)
Figure imgf000018_0001
(1) Ethyl^-oxocyclohexanecarboxylate (10 g, 58.7 mmol) was dissolved in Et2O and cooled with stirring to 00C. Bromine was added dropwise over 15 min and the reaction was allowed to warm to room temperature over 90 min. The reaction mixture was poured into ice-cold saturated aqueous Na2CO3 and extracted with EtOAc (3x). The combined organic layers were dried (Na2SO4), the solvent was evaporated to yield the final product, 3-Bromo-2-oxo-cyclohexane-carboxylic acid ethylester (1) (14.33 g), which was used without further purification.
General procedure (I):
Synthesis of intermediate 2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid ethyl esters
Figure imgf000018_0002
In a flask or a sealable reaction vessel 3-bromo-2-oxo-cyclohexane-carboxylic acid ethylester (1 eq) and an aniline (2.5 eq) were mixed without solvent and the mixture was heated to 150° C in an oil bath for 30 min to 3h under argon. The reaction was monitored either by LCMS and/or TLC. After the heating was stopped, the reaction mixture was purified by flash-chromatography (AcOEt/cyclohexane) to yield the intermediate 2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid ethylesters. In the case of polysubstituted anilines the resulting mixtures of isomeric 2,3,4,9-tetrahydro-1 H- carbazoles were separated as described, if possible. In other cases the isomers were used without separation in the following steps and were separated after the last step via preparative HPLC.
Example 2: Synthesis of 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid ethylester (2)
Figure imgf000019_0001
(2) According to the general procedure (I)1 3-bromo-2-oxo-cyclohexane-carboxylic acid ethylester (800 mg) and 4-chloroaniline (1024 mg) were reacted for 3h to yield 6- chloro-2,3,4,9-tetrahydro-1 H-carbazole-1-carboxylic acid ethylester (2) (422 mg, 45% yield) after purification.
Example 3: Synthesis of β-chloro-S-fluoro^AS-tetrahydro-IH-carbazole-i- carboxylic acid ethylester (3) and 6-chloro-7-fluoro-2,3,4,9-tetrahydro-1H- carbazole-1-carboxylic acid ethylester (4)
Figure imgf000019_0002
(3) (4) According to the general procedure (I), 3-bromo-2-oxo-cyclohexane-carboxylic acid ethylester (1000 mg) and 4-chloro-3-fluoro-aniline (1455 mg) were reacted for 3h to yield a mixture of δ-chloro-S-fluoro^.S^.Θ-tetrahydro-IH-carbazole-i-carboxylic acid ethylester (3) and e-chloro-T-fluoro^.SAΘ-tetrahydro-IH-carbazole-i-carboxylic acid ethylester (4) (1250 mg) after purification. The two isomers could not be separated by column chromatography and were used as a mixture in the next step. Example 4: Synthesis of S-chloro-e-fluoro^AΘ-tetrahydro-IH-carbazole-i- carboxylic acid ethylester (5) and 7-chloro-6-fluoro-2,3,4,9-tetrahydro-1H- carbazole-1-carboxylic acid ethylester (6)
Figure imgf000020_0001
(5) (6)
According to the general procedure (I), 3-bromo-2-oxo-cyclohexanecarboxylic acid ethylester (544 mg) and 3-chloro-4-fluoro-aniline (795 mg) were reacted for 90 min to yield the two isomers 5-chloro-6-fluoro-2,3A9-tetrahydro-1H-carbazole-1 -carboxylic acid ethylester (5) (118mg) and 7-chloro-6-fluoro-2,3A9-tetrahydro-1H-carbazole-1- carboxylic acid ethylester (6) (213 mg) after column chromatography. The two isomers were used separately in the next steps.
Example 5: Synthesis of 5,6-dichloro-2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid ethylester (7) and 6,7-dichloro -2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid ethylester (8)
Figure imgf000020_0002
(7) (8)
According to the general procedure (I), 3-bromo-2-oxo-cyclohexane-carboxylic acid ethylester (500 mg) and 3,4-dichloroaniline (815 mg) were reacted for 2h to yield the two isomeric compounds 5,6-dichloro-2,3,4,9-tetrahydro-1H-carbazole-1 -carboxylic acid ethylester (7) (158 mg) and 6,7-dichloro-2,3A9-tetrahydro-1H-carbazole-1- carboxylic acid ethylester (8) (105 mg) after column chromatography. The two isomers were used separately in the next steps. General procedure (II):
Synthesis of 2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acids
Figure imgf000021_0001
In a flask the intermediate 2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid ethyl- esters were dissolved in a mixture of THF/EtOH/H2O and a 0.5 M solution of LiOH was added. The reaction was stirred at room temperature for a given time and monitored by TLC or HPLC. After the reaction was finished, the solvent was removed by evaporation and the residue was extracted with DCM (2x). The combined organic layers were dried (Na2SO4) and the solvent was evaporated to yield the final product, which was used without further purification.
Example 6: Synthesis of 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid (9)
Figure imgf000021_0002
(9)
According to the general procedure (II) e-chloro^.SAΘ-tetrahydro-IH-carbazole-i- carboxylic acid ethylester (2) (422 mg) was converted to 6-chloro-2,3,4,9-tetrahydro- 1 H-carbazole-1-carboxylic acid (9), which was used directly in the next step. Example 7: Synthesis of 6-chloro-5-fluoro-2,3,4,9-tetrahydro-1 H-carbazole-1- carboxylic acid (10) and 6-chloro-7-fluoro-2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid (11)
Figure imgf000022_0001
