WO2009080242A1 - Substituted 4-aminopyrimidine-5-carboxylic acid and use thereof - Google Patents

Substituted 4-aminopyrimidine-5-carboxylic acid and use thereof Download PDF

Info

Publication number
WO2009080242A1
WO2009080242A1 PCT/EP2008/010691 EP2008010691W WO2009080242A1 WO 2009080242 A1 WO2009080242 A1 WO 2009080242A1 EP 2008010691 W EP2008010691 W EP 2008010691W WO 2009080242 A1 WO2009080242 A1 WO 2009080242A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydrogen
formula
mmol
methyl
fluorine
Prior art date
Application number
PCT/EP2008/010691
Other languages
German (de)
French (fr)
Inventor
Lars BÄRFACKER
Barbara ALBRECHT-KÜPPER
Peter Kolkhof
Yolanda Cancho Grande
Klemens Lustig
Original Assignee
Bayer Schering Pharma Aktiengesellschaft
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bayer Schering Pharma Aktiengesellschaft filed Critical Bayer Schering Pharma Aktiengesellschaft
Publication of WO2009080242A1 publication Critical patent/WO2009080242A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present application relates to novel substituted 4-aminopyrimidine-5-carboxylic acid derivatives, processes for their preparation, their use for the treatment and / or prophylaxis of diseases and their use for the preparation of medicaments for the treatment and / or prophylaxis of diseases, preferably for treatment and / or prophylaxis of cardiovascular diseases, especially dyslipidaemias, atherosclerosis and cardiac insufficiency.
  • fibrates are currently the only treatment option for patients in these risk groups. They lower elevated triglycerides by 20-50%, lower LDL-C by 10-15%, change the LDL particle size from low density atherogenic LDL to normal dense and less atherogenic LDL and increase the HDL concentration by 10-15%.
  • Fibrates act as weak agonists of the peroxisome proliferator-activated receptor (PPAR) - ⁇ (Nature 1990, 347, 645-50).
  • PPAR-alpha is a nuclear receptor that regulates the expression of target genes by binding to DNA sequences in the promoter region of these genes [also called PPAR response elements (PPRE)].
  • PPREs have been identified in a number of genes that encode proteins that regulate lipid metabolism.
  • PPAR-alpha is highly expressed in the liver and its activation leads, inter alia, to decreased VLDL production / secretion and reduced apolipoprotein CHI (ApoCIII) synthesis. In contrast, the synthesis of apolipoprotein Al (ApoAl) is increased.
  • a disadvantage of previously approved fibrates is their weak interaction with the receptor (EC 50 in the ⁇ M range), which in turn leads to the relatively low pharmacological effects described above.
  • WO 99/41253 discloses substituted pyrimidines for the treatment of viral infections.
  • JP 2001-89452 describes substituted pyrimidines for the treatment of autoimmune diseases.
  • WO 03/045941 discloses pyrimidines for the treatment of immunological and inflammatory diseases.
  • WO 2004/111014 claims substituted pyrimidines for the treatment of cystic fibrosis.
  • 4-aminopyrimidines are described in WO 2005/003099 as ion channel modulators for the treatment of pain, arthritis, migraine, epilepsy and incontinence described.
  • WO 2005/040133 claims aminopyrimidines for the treatment of inflammatory disorders.
  • WO 2005/110416 discloses 4,5-disubstituted 2-arylpyrimidines as C5a receptor ligands for the treatment of inflammatory, immunological and cardiovascular diseases. Inter alia, substituted pyrimidines for the treatment of cancer are described in WO 2006/124874.
  • WO 2006/097220 claims 4-phenoxy-2-phenylpyrimidinecarboxylic acids and, in WO 2008/031500 and WO 2008/031501, 4-phenoxy- or 2-phenoxynicotinic acids as PPAR-alpha modulators for the treatment of dyslipidemias and arteriosclerosis.
  • the present invention relates to compounds of the general formula (I)
  • R 1 is hydrogen or (C r C 3 ) -alkyl
  • R 2 is (C r C6) alkyl, (C 3 -C 6) alkenyl or (C 3 -C 7) cycloalkyl,
  • (C 1 -C 4 ) -alkyl having 1 or 2 substituents independently of one another can be substituted from the group of fluorine, trifluoromethyl, hydroxyl, (C 1 -C 4 ) -alkoxy, trifluoromethoxy and (C 3 -C 7 ) -cycloalkyl,
  • (C 3 -C 7 ) -cycloalkyl in turn having 1 or 2 substituents independently selected from the group fluorine, hydroxy, oxo, (C r C 4 ) alkyl, trifluoromethyl, 2,2,2-trifluoroethyl, (C r C 4 ) alkoxy and trifluoromethoxy can be substituted, and
  • R 1 and R 2 together with the nitrogen atom to which they are attached form a pyrrolidine or piperidine ring, which in turn is independently selected from the group of fluorine, trifluoromethyl, (C 1 -C 4 ) -alkyl by 1 or 2 substituents , Hydroxy, (C 1 -C 4 ) -alkoxy and trifluoromethoxy may be substituted,
  • R 3 is (C r C 4) -alkyl or cyclopropyl
  • R 4 is hydrogen or fluorine
  • R 5 is hydrogen, fluorine, chlorine or methyl
  • R 6 is hydrogen, halogen, nitro, cyano, trifluoromethyl, methyl, ethyl, trifluoromethoxy or methoxy,
  • R 7 is hydrogen, fluorine, chlorine or methyl
  • R 4 , R 5 , R 6 and R 7 is different from hydrogen
  • Compounds according to the invention are the compounds of the formula (I) and their salts, solvates and solvates of the salts comprising the compounds of the formulas below and their salts, solvates and solvates of the salts and of the formula (I) encompassed by formula (I), hereinafter referred to as exemplary compounds and their salts, solvates and solvates of the salts, as far as the compounds of formula (I), the compounds mentioned below are not already salts, solvates and solvates of the salts.
  • the compounds of the invention may exist in stereoisomeric forms (enantiomers, diastereomers).
  • the invention therefore includes the enantiomers or diastereomers and their respective mixtures. From such mixtures of enantiomers and / or diastereomers, the stereoisomerically uniform components can be isolated in a known manner.
  • the present invention encompasses all tautomeric forms.
  • Salts used in the context of the present invention are physiologically acceptable salts of the compounds according to the invention. Also included are salts which are themselves unsuitable for pharmaceutical applications but can be used, for example, for the isolation or purification of the compounds of the invention.
  • Physiologically acceptable salts of the compounds of the invention include acid addition salts of mineral acids, carboxylic acids and sulfonic acids, e.g. Salts of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, benzenesulfonic acid, naphthalenedisulfonic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
  • salts of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, benzenesulfonic acid, naphthalenedisulfonic acid acetic acid, trifluoroacetic acid, propionic acid
  • Physiologically acceptable salts of the compounds according to the invention also include salts of customary bases, such as, by way of example and by way of preference, alkali metal salts (for example sodium and potassium salts), alkaline earth salts (for example calcium and magnesium salts) and ammonium salts derived from ammonia or organic amines having from 1 to 16 carbon atoms.
  • alkali metal salts for example sodium and potassium salts
  • alkaline earth salts for example calcium and magnesium salts
  • ammonium salts derived from ammonia or organic amines having from 1 to 16 carbon atoms such as, by way of example and by way of preference, alkali metal salts (for example sodium and potassium salts), alkaline earth salts (for example calcium and magnesium salts) and ammonium salts derived from ammonia or organic amines having from 1 to 16 carbon atoms.
  • Atoms such as, by way of example and by way of preference, ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methylmorpholine, arginine, lysine, ethylenediamine and N-methylpiperidine.
  • Solvates in the context of the invention are those forms of the compounds according to the invention which form a complex in the solid or liquid state by coordination with solvent molecules. Hydrates are a special form of solvates that coordinate with water. As solvates, hydrates are preferred in the context of the present invention.
  • the present invention also includes prodrugs of the compounds of the invention.
  • prodrugs includes compounds which themselves are biologically active or inactive may, however, be converted into compounds of the invention during their residence time in the body (for example metabolically or hydrolytically).
  • the present invention also includes hydrolyzable ester derivatives of the carboxylic acids of the formula (I).
  • esters which can be hydrolyzed in physiological media and in particular in vivo enzymatically or chemically to the free carboxylic acids.
  • straight-chain or branched (C 1 -C 6 ) -alkyl esters in which the alkyl group is hydroxyl, Amino, mono- (Ci-C 4 ) -alkylamino and / or di- (C iC 4 ) -alkylamino may be substituted.
  • Particularly preferred are the methyl or ethyl esters of the compounds of formula (I).
  • alkyl is a linear or branched alkyl radical having in each case the number of carbon atoms specified.
  • alkyl is a linear or branched alkyl radical having in each case the number of carbon atoms specified.
  • Alkenyl in the context of the invention is a straight-chain or branched alkenyl radical having 3 to 6 carbon atoms and one or two double bonds. Preference is given to a straight-chain or branched alkenyl radical having 3 or 4 carbon atoms and one double bond. By way of example and by way of preference: allyl, isopropenyl and n-but-2-en-1-yl.
  • Alkoxy in the context of the invention is a linear or branched alkoxy radical having 1 to 4 carbon atoms. Examples which may be mentioned are: methoxy, ethoxy, n-propoxy, isopropoxy, 1-methylpropoxy, n-butoxy, isobutoxy and tert-butoxy.
  • Cycloalkyl in the context of the invention is a monocyclic, saturated alkyl radical having 3 to 7 carbon atoms. Examples which may be mentioned by way of example include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • Halogen in the context of the invention includes fluorine, chlorine, bromine and iodine. Preference is given to chlorine or fluorine.
  • radicals are substituted in the compounds according to the invention, the radicals can, unless otherwise specified, be monosubstituted or polysubstituted. Within the scope of the present invention It holds true that for all radicals which occur several times, their meaning is independent of one another. Substitution with one, two or three identical or different substituents is preferred. Very particular preference is given to the substitution with a substituent.
  • R 1 is hydrogen, methyl or ethyl
  • R 2 is (C 1 -C 6 ) -alkyl, cyclopropyl, cyclopentyl or cyclohexyl,
  • (C 1 -C 6 ) -alkyl may be substituted by a substituent selected from the group consisting of fluorine, trifluoromethyl, cyclopropyl, cyclopentyl and cyclohexyl,
  • cyclopropyl, cyclopentyl and cyclohexyl may themselves be substituted by a substituent selected from the group consisting of fluorine, methyl, ethyl and trifluoromethyl,
  • cyclopropyl, cyclopentyl and cyclohexyl can be substituted by a substituent selected from the group consisting of fluorine, methyl, ethyl and trifluoromethyl,
  • R 1 and R 2 together with the nitrogen atom to which they are attached form a pyrrolidine or piperidine ring, which in turn may be substituted by a substituent selected from the group consisting of fluorine, trifluoromethyl, methyl and ethyl,
  • R 3 is (C 1 -O-alkyl or trifluoromethyl
  • R 4 is hydrogen
  • R 5 is hydrogen or fluorine
  • R 6 is hydrogen, fluorine, chlorine, trifluoromethyl or methyl
  • R 7 is hydrogen or methyl
  • R 2 is ethyl, iso-propyl, cyclopropyl or cyclopropylmethyl
  • R 3 is methyl, ethyl or iso-propyl
  • R 4 is hydrogen
  • R 5 is hydrogen
  • R 6 is hydrogen, chlorine or methyl
  • R 7 is hydrogen or methyl
  • R 6 and R 7 are different from hydrogen
  • R 1 is hydrogen or ethyl
  • R 2 is ethyl, iso-propyl, iso-butyl or cyclopropylmethyl
  • R 3 is iso-butyl
  • R 4 is hydrogen
  • R 5 is hydrogen
  • R 6 is hydrogen, chlorine or methyl
  • R 7 is hydrogen or methyl
  • R 6 and R 7 are different from hydrogen
  • the invention further provides a process for the preparation of the compounds of the formula (I) according to the invention, which comprises reacting a compound of the formula (II)
  • R 8 is (C 1 -C 4 ) -alkyl
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 and R 7 each have the meanings given above,
  • the compounds of the formula (IV) are commercially available, known from the literature or can be prepared in analogy to processes known from the literature.
  • the compounds of the formula (II) can be prepared by reacting compounds of the formula (VI)
  • the reaction (II) -> (ET) is carried out without a solvent or optionally in an inert solvent which is suitable under the reaction conditions, for example hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane or petroleum fractions, or other solvents such as dimethylformamide, dimethylsulfoxide, N-methylpyrrolidone ( ⁇ MP) or acetonitrile. It is likewise possible to use mixtures of the solvents mentioned.
  • the reaction preferably takes place without solvent.
  • the reaction (II) -> (IH) is generally carried out in a temperature range from 0 0 C to +160 0 C, preferably at +20 0 C to +120 0 C, optionally in a microwave.
  • the reaction can be carried out at normal, elevated or reduced pressure (eg from 0.5 to 5 bar). Generally, one works at normal pressure.
  • Inert solvents for process step (IE) + (IV) ⁇ (V) are, for example, ethers, such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons, such as benzene, toluene, xylene, hexane, cyclohexane or petroleum fractions, or other solvents, such as dimethylformamide , Dimethyl sulfoxide, N, N'-dimethylpropyleneurea (DMPU), N-methylpyrrolidinone ( ⁇ MP), pyridine, acetone, 2-butanone or acetonitrile. It is likewise possible to use mixtures of the solvents mentioned. Preference is given to using dimethylformamide or tetrahydrofuran.
  • ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol
  • Suitable bases for process step (HT) + (IV) -> • (V) are customary inorganic and organic bases. These include in particular alkali metal hydroxides such as lithium, sodium or potassium hydroxide, alkali metal or alkaline earth metal carbonates such as lithium, sodium, potassium, calcium or cesium carbonate, alkali metal hydrides such as sodium or potassium hydride, organometallic bases such as n-butyl lithium or tert. organic amines such as diisopropylethylamine or triethylamine. Preferred is triethylamine.
  • the base is used here in an amount of 1 to 5 mol, preferably in an amount of 1.2 to 3 mol, based on 1 mol of the compound of formula (IV).
  • the reaction (IH) + (FV) -> (V) is generally carried out in a temperature range from 0 0 C to +150 0 C, preferably at +20 0 C to +120 0 C.
  • the reaction can be at normal, elevated or be carried out at reduced pressure (eg from 0.5 to 5 bar). Generally, one works at normal pressure.
  • the hydrolysis of the carboxylic acid esters in process steps (V) -> (I) by conventional methods, optionally in a microwave, by treating the esters in inert solvents with acids or bases, wherein the salts initially formed in the latter by subsequent treatment with acid be converted into the free carboxylic acids. In the case of the tert-butyl ester ester cleavage is preferably carried out with acids.
  • Suitable inert solvents for the hydrolysis of the carboxylic acid esters are water or the organic solvents customary for ester cleavage. These include in particular alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, tetrahydrofuran, dioxane or glycol dimethyl ether, or other solvents such as acetone, acetonitrile, dichloromethane, dimethylformamide or dimethyl sulfoxide. It is likewise possible to use mixtures of the solvents mentioned.
  • Suitable bases for the ester hydrolysis are the customary inorganic bases. These include in particular alkali metal or alkaline earth metal hydroxides such as sodium, lithium, potassium or barium hydroxide, or alkali metal or alkaline earth metal carbonates such as sodium, potassium or calcium carbonate. Preference is given to using sodium hydroxide or potassium hydroxide.
  • Suitable acids for the ester cleavage are generally sulfuric acid, hydrochloric acid / hydrochloric acid, hydrobromic / hydrobromic acid, phosphoric acid, acetic acid, trifluoroacetic acid, toluenesulfonic acid, methanesulfonic acid or trifluoromethanesulfonic acid or mixtures thereof, optionally with the addition of water.
  • Hydrogen chloride or trifluoroacetic acid are preferred in the case of the tert-butyl esters and hydrochloric acid in the case of the methyl esters.
  • the Esterspaltung is generally carried out in a temperature range of 0 0 C to +100 0 C, preferably from 0 0 C to +50 0 C.
  • the reaction may be at atmospheric, elevated or reduced pressure is performed (for example from 0.5 to 5 bar) ,
  • the compounds according to the invention have valuable pharmacological properties and can be used for the prevention and treatment of diseases in humans and animals.
  • the compounds according to the invention are highly effective PPAR-alpha modulators and moreover have increased metabolic stability. They are particularly suitable for primary and / or secondary prevention and treatment of cardiovascular diseases caused by Disturbances in fatty acid and glucose metabolism are caused. Such disorders include dyslipidaemias (hypercholesterolemia, hypertriglyceridemia, elevated levels of postprandial plasma triglycerides, hypoalphalipoproteinemia, combined hyperlipidemias), arteriosclerosis, and metabolic disorders (metabolic syndrome, hyperglycemia, insulin-dependent diabetes, non-insulin-dependent diabetes, gestational diabetes, hyperinsulinemia, insulin resistance , Glucose intolerance, obesity (obesity) and diabetic sequelae such as retinopathy, nephropathy and neuropathy).
  • dyslipidaemias hypercholesterolemia, hypertriglyceridemia, elevated levels of postprandial plasma triglycerides, hypoalphalipoproteinemia, combined hyperlipidemias
  • arteriosclerosis and metabolic disorders (
  • the compounds according to the invention are also particularly suitable for primary and / or secondary prevention and treatment of cardiac insufficiency.
  • cardiac insufficiency also encompasses more specific or related forms of disease such as right heart failure, left heart failure, global insufficiency, hypertension-induced heart failure, ischemic cardiomyopathy, dilated cardiomyopathy, congenital heart defects, valvular heart failure, valvular heart failure, mitral valve stenosis, mitral valve insufficiency, aortic valve stenosis, aortic valve insufficiency, tricuspid stenosis , Tricuspid insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined heart valve defects, myocarditis, chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic cardiomyopathy, cardiac storage disorders, diastolic heart failure, and systolic heart failure.
  • diseases such as right heart failure, left heart failure, global insufficiency, hypertension-induced heart failure, ischemic cardiomyopathy,
  • the compounds of the invention may also be used for the treatment and / or prevention of cancers such as skin cancer, breast cancer, brain tumors, head and neck cancer, liposarcoma, carcinoma of the eye, gastrointestinal tract, thyroid, liver, pancreas of the respiratory organs, the kidney, the ureter, the prostate, the genital tract and their distant metastases as well as lyphomas, sarcomas and leukemias.
  • cancers such as skin cancer, breast cancer, brain tumors, head and neck cancer, liposarcoma, carcinoma of the eye, gastrointestinal tract, thyroid, liver, pancreas of the respiratory organs, the kidney, the ureter, the prostate, the genital tract and their distant metastases as well as lyphomas, sarcomas and leukemias.
  • the compounds according to the invention can also be used for the treatment and / or prevention of micro- and macrovascular damage (vasculitis), reperfusion damage, arterial as well as venous thromboses, edema, diseases of the central nervous system and neurodegenerative disorders (stroke, Alzheimer's disease, Parkinson's disease, dementia, epilepsy, depression, multiple sclerosis), inflammatory diseases, immune diseases (Crohn's disease, ulcerative colitis, lupus erythematosus, rheumatoid arthritis, asthma), chronic obstructive pulmonary diseases (chronic bronchitis, COPD), kidney disease (glomerulonephritis), thyroid disease (hyperthyroidism), diseases of the pancreas (pancreatitis), liver fibrosis, skin diseases (psoriasis, acne, eczema, atopic dermatitis, dermatitis, Keratitis, scarring, wart formation, chilblains), sepsis, viral diseases (vas
  • the effectiveness of the compounds of the invention can be e.g. in vitro by the transactivation assay described in the Examples section.
  • the efficacy of the compounds of the invention in vivo can be e.g. Check by the tests described in the example section.
  • Another object of the present invention is the use of the compounds of the invention for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
  • Another object of the present invention is the use of the erf ⁇ ndungswashen compounds for the manufacture of a medicament for the treatment and / or prevention of Erkran- kungen, in particular the aforementioned diseases.
  • Another object of the present invention is a method for the treatment and / or prevention of diseases, in particular the aforementioned diseases, using an effective amount of at least one of the inventive compounds.
  • Another object of the present invention are the compounds of the invention for use in a method for the treatment and / or prophylaxis of dyslipidaemias, arteriosclerosis and heart failure.
  • the compounds of the invention may be used alone or as needed in combination with other agents.
  • Another object of the present invention are pharmaceutical compositions containing at least one of the compounds of the invention and one or more other active ingredients, in particular for the treatment and / or prevention of the aforementioned diseases.
  • AIs suitable combination active ingredients are exemplary and preferably mentioned: fat metabolism-altering agents, antidiabetics, blood pressure lowering agents, circulation-promoting and / or antithrombotic agents and antioxidants, chemokine receptor antagonists, p38 kinase inhibitors, NPY agonists, orexin agonists, Anorectics, PAF-AH inhibitors, anti-inflammatory drugs (COX inhibitors, LTB 4 receptor antagonists), analgesics (aspirin), antidepressants and other psychotropic drugs.
  • the present invention relates, in particular, to combinations of at least one of the compounds according to the invention with at least one lipid metabolism-altering active ingredient, an antidiabetic agent, a hypotensive agent and / or an antithrombotic agent.
  • the compounds of the invention may preferably be with one or more
  • the substances which modify the lipid metabolism by way of example and preferably from the group of HMG-CoA reductase inhibitors, inhibitors of HMG-CoA reductase expression, squalene synthesis inhibitors, ACAT inhibitors, LDL receptor inducers, cholesterol Absorption inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors,
  • MTP inhibitors lipase inhibitors, LpL activators, fibrates, niacin, CETP inhibitors, PPAR- ⁇ and / or PPAR- ⁇ agonists, RXR modulators, FXR modulators, LXR modulators, thyroid hormones and / or thyroid mimetics, ATP citrate lyase inhibitors, Lp (a) antagonists, cannabinoid receptor 1 antagonists, leptin receptor agonists, bombesin receptor agonists, histamine receptor agonists and the antioxidants / free radical scavengers;
  • hypotensive agents by way of example and preferably from the group of calcium antagonists, angiotensin Aue antagonists, ACE inhibitors, beta-receptor blockers, alpha-receptor blockers, ECE inhibitors and the vasopeptidase inhibitors;
  • Antithrombotic agents by way of example and preferably from the group of platelet aggregation inhibitors or anticoagulants; • diuretics;
  • cGMP cyclic guanosine monophosphate
  • cAMP cyclic adenosine monophosphate
  • PDE phosphodiesterases
  • sildenafil sildenafil
  • Vardenafil tadalafil
  • PDE 3 inhibitors such as milrinone
  • Natriuretic peptides e.g. atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP, Nesiritide), C-type natriuretic peptide (CNP) and urodilatin;
  • ABP atrial natriuretic peptide
  • BNP B-type natriuretic peptide
  • CNP C-type natriuretic peptide
  • urodilatin urodilatin
  • Calcium sensitizers such as by way of example and preferably levosimendan
  • NO-independent, but heme-dependent guanylate cyclase stimulators in particular the compounds described in WO 00/06568, WO 00/06569, WO 02/42301 and WO 03/095451;
  • Guanylate cyclase NO- and heme-independent activators in particular the compounds described in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 and WO 02/070510;
  • HNE human neutrophil elastase
  • the signal transduction cascade inhibiting compounds such as tyrosine kinase inhibitors, especially sorafenib, imatinib, Gef ⁇ tinib and erlotinib; and or
  • lipid metabolism-changing active compounds are preferably compounds from the group of HMG-CoA reductase inhibitors, squalene synthesis inhibitors, ACAT inhibitors, cholesterol Abso ⁇ tionhemmer, MTP inhibitors, lipase inhibitors, thyroid hormones and / or thyroid mimetics, niacin receptor Agonists, CETP inhibitors, PPAR gamma agonists, PPAR delta agonists, polymeric bile acid adsorbers, bile acid reabsorption inhibitors, antioxidants / radical scavengers, and the cannabinoid receptor 1 antagonists.
  • HMG-CoA reductase inhibitors preferably compounds from the group of HMG-CoA reductase inhibitors, squalene synthesis inhibitors, ACAT inhibitors, cholesterol Abso ⁇ tionhemmer, MTP inhibitors, lipase inhibitors, thyroid hormones and / or thyroid mimetics, niacin receptor Agonists, CETP inhibitors,
  • the compounds according to the invention are administered in combination with an HMG-CoA reductase inhibitor from the class of statins, such as by way of example and preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin, cerivastatin or pitavastatin ,
  • statins such as by way of example and preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin, cerivastatin or pitavastatin ,
  • the compounds according to the invention are administered in combination with a squalene synthesis inhibitor, such as by way of example and preferably BMS-188494 or TAK-475.
  • a squalene synthesis inhibitor such as by way of example and preferably BMS-188494 or TAK-475.
  • the compounds according to the invention are administered in combination with an ACAT inhibitor, such as by way of example and preferably melinamide, pactimibe, eflucimibe or SMP-797.
  • an ACAT inhibitor such as by way of example and preferably melinamide, pactimibe, eflucimibe or SMP-797.
  • the compounds according to the invention are administered in combination with a cholesterol absorption inhibitor, such as by way of example and preferably ezetimibe, tiqueside or pamaqueside.
  • a cholesterol absorption inhibitor such as by way of example and preferably ezetimibe, tiqueside or pamaqueside.
  • the compounds according to the invention are administered in combination with an MTP inhibitor, such as by way of example and preferably implitapide or JTT-130.
  • the compounds according to the invention are administered in combination with a lipase inhibitor, such as, for example and preferably, orlistat.
  • a lipase inhibitor such as, for example and preferably, orlistat.
  • the compounds according to the invention are administered in combination with a thyroid hormone and / or thyroid mimetic, such as by way of example and preferably D-thyroxine or 3,5,3'-triiodothyronine (T3).
  • a thyroid hormone and / or thyroid mimetic such as by way of example and preferably D-thyroxine or 3,5,3'-triiodothyronine (T3).
  • the compounds according to the invention are administered in combination with an agonist of the niacin receptor, such as by way of example and preferably niacin, Acipimox, A mecanical or Radecol.
  • an agonist of the niacin receptor such as by way of example and preferably niacin, Acipimox, A mecanical or Radecol.
  • the compounds according to the invention are administered in combination with a CETP inhibitor, such as, by way of example and by way of preference, torcetrapib, JTT-705 or CETP vaccine (Avant).
  • the compounds according to the invention are administered in combination with a PPAR-gamma agonist, such as by way of example and preferably pioglitazone or rosiglitazone.
  • a PPAR-gamma agonist such as by way of example and preferably pioglitazone or rosiglitazone.
  • the compounds of the invention are administered in combination with a PPAR delta agonist such as, for example and preferably, GW-501516.
  • a PPAR delta agonist such as, for example and preferably, GW-501516.
  • the compounds according to the invention are administered in combination with a polymeric bile acid adsorbent such as, by way of example and by way of preference, cholestyramine, colestipol, colesolvam, cholesta gel or colestimide.
  • a polymeric bile acid adsorbent such as, by way of example and by way of preference, cholestyramine, colestipol, colesolvam, cholesta gel or colestimide.
  • the compounds according to the invention are administered in combination with an antioxidant / radical catalyst such as, by way of example and by way of preference, probucol, AGI-1067, BO-653 or AEOL-10150.
  • an antioxidant / radical catalyst such as, by way of example and by way of preference, probucol, AGI-1067, BO-653 or AEOL-10150.
  • the compounds of the invention are administered in combination with a cannabinoid receptor 1 antagonist, such as by way of example and preferably rimonabant or SR-147778.
  • a cannabinoid receptor 1 antagonist such as by way of example and preferably rimonabant or SR-147778.
  • Antidiabetic agents are preferably understood as meaning insulin and insulin derivatives as well as orally active hypoglycemic agents.
  • Insulin and insulin derivatives here include both insulins of animal, human or biotechnological origin as well as mixtures thereof.
  • the orally active hypoglycemic agents preferably include sulphonylureas, biguanides, meglitinide derivatives, glucosidase inhibitors and PPAR-gamma agonists.
  • the compounds according to the invention are administered in combination with insulin.
  • the compounds according to the invention are administered in combination with a sulphonylurea, such as, by way of example and by way of preference, tolbutamide, glibenclamide, glimepiride, glipizide or gliclazide.
  • the compounds according to the invention are administered in combination with a biguanide, such as by way of example and preferably metformin.
  • the compounds of the invention are administered in combination with a meglitinide derivative, such as by way of example and preferably repaglinide or nateglinide.
  • a meglitinide derivative such as by way of example and preferably repaglinide or nateglinide.
  • the compounds according to the invention are administered in combination with a glucosidase inhibitor, such as by way of example and preferably miglitol or acarbose.
  • a glucosidase inhibitor such as by way of example and preferably miglitol or acarbose.
  • the compounds according to the invention are administered in combination with a PPAR-gamma agonist, for example from the class of thiazolidinediones, such as, by way of example and by way of preference, pioglitazone or rosiglitazone.
  • a PPAR-gamma agonist for example from the class of thiazolidinediones, such as, by way of example and by way of preference, pioglitazone or rosiglitazone.
  • the blood pressure lowering agents are preferably understood as meaning compounds from the group of calcium antagonists, angiotensin AQ antagonists, ACE inhibitors, beta-receptor blockers, alpha-receptor blockers and diuretics.
  • the compounds of the invention are administered in combination with a diuretic, such as by way of example and preferably a loop diuretic such as furosemide, bumetanide or torsemide, or a thiazide or thiazide-like diuretic such as chlorothiazide or hydrochlorothiazide.
  • a diuretic such as by way of example and preferably a loop diuretic such as furosemide, bumetanide or torsemide, or a thiazide or thiazide-like diuretic such as chlorothiazide or hydrochlorothiazide.
  • the compounds according to the invention are administered in combination with an aldosterone or mineralocorticoid receptor antagonist, such as by way of example and preferably spironolactone or eplerenone.
  • an aldosterone or mineralocorticoid receptor antagonist such as by way of example and preferably spironolactone or eplerenone.
  • the compounds according to the invention are administered in combination with a vasopressin receptor antagonist, such as by way of example and preferably Conivaptan, tolvaptan, lixivaptan or SR-121463.
  • a vasopressin receptor antagonist such as by way of example and preferably Conivaptan, tolvaptan, lixivaptan or SR-121463.
  • the compounds according to the invention are used in combination with an organic nitrate or NO donor, such as by way of example and by way of example.
  • an organic nitrate or NO donor such as by way of example and by way of example.
  • the compounds according to the invention are used in combination with a positive inotropically active compound, such as by way of example and preferably cardiac glycosides (digoxin), beta-adrenergic and dopaminergic agonists such as isoproterenol, adrenaline, norepinephrine, dopamine or dobutamine, administered.
  • a positive inotropically active compound such as by way of example and preferably cardiac glycosides (digoxin), beta-adrenergic and dopaminergic agonists such as isoproterenol, adrenaline, norepinephrine, dopamine or dobutamine, administered.
  • the compounds according to the invention are administered in combination with a calcium antagonist, such as, by way of example and by way of preference, nifedipine, amlodipine, verapamil or diltiazem.
  • a calcium antagonist such as, by way of example and by way of preference, nifedipine, amlodipine, verapamil or diltiazem.
  • the compounds according to the invention are administered in combination with an angiotensin Aü antagonist, such as by way of example and preferably losartan, valsartan, candesartan, embusartan or telmisartan.
  • angiotensin Aü antagonist such as by way of example and preferably losartan, valsartan, candesartan, embusartan or telmisartan.
  • the compounds according to the invention are administered in combination with an ACE inhibitor such as, for example and preferably, enalapril, captopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
  • an ACE inhibitor such as, for example and preferably, enalapril, captopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
  • the compounds according to the invention are used in combination with a beta-receptor blocker, such as by way of example and preferably propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipranolol, nadolol, mepindolol, carazalol, Sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, Carteolol, esmolol, labetalol, carvedilol, adaprolol, landiolol, nebivolol, epanolol or bucine dolol administered.
  • a beta-receptor blocker such as by way of example and preferably propranolol, atenolol, timolo
  • the compounds according to the invention are administered in combination with an alpha-receptor blocker, such as by way of example and preferably prazosin.
  • the compounds according to the invention are used in combination with an antisympathotonicum, such as by way of example and preferably reserpine, clonidine or alpha-methyl-dopa, or in combination with a potassium channel agonist such as, for example and preferably, minoxidil, diazoxide, dihydralazine or hydralazine.
  • an antisympathotonicum such as by way of example and preferably reserpine, clonidine or alpha-methyl-dopa
  • a potassium channel agonist such as, for example and preferably, minoxidil, diazoxide, dihydralazine or hydralazine.
  • Antithrombotic agents are preferably understood as meaning compounds from the group of platelet aggregation inhibitors or anticoagulants.
  • the compounds according to the invention are administered in combination with a platelet aggregation inhibitor, such as, by way of example and by way of preference, aspirin, clopidogrel, ticlopidine or dipyridamole.
  • the compounds according to the invention are administered in combination with a thrombin inhibitor, such as by way of example and preferably ximelagarran, melagatran, bivalirudin or Clexane.
  • a thrombin inhibitor such as by way of example and preferably ximelagarran, melagatran, bivalirudin or Clexane.
  • the compounds according to the invention are administered in combination with a GPUb / IIIa antagonist, such as by way of example and preferably tirofiban or abciximab.
  • the compounds according to the invention are used in combination with a factor Xa inhibitor, such as by way of example and preferably rivaroxaban (BAY 59-7939), DU-176b, apixaban, otamixaban, fidexaban, razaxaban, fondaparinux, idraparinux, PMD No. 3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
  • a factor Xa inhibitor such as by way of example and preferably rivaroxaban (BAY 59-7939), DU-176b, apixaban, otamixaban, fidexaban, razaxaban, fondaparinux, idraparinux, PMD No. 3112, YM-150, KFA-1982, EMD-503982, MCM
  • the compounds according to the invention are administered in combination with heparin or a low molecular weight (LMW) heparin derivative.
  • LMW low molecular weight
  • the compounds according to the invention are administered in combination with a vitamin K antagonist, such as by way of example and preferably coumarin.
  • HMG-CoA reductase inhibitors statins
  • diuretics beta-receptor blockers
  • organic nitrates and NO Donors ACE inhibitors
  • angiotensin Aü antagonists aldosterone and mineralocorticoid receptor antagonists
  • vasopressin receptor antagonists platelet aggregation inhibitors and anticoagulants, and their use for the treatment and / or prevention of the aforementioned diseases.
  • the compounds according to the invention can be used for the treatment and / or prophylaxis of cancers alone or as needed in combination with other anti-tumor agents.
  • the present invention particularly relates to combinations of at least one of the compounds according to the invention with at least one other anti-tumor active ingredient selected from the group consisting of alkylating agents, antimetabolites, of Plant derived anti-tumor agents, hormone therapy agents, topoisomerase inhibitors, camptothecin derivatives, kinase inhibitors, targeted drugs, antibodies, immunoconjugates, interferon and / or immunomodulators, antiangiogenic compounds, antisense RNA and RNA interference, and others Tumor medication.
  • Alkylating agents such as chloromethine N-oxide, cyclophosphamide, ifosfamide, thiotepa, ranimustine, ⁇ imustin, temozolomide, altretamine, apaciquin, brostallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, mafosfamide and mitolactol;
  • Platinum-coordinated alkylating agents such as, for example, cisplatin, carboplatin, eptaplatin, lobaplatin, ⁇ edaplatin, oxaliplatin and satraplatin;
  • Antimetabolites such as methotrexate, 6-mercaptopurine riboside, mercaptopurine, 5-fluorouracil alone or in combination with leucovorin, tegafur, doxifluridine, carmofur, cytarabine, cytarabine ocfosfate, enocitabine, gemcitabine, fludarabine, 5-azacitidine, capecitabine, cladribine, clofarabine, Decitabine, eflornithine, ethynylcytidine, cytosine arabinoside, hydroxyurea, melphalan, ⁇ elarabine, ⁇ olatrexed, ocfosfit, disodium
  • Premetrexed pentostatin, pelitrexol, raltitrexed, triapine, trimetrexate, vidarabine, vincristine, and vinorelbine;
  • hormonal agents such as exemestane, lupron, anastrozole, doxercalciferol, fadrozole, formestan, 11-beta hydroxysteroid dehydrogenase-1 inhibitors, 17-alpha hydroxylase / 17,20 lyase inhibitors such as abiraterone acetate, 5-alpha reductase
  • Inhibitors such as finasteride and epristeride, anti-estrogens such as tamoxifen citrate and fulvestrant, trelstar, toremifene, raloxifene, lasofoxifene, letrozole, anti-androgens such as bicalutamide, flutamide, mifepristone, ilutamide, Casodex and anti-progesterone, and combinations thereof;
  • anti-estrogens such as tamoxifen citrate and fulvestrant, trelstar, toremifene, raloxifene, lasofoxifene, letrozole
  • anti-androgens such as bicalutamide, flutamide, mifepristone, ilutamide, Casodex and anti-progesterone, and combinations thereof;
  • plant-derived antitumor agents such as mitotic inhibitors such as epothilones (sagopilone, ixabepilone and epothilone B), vinblastine, vinflunine, docetaxel, and paclitaxel;
  • mitotic inhibitors such as epothilones (sagopilone, ixabepilone and epothilone B), vinblastine, vinflunine, docetaxel, and paclitaxel;
  • Cytotoxic topoisomerase inhibitors such as aclarubicin, doxorubicin, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflomotecan, irinotecan, topotecan, edotecarin, epimbicin, etoposide, exatecan, germantcan, lurtotecan, mitoxantrone, pirambicin, pixantrone, Rubitecan, Sobuzoxan, Tafluposide, and combinations thereof; • Immunological agents exemplified and preferably from the group of interferons such.
  • Ibritumomab Imiquimod, Lenograstim, Lentinan, Melanoma Vaccine (Corixa), Molgramostim, Sargramostim, Tasonermin, Tecleukin, Thymalasin, Tositumomab, Vimlizine, Epratuzumab, Mitumomab, Oregovomab, Pemtumomab and Proveng;
  • Immunomodulators such as Krestin, Lentinan, Sizofiran, Picibanil, ProMun and Ubenimex;
  • Antiangiogenic compounds such as, for example, acitretin, aflibercept, angiostatin, aplidine, asentar, axitinib, recentin, bevacizumab, brivanib alaninate, cilengtide, combretastatin, DAST, endostatin, fenretinide, halofuginone, pazopanib, ranibizumab, rebarastat, Removab, Revlimid , Sorafenib, vatalanib, squalamine, sunitinib, telatinib, thalidomide, Ukrain and vitaxin;
  • VEGF inhibitors such as sorafenib, DAST, bevacizumab, sunitinib, recentin, axitinib, aflibercept, telatinib, brivanib alaninate, vatalanib, pazopanib, and ranibizumab;
  • antibodies such as trastuzumab, cetuximab, bevacizumab, rituximab, ticilimumab, ipilimumab, lumiliximab, catumaxomab, atacicept, orregovomab and alemtumab;
  • EGFR (HERI) inhibitors such as cetuximab, panitumumab, vectibix, gefitinib, erlotinib and Zactima;
  • HER2 inhibitors such as lapatinib, tratuzumab and pertuzumab;
  • mTOR inhibitors such as temsirolimus, sirolimus / rapamycin and everolimus;
  • CDK inhibitors such as roscovitine and flavopiridol; • Spindle assembly checkpoint inhibitors and targeted mitotic inhibitors such as PLK inhibitors, Aurora inhibitors (eg hesperadine), checkpoint kinase inhibitors and KSP inhibitors;
  • HDAC inhibitors such as Panobinostat, Vorinostat, MS275, Belinostat and LBH589;
  • Proteasome inhibitors such as bortezomib and carf ⁇ lzomib;
  • Serine / threonine kinase inhibitors such as MEK inhibitors and Raf inhibitors such as sorafenib;
  • Tyrosine kinase inhibitors such as dasatinib, nilotibib, DAST, bosutinib, sorafenib, bevacizumab, sunitinib, AZD2171, axitinib, aflibercept, telatinib, imatinib mesylate, brivanib alaninate, pazopanib, ranibizumab, vatalanib, cetuximab, panitumumab, vectibix, gefitinib, erlotinib , Lapatinib, tratuzumab, pertuzumab and c-kit inhibitors;
  • Corticoids e.g. dexamethasone
  • Thalidomide or thalidolide analogs e.g. lenalidomide
  • Bcl-2 protein inhibitors such as Obatoclax, Oblimersen sodium and Gossypol;
  • CD20 receptor antagonists such as rituximab
  • Ribonucleotide reductase inhibitors such as gemcitabine
  • Tumor necrosis apoptosis inducing ligand receptor 1 agonists such as Mapatumumab
  • 5-hydroxytryptamine receptor antagonists such as rEV598, xaliprod, palonosetron hydrochloride, granisetron, zindol and AB-1001; • Integrin inhibitors including Alpha5-betal integrin inhibitors. E7820, JSM 6425, Volociximab and Endostatin;
  • Androgen receptor antagonists including e.g. Nandrolone Decanoate, Fluoxymesterone, Android, Prost-Aid, Andromustine, Bicalutamide, Flutamide, Apo-cyproterone, Apo-Flutamide, Chlormadinone Acetate, Androcur, Tabi, Cyproterone Acetate and Nilutamide;
  • aromatase inhibitors such as Anastrozole, letrozole, testolactone, exemestane, amino-glutethimide and formestane;
  • the compounds of the invention may also be used to treat cancers associated with radiotherapy and / or surgery.
  • compositions containing at least one compound of the invention usually together with one or more inert, non-toxic, pharmaceutically suitable excipients, and their use for the purposes mentioned above.
  • the compounds according to the invention can act systemically and / or locally.
  • they may be applied in a suitable manner, e.g. oral, parenteral, pulmonary, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival, otic or as an implant or stent.
  • the compounds according to the invention can be administered in suitable administration forms.
  • the inventive compounds rapidly and / or modified donating application forms containing the compounds of the invention in crystalline and / or amorphized and / or dissolved form, such as tablets (uncoated or coated tablets, for example with enteric or delayed-dissolving or insoluble coatings, which control of the compound according to the invention), rapidly disintegrating tablets or films / wafers, films / lyophilisates, capsules (for example hard or soft gelatine capsules), dragees, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
  • tablets uncoated or coated tablets, for example with enteric or delayed-dissolving or insoluble coatings, which control of the compound according to the invention
  • rapidly disintegrating tablets or films / wafers, films / lyophilisates capsules (for example hard or soft gelatine capsules), dragees, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
  • Parenteral administration can be accomplished by bypassing a resorption step (e.g., intravenous, intraarterial, intracardiac, intraspinal, or intralumbar) or by resorting to absorption (e.g., intramuscular, subcutaneous, intracutaneous, percutaneous, or intraperitoneal).
  • a resorption step e.g., intravenous, intraarterial, intracardiac, intraspinal, or intralumbar
  • absorption e.g., intramuscular, subcutaneous, intracutaneous, percutaneous, or intraperitoneal.
  • parenteral administration are suitable as application forms u.a. Injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilisates or sterile powders.
  • Inhalation medicaments including powder inhalers, nebulizers
  • nasal drops solutions or sprays
  • lingual, sublingual or buccal tablets films / wafers or capsules
  • suppositories ear or ophthalmic preparations
  • vaginal capsules aqueous suspensions (lotions, shake mixtures), lipophilic suspensions
  • Ointments creams, transdermal therapeutic systems (eg patches), milk, pastes, foams, scattering powders, implants or stents.
  • the compounds according to the invention can be converted into the stated administration forms. This can be done in a conventional manner by mixing with inert, non-toxic, pharmaceutically suitable excipients.
  • excipients for example microcrystalline cellulose, lactose, mannitol
  • solvents for example liquid polyethylene glycols
  • emulsifiers and dispersants or wetting agents for example sodium dodecyl sulfate, polyoxysorbitanoleate
  • binders for example polyvinylpyrrolidone
  • synthetic and natural polymers for example albumin
  • Stabilizers eg antioxidants such as ascorbic acid
  • dyes eg inorganic pigments such as iron oxides
  • flavor and / or odoriferous include, among others.
  • Excipients for example microcrystalline cellulose, lactose, mannitol
  • solvents for example liquid polyethylene glycols
  • emulsifiers and dispersants or wetting agents for example sodium dodecyl
  • the dosage is about 0.01 to 100 mg / kg, preferably about 0.01 to 20 mg / kg and most preferably 0.1 to 10 mg / kg of body weight.
  • UV ultraviolet spectrometry v / v volume-to-volume ratio (of a mixture)
  • Method 1 Device Type MS: Micromass ZQ; Device type HPLC: HP 1100 Series; UV DAD; Column: Phenomenex Gemini 3 ⁇ 30 mm x 3.00 mm; Eluent A: 1 l of water + 0.5 ml of 50% formic acid, eluent B: 1 l of acetonitrile + 0.5 ml of 50% formic acid; Gradient: 0.0 min 90% A ⁇ 2.5 min 30% A ⁇ 3.0 min 5% A ⁇ 4.5 min 5% A; Flow: 0.0 min 1 ml / min ⁇ 2.5 min / 3.0 min / 4.5 min 2 ml / min; Oven: 50 ° C .; UV detection: 210 nm.
  • Method 2 Device Type MS: Micromass ZQ; Device type HPLC: Waters Alliance 2795; Column: Phenomenex Synergi 2.5 ⁇ MAX-RP 100A Mercury 20 mm x 4 mm; Eluent A: 1 l of water + 0.5 ml of 50% formic acid, eluent B: 1 l of acetonitrile + 0.5 ml of 50% formic acid; Gradient: 0.0 min 90% A - »0.1 min 90% A -» 3.0 min 5% A - »4.0 min 5% A -» 4.01 min 90% A; Flow: 2 ml / min ;; Oven: 50 ° C .; UV detection: 210 nm.
  • Method 3 Device Type MS: Micromass ZQ; Device type HPLC: Waters Alliance 2795; Column: Merck Chromolith SpeedROD RP-18e 100 x 4.6 mm; Eluent A: 1 liter of water + 0.5 ml of 50% formic acid; Eluent B: 1 liter acetonitrile + 0.5 ml 50% formic acid; Gradient: 0.0 min 10% B- * 7.0 min 95% B- * 9.0 min 95% B; Oven: 35 ° C; Flow: 0.0 min 1.0 ml / min - * 7.0 min 2.0 ml / min - * 9.0 min 2.0 ml / min; UV detection: 210 nm
  • Example 2A 1.5 g (5.124 mmol) of Example 2A in 19.1 ml (204.971 mmol) of phosphorus oxychloride are stirred for 2 hours at reflux temperature. The reaction mixture is evaporated. The residue is treated with a 25% aqueous ammonium hydroxide solution, adjusted to pH 7 with 1N hydrochloric acid and then extracted with dichloromethane. The organic phase is dried over sodium sulfate and concentrated. This gives 1.38 g (87% of theory) of the target compound.
  • Example 5A To a solution of 250 mg (0.803 mmol) of Example 5A in 4 ml of tetrahydrofuran is added 203 mg (2.01 mmol) of triethylamine and 71 mg (1.205 mmol) of isopropylamine. The reaction mixture is stirred overnight at 80 0 C. The mixture is concentrated by rotary evaporation and used without further workup. This gives 250 mg (91% of theory) of the target compound
  • Example 4A 1.5 g (about 2151 mmol) of Example 4A in 8 ml (86.041 mmol) of phosphorus oxychloride are stirred for 2 h at reflux temperature. The reaction mixture is evaporated. The residue is treated with a 25% aqueous ammonium hydroxide solution, adjusted to pH 7 with 1N hydrochloric acid and then extracted with dichloromethane. The organic phase is dried over sodium sulfate and concentrated. The residue is column chromatographed on silica gel (mobile phase: cyclohexane / ethyl acetate 50/1 ⁇ 20/1). , This gives 540 mg (55% of theory) of crude product in 74% purity (LC-MS), which is reacted without further purification operation.
  • LC-MS LC-MS
  • Example 5 A To a solution of 100 mg (0.321 mmol) of Example 5 A in 2 ml of tetrahydrofuran is added 1.8 ml (13,498 mmol) of triethylamine and 35 mg (0.482 mmol) of diethylamine. The reaction mixture is stirred at 80 0 C overnight. The mixture is concentrated by rotary evaporation and used without further workup. This gives 100 mg (90% of theory) of the target compound
  • Example 8A To a solution of 60 mg (about 0.131 mmol) of Example 8A in 0.8 ml of THF are added 9 mg (0.196 mmol) of ethylamine and 0.8 ml (5.498 mmol) of triethylamine. The reaction mixture is stirred overnight at 80 0 C. The reaction mixture is concentrated by rotary evaporation and the residue is reacted without further work-up. This gives 55 mg (100% Th.) Of the target compound in 90% - purity (LC-MS).
  • Example 8A To a solution of 60 mg (about 0.131 mmol) of Example 8A in 0.8 ml of THF are added 11 mg (0.196 mmol) of cyclopropanamine and 556 mg (5.498 mmol) of triethylamine. The reaction mixture is stirred overnight at 80 0 C. The mixture is purified by preparative HPLC without elution (eluent: acetonitrile / water, gradient 10/90 ⁇ 90/10). This gives 40 mg (85% Th.) Of the target compound.
  • Example 8A To a solution of 60 mg (about 0.131 mmol) of Example 8A in 0.8 ml of THF are added 0.09 ml (0.196 mmol) of methanamine solution (2M in THF) and 556 mg (5.498 mmol) of triethylamine. The reaction mixture is stirred overnight at 80 0 C. The mixture is purified by preparative HPLC without further workup (eluent: acetonitrile / water, gradient 10:90 ⁇ 90:10). This gives 60 mg (100% Th.) Of the target compound.
  • Example 13A 1.38 g (4.47 mmol) of Example 13A is dissolved in 40 ml of tetrachloromethane and under argon atmosphere with 0.835 g (4.69 mmol) of N-bromosuccinimide, 0.054 mg (0.223 mmol).
  • Residue is purified by preparative MPLC (Biotage 4OM cartridge;
  • Example 15A 39 ul (0:46 mmol) of isopropylamine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted overnight at 80 0 C. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 ⁇ 90:10). This gives 42 mg (39% of theory) of the target compound.
  • Example 15A 35 ul (0:46 mmol) of allylamine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted overnight at 80 0 C. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 70 mg (66% of theory) of the target compound.
  • Example 15A 48 ul (0:46 mmol) of diethylamine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted overnight at 80 0 C. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 90 mg (81% of theory) of the target compound.
  • Example 15A 53 ul (0:46 mmol) of cyclohexylamine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted overnight at 80 0 C. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 43 mg (36% of theory) of the target compound.
  • Example 15 A 100 mg (0.31 mmol) of Example 15 A, 46 .mu.l (0.46 mmol) of piperidine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran and then reacted at 80 0 C overnight. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 123 mg (98% of theory) of the target compound.
  • preparative HPLC eluent: acetonitrile / water, gradient 10:90 -> 90:10
  • Example 21A 1.32 g (4.58 mmol) of Example 21A are dissolved in 40 ml of tetrachloromethane and treated under argon with 0.856 g (4.81 mmol) of N-bromosuccinimide, 0.055 mg (0.229 mmol) of dibenzoyl peroxide and 6.32 g (45.78 mmol) of potassium carbonate. The mixture is then stirred at reflux temperature for 30 minutes. After cooling, the batch is mixed with 100 ml of water and extracted with dichloromethane (3x). The combined organic phases are dried with magnesium sulfate. Then, the volatile components are separated by distillation under reduced pressure.
  • Example 22A 450 mg (1.60 mmol) of Example 22A and 5.96 ml of phosphoxyl chloride are added for 2 h
  • Example 23A 100 mg (0.328 mmol) of Example 23A, 51 ul (0.492 mmol) of diethylamine and 114 .mu.l (0.820 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted at 80 0 C for 3 h. Then the volatile components are separated on a rotary evaporator. The residue will be purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 78 mg (66% of theory) of the target compound.
  • preparative HPLC eluent: acetonitrile / water, gradient 10:90 -> 90:10
  • Example 23A 100 mg (0.328 mmol) of Example 23A, 42 ul (0.492 mmol) of diethylamine and 114 .mu.l (0.820 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted at 80 0 C for 2 h. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 ⁇ 90:10). This gives 68 mg (62% of theory) of the target compound.
  • preparative HPLC eluent: acetonitrile / water, gradient 10:90 ⁇ 90:10
  • Example 26A A mixture of 5.02 g (-10.27 mmol) of Example 26A, 1.83 g (10.27 mmol) of N-bromosuccinimide, 497 mg (2.05 mmol) of dibenzoyl peroxide and 2.13 g (15.40 mmol) of ground potassium carbonate in 120 ml of dioxane is refluxed under argon atmosphere for 3 hours touched. After cooling, the mixture is treated with a saturated aqueous sodium thiosulfate solution and then concentrated. The aqueous phase is extracted with dichloromethane and the organic phase is washed first with water and then with a saturated aqueous sodium chloride solution. It is dried with sodium sulfate and concentrated.
  • Example 28 A To a solution of 60 mg (0.177 mmol) of Example 28 A in 1.2 ml of THF are added 12 mg (0.265 mmol) of ethylamine and 45 mg (0.442 mmol) of triethylamine. The mixture is stirred at 80 0 C overnight. The reaction mixture is concentrated by rotary evaporation and used further without further work-up. This gives 32 mg (53% of theory) of the target compound.
  • Example 28 A To a solution of 60 mg (0.177 mmol) of Example 28 A in 1.2 ml of THF are added 16 mg (0.265 mmol) of isopropylamine and 45 mg (0.442 mmol) of triethylamine. The mixture is stirred at 80 ° C. for 30 h. Then the volatile components are separated on a rotary evaporator. The residue is taken up in ethyl acetate and washed several times with water. The organic phase is dried over magnesium sulfate, the solvent is evaporated in vacuo and the crude product is purified by chromatography on silica gel (mobile phase: dichloromethane). This gives 35 mg (49% of theory) of the target compound.
  • Example 6A To a solution of 250 mg (0.749 mmol) of Example 6A in 15.6 ml of ethanol is added 7.5 ml (14.978 mmol) of a 2M aqueous sodium hydroxide solution. The reaction mixture is stirred at 80 ° C. for 6 hours. For workup, the reaction mixture is adjusted to pH 1 with 1N hydrochloric acid and the volatile components are distilled off in vacuo. The resulting crystals are filtered off and dried under high vacuum. This gives 195 mg (74% of theory) of the target compound.
  • Example 7A To a solution of 100 mg (0.289 mmol) of Example 7A in 6 ml of ethanol is added 2.9 ml (5.783 mmol) of a 2M aqueous sodium hydroxide solution. The reaction mixture is stirred at 80 ° C. for 6 hours. For workup, the reaction mixture with 1N hydrochloric acid to pH. 1 placed and distilled off the volatile components in vacuo. The resulting crystals are filtered off and dried under high vacuum. This gives 83 mg (79% of theory) of the target compound.
  • Example 9A 100 mg (0.287 mmol) of Example 9A are taken up in 6 ml of ethanol and admixed with 2.8 ml (5.75 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 140 0 C in a single Mojé microwave (Emrys Optimizer) 40th The mixture is concentrated to pH 1 with 1N hydrochloric acid and the residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 90:10). This gives 15 mg (28% of theory) of the target compound.
  • Example 1OA 55 mg (about 0.142 mmol) of Example 1OA are taken up in 2 ml of ethanol and treated with 1.4 ml (2,846 mmol) of a 2M aqueous sodium hydroxide solution. It is then reacted at 140 ° C. for 20 minutes and then at 150 ° C. for 20 minutes in a single-mode microwave (Emrys Optimizer). The mixture is concentrated and the aqueous residue is taken up and adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off and dried under high vacuum. This gives 15 mg (27% of theory) of the target compound.
  • Emrys Optimizer single-mode microwave
  • Example I IA 80 mg (0.222 mmol) of Example I IA are taken up in 3 ml of ethanol and admixed with 2.2 ml (4.446 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 140 0 C in a single ⁇ forfe microwave (Emrys Optimizer) 20th The mixture is concentrated and the aqueous residue is taken up and adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off and dried under high vacuum. This gives 82 mg (91% of theory) of the target compound.
  • Example 12A 60 mg (0.180 mmol) of Example 12A are taken up in 2 ml of ethanol and admixed with 1.8 ml (3.595 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 140 0 C in a single Mwfe microwave (Emrys Optimizer) 20th The mixture is concentrated and the aqueous residue is taken up and adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off and dried under high vacuum. This gives 60 mg (97% of theory) of the target compound.
  • Example 16A 42 mg (0.121 mmol) of Example 16A are taken up in 5 ml of ethanol and admixed with 193 mg (4.83 mmol) of sodium hydroxide. After addition of a few drops of water is reacted for 2 hours at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigklareethyester (3x). Then the combined organic phases are dried with magnesium sulfate. The solvent is removed on a rotary evaporator and the residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 90:10). This gives 8 mg (21% of theory) of the target compound.
  • preparative HPLC eluent: acetonitrile / water, gradient 90:10
  • Example 17A 70 mg (0.202 mmol) of Example 17A are taken up in 5 ml of ethanol and admixed with 323 mg (8.10 mmol) of sodium hydroxide. After addition of a few drops of water is reacted for 2 hours at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigklareethyester (3x). Then the combined organic phases are dried with magnesium sulfate. The solvent is separated on a rotary evaporator. This gives 55 mg (86% of theory) of the target compound.
  • Example 18A 90 mg (0.249 mmol) of Example 18A are taken up in 5 ml of ethanol and admixed with 398 mg (9.95 mmol) of sodium hydroxide. After addition of a few drops of water is reacted overnight at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigklareethyester (3x). The combined organic phases are dried with magnesium sulfate and the solvent is removed on a rotary evaporator. The residue is finally purified by preparative HPLC (eluent: acetonitrile / water, gradient 90:10). This gives 54 mg (65% of theory) of the target compound.
  • preparative HPLC eluent: acetonitrile / water, gradient 90:10
  • Example 19A 43 mg (0.111 mmol) of Example 19A are taken up in 5 ml of ethanol and admixed with 177 mg (4.43 mmol) of sodium hydroxide. After addition of a few drops of water is reacted overnight at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigklareethyester (3x): Then the combined organic phases are dried with magnesium sulfate and the solvent is removed on a rotary evaporator. The residue was purified by preparative HPLC (eluent: acetonitrile / water, gradient 90:10). This gives 7 mg (18% of theory) of the target compound.
  • preparative HPLC eluent: acetonitrile / water, gradient 90:10
  • Example 2OA 126 mg (0.337 mmol) of Example 2OA are taken up in 5 ml of ethanol and treated with 539 mg (13.48 mmol) of sodium hydroxide. After addition of a few drops of water is reacted overnight at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. It is then extracted with Essigklareethyester (3x) and the combined organic phases are dried with magnesium sulfate. Then the solvent is separated on a rotary evaporator. This gives 104 mg (87% of theory) of the target compound.
  • Example 24A 78 mg (0.228 mmol) of Example 24A are taken up in 3 ml of ethanol and admixed with 365 mg (9.14 mmol) of sodium hydroxide. It is then reacted overnight at reflux temperature. Since the conversion remains incomplete, 15 min at 140 0 C in a single-mode microwave (Emrys Optimizer) tempered. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigklareethyester (3x). Then the combined organic phases are dried with magnesium sulfate. The solvent is separated on a rotary evaporator. Finally, it is dried in a high vacuum. This gives 60 mg (80% of theory) of the target compound.
  • Emrys Optimizer Emrys Optimizer
  • Example 25A 68 mg (0.208 mmol) of Example 25A are taken up in 3 ml of ethanol and admixed with 332 mg (8.31 mmol) of sodium hydroxide. After addition of a few drops of water is reacted overnight at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. It is then extracted with ethyl acetate (3x), The combined organic phases are dried with magnesium sulfate. Then the solvent is separated on a rotary evaporator. This gives 54 mg (87% of theory) of the target compound.
  • Example 30A 41 mg (0.109 mmol) of Example 30A are taken up in 1.2 ml of dioxane and admixed with 2.2 ml (4.64 mmol) of a 2M aqueous sodium hydroxide solution. The mixture is then reacted for 40 min at 150 0 C in a single ⁇ / brfe microwave (Emrys Optimizer). The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off, re-absorbed with water and extracted with Essigklareethyester (3x). The organic phase is concentrated. This gives 16 mg (36% of theory) of the target compound.
  • Example 31A 41 mg (0.106 mmol) of Example 31A are taken up in 1.2 ml of dioxane and admixed with 2.1 ml (4.23 mmol) of a 2M aqueous sodium hydroxide solution. It is then 40 min at 150 0 C in a single ⁇ / oJe microwave (Emrys Optimizer) implemented. The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off, re-absorbed with water and extracted with Essigklareethyester (3x). The organic phase is concentrated. This gives 21 mg (47% of theory) of the target compound.
  • Emrys Optimizer Emrys Optimizer
  • Example 32A 31 mg (0.089 mmol) of Example 32A are taken up in 1.0 ml of dioxane and admixed with 1.8 ml (3.565 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 150 0 C in a single Aforfe microwave (Emrys Optimizer) 40th The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid posed. The resulting crystals are filtered off, re-absorbed with water and extracted with Essigklareethyester (3x). The organic phase is concentrated. This gives 15 mg (45% of theory) of the target compound.
  • Example 33A 30 mg (0.080 mmol) of Example 33A are taken up in 0.9 ml of dioxane and admixed with 1.6 ml (3.210 mmol) of a 2M aqueous sodium hydroxide solution. It is then 40 min at 150 0 C in a single-Mwfe microwave (Emrys Optimizer) implemented. The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off, re-absorbed with water and extracted with Essigklareethyester (3x). The organic phase is concentrated. This gives 16 mg (50% of theory) of the target compound.
  • Emrys Optimizer single-Mwfe microwave
  • Example 34A 33 mg (0.091 mmol) of Example 34A are taken up in 1.0 ml of dioxane and admixed with 1.8 ml (3.648 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 150 0 C in a single Mocfe microwave (Emrys Optimizer) 40th The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off with suction. This gives 24 mg (68% of theory) of the target compound.
  • Mocfe microwave Emrys Optimizer
  • Example 29A 36 mg (0.091 mmol) of Example 29A are taken up in 1.0 ml of dioxane and admixed with 1.9 ml (3.83 mmol) of a 2M aqueous sodium hydroxide solution. This is followed by 40 min 150 0 C and 40 min at 170 0 C in a single Afocfe microwave (Emrys Optimizer) implemented. The mixture is concentrated and residue is taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off with suction and purified by HPLC (Sunfire C 1, eluent acetonitrile / 0.2% aqueous TFA). This gives 0.6 mg (2% of theory) of the target compound.
  • HPLC Hydrochloric acid
  • a cellular assay is used to identify activators of the peroxisome proliferator-activated receptor alpha (PPAR-alpha).
  • the GAL4-PPAR ⁇ expression construct contains the ligand binding domain of PPARa (amino acids 167-468), which is PCR amplified and cloned into the vector pcDNA3.1. This vector already contains the GAL4 DNA binding domain (amino acids 1-147) of the vector pFC2-dbd (Stratagene).
  • the reporter construct containing five copies of the GAL4 binding site upstream of a thymidine kinase promoter, expresses the firefly luciferase (Photinus pyralis) upon activation and binding of GAL4-PPAR ⁇ .
  • CHO (Chinese hamster ovary) cells stably expressing the GAL4-PPAR ⁇ chimera and the luciferase reporter gene construct described above are in the medium (Optimem, GIBCO), 2% activated charcoal-purified fetal calf serum (Hyclone) the day before the test. , 1.35 mM sodium pyruvate (GIBCO), 0.2% sodium bicarbonate (GIBCO) with 1 x 10 3 cells plated in 96-well microtitre plates and maintained in a cell incubator (96% humidity, 5% v / v CO 2 , 37 ° C).
  • the substances to be tested are taken up in the above-mentioned medium, but without the addition of calf serum, and added to the cells.
  • the luciferase activity is measured using a video camera.
  • the measured relative light units give as a function of the substance concentration a sigmoidal stimulation curve.
  • the EC 50 values are calculated using the computer program GraphPad PRISM (version 3.02).
  • B-3 Test description for the discovery of pharmacologically active substances which increase the apoprotein Al (ApoAl) and HDL-cholesterol (HDL-C) in the serum of transgenic mice transfected with the human ApoAl gene (hApoAl) .
  • the substances to be tested for their HDL-C increasing activity in vivo are orally administered to male transgenic hApoAl mice.
  • the substances are administered orally once a day for 7 days.
  • the test substances are dissolved in a solution of Solutol HS 15 + ethanol + saline (0.9%). in the ratio 1 + 1 + 8 or dissolved in a solution of Solutol HS 15 + saline (0.9%) in the ratio 2 + 8.
  • the application of the dissolved substances takes place in a volume of 10 ml / kg body weight with a gavage. Animals which are treated in the same way, but only the solvent (10 ml / kg body weight) without test substance, serve as a control group.
  • each mouse Before the first substance administration, each mouse is sampled for the determination of ApoAl, serum cholesterol, HDL-C and serum triglycerides (TG) by puncture of the retroorbital venous plexus (initial value). Subsequently, the animals are given the test substance for the first time with a gavage. 24 hours after the last substance application (on the 8th day after the start of treatment), each animal is again drawn by puncture of the retroorbital venous plexus to determine the same parameters.
  • the blood samples are centrifuged and, after recovery of the serum, TG, cholesterol, HDL-C and human ApoAl are incubated with a Cobas Integra 400 plus instrument (Cobas Integra, Roche Diagnostics GmbH, Mannheim) using the respective cassettes (TRIGL, CHOL2, HDL-C and APOAT).
  • HDL-C is purified by gel filtration and post-column derivatization with MEGA cholesterol reagent (Merck KGaA) analogously to the method of Garber et al. [J. Lipid Res. 4L, 1020-1026 (2000)].
  • the effect of the test substances on the HDL-C, hApoAl or TG concentrations is determined by subtracting the measured value of the first blood sample (pre-value) from the measured value of the second blood sample (after treatment).
  • the differences of all HDL-C, hApoAl and TG values of a group are averaged and compared with the mean of the differences of the control group.
  • the statistical evaluation is done with Student's t-test after checking the variances for homogeneity.
  • Substances which increase the HDL-C of the treated animals statistically significantly (p ⁇ 0.05) by at least 20% or decrease the TG statistically significantly (p ⁇ 0.05) by at least 25% compared to those of the control group are considered to be pharmacologically active .
  • DHA desoxycorticosterone acetate
  • Uninephrectomized SD rats receive 1% sodium chloride in drinking water and once weekly a subcutaneous injection of desoxycorticosterone acetate (dissolved in sesame oil, Sigma) injected between the shoulder blades (high dose: 100 mg / kg / week sc, normal dose: 30 mg / kg / week sc).
  • the substances that are to be tested for their protective effect in vivo are administered by gavage or via the feed (Ssniff) or drinking water.
  • the substances are administered once a day for 4-6 weeks by gavage, food or drinking water.
  • the placebo group used is animals that are treated in the same way, but which either only receive the solvent or the feed or drinking water without the test substance.
  • the effect of the test substances is determined by measuring hemodynamic parameters [blood pressure, heart rate, inotropy (dp / dt), relaxation time (tau), maximum left ventricular pressure, left ventricular end-diastolic pressure (LVEDP)], weight determination of heart, kidney and lung Protein excretion and by measuring the gene expression of biomarkers (eg ANP, Atrial Natriuretic Peptide, and BNP, Brain Natriuretic Peptide) by RT / TaqMan PCR after RNA isolation from cardiac tissue determined.
  • biomarkers eg ANP, Atrial Natriuretic Peptide, and BNP, Brain Natriuretic Peptide
  • the statistical evaluation is done with Student's t-test after checking the variances for homogeneity.
  • test compounds are incubated in vitro with liver microsomes or preferably with primary fresh hepatocytes of various animal species (eg from rat and dog) as well as of human origin to obtain metabolite profiles of a complete hepatic phase I and phase II metabolism and compare.
  • the test compounds are incubated at a concentration of 10-20 ⁇ M.
  • stock solutions of the substances are prepared in a concentration of 1-2 mM in acetonitrile and then pipetted with a 1: 100 dilution in the incubation mixture.
  • Liver microsomes are incubated in 50 mM potassium phosphate buffer (pH 7.4) with and without NADPH-generating system consisting of 1 mM NADP + , 10 mM glucose-6-phosphate and 1 unit of glucose-6-phosphate dehydrogenase at 37 ° C incubated.
  • Primary hepatocytes are also incubated in suspension in Williams E medium at 37 ° C. After an incubation period of 0-4 hours, the incubation approaches are stopped with acetonitrile (final concentration about 30%) and the protein is centrifuged off at about 15,000 ⁇ g. The thus-stopped samples are analyzed either directly or stored at -20 0 C until analysis.
  • the analysis is carried out by high performance liquid chromatography with ultraviolet and mass spectrometric detection (HPLC-UV-MS / MS). For this, the supernatants of the incubation samples are chromatographed with suitable C18 reversed-phase columns and variable eluent mixtures of acetonitrile and 10 mM aqueous ammonium formate solution. The UV chromatograms in combination with mass spectrometric MS / MS data are used to identify and structure the metabolites.
  • HPLC-UV-MS / MS ultraviolet and mass spectrometric detection
  • the compounds according to the invention can be converted into pharmaceutical preparations as follows:
  • the mixture of compound of the invention, lactose and starch is granulated with a 5% solution (m / m) of the PVP in water.
  • the granules are mixed after drying with the magnesium stearate for 5 minutes.
  • This mixture is compressed with a conventional tablet press (for the tablet format see above).
  • a pressing force of 15 kN is used as a guideline for the compression.
  • a single dose of 100 mg of the compound of the invention corresponds to 10 ml of oral suspension.
  • the rhodigel is suspended in ethanol, the compound according to the invention is added to the suspension. While stirring, the addition of water. Until the completion of the swelling of Rhodigels is stirred for about 6 h.
  • a single dose of 100 mg of the compound according to the invention corresponds to 20 g of oral solution.
  • the compound of the invention is suspended in the mixture of polyethylene glycol and polysorbate with stirring. The stirring is continued until complete dissolution of the compound according to the invention.
  • the compound of the invention is dissolved in a concentration below saturation solubility in a physiologically acceptable solvent (e.g., isotonic saline, glucose solution 5% and / or PEG 400 solution 30%).
  • a physiologically acceptable solvent e.g., isotonic saline, glucose solution 5% and / or PEG 400 solution 30%.
  • the solution is sterile filtered and filled into sterile and pyrogen-free injection containers.

