WO2009153261A1 - HYDROXYPHENYL-SUBSTITUTED PYRROLO[2,3d]PYRIMIDINE DERIVATIVES, PROCESSES AND INTERMEDIATE PRODUCTS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THESE COMPOUNDS - Google Patents

HYDROXYPHENYL-SUBSTITUTED PYRROLO[2,3d]PYRIMIDINE DERIVATIVES, PROCESSES AND INTERMEDIATE PRODUCTS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THESE COMPOUNDS Download PDF

Info

Publication number
WO2009153261A1
WO2009153261A1 PCT/EP2009/057473 EP2009057473W WO2009153261A1 WO 2009153261 A1 WO2009153261 A1 WO 2009153261A1 EP 2009057473 W EP2009057473 W EP 2009057473W WO 2009153261 A1 WO2009153261 A1 WO 2009153261A1
Authority
WO
WIPO (PCT)
Prior art keywords
compounds
renal
formula
heart failure
compound
Prior art date
Application number
PCT/EP2009/057473
Other languages
French (fr)
Inventor
Ruben De Kanter
Pieter Spaans
Mayke Hesselink
Marcel Van Aar
Yvan Fischer
Original Assignee
Solvay Pharmaceuticals Gmbh
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 Solvay Pharmaceuticals Gmbh filed Critical Solvay Pharmaceuticals Gmbh
Publication of WO2009153261A1 publication Critical patent/WO2009153261A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention relates to novel hydroxyphenyl-substituted pyrrolo[2,3d] pyrimidine derivatives which have been found to be selective adenosine A 1 receptor antagonists ("AA 1 RAs").
  • the novel compounds described herein are e.g. useful for the prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases.
  • the novel compounds described herein may furthermore be useful in the prophylaxis and/or the treatment of metabolic disorders or diseases.
  • the present invention further relates to medicaments or pharmaceutical compositions containing said novel compounds.
  • the invention relates to processes for the preparation of said novel compounds and to intermediate products for producing them.
  • the invention relates to certain pharmaceutical combination treatments involving the novel compounds of the present invention together with other active agents useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases.
  • Adenosine is an ubiquitous modulator of numerous physiological activities, particularly within the cardiovascular and nervous systems. The effects of extracellular adenosine are mediated by specific cell surface receptor proteins. Adenosine modulates diverse physiological functions including induction of sedation, vasodilation, modulation of cardiac rate and contractility, inhibition of platelet aggregability, stimulation of gluconeo- genesis and inhibition of lipolysis. In addition to its effects on adenylate cyclase, adenosine has been shown to open potassium channels, reduce flux through calcium channels, and inhibit or stimulate phosphoinositide turnover through receptor-mediated mechanisms (See for example, CE. Muller and B.
  • Adenosine receptors belong to the superfamily of purine receptors which are currently subdivided into P 1 (adenosine) and P 2 (ATP, ADP, and other nucleotides) receptors.
  • P 1 adenosine
  • P 2 ATP, ADP, and other nucleotides
  • a 1 and A 2 Two receptor subtypes exhibit affinity for adenosine in the nanomolar range while two other known subtypes A 2b and A 3 are low-affinity receptors, with affinity for adenosine in the low-micromolar range.
  • a 1 and A 3 adenosine receptor activation can lead to an inhibition of adenylate cyclase activity, while A 2a , and A 2 b activation causes a stimulation of adenylate cyclase.
  • the kidney produces adenosine constitutively to regulate glomerular filtration and electrolyte reabsorption via the adenosine A 1 receptor system.
  • Activation of renal adenosine A 1 receptors has two main functional consequences: (i) a stronger reabsorption of sodium and other electrolytes in proximal tubules resulting in a decreased natriuresis, and (ii) a vasoconstriction of afferent arterioles which leads to a reduction of renal blood flow and glomerular filtration rate ("GFR").
  • AA 1 RAs are potentially renoprotective, useful for the treatment of renal failure (including acute renal failure), renal dysfunction, nephritis, hypertension, and edema.
  • Certain compounds which specifically inhibit the adenosine A 1 , A 2a , and A 3 receptors and the use of these compounds to treat certain diseases are e.g. described in published International Patent Application WO 01/39777 (OSI Pharmaceuticals; corresponding e.g. to US 6,800,633).
  • AA 1 RA The prolonged improvement of renal function comprising infrequent administration of an AA 1 RA is disclosed in published International Patent Application WO 2007/149366 (Novacardia). It is also known that adenosine modulates seizures. Seizures and convulsions are the consequence of temporary abnormal electrophysiologic phenomena of the brain, resulting in abnormal synchronization of electrical neuronal activity. Every individual has a seizure threshold, i.e. a tolerance point beyond which a seizure can be induced. For example, individuals who develop seizure disorders have a lower threshold for seizures than others. AA 1 RAs with an ability to cross the blood-brain barrier like e.g.
  • theophylline or caffeine are in principle known to have a certain potential for lowering the seizure threshold and/or to induce seizures.
  • Different approaches have been reported to reduce the potential of AA 1 RAs for lowering seizure thresholds, e.g. by co-administration of an AA 1 RA with anticonvulsants (see e.g. published International Patent Application WO 2007/1 17549) or by providing certain conjugates of AA 1 RAs that would no longer cross the blood-brain barrier (see e.g. published International Patent Application WO 2008/ 024277).
  • AA 1 RAs with a reduced ability to cross the blood-brain barrier can retain the beneficial pharmacological properties in the therapy of cardiovascular, renal and/or hepatic disorders or diseases, but would reduce or eliminate the undesired central nervous system ("CNS") side effects of adenosine A 1 receptor antagonism.
  • CNS central nervous system
  • the invention thus concerns novel hydroxyphenyl-substituted pyr- rolo[2,3d] pyrimidine derivatives of the general Formula I,
  • R 1 is hydrogen or hydroxy, and physiologically acceptable salts, tautomers, isotopically-labelled analogues, solvates and/or esters thereof.
  • the invention concerns medicaments containing the compounds of Formula I; processes for the preparation of the compounds of Formula I and intermediate products of this process; and/or certain pharmaceutical combination treatments involving the novel compounds of the present invention together with other active agents useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases.
  • R 1 preferably has the meaning hydrogen.
  • R 1 is hydrogen
  • a compound according to Formula I is preferred wherein the hydroxy substituent at the phenyl group is in the para-position.
  • Particularly preferred compounds of Formula I are selected from the group consisting of
  • novel compounds of Formula I and their salts, isotopically- labelled analogues, solvates and/or esters are obtained by reacting a compound of general Formula II,
  • R 1 has the above meaning and X stands for a cleavable leaving group, with a compound of general Formula III,
  • R 2 is hydrogen or a protective group for the hydroxy group; and - where R 2 does not represent a desired physiologically acceptable ester - cleaving off the protective group for the hydroxy group R 2 in the resulting compounds, and if desired, converting resulting compounds of formula I into their physiologically acceptable salts, tautomers, isotopically-labelled analogues, solvates and/or esters; or converting salts, isotopically- labelled analogues, solvates and/or esters of the compounds of formula I into the free compounds of formula I.
  • the aforementioned reaction can be carried out using a conventional wet-chemical process in an organic solvent which is inert under the reaction conditions, in particular in a polar protic solvent such as lower alcohols like methanol, ethanol, n-propanol, isopro- panol, n-butanol, tert. butanol, n-pentanol; or in a mixture of such solvents, and in the presence of a base.
  • N-pentanol is a preferred solvent.
  • Suitable bases are e.g. weak in- organic bases like alkali metal carbonates, e.g. sodium carbonate or potassium carbonate. Sodium carbonate is a preferred base.
  • Suitable reaction temperatures are between room temperature and approximately 150 0 C, e.g. between 40 0 C and 140 0 C.
  • Suitable protective groups R 2 are known in the art (see e.g. T. W. Greene, P. G. M. Wuts, "Protective Groups in Organic Synthesis", Wiley Interscience, in its latest edition) and can routinely be selected by a person skilled in the art according to the respective needs.
  • R 2 can e.g. be acetyl or n-propanoyl.
  • R 2 is hydrogen.
  • Suitable physiologically acceptable salts are those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of mammalian subjects, including humans, without undue toxicity, irritation, allergic response, etc., and are commensurate with a reasonable benefit/risk ratio.
  • a number of physiologically acceptable salts are well-known in the art. They can be prepared in situ when finally isolating and purifying the compounds of the invention, or separately by reacting them with physiologically acceptable non-toxic acids, including inorganic or organic acids.
  • Suitable physiologically acceptable salts of compounds of Formula I are e.g.
  • inorganic acids for example sulphuric acid (sulfates), phosphoric acid (phosphates) or hydrohalic acids, preferably hydrochloric acid (hydrochlorides), or with organic acids, for example lower aliphatic carboxylic acids such as citric acid (citrates), or with sulphonic acids, for example lower alkanesulphonic acids such as methanesulphonic acid (mesylates) or benzenesulphonic acid (besylates), optionally substitued in the benzene ring by halogen or lower alkyl, e.g. p-toluenesulphonic acid (tosylates).
  • inorganic acids for example sulphuric acid (sulfates), phosphoric acid (phosphates) or hydrohalic acids, preferably hydrochloric acid (hydrochlorides), or with organic acids, for example lower aliphatic carboxylic acids such as citric acid (citrates), or with sulphonic acids, for example
  • Cleavable leaving groups X in compounds of Formula Il are known in the art and are usually halogen, preferably chlorine or bromine.
  • Compounds of Formula Il are novel compounds which are advantageously suitable as intermediate products for the preparation of novel active substances, for example for the preparation of the pharmacologically active compounds of Formula I.
  • the compounds of Formula Il can be prepared by reacting compounds of the general Formula IV,
  • R 1 has the above meaning, with compounds suitable to exchange a hydroxy group for a cleavable leaving group, usually with a halogenating agent like a phosphorus oxyhalide, preferably phosphorus oxychloride, to exchange the hydroxy group in 4- position of the pyrrolo [2,3d]pyrimidine moiety for a halide.
  • a halogenating agent like a phosphorus oxyhalide, preferably phosphorus oxychloride
  • the reaction can be carried out in known manner using a conventional wet-chemical process in an organic solvent which is inert under the reaction conditions, in particular in a polar aprotic solvent such as dimethylformamide (“DMF”), dimethylsulphoxide (“DMSO”), acetonitrile, acetone, di- chloromethane, sulfur dioxide, and/or hexamethylphosphoramide, or in a mixture of said solvents.