(10) (11 )
According to the general procedure (II) a mixture of the two isomers 6-chloro-5-fluoro- 2,3,4,9-tetrahydro-1H-carbazole-1 -carboxylic acid ethylester (3) and 6-chloro-7-fluoro- 2,3,4,9-tetrahydro-1H-carbazole-1 -carboxylic acid ethylester (4) (1250 mg) was converted to their corresponding acids β-chloro-δ-fluoro^.SΛΘ-tetrahydro-I H- carbazole-1 -carboxylic acid (10) and e-chloro-T-fluoro^.SAΘ-tetrahydro-IH-carbazole- 1 -carboxylic acid (11) (1000 mg total), which were used directly as a mixture in the next step.
Further compounds which were synthesized according to general procedure (II) are:
5-Chloro-6-fluoro-2,3A9-tetrahydro-1 H-carbazole-1 -carboxylic acid (12) 7-Chloro-6-fluoro-2,3 A9-tetrahydro-1 H-carbazole-1 -carboxylic acid (13) 5,6-Dichloro-2,3A9-tetrahydro-1 H-carbazole-1 -carboxylic acid (14) 6,7-Dichloro-2,3,4,9-tetrahydro-1 H-carbazole-1 -carboxylic acid (15)
General procedure (III):
Synthesis of 2,3,4,9-tetrahydro-1 H-carbazole-1 -carboxylic acid amides
Figure imgf000022_0002
In a flask the intermediate 2,3,4,9-tetrahydro-IH-carbazole-i-carboxylic acid was dissolved in dry DCM. If necessary, some dry DMF was added to dissolve the starting material. To this mixture, NHS (1.4 eq) and EDC (2 eq) were added and the reaction was stirred at room temperature for 1h. The formation of the intermediate NHS-ester was monitored by TLC and/or LCMS. The solution was then washed with H2O1 the aqueous layer was extracted with DCM. From the combined organic layers the solvent was removed in vacuo. The resulting residue was redissolved in THF and a solution of NH3 in H2O was added. The reaction mixture was stirred at room temperature for a given time. After the reaction was finished, the solvent was evaporated and the final product was purified by prep. HPLC.
Example 8: Synthesis of 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid amide (16)
Figure imgf000023_0001
(16) According to the general procedure (III), δ-chloro^.SAΘ-tetrahydro-IH-carbazole-i- carboxylic acid (9) (400 mg) was converted to 6-chloro-2,3,4,9-tetrahydro-1 H- carbazole-1-carboxylic acid-amide (16) (183 mg).
Example 9: Synthesis of 5-chloro-6-fluoro-2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid amide (17)
Figure imgf000023_0002
(17)
According to the general procedure (III) 5-chloro-6-fluoro-2,3,4,9-tetrahydro-1 H- carbazole-1 -carboxylic acid (12) (97 mg) was converted to the corresponding 5-chloro- 6-fluoro-2,3,4,9-tetrahydro-1 H-carbazole-1 -carboxylic acid amide (17) (22 mg). Example 10: Synthesis of 7-chloro-6-fluoro-2,3,4,9-tetrahydro-1 H-carbazole-1- carboxylic acid amide (18)
Figure imgf000024_0001
(18)
According to the general procedure (III) 7-chloro-6-fluoro-2,3,4,9-tetrahydro-1 H- carbazole-1 -carboxylic acid (13) (187mg) was converted to the corresponding 7-chloro- 6-fluoro-2,3,4,9-tetrahydro-1 H-carbazole-1 -carboxylic acid-amide (18) (48 mg).
Example 11: Synthesis of 5,6-dichloro-2,3,4,9-tetrahydro-1 H-carbazole-1 - carboxylic acid amide (19)
Figure imgf000024_0002
(19)
According to the general procedure (III) 5,6-dichloro-2,3,4,9-tetrahydro-1 H-carbazole-1 - carboxylic acid (14) (110mg) was converted to the corresponding 5,6-dichloro-2, 3,4,9- tetrahydro-1 H-carbazole-1 -carboxylic acid amide (19) (86 mg).
Example 12: Synthesis of 6,7-dichloro-2,3,4,9-tetrahydro-1 H-carbazole-1 - carboxylic acid amide (20)
Figure imgf000024_0003
(20)
According to the general procedure (III) 6,7-dichloro-2,3,4,9-tetrahydro-1 H-carbazole-1 - carboxylic acid (15) (110 mg) was converted to the corresponding 6,7-dichloro -2,3,4,9- tetrahydro-1 H-carbazole-1 -carboxylic acid amide (20) (35 mg). Example 13: Synthesis of β-chloro-S-fluoro^.SAS-tetrahydro-IH-carbazole-i- carboxylic acid-amide (21) and 6-chloro-7-fluoro-2,3,4,9-tetrahydro-1H-carbazole- 1-carboxylic acid-amide (22)
Figure imgf000025_0001
(21 ) (22)
According to the general procedure (III) a mixture of 6-chloro-5-fluoro-2, 3,4,9- tetrahydro-1 H-carbazole-1-carboxylic acid (10) and 6-chloro-7-fluoro-2,3,4,9- tetrahydro-1 H-carbazole-1-carboxylic acid (11) was converted to the corresponding mixture of 6-chloro-5-fluoro-2,3,4,9-tetrahydro-1 H-carbazole-1-carboxylic acid amide (21) and β-chloro-T-fluoro^.S^.Θ-tetrahydro-I H-carbazole-i-carboxylic acid amide (22), which could not be separated by either column chromatography or preparative HPLC. The mixture was further resolved by chiral separation, as described on pages 26 to 27.
General procedure (IV): Chiral separation of 2,3,4,9-tetrahydro-1H-carbazole-1- carboxylic acid-amides
In order to obtain the pure enantiomers of the corresponding racemic 2,3,4,9- tetrahydro-1H-carbazole-1-carboxylic acid-amides the racemate was submitted to chiral separation using a given method (Method H or K) described on page 29.
The absolute stereochemistry of the compounds was not determined.
Example 14: 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid amides (16a) and (16b)
Figure imgf000025_0002
(16a) (16b) According to the general procedure (IV) chiral separation (Method H) of the racemic 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid amide gave the two enantiomers 16a and 16b. Analytical data: The two compounds were analysed (Method G) and had a retention time of 10.8 (purity > 90%) and 16.8 (purity > 90% ), respectively.