Abstract

The present invention relates to novel substituted 4-aminopyrimidine-5-carboxylic acid derivatives, method for the production thereof, use thereof for treating and/or preventing illnesses, and use for producing pharmaceuticals for treating and/or preventing illnesses, preferably for treating and/or preventing cardiovascular diseases, particularly dyslipidemias, atherosclerosis, and heart failure.

Description

Substituierte 4-Aminopyrimidin-5-carbonsäuren und ihre Verwendung Substituted 4-aminopyrimidine-5-carboxylic acids and their use
Die vorliegende Anmeldung betrifft neue substituierte 4-Aminopyrimidin-5-carbonsäure-Derivate, Verfahren zu ihrer Herstellung, ihre Verwendung zur Behandlung und/oder Prophylaxe von Krankheiten sowie ihre Verwendung zur Herstellung von Arzneimitteln zur Behandlung und/oder Prophylaxe von Krankheiten, vorzugsweise zur Behandlung und/oder Prophylaxe kardiovaskulärer Erkrankungen, insbesondere von Dyslipidämien, Arteriosklerose und Herzinsuffizienz.The present application relates to novel substituted 4-aminopyrimidine-5-carboxylic acid derivatives, processes for their preparation, their use for the treatment and / or prophylaxis of diseases and their use for the preparation of medicaments for the treatment and / or prophylaxis of diseases, preferably for treatment and / or prophylaxis of cardiovascular diseases, especially dyslipidaemias, atherosclerosis and cardiac insufficiency.
Trotz vielfacher Therapieerfolge bleiben kardiovaskuläre Erkrankungen ein ernstes Problem der öffentlichen Gesundheit. Während die Behandlung mit Statinen durch Hemmung der HMG-CoA- Reduktase sehr erfolgreich sowohl die Plasmakonzentrationen von LDL-Cholesterin (LDL-C) als auch die Mortalität von Risikopatienten senken, so fehlen heute überzeugende Behandlungsstrategien zur Therapie von Patienten mit ungünstigem HDL-C/LDL-C-Verhältnis oder der Hyper- triglyceridämie.Despite multiple therapeutic successes, cardiovascular disease remains a serious public health problem. While treatment with statins by inhibition of HMG-CoA reductase very successfully reduces both the plasma concentrations of LDL-cholesterol (LDL-C) and the mortality of high-risk patients, today there are no convincing treatment strategies for the treatment of patients with unfavorable HDL-C / LDL-C ratio or hyper-triglyceridemia.
Fibrate stellen neben Niacin bisher die einzige Therapieoption für Patienten dieser Risikogruppen dar. Sie senken erhöhte Triglyceride um 20-50%, erniedrigen LDL-C um 10-15%, verändern die LDL-Partikelgröße von atherogenem LDL geringer Dichte zu normal dichtem und weniger atherogenem LDL und erhöhen die HDL-Konzentration um 10-15%.In addition to niacin, fibrates are currently the only treatment option for patients in these risk groups. They lower elevated triglycerides by 20-50%, lower LDL-C by 10-15%, change the LDL particle size from low density atherogenic LDL to normal dense and less atherogenic LDL and increase the HDL concentration by 10-15%.
Fibrate wirken als schwache Agonisten des Peroxisom-Proliferator-aktivierten Rezeptors (PPAR)- alpha (Nature 1990, 347, 645-50). PPAR-alpha ist ein nuklearer Rezeptor, der die Expression von Zielgenen durch Bindung an DNA-Sequenzen im Promoter-Bereich dieser Gene [auch PPAR Response-Elemente (PPRE) genannt] reguliert. PPREs sind in einer Reihe von Genen identifiziert worden, welche für Proteine kodieren, die den Lipid-Metabolismus regulieren. PPAR-alpha ist hoch in der Leber exprimiert und seine Aktivierung führt unter anderem zu einer gesenkten VLDL- Produktion/-Sekretion sowie zu einer reduzierten Apolipoprotein CHI (ApoCIII)-Synthese. Im Gegensatz dazu wird die Synthese von Apolipoprotein Al (ApoAl) gesteigert.Fibrates act as weak agonists of the peroxisome proliferator-activated receptor (PPAR) -αα (Nature 1990, 347, 645-50). PPAR-alpha is a nuclear receptor that regulates the expression of target genes by binding to DNA sequences in the promoter region of these genes [also called PPAR response elements (PPRE)]. PPREs have been identified in a number of genes that encode proteins that regulate lipid metabolism. PPAR-alpha is highly expressed in the liver and its activation leads, inter alia, to decreased VLDL production / secretion and reduced apolipoprotein CHI (ApoCIII) synthesis. In contrast, the synthesis of apolipoprotein Al (ApoAl) is increased.
Ein Nachteil von bisher zugelassenen Fibraten ist ihre nur schwache Interaktion mit dem Rezeptor (EC50 im μM-Bereich), was wiederum zu den oben beschriebenen relativ geringen pharmakologischen Effekten führt.A disadvantage of previously approved fibrates is their weak interaction with the receptor (EC 50 in the μM range), which in turn leads to the relatively low pharmacological effects described above.
WO 99/41253 offenbart substituierte Pyrimidine zur Behandlung viraler Infektionen. JP 2001- 89452 beschreibt substituierte Pyrimidine zur Behandlung von Autoimmun-Erkrankungen. In WO 03/045941 werden Pyrimidine zur Behandlung von immunologischen und inflammatorischen Erkrankungen offenbart. In WO 2004/111014 werden substituierte Pyrimidine zur Behandlung von cystischer Fibrose beansprucht. 4-Aminopyrimidine werden in WO 2005/003099 als Ionenkanal- Modulatoren zur Behandlung von Schmerz, Arthritis, Migräne, Epilepsie und Inkontinenz beschrieben. WO 2005/040133 beansprucht Aminopyrimidine zur Behandlung von inflammtorischen Erkrankungen. WO 2005/110416 offenbart 4,5-disubstituierte 2-Arylpyrimidine als C5a-Rezeptorliganden zur Behandlung von inflammatorischen, immunologischen und kardiovaskulären Erkrankungen. In WO 2006/124874 werden unter anderem substituierte Pyrimidine zur Behandlung von Krebs beschrieben. In WO 2006/097220 werden 4-Phenoxy-2- phenylpyrimidincarbonsäuren und in WO 2008/031500 und WO 2008/031501 4-Phenoxy- bzw. 2- Phenoxynikotinsäuren als PPAR-alpha-Modulatoren zur Behandlung von Dyslipidämien und Arteriosklerose beansprucht.WO 99/41253 discloses substituted pyrimidines for the treatment of viral infections. JP 2001-89452 describes substituted pyrimidines for the treatment of autoimmune diseases. WO 03/045941 discloses pyrimidines for the treatment of immunological and inflammatory diseases. WO 2004/111014 claims substituted pyrimidines for the treatment of cystic fibrosis. 4-aminopyrimidines are described in WO 2005/003099 as ion channel modulators for the treatment of pain, arthritis, migraine, epilepsy and incontinence described. WO 2005/040133 claims aminopyrimidines for the treatment of inflammatory disorders. WO 2005/110416 discloses 4,5-disubstituted 2-arylpyrimidines as C5a receptor ligands for the treatment of inflammatory, immunological and cardiovascular diseases. Inter alia, substituted pyrimidines for the treatment of cancer are described in WO 2006/124874. WO 2006/097220 claims 4-phenoxy-2-phenylpyrimidinecarboxylic acids and, in WO 2008/031500 and WO 2008/031501, 4-phenoxy- or 2-phenoxynicotinic acids as PPAR-alpha modulators for the treatment of dyslipidemias and arteriosclerosis.
Aufgabe der vorliegenden Erfindung war die Bereitstellung neuer Verbindungen, die als PPAR- alpha-Modulatoren zur Behandlung und/oder Prophylaxe insbesondere kardiovaskulärer Erkrankungen eingesetzt werden können und eine verbesserte metabolische Stabilität gegenüber Verbindungen aus dem Stand der Technik aufweisen.It is an object of the present invention to provide novel compounds which can be used as PPAR-alpha modulators for the treatment and / or prophylaxis of, in particular, cardiovascular diseases and have improved metabolic stability compared to compounds of the prior art.
Gegenstand der vorliegenden Erfindung sind Verbindungen der allgemeinen Formel (I)The present invention relates to compounds of the general formula (I)
Figure imgf000003_0001
Figure imgf000003_0001
in welcherin which
R1 für Wasserstoff oder (CrC3)-Alkyl steht,R 1 is hydrogen or (C r C 3 ) -alkyl,
R2 für (CrC6)-Alkyl, (C3-C6)-Alkenyl oder (C3-C7)-Cycloalkyl steht,R 2 is (C r C6) alkyl, (C 3 -C 6) alkenyl or (C 3 -C 7) cycloalkyl,
wobei (Ci-Q)-Alkyl mit 1 oder 2 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor, Trifluormethyl, Hydroxy, (Ci-C4)-Alkoxy, Trifluormethoxy und (C3-C7)-Cycloalkyl substituiert sein kann,where (C 1 -C 4 ) -alkyl having 1 or 2 substituents independently of one another can be substituted from the group of fluorine, trifluoromethyl, hydroxyl, (C 1 -C 4 ) -alkoxy, trifluoromethoxy and (C 3 -C 7 ) -cycloalkyl,
worin (C3-C7)-Cycloalkyl seinerseits mit 1 oder 2 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor, Hydroxy, Oxo, (CrC4)-Alkyl, Trifluormethyl, 2,2,2-Trifluorethyl, (CrC4)-Alkoxy und Trifluormethoxy substitutiert sein kann, undwherein (C 3 -C 7 ) -cycloalkyl in turn having 1 or 2 substituents independently selected from the group fluorine, hydroxy, oxo, (C r C 4 ) alkyl, trifluoromethyl, 2,2,2-trifluoroethyl, (C r C 4 ) alkoxy and trifluoromethoxy can be substituted, and
wobei (C3-C7)-Cycloalkyl mit 1 oder 2 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor, Hydroxy, Oxo, (Ci-C4)-Alkyl, Trifluormethyl, 2,2,2-Trifluorethyl, (Ci-C4)-Alkoxy und Trifluormethoxy substitutiert sein kann,where (C 3 -C 7 ) -cycloalkyl having 1 or 2 substituents selected independently of one another from the group fluorine, hydroxyl, oxo, (Ci-C 4 ) alkyl, trifluoromethyl, 2,2,2-trifluoroethyl, (Ci-C 4 ) alkoxy and trifluoromethoxy can be substituted,
undand
wobei in allen genannten Cycloalkyl-Gruppen eine CH2-Einheit gegen Sauerstoff ausgetauscht sein kann,wherein in all said cycloalkyl groups a CH 2 unit may be exchanged for oxygen,
oderor
R1 und R2 zusammen mit dem Stickstoffatom, an das sie gebunden sind, einen Pyrrolidin- oder Piperidin-Ring bilden, welcher seinerseits mit 1 oder 2 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor, Trifluormethyl, (Ci-C4)-Alkyl, Hydroxy, (Ci-C4)-Alkoxy und Trifluormethoxy substituiert sein kann,R 1 and R 2 together with the nitrogen atom to which they are attached form a pyrrolidine or piperidine ring, which in turn is independently selected from the group of fluorine, trifluoromethyl, (C 1 -C 4 ) -alkyl by 1 or 2 substituents , Hydroxy, (C 1 -C 4 ) -alkoxy and trifluoromethoxy may be substituted,
R3 für (CrC4)-Alkyl oder Cyclopropyl steht,R 3 is (C r C 4) -alkyl or cyclopropyl,
wobei (C]-C4)-Alkyl mit 1 bis 3 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor und (Ci-C4)-Alkoxy substituiert sein kann,wherein (C] -C4) alkyl having 1 to 3 substituents independently selected from the group of fluorine and (Ci-C 4) -alkoxy,
R4 für Wasserstoff oder Fluor steht,R 4 is hydrogen or fluorine,
R5 für Wasserstoff, Fluor, Chlor oder Methyl steht,R 5 is hydrogen, fluorine, chlorine or methyl,
R6 für Wasserstoff, Halogen, Nitro, Cyano, Trifluormethyl, Methyl, Ethyl, Trifluormethoxy oder Methoxy steht,R 6 is hydrogen, halogen, nitro, cyano, trifluoromethyl, methyl, ethyl, trifluoromethoxy or methoxy,
R7 für Wasserstoff, Fluor, Chlor oder Methyl steht,R 7 is hydrogen, fluorine, chlorine or methyl,
wobei mindestens einer der Reste R4, R5, R6 und R7 von Wasserstoff verschieden ist,wherein at least one of R 4 , R 5 , R 6 and R 7 is different from hydrogen,
sowie ihre Salze, Solvate und Solvate der Salze.and their salts, solvates and solvates of the salts.
Erfindungsgemäße Verbindungen sind die Verbindungen der Formel (I) und deren Salze, Solvate und Solvate der Salze, die von Formel (I) umfassten Verbindungen der nachfolgend genannten Formeln und deren Salze, Solvate und Solvate der Salze sowie die von Formel (I) umfassten, nachfolgend als Ausführungsbeispiele genannten Verbindungen und deren Salze, Solvate und Solvate der Salze, soweit es sich bei den von Formel (I) umfassten, nachfolgend genannten Verbindungen nicht bereits um Salze, Solvate und Solvate der Salze handelt. Die erfindungsgemäßen Verbindungen können in Abhängigkeit von ihrer Struktur in stereoisomeren Formen (Enantiomere, Diastereomere) existieren. Die Erfindung umfasst deshalb die Enantiomeren oder Diastereomeren und ihre jeweiligen Mischungen. Aus solchen Mischungen von Enantiomeren und/oder Diastereomeren lassen sich die stereoisomer einheitlichen Bestandteile in bekannter Weise isolieren.Compounds according to the invention are the compounds of the formula (I) and their salts, solvates and solvates of the salts comprising the compounds of the formulas below and their salts, solvates and solvates of the salts and of the formula (I) encompassed by formula (I), hereinafter referred to as exemplary compounds and their salts, solvates and solvates of the salts, as far as the compounds of formula (I), the compounds mentioned below are not already salts, solvates and solvates of the salts. Depending on their structure, the compounds of the invention may exist in stereoisomeric forms (enantiomers, diastereomers). The invention therefore includes the enantiomers or diastereomers and their respective mixtures. From such mixtures of enantiomers and / or diastereomers, the stereoisomerically uniform components can be isolated in a known manner.
Sofern die erfindungsgemäßen Verbindungen in tautomeren Formen vorkommen können, umfasst die vorliegende Erfindung sämtliche tautomere Formen.If the compounds according to the invention can occur in tautomeric forms, the present invention encompasses all tautomeric forms.
Als Salze sind im Rahmen der vorliegenden Erfindung physiologisch unbedenkliche Salze der erfindungsgemäßen Verbindungen bevorzugt. Umfasst sind auch Salze, die für pharmazeutische Anwendungen selbst nicht geeignet sind, jedoch beispielsweise für die Isolierung oder Reinigung der erfindungsgemäßen Verbindungen verwendet werden können.Salts used in the context of the present invention are physiologically acceptable salts of the compounds according to the invention. Also included are salts which are themselves unsuitable for pharmaceutical applications but can be used, for example, for the isolation or purification of the compounds of the invention.
Physiologisch unbedenkliche Salze der erfindungsgemäßen Verbindungen umfassen Säureadditionssalze von Mineralsäuren, Carbonsäuren und Sulfonsäuren, z.B. Salze der Chlorwasserstoffsäure, Bromwasserstoffsäure, Schwefelsäure, Phosphorsäure, Methansulfonsäure, Ethan- sulfonsäure, Toluolsulfonsäure, Benzolsulfonsäure, Naphthalindisulfonsäure, Essigsäure, Trifluor- essigsäure, Propionsäure, Milchsäure, Weinsäure, Äpfelsäure, Zitronensäure, Fumarsäure, Maleinsäure und Benzoesäure.Physiologically acceptable salts of the compounds of the invention include acid addition salts of mineral acids, carboxylic acids and sulfonic acids, e.g. Salts of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, benzenesulfonic acid, naphthalenedisulfonic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
Physiologisch unbedenkliche Salze der erfindungsgemäßen Verbindungen umfassen auch Salze üblicher Basen, wie beispielhaft und vorzugsweise Alkalimetallsalze (z.B. Natrium- und Kalium- salze), Erdalkalisalze (z.B. Calcium- und Magnesiumsalze) und Ammoniumsalze, abgeleitet von Ammoniak oder organischen Aminen mit 1 bis 16 C-Atomen, wie beispielhaft und vorzugsweise Ethylamin, Diethylamin, Triethylamin, Ethyldiisopropylamin, Monoethanolamin, Diethanolamin, Triethanolamin, Dicyclohexylamin, Dimethylaminoethanol, Prokain, Dibenzylamin, N-Methyl- morpholin, Arginin, Lysin, Ethylendiamin und N-Methylpiperidin.Physiologically acceptable salts of the compounds according to the invention also include salts of customary bases, such as, by way of example and by way of preference, alkali metal salts (for example sodium and potassium salts), alkaline earth salts (for example calcium and magnesium salts) and ammonium salts derived from ammonia or organic amines having from 1 to 16 carbon atoms. Atoms, such as, by way of example and by way of preference, ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methylmorpholine, arginine, lysine, ethylenediamine and N-methylpiperidine.
Als Solvate werden im Rahmen der Erfindung solche Formen der erfindungsgemäßen Verbindungen bezeichnet, welche in festem oder flüssigem Zustand durch Koordination mit Lösungsmittelmolekülen einen Komplex bilden. Hydrate sind eine spezielle Form der Solvate, bei denen die Koordination mit Wasser erfolgt. Als Solvate sind im Rahmen der vorliegenden Erfindung Hydrate bevorzugt.Solvates in the context of the invention are those forms of the compounds according to the invention which form a complex in the solid or liquid state by coordination with solvent molecules. Hydrates are a special form of solvates that coordinate with water. As solvates, hydrates are preferred in the context of the present invention.
Außerdem umfasst die vorliegende Erfindung auch Prodrugs der erfindungsgemäßen Verbindungen. Der Begriff "Prodrugs" umfaßt Verbindungen, welche selbst biologisch aktiv oder inaktiv sein können, jedoch während ihrer Verweilzeit im Körper zu erfindungsgemäßen Verbindungen umgesetzt werden (beispielsweise metabolisch oder hydrolytisch).In addition, the present invention also includes prodrugs of the compounds of the invention. The term "prodrugs" includes compounds which themselves are biologically active or inactive may, however, be converted into compounds of the invention during their residence time in the body (for example metabolically or hydrolytically).
Insbesondere umfasst die vorliegende Erfindung auch hydrolysierbare Ester-Derivate der Carbonsäuren der Formel (I). Hierunter werden Ester verstanden, die in physiologischen Medien und ins- besondere in vivo auf enzymatischem oder chemischem Wege zu den freien Carbonsäuren hydro- lysiert werden können. Als solche Ester werden geradkettige oder verzweigte (d-C6)-Alkylester, in denen die Alkylgruppe mit Hydroxy,
Figure imgf000006_0001
Amino, Mono-(Ci-C4)-alkylamino und/ oder Di-(C i-C4)-alkylamino substituiert sein kann, bevorzugt. Besonders bevorzugt sind die Methyl- oder Ethylester der Verbindungen der Formel (I).
In particular, the present invention also includes hydrolyzable ester derivatives of the carboxylic acids of the formula (I). These are understood to mean esters which can be hydrolyzed in physiological media and in particular in vivo enzymatically or chemically to the free carboxylic acids. As such esters, straight-chain or branched (C 1 -C 6 ) -alkyl esters in which the alkyl group is hydroxyl,
Figure imgf000006_0001
Amino, mono- (Ci-C 4 ) -alkylamino and / or di- (C iC 4 ) -alkylamino may be substituted. Particularly preferred are the methyl or ethyl esters of the compounds of formula (I).
Im Rahmen der vorliegenden Erfindung haben die Substituenten, soweit nicht anders spezifiziert, die folgende Bedeutung:Unless otherwise specified, in the context of the present invention, the substituents have the following meaning:
Alkyl steht im Rahmen der Erfindung für einen linearen oder verzweigten Alkylrest mit der jeweils angegebenen Anzahl an Kohlenstoffatomen. Beispielhaft und vorzugsweise seien genannt: Methyl, Ethyl, n-Propyl, Isopropyl, n-Butyl, iso-Butyl, 1-Methylpropyl, tert.-Butyl, n-Pentyl, iso-Pentyl, 1- Ethylpropyl, 1-Methylbutyl, 2-Methylbutyl, 3-Methylbutyl, n-Hexyl, 1-Methylpentyl, 2- Methylpentyl, 3-Methylpentyl, 4-Methylpentyl, 3,3-Dimethylbutyl, 1-Ethylbutyl, 2-Ethylbutyl, 1 ,4-Dimethylpentyl, 4,4-Dimethylpentyl und 1 ,4,4-Trimethylpentyl.In the context of the invention, alkyl is a linear or branched alkyl radical having in each case the number of carbon atoms specified. By way of example and preferably mention may be made of: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, 1-methylpropyl, tert-butyl, n-pentyl, isopentyl, 1-ethylpropyl, 1-methylbutyl, 2 Methylbutyl, 3-methylbutyl, n -hexyl, 1-methylpentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 3,3-dimethylbutyl, 1-ethylbutyl, 2-ethylbutyl, 1, 4-dimethylpentyl, 4.4 Dimethylpentyl and 1,4,4-trimethylpentyl.
Alkenyl stehen im Rahmen der Erfindung für einen geradkettigen oder verzweigten Alkenylrest mit 3 bis 6 Kohlenstoffatomen und einer oder zwei Doppelbindungen. Bevorzugt ist ein gerad- kettiger oder verzweigter Alkenylrest mit 3 oder 4 Kohlenstoffatomen und einer Doppelbindung. Beispielhaft und vorzugsweise seien genannt: Allyl, Isopropenyl und n-But-2-en-l-yl.Alkenyl in the context of the invention is a straight-chain or branched alkenyl radical having 3 to 6 carbon atoms and one or two double bonds. Preference is given to a straight-chain or branched alkenyl radical having 3 or 4 carbon atoms and one double bond. By way of example and by way of preference: allyl, isopropenyl and n-but-2-en-1-yl.
Alkoxy steht im Rahmen der Erfindung für einen linearen oder verzweigten Alkoxyrest mit 1 bis 4 Kohlenstoffatomen. Beispielhaft und vorzugsweise seien genannt: Methoxy, Ethoxy, n-Propoxy, Isopropoxy, 1-Methylpropoxy, n-Butoxy, iso-Butoxy und tert.-Butoxy.Alkoxy in the context of the invention is a linear or branched alkoxy radical having 1 to 4 carbon atoms. Examples which may be mentioned are: methoxy, ethoxy, n-propoxy, isopropoxy, 1-methylpropoxy, n-butoxy, isobutoxy and tert-butoxy.
Cvcloalkyl steht in Rahmen der Erfindung für einen monocyclischen, gesättigten Alkylrest mit 3 bis 7 Kohlenstoffatomen. Beispielhaft und vorzugsweise seien genannt: Cyclopropyl, Cyclobutyl, Cyclopentyl, Cyclohexyl und Cycloheptyl.Cycloalkyl in the context of the invention is a monocyclic, saturated alkyl radical having 3 to 7 carbon atoms. Examples which may be mentioned by way of example include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
Halogen schließt im Rahmen der Erfindung Fluor, Chlor, Brom und Iod ein. Bevorzugt sind Chlor oder Fluor.Halogen in the context of the invention includes fluorine, chlorine, bromine and iodine. Preference is given to chlorine or fluorine.
Wenn Reste in den erfindungsgemäßen Verbindungen substituiert sind, können die Reste, soweit nicht anders spezifiziert, ein- oder mehrfach substituiert sein. Im Rahmen der vorliegenden Erfin- dung gilt, dass für alle Reste, die mehrfach auftreten, deren Bedeutung unabhängig voneinander ist. Eine Substitution mit ein, zwei oder drei gleichen oder verschiedenen Substituenten ist bevorzugt. Ganz besonders bevorzugt ist die Substitution mit einem Substituenten.If radicals are substituted in the compounds according to the invention, the radicals can, unless otherwise specified, be monosubstituted or polysubstituted. Within the scope of the present invention It holds true that for all radicals which occur several times, their meaning is independent of one another. Substitution with one, two or three identical or different substituents is preferred. Very particular preference is given to the substitution with a substituent.
Bevorzugt im Rahmen der vorliegenden Erfindung sind Verbindungen der Formel (I), in welcherPreferred in the context of the present invention are compounds of the formula (I) in which
R1 für Wasserstoff, Methyl oder Ethyl stehtR 1 is hydrogen, methyl or ethyl
R2 für (Ci-C6)-Alkyl, Cyclopropyl, Cyclopentyl oder Cyclohexyl steht,R 2 is (C 1 -C 6 ) -alkyl, cyclopropyl, cyclopentyl or cyclohexyl,
wobei (Ci-C6)-Alkyl mit einem Substituenten ausgewählt aus der Gruppe Fluor, Trifiuormethyl, Cyclopropyl, Cyclopentyl und Cyclohexyl substituiert sein kann,where (C 1 -C 6 ) -alkyl may be substituted by a substituent selected from the group consisting of fluorine, trifluoromethyl, cyclopropyl, cyclopentyl and cyclohexyl,
worin Cyclopropyl, Cyclopentyl und Cyclohexyl ihrerseits mit einem Substituenten ausgewählt aus der Gruppe Fluor, Methyl, Ethyl und Trifluormethyl substitutiert sein können,in which cyclopropyl, cyclopentyl and cyclohexyl may themselves be substituted by a substituent selected from the group consisting of fluorine, methyl, ethyl and trifluoromethyl,
undand
wobei Cyclopropyl, Cyclopentyl und Cyclohexyl mit einem Substituenten ausgewählt aus der Gruppe Fluor, Methyl, Ethyl und Trifluormethyl substitutiert sein können,where cyclopropyl, cyclopentyl and cyclohexyl can be substituted by a substituent selected from the group consisting of fluorine, methyl, ethyl and trifluoromethyl,
oderor
R1 und R2 zusammen mit dem Stickstoffatom, an das sie gebunden sind, einen Pyrrolidin- oder Piperidin-Ring bilden, welcher seinerseits mit einem Substituenten ausgewählt aus der Gruppe Fluor, Trifluormethyl, Methyl und Ethyl substituiert sein kann,R 1 and R 2 together with the nitrogen atom to which they are attached form a pyrrolidine or piperidine ring, which in turn may be substituted by a substituent selected from the group consisting of fluorine, trifluoromethyl, methyl and ethyl,
R3 für (Ci -O-Alkyl oder Trifluormethyl steht,R 3 is (C 1 -O-alkyl or trifluoromethyl,
R4 für Wasserstoff steht,R 4 is hydrogen,
R5 für Wasserstoff oder Fluor steht,R 5 is hydrogen or fluorine,
R6 für Wasserstoff, Fluor, Chlor, Trifluormethyl oder Methyl steht,R 6 is hydrogen, fluorine, chlorine, trifluoromethyl or methyl,
R7 für Wasserstoff oder Methyl steht,R 7 is hydrogen or methyl,
wobei mindestens einer der Reste R5, R6 und R7 von Wasserstoff verschieden ist,wherein at least one of R 5 , R 6 and R 7 is different from hydrogen,
sowie ihre Salze, Solvate und Solvate der Salze.and their salts, solvates and solvates of the salts.
Besonders bevorzugt sind Verbindungen der Formel (I), in welcher R1 für Wasserstoff oder Ethyl steht,Particular preference is given to compounds of the formula (I) in which R 1 is hydrogen or ethyl,
R2 für Ethyl, iso-Propyl, Cyclopropyl oder Cyclopropylmethyl steht,R 2 is ethyl, iso-propyl, cyclopropyl or cyclopropylmethyl,
R3 für Methyl, Ethyl oder iso-Propyl steht,R 3 is methyl, ethyl or iso-propyl,
R4 für Wasserstoff steht,R 4 is hydrogen,
R5 für Wasserstoff steht,R 5 is hydrogen,
R6 für Wasserstoff, Chlor oder Methyl steht,R 6 is hydrogen, chlorine or methyl,
R7 für Wasserstoff oder Methyl steht,R 7 is hydrogen or methyl,
wobei mindestens einer der Reste R6 und R7 von Wasserstoff verschieden ist,wherein at least one of R 6 and R 7 is different from hydrogen,
sowie ihre Salze, Solvate und Solvate der Salze.and their salts, solvates and solvates of the salts.
Besonders bevorzugt sind auch Verbindungen der Formel (I), in welcherParticular preference is also given to compounds of the formula (I) in which
R1 für Wasserstoff oder Ethyl steht,R 1 is hydrogen or ethyl,
R2 für Ethyl, iso-Propyl, iso-Butyl oder Cyclopropylmethyl steht,R 2 is ethyl, iso-propyl, iso-butyl or cyclopropylmethyl,
R3 für iso-Butyl steht,R 3 is iso-butyl,
R4 für Wasserstoff steht,R 4 is hydrogen,
R5 für Wasserstoff steht,R 5 is hydrogen,
R6 für Wasserstoff, Chlor oder Methyl steht,R 6 is hydrogen, chlorine or methyl,
R7 für Wasserstoff oder Methyl steht,R 7 is hydrogen or methyl,
wobei mindestens einer der Reste R6 und R7 von Wasserstoff verschieden ist,wherein at least one of R 6 and R 7 is different from hydrogen,
sowie ihre Salze, Solvate und Solvate der Salze.and their salts, solvates and solvates of the salts.
Die in den jeweiligen Kombinationen bzw. bevorzugten Kombinationen von Resten im einzelnen angegebenen Reste-Definitionen werden unabhängig von den jeweiligen angegebenen Kombinationen der Reste beliebig auch durch Reste-Definitionen anderer Kombinationen ersetzt.The residue definitions given in detail in the respective combinations or preferred combinations of residues are also replaced by residue definitions of other combinations, regardless of the particular combinations of the residues indicated.
Ganz besonders bevorzugt sind Kombinationen von zwei oder mehreren der oben genannten Vorzugsbereiche. Weiterer Gegenstand der Erfindung ist ein Verfahren zur Herstellung der erfindungsgemäßen Verbindungen der Formel (I), dadurch gekennzeichnet, dass man eine Verbindung der Formel (II)Very particular preference is given to combinations of two or more of the abovementioned preferred ranges. The invention further provides a process for the preparation of the compounds of the formula (I) according to the invention, which comprises reacting a compound of the formula (II)
Figure imgf000009_0001
Figure imgf000009_0001
in welcher R , R , R , R und R jeweils die oben angegebenen Bedeutungen haben,in which R, R, R, R and R each have the meanings given above,
undand
R8 für (C,-C4)-Alkyl steht,R 8 is (C 1 -C 4 ) -alkyl,
mit Hilfe eines geeigneten Chlorierungsmittels, wie beispielsweise Phosphoroxychlorid, in eine Verbindung der Formel (IH)with the aid of a suitable chlorinating agent, such as phosphorus oxychloride, into a compound of formula (IH)
Figure imgf000009_0002
Figure imgf000009_0002
in welcher R , R , R , R , R und R jeweils die oben angegebenen Bedeutungen haben, überfuhrtin which R, R, R, R, R and R each have the meanings given above, überfuhrt
und diese anschliessend in einem inerten Lösungsmittel in Gegenwart einer Base mit einer Verbindung der Formel (IV)and this then in an inert solvent in the presence of a base with a compound of formula (IV)
KK
\\
N-H y (IV),N-H y (IV),
in welcher R1 und R2 jeweils die oben angegebenen Bedeutungen haben,in which R 1 and R 2 each have the meanings given above,
zu Verbindungen der Formel (V)
Figure imgf000010_0001
to compounds of the formula (V)
Figure imgf000010_0001
in welcher R > ', D R^, T Rt I, τ R>4, R τ> 5, τ R> 6, r R> 7 und R jeweils die oben angegebenen Bedeutungen haben,in which R>, D R ^, T Rt I, Rτ4, R 5, Rτ6, R R7 and R each have the meanings given above,
umsetzt und diese durch basische oder saure Hydrolyse in die Carbonsäuren der Formel (I)and these by basic or acidic hydrolysis into the carboxylic acids of the formula (I)
Figure imgf000010_0002
Figure imgf000010_0002
in welcher R1, R2, R3, R4, R5, R6 und R7 jeweils die oben angegebenen Bedeutungen haben,in which R 1 , R 2 , R 3 , R 4 , R 5 , R 6 and R 7 each have the meanings given above,
überführttransferred
und die Verbindungen der Formel (I) gegebenenfalls mit den entsprechenden (i) Lösungsmitteln und/oder (ii) Basen oder Säuren zu ihren Solvaten, Salzen und/oder Solvaten der Salze umsetzt.and optionally reacting the compounds of the formula (I) with the corresponding (i) solvents and / or (ii) bases or acids to their solvates, salts and / or solvates of the salts.
Die Verbindungen der Formel (IV) sind kommerziell erhältlich, literaturbekannt oder können in Analogie zu literaturbekannten Verfahren hergestellt werden.The compounds of the formula (IV) are commercially available, known from the literature or can be prepared in analogy to processes known from the literature.
Die Verbindungen der Formel (II) können hergestellt werden, indem man Verbindungen der Formel (VI)The compounds of the formula (II) can be prepared by reacting compounds of the formula (VI)
Figure imgf000010_0003
(VI), in welcher R3 und R8 die oben angegebenen Bedeutungen haben,
Figure imgf000010_0003
(VI) in which R 3 and R 8 have the meanings given above,
zunächst in einem inerten Lösungsmittel in Gegenwart einer Base mit einer Verbindung der Formel (VΗ)first in an inert solvent in the presence of a base with a compound of the formula (VΗ)
Figure imgf000011_0001
Figure imgf000011_0001
in welcher R4, R5, R6 und R7 die oben angegebenen Bedeutungen haben,in which R 4 , R 5 , R 6 and R 7 have the meanings given above,
zu einer Verbindung der Formel (VIII)to a compound of formula (VIII)
Figure imgf000011_0002
Figure imgf000011_0002
in welcher R , R , R 1 R , R und R jeweils die oben angegebenen Bedeutungen haben,in which R, R, R 1 R, R and R each have the meanings given above,
umsetzt und diese anschliessend in einem inerten Lösungsmittel in Gegenwart einer Base und einer Bromquelle, wie beispielsweise N-Bromsuccinimid, sowie eines geeigneten Radikalstarters, wie beispielsweise Dibenzoylperoxid, zu einer Verbindung der Formel (IX)and then these in an inert solvent in the presence of a base and a bromine source, such as N-bromosuccinimide, and a suitable radical initiator, such as dibenzoyl peroxide, to give a compound of formula (IX)
Figure imgf000011_0003
Figure imgf000011_0003
in welcher R , R , R , R , R und R jeweils die oben angegebenen Bedeutungen haben,in which R, R, R, R, R and R each have the meanings given above,
oxidiert [ vgl. z.B. Veale C. A. et al., J. Org. Chem. 1993, 58, 4490-4493]. Die Verbindungen der Formeln (VI) und (VII} sind kommerziell erhältlich, literaturbekannt oder können in Analogie zu literaturbekannten Verfahren hergestellt werden.oxidized [cf. eg, Veale CA et al., J. Org. Chem. 1993, 58, 4490-4493]. The compounds of the formulas (VI) and (VII) are commercially available, known from the literature or can be prepared in analogy to processes known from the literature.
Die Umsetzung (II) — > (ET) erfolgt ohne Lösungsmittel oder gegebenenfalls in einem unter den Reaktionsbedingungen geeigneten inerten Lösungsmittel wie beispielsweise Kohlenwasserstoffe wie Benzol, Toluol, Xylol, Hexan, Cyclohexan oder Erdölfraktionen, oder andere Lösungsmittel wie Dimethylformamid, Dimethylsulfoxid, N-Methylpyrrolidon (ΝMP) oder Acetonitril. Ebenso ist es möglich, Gemische der genannten Lösungsmittel einzusetzen. Bevorzugt erfolgt die Reaktion ohne Lösungsmittel.The reaction (II) -> (ET) is carried out without a solvent or optionally in an inert solvent which is suitable under the reaction conditions, for example hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane or petroleum fractions, or other solvents such as dimethylformamide, dimethylsulfoxide, N-methylpyrrolidone (ΝMP) or acetonitrile. It is likewise possible to use mixtures of the solvents mentioned. The reaction preferably takes place without solvent.
Die Umsetzung (II) — > (IH) erfolgt im Allgemeinen in einem Temperaturbereich von 00C bis +1600C, bevorzugt bei +200C bis +1200C, gegebenenfalls in einer Mikrowelle. Die Reaktion kann bei normalem, erhöhtem oder bei erniedrigtem Druck durchgeführt werden (z.B. von 0.5 bis 5 bar). Im Allgemeinen arbeitet man bei Normaldruck.The reaction (II) -> (IH) is generally carried out in a temperature range from 0 0 C to +160 0 C, preferably at +20 0 C to +120 0 C, optionally in a microwave. The reaction can be carried out at normal, elevated or reduced pressure (eg from 0.5 to 5 bar). Generally, one works at normal pressure.
Inerte Lösungsmittel für den Verfahrensschritt (IE) + (IV) — > (V) sind beispielsweise Ether wie Diethylether, Dioxan, Tetrahydrofuran, Glykoldimethylether oder Diethylenglykoldimethylether, Kohlenwasserstoffe wie Benzol, Toluol, Xylol, Hexan, Cyclohexan oder Erdölfraktionen, oder andere Lösungsmittel wie Dimethylformamid, Dimethylsulfoxid, NN'-Dimethylpropylenharnstoff (DMPU), N-Methylpyrrolidinon (ΝMP), Pyridin, Aceton, 2-Butanon oder Acetonitril. Ebenso ist es möglich, Gemische der genannten Lösungsmittel einzusetzen. Bevorzugt wird Dimethylformamid oder Tetrahydrofuran verwendet.Inert solvents for process step (IE) + (IV) → (V) are, for example, ethers, such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons, such as benzene, toluene, xylene, hexane, cyclohexane or petroleum fractions, or other solvents, such as dimethylformamide , Dimethyl sulfoxide, N, N'-dimethylpropyleneurea (DMPU), N-methylpyrrolidinone (ΝMP), pyridine, acetone, 2-butanone or acetonitrile. It is likewise possible to use mixtures of the solvents mentioned. Preference is given to using dimethylformamide or tetrahydrofuran.
Als Base für den Verfahrensschritt (HT) + (IV) — »• (V) eignen sich übliche anorganische und organische Basen. Hierzu gehören insbesondere Alkalihydroxide wie beispielsweise Lithium-, Natrium- oder Kaliumhydroxid, Alkali- oder Erdalkalicarbonate wie Lithium-, Natrium-, Kalium-, Calcium- oder Cäsiumcarbonat, Alkalihydride wie Natrium- oder Kaliumhydrid, metallorganische Basen wie n-Butyllithium oder tert. organische Amine wie Diisopropylethylamin oder Triethylamin.. Bevorzugt ist Triethylamin. Die Base wird hierbei in einer Menge von 1 bis 5 Mol, bevorzugt in einer Menge von 1.2 bis 3 Mol, bezogen auf 1 Mol der Verbindung der Formel (IV), eingesetzt.Suitable bases for process step (HT) + (IV) -> • (V) are customary inorganic and organic bases. These include in particular alkali metal hydroxides such as lithium, sodium or potassium hydroxide, alkali metal or alkaline earth metal carbonates such as lithium, sodium, potassium, calcium or cesium carbonate, alkali metal hydrides such as sodium or potassium hydride, organometallic bases such as n-butyl lithium or tert. organic amines such as diisopropylethylamine or triethylamine. Preferred is triethylamine. The base is used here in an amount of 1 to 5 mol, preferably in an amount of 1.2 to 3 mol, based on 1 mol of the compound of formula (IV).