  • a polar aprotic solvent such as dimethylformamide (“DMF"), dimethylsulphoxide (“DMSO”), acetonitrile, acetone, di- chloromethane, sulfur dioxide, and/or hexamethylphosphoramide, or in a mixture of said solvents.
  • DMF dimethylformamide
  • DMSO dimethylsulphoxide
  • acetonitrile acetone
  • di- chloromethane sulfur dioxide
  • sulfur dioxide sulfur dioxide
  • hexamethylphosphoramide sulfur dioxide
  • hexamethylphosphoramide
  • the compounds of Formula IV can be prepared by reacting hydroxybenzamidine compounds of the general Formula V,
  • R 1 has the above meaning, or an acid addition salt of a compound of Formula V, e.g. a hydrohalide thereof, preferably a hydrochloride; with a cyano-acetal compound of general Formula Vl,
  • each of R 3 , R 4 and R 5 independently stands for straight-chain or branched Ci -6 - alkyl, preferably for ethyl, under the conditions of a per se known "Michael condensation".
  • the reaction can be carried out using a conventional wet-chemical process in an organic solvent which is inert under the reaction conditions, in particular in a polar protic solvent such as lower alcohols like methanol, ethanol, n-propanol, isopropanol, n- butanol, tert. butanol, or n-pentanol; or in a mixture of such solvents and in the presence of a base.
  • Ethanol is a preferred solvent.
  • Suitable bases are non-nucleophilic bases e.g. bicyclic amidine bases like 1 ,5-diazabicyclo[3.4.0]non-5-ene ("DBN") or 1 ,8-diazabicyclo- [5.4.0]undecene (“DBU”); or organic nitrogen bases such as tertiary lower alkylamines, e.g. triethyl amine; or mixtures of said non-nucleophilic bases. DBU, triethyl amine and a mixture of DBU and triethyl amine are preferred bases.
  • Suitable reaction temperatures are between room temperature and approximately 150 0 C, e.g. between 40 0 C and 120 0 C, e.g. 90 0 C or 100 0 C.
  • Compounds of Formula V can e.g. be prepared by reacting in a first step a hy- droxybenzonitrile compound of general Formula VIII,
  • R 1 has the above meaning, in an acidic environment and in the presence of a straight-chain or branched Ci -6 -alkanol reagent to produce an intermediate acetimidoal- kylester compound of general Formula VII,
  • the first step of the reaction can be carried out using a conventional wet-chemical process in an organic solvent, in particular in a polar protic solvent such as a lower alcohol like methanol, ethanol, n-propanol, isopropanol, n-butanol, tert.
  • a polar protic solvent such as a lower alcohol like methanol, ethanol, n-propanol, isopropanol, n-butanol, tert.
  • the solvent can also be used as the Ci -6 -alkanol reagent.
  • the acidic environment is usually provided by a hydrohalic acid, preferably by gaseous hydrochloric acid.
  • Suitable reaction temperatures for the first step are between room temperature and approximately 100 0 C, e.g. between room temperature and 80 0 C, e.g. room temperature.
  • the intermediate compound of Formula VII is then isolated, purified, e.g. by washing with a suitable liquid, e.g.
  • Suitable amination reagents in the second step are e.g. ammonia or a salt thereof, e.g. a lower carboxyl salt thereof, and may e.g. be ammonium acetate.
  • Suitable reaction temperatures for the second step are between room temperature and approximately 100 0 C, e.g. between 40 0 C and 80 0 C, e.g. 60 0 C.
  • the cyano-acetal derivatives of Formula Vl are known per se or can be prepared in known manner from known compounds.
  • hydroxybenzonitrile compounds of Formula VIII are known per se or can be prepared in known manner from known compounds.
  • the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters are distinguished by advantageous pharmacological properties.
  • the compounds of Formula I are selective AAiRAs with a high effectiveness, good compatibility and a favourable activity profile.
  • the compounds of Formula I as described herein are therefore suitable for the prophylaxis and/or treatment of adenosine receptor mediated disorders or diseases, in particular for the prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases in mammalian subjects, including humans.
  • the compounds of Formula I as described herein may furthermore be useful for the prophylaxis and/or treatment of metabolic disorders or diseases in mammalian subjects, including humans.
  • the use of the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters in humans is preferred.
  • the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of cardiovascular disorders or diseases, comprising e.g. acute decompensated heart failure; acute heart failure; compensated chronic heart failure; congestive heart failure; decompensated chronic heart failure; forward heart failure; heart failure; high- output heart failure; hypotension on hemodialysis; ischemia reperfusion injury; left-sided heart failure; right-sided heart failure, or combinations of any of the foregoing.
  • cardiovascular disorders or diseases comprising e.g. acute decompensated heart failure; acute heart failure; compensated chronic heart failure; congestive heart failure; decompensated chronic heart failure; forward heart failure; heart failure; high- output heart failure; hypotension on hemodialysis; ischemia reperfusion injury; left-sided heart failure; right-sided heart failure, or combinations of any of the foregoing.
  • the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of hypertension and/or edema, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt, solvate and/or ester.
  • the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of renal disorders or diseases, comprising e.g. acute renal failure; contrast media-induced renal failure; diabetic nephropathy; ischemic renal failure; nephritis; renal disease; renal dysfunction; renal failure; renal hypertension; renal impairment; or combinations of any of the foregoing, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt, solvate and/or ester.
  • renal disorders or diseases comprising e.g. acute renal failure; contrast media-induced renal failure; diabetic nephropathy; ischemic renal failure; nephritis; renal disease; renal dysfunction; renal failure; renal hypertension; renal impairment; or combinations of any of the foregoing, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt
  • the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of hepatic disorders or diseases, comprising e.g. hepatorenal syndrome; liver cirrhosis; liver cirrhosis with ascites; or combinations of any of the foregoing, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt, solvate and/or ester.
  • the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases selected from the group consisting of acute decompensated heart failure; acute heart failure; acute renal failure; compensated chronic heart failure; congestive heart failure; contrast media- induced renal failure; decompensated chronic heart failure; diabetic nephropathy; edema; forward heart failure; heart failure; hepatorenal syndrome; high-output heart failure; hypertension; hypotension on hemodyalisis; ischemia reperfusion injury; ischemic renal failure; left-sided heart failure; liver cirrhosis; liver cirrhosis with ascites; nephritis; renal disease; renal dysfunction; renal failure; renal hypertension; renal impairment; right-sided heart failure, or combinations of any of the foregoing.
  • acute decompensated heart failure selected from the group consisting of acute decompensated heart failure
  • the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of metabolic disorders or diseases, comprising e.g. diabetes mellitus type II, diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic microangiopathy, diabetic macroangiopathy, the metabolic syndrome and/or syndrome X in mammalian subjects, including humans, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt, solvate and/or ester.
  • metabolic disorders or diseases comprising e.g. diabetes mellitus type II, diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic microangiopathy, diabetic macroangiopathy, the metabolic syndrome and/or syndrome X in mammalian subjects, including humans, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its
  • metabolic syndrome as used herein is meant to cover a complex of clinical pictures which mainly comprises central obesity, hypertension, in particular arterial hypertension; insulin resistance, in particular diabetes mellitus type II; glucose intolerance; dyslipoproteinaemia, in particular as hypertriglyceridaemia, accompanied by dysli- poproteinaemia occurring with lowered HDL-cholesterol, and also hyperuricaemia, which can lead to gout.
  • the metabolic syndrome is closely linked to insulin resistance. Some people are genetically predisposed to insulin resistance. Acquired factors, such as excess body fat and physical inactivity, can elicit insulin resistance and the metabolic syndrome in these people. Most people with insulin resistance have central obesity.
  • the biologic mechanisms at the molecular level between insulin resistance and metabolic risk factors are not fully understood and appear to be complex.
  • One group of people at risk for developing metabolic syndrome is those with diabetes who have a defect in insulin action and cannot maintain a proper level of glucose in their blood.
  • a third group is heart attack survivors who, unlike hypertensives, have hyperinsulinemia without having abnormal glucose levels.
  • the metabolic syndrome has become increasingly common in higher developed countries like the United States, where it is estimated that about 20-25 percent of adults have it. There are no well-accepted criteria for diagnosing the metabolic syndrome.
  • Fasting blood triglycerides greater than or equal to 150 mg/dL.
  • the term “syndrome X” is closely related to the term “metabolic syndrome” and usually is supposed to denominate the identical disease or condition. According to information from the American Heart Association, the term “Syndrome X” refers, however, additionally to a heart condition where chest pain and electrocardiographic changes that suggest ischemic heart disease are present, but where there are no angiographic findings of coronary disease. Patients with cardiac syndrome X also sometimes have lipid abnormalities. As used herein "obesity” refers to a condition whereby a person has a Body Mass Index (“BMI”), calculated as weight per height squared (km/m 2 ), of at least 25.9.
  • BMI Body Mass Index
  • the obesity herein may be due to any cause, whether genetic of environmental.
  • disorders that may result in obesity or be the cause of obesity include overeating and bulimia, polycystic ovarian disease, craniopharyngioma, the Prader-Willi syndrome, Frohlich's syndrome, Type-ll diabetes, GH-deficient subjects, normal variant short stature, Turners syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g. children with acute lymphoblastic leukemia.
  • the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters may also be used for the prophylaxis and/or treatment of any cardiovascular, renal, hepatic and/or metabolic disorder or disease mentioned above which may occur as concomitant or secondary disorder or disease in addition to any of the other mentioned cardiovascular, renal, hepatic and/or metabolic disorders or diseases in mammalian subjects, including humans.
  • novel hydroxyphenyl- substituted pyrrolo[2,3d]pyrimidine derivatives as described herein is distinguished by high effectiveness, good compatibility, and a favourable activity profile with a reduced potential for lowering seizure thresholds in mammalian subjects, including humans.
  • Said group of novel hydroxyphenyl-substituted pyrrolo[2,3d]pyrimidine derivatives of Formula I therefore appears suitable for the improved prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases in mammalian subjects, including humans with reduced unwanted side effects.
  • the novel hydroxyphenyl-substituted pyrrolo[2,3d]pyrimidine derivatives as described herein are distinguished by a relatively good solubility in aqueous media.
  • selective and “selectivity” refer to compounds that display reactivity towards a particular receptor (e.g. an adenosine A 1 receptor) without displaying substantial cross-reactivity towards another receptor (e.g. adenosine A 2 receptor).
  • a particular receptor e.g. an adenosine A 1 receptor
  • another receptor e.g. adenosine A 2 receptor
  • selective compounds of the present invention may display reactivity towards adenosine A 1 receptors without displaying substantial cross-reactivity towards other adenosine receptors.