Example 15: 6-chloro-5 fluoro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid- amides (21a) and (21b)
Figure imgf000026_0001
(21a) (21 b)
According to the general procedure (IV) a chiral separation (Method K) of the racemic 6-chloro-5-fluoro-2,3,4,9-tetrahydro-1 H-carbazole-1-carboxylic acid amide mixture gave the two enantiomers 21a and 21b. Analytical data: The two compounds were analysed (Method J) and had a retention time of 9.3 (purity > 90%) and 11.9 ( purity > 90%), respectively.
Example 16: 6-chloro-7-fluoro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid amides (22a) and (22b)
Figure imgf000026_0002
(22a) (22b)
According to the general procedure (IV) a chiral separation (Method K) of the racemic 6-chloro-7-fluoro-2,3A9-tetrahydro-1H-carbazole-1-carboxylic acid amide gave the two enantiomers 22a and 22b. Analytical data: The two compounds were analysed (Method J), and had a retention time of 7.7 (purity > 90%) and 10.6 (purity > 90%), respectively.
Analytical LC-MS
The compounds of the present invention according to formula (I) were analyzed by analytical LC-MS. The conditions are summarized below.
Analytical conditions summary:
LC10Advp-Pump (Shimadzu) with SPD-M10Avp (Shimadzu) UV/Vis diode array detector and QP2010 MS-detector (Shimadzu) in ESI+ modus with UV-detection at 214, 254 and 275 nm,
Column: Waters XTerra MS C18, 3.5 μm, 2.1 * 100 mm, linear gradient with acetonitrile in water (0.15% HCOOH) Flow rate of 0,4 ml/min; Mobile Phase A: water (0.15% HCOOH) Mobile Phase B: acetonitrile (0.15% HCOOH)
Methods are:
A : linear gradient from 5% to 95% acetonitrile in water (0.1% HCOOH)
0.00 mm 5% B
5.00 min 95 % B
5.10 min 99 % B
6.40 min 99 % B
6.50 min 5 % B
8.00 min Pump STOP
B : linear gradient from 5% to 95% acetonitrile in water (0.1% HCOOH) 0.00 min 10% B
5.00 min 90 % B 5.10 min 99 % B 6.40 min 99 % B
6.50 min 5 % B
8.00 min Pump STOP
C : linear gradient from 5% to 95% acetonitrile in water (0.1% HCOOH) 0.00 min 5% B
10.00 min 95% B
10.10 min 99% B
11 .40 min 99% B I
11 .50 min 5% B
13 .00 min Pump STOP
D : start concentration 1 % acetonitrile
9.00 B.Conc 30
10 .00 B.Curve 3
12 .00 B.Conc 99
15. 00 B.Conc 99
15. 20 B.Conc 1
18.00 Pump STOP
E : start concentration 10% acetonitrile
10.00 B.Conc 60
11.00 B.Curve 2
12.00 B.Conc 99
15.00 B.Conc 99
15.20 B.Conc 10
18.00 Pump STOP
F : start concentration 15% acetonitrile 12.00 B.Conc 99 15.00 B.Conc 99 15.20 B.Conc 15
18.00 STOP 0
G;
Analytical conditions:
LC10Advp-Pump (Shimadzu) with SPD-M10Avp (Shimadzu) UV/Vis diode array detector and QP2010 MS-detector (Shimadzu) in ESI+ modus with UV-detection at 214, 254 and
220 nm, Column: Daicel CHIRALPAK AD-H 0.46cm *25cm
Gradient: lsocratic with 20% Ethanol (0.1% DEA) + 80% Heptan (0.1%DEA)
Flow rate: 0,6 ml/min
Runtime: 30 min
Mobile Phase A: Ethanol + 0.1% DEA Mobile Phase B: Heptan + 0.1% DEA
H;
Prep, conditions :
LC8A-Pump (Shimadzu) with SPD-M 10A vp (Shimadzu) UV/Vis diode array detector and QP2010 MS-detector in ESI+ modus with UV-detection at 214 and 254, 275 nm
Column: Daicel CHIRALPAK AD-H 2.0cm *25cm
Gradient: lsocratic with 20% Ethanol (0.1% DEA) + 80% Heptan (0.1%DEA)
Flow rate: 6 ml/min
Runtime: 40 min
J;
Analytical conditions:
LC10Advp-Pump (Shimadzu) with SPD-M 10A vp (Shimadzu) UV/Vis diode array detector and QP2010 MS-detector (Shimadzu) in ESI+ modus with UV-detection at 214, 254 and 220 nm,
Column: Daicel CHIRALPAK AD-H 0.46cm *25cm
Gradient: lsocratic with 80% Ethanol (0.1% DEA) + 20% Methanol (0.1%DEA)
Flow rate: 0,6 ml/min
Runtime: 30min Mobile Phase A: Ethanol + 0.1% DEA
Mobile Phase B: Methanol + 0.1 % DEA K;
Prep, conditions :
LC8A-Pump (Shimadzu) with SPD-M10Avp (Shimadzu) UVΛ/is diode array detector and
QP2010 MS-detector in ESI+ modus with UV-detection at 214 and 254, 275 nm
Column: Daicel CHIRALPAK AD-H 2.0cm *25cm
Gradient: lsocratic with 80% Ethanol (0.1% DEA) + 20% Methanol (0.1%DEA)
Flow rate: 6 ml/min
Runtime: 60 min
In table 1 below detailed examples of the invention are described which can be prepared via the reaction schemes 1 to 3.
The positions of the substituents (Pos 5-7) refer to the scheme shown below.
Table 1:
Figure imgf000030_0001
Figure imgf000030_0002
Figure imgf000031_0001
BIOLOGICAL ASSAYS
A. Enzymatic Assay - SIRT1 Deacetylase Activity Assay
The SIRT1 deacetylase activity assay is an assay system designed to measure the lysine deacetylase activity of recombinant human SIRT1. The assay procedure has two main steps. First, the Fluor-de Lys-SIRT1 substrate, which comprises the p53 sequence Arg-His-Lys-Lys(ε-acetyl), is incubated with human recombinant SIRT1 together with the cosubstrate NAD+. Deacetylation of Fluor de Lys-SIRT1 sensitizes it so that, in the second step, treatment with the Fluor de Lys™ developer (trypsin) produces a fluorophore. The fluorophore is excited with 360 nm light and the emitted light (460 nm) is detected on a fluorometric plate reader. NAD+ is consumed in the reaction to produce nicotinamide (NAM) and O-acetyl-ADP-ribose.