Die Umsetzung (IH) + (FV) — > (V) erfolgt im Allgemeinen in einem Temperaturbereich von 00C bis +1500C, bevorzugt bei +200C bis +1200C. Die Reaktion kann bei normalem, erhöhtem oder bei erniedrigtem Druck durchgeführt werden (z.B. von 0.5 bis 5 bar). Im Allgemeinen arbeitet man bei Normaldruck. Die Hydrolyse der Carbonsäureester in den Verfahrensschritten (V) — > (I) erfolgt nach üblichen Methoden, gegebenenfalls in einer Mikrowelle, indem man die Ester in inerten Lösungsmitteln mit Säuren oder Basen behandelt, wobei die bei letzterem zunächst entstehenden Salze durch nachfolgendes Behandeln mit Säure in die freien Carbonsäuren überführt werden. Im Falle der tert.-Butylester erfolgt die Esterspaltung bevorzugt mit Säuren.The reaction (IH) + (FV) -> (V) is generally carried out in a temperature range from 0 0 C to +150 0 C, preferably at +20 0 C to +120 0 C. The reaction can be at normal, elevated or be carried out at reduced pressure (eg from 0.5 to 5 bar). Generally, one works at normal pressure. The hydrolysis of the carboxylic acid esters in process steps (V) -> (I) by conventional methods, optionally in a microwave, by treating the esters in inert solvents with acids or bases, wherein the salts initially formed in the latter by subsequent treatment with acid be converted into the free carboxylic acids. In the case of the tert-butyl ester ester cleavage is preferably carried out with acids.
Als inerte Lösungsmittel eignen sich für die Hydrolyse der Carbonsäureester Wasser oder die für eine Esterspaltung üblichen organischen Lösungsmittel. Hierzu gehören insbesondere Alkohole wie Methanol, Ethanol, n-Propanol, Isopropanol, n-Butanol oder tert.-Butanol, Ether wie Diethyl- ether, Tetrahydrofuran, Dioxan oder Glykoldimethylether, oder andere Lösungsmittel wie Aceton, Acetonitril, Dichlormethan, Dimethylformamid oder Dimethylsulfoxid. Ebenso ist es möglich, Gemische der genannten Lösungsmittel einzusetzen. Im Falle einer basischen Ester-Hydrolyse werden bevorzugt Gemische von Wasser mit Dioxan, Tetrahydrofuran, Methanol und/oder Ethanol eingesetzt. Im Falle der Umsetzung mit Trifluoressigsäure wird bevorzugt Dichlormethan und im Falle der Umsetzung mit Chlorwasserstoff bevorzugt Tetrahydrofuran, Diethylether, Dioxan oder Wasser verwendet.Suitable inert solvents for the hydrolysis of the carboxylic acid esters are water or the organic solvents customary for ester cleavage. These include in particular alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, tetrahydrofuran, dioxane or glycol dimethyl ether, or other solvents such as acetone, acetonitrile, dichloromethane, dimethylformamide or dimethyl sulfoxide. It is likewise possible to use mixtures of the solvents mentioned. In the case of basic ester hydrolysis, preference is given to using mixtures of water with dioxane, tetrahydrofuran, methanol and / or ethanol. In the case of the reaction with trifluoroacetic acid, preference is given to using dichloromethane and, in the case of the reaction with hydrogen chloride, preferably tetrahydrofuran, diethyl ether, dioxane or water.
Als Basen eignen sich für die Ester-Hydrolyse die üblichen anorganischen Basen. Hierzu gehören insbesondere Alkali- oder Erdalkalihydroxide wie beispielsweise Natrium-, Lithium-, Kaliumoder Bariumhydroxid, oder Alkali- oder Erdalkalicarbonate wie Natrium-, Kalium- oder Calciumcarbonat. Bevorzugt werden Natriumhydroxid oder Kaliumhydroxid eingesetzt.Suitable bases for the ester hydrolysis are the customary inorganic bases. These include in particular alkali metal or alkaline earth metal hydroxides such as sodium, lithium, potassium or barium hydroxide, or alkali metal or alkaline earth metal carbonates such as sodium, potassium or calcium carbonate. Preference is given to using sodium hydroxide or potassium hydroxide.
Als Säuren eignen sich für die Esterspaltung im Allgemeinen Schwefelsäure, Chlorwasserstoff/ Salzsäure, Bromwasserstoff/Bromwasserstoffsäure, Phosphorsäure, Essigsäure, Trifluoressigsäure, Toluolsulfonsäure, Methansulfonsäure oder Trifluormethansulfonsäure oder deren Gemische gegebenenfalls unter Zusatz von Wasser. Bevorzugt sind Chlorwasserstoff oder Trifluoressigsäure im Falle der tert.-Butylester und Salzsäure im Falle der Methylester.Suitable acids for the ester cleavage are generally sulfuric acid, hydrochloric acid / hydrochloric acid, hydrobromic / hydrobromic acid, phosphoric acid, acetic acid, trifluoroacetic acid, toluenesulfonic acid, methanesulfonic acid or trifluoromethanesulfonic acid or mixtures thereof, optionally with the addition of water. Hydrogen chloride or trifluoroacetic acid are preferred in the case of the tert-butyl esters and hydrochloric acid in the case of the methyl esters.
Die Esterspaltung erfolgt im Allgemeinen in einem Temperaturbereich von 00C bis +1000C, bevorzugt bei 00C bis +500C. Die Umsetzung kann bei normalem, erhöhtem oder bei erniedrigtem Druck durchgeführt werden (z.B. von 0.5 bis 5 bar).The Esterspaltung is generally carried out in a temperature range of 0 0 C to +100 0 C, preferably from 0 0 C to +50 0 C. The reaction may be at atmospheric, elevated or reduced pressure is performed (for example from 0.5 to 5 bar) ,
Die Herstellung der erfindungsgemäßen Verbindungen kann durch das folgende Syntheseschema veranschaulicht werden: Schema 1The preparation of the compounds according to the invention can be illustrated by the following synthesis scheme: Scheme 1
Figure imgf000014_0001
Figure imgf000014_0002
Figure imgf000014_0003
Figure imgf000014_0001
Figure imgf000014_0002
Figure imgf000014_0003
[a): Natriumethanolat, Ethanol, Rückflußtemperatur; b): N-Bromsuccinimid, K2CO3, kat. Dibenzoylperoxid, Rückflußtemperatur; c): POCl3, Rückflußtemperatur; d): Triethylamin, Tetrahydrofüran, Rückflußtemperatur; e): NaOH, Ethanol/Wasser, Rückflußtemperatur oder Mikrowelle, 1400C].[a): sodium ethoxide, ethanol, reflux temperature; b): N-bromosuccinimide, K 2 CO 3 , cat. Dibenzoyl peroxide, reflux temperature; c): POCl 3 , reflux temperature; d): triethylamine, tetrahydrofuran, reflux temperature; e): NaOH, ethanol / water, reflux temperature or microwave, 140 0 C].
Die erfindungsgemäßen Verbindungen besitzen wertvolle pharmakologische Eigenschaften und können zur Vorbeugung und Behandlung von Erkrankungen bei Menschen und Tieren verwendet werden.The compounds according to the invention have valuable pharmacological properties and can be used for the prevention and treatment of diseases in humans and animals.
Die erfindungsgemäßen Verbindungen sind hochwirksame PPAR-alpha-Modulatoren und weisen zudem eine erhöhte metabolische Stabilität auf. Sie eignen sich insbesondere zur primären und/oder sekundären Prävention sowie Behandlung von kardiovaskulären Erkrankungen, die durch Störungen im Fettsäure- und Glukose-Metabolismus hervorgerufen werden. Solche Erkrankungen umfassen Dyslipidämien (Hypercholesterolämie, Hypertriglyceridämie, erhöhte Konzentrationen der postprandialen Plasma-Triglyceride, Hypoalphalipoproteinämie, kombinierte Hyperlipidämien), Arteriosklerose sowie metabolische Erkrankungen (Metabolisches Syndrom, Hyperglykämie, Insulin-abhängiger Diabetes, Nicht-Insulin-abhängiger Diabetes, Gestationsdiabetes, Hyperinsulinämie, Insulinresistenz, Glukose-Intoleranz, Fettsucht (Adipositas) und diabetische Spätfolgen wie Retinopathie, Nephropathie und Neuropathie).The compounds according to the invention are highly effective PPAR-alpha modulators and moreover have increased metabolic stability. They are particularly suitable for primary and / or secondary prevention and treatment of cardiovascular diseases caused by Disturbances in fatty acid and glucose metabolism are caused. Such disorders include dyslipidaemias (hypercholesterolemia, hypertriglyceridemia, elevated levels of postprandial plasma triglycerides, hypoalphalipoproteinemia, combined hyperlipidemias), arteriosclerosis, and metabolic disorders (metabolic syndrome, hyperglycemia, insulin-dependent diabetes, non-insulin-dependent diabetes, gestational diabetes, hyperinsulinemia, insulin resistance , Glucose intolerance, obesity (obesity) and diabetic sequelae such as retinopathy, nephropathy and neuropathy).
Als hochwirksame PPAR-alpha-Modulatoren eignen sich die erfmdungsgemäßen Verbindungen insbesondere auch zur primären und/oder sekundären Prävention sowie Behandlung der Herzinsuf- fizienz.As a highly effective PPAR-alpha modulators, the compounds according to the invention are also particularly suitable for primary and / or secondary prevention and treatment of cardiac insufficiency.
Im Sinne der vorliegenden Erfindung umfasst der Begriff Herzinsuffizienz auch spezifischere oder verwandte Krankheitsformen wie Rechtsherzinsuffizienz, Linksherzinsuffizienz, Globalinsuffizienz, durch Hypertonie induzierte Herzinsuffizienz, ischämische Kardiomyopathie, dilatative Kardiomyopathie, angeborene Herzfehler, Herzklappenfehler, Herzinsuffizienz bei Herzklappenfehlern, Mitralklappenstenose, Mitralklappeninsuffizienz, Aortenklappenstenose, Aortenklappeninsuffϊzienz, Trikuspidalstenose, Trikuspidalinsuffϊzienz, Pulmonalklappenstenose, Pulmonalklappeninsuffϊzienz, kombinierte Herzklappenfehler, Herzmuskelentzündung (Myokarditis), chronische Myokarditis, akute Myokarditis, virale Myokarditis, diabetische Herzinsuffizienz, alkoholtoxische Kardiomyopathie, kardiale Speichererkrankungen, diastolische Herzinsuffizienz sowie systolische Herzinsuffizienz.For the purposes of the present invention, the term cardiac insufficiency also encompasses more specific or related forms of disease such as right heart failure, left heart failure, global insufficiency, hypertension-induced heart failure, ischemic cardiomyopathy, dilated cardiomyopathy, congenital heart defects, valvular heart failure, valvular heart failure, mitral valve stenosis, mitral valve insufficiency, aortic valve stenosis, aortic valve insufficiency, tricuspid stenosis , Tricuspid insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined heart valve defects, myocarditis, chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic cardiomyopathy, cardiac storage disorders, diastolic heart failure, and systolic heart failure.
Weitere unabhängige Risikofaktoren für kardiovaskuläre Erkrankungen, welche sich durch die erfϊndungsgemäßen Verbindungen behandeln lassen, sind Bluthochdruck, Ischämie, Myokardinfarkt, Angina pectoris, Herzmuskelschwäche, Restenose, pulmonale Hypertonie, erhöhte Spiegel von Fibrinogen und von LDL geringer Dichte sowie erhöhte Konzentrationen von Plasminogen- aktivator-Inhibitor 1 (PAI-I).Other independent cardiovascular disease risk factors that can be treated by the compounds of the present invention are hypertension, ischemia, myocardial infarction, angina pectoris, cardiac insufficiency, restenosis, pulmonary hypertension, increased levels of fibrinogen and low density LDL, and elevated levels of plasminogen activator. Inhibitor 1 (PAI-I).
Darüber hinaus können die erfindungsgemäßen Verbindungen auch zur Behandlung und/oder Prävention von Krebserkrankungen wie beispielsweise Hautkrebs, Brustkrebs, Hirntumoren, Kopf- Hals-Tumoren, Liposarcomen, Karzinomen des Auges, des Magen-Darm-Traktes, der Schilddrüse, der Leber, der Bauchspeicheldrüse, der Atemwegsorgane, der Niere, der Harnleiter, der Prostata, des Genitaltraktes und deren Fernmetastasen sowie Lyphome, Sarkome und Leukämien eingesetzt werden.In addition, the compounds of the invention may also be used for the treatment and / or prevention of cancers such as skin cancer, breast cancer, brain tumors, head and neck cancer, liposarcoma, carcinoma of the eye, gastrointestinal tract, thyroid, liver, pancreas of the respiratory organs, the kidney, the ureter, the prostate, the genital tract and their distant metastases as well as lyphomas, sarcomas and leukemias.
Darüber hinaus können die erfϊndungsgemäßen Verbindungen auch zur Behandlung und/oder Prävention von mikro- und makrovaskulären Schädigungen (Vasculitis), Reperfusionsschäden, arteri- ellen sowie venösen Thrombosen, Ödemen, von Erkrankungen des Zentralen Nervensystems und neurodegenerativen Störungen (Schlaganfall, Alzheimer'sche Krankheit, Parkinson'sche Krankheit, Demenz, Epilepsie, Depressionen, Multiple Sklerose), von Entzündungserkrankungen, Immunerkrankungen (Morbus Crohn, Colitis ulcerosa, Lupus erythematodes, rheumatoide Arthritis, Asthma), chronisch-obstruktiven Atemwegserkrankungen (chronische Bronchitis, COPD), Nierenerkrankungen (Glomerulonephritis), Schilddrüsenerkrankungen (Hyperthyreose), Erkrankungen der Bauchspeicheldrüse (Pankreatitis), Leberfibrose, Hauterkrankungen (Psoriasis, Akne, Ekzeme, Neurodermitis, Dermatitis, Keratitis, Narbenbildung, Warzenbildung, Frostbeulen), Sepsis, viralen Erkrankungen (HPV, HCMV, HTV), Kachexie, Osteoporose, Gicht, Inkontinenz sowie zur Wundheilung, Angiogenese und zum Anti-Aging eingesetzt werden.In addition, the compounds according to the invention can also be used for the treatment and / or prevention of micro- and macrovascular damage (vasculitis), reperfusion damage, arterial as well as venous thromboses, edema, diseases of the central nervous system and neurodegenerative disorders (stroke, Alzheimer's disease, Parkinson's disease, dementia, epilepsy, depression, multiple sclerosis), inflammatory diseases, immune diseases (Crohn's disease, ulcerative colitis, lupus erythematosus, rheumatoid arthritis, asthma), chronic obstructive pulmonary diseases (chronic bronchitis, COPD), kidney disease (glomerulonephritis), thyroid disease (hyperthyroidism), diseases of the pancreas (pancreatitis), liver fibrosis, skin diseases (psoriasis, acne, eczema, atopic dermatitis, dermatitis, Keratitis, scarring, wart formation, chilblains), sepsis, viral diseases (HPV, HCMV, HTV), cachexia, osteoporosis, gout, incontinence as well as wound healing, angiogenesis and anti-aging.
Die Wirksamkeit der erfϊndungsgemäßen Verbindungen lässt sich z.B. in vitro durch den im Beispielteil beschriebenen Transaktivierungsassay prüfen.The effectiveness of the compounds of the invention can be e.g. in vitro by the transactivation assay described in the Examples section.
Die Wirksamkeit der erfindungsgemäßen Verbindungen in vivo lässt sich z.B. durch die im Beispielteil beschriebenen Untersuchungen prüfen.The efficacy of the compounds of the invention in vivo can be e.g. Check by the tests described in the example section.
Weiterer Gegenstand der vorliegenden Erfindung ist die Verwendung der erfindungsgemäßen Verbindungen zur Behandlung und/oder Prävention von Erkrankungen, insbesondere der zuvor genannten Erkrankungen.Another object of the present invention is the use of the compounds of the invention for the treatment and / or prevention of diseases, in particular the aforementioned diseases.
Weiterer Gegenstand der vorliegenden Erfindung ist die Verwendung der erfϊndungsgemäßen Verbindungen zur Herstellung eines Arzneimittels zur Behandlung und/oder Prävention von Erkran- kungen, insbesondere der zuvor genannten Erkrankungen.Another object of the present invention is the use of the erfϊndungsgemäßen compounds for the manufacture of a medicament for the treatment and / or prevention of Erkran- kungen, in particular the aforementioned diseases.
Weiterer Gegenstand der vorliegenden Erfindung ist ein Verfahren zur Behandlung und/oder Prävention von Erkrankungen, insbesondere der zuvor genannten Erkrankungen, unter Verwendung einer wirksamen Menge von mindestens einer der erfϊndungsgemäßen Verbindungen.Another object of the present invention is a method for the treatment and / or prevention of diseases, in particular the aforementioned diseases, using an effective amount of at least one of the inventive compounds.
Weiterer Gegenstand der vorliegenden Erfindung sind die erfindungsgemäßen Verbindungen zur Verwendung in einem Verfahren zur Behandlung und/oder Prophylaxe von Dyslipidämien, Arteriosklerose und Herzinsuffizienz.Another object of the present invention are the compounds of the invention for use in a method for the treatment and / or prophylaxis of dyslipidaemias, arteriosclerosis and heart failure.
Die erfindungsgemäßen Verbindungen können allein oder bei Bedarf in Kombination mit anderen Wirkstoffen eingesetzt werden. Weiterer Gegenstand der vorliegenden Erfindung sind Arzneimittel, enthaltend mindestens eine der erfindungsgemäßen Verbindungen und einen oder mehrere weitere Wirkstoffe, insbesondere zur Behandlung und/oder Prävention der zuvor genannten Erkrankungen. AIs geeignete Kombinationswirkstoffe seien beispielhaft und vorzugsweise genannt: den Fettstoffwechsel verändernde Wirkstoffe, Antidiabetika, Blutdruck-Senker, durchblutungsfördernd und/ oder antithrombotisch wirkende Mittel sowie Antioxidantien, Chemokin-Rezeptor-Antagonisten, p38-Kinase-Inhibitoren, NPY-Agonisten, Orexin-Agonisten, Anorektika, PAF-AH-Inhibitoren, Antiphlogistika (COX-Inhibitoren, LTB4-Rezeptor-Antagonisten), Analgetika (Aspirin), Antidepressiva und andere Psychopharmaka.The compounds of the invention may be used alone or as needed in combination with other agents. Another object of the present invention are pharmaceutical compositions containing at least one of the compounds of the invention and one or more other active ingredients, in particular for the treatment and / or prevention of the aforementioned diseases. AIs suitable combination active ingredients are exemplary and preferably mentioned: fat metabolism-altering agents, antidiabetics, blood pressure lowering agents, circulation-promoting and / or antithrombotic agents and antioxidants, chemokine receptor antagonists, p38 kinase inhibitors, NPY agonists, orexin agonists, Anorectics, PAF-AH inhibitors, anti-inflammatory drugs (COX inhibitors, LTB 4 receptor antagonists), analgesics (aspirin), antidepressants and other psychotropic drugs.
Gegenstand der vorliegenden Erfindung sind insbesondere Kombinationen mindestens einer der erfindungsgemäßen Verbindungen mit mindestens einem den Fettstoffwechsel verändernden Wirkstoff, einem Antidiabetikum, einem blutdrucksenkenden Wirkstoff und/oder einem antithrombo- tisch wirkenden Mittel.The present invention relates, in particular, to combinations of at least one of the compounds according to the invention with at least one lipid metabolism-altering active ingredient, an antidiabetic agent, a hypotensive agent and / or an antithrombotic agent.
Die erfindungsgemäßen Verbindungen können vorzugsweise mit einem oder mehrerenThe compounds of the invention may preferably be with one or more
• den Fettstoffwechsel verändernden Wirkstoffen, beispielhaft und vorzugsweise aus der Gruppe der HMG-CoA-Reduktase-Inhibitoren, Inhibitoren der HMG-CoA-Reduktase-Expres- sion, Squalensynthese-Inhibitoren, ACAT-Inhibitoren, LDL-Rezeptor-Induktoren, Choleste- rin- Absorptionshemmer, polymeren Gallensäureadsorber, Gallensäure-Reabsorptionshemmer,The substances which modify the lipid metabolism, by way of example and preferably from the group of HMG-CoA reductase inhibitors, inhibitors of HMG-CoA reductase expression, squalene synthesis inhibitors, ACAT inhibitors, LDL receptor inducers, cholesterol Absorption inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors,
MTP-Inhibitoren, Lipase-Inhibitoren, LpL-Aktivatoren, Fibrate, Niacin, CETP-Inhibitoren, PPAR-γ- und/oder PPAR-δ-Agonisten, RXR-Modulatoren, FXR-Modulatoren, LXR-Modula- toren, Thyroidhormone und/oder Thyroidmimetika, ATP-Citrat-Lyase-Inhibitoren, Lp(a)- Antagonisten, Cannabinoid-Rezeptor 1 -Antagonisten, Leptin-Rezeptor-Agonisten, Bombesin- Rezeptor-Agonisten, Histamin-Rezeptor-Agonisten sowie der Antioxidantien / Radikalfänger;MTP inhibitors, lipase inhibitors, LpL activators, fibrates, niacin, CETP inhibitors, PPAR-γ and / or PPAR-δ agonists, RXR modulators, FXR modulators, LXR modulators, thyroid hormones and / or thyroid mimetics, ATP citrate lyase inhibitors, Lp (a) antagonists, cannabinoid receptor 1 antagonists, leptin receptor agonists, bombesin receptor agonists, histamine receptor agonists and the antioxidants / free radical scavengers;
• Antidiabetika, die in der Roten Liste 2004/Η, Kapitel 12 genannt sind, sowie beispielhaft und vorzugsweise jenen aus der Gruppe der Sulphonylharnstoffe, Biguanide, Meglitinid-Derivate, Glukosidase-Inhibitoren, Oxadiazolidinone, Thiazolidindione, GLP 1 -Rezeptor-Agonisten, Glukagon- Antagonisten, Insulin-Sensitizer, CCK 1 -Rezeptor-Agonisten, Leptin-Rezeptor- Agonisten, Inhibitoren von Leberenzymen, die an der Stimulation der Glukoneogenese und/ oder Glykogenolyse beteiligt sind, Modulatoren der Glukoseaufnahme sowie der Kaliumkanalöffner, wie z.B. denjenigen, die in WO 97/26265 und WO 99/03861 offenbart sind;• Antidiabetics listed in the Red List 2004 / Η, Chapter 12, and by way of example and preferably those of the group of sulfonylureas, biguanides, meglitinide derivatives, glucosidase inhibitors, oxadiazolidinones, thiazolidinediones, GLP 1 receptor agonists, glucagon Antagonists, insulin sensitizers, CCK 1 receptor agonists, leptin receptor agonists, inhibitors of liver enzymes involved in the stimulation of gluconeogenesis and / or glycogenolysis, modulators of glucose uptake and potassium channel openers, such as those disclosed in WO 97/26265 and WO 99/03861;
• den Blutdruck senkenden Wirkstoffen, beispielhaft und vorzugsweise aus der Gruppe der Calcium-Antagonisten, Angiotensin Aü-Antagonisten, ACE-Inhibitoren, beta-Rezeptoren- Blocker, alpha-Rezeptoren-Blocker, ECE-Inhibitoren und der Vasopeptidase-Inhibitoren;The hypotensive agents, by way of example and preferably from the group of calcium antagonists, angiotensin Aue antagonists, ACE inhibitors, beta-receptor blockers, alpha-receptor blockers, ECE inhibitors and the vasopeptidase inhibitors;
• antithrombotisch wirkenden Mitteln, beispielhaft und vorzugsweise aus der Gruppe der Thrombozytenaggregationshemmer oder der Antikoagulantien; • Diuretika;Antithrombotic agents, by way of example and preferably from the group of platelet aggregation inhibitors or anticoagulants; • diuretics;
• Aldosteron- und Mineralokorticoid-Rezeptor- Antagonisten;• aldosterone and mineralocorticoid receptor antagonists;
• Vasopressin-Rezeptor-Antagonisten;Vasopressin receptor antagonists;
• organischen Nitraten und NO-Donatoren;• organic nitrates and NO donors;
• positiv-inotrop wirksamen Verbindungen;• positive inotropic compounds;
• Verbindungen, die den Abbau von cyclischem Guanosinmonophosphat (cGMP) und/oder cyclischem Adenosinmonophosphat (cAMP) inhibieren, wie beispielsweise Inhibitoren der Phosphodiesterasen (PDE) 1, 2, 3, 4 und/oder 5, insbesondere PDE 5-Inhibitoren wie Sildenafil, Vardenafil und Tadalafil sowie PDE 3-Inhibitoren wie Milrinone;Compounds which inhibit the degradation of cyclic guanosine monophosphate (cGMP) and / or cyclic adenosine monophosphate (cAMP), such as inhibitors of phosphodiesterases (PDE) 1, 2, 3, 4 and / or 5, in particular PDE 5 inhibitors such as sildenafil, Vardenafil and tadalafil and PDE 3 inhibitors such as milrinone;
• natriuretischen Peptiden, wie z.B. "atrial natriuretic peptide" (ANP, Anaritide), "B-type natriuretic peptide" oder "brain natriuretic peptide" (BNP, Nesiritide), "C-type natriuretic peptide" (CNP) sowie Urodilatin;Natriuretic peptides, e.g. atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP, Nesiritide), C-type natriuretic peptide (CNP) and urodilatin;
• Calcium-Sensitizern, wie beispielhaft und vorzugsweise Levosimendan;Calcium sensitizers, such as by way of example and preferably levosimendan;
• Kalium-Supplements;• potassium supplements;
• NO-unabhängigen, jedoch Häm-abhängigen Stimulatoren der Guanylatcyclase, wie insbesondere den in WO 00/06568, WO 00/06569, WO 02/42301 und WO 03/095451 beschriebenen Verbindungen;NO-independent, but heme-dependent guanylate cyclase stimulators, in particular the compounds described in WO 00/06568, WO 00/06569, WO 02/42301 and WO 03/095451;
• NO- und Häm-unabhängigen Aktivatoren der Guanylatcyclase, wie insbesondere den in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 und WO 02/070510 beschriebenen Verbindungen;Guanylate cyclase NO- and heme-independent activators, in particular the compounds described in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 and WO 02/070510;
• Inhibitoren der humanen neutrophilen Elastase (HNE), wie beispielsweise Sivelestat und DX- 890 (Reltran);• inhibitors of human neutrophil elastase (HNE), such as Sivelestat and DX-890 (Reltran);
• die Signaltransduktionskaskade inhibierenden Verbindungen, wie beispielsweise Tyrosin- kinase-Inhibitoren, insbesondere Sorafenib, Imatinib, Gefϊtinib und Erlotinib; und/oderThe signal transduction cascade inhibiting compounds, such as tyrosine kinase inhibitors, especially sorafenib, imatinib, Gefϊtinib and erlotinib; and or
• den Energiestoffwechsel des Herzens beeinflussenden Verbindungen, wie beispielweise Eto- moxir, Dichloracetat, Ranolazine und Trimetazidine• compounds affecting the energy metabolism of the heart, such as estimoxir, dichloroacetate, ranolazines and trimetazidines
kombiniert werden. Unter den Fettstoffwechsel verändernden Wirkstoffen werden vorzugsweise Verbindungen aus der Gruppe der HMG-CoA-Reduktase-Inhibitoren, Squalensynthese-Inhibitoren, ACAT-Inhibitoren, Cholesterin-Absoφtionshemmer, MTP-Inhibitoren, Lipase-Inhibitoren, Thyroidhormone und/oder Thyroidmimetika, Niacin-Rezeptor-Agonisten, CETP-Inhibitoren, PPAR-gamma-Agonisten, PPAR-delta-Agonisten, polymeren Gallensäureadsorber, Gallensäure-Reabsorptionshemmer, Anti- oxidantien / Radikalfänger sowie der Cannabinoid-Rezeptor 1 -Antagonisten verstanden.be combined. Among the lipid metabolism-changing active compounds are preferably compounds from the group of HMG-CoA reductase inhibitors, squalene synthesis inhibitors, ACAT inhibitors, cholesterol Absoφtionhemmer, MTP inhibitors, lipase inhibitors, thyroid hormones and / or thyroid mimetics, niacin receptor Agonists, CETP inhibitors, PPAR gamma agonists, PPAR delta agonists, polymeric bile acid adsorbers, bile acid reabsorption inhibitors, antioxidants / radical scavengers, and the cannabinoid receptor 1 antagonists.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfϊndungsgemäßen Verbindungen in Kombination mit einem HMG-CoA-Reduktase-Inhibitor aus der Klasse der Statine, wie beispielhaft und vorzugsweise Lovastatin, Simvastatin, Pravastatin, Fluvastatin, Atorvastatin, Rosu- vastatin, Cerivastatin oder Pitavastatin, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an HMG-CoA reductase inhibitor from the class of statins, such as by way of example and preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin, cerivastatin or pitavastatin ,
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfϊndungsgemäßen Verbindungen in Kombination mit einem Squalensynthese-Inhibitor, wie beispielhaft und vorzugsweise BMS-188494 oder TAK-475, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a squalene synthesis inhibitor, such as by way of example and preferably BMS-188494 or TAK-475.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindun- gen in Kombination mit einem ACAT-Inhibitor, wie beispielhaft und vorzugsweise Melinamide, Pactimibe, Eflucimibe oder SMP-797, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an ACAT inhibitor, such as by way of example and preferably melinamide, pactimibe, eflucimibe or SMP-797.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Cholesterin-Absorptionshemmer, wie beispielhaft und vorzugsweise Ezetimibe, Tiqueside oder Pamaqueside, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a cholesterol absorption inhibitor, such as by way of example and preferably ezetimibe, tiqueside or pamaqueside.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem MTP-Inhibitor, wie beispielhaft und vorzugsweise Implitapide oder JTT-130, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an MTP inhibitor, such as by way of example and preferably implitapide or JTT-130.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Lipase-Inhibitor, wie beispielhaft und vorzugsweise Orlistat, verab- reicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a lipase inhibitor, such as, for example and preferably, orlistat.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Thyroidhormon und/oder Thyroidmimetikum, wie beispielhaft und vorzugsweise D-Thyroxin oder 3,5,3'-Triiodothyronin (T3), verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thyroid hormone and / or thyroid mimetic, such as by way of example and preferably D-thyroxine or 3,5,3'-triiodothyronine (T3).
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindun- gen in Kombination mit einem Agonisten des Niacin-Rezeptors, wie beispielhaft und vorzugsweise Niacin, Acipimox, Acifran oder Radecol, verabreicht. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem CETP-Inhibitor, wie beispielhaft und vorzugsweise Torcetrapib, JTT-705 oder CETP Vaccine (Avant), verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an agonist of the niacin receptor, such as by way of example and preferably niacin, Acipimox, Acifran or Radecol. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a CETP inhibitor, such as, by way of example and by way of preference, torcetrapib, JTT-705 or CETP vaccine (Avant).
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindun- gen in Kombination mit einem PPAR-gamma-Agonisten, wie beispielhaft und vorzugsweise Pio- glitazone oder Rosiglitazone, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a PPAR-gamma agonist, such as by way of example and preferably pioglitazone or rosiglitazone.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem PPAR-delta-Agonisten, wie beispielhaft und vorzugsweise GW- 501516, verabreicht.In a preferred embodiment of the invention, the compounds of the invention are administered in combination with a PPAR delta agonist such as, for example and preferably, GW-501516.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem polymeren Gallensäureadsorber, wie beispielhaft und vorzugsweise Cholestyramin, Colestipol, Colesolvam, CholestaGel oder Colestimide, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a polymeric bile acid adsorbent such as, by way of example and by way of preference, cholestyramine, colestipol, colesolvam, cholesta gel or colestimide.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Gallensäure-Reabsorptionshemmer, wie beispielhaft und vorzugs- weise ASBT (= L3AT)-Inhibitoren wie z.B. AZD-7806, S-8921, AK-105, BARI-1741, SC-435 oder SC-635, verabreicht.In a preferred embodiment of the invention, the compounds of the invention are used in combination with a bile acid reabsorption inhibitor such as, for example, and preferably ASBT (= L3AT) inhibitors such as e.g. AZD-7806, S-8921, AK-105, BARI-1741, SC-435 or SC-635.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Antioxidans / Radikallanger, wie beispielhaft und vorzugsweise Probucol, AGI-1067, BO-653 oder AEOL-10150, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an antioxidant / radical catalyst such as, by way of example and by way of preference, probucol, AGI-1067, BO-653 or AEOL-10150.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Cannabinoid-Rezeptor 1 -Antagonisten, wie beispielhaft und vorzugsweise Rimonabant oder SR-147778, verabreicht.In a preferred embodiment of the invention, the compounds of the invention are administered in combination with a cannabinoid receptor 1 antagonist, such as by way of example and preferably rimonabant or SR-147778.
Unter Antidiabetika werden vorzugsweise Insulin und Insulinderivate sowie oral wirksame hypo- glykämische Wirkstoffe verstanden. Insulin und Insulinderivate umfasst hierbei sowohl Insuline tierischen, menschlichen oder biotechnologischen Ursprungs als auch Gemische hieraus. Die oral wirksamen hypoglykämischen Wirkstoffe umfassen vorzugsweise Sulphonylharnstoffe, Biguanide, Meglitinid-Derivate, Glukosidase-Inhibitoren und PPAR-gamma-Agonisten.Antidiabetic agents are preferably understood as meaning insulin and insulin derivatives as well as orally active hypoglycemic agents. Insulin and insulin derivatives here include both insulins of animal, human or biotechnological origin as well as mixtures thereof. The orally active hypoglycemic agents preferably include sulphonylureas, biguanides, meglitinide derivatives, glucosidase inhibitors and PPAR-gamma agonists.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit Insulin verabreicht. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Sulphonylharnstoff, wie beispielhaft und vorzugsweise Tolbutamid, Glibenclamid, Glimepirid, Glipizid oder Gliclazid, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with insulin. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a sulphonylurea, such as, by way of example and by way of preference, tolbutamide, glibenclamide, glimepiride, glipizide or gliclazide.
Bei einer bevorzugten Ausfuhrungsform der Erfindung werden die erfindungsgemäßen Verbindun- gen in Kombination mit einem Biguanid, wie beispielhaft und vorzugsweise Metformin, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a biguanide, such as by way of example and preferably metformin.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Meglitinid-Derivat, wie beispielhaft und vorzugsweise Repaglinid oder Nateglinid, verabreicht.In a preferred embodiment of the invention, the compounds of the invention are administered in combination with a meglitinide derivative, such as by way of example and preferably repaglinide or nateglinide.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Glukosidase-Inhibitor, wie beispielhaft und vorzugsweise Miglitol oder Acarbose, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a glucosidase inhibitor, such as by way of example and preferably miglitol or acarbose.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem PPAR-gamma-Agonisten beispielsweise aus der Klasse der Thia- zolidindione, wie beispielhaft und vorzugsweise Pioglitazone oder Rosiglitazone, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a PPAR-gamma agonist, for example from the class of thiazolidinediones, such as, by way of example and by way of preference, pioglitazone or rosiglitazone.
Unter den Blutdruck senkenden Mitteln werden vorzugsweise Verbindungen aus der Gruppe der Calcium-Antagonisten, Angiotensin AQ-Antagonisten, ACE-Inhibitoren, beta-Rezeptoren-Blocker, alpha-Rezeptoren-Blocker sowie der Diuretika verstanden.The blood pressure lowering agents are preferably understood as meaning compounds from the group of calcium antagonists, angiotensin AQ antagonists, ACE inhibitors, beta-receptor blockers, alpha-receptor blockers and diuretics.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindun- gen in Kombination mit einem Diuretikum, wie beispielhaft und vorzugsweise einem Schleifen- diuretikum wie Furosemid, Bumetanid oder Torsemid, oder einem Thiazid- oder Thiazid-ähnlichen Diuretikum wie Chlorthiazid oder Hydrochlorthiazid, verabreicht.In a preferred embodiment of the invention, the compounds of the invention are administered in combination with a diuretic, such as by way of example and preferably a loop diuretic such as furosemide, bumetanide or torsemide, or a thiazide or thiazide-like diuretic such as chlorothiazide or hydrochlorothiazide.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Aldosteron- oder Mineralokortikoid-Rezeptor-Antagonisten, wie beispielhaft und vorzugsweise Spironolacton oder Eplerenon, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an aldosterone or mineralocorticoid receptor antagonist, such as by way of example and preferably spironolactone or eplerenone.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Vasopressin-Rezeptor-Antagonisten, wie beispielhaft und vorzugsweise Conivaptan, Tolvaptan, Lixivaptan oder SR-121463, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a vasopressin receptor antagonist, such as by way of example and preferably Conivaptan, tolvaptan, lixivaptan or SR-121463.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindun- gen in Kombination mit einem organischen Nitrat oder NO-Donator, wie beispielhaft und Vorzugs- weise Natriumnitroprussid, Nitroglycerin, Isosorbidmononitrat, Isosorbiddinitrat, Molsidomin oder SIN-I, oder in Kombination mit inhalativem NO verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are used in combination with an organic nitrate or NO donor, such as by way of example and by way of example. Sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-I, or administered in combination with inhaled NO.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einer positiv-inotrop wirksamen Verbindung, wie beispielhaft und vor- zugsweise Herzglycosiden (Digoxin), beta-adrenergen und dopaminergen Agonisten wie Isopro- terenol, Adrenalin, Noradrenalin, Dopamin oder Dobutamin, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are used in combination with a positive inotropically active compound, such as by way of example and preferably cardiac glycosides (digoxin), beta-adrenergic and dopaminergic agonists such as isoproterenol, adrenaline, norepinephrine, dopamine or dobutamine, administered.