  • treatment refers to any treatment of a mammalian, for example human condition or disease, and includes: (1 ) inhibiting the disease or condition, i.e., arresting its development, (2) relieving the disease or condition, i.e., causing the condition to regress, or (3) stopping the symptoms of the disease.
  • inhibit includes its generally accepted meaning which includes prohibiting, preventing, restraining, alleviating, ameliorating, and slowing, stopping or reversing progression, severity, or a resultant symptom.
  • the present method includes both medical therapeutic and/or prophylactic administration, as appropriate.
  • the term “medical therapy” includes prophylactic, diagnostic and therapeutic regimens carried out in vivo or ex vivo on humans or other mammals.
  • “Mammals” or “mammalian subjects” include animals of economic importance such as bovine, ovine, and porcine animals, especially those that produce meat, as well as domestic animals, sports animals, zoo animals; and humans, the latter being preferred.
  • terapéuticaally effective amount refers to an amount of a therapeutic agent to treat a condition treatable by administrating a composition of the invention. That amount is the amount sufficient to exhibit a detectable therapeutic or ameliorative response in a tissue system, animal or human. The effect may include, for example, treating the conditions listed herein.
  • the precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician (researcher, veterinarian, medical doctor or other clinician), and the therapeutics, or combination of therapeutics, selected for administration. Thus, an exact effective amount may not always be specified in advance.
  • the beneficial pharmacological properties of the compounds of Formula I can e.g. be shown by the following pharmacological test results.
  • the example numbers quoted relate to the preparation examples described below. 1. In-vitro investigation of the receptor binding affinity of the substances
  • the receptor binding affinities of the test substances (compounds of Formula I) to different human adenosine receptors were evaluated by quantifying the displacement of radiolabeled reference agonists from these receptors.
  • the A 1 adenosine receptor was expressed in Chinese hamster ovary cells, and A 2A , A 2B , and A 3 receptors were expressed in HEK293 cells.
  • the specific reference ligands were 3 H-8-cyclopentyl-1 ,3- dipropylxanthine (an adenosine A 1 and A 26 receptor agonist), 3 H-CGS-21680 (an adenosine A 2A agonist), and 4-(amino-3-iodobenzyl)adenosine-5-N-methyl-uronamide (an adenosine A 3 agonist). All test substances were dissolved in DMSO (10 mM) and diluted in a suitable assay buffer to the test concentration. The highest concentration tested was 10 ⁇ M.
  • receptor binding assays were conducted as follows: following incubation of test substance with the receptor preparation and the ligand at the appropriate time and temperature, the receptor preparations were rapidly filtered under vacuum through glass fibre filters; the filters were washed extensively with an ice-cold buffer using a harvester. Bound radioactivity was measured by scintillation counting using a liquid scintillation cocktail. Results were expressed as percentage of total ligand binding and that of nonspecific binding per concentration of test substance (duplicates). From the concentration-displacement curves, IC 50 values were determined by nonlinear regression analysis using Hill equation curve fitting.
  • test substances of examples 1 , 2 and 3 were highly potent antagonists of the human A 1 receptors, and displayed a high selectivity for these receptors as compared to adenosine A 2A , A 26 , and A 3 receptors.
  • test substances compounds of Formula I
  • a functional adenosine A 1 receptor assay namely in the Guinea pig left atrium bioassay.
  • the ability of the compounds is assessed to bind to A 1 receptors and whether this binding would affect the biological function of this receptor.
  • this is done using a biologically relevant, A 1 receptor-mediated response to adenosine, namely a decrease in contraction force (negative inotropic effect) in the Guinea pig atrium. This effect can be prevented by A 1 receptor blockade.
  • Guinea pig left atria were suspended in 20-ml organ baths filled with an oxygenated (95 % O 2 and 5% CO 2 ) and pre-warmed (30 0 C) physiological salt solution of the following composition (in mM): NaCI 118.0, KCI 4.7, MgSO 4 1.2, CaCI 2 2.5, KH 2 PO 4 1.2, NaHCO 3 25.0 and glucose 1 1.0 (pH 7.4).
  • Propranolol (0.1 ⁇ M), cimetidine (1 ⁇ M), atropine (0.1 ⁇ M) and methysergide (0.1 ⁇ M) were also present throughout the experiments to block the beta-adrenergic, histamine H 2 , muscarinic and 5-HT 2 receptors, respectively.
  • the tissues were connected to force transducers for isometric tension recordings. They were stretched to a resting tension of 1 g, then allowed to equilibrate for at least 30 minutes ("min.") during which time they were washed repeatedly and the tension readjusted. Thereafter, they were paced electrically with square wave pulses of maximal intensity and 5 ms duration, applied at a frequency of 2.5 Hz using a constant current stimulator.
  • the experiments were carried out using semi-automated isolated organ systems possessing eight organ baths, with multichannel data acquisition.
  • the tissues were exposed to increasing concentrations of the reference adenosine agonist cyclopentyladenosine ("CPA)", which were added cumulatively by half-log increments to generate control concentration-response curves (CPA produces a negative inotropic response, i.e. a reduction in contraction force in atria).
  • CPA adenosine agonist cyclopentyladenosine
  • the parameter measured was the maximum change in the amplitude of the electrically-evoked contractions induced by each compound concentration. The results are expressed as percentage of the control response.
  • the EC 50 values of CPA concentration producing a half-maximum response
  • the antagonist potencies of the test substances were expressed in terms of pA 2 values (-log concentration producing a twofold shift to the right of the agonist concentration-response curve), which were calculated according to Arunl- akshana and Schild (see Brit. J. Pharmacol. Chemother., 14: 48-58, 1959).
  • the values are given as the mean ⁇ standard error of the mean ("SEM") of three determinations.
  • SEM standard error of the mean
  • B/P brain / plasma exposure
  • test substance of example no. 1 is a p-gp substrate, using an in vitro human p-gp test system.
  • the flux of the test substance of example no. 1 across a monolayer of PK1 cells, expressing p-gp was determined at a start concentration of 1 ug/ml.
  • P-gp is expressed on the Basolateral (B) membrane and therefore the ratio of Apical (A) -> B to B->A is a measure of p-gp activity. This ratio was > 3 for the test substance of example no. 1.
  • P-gp MDR1 orABCBI
  • test substances' pro-convulsive potential is evaluated in a standard pentylenetetrazol (“PTZ") test model.
  • PTZ pentylenetetrazol
  • the dose response for PTZ is established following administration of vehicle and number and types of convulsions observed in the test animals following a 10 min. period are recorded. The percentage of mice showing tonic-extensor convulsions is analyzed and reported.
  • Vehicle treated animals are administered further doses of PTZ, in increasing or decreasing increments, in order to produce a linear response curve.
  • a dose of PTZ which is close to the middle of the linear response curve i.e. that inducing convulsions in around half the animals tested, is selected as the starting dose level of PTZ to be administered to animals receiving the test substances.
  • Test substance treated animals are then administered further doses of PTZ, in increasing or decreasing increments, in order to produce a linear response curve. Selection of the subsequent PTZ dose is dependent on the response displayed by the preceding sub group of animals.
  • test model is suitable to show the favourable activity profile of the test substances, e.g. of the test substance of example no. 1 , in particular their low pro-convulsive potential.
  • test substances can also be demonstrated in other pharmacological test models, e.g. in suitable in vivo animal test models.
  • a pharmaceutical pack or kit comprising one or more container(s) filled with one or more of the ingredients of a pharmaceutical composition of the invention.
  • container(s) filled with one or more of the ingredients of a pharmaceutical composition of the invention.
  • Associated with such containers can be various written materials such as instructions for use, or a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals products, which notice reflects approval by the agency of manufacture, use, or sale for human or veterinary administration.
  • composition encompasses a product comprising specified ingredients in predetermined amounts or proportions, as well as any product that results, directly or indirectly, from combining specified ingredients in specified amounts.
  • this term encompasses a product comprising one or more pharmacologically active ingredients, in particular one or more compounds of Formula I, and additionally conventional pharmaceutically acceptable auxiliaries and/or carriers comprising inert ingredients, as well as any product that results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or with both, and then, if necessary, shaping the product into the desired formulation.
  • the pharma- ceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable auxiliary and/or carrier.
  • the pharmaceutical compositions may be designed to be ready for oral, intravenous ("i.v.”), subcutaneous, tracheal, bronchial, intranasal, pulmonary, transdermal, buccal, rectal and/or parenteral use and/or other ways to administer.
  • the total amount of compounds of Formula I in a pharmaceutical composition suitably is in the range of from about 0.1 % (w/w) to about 95% (w/w) of the pharmaceutical composition, suitably from 0.5% to 50% (w/w) and preferably from 1 % to 25% (w/w).
  • the amount of active ingredient of compound of Formula I is greater than about 95% (w/w) or less than about 0.1 % (w/w).
  • the active ingredients may be separately premixed with the other non-active ingredients, before being mixed to form a formulation.
  • the active ingredients may also be mixed with each other, before being mixed with the non-active ingredients to form a formulation.
  • the compounds of Formula I may be contained according to the invention, together with conventional pharmaceutically acceptable auxiliaries and/or carriers, in solid or liquid pharmaceutical compositions.
  • pharmaceutically acceptable auxiliaries and/or carriers as used herein is meant to comprise solid and liquid auxiliaries, carriers, excipi- ents, adjuvants, diluents etc. which are usual or customary in the art.
  • the auxiliaries and/or carriers must further be compliant or compatible with the other ingredients of the pharmaceutical composition and must not be deleterious to the recipient thereof, i.e. auxiliaries and/or carriers must also be physiologically acceptable when administered to mammalian subjects, including humans.
  • compositions which can be administered orally such as tablets, microtablets, coated tablets, capsules, powders or granules, or alternatively suppositories.
  • These pharmaceutical compositions may contain conventional pharmaceutical inorganic and/or organic excipients, such as talcum, lactose or starch, in addition to conventional pharmaceutical auxiliaries, for example lubricants or tablet disintegrating agents.
  • the active substances may for example be mixed with the auxiliaries and/or excipients in conventional manner and may be wet or dry granulated.
  • the granules or powder may be poured directly into capsules or be pressed into tablet cores in conventional manner. These may be coated in known manner if desired.
  • Liquid pharmaceutical compositions such as solutions, parenteral solutions, suspensions or emulsions of the active substances may contain the usual diluents such as water, oils and/or suspension agents such as polyethylene glycols and the like. Other auxiliaries may additionally be added, such as preservatives, taste correctives and the like.