In detail: recombinant human SIRT1 is incubated for 60 min at 37°C with 25 μM of the indicated fluorogenic acetylated peptide substrate and 1.25 mM NAD+. Reactions were stopped by the addition of trypsin/2 mM nicotinamide and the deacetylation-dependent fluorescent signal was allowed to develop for 45 min. Fluorescence was then measured in the wells of a white microplate with a FARCyte fluorescence plate reader (Ex. 360 nm, Em. 450 nm, gain=24).
Table 2: Biological data for selected examples of the invention
In the table are listed the IC50 values of the SIRT 1 enzymatic assay. The IC50 values were grouped in 2 classes: a < 0.1 μM and 0.1 < b < 1 μM.
Figure imgf000032_0001
B. Cell based assay
Atrogin-1 levels in C2C12 myotubes
For the determination of atrogin-1 mRNA levels, C2C12 myoblasts were seeded at a density of 40O00 cells per well in 6-well plates previously coated with 0.1% gelatin. The cells were then grown for 2 days in growth medium containing 20% fetal calf serum. To initiate myotube differentiation, the growth medium was replaced by fusion medium containing 5% horse serum and the cells were incubated for another 4 days until myotubes formed.
Differentiated myotubes were then pre-treated with 10 ng/ml IGF-1 , 10 μM Compound 16 (6-Chloro-2,3,4,9-tetrahydro-1 H-carbazole-1-carboxylic acid amide) or solvent only (1% DMSO) for 1 hour. After this pre-incubation, myotube atrophy was induced by adding 10 μM of the glucocorticoid dexamethasone (Dex) and the cultures were incubated for another 24 hours.
At the end of this treatment the cultures were rinsed once with PBS and total RNA was extracted. For each culture, 0.5 μg RNA were then reversed transcribed and atrogin-1 mRNA levels were determined relative to the 18 S mRNA levels using real-time PCR. For each treatment condition atrogin-1 mRNA levels were quantified in three separate cultures. The results are shown in Figure 1.
As was also shown by others (Sandri et al., Cell 117, 2004, 399-412) treatment of differentiated myotubes with the glucocorticoid dexamethasone (Dex) induces atrogin-1 mRNA levels 2-fold. This increase in atrogin-1 ^mRNA levels can be suppressed by co- treatment with 10 ng/ml of IGF-1 to the level measured in solvent treated myotubes. IGF-1 is a protein which acts through the IGF-Akt pathway.
As can be taken from Figure 1 , co-treatment of differentiated myotubes with compound 16 (6-Chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxylic acid amide, 10μM) for 1h also suppresses the increase in atrogin-1 mRNA in Dex treated myotubes to the level measured in solvent treated myotubes or IGF-1 treated myotubes.
C. In vivo assay
1) In vivo starvation experiment Food deprivation/starvation is known to induce the transcription of the atrophy relevant atrogin-1 messenger RNA (Sandri et al., Cell 117, 2004, 399-412). In a typical setup 2- month-old female C57/BI6 mice arestarved for 1 day. The food is removed at 8.00 am and the mice are sacrificed on the next day at 2.00 pm. Total RNA is isolated from the medial gastrocnemius muscles (using the RNeasy Fibrous Tissue Mini Kit from Qiagen). From each muscle 0.5 μg RNA is reversed transcribed and atrogin-1 mRNA levels can be determined relative to the 18S mRNA levels using real-time PCR. Four groups of mice can be included in this analysis: untreated non-starved mice, compound treated non-starved mice, untreated starved mice and compound treated starved mice. The bi-daily treatment typically starts on the day before the food deprivation and the last application takes place one hour before the mice are sacrificed. As vehicle 10% Cyclodextrin 4% DMSO in PBS can be used. A volume of 10 micro liters per gram bodyweight can be applied.
D. In vitro ADME Assays
1. Microsomal Stability
Experimental Procedure
Pooled human liver microsomes (pooled male and female) and pooled rat liver microsomes (male Sprague Dawley rats) are prepared. Microsomes are stored at - 800C prior to use.
Microsomes (final concentration 0.5 mg/ml), 0.1 M phosphate buffer pH7.4 and test compound (final substrate concentration = 3 μM; final DMSO concentration = 0.25%) are pre-incubated at 37°C prior to the addition of NADPH (final concentration = 1 mM) to initiate the reaction. The final incubation volume is 25 μl. A control incubation is included for each compound tested where 0.1 M phosphate buffer pH7.4 is added instead of NADPH (minus NADPH). Two control compounds are included with each species. All incubations are performed singularly for each test compound.
Each compound is incubated for O1 5, 15, 30 and 45 min. The control (minus NADPH) is incubated for 45 min only. The reactions are stopped by the addition of 50 μl methanol containing internal standard at the appropriate time points. The incubation plates are centrifuged at 2,500 rpm for 20 min at 4°C to precipitate the protein.
Quantitative Analysis
Following protein precipitation, the sample supernatants are combined in cassettes of up to 4 compounds and analysed using generic LC-MS/MS conditions. Data Analysis
From a plot of the peak area ratio (compound peak area/internal standard peak area) against time, the gradient of the line is determined. Subsequently, half-life and intrinsic clearance are calculated using the equations below:
Elimination rate constant (k) = (- gradient)
0.693 Half life (t1/2) (min) = k
VxO.693
Intrinsic Clearance (CLint) (μl/min/mg protein) = tl/2 where V = Incubation volume μl/mg microsomal protein.
Two control compounds are included in the assay and if the values for these compounds are not within the specified limits the results are rejected and the experiment repeated.
2. Hepatocyte Stability
Experimental Procedure
Suspensions of cryopreserved hepatocytes are used for human hepatocyte stability assay (pooled from 3 individuals). All cryopreserved hepatocytes are purchased from In Vitro Technologies, Xenotech or TCS.
Incubations are performed at a test or control compound concentration of 3 μM at a cell density of 0.5x106 viable cells/mlL The final DMSO concentration in the incubation is 0.25%. Control incubations are also performed in the absence of cells to reveal any non-enzymatic degradation.
Duplicate samples (50 μl) are removed from the incubation mixture at 0, 5, 10, 20, 40 and 60 min (control sample at 60 min only) and added to methanol, containing internal standard (100 μl), to stop the reaction. Tolbutamide, 7-hydroxycoumarin, and testosterone are used as control compounds. The samples are centrifuged (2500 rpm at 40C for 20 min) and the supernatants at each time point are pooled for cassette analysis by LC-MS/MS using generic methods. Data Analysis
From a plot of In peak area ratio (compound peak area/internal standard peak area) against time, the gradient of the line is determined. Subsequently, half-life and intrinsic clearance are calculated using the equations below:
Elimination rate constant (k) = (- gradient)
0.693 Half life (t1/2)(min) = k
Figure imgf000036_0001
Intrinsic Clearance (CLint)(μl/min/million cells) = tl/2 where V = Incubation volume (μl)/number of cells
3. Caco-2 Permeability (Bi-directional)
Experimental Procedure
Caco-2 cells obtained from the ATCC at passage number 27 are used. Cells (passage number 40-60) are seeded on to Millipore Multiscreen Caco-2 plates at 1 x 105 cells/cm2. They are cultured for 20 days in DMEM and media is changed every two or three days. On day 20 the permeability study is performed.
Hanks Balanced Salt Solution (HBSS) pH7.4 buffer with 25 mM HEPES and 10 mM glucose at 37°C is used as the medium in permeability studies. Incubations are carried out in an atmosphere of 5% CO2 with a relative humidity of 95%. On day 20, the monolayers are prepared by rinsing both basolateral and apical surfaces twice with HBSS at 370C. Cells are then incubated with HBSS in both apical and basolateral compartments for 40 min to stabilize physiological parameters. HBSS is then removed from the apical compartment and replaced with test compound dosing solutions. The solutions are made by diluting 10 mM test compound in DMSO with HBSS to give a final test compound concentration of 10 μM (final DMSO concentration 1 %). The fluorescent integrity marker lucifer yellow is also included in the dosing solution. Analytical standards are made from dosing solutions. Test compound permeability is assessed in duplicate. On each plate compounds of known permeability characteristics are run as controls. The apical compartment inserts are then placed into 'companion' plates containing fresh HBSS. For basolateral to apical (B-A) permeability determination the experiment is initiated by replacing buffer in the inserts then placing them in companion plates containing dosing solutions. At 120 min the companion plate is removed and apical and basolateral samples diluted for analysis by LC-MS/MS. The starting concentration (C0) and experimental recovery is calculated from both apical and basolateral compartment concentrations.
The integrity of the monolayers throughout the experiment is checked by monitoring lucifer yellow permeation using fluorimetric analysis. Lucifer yellow permeation is low if monolayers have not been damaged. Test and control compounds are quantified by LC-MS/MS cassette analysis using a 5-point calibration with appropriate dilution of the samples. Generic analytical conditions are used.
If a lucifer yellow Papp value is above QC limits in one individual test compound well, then an n=1 result is reported. If lucifer yellow Papp values are above QC limits in both replicate wells for a test compound, the compound is re-tested. Consistently high lucifer yellow permeation for a particular compound in both wells indicates toxicity. No further experiments are performed in this case.
Data Analysis
The permeability coefficient for each compound (Papp) is calculated from the following equation:
Figure imgf000037_0001
C0 X A
Where dQ/dt is the rate of permeation of the drug across the cells, C0 is the donor compartment concentration at time zero and A is the area of the cell monolayer. C0 is obtained from analysis of donor and receiver compartments at the end of the incubation period. It is assumed that all of the test compound measured after 120 min incubation was initially present in the donor compartment at 0 min. An asymmetry index (Al) is derived as follows:
Al = PapP (B-A) Papp (A-B) An asymmetry index above unity shows efflux from the Caco-2 cells, which indicates that the compound may have potential absorption problems in vivo.