Bei einer bevorzugten Ausfuhrungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Calcium-Antagonisten, wie beispielhaft und vorzugsweise Nifedipin, Amlodipin, Verapamil oder Diltiazem, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a calcium antagonist, such as, by way of example and by way of preference, nifedipine, amlodipine, verapamil or diltiazem.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Angiotensin Aü-Antagonisten, wie beispielhaft und vorzugsweise Losartan, Valsartan, Candesartan, Embusartan oder Telmisartan, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an angiotensin Aü antagonist, such as by way of example and preferably losartan, valsartan, candesartan, embusartan or telmisartan.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem ACE-Inhibitor, wie beispielhaft und vorzugsweise Enalapril, Capto- pril, Ramipril, Delapril, Fosinopril, Quinopril, Perindopril oder Trandopril, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an ACE inhibitor such as, for example and preferably, enalapril, captopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem beta-Rezeptoren-Blocker, wie beispielhaft und vorzugsweise Propranolol, Atenolol, Timolol, Pindolol, Alprenolol, Oxprenolol, Penbutolol, Bupranolol, Metipra- nolol, Nadolol, Mepindolol, Carazalol, Sotalol, Metoprolol, Betaxolol, Celiprolol, Bisoprolol, Carteolol, Esmolol, Labetalol, Carvedilol, Adaprolol, Landiolol, Nebivolol, Epanolol oder Bucin- dolol, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are used in combination with a beta-receptor blocker, such as by way of example and preferably propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipranolol, nadolol, mepindolol, carazalol, Sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, Carteolol, esmolol, labetalol, carvedilol, adaprolol, landiolol, nebivolol, epanolol or bucine dolol administered.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem alpha-Rezeptoren-Blocker, wie beispielhaft und vorzugsweise Prazosin, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an alpha-receptor blocker, such as by way of example and preferably prazosin.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Antisympathotonikum, wie beispielhaft und vorzugsweise Reser- pin, Clonidin oder alpha-Methyl-Dopa, oder in Kombination mit einem Kaliumkanal-Agonisten, wie beispielhaft und vorzugsweise Minoxidil, Diazoxid, Dihydralazin oder Hydralazin, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are used in combination with an antisympathotonicum, such as by way of example and preferably reserpine, clonidine or alpha-methyl-dopa, or in combination with a potassium channel agonist such as, for example and preferably, minoxidil, diazoxide, dihydralazine or hydralazine.
Unter antithrombotisch wirkenden Mitteln werden vorzugsweise Verbindungen aus der Gruppe der Thrombozytenaggregationshemmer oder der Antikoagulantien verstanden. Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Thrombozytenaggregationshemmer, wie beispielhaft und vorzugsweise Aspirin, Clopidogrel, Ticlopidin oder Dipyridamol, verabreicht.Antithrombotic agents are preferably understood as meaning compounds from the group of platelet aggregation inhibitors or anticoagulants. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a platelet aggregation inhibitor, such as, by way of example and by way of preference, aspirin, clopidogrel, ticlopidine or dipyridamole.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindun- gen in Kombination mit einem Thrombin-Inhibitor, wie beispielhaft und vorzugsweise Ximelaga- tran, Melagatran, Bivalirudin oder Clexane, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thrombin inhibitor, such as by way of example and preferably ximelagarran, melagatran, bivalirudin or Clexane.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem GPÜb/IIIa-Antagonisten, wie beispielhaft und vorzugsweise Tiro- fiban oder Abciximab, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a GPUb / IIIa antagonist, such as by way of example and preferably tirofiban or abciximab.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Faktor Xa-Inhibitor, wie beispielhaft und vorzugsweise Rivaroxa- ban (BAY 59-7939), DU-176b, Apixaban, Otamixaban, Fidexaban, Razaxaban, Fondaparinux, Idraparinux, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 oder SSR-128428, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are used in combination with a factor Xa inhibitor, such as by way of example and preferably rivaroxaban (BAY 59-7939), DU-176b, apixaban, otamixaban, fidexaban, razaxaban, fondaparinux, idraparinux, PMD No. 3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit Heparin oder einem low molecular weight (LMW)-Heparin-Derivat verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with heparin or a low molecular weight (LMW) heparin derivative.
Bei einer bevorzugten Ausführungsform der Erfindung werden die erfindungsgemäßen Verbindungen in Kombination mit einem Vitamin K-Antagonisten, wie beispielhaft und vorzugsweise Coumarin, verabreicht.In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a vitamin K antagonist, such as by way of example and preferably coumarin.
Besonders bevorzugt im Rahmen der vorliegenden Erfindung sind Kombinationen enthaltend mindestens eine der erfindungsgemäßen Verbindungen sowie einen oder mehrere weitere Wirkstoffe ausgewählt aus der Gruppe bestehend aus HMG-CoA-Reduktase-Inhibitoren (Statine), Diuretika, beta-Rezeptoren-Blocker, organische Nitrate und NO-Donatoren, ACE-Inhibitoren, Angiotensin Aü-Antagonisten, Aldosteron- und Mineralokortikoid-Rezeptor-Antagonisten, Vasopressin-Rezep- tor-Antagonisten, Thrombozytenaggregationshemmer und Antikoagulantien, sowie deren Verwendung zur Behandlung und/oder Prävention der zuvor genannten Erkrankungen.Particularly preferred in the context of the present invention are combinations containing at least one of the compounds according to the invention and one or more further active compounds selected from the group consisting of HMG-CoA reductase inhibitors (statins), diuretics, beta-receptor blockers, organic nitrates and NO Donors, ACE inhibitors, angiotensin Aü antagonists, aldosterone and mineralocorticoid receptor antagonists, vasopressin receptor antagonists, platelet aggregation inhibitors and anticoagulants, and their use for the treatment and / or prevention of the aforementioned diseases.
Die erfindungsgemäßen Verbindungen können zur Behandlung und/oder Prophylaxe von Krebserkrankungen allein oder bei Bedarf in Kombination mit anderen Anti-Tumor Wirkstoffen eingesetzt werden. Gegenstand der vorliegenden Erfindung sind besonders Kombinationen mindestens einer der erfindungsgemäßen Verbindungen mit mindestens einem anderen AntiTumor Wirkstoff ausgewählt aus der Gruppe bestehend aus Alkylanzien, Antimetaboliten, von Pflanzen abgeleitete Anti-Tumor Wirkstoffe, Wirkstoffe der Hormontherapie, Topoisomerase Inhibitoren, Camptothecin-Derivate, Kinase Inhibitoren, zielgerichtete Medikamente, Antikörper, Immunokonjugate, Interferon und/oder Immunmodulatoren, Antiangiogen-wirksame Verbindungen, Antisense-RNA und RNA- Interferenz, und weitere Anti-Tumor Medikamente.The compounds according to the invention can be used for the treatment and / or prophylaxis of cancers alone or as needed in combination with other anti-tumor agents. The present invention particularly relates to combinations of at least one of the compounds according to the invention with at least one other anti-tumor active ingredient selected from the group consisting of alkylating agents, antimetabolites, of Plant derived anti-tumor agents, hormone therapy agents, topoisomerase inhibitors, camptothecin derivatives, kinase inhibitors, targeted drugs, antibodies, immunoconjugates, interferon and / or immunomodulators, antiangiogenic compounds, antisense RNA and RNA interference, and others Tumor medication.
Als geeignete Kombinationswirkstoffe seien beispielhaft und vorzugsweise genannt:As suitable combination active ingredients may be mentioned by way of example and preferably:
• Alkylanzien wie beispielsweise Chlormethin-N-oxid, Zyklophosphamid, Ifosfamid, Thiotepa, Ranimustin, Νimustin, Temozolomid, Altretamin, Apaziquon, Brostallicin, Bendamustin, Carmustin, Estramustin, Fotemustin, Glufosfamid, Mafosfamid und Mitolactol; Platinkoordinierte Alkylanzien wie beispielhaft Cisplatin, Carboplatin, Eptaplatin, Lobaplatin, Νedaplatin, Oxaliplatin und Satraplatin;Alkylating agents such as chloromethine N-oxide, cyclophosphamide, ifosfamide, thiotepa, ranimustine, Νimustin, temozolomide, altretamine, apaciquin, brostallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, mafosfamide and mitolactol; Platinum-coordinated alkylating agents such as, for example, cisplatin, carboplatin, eptaplatin, lobaplatin, Νedaplatin, oxaliplatin and satraplatin;
• Antimetabolite wie beispielsweise Methotrexat, 6-Mercaptopurin ribosid, Mercaptopurin, 5- Fluoruracil allein oder in Kombination mit Leucovorin, Tegafur, Doxifluridin, Carmofur, Cytarabin, Cytarabin Ocfosfat, Enocitabin, Gemcitabin, Fludarabin, 5-Azacitidin, Capecitabin, Cladribin, Clofarabin, Decitabin, Eflornithin, Ethynylcytidin, Cytosin- Arabinosid, Hydroxyharnstoff, Melphalan, Νelarabin, Νolatrexed, Ocfosfit, DinatriumAntimetabolites such as methotrexate, 6-mercaptopurine riboside, mercaptopurine, 5-fluorouracil alone or in combination with leucovorin, tegafur, doxifluridine, carmofur, cytarabine, cytarabine ocfosfate, enocitabine, gemcitabine, fludarabine, 5-azacitidine, capecitabine, cladribine, clofarabine, Decitabine, eflornithine, ethynylcytidine, cytosine arabinoside, hydroxyurea, melphalan, Νelarabine, Νolatrexed, ocfosfit, disodium
Premetrexed, Pentostatin, Pelitrexol, Raltitrexed, Triapin, Trimetrexat, Vidarabin, Vincristin, und Vinorelbin;Premetrexed, pentostatin, pelitrexol, raltitrexed, triapine, trimetrexate, vidarabine, vincristine, and vinorelbine;
• Wirkstoffe der Hormontherapie wie beispielsweise Exemestan, Lupron, Anastrozol, Doxercalciferol, Fadrozol, Formestan, 11-beta Hydroxysteroid Dehydrogenase- 1 Inhibitoren, 17-alpha Hydroxylase/ 17,20 Lyase Inhibitoren wie Abirateron Acetat, 5-alpha-Reduktase• hormonal agents such as exemestane, lupron, anastrozole, doxercalciferol, fadrozole, formestan, 11-beta hydroxysteroid dehydrogenase-1 inhibitors, 17-alpha hydroxylase / 17,20 lyase inhibitors such as abiraterone acetate, 5-alpha reductase
Inhibitoren wie beispielsweise Finasterid und Epristerid, Anti-Östrogene wie Tamoxifen- Zitrat und Fulvestrant, Trelstar, Toremifen, Raloxifen, Lasofoxifen, Letrozol, Anti-Androgene wie Bicalutamid, Flutamid, Mifepriston, Νilutamid, Casodex sowie Anti-Progesterone, und Kombinationen davon;Inhibitors such as finasteride and epristeride, anti-estrogens such as tamoxifen citrate and fulvestrant, trelstar, toremifene, raloxifene, lasofoxifene, letrozole, anti-androgens such as bicalutamide, flutamide, mifepristone, ilutamide, Casodex and anti-progesterone, and combinations thereof;
• von Pflanzen abgeleitete Anti-Tumor Wirkstoffe wie beispielsweise Mitosehemmer wie Epothilone (Sagopilon, Ixabepilon und Epothilon B), Vinblastin, Vinflunin, Docetaxel, und Paclitaxel;• plant-derived antitumor agents such as mitotic inhibitors such as epothilones (sagopilone, ixabepilone and epothilone B), vinblastine, vinflunine, docetaxel, and paclitaxel;
• Zytotoxische Topoisomerase Inhibitoren wie beispielsweise Aclarubicin, Doxorubicin, Amonafid, Belotecan, Camptothecin, 10-Hydroxycamptothecin, 9-Aminocamptothecin, Diflomotecan, Irinotecan, Topotecan, Edotecarin, Epimbicin, Etoposide, Exatecan, Gima- tecan, Lurtotecan, Mitoxantron, Pirambicin, Pixantron, Rubitecan, Sobuzoxan, Tafluposid, und Kombinationen davon; • Immunologische Wirkstoffe beispielhaft und vorzugsweise aus der Gruppe der Interferone wie z. B. Interferon alpha, Interferon alpha-2a, Interferon alpha-2b, Interferon beta, Interferon gamma- Ia und Interferon gamma-nl, und andere Immunstimulanzien wie z. B. L19-IL2 und andere IL2 Derivative, Filgrastim, Lentinan, Sizofilan, TheraCys, Ubenimex, Aldesleukin, Alemtuzumab, BAM-002, Dacarbazin, Daclizumab, Denileukin, Gemtuzumab, Ozogamicin,Cytotoxic topoisomerase inhibitors such as aclarubicin, doxorubicin, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflomotecan, irinotecan, topotecan, edotecarin, epimbicin, etoposide, exatecan, germantcan, lurtotecan, mitoxantrone, pirambicin, pixantrone, Rubitecan, Sobuzoxan, Tafluposide, and combinations thereof; • Immunological agents exemplified and preferably from the group of interferons such. Interferon alpha, Interferon alpha-2a, Interferon alpha-2b, Interferon beta, Interferon gamma- Ia and interferon gamma-nl, and other immune stimulants such. B. L19-IL2 and other IL2 Derivatives, Filgrastim, Lentinan, Sizofilan, TheraCys, Ubenimex, Aldesleukin, Alemtuzumab, BAM-002, Dacarbazine, Daclizumab, Denileukin, Gemtuzumab, Ozogamicin,
Ibritumomab, Imiquimod, Lenograstim, Lentinan, Melanom-Impfstoff (Corixa), Molgramostim, Sargramostim, Tasonermin, Tecleukin, Thymalasin, Tositumomab, Vimlizin, Epratuzumab, Mitumomab, Oregovomab, Pemtumomab und Proveng;Ibritumomab, Imiquimod, Lenograstim, Lentinan, Melanoma Vaccine (Corixa), Molgramostim, Sargramostim, Tasonermin, Tecleukin, Thymalasin, Tositumomab, Vimlizine, Epratuzumab, Mitumomab, Oregovomab, Pemtumomab and Proveng;
• Immunmodulatoren wie beispielsweise Krestin, Lentinan, Sizofiran, Picibanil, ProMun und Ubenimex;Immunomodulators such as Krestin, Lentinan, Sizofiran, Picibanil, ProMun and Ubenimex;
• Antiangiogen-wirksame Verbindungen wie beispielsweise Acitretin, Aflibercept, Angiostatin, Aplidin, Asentar, Axitinib, Recentin, Bevacizumab, Brivanib-Alaninat, Cilengtid, Combreta- statin, DAST, Endostatin, Fenretinid, Halofuginon, Pazopanib, Ranibizumab, Rebimastat, Removab, Revlimid, Sorafenib, Vatalanib, Squalamin, Sunitinib, Telatinib, Thalidomid, Ukrain und Vitaxin;Antiangiogenic compounds such as, for example, acitretin, aflibercept, angiostatin, aplidine, asentar, axitinib, recentin, bevacizumab, brivanib alaninate, cilengtide, combretastatin, DAST, endostatin, fenretinide, halofuginone, pazopanib, ranibizumab, rebarastat, Removab, Revlimid , Sorafenib, vatalanib, squalamine, sunitinib, telatinib, thalidomide, Ukrain and vitaxin;
• VEGF Inhibitoren wie beispielsweise Sorafenib, DAST, Bevacizumab, Sunitinib, Recentin, Axitinib, Aflibercept, Telatinib, Brivanib alaninat, Vatalanib, Pazopanib und Ranibizumab;VEGF inhibitors such as sorafenib, DAST, bevacizumab, sunitinib, recentin, axitinib, aflibercept, telatinib, brivanib alaninate, vatalanib, pazopanib, and ranibizumab;
• Antikörper wie beispielsweise Trastuzumab, Cetuximab, Bevacizumab, Rituximab, Ticilimumab, Ipilimumab, Lumiliximab, Catumaxomab, Atacicept, Oregovomab und Alemtu- zumab;• antibodies such as trastuzumab, cetuximab, bevacizumab, rituximab, ticilimumab, ipilimumab, lumiliximab, catumaxomab, atacicept, orregovomab and alemtumab;
• EGFR (HERl) Inhibitoren wie beispielsweise Cetuximab, Panitumumab, Vectibix, Gefitinib, Erlotinib und Zactima;EGFR (HERI) inhibitors such as cetuximab, panitumumab, vectibix, gefitinib, erlotinib and Zactima;
• HER2 Inhibitoren wie beispielsweise Lapatinib, Tratuzumab und Pertuzumab;• HER2 inhibitors such as lapatinib, tratuzumab and pertuzumab;
• mTOR Inhibitoren wie beispielsweise Temsirolimus, Sirolimus/Rapamycin und Everolimus;• mTOR inhibitors such as temsirolimus, sirolimus / rapamycin and everolimus;
• cMet Inhibitoren;• cMet inhibitors;
• PBK und AKT Inhibitoren;PBK and AKT inhibitors;
• CDK Inhibitoren wie beispielsweise Roscovitin und Flavopiridol; • Spindle assembly Checkpoint Inhibitoren und zielgerichtete Mitosehemmer wie PLK Inhibitoren, Aurora Inhibitoren (z.B. Hesperadin), Checkpoint-Kinase Inhibitoren und KSP Inhibitoren;CDK inhibitors such as roscovitine and flavopiridol; • Spindle assembly checkpoint inhibitors and targeted mitotic inhibitors such as PLK inhibitors, Aurora inhibitors (eg hesperadine), checkpoint kinase inhibitors and KSP inhibitors;
• HDAC Inhibitoren wie beispielsweise Panobinostat, Vorinostat, MS275, Belinostat und LBH589;• HDAC inhibitors such as Panobinostat, Vorinostat, MS275, Belinostat and LBH589;
• Inhibitoren der Histon-Methyltransferasen wie beispielsweise Vidaza;Inhibitors of histone methyltransferases such as Vidaza;
• HSP90 und HSP70 Inhibitoren;• HSP90 and HSP70 inhibitors;
• Proteasom Inhibitoren wie Bortezomib und Carfϊlzomib;Proteasome inhibitors such as bortezomib and carfϊlzomib;
• Serin-/Threonin-Kinase Inhibitoren wie beispielsweise MEK Inhibitoren und Raf Inhibitoren wie Sorafenib;Serine / threonine kinase inhibitors such as MEK inhibitors and Raf inhibitors such as sorafenib;
• Farnesyl Transferase Inhibitoren wie beispielsweise Tipifarnib;• farnesyl transferase inhibitors such as tipifarnib;
• Tyrosin-Kinase Inhibitoren wie beispielsweise Dasatinib, Nilotibib, DAST, Bosutinib, Sorafenib, Bevacizumab, Sunitinib, AZD2171, Axitinib, Aflibercept, Telatinib, Imatinib Mesylat, Brivanib Alaninat, Pazopanib, Ranibizumab, Vatalanib, Cetuximab, Panitumumab, Vectibix, Gefitinib, Erlotinib, Lapatinib, Tratuzumab, Pertuzumab und c-Kit Inhibitoren;Tyrosine kinase inhibitors such as dasatinib, nilotibib, DAST, bosutinib, sorafenib, bevacizumab, sunitinib, AZD2171, axitinib, aflibercept, telatinib, imatinib mesylate, brivanib alaninate, pazopanib, ranibizumab, vatalanib, cetuximab, panitumumab, vectibix, gefitinib, erlotinib , Lapatinib, tratuzumab, pertuzumab and c-kit inhibitors;
• Vitamin D Rezeptor Agonisten;• Vitamin D receptor agonists;
• Kortikoide, z.B. Dexamethason;Corticoids, e.g. dexamethasone;
• Thalidomid oder Thalidolid Analoga, z.B. LenalidomidThalidomide or thalidolide analogs, e.g. lenalidomide
• Bcl-2 Protein Inhibitoren wie beispielsweise Obatoclax, Oblimersen Natrium und Gossypol;Bcl-2 protein inhibitors such as Obatoclax, Oblimersen sodium and Gossypol;
• CD20 Rezeptor Antagonisten wie beispielsweise Rituximab;CD20 receptor antagonists such as rituximab;
• Ribonukleotid Reduktase Inhibitoren wie Gemcitabin;Ribonucleotide reductase inhibitors such as gemcitabine;
• Tumor Nekrose Apoptose einleitende Ligand-Rezeptor 1 Agonisten wie beispielsweise Mapatumumab;• Tumor necrosis apoptosis inducing ligand receptor 1 agonists such as Mapatumumab;
• 5-Hydroxytryptamin Rezeptor Antagonisten wie beispielsweise rEV598, Xaliprod, Palonosetron-Hydrochlorid, Granisetron, Zindol und AB-1001 ; • Integrin Inhibitoren einschliesslich Alpha5-betal Integrin Inhibitoren z. B. E7820, JSM 6425, Volociximab und Endostatin;5-hydroxytryptamine receptor antagonists such as rEV598, xaliprod, palonosetron hydrochloride, granisetron, zindol and AB-1001; • Integrin inhibitors including Alpha5-betal integrin inhibitors. E7820, JSM 6425, Volociximab and Endostatin;
• Androgen Rezeptor Antagonisten einschliesslich z.B. Nandrolon Decanoat, Fluoxymesteron, Android, Prost-aid, Andromustin, Bicalutamid, Flutamid, Apo-cyproteron, Apo-flutamid, Chlormadinon Acetat, Androcur, Tabi, Cyproteron Acetat und Nilutamid;Androgen receptor antagonists including e.g. Nandrolone Decanoate, Fluoxymesterone, Android, Prost-Aid, Andromustine, Bicalutamide, Flutamide, Apo-cyproterone, Apo-Flutamide, Chlormadinone Acetate, Androcur, Tabi, Cyproterone Acetate and Nilutamide;
• Aromatase Inhibitoren wie z. B. Anastrozol, Letrozol, Testolacton, Exemestan, Amino- glutethimid und Formestan;• aromatase inhibitors such. Anastrozole, letrozole, testolactone, exemestane, amino-glutethimide and formestane;
• Matrix Metalloproteinase Inhibitoren;• matrix metalloproteinase inhibitors;
• Weitere zur Krebstherapie eingesetzte Wirkstoffe einschliesslich z. B. Alitretinoin, Ampligen, Atrasentan Bexaroten, Bortezomib, Bosentan, Calcitriol, Exisulind, Fotemustin, Brondonat,• Other active ingredients used in cancer therapy, including Alitretinoin, Ampligen, Atrasentan Bexaroten, Bortezomib, Bosentan, Calcitriol, Exisulind, Fotemustin, Brondonat,
Miltefosin, Mitoxantron, I-Asparaginase, Procarbazin, Dacarbazin, Hydroxycarbamid, Pegaspargase, Pentostatin, Tazaroten, Velcad, Gallium-Nitrat, Canfosfamid, Darinaparsin und Tretinoin.Miltefosine, mitoxantrone, I-asparaginase, procarbazine, dacarbazine, hydroxycarbamide, pegaspargase, pentostatin, tazarotene, velcad, gallium nitrate, canfosfamide, darinaparsine and tretinoin.
Die erfindungsgemäßen Verbindungen können auch zur Behandlung von Krebserkrankungen in Verbindung mit Strahlentherapie und/ oder chirurgischen Eingriffen eingesetzt werden.The compounds of the invention may also be used to treat cancers associated with radiotherapy and / or surgery.
Weiterer Gegenstand der vorliegenden Erfindung sind Arzneimittel, die mindestens eine erfindungsgemäße Verbindung, üblicherweise zusammen mit einem oder mehreren inerten, nichttoxischen, pharmazeutisch geeigneten Hilfsstoffen enthalten, sowie deren Verwendung zu den zuvor genannten Zwecken.Another object of the present invention are pharmaceutical compositions containing at least one compound of the invention, usually together with one or more inert, non-toxic, pharmaceutically suitable excipients, and their use for the purposes mentioned above.
Die erfindungsgemäßen Verbindungen können systemisch und/oder lokal wirken. Zu diesem Zweck können sie auf geeignete Weise appliziert werden, wie z.B. oral, parenteral, pulmonal, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival, otisch oder als Implantat bzw. Stent.The compounds according to the invention can act systemically and / or locally. For this purpose, they may be applied in a suitable manner, e.g. oral, parenteral, pulmonary, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival, otic or as an implant or stent.
Für diese Applikationswege können die erfindungsgemäßen Verbindungen in geeigneten Appli- kationsformen verabreicht werden.For these administration routes, the compounds according to the invention can be administered in suitable administration forms.
Für die orale Applikation eignen sich nach dem Stand der Technik funktionierende, die erfin- dungsgemäßen Verbindungen schnell und/oder modifiziert abgebende Applikationsformen, die die erfindungsgemäßen Verbindungen in kristalliner und/oder amorphisierter und/oder gelöster Form enthalten, wie z.B. Tabletten (nicht-überzogene oder überzogene Tabletten, beispielsweise mit magensaftresistenten oder sich verzögert auflösenden oder unlöslichen Überzügen, die die Frei- setzung der erfϊndungsgemäßen Verbindung kontrollieren), in der Mundhöhle schnell zerfallende Tabletten oder Filme/Oblaten, Filme/Lyophylisate, Kapseln (beispielsweise Hart- oder Weichgelatinekapseln), Dragees, Granulate, Pellets, Pulver, Emulsionen, Suspensionen, Aerosole oder Lösungen.For the oral administration are according to the prior art functioning, the inventive compounds rapidly and / or modified donating application forms containing the compounds of the invention in crystalline and / or amorphized and / or dissolved form, such as tablets (uncoated or coated tablets, for example with enteric or delayed-dissolving or insoluble coatings, which control of the compound according to the invention), rapidly disintegrating tablets or films / wafers, films / lyophilisates, capsules (for example hard or soft gelatine capsules), dragees, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
Die parenterale Applikation kann unter Umgehung eines Resorptionsschrittes geschehen (z.B. intravenös, intraarteriell, intrakardial, intraspinal oder intralumbal) oder unter Einschaltung einer Resorption (z.B. intramuskulär, subcutan, intracutan, percutan oder intraperitoneal). Für die parenterale Applikation eignen sich als Applikationsformen u.a. Injektions- und Infusionszubereitungen in Form von Lösungen, Suspensionen, Emulsionen, Lyophilisaten oder sterilen Pulvern.Parenteral administration can be accomplished by bypassing a resorption step (e.g., intravenous, intraarterial, intracardiac, intraspinal, or intralumbar) or by resorting to absorption (e.g., intramuscular, subcutaneous, intracutaneous, percutaneous, or intraperitoneal). For parenteral administration are suitable as application forms u.a. Injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilisates or sterile powders.
Für die sonstigen Applikationswege eignen sich z.B. Inhalationsarzneiformen (u.a. Pulverinhalatoren, Nebulizer), Nasentropfen, -lösungen oder -sprays, lingual, sublingual oder buccal zu applizierende Tabletten, Filme/Oblaten oder Kapseln, Suppositorien, Ohren- oder Augenpräparationen, Vaginalkapseln, wäßrige Suspensionen (Lotionen, Schüttelmixturen), lipophile Suspensionen, Salben, Cremes, transdermale therapeutische Systeme (z.B. Pflaster), Milch, Pasten, Schäume, Streu- puder, Implantate oder Stents.For the other routes of administration are suitable, for example Inhalation medicaments (including powder inhalers, nebulizers), nasal drops, solutions or sprays, lingual, sublingual or buccal tablets, films / wafers or capsules, suppositories, ear or ophthalmic preparations, vaginal capsules, aqueous suspensions (lotions, shake mixtures), lipophilic suspensions , Ointments, creams, transdermal therapeutic systems (eg patches), milk, pastes, foams, scattering powders, implants or stents.
Bevorzugt sind die orale oder parenterale Applikation, insbesondere die orale und die intravenöse Applikation.Preference is given to oral or parenteral administration, in particular oral and intravenous administration.
Die erfϊndungsgemäßen Verbindungen können in die angeführten Applikationsformen überführt werden. Dies kann in an sich bekannter Weise durch Mischen mit inerten, nichttoxischen, pharma- zeutisch geeigneten Hilfsstoffen geschehen. Zu diesen Hilfsstoffen zählen u.a. Trägerstoffe (beispielsweise mikrokristalline Cellulose, Lactose, Mannitol), Lösungsmittel (z.B. flüssige PoIy- ethylenglycole), Emulgatoren und Dispergier- oder Netzmittel (beispielsweise Natriumdodecyl- sulfat, Polyoxysorbitanoleat), Bindemittel (beispielsweise Polyvinylpyrrolidon), synthetische und natürliche Polymere (beispielsweise Albumin), Stabilisatoren (z.B. Antioxidantien wie beispiels- weise Ascorbinsäure), Farbstoffe (z.B. anorganische Pigmente wie beispielsweise Eisenoxide) und Geschmacks- und/oder Geruchskorrigentien.The compounds according to the invention can be converted into the stated administration forms. This can be done in a conventional manner by mixing with inert, non-toxic, pharmaceutically suitable excipients. These adjuvants include, among others. Excipients (for example microcrystalline cellulose, lactose, mannitol), solvents (for example liquid polyethylene glycols), emulsifiers and dispersants or wetting agents (for example sodium dodecyl sulfate, polyoxysorbitanoleate), binders (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), Stabilizers (eg antioxidants such as ascorbic acid), dyes (eg inorganic pigments such as iron oxides) and flavor and / or odoriferous.
Im Allgemeinen hat es sich als vorteilhaft erwiesen, bei parenteraler Applikation Mengen von etwa 0.001 bis 1 mg/kg, vorzugsweise etwa 0.01 bis 0.5 mg/kg Körpergewicht zur Erzielung wirksamer Ergebnisse zu verabreichen. Bei oraler Applikation beträgt die Dosierung etwa 0.01 bis 100 mg/kg, vorzugsweise etwa 0.01 bis 20 mg/kg und ganz besonders bevorzugt 0.1 bis 10 mg/kg Körpergewicht. Trotzdem kann es gegebenenfalls erforderlich sein, von den genannten Mengen abzuweichen, und zwar in Abhängigkeit von Körpergewicht, Applikationsweg, individuellem Verhalten gegenüber dem Wirkstoff, Art der Zubereitung und Zeitpunkt bzw. Intervall, zu welchem die Applikation erfolgt. So kann es in einigen Fällen ausreichend sein, mit weniger als der vorgenannten Mindest- menge auszukommen, während in anderen Fällen die genannte obere Grenze überschritten werden muss. Im Falle der Applikation größerer Mengen kann es empfehlenswert sein, diese in mehreren Einzelgaben über den Tag zu verteilen.In general, it has proven to be advantageous, when administered parenterally, to administer amounts of about 0.001 to 1 mg / kg, preferably about 0.01 to 0.5 mg / kg of body weight, in order to achieve effective results. When administered orally, the dosage is about 0.01 to 100 mg / kg, preferably about 0.01 to 20 mg / kg and most preferably 0.1 to 10 mg / kg of body weight. Nevertheless, it may be necessary to deviate from the stated amounts, depending on body weight, route of administration, individual behavior towards the active ingredient, type of preparation and time or interval at which the application is carried out. For example, in some cases it may be sufficient to make do with less than the minimum amount mentioned above, while in other cases this upper limit must be exceeded. In the case of the application of larger quantities, it may be advisable to distribute these in several single doses throughout the day.
Die nachfolgenden Ausführungsbeispiele erläutern die Erfindung. Die Erfindung ist nicht auf die Beispiele beschränkt.The following embodiments illustrate the invention. The invention is not limited to the examples.
Die Prozentangaben in den folgenden Tests und Beispielen sind, sofern nicht anders angegeben, Gewichtsprozente; Teile sind Gewichtsteile. Lösungsmittelverhältnisse, Verdünnungsverhältnisse und Konzentrationsangaben von flüssig/flüssig-Lösungen beziehen sich jeweils auf das Volumen. The percentages in the following tests and examples are by weight unless otherwise indicated; Parts are parts by weight. Solvent ratios, dilution ratios and concentration data of liquid / liquid solutions are based on volume.
Abkürzungen und Akronyme:Abbreviations and acronyms:
abs. absolutSection. absolutely
Ac2O AcetanhydridAc 2 O acetic anhydride
AcOH Essigsäure aq. wässrig d TageAcOH acetic acid aq. Aqueous d days
DC DünnschichtchromatographieTLC thin layer chromatography
DCI direkte chemische Ionisation (bei MS) dd Dublett von Dublett (bei NMR)DCI direct chemical ionization (in MS) dd doublet of doublet (in NMR)
DDQ 2,3-Dichlor-5,6-dicyano-l,4-benzochinonDDQ 2,3-dichloro-5,6-dicyano-1,4-benzoquinone
DIAD DiisopropylazodicarboxylatDIAD diisopropyl azodicarboxylate
DMF DimethylformamidDMF dimethylformamide
DMSO Dimethylsulfoxid dt Dublett von Triplett (bei NMR) d. Th. der Theorie (bei Ausbeute) eq. Äquivalent(e)DMSO dimethyl sulfoxide dt doublet of triplet (by NMR) d. Th. Of theory (in yield) eq. Equivalent (s)
ESI Elektrospray-Ionisation (bei MS) h Stunde(n)ESI electrospray ionization (in MS) h hour (s)
HPLC Hochdruck-, HochleistungsflüssigchromatographieHPLC high pressure, high performance liquid chromatography
LHMDS Lithium-N,N-bistrimethylsilylamidLHMDS lithium N, N-bistrimethylsilylamide
LC-MS Flüssigchromatographie-gekoppelte Massenspektrometrie min Minute(n)LC-MS liquid chromatography-coupled mass spectrometry min minute (s)
MPLC MitteldruckchromatographieMPLC medium pressure chromatography
MS Massenspektrometrie mz Multiple«, zentriert (bei NMR) n-Bu n-ButylMS mass spectrometry mz multiple «, centered (by NMR) n-Bu n-butyl
NMR Kernresonanzspektrometrie o-Tol ortho-TolylNMR nuclear magnetic resonance spectrometry o-Tol ortho-tolyl
Ph PhenylPh phenyl
RP reverse phase (bei HPLC)RP reverse phase (on HPLC)
RT RaumtemperaturRT room temperature
R. Retentionszeit (bei HPLC) sbr Singulett, breit (bei NMR) sept Septett (bei NMR) t-Bu tert.-Butyl THF Tetrahydrofuran tt Triplett von Triplett (bei NMR)R. Retention time (by HPLC) sbr singlet, broad (by NMR) sept septet (by NMR) t-Bu tert-butyl THF tetrahydrofuran tt triplet of triplet (by NMR)
UV Ultraviolett-Spektrometrie v/v Volumen-zu- Volumen-Verhältnis (einer Mischung)UV ultraviolet spectrometry v / v volume-to-volume ratio (of a mixture)
LC-MS- und HPLC-Methoden:LC-MS and HPLC methods:
Methode 1 (LC-MS): Gerätetyp MS: Micromass ZQ; Gerätetyp HPLC: HP 1100 Series; UV DAD; Säule: Phenomenex Gemini 3μ 30 mm x 3.00 mm; Eluent A: 1 1 Wasser + 0.5 ml 50%-ige Ameisensäure, Eluent B: 1 1 Acetonitril + 0.5 ml 50%-ige Ameisensäure; Gradient: 0.0 min 90% A → 2.5 min 30% A → 3.0 min 5% A → 4.5 min 5% A; Fluss: 0.0 min 1 ml/min → 2.5 min/3.0 min/4.5 min 2 ml/min; Ofen: 500C; UV-Detektion: 210 nm.Method 1 (LC-MS): Device Type MS: Micromass ZQ; Device type HPLC: HP 1100 Series; UV DAD; Column: Phenomenex Gemini 3μ 30 mm x 3.00 mm; Eluent A: 1 l of water + 0.5 ml of 50% formic acid, eluent B: 1 l of acetonitrile + 0.5 ml of 50% formic acid; Gradient: 0.0 min 90% A → 2.5 min 30% A → 3.0 min 5% A → 4.5 min 5% A; Flow: 0.0 min 1 ml / min → 2.5 min / 3.0 min / 4.5 min 2 ml / min; Oven: 50 ° C .; UV detection: 210 nm.
Methode 2 (LC-MS): Gerätetyp MS: Micromass ZQ; Gerätetyp HPLC: Waters Alliance 2795; Säule: Phenomenex Synergi 2.5 μ MAX-RP 100A Mercury 20 mm x 4mm; Eluent A: 1 1 Wasser + 0.5 ml 50%ige Ameisensäure, Eluent B: 1 1 Acetonitril + 0.5 ml 50%ige Ameisensäure; Gradient: 0.0 min 90%A -» 0.1 min 90%A -» 3.0 min 5%A -» 4.0 min 5%A -» 4.01 min 90%A; Fluss: 2 ml/min;; Ofen: 500C; UV-Detektion: 210 nm.Method 2 (LC-MS): Device Type MS: Micromass ZQ; Device type HPLC: Waters Alliance 2795; Column: Phenomenex Synergi 2.5 μ MAX-RP 100A Mercury 20 mm x 4 mm; Eluent A: 1 l of water + 0.5 ml of 50% formic acid, eluent B: 1 l of acetonitrile + 0.5 ml of 50% formic acid; Gradient: 0.0 min 90% A - »0.1 min 90% A -» 3.0 min 5% A - »4.0 min 5% A -» 4.01 min 90% A; Flow: 2 ml / min ;; Oven: 50 ° C .; UV detection: 210 nm.
Methode 3 (LC-MS): Gerätetyp MS: Micromass ZQ; Gerätetyp HPLC: Waters Alliance 2795; Säule: Merck Chromolith SpeedROD RP-18e 100 x 4.6 mm; Eluent A: 1 1 Wasser + 0.5 ml 50%ige Ameisensäure; Eluent B: 1 1 Acetonitril + 0.5 ml 50%ige Ameisensäure; Gradient: 0.0 min 10% B-* 7.0 min 95% B-* 9.0 min 95% B; Ofen: 350C; Fluss: 0.0 min 1.0 ml/min -* 7.0 min 2.0 ml/min-* 9.0 min 2.0 ml/min; UV-Detektion: 210 nm Method 3 (LC-MS): Device Type MS: Micromass ZQ; Device type HPLC: Waters Alliance 2795; Column: Merck Chromolith SpeedROD RP-18e 100 x 4.6 mm; Eluent A: 1 liter of water + 0.5 ml of 50% formic acid; Eluent B: 1 liter acetonitrile + 0.5 ml 50% formic acid; Gradient: 0.0 min 10% B- * 7.0 min 95% B- * 9.0 min 95% B; Oven: 35 ° C; Flow: 0.0 min 1.0 ml / min - * 7.0 min 2.0 ml / min - * 9.0 min 2.0 ml / min; UV detection: 210 nm
Ausgangsverbindungen und Intermediate:Starting compounds and intermediates:
Beispiel IAExample IA
2-(4-Chloφhenyl)-4-methyl-6-oxo-l,4,5,6-tetrahydropyrimidin-5-carbonsäureethylester2- (4-Chloφhenyl) -4-methyl-6-oxo-l, 4,5,6-tetrahydropyrimidine-5-carboxylate
Figure imgf000032_0001
Figure imgf000032_0001
Zu einer Lösung aus 3.66 g (53.703 mmol) Natriumethylat und 5.00 g (29.537 mmol) 4-Chlorbenzamidin-Hydrochlorid in 50 ml Ethanol werden unter Argonatmosphäre 5.00 g (26.851 mmol) Ethylidenmalonsäurediethylester zugegeben. Das Reaktionsgemisch wird 1.5 h bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird der Ansatz eingeengt, in Dichlormethan aufgenommen und anschließend mit gesättigter wässriger Natriumchlorid-Lösung gewaschen, über Natriumsulfat getrocknet und eingeengt. Man erhält so 6.86 g (75% d. Th.) Rohprodukt in 86%iger Reinheit (LC-MS), welches ohne weitere Reinigungsoperation umgesetzt wird.To a solution of 3.66 g (53.703 mmol) of sodium ethylate and 5.00 g (29.537 mmol) of 4-chlorobenzamidine hydrochloride in 50 ml of ethanol are added under argon atmosphere 5.00 g (26.851 mmol) Ethylidenmalonsäurediethylester. The reaction mixture is stirred for 1.5 hours at reflux temperature. After cooling, the mixture is concentrated, taken up in dichloromethane and then washed with saturated aqueous sodium chloride solution, dried over sodium sulfate and concentrated. This gives 6.86 g (75% of theory) of crude product in 86% purity (LC-MS), which is reacted without further purification.
LC-MS (Methode 1): R, = 0.87 min; MS (ESIpos): m/z = 295 [M+H]+.LC-MS (Method 1): R, = 0.87 min; MS (ESIpos): m / z = 295 [M + H] + .
1H-NMR (400 MHz, DMSO-(I6): δ [ppm] = 1.20 (t, 3H), 1.28 (d, 3H), 3.40 (d, IH), 3.97-4.03 (m, IH), 4.16 (q, 2H), 7.52 (d, 2H), 7.85 (d, 2H), 9.53 (sbr, IH). 1 H-NMR (400 MHz, DMSO- (I 6 ): δ [ppm] = 1.20 (t, 3H), 1.28 (d, 3H), 3.40 (d, IH), 3.97-4.03 (m, IH), 4.16 (q, 2H), 7.52 (d, 2H), 7.85 (d, 2H), 9.53 (sbr, IH).
Beispiel 2AExample 2A
2-(4-Chlorphenyl)-4-methyl-6-oxo-l,6-dihydropyrimidin-5-carbonsäureethylester2- (4-chlorophenyl) -4-methyl-6-oxo-l, 6-dihydropyrimidine-5-carboxylate
Figure imgf000032_0002
Figure imgf000032_0002
Eine Lösung aus 3.00 g (ca. 8.753 mmol) Beispiel IA, 1.56g (8.753 mmol) N-Bromsuccinimid,A solution of 3.00 g (about 8.753 mmol) Example IA, 1.56 g (8.753 mmol) of N-bromosuccinimide,
212 mg (0.875 mmol) Dibenzoylperoxid und 1.81g (13.130 mmol) im Mörser gemahlenes Kaliumcarbonat in 150 ml Dioxan wird Ih bei Rückflußtemperatur unter Argonatmosphäre gerührt. Nach dem Abkühlen wird der Ansatz mit Wasser versetzt und anschließend mit Dichlormethan und Essigsäureethylester extrahiert. Die organische Phase wird mit einer gesättigten wässrigen Natriumchlorid-Lösung gewaschen, über Natriumsulfat getrocknet und eingeengt. Man erhält so 2.5g (66% d. Th.) Rohprodukt in 71%iger Reinheit (LC-MS), welches ohne weitere Reinigungsoperation umgesetzt wird.212 mg (0.875 mmol) of dibenzoyl peroxide and 1.81 g (13,130 mmol) of potassium carbonate ground in a mortar in 150 ml of dioxane are stirred at reflux temperature under an argon atmosphere. After cooling, the batch is mixed with water and then with Dichloromethane and ethyl acetate extracted. The organic phase is washed with a saturated aqueous sodium chloride solution, dried over sodium sulfate and concentrated. This gives 2.5 g (66% of theory) of crude product in 71% purity (LC-MS), which is reacted without further purification operation.
LC-MS (Methode 1): R, = 1.04 min; MS (ESIpos): m/z = 293 [M+H]+.LC-MS (Method 1): R, = 1.04 min; MS (ESIpos): m / z = 293 [M + H] + .