  • the active substances may be mixed and formulated with the pharmaceutical auxiliaries and/or excipients in known manner.
  • Liquid pharmaceutical compositions may also be prepared in the form of syrups, elixirs, concentrated drops or suspensions, e.g.
  • liquid pharmaceutical composition may contain coloring agents, flavoring agents, preservatives, saccharine and carboxymethyl cellulose or other thickening agents.
  • Liquid pharmaceutical compositions may further be prepared in the form of a dry powder, reconstituted with a suitable solvent prior to use. Solutions for parenteral administration may be prepared as a solution of a formulation of the invention in a pharmaceutically acceptable solvent. These solutions may also contain stabilizing ingredients, preservatives and/or buffering ingredients.
  • Solutions for parenteral administration may also be prepared as a dry preparation, reconstituted with a suitable solvent before use.
  • Liquid pharmaceutical compositions may be filled into suitable administration forms for e.g. i.v. or for oral administration, like e.g. into ampoules, capsules, flasks, infusion bags, or vials.
  • the liquid pharmaceutical compositions may subsequently be administered i.v., e.g. via a syringe, or orally, or may further be processed into solid administration forms like granules or powders.
  • Suitable capsules for use with solid or for liquid pharmaceutical compositions may e.g. be hard gelatine capsules or soft gelatine capsules.
  • Soft gelatin capsules may be prepared with capsules containing a mixture of the active ingredients of the invention, vegetable oil, fat, or other suitable vehicle for soft gelatin capsules.
  • Hard gelatin capsules may contain granules of the active ingredients.
  • Hard gelatin capsules may also contain the active ingredients together with solid powdered ingredients such as lactose, saccharose, sorbitol, mannitol, potato starch, corn starch, amylopectin, cellulose derivatives or gelatin.
  • Dosage units for rectal administration may be prepared (i) in the form of suppositories that contain the active substance mixed with a neutral fat base; (ii) in the form of a gelatin rectal capsule that contains the active substance in a mixture with a vegetable oil, paraffin oil or other suitable vehicle for gelatin rectal capsules; (iii) in the form of a ready- made micro enema; or (iv) in the form of a dry micro enema formulation to be reconstituted in a suitable solvent just prior to administration.
  • Administration forms for transdermal or topical use are e.g. creams or ointments which can be prepared in a manner known per se.
  • composition may be in any form including a tablet, a liquid, a capsule, and a powder or in the form of a food product, e.g. a functional food. In the latter case the food product itself may act as the pharmaceutically acceptable carrier.
  • the compound of Formula I itself or the pharmaceutical composition comprising a compound of Formula I is preferably administered to a patient in need thereof and in a quantity sufficient to prevent and/or treat the symptoms of the cardiovascular, renal, hepatic, and/or metabolic disorder or disease.
  • the administration of a compound of Formula I itself or a pharmaceutical composition comprising a compound of Formula I has a dosage regime which will ultimately be determined by the attending physician and will take into consideration such factors as the compound being used, type of mammalian subject, age, weight, severity of symptoms, method of administration, potential adverse reactions and/or other contraindications. Specific defined dosage ranges can be determined by standard design clinical trials with patient progress and recovery being fully monitored.
  • the compounds of Formula I will normally be administered in a daily dosage regimen (for an adult patient) of, for example, an oral dose of between 1 mg and 2000 mg, preferably between 30 mg and 1000 mg, e.g. between 10 and 250 mg or an intravenous, subcutaneous, or intramuscular dose of between 0.1 mg and 100 mg, preferably between 0.1 mg and 50 mg, e.g. between 1 and 25 mg of the compound of the Formula I or a physiologically acceptable salt thereof calculated as the free base, the compound being usually administered 1 to 4 times per day, e.g. once daily or twice daily.
  • the compounds of Formula I according to the invention can also be administered to children or juveniles while the individual dosage regimens in these cases will need to be particularly thoroughly adjusted by the physician and may usually comprise lower doses than will be administered to adults.
  • pharmaceutical compositions with an active substance content of 0.2 to 500 mg, in particular 10 to 200 mg, compound of Formula I per individual dose are suitable for administration to mammalian subjects, in particular to humans.
  • the compounds of Formula I of the present invention may also be administered by intravenous infusion, at a dose which is likely to range from 0.001-10 mg/kg/hr.
  • the above dosages are exemplary of the average case.
  • hydroxyphenyl-substituted pyrrolo[2,3d] pyrimidine derivatives of Formula I are administered in combination with at least one other active agent useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases.
  • the compounds of Formula I and the other active agents may e.g. be administered as a combination preparation.
  • the term "combination preparation” comprises both true combinations, meaning compounds of Formula I and other active agents physically combined in one preparation such as a tablet or injection fluid; as well as a "kit-of-parts", comprising compounds of Formula I and other active agents useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases in separate dosage forms, together with instructions for use, with or without further means for facilitating compliance with the administration of the component compounds, e.g. labels or drawings.
  • the pharmacotherapy by definition is simultaneous.
  • True combinations comprise fixed combinations of compounds of Formula I and at least one other active agent useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases.
  • kits-of-parts can be administered either simultaneously or at different time intervals.
  • the therapy being either concomitant or sequential, will be dependant on the characteristics of the other medicaments used, characteristics such as onset and duration of action, plasma levels, clearance, etc., as well as on the disease, its stage, and characteristics of the individual patient.
  • pharmacologically active agents useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases which may be suitable for administration in combination with the compounds of Formula I as described herein comprise e.g. the following:
  • ACE-inhibitors e.g. benazepril, captopril, cilazapril, enalapril, fosinopril, imidapril, lisino- pril, moexipril, quinapril, perindopril, ramipril, spirapril or trandolapril;
  • ACE/NEP mixed inhibitors e.g. omapatrilat; adenosine; aldosterone receptor antagonists, e.g. eplerenone; alpha-adrenoceptor antagonists (non-selective), e.g. tolazoline or phenoxybenzamine; alpha-adrenoceptor antagonists (selective), e.g. doxazosin (mesylate), prazosin (hydrochloride and polythiazide), terazosin (hydrochloride) or urapidil; alpha2-adrenoceptor agonists (including centrally acting alpha2-adrenoceptor agonists), e.g.
  • anti-anginal drugs e.g. bepridil, beta blockers, diltiazem, nicardipine, nifedipine, nitrates
  • anticoagulants e.g. dalteparin, danaparoid, enoxaparin, heparin, tinzaparin, warfarin
  • antidiabetics e.g. insulins, amylin, derivatives of GLP-1 and GLP-2 such as, for example, those disclosed in WO 98/08871 and orally active hypoglycemic active ingredients.
  • the orally active hypoglycemic active ingredients preferably comprise sulfonylureas, e.g tolbutamide, glibenclamide, glimepiride, glipizide, gliquidone, gli- soxepide, glibomuride or gliclazide; biguanides, e.g. metformin; meglitinides, e.g. repaglinide; beta3 adrenergic agonists; oxadiazolidinediones; glucosidase inhibitors e.g.
  • sulfonylureas e.g tolbutamide, glibenclamide, glimepiride, glipizide, gliquidone, gli- soxepide, glibomuride or gliclazide
  • biguanides e.g. metformin
  • meglitinides e.g. repaglinide
  • alpha-glucosidase inhibitors such as miglitol or acarbose
  • glucagon receptor antagonists such as GLP-1 agonists, potassium channel openers like diazoxide or those disclosed in WO 97/26265 or WO 99/03861
  • CB-1 (cannabinoid-1 receptor) antagonists/inverse agonists insulin sensitizers like thiazolidinediones, e.g.
  • troglita- zone ciglitazone, pioglitazone, rosiglitazone or the compounds disclosed in WO 97/41097, in particular 5-[[4-[(3,4-dihydro-3-methyl-4-oxo-2- quinazolinylmethoxy]pheny-l]methyl]-2,4-thiazolidinedione; activators of insulin receptor kinase; inhibitors of liver enzymes involved in the stimulation of gluconeo- genesis and/or glycogenosis, for example inhibitors of glycogen phosphorylase; and modulators of glucose uptake and glucose excretion; antiplatelet drugs, e.g.
  • abciximab aspirin, aspirin and dipyridamole, cilostazol, clopido- grel, dipyridamole, eptifibatide, ticlodipine, tirofiban; antiarrhythmic drugs like class I antiarrhythmics, e.g. sodium channel blockers, disopyra- mide, flecainide, lidocaine, mexiletine, moricizine, procainamide, propafenone, quinidine, tocainide; or class Il antiarrhythmics, e.g. beta-adrenoceptor antagonists (see below); or class III antiarrhythmics, e.g.
  • class I antiarrhythmics e.g. sodium channel blockers, disopyra- mide, flecainide, lidocaine, mexiletine, moricizine, procainamide, propafenone, quinidine, tocainide
  • class Il antiarrhythmics e
  • potassium channel blockers like amio- darone, azimilide, bepridil, dofetilide, ibutalide, sotalol, tedisamil; or class IV antiarrhythmics, e.g. calcium channel blockers (see below); ATi receptor antagonists ("sartans”), e.g.
  • ECE-inhbitors e.g. FR-901533; PD-069185; CGS-26303; CGS-34043; CGS-35066; CGS-30084; CGS-35066; SM-19712; Ro0677447;
  • ECE/NEP-mixed inhibitors e.g. daglutril and the compounds disclosed in European Patent Application EP 733642 or in International Patent Application WO 2005/030795; endothelin receptor antagonists (mixed or selective) e.g. atrasentan, bosentan, clazosen- tan, darusentan, sitaxsentan, tezosentan, BMS-193884 or J-104132; ganglion blockers, e.g. reserpine or guanethidine;
  • HMG CoA reductase inhibitors e.g. atorvastatin, berivastatin, cerivastatin, cril- vastatin, fluvastatin, glenvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin, tempostatin or any physiologically compatible salts, solvates, prodrugs or esters thereof; natriuretic peptides, e.g. ANP, nesiritide and/or ularitide; NEP-inhibitors (selective); non-adenosine modifying diuretics, e.g.