The apparent permeability (Papp (A-B)) values of test compounds are compared to those of control compounds, atenolol and propranolol, that have human absorption of approximately 50 and 90% respectively (Zhao, Y.H., et a/., (2001). Evaluation of Human Intestinal Absorption Data and Subsequent Derivation of a Quantitative Structure-Activity Relationship (QSAR) with the Abraham Descriptors. Journal of Pharmaceutical Sciences. 90 (6), 749-784). Talinolol (a known P-gp substrate (Deferme, S., MoIs, R., Van Driessche, W., Augustijns, P. (2002). Apricot Extract Inhibits the P-gp-Mediated Efflux of Talinolol. Journal of Pharmaceutical Sciences. 91(12), 2539-48)) is also included as a control compound to assess whether functional P-gp is present in the Caco-2 cell monolayer.
4. Cytochrome P450 Inhibition (5 lsoform IC50 Determination))
Experimental Procedure
CYP1A Inhibition Six test compound concentrations (0.05, 0.25, 0.5, 2.5, 5, 25 μM in DMSO; final DMSO concentration = 0.35%) are incubated with human liver microsomes (0.25 mg/ml) and NADPH (1 mM) in the presence of the probe substrate ethoxyresorufin (0.5 μM) for 5 min at 37°C. The selective CYP1A inhibitor, alpha-naphthoflavone, is screened alongside the test compounds as a positive control. CYP2C9 Inhibition
Six test compound concentrations (0.05, 0.25, 0.5, 2.5, 5, 25 μM in DMSO; final DMSO concentration = 0.25%) are incubated with human liver microsomes (1 mg/ml) and NADPH (1 mM) in the presence of the probe substrate tolbutamide (120 μM) for 60 min at 37°C. The selective CYP2C9 inhibitor, sulphaphenazole, is screened alongside the test compounds as a positive control. CYP2C19 Inhibition
Six test compound concentrations (0.05, 0.25, 0.5, 2.5, 5, 25 μM in DMSO; final DMSO concentration = 0.25%) are incubated with human liver microsomes (0.5 mg/ml) and NADPH' (1 mM) in the presence of the probe substrate mephenytoin (25 μM) for 60 min at 370C. The selective CYP2C19 inhibitor, tranylcypromine, is screened alongside the test compounds as a positive control.
CYP2D6 Inhibition
Six test compound concentrations (0.05, 0.25, 0.5, 2.5, 5, 25 μM in DMSO; final DMSO concentration = 0.25%) are incubated with human liver microsomes (0.5 mg/ml) and
NADPH (1 mM) in the presence of the probe substrate dextromethorphane (5 μM) for
30 min at 37°C. The selective CYP2D6 inhibitor, quinidine, is screened alongside the test compounds as a positive control.
CYP3A4 Inhibition Six test compound concentrations (0.05, 0.25, 0.5, 2.5, 5, 25 μM in DMSO; final DMSO concentration 0.26%) are incubated with human liver microsomes (0.25 mg/ml) and
NADPH (1 mM) in the presence of the probe substrate midazolam (2.5 μM) for 5 min at
37°C. The selective CYP3A4 inhibitor, ketoconazole, is screened alongside the test compounds as a positive control.
For the CYP1A incubations, the reactions are terminated by the addition of methanol, and the formation of the metabolite, resorufin, is monitored by fluorescence (excitation wavelength = 535 nm, emission wavelength = 595 nm). For the CYP2C9, CYP2C19, CYP2D6, and CYP3A4 incubations, the reactions are terminated by the addition of methanol containing internal standard. The samples are then centrifuged, and the supematants are combined, for the simultaneous analysis of 4-hydroxytolbutamide, 4- hydroxymephenytoin, dextrorphan, and 1 -hydroxymidazolam plus internal standard by LC-MS/MS. Generic LC-MS/MS conditions are used. Formic acid in deionised water (final concentration = 0.1%) is added to the final sample prior to analysis. A decrease in the formation of the metabolites compared to vehicle control is used to calculate an IC5O value (test compound concentration which produces 50% inhibition).
5. Plasma Protein Binding (10%)
Experimental Procedure
Solutions of test compound (5 μM, 0.5% final DMSO concentration) are prepared in buffer (pH 7.4) and 10% plasma (v/v in buffer). The experiment is performed using equilibrium dialysis with the two compartments separated by a semi-permeable membrane. The buffer solution is added to one side of the membrane and the plasma solution to the other side. Standards are prepared in plasma and buffer and are incubated at 37°C. Corresponding solutions for each compound are analyzed in cassettes by LC-MS/MS.
Quantitative Analysis
After equilibration, samples are taken from both sides of the membrane. The solutions for each batch of compounds are combined into two groups (plasma-free and plasma- containing) then cassette analyzed by LC-MS/MS using two sets of calibration, standards for plasma-free (7 points) and plasma-containing solutions (6 points). Generic LC-MS/MS conditions are used. Samples are quantified using standard curves prepared in the equivalent matrix. The compounds are tested in duplicate. A control compound is included in each experiment. Data Analysis
fu = 1 - (( PC - PF))
(PC) fu = fraction unbound
PC = sample concentration in protein containing side PF = sample concentration in protein free side fu at 10% plasma is converted to fu 100% plasma using the following equation:
fUioo% = f U 10%
Figure imgf000040_0001