1H-NMR (400 MHz, DMSO-d*): δ [ppm] = 1.28 (t, 3H), 2.32 (s, 3H), 4.28 (q, 2H), 7.62 (d, 2H), 8.13 (d, 2H), 13.11 (sbr, IH). 1 H-NMR (400 MHz, DMSO-d *): δ [ppm] = 1.28 (t, 3H), 2.32 (s, 3H), 4.28 (q, 2H), 7.62 (d, 2H), 8.13 (i.e. , 2H), 13.11 (sbr, IH).
Beispiel 3AExample 3A
2-(4-Chlorphenyl)-4-(l-methylethyl)-6-oxo-l,4,5,6-tetrahydropyrimidin-5-carbonsäureethylester2- (4-chlorophenyl) -4- (l-methylethyl) -6-oxo-l, 4,5,6-tetrahydropyrimidine-5-carboxylate
Figure imgf000033_0001
Figure imgf000033_0001
Zu einer Lösung aus 1.59 g (23.336 mmol) Natriumethylat und 2.45 g (12.835 mmol) 4-Chlorbenzamidin-Hydrochlorid in 10 ml Ethanol unter Argonatmosphäre werden 2.5 g (11.668 mmol) (2-Methylpropyliden)malonsäuredieethylester zugegeben. Das Reaktionsgemisch wird 6.5h bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird der Ansatz eingeengt, in Dichlormethan aufgenommen und anschließend mit gesättigter wässriger Natriumchlorid-Lösung gewaschen, über Natriumsulfat getrocknet und eingeengt. Man erhält so 3.03 g (60% d. Th.) Rohprodukt (75 %ige Reinheit (LC-MS)), welches ohne weitere Reinigungsoperation umgesetzt wird.To a solution of 1.59 g (23.336 mmol) of sodium ethylate and 2.45 g (12.835 mmol) of 4-chlorobenzamidine hydrochloride in 10 ml of ethanol under an argon atmosphere, 2.5 g (11.668 mmol) of (2-methylpropylidene) malonate are added. The reaction mixture is stirred for 6.5 hours at reflux temperature. After cooling, the mixture is concentrated, taken up in dichloromethane and then washed with saturated aqueous sodium chloride solution, dried over sodium sulfate and concentrated. This gives 3.03 g (60% of theory) of crude product (75% purity (LC-MS)), which is reacted without further purification operation.
LC-MS (Methode 3): R, = 2.23 min; MS (ESIpos): m/z = 323 [M+H]+.LC-MS (Method 3): R, = 2.23 min; MS (ESIpos): m / z = 323 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 0.87 (d, 3H), 1.07 (d, 3H), 1.20 (t, 3H), 1.70-1.80 (m, IH), 3.50 (d, IH), 3.76 (dd, IH), 4.16 (q, 2H), 7.53 (d, 2H), 7.88 (d, 2H), 11.01 (sbr, IH). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 0.87 (d, 3H), 1.07 (d, 3H), 1.20 (t, 3H), 1.70-1.80 (m, IH), 3.50 (i.e. , IH), 3.76 (dd, IH), 4.16 (q, 2H), 7.53 (d, 2H), 7.88 (d, 2H), 11.01 (sbr, IH).
Beispiel 4AExample 4A
2-(4-Chlorphenyl)-4-( 1 -methylethyl)-6-oxo- 1 ,6-dihydropyrimidin-5 -carbonsäureethylester
Figure imgf000034_0001
Ethyl 2- (4-chlorophenyl) -4- (1-methylethyl) -6-oxo-1,6-dihydropyrimidine-5-carboxylate
Figure imgf000034_0001
Eine Lösung aus 3.00 g (ca. 6.970 mmol) Beispiel 3A, 1.24 g (6.970 mmol) N-Bromsuccinimid,A solution of 3.00 g (about 6.970 mmol) Example 3A, 1.24 g (6.970 mmol) of N-bromosuccinimide,
0.34 mg (1.394 mmol) Dibenzoylperoxid und 1.44 g (10.456 mmol) im Mörser gemahlenes0.34 mg (1.394 mmol) of dibenzoyl peroxide and 1.44 g (10.456 mmol) ground in a mortar
Kaliumcarbonat in 100 ml Dioxan wird über Nacht unter Argonatmosphäre bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird der Ansatz mit einer gesättigten wässrigenPotassium carbonate in 100 ml of dioxane is stirred overnight under argon atmosphere at reflux temperature. After cooling, the batch is washed with a saturated aqueous solution
Natriumthiosulfat-Lösung versetzt und anschließend die flüchtigen Komponenten amSodium thiosulfate solution and then the volatile components on
Rotationsverdampfer abgetrennt. Die wässrige Phase wird mit Essigsäureethylester extrahiert und die vereinigten organischen Phasen werden über Natriumsulfat getrocknet und dann eingeengt.Separated rotary evaporator. The aqueous phase is extracted with ethyl acetate and the combined organic phases are dried over sodium sulfate and then concentrated.
Man erhält so 2.46 g (51% d. Th.) Rohprodukt in 46 %iger Reinheit (LC-MS), welches ohne weitere Reinigungsoperation umgesetzt wird.This gives 2.46 g (51% of theory) of crude product in 46% purity (LC-MS), which is reacted without further purification operation.
LC-MS (Methode 3): R, = 2.50 min; MS (ESIpos): m/z = 321 [M+H]+.LC-MS (Method 3): R, = 2.50 min; MS (ESIpos): m / z = 321 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.18 (d, 6H), 1.26 (t, 3H), 4.23 (q, 2H), 7.54 (d, 2H), 8.20 (d, 2H). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.18 (d, 6H), 1.26 (t, 3H), 4.23 (q, 2H), 7.54 (d, 2H), 8.20 (d, 2H ).
Beispiel 5AExample 5A
4-Chlor-2-(4-Chlθφhenyl)-6-methylpyrimidin-5-carbonsäureethylester4-chloro-2- (4-Chlθφhenyl) -6-methylpyrimidine-5-carboxylate
Figure imgf000034_0002
Figure imgf000034_0002
1.5 g (5.124 mmol) Beispiel 2A in 19.1 ml (204.971 mmol) Phosphoroxychlorid werden 2h bei Rückflußtemperatur gerührt. Das Reaktionsgemisch wird einrotiert. Der Rückstand wird mit einer 25%igen wässrigen Ammoniumhydroxid-Lösung versetzt, mit IN Salzsäure auf pH 7 gestellt und anschließend mit Dichlormethan extrahiert. Die organische Phase wird über Natriumsulfat getrocknet und eingeengt. Man erhält so 1.38 g (87% d. Th.) der Zielverbindung.1.5 g (5.124 mmol) of Example 2A in 19.1 ml (204.971 mmol) of phosphorus oxychloride are stirred for 2 hours at reflux temperature. The reaction mixture is evaporated. The residue is treated with a 25% aqueous ammonium hydroxide solution, adjusted to pH 7 with 1N hydrochloric acid and then extracted with dichloromethane. The organic phase is dried over sodium sulfate and concentrated. This gives 1.38 g (87% of theory) of the target compound.
LC-MS (Methode 3): R, = 3.10 min; MS (ESIpos): m/z = 311 [M+H]+. Beispiel 6ALC-MS (Method 3): R, = 3.10 min; MS (ESIpos): m / z = 311 [M + H] + . Example 6A
2-(4-Chloφhenyl)-4-methyl-6-[(l-methylethyl)amino]pyrimidin-5-carbonsäureethylester2- (4-Chloφhenyl) -4-methyl-6 - [(l-methylethyl) amino] pyrimidin-5-carboxylate
Figure imgf000035_0001
Figure imgf000035_0001
Zu einer Lösung aus 250 mg (0.803 mmol) Beispiel 5A in 4 ml Tetrahydrofuran wird 203 mg (2.01 mmol) Triethylamin und 71 mg (1.205 mmol) Isopropylamin addiert. Das Reaktionsgemisch wird über Nacht bei 800C gerührt. Die Mischung wird einrotiert und ohne weitere Aufarbeitung weiter eingesetzt. Man erhält so 250 mg (91% d. Th.) der ZielverbindungTo a solution of 250 mg (0.803 mmol) of Example 5A in 4 ml of tetrahydrofuran is added 203 mg (2.01 mmol) of triethylamine and 71 mg (1.205 mmol) of isopropylamine. The reaction mixture is stirred overnight at 80 0 C. The mixture is concentrated by rotary evaporation and used without further workup. This gives 250 mg (91% of theory) of the target compound
LC-MS (Methode 3): R, = 3.29 min; MS (ESIpos): m/z = 334 [M+H]+.LC-MS (Method 3): R, = 3.29 min; MS (ESIpos): m / z = 334 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] =1.26 (d, 6H), 1.34 (t, 3H), 2.60 (t, 3H), 3.23-3.30 (m, IH), 4.32 (q, 2H), 7.56 (d, 2H), 7.90 (sbr, IH), 8.36 (d, 2H). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.26 (d, 6H), 1.34 (t, 3H), 2.60 (t, 3H), 3.23-3.30 (m, IH), 4.32 (q , 2H), 7.56 (d, 2H), 7.90 (sbr, IH), 8.36 (d, 2H).
Beispiel 7AExample 7A
2-(4-Chloφhenyl)-4-[(cyclopropyhnethyl)amino]-6-methylpyrimidin-5-carbonsäureethylester2- (4-Chloφhenyl) -4 - [(cyclopropyhnethyl) amino] -6-methylpyrimidine-5-carboxylate
Figure imgf000035_0002
Figure imgf000035_0002
Zu einer Lösung aus 100 mg (0.321 mmol) Beispiel 5A in 1.6 ml Tetrahydrofuran wird 81 mg (0.803 mmol) Triethylamin und 34 mg (0.482 mmol) Cyclopropylmethylamin addiert. Das Reaktionsgemisch wird über Nacht bei 800C gerührt. Die Mischung wird einrotiert und ohne weitere Aufarbeitung weiter eingesetzt. Man erhält so 100 mg (97% d. Th.) der Zielverbindung LC-MS (Methode 3): R, = 3.28 min; MS (ESIpos): m/z = 346 [M+H]+.To a solution of 100 mg (0.321 mmol) of Example 5A in 1.6 ml of tetrahydrofuran is added 81 mg (0.803 mmol) of triethylamine and 34 mg (0.482 mmol) of cyclopropylmethylamine. The reaction mixture is stirred overnight at 80 0 C. The mixture is concentrated by rotary evaporation and used without further workup. This gives 100 mg (97% of theory) of the target compound LC-MS (Method 3): R, = 3.28 min; MS (ESIpos): m / z = 346 [M + H] + .
1H-NMR (400 MHz, DMSO-Cl6): δ [ppm] = 0.29-0.32 (m, 2H), 0.46-0.50 (m, 2H), 1.35 (t, 3H), 2.60 (s, 3H), 3.45 (dd, 2H), 4.36 (q, 2H), 7.57 (d, 2H), 8.22 (sbr, IH), 8.37 (d, 2H). 1 H-NMR (400 MHz, DMSO-Cl 6 ): δ [ppm] = 0.29-0.32 (m, 2H), 0.46-0.50 (m, 2H), 1.35 (t, 3H), 2.60 (s, 3H) , 3.45 (dd, 2H), 4.36 (q, 2H), 7.57 (d, 2H), 8.22 (sbr, IH), 8.37 (d, 2H).
Beispiel 8AExample 8A
4-Chlor-2-(4-chlorphenyl)-6-( 1 -methylethyl)pyrimidin-5- carbonsäureethylester4-Chloro-2- (4-chlorophenyl) -6- (1-methylethyl) pyrimidine-5-carboxylic acid ethyl ester
Figure imgf000036_0001
Figure imgf000036_0001
1.5 g (ca. 2.151 mmol) Beispiel 4A in 8 ml (86.041 mmol) Phosphoroxychlorid werden 2h bei Rückflußtemperatur gerührt. Das Reaktionsgemisch wird einrotiert. Der Rückstand wird mit einer 25%igen wässrigen Ammoniumhydroxid-Lösung versetzt, mit IN Salzsäure auf pH 7 gestellt und anschließend mit Dichlormethan extrahiert. Die organische Phase wird über Natriumsulfat getrocknet und eingeengt. Der Rückstand wird an Kieselgel säulenchromatographiert (Laufmittel: Cyclohexan/Essigsäureethylester 50/1 → 20/1). . Man erhält so 540 mg (55% d. Th.) Rohprodukt in 74 %iger Reinheit (LC-MS), welches ohne weitere Reinigungsoperation umgesetzt wird.1.5 g (about 2151 mmol) of Example 4A in 8 ml (86.041 mmol) of phosphorus oxychloride are stirred for 2 h at reflux temperature. The reaction mixture is evaporated. The residue is treated with a 25% aqueous ammonium hydroxide solution, adjusted to pH 7 with 1N hydrochloric acid and then extracted with dichloromethane. The organic phase is dried over sodium sulfate and concentrated. The residue is column chromatographed on silica gel (mobile phase: cyclohexane / ethyl acetate 50/1 → 20/1). , This gives 540 mg (55% of theory) of crude product in 74% purity (LC-MS), which is reacted without further purification operation.
LC-MS (Methode 3): R, = 3.32 min; MS (ESIpos): m/z = 339 [M+H]+.LC-MS (Method 3): R, = 3.32 min; MS (ESIpos): m / z = 339 [M + H] + .
Beispiel 9AExample 9A
2-(4-Chloφhenyl)-4-(diethylamino)-6-methylpyrimidin-5-carbonsäureethylester2- (4-Chloφhenyl) -4- (diethylamino) -6-methyl-pyrimidin-5-carboxylate
Figure imgf000036_0002
Figure imgf000036_0002
Zu einer Lösung aus 100 mg (0.321 mmol) Beispiel 5 A in 2 ml Tetrahydrofuran wird 1.8 ml (13.498 mmol) Triethylamin und 35 mg (0.482 mmol) Diethylamin addiert. Das Reaktionsgemisch wird über Nacht bei 800C gerührt. Die Mischung wird einrotiert und ohne weitere Aufarbeitung weiter eingesetzt. Man erhält so 100 mg (90 % d. Th.) der ZielverbindungTo a solution of 100 mg (0.321 mmol) of Example 5 A in 2 ml of tetrahydrofuran is added 1.8 ml (13,498 mmol) of triethylamine and 35 mg (0.482 mmol) of diethylamine. The reaction mixture is stirred at 80 0 C overnight. The mixture is concentrated by rotary evaporation and used without further workup. This gives 100 mg (90% of theory) of the target compound
LC-MS (Methode 3): R, = 2.95 min; MS (ESIpos): m/z = 348 [M+H]+.LC-MS (Method 3): R, = 2.95 min; MS (ESIpos): m / z = 348 [M + H] + .
1H-NMR (400 MHz, DMSO-(I6): δ [ppm] = 1.17 (t, 6H), 1.32 (t, 3H), 2.34 (s, 3H), 3.50 (q, 4H), 4.33 (q, 2H), 7.56 (d, 2H), 8.32 (d, 2H), 8.59 (sbr, IH). 1 H-NMR (400 MHz, DMSO- (I 6 ): δ [ppm] = 1.17 (t, 6H), 1.32 (t, 3H), 2.34 (s, 3H), 3.50 (q, 4H), 4.33 ( q, 2H), 7.56 (d, 2H), 8.32 (d, 2H), 8.59 (sbr, IH).
Beispiel IQAExample IQA
2-(4-Chlθφhenyl)-4-(ethylamino)-6-( 1 -methylethyl)pyrimidin-5 carbonsäureethylesterEthyl 2- (4-chloro-phenyl) -4- (ethylamino) -6- (1-methylethyl) pyrimidine-5-carboxylate
Figure imgf000037_0001
Figure imgf000037_0001
Zu einer Lösung aus 60mg (ca. 0.131 mmol) Beispiel 8A in 0.8 ml THF werden 9 mg (0.196 mmol) Ethylamin und 0.8 ml (5.498 mmol) Triethylamin zugegeben. Das Reaktionsgemisch wird über Nacht bei 800C gerührt. Das Reaktionsgemisch wird einrotiert und der Rückstand ohne weitere Aufarbeitung umgesetzt. Man erhält so 55 mg (100 % Th.) der Zielverbindung in 90 %- iger Reinheit (LC-MS).To a solution of 60 mg (about 0.131 mmol) of Example 8A in 0.8 ml of THF are added 9 mg (0.196 mmol) of ethylamine and 0.8 ml (5.498 mmol) of triethylamine. The reaction mixture is stirred overnight at 80 0 C. The reaction mixture is concentrated by rotary evaporation and the residue is reacted without further work-up. This gives 55 mg (100% Th.) Of the target compound in 90% - purity (LC-MS).
LC-MS (Methode 3): R, = 3.51 min; MS (ESIpos): m/z = 348 [M+H]+.LC-MS (Method 3): R, = 3.51 min; MS (ESIpos): m / z = 348 [M + H] + .
Beispiel IIAExample IIA
2-(4-Chlθφhenyl)-4-(cyclopropylamino)-6-(l-methylethyl)pyrimidin-5-carbonsäureethylester2- (4-Chlθφhenyl) -4- (cyclopropylamino) -6- (l-methylethyl) pyrimidin-5-carboxylate
Figure imgf000037_0002
Zu einer Lösung aus 60 mg (ca. 0.131 mmol) Beispiel 8A in 0.8 ml THF werden 11 mg (0.196 mmol) Cyclopropanamin und 556 mg (5.498 mmol) Triethylamin zugegeben. Das Reaktionsgemisch wird über Nacht bei 800C gerührt. Die Mischung wird ohne weitere Aufarbeitung mittels präparativer HPLC aufgereinigt (Eluent: Acetonitril/Wasser, Gradient 10/90 → 90/10). Man erhält so 40 mg (85 % Th.) der Zielverbindung .
Figure imgf000037_0002
To a solution of 60 mg (about 0.131 mmol) of Example 8A in 0.8 ml of THF are added 11 mg (0.196 mmol) of cyclopropanamine and 556 mg (5.498 mmol) of triethylamine. The reaction mixture is stirred overnight at 80 0 C. The mixture is purified by preparative HPLC without elution (eluent: acetonitrile / water, gradient 10/90 → 90/10). This gives 40 mg (85% Th.) Of the target compound.
LC-MS (Methode 1): R, = 1.90 min; MS (ESIpos): m/z = 360 [M+H]+.LC-MS (Method 1): R, = 1.90 min; MS (ESIpos): m / z = 360 [M + H] + .
Beispiel 12AExample 12A
2-(4-Chloφhenyl)-4-(methylamino)-6-(l-methylethyl)pyrimidin-5-carbonsäureethylester2- (4-Chloφhenyl) -4- (methylamino) -6- (l-methylethyl) pyrimidin-5-carboxylate
Figure imgf000038_0001
Figure imgf000038_0001
Zu einer Lösung aus 60 mg (ca. 0.131 mmol) Beispiel 8A in 0.8 ml THF werden 0.09 ml (0.196 mmol) Methanamin-Lösung (2M in THF) und 556 mg (5.498 mmol) Triethylamin zugegeben. Das Reaktionsgemisch wird über Nacht bei 800C gerührt. Die Mischung wird ohne weitere Aufarbeitung mittels präparativer HPLC auf gereinigt (Eluent: Acetonitril/Wasser, Gradient 10:90 → 90:10). Man erhält so 60 mg (100 % Th.) der Zielverbindung.To a solution of 60 mg (about 0.131 mmol) of Example 8A in 0.8 ml of THF are added 0.09 ml (0.196 mmol) of methanamine solution (2M in THF) and 556 mg (5.498 mmol) of triethylamine. The reaction mixture is stirred overnight at 80 0 C. The mixture is purified by preparative HPLC without further workup (eluent: acetonitrile / water, gradient 10:90 → 90:10). This gives 60 mg (100% Th.) Of the target compound.
LC-MS (Methode 1): R, = 1.82 min; MS (ESIpos): m/z = 334 [M+H]+.LC-MS (Method 1): R, = 1.82 min; MS (ESIpos): m / z = 334 [M + H] + .
Beispiel 13AExample 13A
2-(4-Chloφhenyl)-4-ethyl-6-oxo-l,4,5,6-tetrahydropyrimidin-5-carbonsäureethylester2- (4-Chloφhenyl) -4-ethyl-6-oxo-l, 4,5,6-tetrahydropyrimidine-5-carboxylate
Figure imgf000038_0002
Figure imgf000038_0002
7.09 ml (18.98 mmol) Natriumethylat-Lösung (21 Gew-% in Ethanol) wird vorgelegt. Dann werden 2.10 g (10.43 mmol) 4-Chlorbenzamidin-Hydrochlorid als Lösung in 50 ml Ethanol und 1.90 g (9.49 mmol) Propylidenmalonsäuredieethylester [B. C. Ranu, R. Jana, Eur. J. Org. Chem. (2006) 3767-3770] unter Argonatmosphäre zugegeben. Das Reaktionsgemisch wird 2h bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird der Ansatz eingeengt, in Wasser aufgenommen, mit IN Salzsäure auf pH 4-5 eingestellt und anschließend mit Essigsäureethylester extrahiert (3x). Die vereinigten organischen Phasen werden mit Magnesiumsulfat getrocknet und das Lösemittel am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels einer präparativen MPLC aufgereinigt (Biotage 4OM Kartusche; Laufrnittel: Isohexan / Essigsäureethylester = 1 / 1). Man erhält so 1.37 g (35% d. Th.) Rohprodukt (82 %ige Reinheit (LC-MS)), welches ohne weitere Reinigungsoperation umgesetzt wird.7.09 ml (18.98 mmol) of sodium ethylate solution (21% by weight in ethanol) are initially charged. Then 2.10 g (10.43 mmol) of 4-chlorobenzamidine hydrochloride as a solution in 50 ml of ethanol and 1.90 g (9.49 mmol) of propylidenemalonic acid diester [BC Ranu, R. Jana, Eur. J. Org. Chem. (2006) 3767-3770] are added under an argon atmosphere. The reaction mixture is stirred for 2 h at reflux temperature. After cooling, the mixture is concentrated, taken up in water, adjusted to pH 4-5 with 1N hydrochloric acid and then extracted with ethyl acetate (3x). The combined organic phases are dried with magnesium sulfate and the solvent is removed on a rotary evaporator. The residue is purified by preparative MPLC (Biotage 4OM cartridge, eluent: isohexane / ethyl acetate = 1/1). This gives 1.37 g (35% of theory) of crude product (82% purity (LC-MS)), which is reacted without further purification operation.
LC-MS (Methode 3): R, = 1.92 min; MS (ESIpos): m/z = 309 [M+H]+.LC-MS (Method 3): R, = 1.92 min; MS (ESIpos): m / z = 309 [M + H] + .
Beispiel 14AExample 14A
2-(4-Chloφhenyl)-4-ethyl-6-oxo-l,6-dihydropyrimidin-5-carbonsäureethylester2- (4-Chloφhenyl) -4-ethyl-6-oxo-l, 6-dihydropyrimidine-5-carboxylate
Figure imgf000039_0001
Figure imgf000039_0001
1.38 g (4.47 mmol) Beispiel 13A wird in 40 ml Tetrachlormethan gelöst und unter Argonatmosphäre mit 0.835 g (4.69 mmol) N-Bromsuccinirnid, 0.054 mg (0.223 mmol)1.38 g (4.47 mmol) of Example 13A is dissolved in 40 ml of tetrachloromethane and under argon atmosphere with 0.835 g (4.69 mmol) of N-bromosuccinimide, 0.054 mg (0.223 mmol).
Dibenzoylperoxid und 6.18 g (44.69 mmol) Kaliumcarbonat versetzt. Anschließend wird beiDibenzoyl peroxide and 6.18 g (44.69 mmol) of potassium carbonate. Subsequently, at
Rückflußtemperatur 30 min gerührt. Nach dem Abkühlen wird der Ansatz mit 100 ml Wasser versetzt und mit Dichlormethan (3x) extrahiert. Die vereinigten organischen Phasen werden mitReflux temperature for 30 min stirred. After cooling, the batch is mixed with 100 ml of water and extracted with dichloromethane (3x). The combined organic phases are with
Magnesiumsulfat getrocknet. Dann werden die flüchtigen Komponenten durch Destillation bei vermindertem Druck abgetrennt. Man erhält so 1.25 g (73% d. Th.) Rohprodukt in 80-%igerDried magnesium sulfate. Then, the volatile components are separated by distillation under reduced pressure. This gives 1.25 g (73% of theory) of crude product in 80% strength
Reinheit (LC-MS), welches ohne weitere Reinigungsoperation umgesetzt wird.Purity (LC-MS), which is reacted without further purification operation.
LC-MS (Methode 1): R, = 1.17 min; MS (ESIpos): m/z = 307 [M+H]+.LC-MS (Method 1): R, = 1.17 min; MS (ESIpos): m / z = 307 [M + H] + .
1H-NMR (400 MHz, DMSO-Cl6): δ [ppm] = 1.34 (t, 3H), 1.42 (t, 3H), 2.83 (mz, 2H), 4.45 (q, 2H), 7.48 (d, 2H), 8.27 (d, 2H), 12.78 (sbr, IH). 1 H-NMR (400 MHz, DMSO-Cl 6 ): δ [ppm] = 1.34 (t, 3H), 1.42 (t, 3H), 2.83 (mz, 2H), 4.45 (q, 2H), 7.48 (i.e. , 2H), 8.27 (d, 2H), 12.78 (sbr, IH).
Beispiel 15AExample 15A
4-Chlor-2-(4-chloφhenyl)-6-ethylpyrimidin-5-carbonsäureethylester
Figure imgf000040_0001
4-chloro-2- (4-chloφhenyl) -6-ethylpyrimidine-5-carboxylate
Figure imgf000040_0001
1.25 g (3.26 mmol) Beispiel 14A und 12.2 ml Phosphoxylchlorid werden 2h bei1.25 g (3.26 mmol) Example 14A and 12.2 ml of phosphoxyl chloride are added for 2 h
Rückflußtemperatur gerührt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt und der Rückstand vorsichtig und unter Eiskühlung in Wasser aufgenommen. Anschließend wird mit 25%-iger wässriger Ammoniak-Lösung versetzt, dann mit IN Salzsäure neutralisiert und mit Dichlormethan extrahiert (3x). Die vereinigten organischen Phasen werden mit Magnesiumsulfat getrocknet. Dann wird der Ansatz am Rotationsverdampfer eingeengt. DerReflux temperature stirred. Then, the volatile components are separated on a rotary evaporator and the residue was added carefully and under ice cooling in water. Then it is mixed with 25% aqueous ammonia solution, then neutralized with 1N hydrochloric acid and extracted with dichloromethane (3x). The combined organic phases are dried with magnesium sulfate. Then the batch is concentrated on a rotary evaporator. Of the
Rückstand wird mittels einer präparativen MPLC aufgereinigt (Biotage 4OM Kartusche;Residue is purified by preparative MPLC (Biotage 4OM cartridge;
Laufmittel: Isohexan / Essigsäureethylester = 7 / 3). Man erhält so 560 mg (52% d. Th.) der Zielverbindung.Eluent: isohexane / ethyl acetate = 7/3). This gives 560 mg (52% of theory) of the target compound.
LC-MS (Methode 3): R, = 3.32 min; MS (ESIpos): m/z = 325 [M]+.LC-MS (Method 3): R, = 3.32 min; MS (ESIpos): m / z = 325 [M] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.31 (t, 3H), 1.35 (t, 3H), 2.84 (q, 2H), 4.45 (q, 2H), 7.64 (d, 2H), 8.37 (d, 2H). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 1.31 (t, 3H), 1.35 (t, 3H), 2.84 (q, 2H), 4.45 (q, 2H), 7.64 (i.e. , 2H), 8.37 (d, 2H).
Beispiel 16AExample 16A
2-(4-Chlθφhenyl)-4-ethyl-6-[(l-methylethyl)amino]pyrimidin-5-carbonsäureethylester2- (4-Chlθφhenyl) -4-ethyl-6 - [(l-methylethyl) amino] pyrimidin-5-carboxylate
Figure imgf000040_0002
Figure imgf000040_0002
100 mg (0.31 mmol) Beispiel 15A, 39 μl (0.46 mmol) Isopropylamin und 107 μl (0.77 mmol) Triethylamin werden in 3 ml Tetrahydrofuran gelöst und dann über Nacht bei 800C umgesetzt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels präparativer HPLC aufgereinigt (Eluent: Acetonitril/Wasser, Gradient 10:90 → 90:10). Man erhält so 42 mg (39% d. Th.) der Zielverbindung. LC-MS (Methode 1): R, = 1.83 min; MS (ESIpos): m/z = 348 [M+H]+.100 mg (0.31mmol) Example 15A, 39 ul (0:46 mmol) of isopropylamine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted overnight at 80 0 C. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 → 90:10). This gives 42 mg (39% of theory) of the target compound. LC-MS (Method 1): R, = 1.83 min; MS (ESIpos): m / z = 348 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.24 (t, 3H), 1.26 (d, 6H), 1.34 (t, 3H), 2.89 (q, 2H), 4.34 (q, 2H), 4.45 (mz, IH), 7.58 (d, 2H), 7.75 (d, IH), 8.38 (d, 2H). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.24 (t, 3H), 1.26 (d, 6H), 1.34 (t, 3H), 2.89 (q, 2H), 4.34 (q, 2H ), 4.45 (mz, IH), 7.58 (d, 2H), 7.75 (d, IH), 8.38 (d, 2H).
Beispiel 17AExample 17A
2-(4-Chlθφhenyl)-4-ethyl-6-(prop-2-en-l-ylamino)pyrimidin-5-carbonsäureethylester2- (4-Chlθφhenyl) -4-ethyl-6- (prop-2-en-l-ylamino) pyrimidin-5-carboxylate
Figure imgf000041_0001
Figure imgf000041_0001
100 mg (0.31 mmol) Beispiel 15A, 35 μl (0.46 mmol) Allylamin und 107 μl (0.77 mmol) Triethylamin werden in 3 ml Tetrahydrofuran gelöst und dann über Nacht bei 800C umgesetzt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels präparativer HPLC aufgereinigt (Eluent: Acetonitril/Wasser, Gradient 10:90 — > 90:10). Man erhält so 70 mg (66% d. Th.) der Zielverbindung.100 mg (0.31mmol) Example 15A, 35 ul (0:46 mmol) of allylamine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted overnight at 80 0 C. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 70 mg (66% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.77 min; MS (ESIpos): m/z = 346 [M+H]+.LC-MS (Method 1): R, = 1.77 min; MS (ESIpos): m / z = 346 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.25 (t, 3H), 1.34 (t, 3H), 2.87 (q, 2H), 4.20 (mz, 2H), 4.35 (q, 2H), 5.12 (dd, IH), 5.23 (dd, IH), 5.99 (mz, IH), 7.57 (d, 2H), 8.02 (t, IH), 8.37 (d, 2H). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.25 (t, 3H), 1.34 (t, 3H), 2.87 (q, 2H), 4.20 (mz, 2H), 4.35 (q, 2H ), 5.12 (dd, IH), 5.23 (dd, IH), 5.99 (mz, IH), 7.57 (d, 2H), 8.02 (t, IH), 8.37 (d, 2H).
Beispiel 18AExample 18A
2-(4-Chlorphenyl)-4-(diethylamino)-6-ethylpyrimidin-5-carbonsäureethylester2- (4-chlorophenyl) -4- (diethylamino) -6-ethylpyrimidine-5-carboxylate
Figure imgf000041_0002
100 mg (0.31 mmol) Beispiel 15A, 48 μl (0.46 mmol) Diethylamin und 107 μl (0.77 mmol) Triethylamin werden in 3 ml Tetrahydrofuran gelöst und dann über Nacht bei 800C umgesetzt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels präparativer HPLC aufgereinigt (Eluent: Acetonitril/Wasser, Gradient 10:90 — > 90:10). Man erhält so 90 mg (81% d. Th.) der Zielverbindung.
Figure imgf000041_0002
100 mg (0.31mmol) Example 15A, 48 ul (0:46 mmol) of diethylamine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted overnight at 80 0 C. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 90 mg (81% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.71 min; MS (ESIpos): m/z = 362 [M+H]+.LC-MS (method 1): R, = 1.71 min; MS (ESIpos): m / z = 362 [M + H] + .
1H-NMR (400 MHz, DMSO-d«): δ [ppm] = 1.17 (t, 6H), 1.22 (t, 3H), 1.32 (t, 3H), 2.59 (q, 2H), 3.51 (q, 4H), 4.33 (q, 2H), 7.57 (d, 2H), 8.33 (d, 2H). 1 H NMR (400 MHz, DMSO-d "): δ [ppm] = 1.17 (t, 6H), 1.22 (t, 3H), 1.32 (t, 3H), 2.59 (q, 2H), 3.51 (q , 4H), 4.33 (q, 2H), 7.57 (d, 2H), 8.33 (d, 2H).
Beispiel 19AExample 19A
2-(4-Chloφhenyl)-4-(cyclohexylamino)-6-ethylpyrimidin-5 -carbonsäureethylesterEthyl 2- (4-chlorophenyl) -4- (cyclohexylamino) -6-ethylpyrimidine-5-carboxylate
Figure imgf000042_0001
Figure imgf000042_0001
100 mg (0.31 mmol) Beispiel 15A, 53 μl (0.46 mmol) Cyclohexylamin und 107 μl (0.77 mmol) Triethylamin werden in 3 ml Tetrahydrofuran gelöst und dann über Nacht bei 800C umgesetzt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels präparativer HPLC auf gereinigt (Eluent: Acetonitril/Wasser, Gradient 10:90 -» 90:10). Man erhält so 43 mg (36% d. Th.) der Zielverbindung.100 mg (0.31mmol) Example 15A, 53 ul (0:46 mmol) of cyclohexylamine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted overnight at 80 0 C. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 43 mg (36% of theory) of the target compound.
LC-MS (Methode 1): R, = 2.01 min; MS (ESIpos): m/z = 388 [M+H]+.LC-MS (Method 1): R, = 2.01 min; MS (ESIpos): m / z = 388 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.18-1.51 (m, HH), 1.61 (m, IH), 1.72 (m, 2H), 1.96 (mz, 2H), 2.91 (q, 2H), 4.17 (mz, IH), 4.34 (q, 2H), 7.58 (d, 2H), 7.89 (d, IH), 8.36 (d, 2H). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.18-1.51 (m, HH), 1.61 (m, IH), 1.72 (m, 2H), 1.96 (mz, 2H), 2.91 (q , 2H), 4.17 (mz, IH), 4.34 (q, 2H), 7.58 (d, 2H), 7.89 (d, IH), 8.36 (d, 2H).
Beispiel 2OAExample 2OA
2-(4-Chloφhenyl)-4-ethyl-6-piperidin-l-ylpyrimidin-5-carbonsäureethylester
Figure imgf000043_0001
2- (4-Chloφhenyl) -4-ethyl-6-piperidin-l-yl-pyrimidine-5-carboxylate
Figure imgf000043_0001
100 mg (0.31 mmol) Beispiel 15 A, 46 μl (0.46 mmol) Piperidin und 107 μl (0.77 mmol) Triethylamin werden in 3 ml Tetrahydrofuran gelöst und dann über Nacht bei 80 0C umgesetzt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels präparativer HPLC aufgereinigt (Eluent: Acetonitril/Wasser, Gradient 10:90 — > 90:10). Man erhält so 123 mg (98% d. Th.) der Zielverbindung.100 mg (0.31 mmol) of Example 15 A, 46 .mu.l (0.46 mmol) of piperidine and 107 .mu.l (0.77 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran and then reacted at 80 0 C overnight. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 123 mg (98% of theory) of the target compound.
LC-MS (Methode 3): Rt = 3.36 min; MS (ESIpos): m/z = 374 [M+H]+.LC-MS (Method 3): R t = 3.36 min; MS (ESIpos): m / z = 374 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.22 (t, 3H), 1.31 (t, 3H), 1.58 (m, 4H), 1.64 (m, 2H), 2.66 (q, 2H), 3.58 (mz, 4H), 4.32 (q, 2H), 7.56 (d, 2H), 8.34 (d, 2H). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.22 (t, 3H), 1.31 (t, 3H), 1.58 (m, 4H), 1.64 (m, 2H), 2.66 (q, 2H ), 3.58 (mz, 4H), 4.32 (q, 2H), 7.56 (d, 2H), 8.34 (d, 2H).
Beispiel 21AExample 21A
4-Ethyl-2-(4-methylphenyl)-6-oxo-l,4,5,6-tetrahydropyrimidin-5-carbonsäureethylester4-ethyl-2- (4-methylphenyl) -6-oxo-l, 4,5,6-tetrahydropyrimidine-5-carboxylate
Figure imgf000043_0002
Figure imgf000043_0002
7.46 ml (19.98 mmol) Natriumethylat-Lösung (21 Gew-% in Ethanol) wird vorgelegt. Dann werden 1.47 g (10.99 mmol) 4-Methylbenzamidin in 50 ml Ethanol und 2.00 g (9.99 mmol) Propylidenmalonsäuredieethylester [B. C. Ranu, R. Jana, Eur. J. Org. Chem. (2006) 3767-3770] unter Argonatmosphäre zugegeben. Das Reaktionsgemisch wird 2h bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird der Ansatz eingeengt, in Wasser aufgenommen, mit IN Salzsäure auf pH 4-5 eingestellt und anschließend mit Essigsäureethylester extrahiert (3x). Die vereinigten organischen Phasen werden mit Magnesiumsulfat getrocknet und das Lösemittel am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels einer präparativen MPLC aufgereinigt (Biotage 4OM Kartusche; Laufinittel: Isohexan / Essigsäureethylester = 1 / 1). Man erhält so 1.32 g (37% d. Th.) Rohprodukt (88 %-ige Reinheit (LC-MS)), welches ohne weitere Reinigungsoperationen umgesetzt wird.7.46 ml (19.98 mmol) of sodium ethylate solution (21% by weight in ethanol) is initially charged. Then 1.47 g (10.99 mmol) of 4-methylbenzamidine in 50 ml of ethanol and 2.00 g (9.99 mmol) of propylidenemalonic acid diester [BC Ranu, R. Jana, Eur. J. Org. Chem. (2006) 3767-3770] are added under an argon atmosphere. The reaction mixture is stirred for 2 h at reflux temperature. After cooling, the mixture is concentrated, taken up in water, adjusted to pH 4-5 with 1N hydrochloric acid and then extracted with ethyl acetate (3x). The combined organic phases are dried with magnesium sulfate and the solvent is removed on a rotary evaporator. The residue is purified by preparative MPLC Purified (Biotage 4OM cartridge, eluent: isohexane / ethyl acetate = 1/1). This gives 1.32 g (37% of theory) of crude product (88% purity (LC-MS)), which is reacted without further purification operations.
LC-MS (Methode 3): R, = 1.44 min; MS (ESIpos): m/z = 289 [M+H]+.LC-MS (Method 3): R, = 1.44 min; MS (ESIpos): m / z = 289 [M + H] + .
Beispiel 22AExample 22A
4-Ethyl-2-(4-methylphenyl)-6-oxo-l,6-dihydropyrimidin-5-carbonsäureethylester4-ethyl-2- (4-methylphenyl) -6-oxo-l, 6-dihydropyrimidine-5-carboxylate
Figure imgf000044_0001
Figure imgf000044_0001
1.32 g (4.58 mmol) Beispiel 21A werden in 40 ml Tetrachlormethan gelöst und unter Argon mit 0.856 g (4.81 mmol) N-Bromsuccinimid, 0.055 mg (0.229 mmol) Dibenzoylperoxid und 6.32 g (45.78 mmol) Kaliumcarbonat versetzt. Anschließend wird 30 min bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird der Ansatz mit 100 ml Wasser versetzt und mit Dichlormethan (3x) extrahiert. Die vereinigten organischen Phasen werden mit Magnesiumsulfat getrocknet. Dann werden die flüchtigen Komponenten durch Destillation bei vermindertem Druck abgetrennt. Man erhält so 1.31 g Rohprodukt, welches aus 2-[4-(Brommethyl)phenyl]-4-ethyl-6-oxo-l,6- dihydropyrimidin-5-carbonsäureethylester und der Zielsubstanz im Verhältnis 1.5:1 besteht. Das so gewonnene Rohmaterial wird in 40 ml Ethanol aufgenommen und unter Argon mit 26 mg (0.247 mmol) Palladium auf Kohle (10 w/w), als auch mit 779 mg (12.35 mmol) Ammoniumformiat versetzt. Anschließend wird 30 min bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird über Celite filtriert. Das so erhaltene Rohmaterial wird dann abschließend mittels einer präparativen MPLC aufgereinigt (Biotage 4OM Kartusche; Laufmittel: Isohexan / Essigsäureethylester = 1 / 1). Man erhält so 450 mg (35% d. Th.) der Zielverbindung.1.32 g (4.58 mmol) of Example 21A are dissolved in 40 ml of tetrachloromethane and treated under argon with 0.856 g (4.81 mmol) of N-bromosuccinimide, 0.055 mg (0.229 mmol) of dibenzoyl peroxide and 6.32 g (45.78 mmol) of potassium carbonate. The mixture is then stirred at reflux temperature for 30 minutes. After cooling, the batch is mixed with 100 ml of water and extracted with dichloromethane (3x). The combined organic phases are dried with magnesium sulfate. Then, the volatile components are separated by distillation under reduced pressure. This gives 1.31 g of crude product which consists of 2- [4- (bromomethyl) phenyl] -4-ethyl-6-oxo-l, 6-dihydropyrimidine-5-carboxylic acid ethyl ester and the target substance in a ratio of 1.5: 1. The crude material thus obtained is taken up in 40 ml of ethanol and admixed under argon with 26 mg (0.247 mmol) of palladium on carbon (10 w / w) and with 779 mg (12.35 mmol) of ammonium formate. The mixture is then stirred at reflux temperature for 30 minutes. After cooling, it is filtered through Celite. The crude material thus obtained is then finally purified by means of a preparative MPLC (Biotage 4OM cartridge, mobile phase: isohexane / ethyl acetate = 1/1). This gives 450 mg (35% of theory) of the target compound.
LC-MS (Methode 3): R, = 2.18 min; MS (ESIpos): m/z = 287 [M+H]+.LC-MS (Method 3): R, = 2.18 min; MS (ESIpos): m / z = 287 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.21 (t, 3H), 1.28 (t, 3H), 2.39 (s, 3H), 4.28 (q, 2H), 7.35 (d, 2H), 8.04 (d, 2H), 12.97 (sbr, IH). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.21 (t, 3H), 1.28 (t, 3H), 2.39 (s, 3H), 4.28 (q, 2H), 7.35 (d, 2H ), 8.04 (d, 2H), 12.97 (sbr, IH).
Beispiel 23AExample 23A
4-Chlor-6-ethyl-2-(4-methylphenyl)pyrimidin-5-carbonsäureethylester
Figure imgf000045_0001
4-chloro-6-ethyl-2- (4-methylphenyl) pyrimidin-5-carboxylate
Figure imgf000045_0001
450 mg (1.60 mmol) Beispiel 22A und 5.96 ml Phosphoxylchlorid werden 2 h bei450 mg (1.60 mmol) of Example 22A and 5.96 ml of phosphoxyl chloride are added for 2 h
Rückflußtemperatur gerührt. Der Ansatz wird am Rotationsverdampfer eingeengt und derReflux temperature stirred. The mixture is concentrated on a rotary evaporator and the
Rückstand vorsichtig und unter Eiskühlung in Wasser aufgenommen. Anschließend wird mit 25%- iger wässriger Ammoniak-Lösung versetzt, dann mit IN Salzsäure neutralisiert und mitCarefully collect the residue in water and cool with ice. It is then treated with 25% - aqueous ammonia solution, then neutralized with IN hydrochloric acid and with
Dichlormethan extrahiert (3x). Die vereinigten organischen Phasen werden mit Magnesiumsulfat getrocknet. Dann werden die flüchtigen Komponenten durch Destillation bei vermindertem Druck abgetrennt. Der Rückstand wird mittels einer präparativen MPLC aufgereinigt (Biotage 4OMDichloromethane extracted (3x). The combined organic phases are dried with magnesium sulfate. Then, the volatile components are separated by distillation under reduced pressure. The residue is purified by preparative MPLC (Biotage 4OM
Kartusche; Laufmittel: Isohexan / Essigsäureethylester = 7 / 3). Man erhält so 421 mg (85% d. Th.) der Zielverbindung.Cartridge; Eluent: isohexane / ethyl acetate = 7/3). This gives 421 mg (85% of theory) of the target compound.
LC-MS (Methode 3): R, = 3.22 min; MS (ESIpos): m/z = 305 [M]+.LC-MS (Method 3): R, = 3.22 min; MS (ESIpos): m / z = 305 [M] + .
1H-NMR (400 MHz, DMSO-(I6): δ [ppm] = 1.30 (t, 3H), 1.35 (t, 3H), 2.40 (s, 3H), 2.82 (q, 2H), 4.44 (q, 2H), 7.38 (d, 2H), 8.27 (d, 2H). 1 H-NMR (400 MHz, DMSO- (I 6 ): δ [ppm] = 1.30 (t, 3H), 1.35 (t, 3H), 2.40 (s, 3H), 2.82 (q, 2H), 4.44 ( q, 2H), 7.38 (d, 2H), 8.27 (d, 2H).
Beispiel 24AExample 24A
4-(Diethylamino)-6-ethyl-2-(4-methylphenyl)pyrimidin-5-carbonsäureethylester4- (diethylamino) -6-ethyl-2- (4-methylphenyl) pyrimidin-5-carboxylate
Figure imgf000045_0002
Figure imgf000045_0002
100 mg (0.328 mmol) Beispiel 23A, 51 μl (0.492 mmol) Diethylamin und 114 μl (0.820 mmol) Triethylamin werden in 3 ml Tetrahydrofuran gelöst und dann 3h bei 800C umgesetzt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels präparativer HPLC aufgereinigt (Eluent: Acetonitril/Wasser, Gradient 10:90 — > 90:10). Man erhält so 78 mg (66% d. Th.) der Zielverbindung.100 mg (0.328 mmol) of Example 23A, 51 ul (0.492 mmol) of diethylamine and 114 .mu.l (0.820 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted at 80 0 C for 3 h. Then the volatile components are separated on a rotary evaporator. The residue will be purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 -> 90:10). This gives 78 mg (66% of theory) of the target compound.
LC-MS (Methode 2): R, = 2.32 min; MS (ESIpos): m/z = 342 [M+H]+.LC-MS (Method 2): R, = 2.32 min; MS (ESIpos): m / z = 342 [M + H] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.17 (t, 6H), 1.22 (t, 3H), 1.32 (t, 3H), 2.37 (s, 3H), 2.58 (q, 2H), 3.50 (q, 4H), 4.32 (q, 2H), 7.30 (d, 2H), 8.23 (d, 2H). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 1.17 (t, 6H), 1.22 (t, 3H), 1.32 (t, 3H), 2.37 (s, 3H), 2.58 (q , 2H), 3.50 (q, 4H), 4.32 (q, 2H), 7.30 (d, 2H), 8.23 (d, 2H).
Beispiel 25AExample 25A
4-Ethyl-6-[(l-methylethyl)amino]-2-(4-methylphenyl)pyrimidin-5-carbonsäureethylester4-ethyl-6 - [(l-methylethyl) amino] -2- (4-methylphenyl) pyrimidin-5-carboxylate
Figure imgf000046_0001
Figure imgf000046_0001
100 mg (0.328 mmol) Beispiel 23A, 42 μl (0.492 mmol) Diethylamin und 114 μl (0.820 mmol) Triethylamin werden in 3 ml Tetrahydrofuran gelöst und dann 2h bei 800C umgesetzt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt. Der Rückstand wird mittels präparativer HPLC aufgereinigt (Eluent: Acetonitril/Wasser, Gradient 10:90 → 90:10). Man erhält so 68 mg (62% d. Th.) der Zielverbindung.100 mg (0.328 mmol) of Example 23A, 42 ul (0.492 mmol) of diethylamine and 114 .mu.l (0.820 mmol) of triethylamine are dissolved in 3 ml of tetrahydrofuran, and then reacted at 80 0 C for 2 h. Then the volatile components are separated on a rotary evaporator. The residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 10:90 → 90:10). This gives 68 mg (62% of theory) of the target compound.