  • statins e.g. atorvastatin, berivastatin, cerivastatin, cril- vastatin, fluvastatin, glenvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, sim
  • thiazide diuretics like althiazide, bemetizide, ben- droflumethiazide, benzylhydrochlorothiazide, benzthiazide, buthiazide, chlorothiazide, cyclothiazide, cyclopenthiazide, ethiazide, hydrochlorothiazide, hydroflumethiazide, methylclothiazide, paraflutizide, polythiazide, teclothiazide, trichlormethi- azide or any physiologically compatible tautomers, salts, solvates, prodrugs or esters thereof; thiazide analogues like chloraminofenamide, chlortalidone, clofena- mide, clopamide, clorexolone, fenquizone, indapamide, mefruside, metolazone, quinethazone, tripamide and xipamide; loop di
  • Example I solid pharmaceutical composition of the test substances
  • Capsules with the following composition per capsule are produced: 4-[4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol 20 mg
  • the active substance, the corn starch and the lactose are processed into a homogeneous pasty mixture using ethyl acetate.
  • the paste is ground and the resulting granules are placed on a suitable tray and dried at 45°C in order to remove the solvent.
  • the dried granules are passed through a crusher and mixed in a mixer with the further following auxiliaries:
  • Example II liquid pharmaceutical composition of the test substances
  • a solution suitable for pharmaceutical use is produced with the following amounts per dose:
  • the active substance is weighed on an analytical balance into a beaker, the 0.1 M methanesulfonic acid solution and the 5% dextrose solution are added to the active substance and the mixture is then homogenized by ultrasound treatment for approx. 10 min.
  • the resulting yellow, clear solution (pH approximately 2.3) may then be filled into suitable administration forms.
  • MS Mass spectra
  • HPLC-MS high performance liquid chromatography mass spectra
  • the aqueous layer was further extracted with methyl ethyl ketone (2 x 50 ml_), the combined organic layers were washed with water (200 ml.) brine (100 ml.) and dried over Na 2 SO 4 . The dried combined organic layers were partially concentrated and n-heptane (200 ml.) was added. The precipitated crude product was filtered and washed with n-heptane (20 ml_). After drying in vacuo at 40 0 C, the crude product was re-crystallized from n-butanol/methyl ethyl ketone (1 :1 v/v) and n-heptane.

Abstract

Novel compounds are described of the general formula (I), which are selective antagonists of the adenosine A1 receptor and wherein the substituents R1 have the meanings given in the description,and also medicaments containing these compounds, in particular medicaments suitable for treating or preventing cardio-vascular, renal, and/or hepatic disorders or diseases.

Description

Hydroxyphenyl-Substituted Pyrrolo[2,3d] Pyrimidine Derivatives, Processes and Intermediate Products for Their Preparation and Medicaments Containing These
Compounds
The present invention relates to novel hydroxyphenyl-substituted pyrrolo[2,3d] pyrimidine derivatives which have been found to be selective adenosine A1 receptor antagonists ("AA1RAs"). The novel compounds described herein are e.g. useful for the prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases. The novel compounds described herein may furthermore be useful in the prophylaxis and/or the treatment of metabolic disorders or diseases. The present invention further relates to medicaments or pharmaceutical compositions containing said novel compounds. Furthermore, the invention relates to processes for the preparation of said novel compounds and to intermediate products for producing them. Still further, the invention relates to certain pharmaceutical combination treatments involving the novel compounds of the present invention together with other active agents useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases.
Adenosine is an ubiquitous modulator of numerous physiological activities, particularly within the cardiovascular and nervous systems. The effects of extracellular adenosine are mediated by specific cell surface receptor proteins. Adenosine modulates diverse physiological functions including induction of sedation, vasodilation, modulation of cardiac rate and contractility, inhibition of platelet aggregability, stimulation of gluconeo- genesis and inhibition of lipolysis. In addition to its effects on adenylate cyclase, adenosine has been shown to open potassium channels, reduce flux through calcium channels, and inhibit or stimulate phosphoinositide turnover through receptor-mediated mechanisms (See for example, CE. Muller and B. Stein "Adenosine Receptor Antagonists: Structures and Potential Therapeutic Applications," Current Pharmaceutical Design, 2:50 1 (1996) and CE. Muller "ArAdenosine Receptor Antagonists," Exp. Opin. Then Patents 7 (5):419 (1997).
Adenosine receptors belong to the superfamily of purine receptors which are currently subdivided into P1 (adenosine) and P2 (ATP, ADP, and other nucleotides) receptors. Four adenosine receptor subtypes have been cloned so far from various species including humans. Two receptor subtypes (A1 and A2) exhibit affinity for adenosine in the nanomolar range while two other known subtypes A2b and A3 are low-affinity receptors, with affinity for adenosine in the low-micromolar range. A1 and A3 adenosine receptor activation can lead to an inhibition of adenylate cyclase activity, while A2a, and A2b activation causes a stimulation of adenylate cyclase.
The kidney produces adenosine constitutively to regulate glomerular filtration and electrolyte reabsorption via the adenosine A1 receptor system. Activation of renal adenosine A1 receptors has two main functional consequences: (i) a stronger reabsorption of sodium and other electrolytes in proximal tubules resulting in a decreased natriuresis, and (ii) a vasoconstriction of afferent arterioles which leads to a reduction of renal blood flow and glomerular filtration rate ("GFR"). Accordingly, pharmacological blockade of A1 receptors has been shown to decrease sodium reabsorption, and, thus, to increase natriuresis and diuresis, while preserving renal blood flow and GFR. Thus, AA1RAs are potentially renoprotective, useful for the treatment of renal failure (including acute renal failure), renal dysfunction, nephritis, hypertension, and edema.
Certain deazapurine derivatives which are useful for the treatment of adenosine receptor stimulated diseases are known from published International Patent Application WO 99/62518 (Cadus Pharmaceutical Corp.; corresponding e.g. to US 6,878,716).
Certain compounds which specifically inhibit the adenosine A1, A2a, and A3 receptors and the use of these compounds to treat certain diseases are e.g. described in published International Patent Application WO 01/39777 (OSI Pharmaceuticals; corresponding e.g. to US 6,800,633).
4-(4-trans-hydroxycyclohexyl)amino-2-phenyl-7/-/-pyrrolo[2,3d]pyrimidine hydrogen mesylate and its polymorphic forms are e.g. described in published International Patent Application WO 2004/094428 (Solvay Pharmaceuticals; corresponding e.g. to US 2004- 0248912).
Combinations of certain pyrrolo[2,3d]pyrimidine derivatives which are AA1RAs with certain other pharmacologically active compounds are e.g. described in published International Patent Applications WO 2006/087371 or WO 2007/147809 (Solvay Pharmaceuticals).
The prolonged improvement of renal function comprising infrequent administration of an AA1RA is disclosed in published International Patent Application WO 2007/149366 (Novacardia). It is also known that adenosine modulates seizures. Seizures and convulsions are the consequence of temporary abnormal electrophysiologic phenomena of the brain, resulting in abnormal synchronization of electrical neuronal activity. Every individual has a seizure threshold, i.e. a tolerance point beyond which a seizure can be induced. For example, individuals who develop seizure disorders have a lower threshold for seizures than others. AA1RAs with an ability to cross the blood-brain barrier like e.g. theophylline or caffeine, are in principle known to have a certain potential for lowering the seizure threshold and/or to induce seizures. Different approaches have been reported to reduce the potential of AA1RAs for lowering seizure thresholds, e.g. by co-administration of an AA1RA with anticonvulsants (see e.g. published International Patent Application WO 2007/1 17549) or by providing certain conjugates of AA1RAs that would no longer cross the blood-brain barrier (see e.g. published International Patent Application WO 2008/ 024277). It is assumed that AA1RAs with a reduced ability to cross the blood-brain barrier can retain the beneficial pharmacological properties in the therapy of cardiovascular, renal and/or hepatic disorders or diseases, but would reduce or eliminate the undesired central nervous system ("CNS") side effects of adenosine A1 receptor antagonism.
It was an object of the present invention to provide novel active substances for the treatment of cardiovascular, renal and/or hepatic disorders or diseases which are distinguished by high effectiveness, good compatibility, and a favourable activity profile with a reduced potential for lowering seizure thresholds in mammalian subjects, including humans.
It has now surprisingly been found that a carefully selected group of novel hydroxy- phenyl-substituted pyrrolo[2,3d]pyrimidine derivatives as described herein is distinguished by high effectiveness, good compatibility, and a favourable activity profile with a reduced potential for lowering seizure thresholds in mammalian subjects, including humans. Said group of novel hydroxyphenyl-substituted pyrrolo[2,3d]pyrimidine derivatives therefore appears suitable for the improved prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases in mammalian subjects, including humans.
In one aspect, the invention thus concerns novel hydroxyphenyl-substituted pyr- rolo[2,3d] pyrimidine derivatives of the general Formula I,
Figure imgf000005_0001
wherein
R1 is hydrogen or hydroxy, and physiologically acceptable salts, tautomers, isotopically-labelled analogues, solvates and/or esters thereof.
In other aspects, the invention concerns medicaments containing the compounds of Formula I; processes for the preparation of the compounds of Formula I and intermediate products of this process; and/or certain pharmaceutical combination treatments involving the novel compounds of the present invention together with other active agents useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases.
In the compounds of Formula I, R1 preferably has the meaning hydrogen.
Where R1 is hydrogen, a compound according to Formula I is preferred wherein the hydroxy substituent at the phenyl group is in the para-position.
Compounds of Formula I are preferred wherein the substituents at the cyclohexyl group, namely the hydroxy substituent and the phenyl substituted amino-1 /-/-pyrrolo- [2,3,d]pyrimidin-2-yl substituent, are in the frans-position to each other.
Particularly preferred compounds of Formula I are selected from the group consisting of
4-[4-(4-hydroxy-cyclohexylamino)-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol, in particular 4- [4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol ;
3-[4-(4-hydroxy-cyclohexylamino)-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol, in particular 3- [4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol; and 4-[4-(4-hydroxy-cyclohexylamino)-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl]-benzene-1 ,2-diol, in particular 4-[4-(frans-4-hydroxy-cyclohexylamino)-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl]- benzene-1 ,2-diol; together with their physiologically acceptable salts, tautomers, isotopically-labelled analogues, solvates and/or esters.
In one embodiment, the novel compounds of Formula I and their salts, isotopically- labelled analogues, solvates and/or esters are obtained by reacting a compound of general Formula II,
Figure imgf000006_0001
wherein R1 has the above meaning and X stands for a cleavable leaving group, with a compound of general Formula III,
Figure imgf000006_0002
wherein R2 is hydrogen or a protective group for the hydroxy group; and - where R2 does not represent a desired physiologically acceptable ester - cleaving off the protective group for the hydroxy group R2 in the resulting compounds, and if desired, converting resulting compounds of formula I into their physiologically acceptable salts, tautomers, isotopically-labelled analogues, solvates and/or esters; or converting salts, isotopically- labelled analogues, solvates and/or esters of the compounds of formula I into the free compounds of formula I.