Claims

1. Use of a compound according to formula (I):
Figure imgf000041_0001
wherein each of R1 and R2 is, independently from each other, halo, hydroxy, C1-Ci0 alkyl,
CrC6 haloalkyl, C1-C10 alkoxy, Ci-C6 haloalkoxy, Z-C6-Ci0 aryl, Z-C5-Ci0 heteroaryl,
Z-C3-C8 heterocyclyl, Z-C3-C8 cycloalkyl, C2-Ci2 alkenyl, C2-Ci2 alkynyl, Z-C5-Ci0 cycloalkenyl, Z-C5-Ci0 heterocycloalkenyl, carboxy, carboxylate, cyano, nitro, amino, Ci-C6 alkyl amino, d-C6 dialkyl amino, mercapto, S(O)NH2, S(O)2NH2,
Ci-C4 alkylenedioxy, oxo, acyl, aminocarbonyl, C1-C1Q alkoxycarbonyl, hydrazine- carbonyl, CrC6 alkyl hydrazinocarbonyl, Ci-C6 dialkyl hydrazinocarbonyl, hydroxyamino carbonyl, or alkoxyamino carbonyl, wherein each alkyl, alkenyl, alkynyl, heterocyclyl, heterocycloalkenyl, cycloalkyl and cycloalkenyl is optionally substituted by one or more R3; and wherein each aryl and heteroaryl is optionally substituted with one or more R4;
R3 is independently from each other fluoro, methyl, methoxy, hydroxy or amino;
R4 is independently from each other fluoro, chloro, methyl, methoxy, cyano, hydroxy, amino, CF3, CHF2, CH2F, OCF3, OCHF2 or OCH2F;
Z is a bond or Ci-C6 alkylene, wherein the CrC6 alkylene group is optionally substituted with one or more fluorine atoms; m is 1 , 2, 3 or 4; n is 1 , 2, 3 or 4; and p is O, 1 , 2 or 3 or its tautomeric, enantiomeric, diastereomeric or pharmaceutically acceptable salts thereof for the preparation of a medicament for the prophylaxis and/or treatment of muscular atrophy.
2. The use according to claim 1 , wherein R1 is halo, Ci-C6 alkyl, Z-C6-Ci0 aryl, Z-C5- Cio heteroaryl or Z-C3-C8 cycloalkyl, wherein alkyl, and cycloalkyl are optionally substituted with one or more R3 and wherein aryl and heteroaryl are optionally substituted with one or more R4.
3. The use according to claim 1 or 2, wherein R1 is halo or Z-cyclopropyl optionally substituted with one or more R3.
4. The use according to any one of claims 1 to 3, wherein R2 is SO2NH2 or aminocarbonyl.
5. The use according to any one of claims 1 to 4, wherein p is 1 or 2.
6. Use of sirtuin modulators for regulating atrogin levels in a mammal.
7. Use of sirtuin modulators for the preparation of a medicament for the prophylaxis and/or treatment of disorders associated with elevated atrogin levels.
8. The use according to claim 7 for the prophylaxis and/or treatment of muscular atrophy.
PCT/EP2007/007183 2006-08-14 2007-08-14 Use of tricyclic indole derivatives for the treatment of muscular diseases WO2008019825A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US83749506P 2006-08-14 2006-08-14
EP06016934A EP1891949A1 (en) 2006-08-14 2006-08-14 Use of tricyclic indole derivatives for the treatment of muscular atrophy
EP06016934.9 2006-08-14
US60/837,495 2006-08-14

Publications (1)

Publication Number Publication Date
WO2008019825A1 true WO2008019825A1 (en) 2008-02-21

Family

ID=38606709

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2007/007183 WO2008019825A1 (en) 2006-08-14 2007-08-14 Use of tricyclic indole derivatives for the treatment of muscular diseases

Country Status (1)

Country Link
WO (1) WO2008019825A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7968587B2 (en) 2006-11-20 2011-06-28 Eli Lilly And Company Tetrahydrocyclopenta[b]indole compounds as androgen receptor modulators
WO2013057258A1 (en) * 2011-10-20 2013-04-25 Siena Biotech S.P.A. Processes for the preparation of 6-chloro-2,3,4,9-tetrahydro-1h-carbazole-1-carboxamide and of its precursors
US8486943B2 (en) 2008-05-16 2013-07-16 Eli Lilly And Company Tetrahydrocyclopenta[b]indole androgen receptor modulators
US8551726B2 (en) 2008-12-08 2013-10-08 Northwestern University Method of modulating HSF-1

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005026112A2 (en) * 2003-09-12 2005-03-24 Elixir Pharmaceuticals, Inc. Methods of treating a disorder
WO2005072408A2 (en) * 2004-01-29 2005-08-11 Elixir Pharmaceuticals, Inc Treating a viral disorder
WO2006099245A1 (en) * 2005-03-11 2006-09-21 Elixir Pharmaceuticals, Inc. Sirt inhibitors that bind to nad
WO2007047604A2 (en) * 2005-10-18 2007-04-26 Elixir Pharmaceuticals, Inc. Sirt1 inhibition

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005026112A2 (en) * 2003-09-12 2005-03-24 Elixir Pharmaceuticals, Inc. Methods of treating a disorder
WO2005072408A2 (en) * 2004-01-29 2005-08-11 Elixir Pharmaceuticals, Inc Treating a viral disorder
WO2006099245A1 (en) * 2005-03-11 2006-09-21 Elixir Pharmaceuticals, Inc. Sirt inhibitors that bind to nad
WO2007047604A2 (en) * 2005-10-18 2007-04-26 Elixir Pharmaceuticals, Inc. Sirt1 inhibition