LC-MS (Methode 2): R, = 2.66 min; MS (ESIpos): m/z = 328 [M+H]+.LC-MS (Method 2): R, = 2.66 min; MS (ESIpos): m / z = 328 [M + H] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.24 (t, 3H), 1.26 (d, 6H), 1.34 (t, 3H), 2.38 (s, 3H), 2.90 (q, 2H), 4.34 (q, 2H), 4.45 (mz, IH), 7.31 (d, 2H), 7.75 (d, IH), 8.28 (d, 2H). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 1.24 (t, 3H), 1.26 (d, 6H), 1.34 (t, 3H), 2.38 (s, 3H), 2.90 (q , 2H), 4.34 (q, 2H), 4.45 (mz, IH), 7.31 (d, 2H), 7.75 (d, IH), 8.28 (d, 2H).
Beispiel 26AExample 26A
2-(4-Chloφhenyl)-4-(2-methylpropyl)-6-oxo-l,4,5,6-tetrahydropyrimidin-5- carbonsäuremethylester
Figure imgf000047_0001
2- (4-chloro-phenyl) -4- (2-methyl-propyl) -6-oxo-l, 4,5,6-tetrahydropyrimidine-5-carboxylic acid methyl ester
Figure imgf000047_0001
3.0 g (22.71 mmol) Malonsäuredimethylester , 2.15 g (24.98 mmol) 3-Methylbutanal, 0.22 ml (2.27 mmol) Piperidin und 0.13 ml (2.27 mmol) Essigsäure werden in 40 ml Dichlormethan gelöst und über Nacht am inversen Wasserabscheider bei Rückflußtemperatur umgesetzt. Die flüchtigen Komponenten werden am Rotationsverdampfer abgetrennt. Man erhält so (3-Methylbutyliden)- propansäuredimethylester, welcher ohne weitere Reingungsoperationen umgesetzt wird. Zu einer Mischung von 3.52 g (51.78 mmol) Natriumethylat und 5.44 g (24.48 mmol) 4-Chlorbenzamidin- Hydrochlorid in 50 ml Methanol werden unter Argonatmosphäre 5.18g (-25.89 mmol) des so gewonnenen Rohmaterials zugegeben. Das Reaktionsgemisch wird über Nacht bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird der Ansatz eingeengt und mit Wasser versetzt. Die wässrige Phase wird mit Essigsäureethylester und Dichlormethan extrahiert. Anschließend wird die organische Phase mit Wasser und mit gesättigter wässriger Natriumchlorid- Lösung gewaschen, über Natriumsulfat getrocknet und eingeengt. Man erhält so 5.02 g (40% d. Th.) Rohprodukt in 66%-iger Reinheit (LC-MS), welches ohne weitere Reinigungsschritte umgesetzt wird.3.0 g (22.71 mmol) of dimethyl malonate, 2.15 g (24.98 mmol) of 3-methylbutanal, 0.22 ml (2.27 mmol) of piperidine and 0.13 ml (2.27 mmol) of acetic acid are dissolved in 40 ml of dichloromethane and reacted overnight at the inverse water at reflux temperature. The volatile components are separated on a rotary evaporator. This gives (3-methylbutylidene) - propanoic acid dimethyl ester, which is reacted without further purification operations. To a mixture of 3.52 g (51.78 mmol) of sodium ethylate and 5.44 g (24.48 mmol) of 4-chlorobenzamidine hydrochloride in 50 ml of methanol are added 5.18 g (-25.89 mmol) of the raw material thus obtained under an argon atmosphere. The reaction mixture is stirred overnight at reflux temperature. After cooling, the mixture is concentrated and treated with water. The aqueous phase is extracted with ethyl acetate and dichloromethane. The organic phase is then washed with water and with saturated aqueous sodium chloride solution, dried over sodium sulfate and concentrated. This gives 5.02 g (40% of theory) of crude product in 66% purity (LC-MS), which is reacted without further purification steps.
LC-MS (Methode 2): R, = 1.83 min; MS (EIpos): m/z = 323 [M+H]+.LC-MS (Method 2): R, = 1.83 min; MS (EIpos): m / z = 323 [M + H] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.93 (dd, 6H), 1.15-1.23 (dd, IH), 1.46-1.53 (dd, IH), 2.06 (sbr, IH), 3.44 (d, IH), 3.69 (s, 3H), 3.93-4.00 (dd, IH), 7.52 (d, IH), 7.86 (d, IH), 11.15 (sbr, IH). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 0.93 (dd, 6H), 1.15-1.23 (dd, IH), 1.46-1.53 (dd, IH), 2.06 (sbr, IH) , 3.44 (d, IH), 3.69 (s, 3H), 3.93-4.00 (dd, IH), 7.52 (d, IH), 7.86 (d, IH), 11.15 (sbr, IH).
Beispiel 27AExample 27A
2-(4-Chloφhenyl)-4-(2-methylpropyl)-6-oxo-l,6-dihydropyrimidin-5-carbonsäuremethylester2- (4-Chloφhenyl) -4- (2-methylpropyl) -6-oxo-l, 6-dihydropyrimidine-5-carbonsäuremethylester
Figure imgf000047_0002
Eine Mischung aus 5.02 g (-10.27 mmol) Beispiel 26A, 1.83 g (10.27 mmol) N-Bromsuccinimid, 497 mg (2.05 mmol) Dibenzoylperoxid und 2.13 g (15.40 mmol) gemahlenes Kaliumcarbonat in 120 ml Dioxan wird unter Argonatmosphäre 3 h bei Rückflußtemperatur gerührt. Nach dem Abkühlen wird der Ansatz mit einer gesättigten wässrigen Natriumthiosulfat-Lösung versetzt und anschließend eingeengt. Die wässrige Phase wird mit Dichlormethan extrahiert und die organische Phase wird erst mit Wasser und dann mit einer gesättigten wässrigen Natriumchlorid-Lösung gewaschen. Es wird mit Natriumsulfat getrocknet und eingeengt. Das Rohmaterial wird mit Acetonitril versetzt und die entstandenen Kristalle werden abgesaugt. Dann wird im Hochvakuum bis zur Gewichtskonstanz getrocknet. Man erhält 1.47 g (40% d. Th.) der Zielverbindung in 88%- iger Reinheit (LC-MS), welche ohne weitere Reinigungsschritte umgesetzt wird.
Figure imgf000047_0002
A mixture of 5.02 g (-10.27 mmol) of Example 26A, 1.83 g (10.27 mmol) of N-bromosuccinimide, 497 mg (2.05 mmol) of dibenzoyl peroxide and 2.13 g (15.40 mmol) of ground potassium carbonate in 120 ml of dioxane is refluxed under argon atmosphere for 3 hours touched. After cooling, the mixture is treated with a saturated aqueous sodium thiosulfate solution and then concentrated. The aqueous phase is extracted with dichloromethane and the organic phase is washed first with water and then with a saturated aqueous sodium chloride solution. It is dried with sodium sulfate and concentrated. The crude material is mixed with acetonitrile and the resulting crystals are filtered off with suction. Then it is dried under high vacuum to constant weight. This gives 1.47 g (40% of theory) of the target compound in 88% purity (LC-MS), which is reacted without further purification steps.
LC-MS (Methode 2): Rt = 2.02 min; MS (EIpos): m/z = 321 [M+H]+.LC-MS (method 2): R t = 2.02 min; MS (EIpos): m / z = 321 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 0.92 (d, 6H), 2.11-2.21 (m, IH), 2.41 (d, 2H), 3.80 (s, 3H), 7.61 (d, 2H), 8.14 (d, 2H), 13.13 (sbr, IH). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 0.92 (d, 6H), 2.11-2.21 (m, IH), 2.41 (d, 2H), 3.80 (s, 3H), 7.61 (i.e. , 2H), 8.14 (d, 2H), 13.13 (sbr, IH).
Beispiel 28AExample 28A
4-Chlor-2-(4-chloφhenyl)-6-(2-methylpropyl)pyrimidine-5- carbonsäuremethylester4-Chloro-2- (4-chloro-phenyl) -6- (2-methyl-propyl) -pyrimidines-5-carboxylic acid methyl ester
Figure imgf000048_0001
Figure imgf000048_0001
400 mg (-1.10 mmol) Beispiel 27A und 4.09 ml (43.89 mmol) Phosphoxylchlorid werden Ih bei Rückflußtemperatur gerührt. Das Reaktionsgemisch wird einrotiert. Der Rückstand wird mit einer Ammoniumhydroxid-Lösung (25%ig in Wasser) versetzt, mit 1 N Salzsäure auf pH 7 gestellt und anschließend mit Dichlormethan extrahiert. Die organische Phase wird über Natriumsulfat getrocknet und eingeengt. Man erhält so 416 mg (98% d. Th.) Rohprodukt in 88%-iger Reinheit (LC-MS), welches ohne weitere Reinigungsoperation umgesetzt wird.400 mg (-1.10 mmol) of Example 27A and 4.09 ml (43.89 mmol) of phosphoxyl chloride are stirred at reflux temperature for 1 h. The reaction mixture is evaporated. The residue is mixed with an ammonium hydroxide solution (25% in water), adjusted to pH 7 with 1 N hydrochloric acid and then extracted with dichloromethane. The organic phase is dried over sodium sulfate and concentrated. This gives 416 mg (98% of theory) of crude product in 88% purity (LC-MS), which is reacted without further purification operation.
LC-MS (Methode 1): R, = 1.76 min; MS (EIpos): m/z = 339 [M+H]+.LC-MS (Method 1): R, = 1.76 min; MS (EIpos): m / z = 339 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 0.93 (d, 6H), 2.20-2.29 (m, IH), 2.68 (d, 2H), 3.97 (s, 3H), 7.64 (d, 2H), 8.36 (d, 2H). Beispiel 29A 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 0.93 (d, 6H), 2.20-2.29 (m, IH), 2.68 (d, 2H), 3.97 (s, 3H), 7.64 (i.e. , 2H), 8.36 (d, 2H). Example 29A
2-(4-Chlorphenyl)-4-(diethylamin)-6-(2-methylpropyl)pyrimidin-5-carbonsäuremethylester2- (4-chlorophenyl) -4- (diethylamino) -6- (2-methylpropyl) pyrimidine-5-carbonsäuremethylester
Figure imgf000049_0001
Figure imgf000049_0001
Zu einer Lösung aus 60 mg (0.177 mmol) Beispiel 28A in 1.2 ml THF werden 19 mg (0.265 mmol) Diethylamin und 45 mg (0.442 mmol) Triethylamin zugegeben. Die Mischung wird über Nacht bei 800C gerührt. Das Reaktionsgemisch wird einrotiert und ohne weitere Aufarbeitung weiter eingesetzt. Man erhält so 36 mg (54 % d. Th.) der Zielverbindung.19 mg (0.265 mmol) of diethylamine and 45 mg (0.442 mmol) of triethylamine are added to a solution of 60 mg (0.177 mmol) of Example 28A in 1.2 ml of THF. The mixture is stirred overnight at 80 0 C. The reaction mixture is concentrated by rotary evaporation and used further without further work-up. This gives 36 mg (54% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.79 min; MS (EIpos): m/z = 376 [M+H]+.LC-MS (Method 1): R, = 1.79 min; MS (EIpos): m / z = 376 [M + H] +.
IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.89 (d, 6H), 1.17 (t, 6H), 2.15-2.23 (m, IH), 2.44 (d, 2H), 3.49 (q, 4H), 3.85 (s, 3H), 7.56 (d, 2H), 8.32 (d, 2H).IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.89 (d, 6H), 1.17 (t, 6H), 2.15-2.23 (m, IH), 2.44 (d, 2H), 3.49 (q , 4H), 3.85 (s, 3H), 7.56 (d, 2H), 8.32 (d, 2H).
Beispiel 3OAExample 3OA
2-(4-Chlorphenyl)-4-(2-methylpropyl)-6-[(2-methylpropyl)amino]pyrimidin-5- carbonsäuremethylester2- (4-Chloro-phenyl) -4- (2-methyl-propyl) -6 - [(2-methyl-propyl) -amino] -pyrimidine-5-carboxylic acid, methyl ester
Figure imgf000049_0002
Figure imgf000049_0002
Zu einer Lösung aus 60 mg (0.177 mmol) Beispiel 28A in 1.2 ml THF werden 19 mg (0.265 mmol) Isobutylamin und 45 mg (0.442 mmol) Triethylamin zugegeben. Die Mischung wird über Nacht bei 800C gerührt. Das Reaktionsgemisch wird einrotiert und ohne weitere Aufarbeitung weiter eingesetzt. Man erhält so 41 mg (62 % d. Th.) der Zielverbindung.19 mg (0.265 mmol) of isobutylamine and 45 mg (0.442 mmol) of triethylamine are added to a solution of 60 mg (0.177 mmol) of Example 28A in 1.2 ml of THF. The mixture is over Night at 80 0 C stirred. The reaction mixture is concentrated by rotary evaporation and used further without further work-up. This gives 41 mg (62% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.94 min; MS (EIpos): m/z = 376 [M+H]+.LC-MS (Method 1): R, = 1.94 min; MS (EIpos): m / z = 376 [M + H] +.
IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.91 (d, 6H), 0.93 (d, 2H), 1.93-2.00 (m, IH), 2.10- 2.19 (m, IH), 2.73 (d, 2H), 3.39 (t, 2H), 3.87 (s, 3H), 7.57 (d, 2H), 7.85 (t, IH), 8.35 (d, 2H).IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.91 (d, 6H), 0.93 (d, 2H), 1.93-2.00 (m, IH), 2.10-2.29 (m, IH), 2.73 (d, 2H), 3.39 (t, 2H), 3.87 (s, 3H), 7.57 (d, 2H), 7.85 (t, IH), 8.35 (d, 2H).
Beispiel 31 AExample 31A
2-(4-Chloφhenyl)-4-(cyclopentylamino)-6-(2-methylpropyl)pyrimidin-5-carbonsäuremethylester2- (4-Chloφhenyl) -4- (cyclopentylamino) -6- (2-methylpropyl) pyrimidine-5-carbonsäuremethylester
Figure imgf000050_0001
Figure imgf000050_0001
Zu einer Lösung aus 60 mg (0.177 mmol) Beispiel 28A in 1.2 ml THF werden 23 mg (0.265 mmol) Cyclopentylamin und 45 mg (0.442 mmol) Triethylamin zugegeben. Die Mischung wird über Nacht bei 800C gerührt. Das Reaktionsgemisch wird einrotiert und ohne weitere Aufarbeitung weiter eingesetzt. Man erhält so 43 mg (63 % d. Th.) der Zielverbindung.23 mg (0.265 mmol) of cyclopentylamine and 45 mg (0.442 mmol) of triethylamine are added to a solution of 60 mg (0.177 mmol) of Example 28A in 1.2 ml of THF. The mixture is stirred overnight at 80 0 C. The reaction mixture is concentrated by rotary evaporation and used further without further work-up. This gives 43 mg (63% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.99 min; MS (EIpos): m/z = 388 [M+H]+.LC-MS (Method 1): R, = 1.99 min; MS (EIpos): m / z = 388 [M + H] + .
1H-NMR (400 MHz, DMSO-Cl6): δ [ppm] = 0.91 (d, 6H), 1.48-1.76 (6H), 2.03-2.18 (3H), 2.74 (d, 2H), 3.86 (s, 3H), 4.48-4.56 (m, IH), 7.58 (d, 2H), 7.73 (d, IH), 8.36 (d, 2H). 1 H-NMR (400 MHz, DMSO-Cl 6 ): δ [ppm] = 0.91 (d, 6H), 1.48-1.76 (6H), 2.03-2.18 (3H), 2.74 (d, 2H), 3.86 (s , 3H), 4.48-4.56 (m, IH), 7.58 (d, 2H), 7.73 (d, IH), 8.36 (d, 2H).
Beispiel 32AExample 32A
2-(4-Chloφhenyl)-4-(ethylamino)-6-(2-methylpropyl)pyrimidin-5-carbonsäuremethylester
Figure imgf000051_0001
2- (4-Chloφhenyl) -4- (ethylamino) -6- (2-methylpropyl) pyrimidine-5-carbonsäuremethylester
Figure imgf000051_0001
Zu einer Lösung aus 60 mg (0.177 mmol) Beispiel 28 A in 1.2 ml THF werden 12 mg (0.265 mmol) Ethylamin und 45 mg (0.442 mmol) Triethylamin zugegeben. Die Mischung wird bei über Nacht 800C gerührt. Das Reaktionsgemisch wird einrotiert und ohne weitere Aufarbeitung weiter eingesetzt. Man erhält so 32 mg (53 % d. Th.) der Zielverbindung.To a solution of 60 mg (0.177 mmol) of Example 28 A in 1.2 ml of THF are added 12 mg (0.265 mmol) of ethylamine and 45 mg (0.442 mmol) of triethylamine. The mixture is stirred at 80 0 C overnight. The reaction mixture is concentrated by rotary evaporation and used further without further work-up. This gives 32 mg (53% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.79 min; MS (EIpos): m/z = 348 [M+H]+.LC-MS (Method 1): R, = 1.79 min; MS (EIpos): m / z = 348 [M + H] +.
IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.91 (d, 6H), 1,21 (t, 3H), 2.08-2.19 (m, IH), 2.70 (d, 2H), 2.54-3.60 (m, 2H), 3.86 (s, 3H), 7.58 (d, 2H), 7.74 (t, IH), 8.36 (d, 2H).IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.91 (d, 6H), 1.21 (t, 3H), 2.08-2.19 (m, IH), 2.70 (d, 2H), 2.54 -3.60 (m, 2H), 3.86 (s, 3H), 7.58 (d, 2H), 7.74 (t, IH), 8.36 (d, 2H).
Beispiel 33AExample 33A
2-(4-Chloφhenyl)-4-[(cyclopropylmethyl)amino]-6-(2-methylpropyl)pyrimidin-5- carbonsäuremethylester2- (4-chlorophenyl) -4 - [(cyclopropylmethyl) amino] -6- (2-methylpropyl) pyrimidine-5-carboxylic acid methyl ester
Figure imgf000051_0002
Figure imgf000051_0002
Zu einer Lösung aus 60 mg (0.177 mmol) Beispiel 28A in 1.2 ml THF werden 19 mg (0.265 mmol) Cyclopropanmethylamin und 45 mg (0.442 mmol) Triethylamin zugegeben. Die Mischung wird über Nacht bei 8O0C gerührt. Das Reaktionsgemisch wird einrotiert und ohne weitere Aufarbeitung weiter eingesetzt. Man erhält so 31 mg (47 % d. Th.) der Zielverbindung.19 mg (0.265 mmol) of cyclopropanemethylamine and 45 mg (0.442 mmol) of triethylamine are added to a solution of 60 mg (0.177 mmol) of Example 28A in 1.2 ml of THF. The mixture is stirred at 8O 0 C overnight. The reaction mixture is concentrated by rotary evaporation and used further without further work-up. This gives 31 mg (47% of theory) of the target compound.
LC-MS (Methode 1): Rt = 1.88 min; MS (EIpos): m/z = 374 [M+H]+. IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.28-0.32 (m, 2H), 0.43-0.49 (m, 2H), 0.91 (d, 6H), 1.13-1.17 (m, IH), 2.09-2.19 (m, IH), 2.73 (d, 2H), 3.42 (dd, 2H), 3.87 (s, 3H), 7.57 (d, 2H), 7.90 (t, IH), 8.36 (d, 2H).LC-MS (Method 1): R t = 1.88 min; MS (EIpos): m / z = 374 [M + H] +. IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.28-0.32 (m, 2H), 0.43-0.49 (m, 2H), 0.91 (d, 6H), 1.13-1.17 (m, IH) , 2.09-2.19 (m, IH), 2.73 (d, 2H), 3.42 (dd, 2H), 3.87 (s, 3H), 7.57 (d, 2H), 7.90 (t, IH), 8.36 (d, 2H ).
Beispiel 34AExample 34A
2-(4-Chlorphenyl)-4-[( 1 -methylethyl)arrύn]-6-(2-methylpropyl)pyrimidin-5- carbonsäuremethylester2- (4-chlorophenyl) -4 - [(1-methylethyl) arrύn] -6- (2-methylpropyl) pyrimidine-5-carboxylic acid methyl ester
Figure imgf000052_0001
Figure imgf000052_0001
Zu einer Lösung aus 60 mg (0.177 mmol) Beispiel 28 A in 1.2 ml THF werden 16 mg (0.265 mmol) Isopropylamin und 45 mg (0.442 mmol) Triethylamin zugegeben. Die Mischung wird 30 h bei 800C gerührt. Dann werden die flüchtigen Komponenten am Rotationsverdampfer abgetrennt. Der Rückstand wird in Essigsäureethylester aufgenommen und mehrmals mit Wasser gewaschen. Die organische Phase wird über Magnesiumsulfat getrocknet, das Lösungsmittel in Vakuum eingedampft und das Rohprodukt chromatograpisch an Kiesegel gereinigt (Laufinittel: Dichlormethan). Man erhält so 35 mg (49 % d. Th.) der Zielverbindung.To a solution of 60 mg (0.177 mmol) of Example 28 A in 1.2 ml of THF are added 16 mg (0.265 mmol) of isopropylamine and 45 mg (0.442 mmol) of triethylamine. The mixture is stirred at 80 ° C. for 30 h. Then the volatile components are separated on a rotary evaporator. The residue is taken up in ethyl acetate and washed several times with water. The organic phase is dried over magnesium sulfate, the solvent is evaporated in vacuo and the crude product is purified by chromatography on silica gel (mobile phase: dichloromethane). This gives 35 mg (49% of theory) of the target compound.
LC-MS (Methode 3): R, = 3.49 min; MS (EIpos): m/z = 362 [M+H]+.LC-MS (Method 3): R, = 3.49 min; MS (EIpos): m / z = 362 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 0.91 (d, 6H), 1.25 (d, 6H), 2.10-2.18 (m, IH), 2.73 (d, 2H), 3.86 (s, 3H), 4.23-4.51 (m, IH), 7.57 (d, 2H), 8.35 (d, 2H). Ausführungsbeispiele: 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 0.91 (d, 6H), 1.25 (d, 6H), 2.10-2.18 (m, IH), 2.73 (d, 2H), 3.86 (s , 3H), 4.23-4.51 (m, IH), 7.57 (d, 2H), 8.35 (d, 2H). EXAMPLES
Beispiel 1example 1
2-(4-Chlorphenyl)-4-methyl-6-[(l-methylethyl)amino]pyrimidin-5-carbonsäure-Hydrochlorid2- (4-chlorophenyl) -4-methyl-6 - [(l-methylethyl) amino] pyrimidin-5-carboxylic acid hydrochloride
Figure imgf000053_0001
Figure imgf000053_0001
Zu einer Lösung aus 250 mg (0.749 mmol) Beispiel 6A in 15.6 ml Ethanol werden 7.5 ml (14.978 mmol) einer 2M wässrigen Natriumhydroxid-Lösung gegeben. Das Reaktionsgemisch wird 6h bei 800C gerührt. Zur Aufarbeitung wird das Reaktionsgemisch mit IN Salzsäure auf pH 1 gestellt und die flüchtigen Komponenten im Vakuum abdestilliert. Die entstandenen Kristalle werden abgesaugt und im Hochvakuum getrocknet. Man erhält so 195 mg (74% d. Th.) der Zielverbindung.To a solution of 250 mg (0.749 mmol) of Example 6A in 15.6 ml of ethanol is added 7.5 ml (14.978 mmol) of a 2M aqueous sodium hydroxide solution. The reaction mixture is stirred at 80 ° C. for 6 hours. For workup, the reaction mixture is adjusted to pH 1 with 1N hydrochloric acid and the volatile components are distilled off in vacuo. The resulting crystals are filtered off and dried under high vacuum. This gives 195 mg (74% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.01 min; MS (ESIpos): m/z = 306 [M+H-HC1]+.LC-MS (Method 1): R, = 1.01 min; MS (ESIpos): m / z = 306 [M + H-HC1] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.27 (d, 6H), 2,68 (s, 3H), 4.45-4.52 (m, IH), 7.58 (d, 2H), 8.35 (d, 2H), 8.63 (sbr). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 1.27 (d, 6H), 2.68 (s, 3H), 4.45-4.52 (m, IH), 7.58 (d, 2H) , 8.35 (d, 2H), 8.63 (sbr).
Beispiel 2Example 2
2-(4-Chloφhenyl)-4-[(cyclopropylmethyl)amino]-6-methylpyrimidin-5-carbonsäure-Hydrochlorid2- (4-Chloφhenyl) -4 - [(cyclopropylmethyl) amino] -6-methylpyrimidine-5-carboxylic acid hydrochloride
Figure imgf000053_0002
Figure imgf000053_0002
Zu einer Lösung aus 100 mg (0.289 mmol) Beispiel 7A in 6 ml Ethanol werden 2.9 ml (5.783 mmol) einer 2M wässrigen Natriumhydroxid-Lösung gegeben. Das Reaktionsgemisch wird 6h bei 800C gerührt. Zur Aufarbeitung wird das Reaktionsgemisch mit IN Salzsäure auf pH 1 gestellt und die flüchtigen Komponenten im Vakuum abdestilliert. Die entstandenen Kristalle werden abgesaugt und im Hochvakuum getrocknet. Man erhält so 83 mg (79% d. Th.) der Zielverbindung.To a solution of 100 mg (0.289 mmol) of Example 7A in 6 ml of ethanol is added 2.9 ml (5.783 mmol) of a 2M aqueous sodium hydroxide solution. The reaction mixture is stirred at 80 ° C. for 6 hours. For workup, the reaction mixture with 1N hydrochloric acid to pH. 1 placed and distilled off the volatile components in vacuo. The resulting crystals are filtered off and dried under high vacuum. This gives 83 mg (79% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.05 min; MS (ESIpos): m/z = 318 [M+H-HC1]+.LC-MS (Method 1): R, = 1.05 min; MS (ESIpos): m / z = 318 [M + H-HC1] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 0.29-0.33 (m, 2H), 0.46-0.51 (m, 2H), 1.14-1.19 (m, IH), 2.65 (s, 3H), 3.48 (dd, 2H), 7.61 (d, 2H), 8.34 (d, 2H), 8.75 (sbr). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 0.29-0.33 (m, 2H), 0.46-0.51 (m, 2H), 1.14-1.19 (m, IH), 2.65 (s, 3H) , 3.48 (dd, 2H), 7.61 (d, 2H), 8.34 (d, 2H), 8.75 (sbr).
Beispiel 3Example 3
2-(4-Chlorphenyl)-4-(diethylamino)-6-methylpyrimidin-5-carbonsäure-Hydrochlorid2- (4-chlorophenyl) -4- (diethylamino) -6-methyl-pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000054_0001
Figure imgf000054_0001
100 mg (0.287 mmol) Beispiel 9A werden in 6 ml Ethanol aufgenommen und mit 2.8 ml (5.75mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 40 min bei 1400C in einer Single MoJe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird mit IN Salzsäure auf pH 1 gestellt eingeengt und der Rückstand mittels präparativer HPLC (Eluent: Acetonitril/Wasser, Gradient 90:10) aufgereinigt. Man erhält so 15 mg (28% d. Th.) der Zielverbindung.100 mg (0.287 mmol) of Example 9A are taken up in 6 ml of ethanol and admixed with 2.8 ml (5.75 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 140 0 C in a single Mojé microwave (Emrys Optimizer) 40th The mixture is concentrated to pH 1 with 1N hydrochloric acid and the residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 90:10). This gives 15 mg (28% of theory) of the target compound.
Die entstandenen Kristalle werden abgesaugt und im Hochvakuum getrocknet. Man erhält so 25 mg (24% d. Th.) der Zielverbindung.The resulting crystals are filtered off and dried under high vacuum. This gives 25 mg (24% of theory) of the target compound.
LC-MS (Methode 3): R, = 1.62 min; MS (ESIpos): m/z = 320 [M+H]+ -HC1.LC-MS (Method 3): R, = 1.62 min; MS (ESIpos): m / z = 320 [M + H] + -HC1.
1H-NMR (400 MHz, DMSO-(I6): δ [ppm] = 1.19 (t, 6H), 2.38 (s, 3H), 3.57 (q, 4H), 7.55 (d, 2H), 8.31 (d, 2H), 13.58 (sbr, IH). 1 H-NMR (400 MHz, DMSO- (I 6 ): δ [ppm] = 1.19 (t, 6H), 2.38 (s, 3H), 3.57 (q, 4H), 7.55 (d, 2H), 8.31 ( d, 2H), 13.58 (sbr, IH).
Beispiel 4Example 4
2-(4-Chloφhenyl)-4-(ethylamino)-6-(l-methylethyl)pyrimidin-5-carbonsäure-Hydrochlorid
Figure imgf000055_0001
2- (4-Chloφhenyl) -4- (ethylamino) -6- (l-methylethyl) -pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000055_0001
55 mg (ca. 0.142 mmol) Beispiel 1OA werden in 2 ml Ethanol aufgenommen und mit 1.4 ml (2.846 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 20 min bei 1400C und dann 20 min bei 1500C in einer Single Mode-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und der wässrige Rückstand aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugt und im Hochvakuum getrocknet. Man erhält so 15 mg (27% d. Th.) der Zielverbindung.55 mg (about 0.142 mmol) of Example 1OA are taken up in 2 ml of ethanol and treated with 1.4 ml (2,846 mmol) of a 2M aqueous sodium hydroxide solution. It is then reacted at 140 ° C. for 20 minutes and then at 150 ° C. for 20 minutes in a single-mode microwave (Emrys Optimizer). The mixture is concentrated and the aqueous residue is taken up and adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off and dried under high vacuum. This gives 15 mg (27% of theory) of the target compound.
LC-MS (Methode 1): Rt = 1.46 min; MS (ESIpos): m/z = 319 [M+H-HC1]+.LC-MS (Method 1): Rt = 1.46 min; MS (ESIpos): m / z = 319 [M + H-HC1] +.
IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.20 (t, 3H), 1.22 (d, 6H), 3.51-3.59 (m, 2H), 3.65- 3.72 (m, IH), 7.57 (d, 2H), 7.97 (sbr, IH), 8.38 (d, 2H), 13.59 (sbr, IH).IH-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.20 (t, 3H), 1.22 (d, 6H), 3.51-3.59 (m, 2H), 3.65-3.72 (m, IH), 7.57 (d, 2H), 7.97 (sbr, IH), 8.38 (d, 2H), 13.59 (sbr, IH).
Beispiel 5Example 5
2-(4-Chloφhenyl)-4-(cyclopropylamino)-6-(l-methylethyl)pyrimidin-5-carbonsäure-Hydrochlorid2- (4-Chloφhenyl) -4- (cyclopropylamino) -6- (l-methylethyl) -pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000055_0002
Figure imgf000055_0002
80 mg (0.222 mmol) Beispiel I IA werden in 3 ml Ethanol aufgenommen und mit 2.2 ml (4.446 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 20 min bei 1400C in einer Single Λforfe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und der wässrige Rückstand aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugt und im Hochvakuum getrocknet. Man erhält so 82 mg (91% d. Th.) der Zielverbindung. LC-MS (Methode 3): R, = 2.89 min; MS (ESIpos): m/z = 332 [M+H-HC1]+.80 mg (0.222 mmol) of Example I IA are taken up in 3 ml of ethanol and admixed with 2.2 ml (4.446 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 140 0 C in a single Λforfe microwave (Emrys Optimizer) 20th The mixture is concentrated and the aqueous residue is taken up and adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off and dried under high vacuum. This gives 82 mg (91% of theory) of the target compound. LC-MS (Method 3): R, = 2.89 min; MS (ESIpos): m / z = 332 [M + H-HC1] + .
1H-NMR (400 MHz, DMSO-dβ): δ [ppm] = 0.56-0.60 (m, 2H), 0.80-0.85 (m, 2H), 1.22 (d, 6H), 2.98-3.03 (m, IH), 3.61-3.66 (m, IH), 7.58 (d, 2H), 7.86 (sbr, IH), 8.43 (d, 2H). 1 H-NMR (400 MHz, DMSO-dβ): δ [ppm] = 0.56-0.60 (m, 2H), 0.80-0.85 (m, 2H), 1.22 (d, 6H), 2.98-3.03 (m, IH ), 3.61-3.66 (m, IH), 7.58 (d, 2H), 7.86 (sbr, IH), 8.43 (d, 2H).
Beispiel 6Example 6
2-(4-Chloφhenyl)-4-(methylamino)-6-( 1 -methylethyl)pyrimidin-5-carbonsäure-Hydrochlorid2- (4-Chlorophenyl) -4- (methylamino) -6- (1-methylethyl) pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000056_0001
Figure imgf000056_0001
60 mg (0.180 mmol) Beispiel 12A werden in 2 ml Ethanol aufgenommen und mit 1.8 ml (3.595 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 20 min bei 1400C in einer Single Mwfe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und der wässrige Rückstand aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugt und im Hochvakuum getrocknet. Man erhält so 60 mg (97% d. Th.) der Zielverbindung.60 mg (0.180 mmol) of Example 12A are taken up in 2 ml of ethanol and admixed with 1.8 ml (3.595 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 140 0 C in a single Mwfe microwave (Emrys Optimizer) 20th The mixture is concentrated and the aqueous residue is taken up and adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off and dried under high vacuum. This gives 60 mg (97% of theory) of the target compound.
LC-MS (Methode 3): R, = 2.46 min; MS (ESIpos): m/z = 306 [M+H-HC1]+.LC-MS (Method 3): R, = 2.46 min; MS (ESIpos): m / z = 306 [M + H-HC1] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.22 (d, 6H), 3.02 (d, 3H), 7.57 (d, 2H), 7.82 (sbr, IH), 8.40 (d, 2H). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 1.22 (d, 6H), 3.02 (d, 3H), 7.57 (d, 2H), 7.82 (sbr, IH), 8.40 (i.e. , 2H).
Beispiel 7Example 7
2-(4-Chloφhenyl)-4-ethyl-6-[(l-methylethyl)amino]pyrimidin-5-carbonsäure2- (4-Chloφhenyl) -4-ethyl-6 - [(l-methylethyl) amino] pyrimidin-5-carboxylic acid
Figure imgf000056_0002
42 mg (0.121 mmol) Beispiel 16A werden in 5 ml Ethanol aufgenommen und mit 193 mg (4.83 mmol) Natriumhydroxid versetzt. Nach Addition von ein paar Tropfen Wasser wird 2h bei Rückflußtemperatur umgesetzt. Der Ansatz wird eingeengt, mit Wasser aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Anschließend wird mit Essigsäureethyester extrahiert (3x). Dann werden die vereinigten organischen Phasen mit Magnesiumsulfat getrocknet. Das Lösemittel wird am Rotationsverdampfer abgetrennt und der Rückstand mittels präparativer HPLC (Eluent: Acetonitril/Wasser, Gradient 90:10) aufgereinigt. Man erhält so 8 mg (21% d. Th.) der Zielverbindung.
Figure imgf000056_0002
42 mg (0.121 mmol) of Example 16A are taken up in 5 ml of ethanol and admixed with 193 mg (4.83 mmol) of sodium hydroxide. After addition of a few drops of water is reacted for 2 hours at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigsäureethyester (3x). Then the combined organic phases are dried with magnesium sulfate. The solvent is removed on a rotary evaporator and the residue is purified by preparative HPLC (eluent: acetonitrile / water, gradient 90:10). This gives 8 mg (21% of theory) of the target compound.
LC-MS (Methode 3): R, = 2.12 min; MS (ESIpos): m/z = 320 [M+H]+.LC-MS (Method 3): R, = 2.12 min; MS (ESIpos): m / z = 320 [M + H] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.17-1.31 (m, 9H), 2.97 (q, 2H), 4.44 (mz, IH), 7.58 (d, 2H), 8.13 (d, IH), 8.38 (d, 2H), 13.57 (sbr, IH). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 1.17-1.31 (m, 9H), 2.97 (q, 2H), 4.44 (mz, IH), 7.58 (d, 2H), 8.13 (d, IH), 8.38 (d, 2H), 13.57 (sbr, IH).
Beispiel 8Example 8
2-(4-Chloφhenyl)-4-ethyl-6-(prop-2-en-l-ylamino)pyrimidin-5-carbonsäure2- (4-Chloφhenyl) -4-ethyl-6- (prop-2-en-l-ylamino) -pyrimidine-5-carboxylic acid
Figure imgf000057_0001
Figure imgf000057_0001
70 mg (0.202 mmol) Beispiel 17A werden in 5 ml Ethanol aufgenommen und mit 323 mg (8.10 mmol) Natriumhydroxid versetzt. Nach Addition von ein paar Tropfen Wasser wird 2h bei Rückflußtemperatur umgesetzt. Der Ansatz wird eingeengt, mit Wasser aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Anschließend wird mit Essigsäureethyester extrahiert (3x). Dann werden die vereinigten organischen Phasen mit Magnesiumsulfat getrocknet. Das Lösemittel am Rotationsverdampfer abgetrennt. Man erhält so 55 mg (86% d. Th.) der Zielverbindung.70 mg (0.202 mmol) of Example 17A are taken up in 5 ml of ethanol and admixed with 323 mg (8.10 mmol) of sodium hydroxide. After addition of a few drops of water is reacted for 2 hours at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigsäureethyester (3x). Then the combined organic phases are dried with magnesium sulfate. The solvent is separated on a rotary evaporator. This gives 55 mg (86% of theory) of the target compound.
LC-MS (Methode 2): R, = 1.71 min; MS (ESIpos): m/z = 318 [M+H]+.LC-MS (Method 2): R, = 1.71 min; MS (ESIpos): m / z = 318 [M + H] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.25 (t, 3H), 2.97 (q, 2H), 4.22 (mz, 2H), 5.12 (dd, IH), 5.23 (dd, IH), 6.00 (mz, IH), 7.57 (d, 2H), 8.32-8.42 (m, 3H), 13.60 (sbr, IH). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 1.25 (t, 3H), 2.97 (q, 2H), 4.22 (mz, 2H), 5.12 (dd, IH), 5.23 (dd , IH), 6.00 (mz, IH), 7.57 (d, 2H), 8.32-8.42 (m, 3H), 13.60 (sbr, IH).
Beispiel 9Example 9
2-(4-Chloφhenyl)-4-(diethylamino)-6-ethylpyrimidin-5 -carbonsäure
Figure imgf000058_0001
2- (4-chlorophenyl) -4- (diethylamino) -6-ethylpyrimidine-5-carboxylic acid
Figure imgf000058_0001
90 mg (0.249 mmol) Beispiel 18A werden in 5 ml Ethanol aufgenommen und mit 398 mg (9.95 mmol) Natriumhydroxid versetzt. Nach Addition von ein paar Tropfen Wasser wird über Nacht bei Rückflußtemperatur umgesetzt. Der Ansatz wird eingeengt, mit Wasser aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Anschließend wird mit Essigsäureethyester extrahiert (3x). Die vereinigten organischen Phasen werden mit Magnesiumsulfat getrocknet und das Lösemittel wird am Rotationsverdampfer abgetrennt. Der Rückstand wird abschließend mittels präparativer HPLC (Eluent: Acetonitril/Wasser, Gradient 90:10) aufgereinigt. Man erhält so 54 mg (65% d. Th.) der Zielverbindung.90 mg (0.249 mmol) of Example 18A are taken up in 5 ml of ethanol and admixed with 398 mg (9.95 mmol) of sodium hydroxide. After addition of a few drops of water is reacted overnight at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigsäureethyester (3x). The combined organic phases are dried with magnesium sulfate and the solvent is removed on a rotary evaporator. The residue is finally purified by preparative HPLC (eluent: acetonitrile / water, gradient 90:10). This gives 54 mg (65% of theory) of the target compound.
LC-MS (Methode 1): R, = 0.94 min; MS (ESIpos): m/z = 334 [M+H]+.LC-MS (Method 1): R, = 0.94 min; MS (ESIpos): m / z = 334 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.16-1.26 (m, 9H), 2.65 (q, 2H), 3.57 (q, 4H), 7.56 (d, 2H), 8.33 (d, 2H), 13.59 (sbr, IH). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.16-1.26 (m, 9H), 2.65 (q, 2H), 3.57 (q, 4H), 7.56 (d, 2H), 8.33 (i.e. , 2H), 13.59 (sbr, IH).
Beispiel 10Example 10
2-(4-Chloφhenyl)-4-(cyclohexylamino)-6-ethylpyrimidin-5-carbonsäure2- (4-Chloφhenyl) -4- (cyclohexylamino) -6-ethylpyrimidine-5-carboxylic acid
Figure imgf000058_0002
Figure imgf000058_0002
43 mg (0.111 mmol) Beispiel 19A werden in 5 ml Ethanol aufgenommen und mit 177 mg (4.43 mmol) Natriumhydroxid versetzt. Nach Addition von ein paar Tropfen Wasser wird über Nacht bei Rückflußtemperatur umgesetzt. Der Ansatz wird eingeengt, mit Wasser aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Anschließend wird mit Essigsäureethyester extrahiert (3x): Dann werden die vereinigten organischen Phasen mit Magnesiumsulfat getrocknet und das Lösemittel wird am Rotationsverdampfer abgetrennt. Der Rückstand mittels präparativer HPLC (Eluent: Acetonitril/Wasser, Gradient 90:10) aufgereinigt. Man erhält so 7 mg (18% d. Th.) der Zielverbindung.43 mg (0.111 mmol) of Example 19A are taken up in 5 ml of ethanol and admixed with 177 mg (4.43 mmol) of sodium hydroxide. After addition of a few drops of water is reacted overnight at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigsäureethyester (3x): Then the combined organic phases are dried with magnesium sulfate and the solvent is removed on a rotary evaporator. The residue was purified by preparative HPLC (eluent: acetonitrile / water, gradient 90:10). This gives 7 mg (18% of theory) of the target compound.
LC-MS (Methode 3): R, = 2.53 min; MS (ESIpos): m/z = 360 [M+H]+.LC-MS (Method 3): R, = 2.53 min; MS (ESIpos): m / z = 360 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.20-1.51 (m, 9H), 1.60 (m, IH), 1.71 (m, 2H), 1.96 (mz, 2H), 2.98 (q, 2H), 7.58 (d, 2H), 8.30 (mz, IH), 8.36 (d, 2H), 13.59 (sbr, IH). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.20-1.51 (m, 9H), 1.60 (m, IH), 1.71 (m, 2H), 1.96 (mz, 2H), 2.98 (q , 2H), 7.58 (d, 2H), 8.30 (mz, IH), 8.36 (d, 2H), 13.59 (sbr, IH).
Beispiel 11Example 11
2-(4-Chlorphenyl)-4-ethyl-6-piperidin-l-ylpyrimidin-5-carbonsäure2- (4-chlorophenyl) -4-ethyl-6-piperidin-l-yl-pyrimidine-5-carboxylic acid
Figure imgf000059_0001
Figure imgf000059_0001
126 mg (0.337 mmol) Beispiel 2OA werden in 5 ml Ethanol aufgenommen und mit 539 mg (13.48 mmol) Natriumhydroxid versetzt. Nach Addition von ein paar Tropfen Wasser wird über Nacht bei Rückflußtemperatur umgesetzt. Der Ansatz wird eingeengt, mit Wasser aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Anschließend wird mit Essigsäureethyester extrahiert (3x) und die vereinigten organischen Phasen werden mit Magnesiumsulfat getrocknet. Dann wird das Lösemittel am Rotationsverdampfer abgetrennt. Man erhält so 104 mg (87% d. Th.) der Zielverbindung.126 mg (0.337 mmol) of Example 2OA are taken up in 5 ml of ethanol and treated with 539 mg (13.48 mmol) of sodium hydroxide. After addition of a few drops of water is reacted overnight at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. It is then extracted with Essigsäureethyester (3x) and the combined organic phases are dried with magnesium sulfate. Then the solvent is separated on a rotary evaporator. This gives 104 mg (87% of theory) of the target compound.
LC-MS (Methode 3): R, = 1.82 min; MS (ESIpos): m/z = 346 [M+H]+.LC-MS (Method 3): R, = 1.82 min; MS (ESIpos): m / z = 346 [M + H] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 1.23 (t, 3H), 1.53-1.73 (m, 6H), 2.73 (q, 2H), 3.65 (mz, 4H), 7.57 (d, 2H), 8.34 (d, 2H), 13.51 (sbr, IH). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 1.23 (t, 3H), 1.53-1.73 (m, 6H), 2.73 (q, 2H), 3.65 (mz, 4H), 7.57 (d, 2H), 8.34 (d, 2H), 13.51 (sbr, IH).
Beispiel 12Example 12
4-(Diethylamino)-6-ethyl-2-(4-methylphenyl)pyrimidin-5-carbonsäure
Figure imgf000060_0001
4- (diethylamino) -6-ethyl-2- (4-methylphenyl) pyrimidine-5-carboxylic acid
Figure imgf000060_0001
78 mg (0.228 mmol) Beispiel 24A werden in 3 ml Ethanol aufgenommen und mit 365 mg (9.14 mmol) Natriumhydroxid versetzt. Anschließend wird über Nacht bei Rückflußtemperatur umgesetzt. Da der Umsatz unvollständig verbleibt, wird 15 min bei 1400C in einer Single Mode- Mikrowelle (Emrys Optimizer) temperiert. Der Ansatz wird eingeengt, mit Wasser aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Anschließend wird mit Essigsäureethyester extrahiert (3x). Dann werden die vereinigten organischen Phasen mit Magnesiumsulfat getrocknet. Das Lösemittel wird am Rotationsverdampfer abgetrennt. Abschließend wird am Hochvakuum getrocknet. Man erhält so 60 mg (80% d. Th.) der Zielverbindung.78 mg (0.228 mmol) of Example 24A are taken up in 3 ml of ethanol and admixed with 365 mg (9.14 mmol) of sodium hydroxide. It is then reacted overnight at reflux temperature. Since the conversion remains incomplete, 15 min at 140 0 C in a single-mode microwave (Emrys Optimizer) tempered. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. Then it is extracted with Essigsäureethyester (3x). Then the combined organic phases are dried with magnesium sulfate. The solvent is separated on a rotary evaporator. Finally, it is dried in a high vacuum. This gives 60 mg (80% of theory) of the target compound.