The aforementioned reaction can be carried out using a conventional wet-chemical process in an organic solvent which is inert under the reaction conditions, in particular in a polar protic solvent such as lower alcohols like methanol, ethanol, n-propanol, isopro- panol, n-butanol, tert. butanol, n-pentanol; or in a mixture of such solvents, and in the presence of a base. N-pentanol is a preferred solvent. Suitable bases are e.g. weak in- organic bases like alkali metal carbonates, e.g. sodium carbonate or potassium carbonate. Sodium carbonate is a preferred base. Suitable reaction temperatures are between room temperature and approximately 150 0C, e.g. between 40 0C and 140 0C. Suitable protective groups R2 are known in the art (see e.g. T. W. Greene, P. G. M. Wuts, "Protective Groups in Organic Synthesis", Wiley Interscience, in its latest edition) and can routinely be selected by a person skilled in the art according to the respective needs. In some embodiments, R2 can e.g. be acetyl or n-propanoyl. Preferably R2 is hydrogen.
Suitable physiologically acceptable salts are those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of mammalian subjects, including humans, without undue toxicity, irritation, allergic response, etc., and are commensurate with a reasonable benefit/risk ratio. A number of physiologically acceptable salts are well-known in the art. They can be prepared in situ when finally isolating and purifying the compounds of the invention, or separately by reacting them with physiologically acceptable non-toxic acids, including inorganic or organic acids. Suitable physiologically acceptable salts of compounds of Formula I are e.g. their conventional stoichometric acid addition salts with inorganic acids, for example sulphuric acid (sulfates), phosphoric acid (phosphates) or hydrohalic acids, preferably hydrochloric acid (hydrochlorides), or with organic acids, for example lower aliphatic carboxylic acids such as citric acid (citrates), or with sulphonic acids, for example lower alkanesulphonic acids such as methanesulphonic acid (mesylates) or benzenesulphonic acid (besylates), optionally substitued in the benzene ring by halogen or lower alkyl, e.g. p-toluenesulphonic acid (tosylates).
Cleavable leaving groups X in compounds of Formula Il are known in the art and are usually halogen, preferably chlorine or bromine.
Compounds of Formula Il are novel compounds which are advantageously suitable as intermediate products for the preparation of novel active substances, for example for the preparation of the pharmacologically active compounds of Formula I. The compounds of Formula Il can be prepared by reacting compounds of the general Formula IV,
IV
Figure imgf000007_0001
wherein R1 has the above meaning, with compounds suitable to exchange a hydroxy group for a cleavable leaving group, usually with a halogenating agent like a phosphorus oxyhalide, preferably phosphorus oxychloride, to exchange the hydroxy group in 4- position of the pyrrolo [2,3d]pyrimidine moiety for a halide. The reaction can be carried out in known manner using a conventional wet-chemical process in an organic solvent which is inert under the reaction conditions, in particular in a polar aprotic solvent such as dimethylformamide ("DMF"), dimethylsulphoxide ("DMSO"), acetonitrile, acetone, di- chloromethane, sulfur dioxide, and/or hexamethylphosphoramide, or in a mixture of said solvents. DMF is a preferred solvent. Suitable reaction temperatures are between room temperature and approximately 150 0C, e.g. between 40 0C and 120 0C, e.g. 90 0C or 100 0C.
The compounds of Formula IV can be prepared by reacting hydroxybenzamidine compounds of the general Formula V,
Figure imgf000008_0001
wherein R1 has the above meaning, or an acid addition salt of a compound of Formula V, e.g. a hydrohalide thereof, preferably a hydrochloride; with a cyano-acetal compound of general Formula Vl,
OR3
Figure imgf000008_0002
wherein each of R3, R4 and R5 independently stands for straight-chain or branched Ci-6- alkyl, preferably for ethyl, under the conditions of a per se known "Michael condensation". The reaction can be carried out using a conventional wet-chemical process in an organic solvent which is inert under the reaction conditions, in particular in a polar protic solvent such as lower alcohols like methanol, ethanol, n-propanol, isopropanol, n- butanol, tert. butanol, or n-pentanol; or in a mixture of such solvents and in the presence of a base. Ethanol is a preferred solvent. Suitable bases are non-nucleophilic bases e.g. bicyclic amidine bases like 1 ,5-diazabicyclo[3.4.0]non-5-ene ("DBN") or 1 ,8-diazabicyclo- [5.4.0]undecene ("DBU"); or organic nitrogen bases such as tertiary lower alkylamines, e.g. triethyl amine; or mixtures of said non-nucleophilic bases. DBU, triethyl amine and a mixture of DBU and triethyl amine are preferred bases. Suitable reaction temperatures are between room temperature and approximately 150 0C, e.g. between 40 0C and 120 0C, e.g. 90 0C or 100 0C.
Compounds of Formula V can e.g. be prepared by reacting in a first step a hy- droxybenzonitrile compound of general Formula VIII,
Figure imgf000009_0001
wherein R1 has the above meaning, in an acidic environment and in the presence of a straight-chain or branched Ci-6-alkanol reagent to produce an intermediate acetimidoal- kylester compound of general Formula VII,
Figure imgf000009_0002
or an acid addition salt thereof, e.g. a hydrohalic salt, preferably a hydrochloride salt thereof; wherein R1 has the above meaning and R6 stands for straight-chain or branched Ci-6-alkyl, preferably for ethyl; and reacting in a second step the compound of Formula VII so received with an amination reagent to arrive at compounds of Formula V. The first step of the reaction can be carried out using a conventional wet-chemical process in an organic solvent, in particular in a polar protic solvent such as a lower alcohol like methanol, ethanol, n-propanol, isopropanol, n-butanol, tert. butanol, n-pentanol; or in a mixture of such solvents. The solvent can also be used as the Ci-6-alkanol reagent. The acidic environment is usually provided by a hydrohalic acid, preferably by gaseous hydrochloric acid. Suitable reaction temperatures for the first step are between room temperature and approximately 100 0C, e.g. between room temperature and 80 0C, e.g. room temperature. Usually, the intermediate compound of Formula VII is then isolated, purified, e.g. by washing with a suitable liquid, e.g. with the solvent used in the first step, and resus- pended or resolved, respectively, in an organic solvent which is inert under the reaction conditions, in particular in a polar protic solvent such as a lower alcohol like methanol, ethanol, n-propanol, isopropanol, n-butanol, tert. butanol, n-pentanol; or in a mixture of such solvents. The subsequent second step can be carried out using a conventional wet- chemical process in the organic solvent as mentioned here before. Suitable amination reagents in the second step are e.g. ammonia or a salt thereof, e.g. a lower carboxyl salt thereof, and may e.g. be ammonium acetate. Suitable reaction temperatures for the second step are between room temperature and approximately 100 0C, e.g. between 40 0C and 80 0C, e.g. 60 0C.
The cyano-acetal derivatives of Formula Vl are known per se or can be prepared in known manner from known compounds.
The hydroxybenzonitrile compounds of Formula VIII are known per se or can be prepared in known manner from known compounds.
The compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters are distinguished by advantageous pharmacological properties. In particular the compounds of Formula I are selective AAiRAs with a high effectiveness, good compatibility and a favourable activity profile. The compounds of Formula I as described herein are therefore suitable for the prophylaxis and/or treatment of adenosine receptor mediated disorders or diseases, in particular for the prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases in mammalian subjects, including humans. The compounds of Formula I as described herein may furthermore be useful for the prophylaxis and/or treatment of metabolic disorders or diseases in mammalian subjects, including humans. The use of the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters in humans is preferred.
In one embodiment, the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of cardiovascular disorders or diseases, comprising e.g. acute decompensated heart failure; acute heart failure; compensated chronic heart failure; congestive heart failure; decompensated chronic heart failure; forward heart failure; heart failure; high- output heart failure; hypotension on hemodialysis; ischemia reperfusion injury; left-sided heart failure; right-sided heart failure, or combinations of any of the foregoing. In another variant of this embodiment, the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of hypertension and/or edema, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt, solvate and/or ester.
In another embodiment, the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of renal disorders or diseases, comprising e.g. acute renal failure; contrast media-induced renal failure; diabetic nephropathy; ischemic renal failure; nephritis; renal disease; renal dysfunction; renal failure; renal hypertension; renal impairment; or combinations of any of the foregoing, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt, solvate and/or ester.
In yet another embodiment the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of hepatic disorders or diseases, comprising e.g. hepatorenal syndrome; liver cirrhosis; liver cirrhosis with ascites; or combinations of any of the foregoing, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt, solvate and/or ester.
In one embodiment, the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases selected from the group consisting of acute decompensated heart failure; acute heart failure; acute renal failure; compensated chronic heart failure; congestive heart failure; contrast media- induced renal failure; decompensated chronic heart failure; diabetic nephropathy; edema; forward heart failure; heart failure; hepatorenal syndrome; high-output heart failure; hypertension; hypotension on hemodyalisis; ischemia reperfusion injury; ischemic renal failure; left-sided heart failure; liver cirrhosis; liver cirrhosis with ascites; nephritis; renal disease; renal dysfunction; renal failure; renal hypertension; renal impairment; right-sided heart failure, or combinations of any of the foregoing.
In another embodiment, the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters can be used for the prophylaxis and/or treatment of metabolic disorders or diseases, comprising e.g. diabetes mellitus type II, diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic microangiopathy, diabetic macroangiopathy, the metabolic syndrome and/or syndrome X in mammalian subjects, including humans, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I or its physiologically acceptable salt, solvate and/or ester.
The term "metabolic syndrome" as used herein is meant to cover a complex of clinical pictures which mainly comprises central obesity, hypertension, in particular arterial hypertension; insulin resistance, in particular diabetes mellitus type II; glucose intolerance; dyslipoproteinaemia, in particular as hypertriglyceridaemia, accompanied by dysli- poproteinaemia occurring with lowered HDL-cholesterol, and also hyperuricaemia, which can lead to gout. According to information from the American Heart Association, the metabolic syndrome is closely linked to insulin resistance. Some people are genetically predisposed to insulin resistance. Acquired factors, such as excess body fat and physical inactivity, can elicit insulin resistance and the metabolic syndrome in these people. Most people with insulin resistance have central obesity. The biologic mechanisms at the molecular level between insulin resistance and metabolic risk factors are not fully understood and appear to be complex. One group of people at risk for developing metabolic syndrome is those with diabetes who have a defect in insulin action and cannot maintain a proper level of glucose in their blood. Another is people, mainly those with high blood pressure, who are nondiabetic and insulin-resistant but who compensate by secreting large amounts of insulin. This condition is known as hyperinsulinemia. A third group is heart attack survivors who, unlike hypertensives, have hyperinsulinemia without having abnormal glucose levels. The metabolic syndrome has become increasingly common in higher developed countries like the United States, where it is estimated that about 20-25 percent of adults have it. There are no well-accepted criteria for diagnosing the metabolic syndrome. The criteria proposed by the Third Report of the National Cholesterol Education Program ("NCEP") Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III, "ATP III") are the most current and widely used. According to the ATP III criteria, the metabolic syndrome is identified by the presence of three or more of these components:
Central obesity as measured by waist circumference (Men - Greater than 40 inches; Women - Greater than 35 inches).