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NAPPER ANDREW D ET AL: "Discovery of indoles as potent and selective inhibitors of the deacetylase SIRT1.", JOURNAL OF MEDICINAL CHEMISTRY. 15 DEC 2005, vol. 48, no. 25, 15 December 2005 (2005-12-15), pages 8045 - 8054, XP002414402, ISSN: 0022-2623 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7968587B2 (en) 2006-11-20 2011-06-28 Eli Lilly And Company Tetrahydrocyclopenta[b]indole compounds as androgen receptor modulators
US8486943B2 (en) 2008-05-16 2013-07-16 Eli Lilly And Company Tetrahydrocyclopenta[b]indole androgen receptor modulators
US8551726B2 (en) 2008-12-08 2013-10-08 Northwestern University Method of modulating HSF-1
WO2013057258A1 (en) * 2011-10-20 2013-04-25 Siena Biotech S.P.A. Processes for the preparation of 6-chloro-2,3,4,9-tetrahydro-1h-carbazole-1-carboxamide and of its precursors
CN103958467A (en) * 2011-10-20 2014-07-30 锡耶纳生物技术股份公司 Processes for the preparation of 6-chloro-2,3,4,9-tetrahydro-1h-carbazole-1-carboxamide and of its precursors
JP2014530839A (en) * 2011-10-20 2014-11-20 シエナ バイオテク エス.ピー.エー. Process for the preparation of 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and its precursor
AU2012324824B2 (en) * 2011-10-20 2016-08-11 Ipsol Ag Processes for the preparation of 6-Chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and of its precursors
CN103958467B (en) * 2011-10-20 2016-10-12 锡耶纳生物技术股份公司 Prepare 6-chloro-2,3,4,9-tetrahydrochysene-1H-carbazole-1-Methanamide and the method for precursor thereof
US10329254B2 (en) 2011-10-20 2019-06-25 Aop Orphan Pharmaceuticals Ag Process for the preparation of 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and intermediates thereof

Similar Documents

Publication Publication Date Title
RU2447063C2 (en) Azabicyclooctane derivatives, method for preparing and using as dipeptidyl peptidase iv inhibitors
JP2008540424A (en) Methods of using acyl hydrazones as sEH inhibitors
US20160318856A1 (en) Substituted Urea eIF2alpha Kinase Activators
CN109705071B (en) HDAC inhibitors and methods of making and using the same
Wen et al. Identification of N-(6-mercaptohexyl)-3-(4-pyridyl)-1H-pyrazole-5-carboxamide and its disulfide prodrug as potent histone deacetylase inhibitors with in vitro and in vivo anti-tumor efficacy
EP2318382A1 (en) Novel imidazo[1,2-a]pyridine derivatives, method for the preparation thereof, use thereof as medicaments, pharmaceutical compositions and novel use in particular as met inhibitors
JP5913651B2 (en) Compounds and methods for the treatment of pain and other diseases
CN102260190B (en) N-benzyl-N&#39;-(terminal carboxylic acid substituted acyloxy)octanediamide compounds with anti-tumor effect and pharmaceutical salts thereof
CA2815855C (en) Novel specific hcv ns3 protease inhibitors
FR2933982A1 (en) NOVEL IMIDAZO-1,2-A! PYRIMIDINE DERIVATIVES, PROCESS FOR THEIR PREPARATION, THEIR USE AS MEDICAMENTS, PHARMACEUTICAL COMPOSITIONS AND NOVEL USE IN PARTICULAR AS MET INHIBITORS
Liao et al. Synthesis and structure–activity relationship of non-phosphorus-based fructose-1, 6-bisphosphatase inhibitors: 2, 5-Diphenyl-1, 3, 4-oxadiazoles
CA2928468A1 (en) Novel dgat2 inhibitors
Kozlov et al. Selective inhibitor of histone deacetylase 6 (tubastatin A) suppresses proliferation of hepatitis C virus replicon in culture of human hepatocytes
WO2008019825A1 (en) Use of tricyclic indole derivatives for the treatment of muscular diseases
Nepali et al. Ring-opened tetrahydro-γ-carbolines display cytotoxicity and selectivity with histone deacetylase isoforms
Wang et al. Discovery of N-substituted sulfamoylbenzamide derivatives as novel inhibitors of STAT3 signaling pathway based on Niclosamide
CN114890990A (en) Compound and application thereof in preparation of NAMPT protein autophagy degradation agent
Xu et al. Design, synthesis and in vitro biological evaluation of marine phidianidine derivatives as potential anti-inflammatory agents
JP2021509399A (en) Indoleamine-2,3-dioxygenase inhibitor and its preparation method and use
JP5700324B2 (en) Small molecule anticancer agent that inhibits p53-mdmx interaction
EP2393792A1 (en) Derivatives of 6-(6-nh-substituted-triazolopyridazine-sulfanyl) benzothiazoles and benzimidazoles, preparation thereof, use thereof as drugs, and use thereof as met inhibitors
Su et al. Design, synthesis and structure-activity relationship studies of pyrido [2, 3-d] pyrimidin-7-ones as potent Janus Kinase 3 (JAK3) covalent inhibitors
US8530453B2 (en) Compounds and methods for the treatment of pain and other diseases
CN113527195B (en) 5-aryl nicotinamide LSD1/HDAC double-target inhibitor, preparation method and application thereof
RU2616610C2 (en) Paroxetine derivative

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07786685

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 07786685

Country of ref document: EP

Kind code of ref document: A1