LC-MS (Methode 3): R, = 1.60 min; MS (ESIpos): m/z = 314 [M+H]+.LC-MS (Method 3): R, = 1.60 min; MS (ESIpos): m / z = 314 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.14-1.27 (m, 9H), 2.37 (s, 3H), 2.66 (q, 2H), 3.58 (q, 4H), 7.30 (d, 2H), 8.22 (d, 2H), 13.53 (sbr, IH). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.14-1.27 (m, 9H), 2.37 (s, 3H), 2.66 (q, 2H), 3.58 (q, 4H), 7.30 (i.e. , 2H), 8.22 (d, 2H), 13.53 (sbr, IH).
Beispiel 13Example 13
4-Ethyl-6-[(l-methylethyl)amino]-2-(4-methylphenyl)pyrimidin-5-carbonsäure4-ethyl-6 - [(l-methylethyl) amino] -2- (4-methylphenyl) pyrimidine-5-carboxylic acid
Figure imgf000060_0002
Figure imgf000060_0002
68 mg (0.208 mmol) Beispiel 25A werden in 3 ml Ethanol aufgenommen und mit 332 mg (8.31 mmol) Natriumhydroxid versetzt. Nach Addition von ein paar Tropfen Wasser wird über Nacht bei Rückflußtemperatur umgesetzt. Der Ansatz wird eingeengt, mit Wasser aufgenommen und mit IN Salzsäure auf pH 1 gestellt. Anschließend wird mit Essigsäureethyester extrahiert (3x), die vereinigten organischen Phasen werden mit Magnesiumsulfat getrocknet. Dann wird das Lösemittel am Rotationsverdampfer abgetrennt. Man erhält so 54 mg (87% d. Th.) der Zielverbindung.68 mg (0.208 mmol) of Example 25A are taken up in 3 ml of ethanol and admixed with 332 mg (8.31 mmol) of sodium hydroxide. After addition of a few drops of water is reacted overnight at reflux temperature. The mixture is concentrated, taken up with water and adjusted to pH 1 with 1N hydrochloric acid. It is then extracted with ethyl acetate (3x), The combined organic phases are dried with magnesium sulfate. Then the solvent is separated on a rotary evaporator. This gives 54 mg (87% of theory) of the target compound.
LC-MS (Methode 3): R, = 1.82 min; MS (ESIpos): m/z = 300 [M+H]+.LC-MS (Method 3): R, = 1.82 min; MS (ESIpos): m / z = 300 [M + H] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 1.23 (t, 3H), 1.26 (d, 6H), 2.38 (s, 3H), 2.97 (q, 2H), 4.44 (mz, IH), 7.31 (d, 2H), 8.17 (sbr, IH), 8.28 (d, 2H), 13.47 (sbr, IH). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 1.23 (t, 3H), 1.26 (d, 6H), 2.38 (s, 3H), 2.97 (q, 2H), 4.44 (mz, IH ), 7.31 (d, 2H), 8.17 (sbr, IH), 8.28 (d, 2H), 13.47 (sbr, IH).
Beispiel 14Example 14
2-(4-Chlθφhenyl)-4-(2-methylpropyl)-6-[(2-methylpropyl)amino]pyrimidin-5-carbonsäure- Hydrochlorid2- (4-Chloro-phenyl) -4- (2-methyl-propyl) -6 - [(2-methyl-propyl) -amino] -pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000061_0001
Figure imgf000061_0001
41 mg (0.109 mmol) Beispiel 30A werden in 1.2 ml Dioxan aufgenommen und mit 2.2 ml (4.64 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 40 min bei 1500C in einer Single Λ/brfe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und der Rückstand mit Wasser aufgenommen. Dann wird mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugt, erneut mit Wasser aufgenommen und mit Essigsäureethyester extrahiert (3x). Die organische Phase wird eingeengt. Man erhält so 16 mg (36% d. Th.) der Zielverbindung.41 mg (0.109 mmol) of Example 30A are taken up in 1.2 ml of dioxane and admixed with 2.2 ml (4.64 mmol) of a 2M aqueous sodium hydroxide solution. The mixture is then reacted for 40 min at 150 0 C in a single Λ / brfe microwave (Emrys Optimizer). The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off, re-absorbed with water and extracted with Essigsäureethyester (3x). The organic phase is concentrated. This gives 16 mg (36% of theory) of the target compound.
LC-MS (Methode 1): R, = 1.40 min; MS (EIpos): m/z = 362 [M+H-HC1]+.LC-MS (Method 1): R, = 1.40 min; MS (EIpos): m / z = 362 [M + H-HC1] + .
1H-NMR (400 MHz, DMSO-(I6): δ [ppm] = 0.91 (d, 6H), 0.94 (d, 6H), 1.89-2.03 (m, IH), 2.13- 2.23 (m, IH), 2.86 (d, 2H), 3.42 (dd, 2H), 7.57 (d, 2H), 8.26 (sbr, IH), 8.36 (d, 2H). 1 H-NMR (400 MHz, DMSO- (I 6 ): δ [ppm] = 0.91 (d, 6H), 0.94 (d, 6H), 1.89-2.03 (m, IH), 2.13-2.23 (m, IH ), 2.86 (d, 2H), 3.42 (dd, 2H), 7.57 (d, 2H), 8.26 (sbr, IH), 8.36 (d, 2H).
Beispiel 15Example 15
2-(4-Chlθφhenyl)-4-(cyclopentylamino)-6-(2-methylpropyl)pyrimidin-5-carbonsäure- Hydrochlorid
Figure imgf000062_0001
2- (4-Chloro-phenyl) -4- (cyclopentylamino) -6- (2-methyl-propyl) -pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000062_0001
41 mg (0.106 mmol) Beispiel 31A werden in 1.2 ml Dioxan aufgenommen und mit 2.1 ml (4.23 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 40 min bei 1500C in einer Single Λ/oJe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und der Rückstand mit Wasser aufgenommen. Dann wird mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugt, erneut mit Wasser aufgenommen und mit Essigsäureethyester extrahiert (3x). Die organische Phase wird eingeengt. Man erhält so 21 mg (47% d. Th.) der Zielverbindung.41 mg (0.106 mmol) of Example 31A are taken up in 1.2 ml of dioxane and admixed with 2.1 ml (4.23 mmol) of a 2M aqueous sodium hydroxide solution. It is then 40 min at 150 0 C in a single Λ / oJe microwave (Emrys Optimizer) implemented. The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off, re-absorbed with water and extracted with Essigsäureethyester (3x). The organic phase is concentrated. This gives 21 mg (47% of theory) of the target compound.
LC-MS (Methode 3): R, = 2.64 min; MS (EIpos): m/z = 374 [M+H-HC1]+.LC-MS (Method 3): R, = 2.64 min; MS (EIpos): m / z = 374 [M + H-HC1] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 0.91 (d, 6H), 1.48-1.73 (m, 6H), 2.05-2.12 (m, 2H), 2.13-2.22 (m, IH), 2.88 (d, 2H), 4.47-4.55 (m, IH), 7.57 (d, 2H), 8.18 (d, IH), 8.37 (d, 2H), 13.63 (sbr, IH). 1 H-NMR (400 MHz, DMSOd 6 ): δ [ppm] = 0.91 (d, 6H), 1.48-1.73 (m, 6H), 2.05-2.12 (m, 2H), 2.13-2.22 (m, IH) , 2.88 (d, 2H), 4.47-4.55 (m, IH), 7.57 (d, 2H), 8.18 (d, IH), 8.37 (d, 2H), 13.63 (sbr, IH).
Beispiel 16Example 16
2-(4-Chloφhenyl)-4-(ethylamino)-6-(2-methylpropyl)pyrimidin-5-carbonsäure-Hydrochlorid2- (4-Chloφhenyl) -4- (ethylamino) -6- (2-methylpropyl) pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000062_0002
Figure imgf000062_0002
31 mg (0.089 mmol) Beispiel 32A werden in 1.0 ml Dioxan aufgenommen und mit 1.8 ml (3.565 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 40 min bei 1500C in einer Single Aforfe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und der Rückstand mit Wasser aufgenommen. Dann wird mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugt, erneut mit Wasser aufgenommen und mit Essigsäureethyester extrahiert (3x). Die organische Phase wird eingeengt. Man erhält so 15 mg (45% d. Th.) der Zielverbindung.31 mg (0.089 mmol) of Example 32A are taken up in 1.0 ml of dioxane and admixed with 1.8 ml (3.565 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 150 0 C in a single Aforfe microwave (Emrys Optimizer) 40th The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid posed. The resulting crystals are filtered off, re-absorbed with water and extracted with Essigsäureethyester (3x). The organic phase is concentrated. This gives 15 mg (45% of theory) of the target compound.
LC-MS (Methode 3): R, = 2.14 min; MS (EIpos): m/z = 324 [M+H-HC1]+.LC-MS (Method 3): R, = 2.14 min; MS (EIpos): m / z = 324 [M + H-HC1] + .
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 0.91 (d, 6H), 1.21 (t, 3H), 2.12-2.28 (m, IH), 2.85 (d, 2H), 3.55-3.66 (m, IH), 7.57 (d, 2H), 8.09 (sbr, IH), 8.36 (d, 2H), 13.52 (sbr, IH). 1 H-NMR (400 MHz, DMSO-d 6 ): δ [ppm] = 0.91 (d, 6H), 1.21 (t, 3H), 2.12-2.28 (m, IH), 2.85 (d, 2H), 3.55 -3.66 (m, IH), 7.57 (d, 2H), 8.09 (sbr, IH), 8.36 (d, 2H), 13.52 (sbr, IH).
Beispiel 17Example 17
2-(4-Chloφhenyl)-4-[(cyclopropylmethyl)amino]-6-(2-methylpropyl)pyrimidin-5-carbonsäure- Hydrochlorid2- (4-Chlorophenyl) -4 - [(cyclopropylmethyl) amino] -6- (2-methylpropyl) pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000063_0001
Figure imgf000063_0001
30 mg (0.080 mmol) Beispiel 33A werden in 0.9 ml Dioxan aufgenommen und mit 1.6 ml (3.210 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 40 min bei 1500C in einer Single Mwfe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und der Rückstand mit Wasser aufgenommen. Dann wird mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugt, erneut mit Wasser aufgenommen und mit Essigsäureethyester extrahiert (3x). Die organische Phase wird eingeengt. Man erhält so 16 mg (50% d. Th.) der Zielverbindung.30 mg (0.080 mmol) of Example 33A are taken up in 0.9 ml of dioxane and admixed with 1.6 ml (3.210 mmol) of a 2M aqueous sodium hydroxide solution. It is then 40 min at 150 0 C in a single-Mwfe microwave (Emrys Optimizer) implemented. The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off, re-absorbed with water and extracted with Essigsäureethyester (3x). The organic phase is concentrated. This gives 16 mg (50% of theory) of the target compound.
LC-MS (Methode 3): R, = 2.48 min; MS (EIpos): m/z = 360 [M+H-HC1]+.LC-MS (Method 3): R, = 2.48 min; MS (EIpos): m / z = 360 [M + H-HC1] + .
1H-NMR (400 MHz, DMSOd6): δ [ppm] = 0.25-0.30 (m, 2H), 0.41-0.48 (m, 2H), 0.91 (d, 6H), 1.06-1.15 (m, IH), 2.13-2.23 (m, IH), 2.87 (d, 2H), 3.43 (dd, 2H), 7.57 (d, 2H), 8.23 (sbr, IH), 8.36 (d, 2H), 13.55 (sbr, IH). Beispiel 18 1 H NMR (400 MHz, DMSOd 6 ): δ [ppm] = 0.25-0.30 (m, 2H), 0.41-0.48 (m, 2H), 0.91 (d, 6H), 1.06-1.15 (m, IH) , 2.13-2.23 (m, IH), 2.87 (d, 2H), 3.43 (dd, 2H), 7.57 (d, 2H), 8.23 (sbr, IH), 8.36 (d, 2H), 13.55 (sbr, IH ). Example 18
2-(4-Chlorphenyl)-4-[(l-methylethyl)amin]-6-(2-methylpropyl)pyrimidin-5-carbonsäure- Hydrochlorid2- (4-chlorophenyl) -4 - [(1-methylethyl) amine] -6- (2-methylpropyl) pyrimidine-5-carboxylic acid hydrochloride
Figure imgf000064_0001
Figure imgf000064_0001
33 mg (0.091 mmol) Beispiel 34A werden in 1.0 ml Dioxan aufgenommen und mit 1.8 ml (3.648 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 40 min bei 1500C in einer Single Mocfe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und der Rückstand mit Wasser aufgenommen. Dann wird mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugt. Man erhält so 24 mg (68% d. Th.) der Zielverbindung.33 mg (0.091 mmol) of Example 34A are taken up in 1.0 ml of dioxane and admixed with 1.8 ml (3.648 mmol) of a 2M aqueous sodium hydroxide solution. Min is then reacted at 150 0 C in a single Mocfe microwave (Emrys Optimizer) 40th The mixture is concentrated and the residue taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off with suction. This gives 24 mg (68% of theory) of the target compound.
LC-MS (Methode 3): R, = 2.38 min; MS (EIpos): m/z = 348 [M+H-HC1]+.LC-MS (Method 3): R, = 2.38 min; MS (EIpos): m / z = 348 [M + H-HC1] + .
1H-NMR (400 MHz, DMSO-O6): δ [ppm] = 0.91 (d, 6H), 1.26 (d, 6H), 2.13-2.23 (m, IH), 2.87 (d, 2H), 4.37-4.47 (m, IH), 7.57 (d, 2H), 8.00 (sbr, IH), 8.36 (d, 2H), 13.57 (sbr, IH). 1 H-NMR (400 MHz, DMSO-O 6 ): δ [ppm] = 0.91 (d, 6H), 1.26 (d, 6H), 2.13-2.23 (m, IH), 2.87 (d, 2H), 4.37 -4.47 (m, IH), 7.57 (d, 2H), 8.00 (sbr, IH), 8.36 (d, 2H), 13.57 (sbr, IH).
Beispiel 19Example 19
2-(4-Chloφhenyl)-4-(diethylamino)-6-(2-methylpropyl)pyrimidin-5-carbonsäure2- (4-Chloφhenyl) -4- (diethylamino) -6- (2-methylpropyl) pyrimidine-5-carboxylic acid
Figure imgf000064_0002
Figure imgf000064_0002
36 mg (0.091 mmol) Beispiel 29A werden in 1.0 ml Dioxan aufgenommen und mit 1.9 ml (3.83 mmol) einer 2M wässrigen Natriumhydroxid-Lösung versetzt. Anschließend wird 40 min bei 1500C und 40 min bei 1700C in einer Single Afocfe-Mikrowelle (Emrys Optimizer) umgesetzt. Der Ansatz wird eingeengt und Rückstand mit Wasser aufgenommen. Dann wird mit IN Salzsäure auf pH 1 gestellt. Die entstandenen Kristalle werden abgesaugtund durch HPLC (Sunfire C 1, Laufmittel Acetonitril /0.2%ige wässrige TFA) gereinigt. Man erhält so 0.6 mg (2% d. Th.) der Zielverbindung.36 mg (0.091 mmol) of Example 29A are taken up in 1.0 ml of dioxane and admixed with 1.9 ml (3.83 mmol) of a 2M aqueous sodium hydroxide solution. This is followed by 40 min 150 0 C and 40 min at 170 0 C in a single Afocfe microwave (Emrys Optimizer) implemented. The mixture is concentrated and residue is taken up in water. Then it is adjusted to pH 1 with 1N hydrochloric acid. The resulting crystals are filtered off with suction and purified by HPLC (Sunfire C 1, eluent acetonitrile / 0.2% aqueous TFA). This gives 0.6 mg (2% of theory) of the target compound.
LC-MS (Methode 2): R, = 2.20 min; MS (EIpos): m/z = 362 [M+H-HC1]+. LC-MS (Method 2): R, = 2.20 min; MS (EIpos): m / z = 362 [M + H-HC1] + .
B. Bewertung der pharmakologischen WirksamkeitB. Evaluation of Pharmacological Activity
Die pharmakologische Wirkung der erfindungsgemäßen Verbindungen kann in folgenden Assays gezeigt werden:The pharmacological activity of the compounds according to the invention can be demonstrated in the following assays:
B-I: Zellulärer Transaktivierungs-Assav:B-I: Cellular Transactivation Assav:
a) Testprinzip:a) test principle:
Ein zellulärer Assay wird eingesetzt zur Identifizierung von Aktivatoren des Peroxisom- Proliferator-aktivierten Rezeptors alpha (PPAR-alpha).A cellular assay is used to identify activators of the peroxisome proliferator-activated receptor alpha (PPAR-alpha).
Da Säugetierzellen verschiedene endogene nukleare Rezeptoren enthalten, die eine eindeutige Interpretation der Ergebnisse komplizieren könnten, wird ein etabliertes Chimärensystem ein- gesetzt, in dem die Liganden-Bindungsdomäne des humanen PPARα-Rezeptors an die DNA- Bindungsdomäne des Hefe-Transkriptionsfaktors GAL4 fusioniert wird. Die so entstehende GAL4-PPARα-Chimäre wird in CHO-Zellen mit einem Reporterkonstrukt co-transfϊziert und stabil exprimiert.Since mammalian cells contain various endogenous nuclear receptors that could complicate unambiguous interpretation of the results, an established chimera system is used in which the ligand-binding domain of the human PPARα receptor is fused to the DNA binding domain of the yeast transcription factor GAL4. The resulting GAL4-PPARα chimera is co-transfected into CHO cells with a reporter construct and stably expressed.
b) Klonierung:b) Cloning:
Das GAL4-PPARα-Expressions-Konstrukt enthält die Ligandenbindungsdomäne von PP ARa (Aminosäuren 167-468), welche PCR-amplifiziert wird und in den Vektor pcDNA3.1 hinein- kloniert wird. Dieser Vektor enthält bereits die GAL4-DNA-Bindungsdomäne (Aminosäuren 1- 147) des Vektors pFC2-dbd (Stratagene). Das Reporterkonstrukt, welches fünf Kopien der GAL4- Bindestelle vorgeschaltet vor einem Thymidinkinase-Promoter enthält, führt zur Expression der Firefly-Luciferase (Photinus pyralis) nach Aktivierung und Bindung von GAL4-PPARα.The GAL4-PPARα expression construct contains the ligand binding domain of PPARa (amino acids 167-468), which is PCR amplified and cloned into the vector pcDNA3.1. This vector already contains the GAL4 DNA binding domain (amino acids 1-147) of the vector pFC2-dbd (Stratagene). The reporter construct, containing five copies of the GAL4 binding site upstream of a thymidine kinase promoter, expresses the firefly luciferase (Photinus pyralis) upon activation and binding of GAL4-PPARα.
c) Testablauf:c) Test procedure:
CHO (chinese hamster ovary)-Zellen, die die oben beschriebene GAL4-PPARα-Chimäre und das Luciferase-Reportergenkonstrukt stabil exprimieren, werden am Tag vor dem Test in Medium (Optimem, GIBCO), 2% Aktivkohle-gereinigtes fötales Kälberserum (Hyclone), 1.35 mM Natriumpyruvat (GIBCO), 0.2% Natriumbicarbonat (GIBCO) mit 1 x 103 Zellen in 96-Loch- Mikrotiteφlatten ausplattiert und in einem Zellinkubator (96% Luftfeuchtigkeit, 5% v/v CO2, 37°C) gehalten. Am Testtag werden die zu prüfenden Substanzen in oben genanntem Medium, allerdings ohne Zusatz von Kälberserum, aufgenommen und zu den Zellen hinzugegeben. Nach einer Stimulationszeit von 6 h wird die Luciferaseaktivität mit Hilfe einer Videokamera gemessen. Die gemessenen relativen Lichteinheiten ergeben in Abhängigkeit von der Substanzkonzentration eine sigmoide Stimulationskurve. Die Berechnung der EC50- Werte erfolgt mit Hilfe des Computerprogramms GraphPad PRISM (Version 3.02).CHO (Chinese hamster ovary) cells stably expressing the GAL4-PPARα chimera and the luciferase reporter gene construct described above are in the medium (Optimem, GIBCO), 2% activated charcoal-purified fetal calf serum (Hyclone) the day before the test. , 1.35 mM sodium pyruvate (GIBCO), 0.2% sodium bicarbonate (GIBCO) with 1 x 10 3 cells plated in 96-well microtitre plates and maintained in a cell incubator (96% humidity, 5% v / v CO 2 , 37 ° C). On the test day, the substances to be tested are taken up in the above-mentioned medium, but without the addition of calf serum, and added to the cells. After a stimulation time of 6 h, the luciferase activity is measured using a video camera. The measured relative light units give as a function of the substance concentration a sigmoidal stimulation curve. The EC 50 values are calculated using the computer program GraphPad PRISM (version 3.02).
In der folgenden Tabelle sind die EC50- Werte repräsentativer Beispielverbindungen aufgeführt:The following table lists the EC 50 values of representative example compounds:
Tabelletable
Figure imgf000067_0001
Figure imgf000067_0001
B-2: Fibrinogenbestimmung:B-2: fibrinogen determination:
Zur Bestimmung der Wirkung auf die Plasma-Fibrinogen-Konzentration werden männliche Wistar-Ratten oder NMRI-Mäuse für einen Zeitraum von 4-9 Tagen per Schlundsonden-Applikation oder über Futterbeimischung mit der zu untersuchenden Substanz behandelt. Anschließend wird in Terminalnarkose Citratblut durch Herzpunktion gewonnen. Die Plasma-Fibrinogen-Spiegel werden nach der Clauss-Methode [A. Clauss, Acta Haematol. J/7, 237-46 (1957)] durch Messung der Thrombinzeit mit humanem Fibrinogen als Standard bestimmt.To determine the effect on the plasma fibrinogen concentration male Wistar rats or NMRI mice are treated for a period of 4-9 days by gavage or via feed admixture with the substance to be examined. Subsequently, in terminal anesthesia citrated blood is obtained by cardiac puncture. The plasma fibrinogen levels are determined by the Clauss method [A. Clauss, Acta Haematol. J / 7, 237-46 (1957)] by measuring the thrombin time with human fibrinogen as a standard.
B-3: Testbeschreibung zur Auffindung von pharmakologisch wirksamen Substanzen, die das Apoprotein Al (ApoAl) und das HDL-Cholesterin (HDL-C) im Serum von transgenen Mäusen, die mit dem humanen ApoAl-Gen (hApoAl) transfiziert sind, erhöhen bzw. dieB-3: Test description for the discovery of pharmacologically active substances which increase the apoprotein Al (ApoAl) and HDL-cholesterol (HDL-C) in the serum of transgenic mice transfected with the human ApoAl gene (hApoAl) . the
Serumtriglyzeride (TG) senken:Lower serum triglycerides (TG):
Die Substanzen, die auf ihre HDL-C erhöhende Wirkung in vivo untersucht werden sollen, werden männlichen transgenen hApoAl -Mäusen oral verabreicht. Die Tiere werden einen Tag vor Versuchsbeginn randomisiert Gruppen mit gleicher Tierzahl, in der Regel n = 7-10, zugeordnet. Während des gesamten Versuches steht den Tieren Trinkwasser und Futter ad libitum zur Verfügung. Die Substanzen werden einmal täglich 7 Tage lang oral verabreicht. Zu diesem Zweck werden die Testsubstanzen in einer Lösung aus Solutol HS 15 + Ethanol + Kochsalzlösung (0.9%) im Verhältnis 1+1+8 oder in einer Lösung aus Solutol HS 15 + Kochsalzlösung (0.9%) im Verhältnis 2+8 gelöst. Die Applikation der gelösten Substanzen erfolgt in einem Volumen von 10 ml/kg Körpergewicht mit einer Schlundsonde. Als Kontrollgruppe dienen Tiere, die genauso behandelt werden, aber nur das Lösungsmittel (10 ml/kg Körpergewicht) ohne Testsubstanz erhalten.The substances to be tested for their HDL-C increasing activity in vivo are orally administered to male transgenic hApoAl mice. The animals are randomly assigned to groups with the same number of animals, usually n = 7-10, one day before the start of the experiment. Throughout the experiment, the animals have access to drinking water and food ad libitum. The substances are administered orally once a day for 7 days. For this purpose, the test substances are dissolved in a solution of Solutol HS 15 + ethanol + saline (0.9%). in the ratio 1 + 1 + 8 or dissolved in a solution of Solutol HS 15 + saline (0.9%) in the ratio 2 + 8. The application of the dissolved substances takes place in a volume of 10 ml / kg body weight with a gavage. Animals which are treated in the same way, but only the solvent (10 ml / kg body weight) without test substance, serve as a control group.
Vor der ersten Substanzapplikation wird jeder Maus zur Bestimmung von ApoAl, Serumcholesterin, HDL-C und Serumtriglyzeriden (TG) Blut durch Punktion des retroorbitalen Venen- plexus entnommen (Vorwert). Anschließend wird den Tieren mit einer Schlundsonde die Testsubstanz zum ersten Mal verabreicht. 24 Stunden nach der letzten Substanzapplikation (am 8. Tag nach Behandlungsbeginn) wird jedem Tier zur Bestimmung der gleichen Parameter erneut Blut durch Punktion des retroorbitalen Venenplexus entnommen. Die Blutproben werden zentrifugiert und nach Gewinnung des Serums werden TG, Cholesterin, HDL-C und humanes ApoAl mit einem Cobas Integra 400 plus-Gerät (Cobas Integra, Fa. Roche Diagnostics GmbH, Mannheim) unter Verwendung der jeweiligen Kassetten (TRIGL, CHOL2, HDL-C und APOAT) bestimmt. HDL-C wird durch Gelfϊltration und Nachsäulenderivatisierung mit MEGA Cholesterol-Reagens (Fa. Merck KGaA) analog zur Methode von Garber et al. [J. Lipid Res. 4L, 1020-1026 (2000)] bestimmt.Before the first substance administration, each mouse is sampled for the determination of ApoAl, serum cholesterol, HDL-C and serum triglycerides (TG) by puncture of the retroorbital venous plexus (initial value). Subsequently, the animals are given the test substance for the first time with a gavage. 24 hours after the last substance application (on the 8th day after the start of treatment), each animal is again drawn by puncture of the retroorbital venous plexus to determine the same parameters. The blood samples are centrifuged and, after recovery of the serum, TG, cholesterol, HDL-C and human ApoAl are incubated with a Cobas Integra 400 plus instrument (Cobas Integra, Roche Diagnostics GmbH, Mannheim) using the respective cassettes (TRIGL, CHOL2, HDL-C and APOAT). HDL-C is purified by gel filtration and post-column derivatization with MEGA cholesterol reagent (Merck KGaA) analogously to the method of Garber et al. [J. Lipid Res. 4L, 1020-1026 (2000)].
Die Wirkung der Testsubstanzen auf die HDL-C-, hApoAl- bzw. TG-Konzentrationen wird durch Subtraktion des Messwertes der 1. Blutentnahme (Vorwert) von dem Messwert der 2. Blutentnahme (nach Behandlung) bestimmt. Es werden die Differenzen aller HDL-C-, hApoAl- bzw. TG- Werte einer Gruppe gemittelt und mit dem Mittelwert der Differenzen der Kontrollgruppe verglichen. Die statistische Auswertung erfolgt mit Student's t-Test nach vorheriger Überprüfung der Varianzen auf Homogenität.The effect of the test substances on the HDL-C, hApoAl or TG concentrations is determined by subtracting the measured value of the first blood sample (pre-value) from the measured value of the second blood sample (after treatment). The differences of all HDL-C, hApoAl and TG values of a group are averaged and compared with the mean of the differences of the control group. The statistical evaluation is done with Student's t-test after checking the variances for homogeneity.
Substanzen, die das HDL-C der behandelten Tiere, verglichen mit dem der Kontrollgruppe, statistisch signifikant (p<0.05) um mindestens 20% erhöhen oder die TG statistisch signifikant (p<0.05) um mindestens 25% senken, werden als pharmakologisch wirksam angesehen.Substances which increase the HDL-C of the treated animals statistically significantly (p <0.05) by at least 20% or decrease the TG statistically significantly (p <0.05) by at least 25% compared to those of the control group are considered to be pharmacologically active ,
B-4: DOCA/Salz-Modell:B-4: DOCA / salt model:
Die Verabreichung von Desoxycorticosteronacetat (DOCA) in Kombination mit einer Hochsalzdiät und einseitiger Nierenentfernung induziert bei der Ratte einen Hypertonus, der durch relativ niedrige Reninspiegel charakterisiert ist. Als Folge dieser endokrinen Hypertonie (DOCA ist eine direkte Vorstufe von Aldosteron) kommt es in Abhängigkeit von der gewählten DOCA-Konzen- tration zu einer Hypertrophie des Herzens und weiteren Endorgan-Schäden, z.B. der Niere, die u.a. durch Proteinurie und Glomerulosklerose charakterisiert sind. In diesem Rattenmodell lassen sich somit Testsubstanzen auf vorhandene antihypertrophe und Endorgan-schützende Wirkung hin untersuchen.The administration of desoxycorticosterone acetate (DOCA) in combination with a high-salt diet and unilateral kidney removal induces hypertension in the rat, characterized by relatively low renin levels. As a result of this endocrine hypertension (DOCA is a direct precursor of aldosterone), hypertrophy of the heart and other end organ damage, eg kidney, characterized by proteinuria and glomerulosclerosis, depending on the DOCA concentration selected. In this rat model can be thus examine test substances for existing antihypertrophic and end organ protective effect.
Etwa 8 Wochen alte (Körpergewicht zwischen 250 und 300 Gramm), männliche Sprague Dawley (SD)-Ratten werden linksseitig uninephrektomiert. Dazu werden die Ratten mit 1.5-2%-igem Iso- fluran in einer Mischung aus 66% N2O und 33% O2 anästhesiert und die Niere über einen Flankenschnitt entfernt. Als spätere Kontrolltiere dienen sogenannte sham-operierte Tiere, denen keine Niere entfernt wird.About 8 weeks old (body weight between 250 and 300 grams), male Sprague Dawley (SD) rats are left uninephrectomized. For this purpose, the rats are anesthetized with 1.5-2% isoflurane in a mixture of 66% N 2 O and 33% O 2 and the kidney is removed via a flank incision. As a later control animals serve so-called sham-operated animals, which no kidney is removed.
Uninephrektomierte SD-Ratten erhalten 1% Natriumchlorid im Trinkwasser und einmal wöchentlich eine subkutane Injektion von Desoxycorticosteronacetat (gelöst in Sesamöl; Fa. Sigma) zwischen die Schulterblätter gespritzt (Hochdosis: 100 mg/kg/Woche s.c; Normaldosis: 30 mg/kg/Woche s.c).Uninephrectomized SD rats receive 1% sodium chloride in drinking water and once weekly a subcutaneous injection of desoxycorticosterone acetate (dissolved in sesame oil, Sigma) injected between the shoulder blades (high dose: 100 mg / kg / week sc, normal dose: 30 mg / kg / week sc).
Die Substanzen, die auf ihre protektive Wirkung in vivo untersucht werden sollen, werden per Gavage oder über das Futter (Fa. Ssniff) oder Trinkwasser verabreicht. Die Tiere werden einen Tag vor Versuchsbeginn randomisiert und Gruppen mit gleicher Tierzahl, in der Regel n = 10, zugeordnet. Während des gesamten Versuchs steht den Tieren Trinkwasser und Futter ad libitum zur Verfügung. Die Substanzen werden einmal täglich 4-6 Wochen lang per Gavage, Futter oder Trinkwasser verabreicht. Als Plazebogruppe dienen Tiere, die genauso behandelt werden, aber entweder nur das Lösungsmittel oder das Futter bzw. Trinkwasser ohne Testsubstanz erhalten.The substances that are to be tested for their protective effect in vivo are administered by gavage or via the feed (Ssniff) or drinking water. The animals are randomized one day before the start of the experiment and assigned to groups with the same number of animals, usually n = 10. Throughout the experiment, the animals have access to drinking water and food ad libitum. The substances are administered once a day for 4-6 weeks by gavage, food or drinking water. The placebo group used is animals that are treated in the same way, but which either only receive the solvent or the feed or drinking water without the test substance.
Die Wirkung der Testsubstanzen wird durch Messung hämodynamischer Parameter [Blutdruck, Herzfrequenz, Inotropie (dp/dt), Relaxationszeit (tau), maximaler linksventrikulärer Druck, links- ventrikulärer enddiastolischer Druck (LVEDP)], Gewichtsbestimmung von Herz, Niere und Lunge, Messung der Proteinausscheidung sowie durch Messung der Genexpression von Bio- markern (z.B. ANP, Atrial Natriuretic Peptide, und BNP, Brain Natriuretic Peptide) mittels RT/TaqMan-PCR nach RNA-Isolation aus kardialem Gewebe bestimmt.The effect of the test substances is determined by measuring hemodynamic parameters [blood pressure, heart rate, inotropy (dp / dt), relaxation time (tau), maximum left ventricular pressure, left ventricular end-diastolic pressure (LVEDP)], weight determination of heart, kidney and lung Protein excretion and by measuring the gene expression of biomarkers (eg ANP, Atrial Natriuretic Peptide, and BNP, Brain Natriuretic Peptide) by RT / TaqMan PCR after RNA isolation from cardiac tissue determined.
Die statistische Auswertung erfolgt mit Student's t-Test nach vorheriger Überprüfung der Varianzen auf Homogenität.The statistical evaluation is done with Student's t-test after checking the variances for homogeneity.
B-5: Bestimmung der metabolischen StabilitätB-5: Determination of metabolic stability
Zur Bestimmung der metabolischen Stabilität von Testverbindungen werden diese in vitro mit Lebermikrosomen oder bevorzugt mit primären frischen Hepatozyten verschiedener Tierspezies (z.B. von Ratte und Hund) als auch humanen Ursprungs inkubiert, um Metabolitenprofile eines möglichst kompletten hepatischen Phase I- und Phase Ü-Metabolismus zu erhalten und zu vergleichen. Die Testverbindungen werden mit einer Konzentration von 10-20 μM inkubiert. Dazu werden Stammlösungen der Substanzen mit einer Konzentration von 1-2 mM in Acetonitril hergestellt und dann mit einer l :100-Verdünnung in den Inkubationsansatz pipettiert. Die Lebermikrosomen werden in 50 mM Kaliumphosphat-Puffer (pH 7.4) mit und ohne NADPH-generierendem System, bestehend aus 1 mM NADP+, 10 mM Glucose-6-phosphat und 1 Einheit Glucose-6-phosphat- Dehydrogenase, bei 37°C inkubiert. Primäre Hepatozyten werden in Suspension in Williams E- Medium ebenfalls bei 37°C inkubiert. Nach einer Inkubationszeit von 0-4 Stunden werden die Inkubationsansätze mit Acetonitril abgestoppt (Endkonzentration ca. 30%) und das Protein bei ca. 15000 x g abzentrifugiert. Die so abgestoppten Proben werden entweder direkt analysiert oder bis zur Analyse bei -200C gelagert.To determine the metabolic stability of test compounds, they are incubated in vitro with liver microsomes or preferably with primary fresh hepatocytes of various animal species (eg from rat and dog) as well as of human origin to obtain metabolite profiles of a complete hepatic phase I and phase II metabolism and compare. The test compounds are incubated at a concentration of 10-20 μM. For this purpose, stock solutions of the substances are prepared in a concentration of 1-2 mM in acetonitrile and then pipetted with a 1: 100 dilution in the incubation mixture. Liver microsomes are incubated in 50 mM potassium phosphate buffer (pH 7.4) with and without NADPH-generating system consisting of 1 mM NADP + , 10 mM glucose-6-phosphate and 1 unit of glucose-6-phosphate dehydrogenase at 37 ° C incubated. Primary hepatocytes are also incubated in suspension in Williams E medium at 37 ° C. After an incubation period of 0-4 hours, the incubation approaches are stopped with acetonitrile (final concentration about 30%) and the protein is centrifuged off at about 15,000 × g. The thus-stopped samples are analyzed either directly or stored at -20 0 C until analysis.
Die Analyse erfolgt mittels Hochleistungsflüssigkeits-Chromatographie mit Ultraviolett- und massenspektrometrischer Detektion (HPLC-UV-MS/MS). Dazu werden die Überstände der Inkubationsproben mit geeigneten C18-reversed-phase-Säulen und variablen Eluenten-Gemischen aus Acetonitril und 10 mM wässriger Ammoniumformiat-Lösung chromatographiert. Die UV-Chroma- togramme in Verbindung mit massenspektrometrischen MS/MS-Daten dienen zur Identifizierung und Strukturaufklärung der Metabolite. The analysis is carried out by high performance liquid chromatography with ultraviolet and mass spectrometric detection (HPLC-UV-MS / MS). For this, the supernatants of the incubation samples are chromatographed with suitable C18 reversed-phase columns and variable eluent mixtures of acetonitrile and 10 mM aqueous ammonium formate solution. The UV chromatograms in combination with mass spectrometric MS / MS data are used to identify and structure the metabolites.
C. Ausführungsbeispiele für pharmazeutische ZusammensetzungenC. Embodiments of Pharmaceutical Compositions
Die erfindungsgemäßen Verbindungen können folgendermaßen in pharmazeutische Zubereitungen überfuhrt werden:The compounds according to the invention can be converted into pharmaceutical preparations as follows:
Tablette:Tablet:
Zusammensetzung:Composition:
100 mg der erfindungsgemäßen Verbindung, 50 mg Lactose (Monohydrat), 50 mg Maisstärke (nativ), 10 mg Polyvinylpyrrolidon (PVP 25) (Fa. BASF, Ludwigshafen, Deutschland) und 2 mg Magnesiumstearat.100 mg of the compound according to the invention, 50 mg of lactose (monohydrate), 50 mg of corn starch (native), 10 mg of polyvinylpyrrolidone (PVP 25) (BASF, Ludwigshafen, Germany) and 2 mg of magnesium stearate.
Tablettengewicht 212 mg. Durchmesser 8 mm, Wölbungsradius 12 mm.Tablet weight 212 mg. Diameter 8 mm, radius of curvature 12 mm.
Herstellung:production:
Die Mischung aus erfindungsgemäßer Verbindung, Lactose und Stärke wird mit einer 5%-igen Lösung (m/m) des PVPs in Wasser granuliert. Das Granulat wird nach dem Trocknen mit dem Magnesiumstearat 5 Minuten gemischt. Diese Mischung wird mit einer üblichen Tablettenpresse verpresst (Format der Tablette siehe oben). Als Richtwert für die Verpressung wird eine Presskraft von 15 kN verwendet.The mixture of compound of the invention, lactose and starch is granulated with a 5% solution (m / m) of the PVP in water. The granules are mixed after drying with the magnesium stearate for 5 minutes. This mixture is compressed with a conventional tablet press (for the tablet format see above). As a guideline for the compression, a pressing force of 15 kN is used.
Oral applizierbare Suspension:Orally administrable suspension:
Zusammensetzung:Composition:
1000 mg der erfϊndungsgemäßen Verbindung, 1000 mg Ethanol (96%), 400 mg Rhodigel® (Xanthan gum der Firma FMC, Pennsylvania, USA) und 99 g Wasser.1000 mg of the inventive compound, 1000 mg of ethanol (96%), 400 mg of Rhodigel ® (xanthan gum of the firm FMC, Pennsylvania, USA) and 99 g of water.
Einer Einzeldosis von 100 mg der erfindungsgemäßen Verbindung entsprechen 10 ml orale Suspension.A single dose of 100 mg of the compound of the invention corresponds to 10 ml of oral suspension.
Herstellung:production:
Das Rhodigel wird in Ethanol suspendiert, die erfindungsgemäße Verbindung wird der Suspension zugefügt. Unter Rühren erfolgt die Zugabe des Wassers. Bis zum Abschluß der Quellung des Rhodigels wird ca. 6 h gerührt. Oral applizierbare Lösung:The rhodigel is suspended in ethanol, the compound according to the invention is added to the suspension. While stirring, the addition of water. Until the completion of the swelling of Rhodigels is stirred for about 6 h. Orally administrable solution:
Zusammensetzung:Composition:
500 mg der erfϊndungsgemäßen Verbindung, 2.5 g Polysorbat und 97 g Polyethylenglycol 400. Einer Einzeldosis von 100 mg der erfindungsgemäßen Verbindung entsprechen 20 g orale Lösung.500 mg of the compound according to the invention, 2.5 g of polysorbate and 97 g of polyethylene glycol 400. A single dose of 100 mg of the compound according to the invention corresponds to 20 g of oral solution.
Herstellung:production:
Die erfindungsgemäße Verbindung wird in der Mischung aus Polyethylenglycol und Polysorbat unter Rühren suspendiert. Der Rührvorgang wird bis zur vollständigen Auflösung der erfindungsgemäßen Verbindung fortgesetzt.The compound of the invention is suspended in the mixture of polyethylene glycol and polysorbate with stirring. The stirring is continued until complete dissolution of the compound according to the invention.
i.v.-Lösung:iv solution:
Die erfindungsgemäße Verbindung wird in einer Konzentration unterhalb der Sättigungslöslichkeit in einem physiologisch verträglichen Lösungsmittel (z.B. isotonische Kochsalzlösung, Glucose- lösung 5% und/oder PEG 400-Lösung 30%) gelöst. Die Lösung wird steril filtriert und in sterile und pyrogenfreie Injektionsbehältnisse abgefüllt. The compound of the invention is dissolved in a concentration below saturation solubility in a physiologically acceptable solvent (e.g., isotonic saline, glucose solution 5% and / or PEG 400 solution 30%). The solution is sterile filtered and filled into sterile and pyrogen-free injection containers.