Fasting blood triglycerides greater than or equal to 150 mg/dL.
Blood HDL cholesterol (Men - Less than 40 mg/dL; Women - Less than 50 mg/dL)
Blood pressure greater than or equal to 130/85 mmHg.
Fasting glucose greater than or equal to 110 mg/dL. The term "syndrome X" is closely related to the term "metabolic syndrome" and usually is supposed to denominate the identical disease or condition. According to information from the American Heart Association, the term "Syndrome X" refers, however, additionally to a heart condition where chest pain and electrocardiographic changes that suggest ischemic heart disease are present, but where there are no angiographic findings of coronary disease. Patients with cardiac syndrome X also sometimes have lipid abnormalities. As used herein "obesity" refers to a condition whereby a person has a Body Mass Index ("BMI"), calculated as weight per height squared (km/m2), of at least 25.9. Conventionally, those persons with normal weight have a BMI of 19.9 to less than 25.9. The obesity herein may be due to any cause, whether genetic of environmental. Examples of disorders that may result in obesity or be the cause of obesity include overeating and bulimia, polycystic ovarian disease, craniopharyngioma, the Prader-Willi syndrome, Frohlich's syndrome, Type-ll diabetes, GH-deficient subjects, normal variant short stature, Turners syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g. children with acute lymphoblastic leukemia.
In a further embodiment, the compounds of Formula I and their physiologically acceptable salts, tautomers, solvates and/or esters may also be used for the prophylaxis and/or treatment of any cardiovascular, renal, hepatic and/or metabolic disorder or disease mentioned above which may occur as concomitant or secondary disorder or disease in addition to any of the other mentioned cardiovascular, renal, hepatic and/or metabolic disorders or diseases in mammalian subjects, including humans.
What is striking is that a carefully selected group of novel hydroxyphenyl- substituted pyrrolo[2,3d]pyrimidine derivatives as described herein is distinguished by high effectiveness, good compatibility, and a favourable activity profile with a reduced potential for lowering seizure thresholds in mammalian subjects, including humans. Said group of novel hydroxyphenyl-substituted pyrrolo[2,3d]pyrimidine derivatives of Formula I therefore appears suitable for the improved prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases in mammalian subjects, including humans with reduced unwanted side effects. Furthermore, the novel hydroxyphenyl-substituted pyrrolo[2,3d]pyrimidine derivatives as described herein are distinguished by a relatively good solubility in aqueous media.
The terms "selective" and "selectivity" refer to compounds that display reactivity towards a particular receptor (e.g. an adenosine A1 receptor) without displaying substantial cross-reactivity towards another receptor (e.g. adenosine A2 receptor). Thus, for exam- pie, selective compounds of the present invention may display reactivity towards adenosine A1 receptors without displaying substantial cross-reactivity towards other adenosine receptors.
The term "treatment" as used herein refers to any treatment of a mammalian, for example human condition or disease, and includes: (1 ) inhibiting the disease or condition, i.e., arresting its development, (2) relieving the disease or condition, i.e., causing the condition to regress, or (3) stopping the symptoms of the disease. The term "inhibit" includes its generally accepted meaning which includes prohibiting, preventing, restraining, alleviating, ameliorating, and slowing, stopping or reversing progression, severity, or a resultant symptom. As such, the present method includes both medical therapeutic and/or prophylactic administration, as appropriate. As used herein, the term "medical therapy" includes prophylactic, diagnostic and therapeutic regimens carried out in vivo or ex vivo on humans or other mammals. "Mammals" or "mammalian subjects" include animals of economic importance such as bovine, ovine, and porcine animals, especially those that produce meat, as well as domestic animals, sports animals, zoo animals; and humans, the latter being preferred.
Throughout the description and the claims of this specification the word "comprise" and variations of the word, such as "comprising" and "comprises" is not intended to exclude other additives, components, integers or steps.
The term "therapeutically effective amount" as used herein refers to an amount of a therapeutic agent to treat a condition treatable by administrating a composition of the invention. That amount is the amount sufficient to exhibit a detectable therapeutic or ameliorative response in a tissue system, animal or human. The effect may include, for example, treating the conditions listed herein. The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician (researcher, veterinarian, medical doctor or other clinician), and the therapeutics, or combination of therapeutics, selected for administration. Thus, an exact effective amount may not always be specified in advance.
Description of the pharmacological test methods
The beneficial pharmacological properties of the compounds of Formula I can e.g. be shown by the following pharmacological test results. The example numbers quoted relate to the preparation examples described below. 1. In-vitro investigation of the receptor binding affinity of the substances
The receptor binding affinities of the test substances (compounds of Formula I) to different human adenosine receptors were evaluated by quantifying the displacement of radiolabeled reference agonists from these receptors. The A1 adenosine receptor was expressed in Chinese hamster ovary cells, and A2A, A2B, and A3 receptors were expressed in HEK293 cells. The specific reference ligands were 3H-8-cyclopentyl-1 ,3- dipropylxanthine (an adenosine A1 and A26 receptor agonist), 3H-CGS-21680 (an adenosine A2A agonist), and 4-(amino-3-iodobenzyl)adenosine-5-N-methyl-uronamide (an adenosine A3 agonist). All test substances were dissolved in DMSO (10 mM) and diluted in a suitable assay buffer to the test concentration. The highest concentration tested was 10 μM. If the ligand displacement by compounds was > 50% at this concentration, further testing was carried out in a 3 log concentration range around a predetermined IC5O for the respective assay. All determinations were performed as duplicates. For test substances found to be active at 10 μM, the subsequent IC50 (and pKi) determination was repeated three times (i.e. 3 duplicates for each concentration, unless indicated otherwise).
In brief, receptor binding assays were conducted as follows: following incubation of test substance with the receptor preparation and the ligand at the appropriate time and temperature, the receptor preparations were rapidly filtered under vacuum through glass fibre filters; the filters were washed extensively with an ice-cold buffer using a harvester. Bound radioactivity was measured by scintillation counting using a liquid scintillation cocktail. Results were expressed as percentage of total ligand binding and that of nonspecific binding per concentration of test substance (duplicates). From the concentration-displacement curves, IC50 values were determined by nonlinear regression analysis using Hill equation curve fitting. The inhibition constants (Ki) were calculated from the Cheng-Prusoff equation Ki = IC50(1 +L/Kd)"1, where L is the concentration of radioligand in the assay and Kd the affinity of the radioligand for the receptor. The results were expressed as mean pKi values ± s.d. (standard deviation) of at least two separate experiments. Where no significant affinity was found at concentrations of 10 μM, test substances were concluded to be inactive as denoted by pKi of < 5. Table 1 : Binding affinity of the test substances at human adenosine receptor subtypes in vitro
Figure imgf000016_0001
test repeated only twice ,.; < (**) ' only 1 value in duplicate
As can be seen from Table 1 , the test substances of examples 1 , 2 and 3 were highly potent antagonists of the human A1 receptors, and displayed a high selectivity for these receptors as compared to adenosine A2A, A26, and A3 receptors.
2. Functional in vitro investigation of the substances in tissue from the Guinea pig
The effects of test substances (compounds of Formula I) were investigated in a functional adenosine A1 receptor assay, namely in the Guinea pig left atrium bioassay. In this assay, the ability of the compounds is assessed to bind to A1 receptors and whether this binding would affect the biological function of this receptor. In the present assay, this is done using a biologically relevant, A1 receptor-mediated response to adenosine, namely a decrease in contraction force (negative inotropic effect) in the Guinea pig atrium. This effect can be prevented by A1 receptor blockade.
Guinea pig left atria were suspended in 20-ml organ baths filled with an oxygenated (95 % O2 and 5% CO2) and pre-warmed (300C) physiological salt solution of the following composition (in mM): NaCI 118.0, KCI 4.7, MgSO4 1.2, CaCI2 2.5, KH2PO4 1.2, NaHCO3 25.0 and glucose 1 1.0 (pH 7.4). Propranolol (0.1 μM), cimetidine (1 μM), atropine (0.1 μM) and methysergide (0.1 μM) were also present throughout the experiments to block the beta-adrenergic, histamine H2, muscarinic and 5-HT2 receptors, respectively. The tissues were connected to force transducers for isometric tension recordings. They were stretched to a resting tension of 1 g, then allowed to equilibrate for at least 30 minutes ("min.") during which time they were washed repeatedly and the tension readjusted. Thereafter, they were paced electrically with square wave pulses of maximal intensity and 5 ms duration, applied at a frequency of 2.5 Hz using a constant current stimulator. The experiments were carried out using semi-automated isolated organ systems possessing eight organ baths, with multichannel data acquisition.
The tissues were exposed to increasing concentrations of the reference adenosine agonist cyclopentyladenosine ("CPA)", which were added cumulatively by half-log increments to generate control concentration-response curves (CPA produces a negative inotropic response, i.e. a reduction in contraction force in atria). Following extensive washings and a subsequent equilibration period, the concentration-response curves to CPA were reproduced in the absence (control tissues) or presence of the test substances (test tissues) which were added at a fixed concentration 30 min. prior to the second addition of CPA. Only one concentration of the test compounds was investigated in each atrium. Each test substance was investigated at three concentrations, each on three tissues (n=3), to evaluate its antagonist potency against CPA.
The parameter measured was the maximum change in the amplitude of the electrically-evoked contractions induced by each compound concentration. The results are expressed as percentage of the control response. The EC50 values of CPA (concentration producing a half-maximum response) were calculated by linear regression analysis of the concentration-response curves. The antagonist potencies of the test substances were expressed in terms of pA2 values (-log concentration producing a twofold shift to the right of the agonist concentration-response curve), which were calculated according to Arunl- akshana and Schild (see Brit. J. Pharmacol. Chemother., 14: 48-58, 1959). The values are given as the mean ± standard error of the mean ("SEM") of three determinations. Statistical significance of the differences was determined using Student's t-test for paired and unpaired data, and p values < 0.05 were considered significant.