Claims

Patentansprüche claims
1. Verbindung der Formel (I)1. Compound of formula (I)
Figure imgf000073_0001
Figure imgf000073_0001
in welcherin which
R1 für Wasserstoff oder (C,-C3)-Alkyl steht,R 1 is hydrogen or (C 1 -C 3 ) -alkyl,
R2 für (CrC6)-Alkyl, (C3-C6)-Alkenyl oder (C3-C7)-Cycloalkyl steht,R 2 is (C r C6) alkyl, (C 3 -C 6) alkenyl or (C 3 -C 7) cycloalkyl,
wobei (Ci-C6)-Alkyl mit 1 oder 2 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor, Trifluormethyl, Hydroxy, (Ci-C4)-Alkoxy, Trifluormethoxy und (C3-C7)-Cycloalkyl substituiert sein kann,where (C 1 -C 6 ) -alkyl having 1 or 2 substituents independently of one another can be substituted from the group of fluorine, trifluoromethyl, hydroxyl, (C 1 -C 4 ) -alkoxy, trifluoromethoxy and (C 3 -C 7 ) -cycloalkyl,
worin (C3-C7)-Cycloalkyl seinerseits mit 1 oder 2 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor, Hydroxy, Oxo, (CrC4)-Alkyl, Trifluormethyl, 2,2,2-Trifluorethyl, (CrC4)-Alkoxy und Trifluormethoxy substitutiert sein kann,wherein (C 3 -C 7 ) -cycloalkyl in turn having 1 or 2 substituents independently selected from the group fluorine, hydroxy, oxo, (C r C 4 ) alkyl, trifluoromethyl, 2,2,2-trifluoroethyl, (C r C 4 ) alkoxy and trifluoromethoxy can be substituted,
undand
wobei (C3-C7)-Cycloalkyl mit 1 oder 2 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor, Hydroxy, Oxo, (Ci-C4)-Alkyl, Trifluormethyl, 2,2,where (C 3 -C 7 ) -cycloalkyl having 1 or 2 substituents independently of one another is selected from the group consisting of fluorine, hydroxyl, oxo, (C 1 -C 4 ) -alkyl, trifluoromethyl, 2,2,
2-Trifluorethyl, (CrC4)-Alkoxy und Trifluormethoxy substitutiert sein kann,2-trifluoroethyl, (C r C 4 ) alkoxy and trifluoromethoxy can be substituted,
undand
wobei in allen genannten Cycloalkyl-Gruppen eine CH2-Einheit gegen Sauerstoff ausgetauscht sein kann,wherein in all said cycloalkyl groups a CH 2 unit may be exchanged for oxygen,
oder R1 und R2 zusammen mit dem Stickstoffatom, an das sie gebunden sind, einen Pyrrolidin- oder Piperidin-Ring bilden, welcher seinerseits mit 1 oder 2 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor, Trifluormethyl, (Ci- C4)-Alkyl, Hydroxy, (Ci-C4)-Alkoxy und Trifluormethoxy substituiert sein kann,or R 1 and R 2 together with the nitrogen atom to which they are attached form a pyrrolidine or piperidine ring, which in turn is independently selected from the group of fluorine, trifluoromethyl, (C 1 -C 4 ) -alkyl by 1 or 2 substituents , Hydroxy, (C 1 -C 4 ) -alkoxy and trifluoromethoxy may be substituted,
R3 für (CrC4)-Alkyl oder Cyclopropyl steht,R 3 is (C r C 4) -alkyl or cyclopropyl,
wobei (Ci-C4)-Alkyl mit 1 bis 3 Substituenten unabhängig voneinander ausgewählt aus der Gruppe Fluor und (CrC4)-Alkoxy substituiert sein kann,wherein (Ci-C 4) -alkyl may be substituted with 1 to 3 substituents independently selected from the group of fluorine and (C r C 4) alkoxy,
R4 für Wasserstoff oder Fluor steht,R 4 is hydrogen or fluorine,
R5 für Wasserstoff, Fluor, Chlor oder Methyl steht,R 5 is hydrogen, fluorine, chlorine or methyl,
R6 für Wasserstoff, Halogen, Nitro, Cyano, Trifluormethyl, Methyl, Ethyl, Trifluormethoxy oder Methoxy steht,R 6 is hydrogen, halogen, nitro, cyano, trifluoromethyl, methyl, ethyl, trifluoromethoxy or methoxy,
R7 für Wasserstoff, Fluor, Chlor oder Methyl steht,R 7 is hydrogen, fluorine, chlorine or methyl,
wobei mindestens einer der Reste R4, R5, R6 und R7 von Wasserstoff verschieden ist,wherein at least one of R 4 , R 5 , R 6 and R 7 is different from hydrogen,
sowie ihre Salze, Solvate und Solvate der Salze.and their salts, solvates and solvates of the salts.
Verbindung der Formel (I) nach Anspruch 1, in welcherA compound of the formula (I) according to claim 1, in which
R1 für Wasserstoff, Methyl oder Ethyl stehtR 1 is hydrogen, methyl or ethyl
R2 für (Ci-C6)-Alkyl, Cyclopropyl, Cyclopentyl oder Cyclohexyl steht,R 2 is (C 1 -C 6 ) -alkyl, cyclopropyl, cyclopentyl or cyclohexyl,
wobei (Ci-C6)-Alkyl mit einem Substituenten ausgewählt aus der Gruppe Fluor, Trifluormethyl, Cyclopropyl, Cyclopentyl und Cyclohexyl substituiert sein kann,where (C 1 -C 6 ) -alkyl may be substituted by a substituent selected from the group consisting of fluorine, trifluoromethyl, cyclopropyl, cyclopentyl and cyclohexyl,
worin Cyclopropyl, Cyclopentyl und Cyclohexyl ihrerseits mit einemwherein cyclopropyl, cyclopentyl and cyclohexyl in turn with a
Substituenten ausgewählt aus der Gruppe Fluor, Methyl, Ethyl und Trifluormethyl substitutiert sein können,Substituents selected from the group fluorine, methyl, ethyl and trifluoromethyl can be substituted,
undand
wobei Cyclopropyl, Cyclopentyl und Cyclohexyl mit einem Substituenten ausgewählt aus der Gruppe Fluor, Methyl, Ethyl und Trifluormethyl substitutiert sein können, oderwhere cyclopropyl, cyclopentyl and cyclohexyl can be substituted by a substituent selected from the group consisting of fluorine, methyl, ethyl and trifluoromethyl, or
R1 und R2 zusammen mit dem Stickstoffatom, an das sie gebunden sind, einen Pyrrolidin- oder Piperidin-Ring bilden, welcher seinerseits mit einem Substituenten ausgewählt aus der Gruppe Fluor, Trifluormethyl, Methyl und Ethyl substituiert sein kann,R 1 and R 2 together with the nitrogen atom to which they are attached form a pyrrolidine or piperidine ring, which in turn may be substituted by a substituent selected from the group consisting of fluorine, trifluoromethyl, methyl and ethyl,
R3 für (CrC4)-Alkyl oder Trifluormethyl steht,R 3 is (C r C 4) -alkyl or trifluoromethyl,
R4 für Wasserstoff steht,R 4 is hydrogen,
R5 für Wasserstoff oder Fluor steht,R 5 is hydrogen or fluorine,
R6 für Wasserstoff, Fluor, Chlor, Trifluormethyl oder Methyl steht,R 6 is hydrogen, fluorine, chlorine, trifluoromethyl or methyl,
R7 für Wasserstoff oder Methyl steht,R 7 is hydrogen or methyl,
wobei mindestens einer der Reste R5, R6 und R7 von Wasserstoff verschieden ist,wherein at least one of R 5 , R 6 and R 7 is different from hydrogen,
sowie ihre Salze, Solvate und Solvate der Salze.and their salts, solvates and solvates of the salts.
3. Verbindung der Formel (I) nach Anspruch 1 oder 2, in welcher3. A compound of the formula (I) according to claim 1 or 2, in which
R1 für Wasserstoff oder Ethyl steht,R 1 is hydrogen or ethyl,
R2 für Ethyl, iso-Propyl, Cyclopropyl oder Cyclopropylmethyl steht,R 2 is ethyl, iso-propyl, cyclopropyl or cyclopropylmethyl,
R3 für Methyl, Ethyl oder iso-Propyl steht,R 3 is methyl, ethyl or iso-propyl,
R4 für Wasserstoff steht,R 4 is hydrogen,
R5 für Wasserstoff steht,R 5 is hydrogen,
R6 für Wasserstoff, Chlor oder Methyl steht,R 6 is hydrogen, chlorine or methyl,
R7 für Wasserstoff oder Methyl steht,R 7 is hydrogen or methyl,
wobei mindestens einer der Reste R6 und R7 von Wasserstoff verschieden ist,wherein at least one of R 6 and R 7 is different from hydrogen,
sowie ihre Salze, Solvate und Solvate der Salze.and their salts, solvates and solvates of the salts.
4. Verbindung der Formel (I) nach Anspruch 1 oder 2, in welcher4. A compound of formula (I) according to claim 1 or 2, in which
R1 für Wasserstoff oder Ethyl steht, R für Ethyl, iso-Propyl, iso-Butyl oder Cyclopropylmethyl steht,R 1 is hydrogen or ethyl, R is ethyl, iso-propyl, iso-butyl or cyclopropylmethyl,
R3 für iso-Butyl steht,R 3 is iso-butyl,
R4 für Wasserstoff steht,R 4 is hydrogen,
R5 für Wasserstoff steht,R 5 is hydrogen,
R6 für Wasserstoff, Chlor oder Methyl steht,R 6 is hydrogen, chlorine or methyl,
R7 für Wasserstoff oder Methyl steht,R 7 is hydrogen or methyl,
wobei mindestens einer der Reste R6 und R7 von Wasserstoff verschieden ist,wherein at least one of R 6 and R 7 is different from hydrogen,
sowie ihre Salze, Solvate und Solvate der Salze.and their salts, solvates and solvates of the salts.
5. Verfahren zur Herstellung von Verbindungen der Formel (I), wie in den Ansprüchen 1 bis 4 definiert, dadurch gekennzeichnet, dass man eine Verbindung der Formel (II)5. A process for the preparation of compounds of the formula (I) as defined in claims 1 to 4, which comprises reacting a compound of the formula (II)
Figure imgf000076_0001
Figure imgf000076_0001
in welcher R3, R4, R5, R6 und R7 jeweils die in den Ansprüchen 1 bis 4 angegebenen Bedeutungen haben,in which R 3 , R 4 , R 5 , R 6 and R 7 each have the meanings given in claims 1 to 4,
undand
R8 für (CrC4)-Alkyl steht,R 8 is (C r C 4 ) -alkyl,
mit Hilfe eines geeigneten Chlorierungsmittels, wie beispielsweise Phosphoroxychlorid, in eine Verbindung der Formel (HI)
Figure imgf000077_0001
with the aid of a suitable chlorinating agent, such as phosphorus oxychloride, in a compound of formula (HI)
Figure imgf000077_0001
in welcher R3, R4, R5, R6, R7 und R8 jeweils die zuvor angegebenen Bedeutungen haben, überfuhrtin which R 3 , R 4 , R 5 , R 6 , R 7 and R 8 each have the meanings given above überfuhrt
und diese anschliessend in einem inerten Lösungsmittel in Gegenwart einer Base mit einer Verbindung der Formel (IV)and this then in an inert solvent in the presence of a base with a compound of formula (IV)
R\R \
N-HN-H
(IV),(IV),
in welcher R1 und R2 jeweils die in den Ansprüchen 1 bis 4 angegebenen Bedeutungen haben,in which R 1 and R 2 each have the meanings given in claims 1 to 4,
zu Verbindungen der Formel (V)to compounds of the formula (V)
Figure imgf000077_0002
Figure imgf000077_0002
in welcher R1, R2, R3, R4, R5, R6, R7 und R8 jeweils die zuvor angegebenen Bedeutungen haben,in which R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 each have the meanings given above,
umsetzt und diese durch basische oder saure Hydrolyse in die Carbonsäuren der Formel (I) and these by basic or acidic hydrolysis into the carboxylic acids of the formula (I)
Figure imgf000078_0001
Figure imgf000078_0001
in welcher R1, R2, R3, R4, R5, R6 und R7 jeweils die in den Ansprüchen 1 bis 4 angegebenen Bedeutungen haben,in which R 1 , R 2 , R 3 , R 4 , R 5 , R 6 and R 7 each have the meanings given in claims 1 to 4,
überführttransferred
und die Verbindungen der Formel (I) gegebenenfalls mit den entsprechenden (i) Lösungsmitteln und/oder (ii) Basen oder Säuren zu ihren Solvaten, Salzen und/oder Solvaten der Salze umsetzt.and optionally reacting the compounds of the formula (I) with the corresponding (i) solvents and / or (ii) bases or acids to their solvates, salts and / or solvates of the salts.
6. Verbindung der Formel (I), wie in einem der Ansprüche 1 bis 4 definiert, zur Behandlung und/oder Prophylaxe von Krankheiten.6. A compound of the formula (I) as defined in any one of claims 1 to 4, for the treatment and / or prophylaxis of diseases.
7. Verbindung der Formel (I), wie in einem der Ansprüche 1 bis 4 definiert, zur Verwendung in einem Verfahren zur Behandlung und/oder Prophylaxe von Dyslipidämien, Arteriosklerose und Herzinsuffizienz.A compound of the formula (I) as defined in any one of claims 1 to 4 for use in a method for the treatment and / or prophylaxis of dyslipidemias, arteriosclerosis and cardiac insufficiency.
8. Verwendung einer Verbindung der Formel (I), wie in einem der Ansprüche 1 bis 4 definiert, zur Herstellung eines Arzneimittels zur Behandlung und/oder Prophylaxe von Dys- lipidämien, Arteriosklerose und Herzinsuffizienz.8. Use of a compound of formula (I) as defined in any one of claims 1 to 4 for the manufacture of a medicament for the treatment and / or prophylaxis of dyslipidaemias, arteriosclerosis and cardiac insufficiency.
9. Arzneimittel enthaltend eine Verbindung der Formel (I), wie in einem der Ansprüche 1 bis 4 definiert, in Kombination mit einem inerten, nicht-toxischen, pharmazeutisch geeigneten Hilfsstoff.A pharmaceutical composition comprising a compound of the formula (I) as defined in any one of claims 1 to 4, in combination with an inert, non-toxic, pharmaceutically acceptable excipient.
10. Arzneimittel enthaltend eine Verbindung der Formel (I), wie in einem der Ansprüche 1 bis 4 definiert, in Kombination mit einem oder mehreren weiteren Wirkstoffen ausgewählt aus der Gruppe bestehend aus HMG-CoA-Reduktase-Inhibitoren, Diuretika, beta-Rezeptoren- Blocker, organische Nitrate und NO-Donatoren, ACE-Inhibitoren, Angiotensin AnAntagonisten, Aldosteron- und Mineralokortikoid-Rezeptor-Antagonisten, Vasopressin- Rezeptor-Antagonisten, Thrombozytenaggregationshemmer sowie Antikoagulantien. 10. A medicament comprising a compound of the formula (I) as defined in any one of claims 1 to 4, in combination with one or more further active compounds selected from the group consisting of HMG-CoA reductase inhibitors, diuretics, beta-receptor Blockers, organic nitrates and NO donors, ACE inhibitors, angiotensin antagonists, aldosterone and mineralocorticoid receptor antagonists, vasopressin receptor antagonists, platelet aggregation inhibitors and anticoagulants.
11. Arzneimittel nach Anspruch 9 oder 10 zur Behandlung und/oder Prophylaxe von Dyslipid- ämien, Arteriosklerose und Herzinsuffizienz.11. Medicament according to claim 9 or 10 for the treatment and / or prophylaxis of dyslipidemia, arteriosclerosis and cardiac insufficiency.
12. Verfahren zur Behandlung und/oder Prophylaxe von Dyslipidämien, Arteriosklerose und Herzinsuffizienz in Menschen und Tieren unter Verwendung einer wirksamen Menge mindestens einer Verbindung der Formel (I), wie in einem der Ansprüche 1 bis 4 definiert, oder eines Arzneimittels, wie in einem der Ansprüche 9 bis 11 definiert. 12. A method for the treatment and / or prophylaxis of dyslipidaemias, arteriosclerosis and cardiac insufficiency in humans and animals using an effective amount of at least one compound of formula (I) as defined in any one of claims 1 to 4, or a drug as in one of claims 9 to 11 defined.
PCT/EP2008/010691 2007-12-20 2008-12-16 Substituted 4-aminopyrimidine-5-carboxylic acid and use thereof WO2009080242A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DE102007061756A DE102007061756A1 (en) 2007-12-20 2007-12-20 Substituted 4-aminopyrimidine-5-carboxylic acids and their use
DE102007061756.0 2007-12-20

Publications (1)

Publication Number Publication Date
WO2009080242A1 true WO2009080242A1 (en) 2009-07-02

Family

ID=40427639

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/010691 WO2009080242A1 (en) 2007-12-20 2008-12-16 Substituted 4-aminopyrimidine-5-carboxylic acid and use thereof

Country Status (2)

Country Link
DE (1) DE102007061756A1 (en)
WO (1) WO2009080242A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100298143A1 (en) * 2009-05-19 2010-11-25 Dow Agrosciences Llc Compounds and methods for controlling fungi
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
CN116496252A (en) * 2022-04-29 2023-07-28 江苏亚虹医药科技股份有限公司 Pyrimidine compound, preparation method and medical application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006097220A1 (en) * 2005-03-12 2006-09-21 Bayer Healthcare Ag Pyrimidine carboxylic acid derivatives and use thereof

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0876379A1 (en) 1996-01-17 1998-11-11 Novo Nordisk A/S Fused 1,2,4-thiadiazine and fused 1,4-thiazine derivatives, their preparation and use
KR20010021936A (en) 1997-07-16 2001-03-15 한센 핀 베네드, 안네 제헤르, 웨이콥 마리안느 Fused 1,2,4-thiadiazine derivatives, their preparation and use
CN1297447A (en) 1998-02-17 2001-05-30 图拉列克股份有限公司 Anti-viral pyrimidine derivs
DE19834047A1 (en) 1998-07-29 2000-02-03 Bayer Ag Substituted pyrazole derivatives
DE19834044A1 (en) 1998-07-29 2000-02-03 Bayer Ag New substituted pyrazole derivatives
DE19943639A1 (en) 1999-09-13 2001-03-15 Bayer Ag Dicarboxylic acid derivatives with novel pharmaceutical properties
DE19943634A1 (en) 1999-09-13 2001-04-12 Bayer Ag Novel dicarboxylic acid derivatives with pharmaceutical properties
DE19943636A1 (en) 1999-09-13 2001-03-15 Bayer Ag Novel dicarboxylic acid derivatives with pharmaceutical properties
DE19943635A1 (en) 1999-09-13 2001-03-15 Bayer Ag Novel aminodicarboxylic acid derivatives with pharmaceutical properties
JP2001089452A (en) 1999-09-22 2001-04-03 Sankyo Co Ltd Pyrimidine derivative
AR031176A1 (en) 2000-11-22 2003-09-10 Bayer Ag NEW DERIVATIVES OF PIRAZOLPIRIDINA SUBSTITUTED WITH PIRIDINE
DE10110749A1 (en) 2001-03-07 2002-09-12 Bayer Ag Substituted aminodicarboxylic acid derivatives
DE10110750A1 (en) 2001-03-07 2002-09-12 Bayer Ag Novel aminodicarboxylic acid derivatives with pharmaceutical properties
GB0127929D0 (en) 2001-11-21 2002-01-16 Celltech R&D Ltd Chemical compounds
DE10220570A1 (en) 2002-05-08 2003-11-20 Bayer Ag Carbamate-substituted pyrazolopyridines
EP1646615B1 (en) 2003-06-06 2009-08-26 Vertex Pharmaceuticals Incorporated Pyrimidine derivatives as modulators of atp-binding cassette transporters
TW200519094A (en) 2003-07-02 2005-06-16 Vertex Pharma Pyrimidines useful as modulators of voltage-gated ion channels
JP2007509126A (en) 2003-10-23 2007-04-12 ファルマシア・コーポレーション Pyrimidine compounds for the treatment of inflammation
AU2005244104B2 (en) 2004-05-08 2012-03-15 Novartis International Pharmaceutical Ltd. 4,5-disubstituted-2-aryl pyrimidines
WO2006124874A2 (en) 2005-05-12 2006-11-23 Kalypsys, Inc. Inhibitors of b-raf kinase

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006097220A1 (en) * 2005-03-12 2006-09-21 Bayer Healthcare Ag Pyrimidine carboxylic acid derivatives and use thereof

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100298143A1 (en) * 2009-05-19 2010-11-25 Dow Agrosciences Llc Compounds and methods for controlling fungi
US8759356B2 (en) * 2009-05-19 2014-06-24 Dow Agrosciences, Llc. Compounds and methods for controlling fungi
AU2010250017B2 (en) * 2009-05-19 2015-02-12 Dow Agrosciences Llc Compounds and methods for controlling fungi
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
CN116496252A (en) * 2022-04-29 2023-07-28 江苏亚虹医药科技股份有限公司 Pyrimidine compound, preparation method and medical application thereof

Also Published As

Publication number Publication date
DE102007061756A1 (en) 2009-06-25

Similar Documents

Publication Publication Date Title
WO2009033561A1 (en) Substituted 6-pheylnicotinic acids and the use thereof
WO2009080248A1 (en) Substituted 2-phenylpyrimidine-5-carboxylic acids and the use thereof
WO2009080242A1 (en) Substituted 4-aminopyrimidine-5-carboxylic acid and use thereof
EP2235011B1 (en) Substituted pyrrolo[2, 3-b]and pyrazolo[3, 4-b]pyridines for use as adenosine receptor ligands
EP2635576B1 (en) Benzyl-substituted carbamates and use thereof
EP3174875A1 (en) Method for the preparation of (4s)-4-(4-cyano-2-methoxyphenyl)-5-ethoxy-2,8-dimethyl-1,4-dihydro-1-6-naphthyridine-3-carbox-amide and the purification thereof for use as an active pharmaceutical ingredient
EP1797045A1 (en) Novel pyrimidine derivatives and their use as ppar-alpha modulators
EP2024361A1 (en) 3-tetrazolyl indazoles, 3-tetrazolyl pyrazolopyridines, and use thereof
EP1866289A1 (en) Pyrimidine carboxylic acid derivatives and use thereof
EP2066634A1 (en) 4-phenoxy-nicotine acid derivatives and use thereof as ppar-modulators
EP3191452A1 (en) Substituted n,2-diarylquinoline-4-carboxamides and the use thereof as anti-inflammatory agents
EP2066635A2 (en) 2-phenoxy nicotine acid derivative and use thereof
EP2556856B1 (en) 6-Cyano-substituted Pyrido[2,3-d]pyrimidines as Adenosin Rezeptor Ligands for the Treatment of Cardiovascular Diseases
EP3292133A1 (en) Process for the preparation of 2-{4-[2-({[2-(4-chlorophenyl)-1,3-thiazol-4-yl]methyl}sulfanyl)-3,5-dicyano-6-(pyrrolidin-1-yl)pyridin-4-yl]phenoxy}ethyl-l-alanyl-l-alaninate monohydrochloride
WO2022224166A1 (en) Compounds as lxr agonists
WO2017153231A1 (en) Substituted n-cyclo-2-aryl-isoquinolinone-4-carboxamides and use thereof
DE102008052013A1 (en) New 4-(4-cyano-2-thioaryl)dihydropyrimidinone compounds are neutrophil elastase inhibitors useful to treat or prevent e.g. pulmonary arterial hypertension, chronic obstructive pulmonary disease, acute lung injury, or cystic fibrosis
WO2020165031A1 (en) Substituted isoquinoline-piperidinylmethanone derivatives
WO2017153235A1 (en) Substituted n-cyclo-3-aryl-1-naphthamides and use thereof
WO2017153234A1 (en) Substituted n-cyclo-2-aryl-quinoline-4-carboxamides and use thereof

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: 08863704

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08863704

Country of ref document: EP

Kind code of ref document: A1