Table 2: Effects of the test compounds in the functional adenosine A1 guinea pig left atrium bioassay
Figure imgf000017_0001
3. In vivo investigation of the test substances' brain penetration potential in the rat
The study was carried out using one treatment group of 12 male Wistar rats. During the study, the animals had free access to food and tap water. The animals were dosed i.v., 1 mg/kg. The dose volume was 2 mL/kg of a liquid formulation (aqueous surfac- tant/co-solvent solution) of the test compound of example no. 1. At each of the time- points, 10 min., 0.5, 1 , 3, 7 and 24 hrs after dosing, 2 animals were sacrificed by exsan- guination and blood and brain samples were harvested. Using this procedure, there remains only neglectable amount of blood in the brain. The concentration of the test substance of example no. 1 was determined in the plasma and brain homogenates. Thereafter, the brain / plasma exposure ("B/P") ratio was determined as the ratio between the AUCo-~ for plasma and AUCo-~ for brain and found to be 0.03. This finding suggests that there is limited potential for any adverse events due to pharmacological activity of the test substances in the brain.
The low brain penetration was confirmed by the finding that the test substance of example no. 1 is a p-gp substrate, using an in vitro human p-gp test system. In this study the flux of the test substance of example no. 1 across a monolayer of PK1 cells, expressing p-gp, was determined at a start concentration of 1 ug/ml. P-gp is expressed on the Basolateral (B) membrane and therefore the ratio of Apical (A) -> B to B->A is a measure of p-gp activity. This ratio was > 3 for the test substance of example no. 1. P-gp (MDR1 orABCBI ) is known to be expressed on the blood brain barrier where it actively transports substrates from the cerebrospinal fluid back into the blood, thus limiting brain exposure.
4. In vivo investigation of the test substances' pro-convulsive potential in mice
The test substances' pro-convulsive potential is evaluated in a standard pentylenetetrazol ("PTZ") test model. This test model is indicative for the potential of the test compounds to induce seizures in mammalian subjects, including humans.
The test is performed in mice using four treatment groups, (n= 10 mice per group ), one group for vehicle control and three groups for different dose levels of test substance. The dose response for PTZ is established following administration of vehicle and number and types of convulsions observed in the test animals following a 10 min. period are recorded. The percentage of mice showing tonic-extensor convulsions is analyzed and reported. Vehicle treated animals are administered further doses of PTZ, in increasing or decreasing increments, in order to produce a linear response curve. A dose of PTZ which is close to the middle of the linear response curve i.e. that inducing convulsions in around half the animals tested, is selected as the starting dose level of PTZ to be administered to animals receiving the test substances. Test substance treated animals are then administered further doses of PTZ, in increasing or decreasing increments, in order to produce a linear response curve. Selection of the subsequent PTZ dose is dependent on the response displayed by the preceding sub group of animals.
This test model is suitable to show the favourable activity profile of the test substances, e.g. of the test substance of example no. 1 , in particular their low pro-convulsive potential.
The favourable action profile and/or the beneficial pharmacological properties of the test substances can also be demonstrated in other pharmacological test models, e.g. in suitable in vivo animal test models.
The compounds of the present invention and their physiologically acceptable salts, tautomers, solvates and/or esters may be administered as pharmaceutical compositions which are important and novel embodiments of the invention because of the presence of the compounds disclosed herein. In embodiments of the invention, a pharmaceutical pack or kit is provided comprising one or more container(s) filled with one or more of the ingredients of a pharmaceutical composition of the invention. Associated with such containers) can be various written materials such as instructions for use, or a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals products, which notice reflects approval by the agency of manufacture, use, or sale for human or veterinary administration.
The term "composition" as used herein encompasses a product comprising specified ingredients in predetermined amounts or proportions, as well as any product that results, directly or indirectly, from combining specified ingredients in specified amounts. In relation to pharmaceutical compositions, this term encompasses a product comprising one or more pharmacologically active ingredients, in particular one or more compounds of Formula I, and additionally conventional pharmaceutically acceptable auxiliaries and/or carriers comprising inert ingredients, as well as any product that results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. In general, pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or with both, and then, if necessary, shaping the product into the desired formulation. Accordingly, the pharma- ceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable auxiliary and/or carrier.
The pharmaceutical compositions may be designed to be ready for oral, intravenous ("i.v."), subcutaneous, tracheal, bronchial, intranasal, pulmonary, transdermal, buccal, rectal and/or parenteral use and/or other ways to administer. The total amount of compounds of Formula I in a pharmaceutical composition suitably is in the range of from about 0.1 % (w/w) to about 95% (w/w) of the pharmaceutical composition, suitably from 0.5% to 50% (w/w) and preferably from 1 % to 25% (w/w). In some embodiments, the amount of active ingredient of compound of Formula I is greater than about 95% (w/w) or less than about 0.1 % (w/w). The active ingredients may be separately premixed with the other non-active ingredients, before being mixed to form a formulation. The active ingredients may also be mixed with each other, before being mixed with the non-active ingredients to form a formulation.
The compounds of Formula I may be contained according to the invention, together with conventional pharmaceutically acceptable auxiliaries and/or carriers, in solid or liquid pharmaceutical compositions. The term "pharmaceutically acceptable auxiliaries and/or carriers" as used herein is meant to comprise solid and liquid auxiliaries, carriers, excipi- ents, adjuvants, diluents etc. which are usual or customary in the art. The auxiliaries and/or carriers must further be compliant or compatible with the other ingredients of the pharmaceutical composition and must not be deleterious to the recipient thereof, i.e. auxiliaries and/or carriers must also be physiologically acceptable when administered to mammalian subjects, including humans.
Examples of solid pharmaceutical compositions are compositions which can be administered orally, such as tablets, microtablets, coated tablets, capsules, powders or granules, or alternatively suppositories. These pharmaceutical compositions may contain conventional pharmaceutical inorganic and/or organic excipients, such as talcum, lactose or starch, in addition to conventional pharmaceutical auxiliaries, for example lubricants or tablet disintegrating agents. For the preparation of solid pharmaceutical compositions, the active substances may for example be mixed with the auxiliaries and/or excipients in conventional manner and may be wet or dry granulated. The granules or powder may be poured directly into capsules or be pressed into tablet cores in conventional manner. These may be coated in known manner if desired. Liquid pharmaceutical compositions such as solutions, parenteral solutions, suspensions or emulsions of the active substances may contain the usual diluents such as water, oils and/or suspension agents such as polyethylene glycols and the like. Other auxiliaries may additionally be added, such as preservatives, taste correctives and the like. The active substances may be mixed and formulated with the pharmaceutical auxiliaries and/or excipients in known manner. Liquid pharmaceutical compositions may also be prepared in the form of syrups, elixirs, concentrated drops or suspensions, e.g. solutions or suspensions containing the active ingredients and the remainder consisting, for example, of sugar or sugar alcohols and a mixture of ethanol, water, glycerol, propylene glycol and polyethylene glycol. If desired, such liquid pharmaceutical composition may contain coloring agents, flavoring agents, preservatives, saccharine and carboxymethyl cellulose or other thickening agents. Liquid pharmaceutical compositions may further be prepared in the form of a dry powder, reconstituted with a suitable solvent prior to use. Solutions for parenteral administration may be prepared as a solution of a formulation of the invention in a pharmaceutically acceptable solvent. These solutions may also contain stabilizing ingredients, preservatives and/or buffering ingredients. Solutions for parenteral administration may also be prepared as a dry preparation, reconstituted with a suitable solvent before use. Liquid pharmaceutical compositions may be filled into suitable administration forms for e.g. i.v. or for oral administration, like e.g. into ampoules, capsules, flasks, infusion bags, or vials. The liquid pharmaceutical compositions may subsequently be administered i.v., e.g. via a syringe, or orally, or may further be processed into solid administration forms like granules or powders.
Suitable capsules for use with solid or for liquid pharmaceutical compositions may e.g. be hard gelatine capsules or soft gelatine capsules. Soft gelatin capsules may be prepared with capsules containing a mixture of the active ingredients of the invention, vegetable oil, fat, or other suitable vehicle for soft gelatin capsules. Hard gelatin capsules may contain granules of the active ingredients. Hard gelatin capsules may also contain the active ingredients together with solid powdered ingredients such as lactose, saccharose, sorbitol, mannitol, potato starch, corn starch, amylopectin, cellulose derivatives or gelatin.
Dosage units for rectal administration may be prepared (i) in the form of suppositories that contain the active substance mixed with a neutral fat base; (ii) in the form of a gelatin rectal capsule that contains the active substance in a mixture with a vegetable oil, paraffin oil or other suitable vehicle for gelatin rectal capsules; (iii) in the form of a ready- made micro enema; or (iv) in the form of a dry micro enema formulation to be reconstituted in a suitable solvent just prior to administration.
Administration forms for transdermal or topical use are e.g. creams or ointments which can be prepared in a manner known per se.
Yet a further aspect of the invention provides a process for the manufacture of a pharmaceutical composition as described here above. The manufacture can be carried out by standard techniques well known in the art and involves combining a compound according to the invention and the pharmaceutically acceptable auxiliaries and/or carriers. The composition may be in any form including a tablet, a liquid, a capsule, and a powder or in the form of a food product, e.g. a functional food. In the latter case the food product itself may act as the pharmaceutically acceptable carrier.
The compound of Formula I itself or the pharmaceutical composition comprising a compound of Formula I is preferably administered to a patient in need thereof and in a quantity sufficient to prevent and/or treat the symptoms of the cardiovascular, renal, hepatic, and/or metabolic disorder or disease. For all aspects of the invention, particularly medical ones, the administration of a compound of Formula I itself or a pharmaceutical composition comprising a compound of Formula I has a dosage regime which will ultimately be determined by the attending physician and will take into consideration such factors as the compound being used, type of mammalian subject, age, weight, severity of symptoms, method of administration, potential adverse reactions and/or other contraindications. Specific defined dosage ranges can be determined by standard design clinical trials with patient progress and recovery being fully monitored. Such trials may use an escalating dose design using a low percentage of the maximum tolerated dose in animals as the starting dose in man. The compounds of Formula I will normally be administered in a daily dosage regimen (for an adult patient) of, for example, an oral dose of between 1 mg and 2000 mg, preferably between 30 mg and 1000 mg, e.g. between 10 and 250 mg or an intravenous, subcutaneous, or intramuscular dose of between 0.1 mg and 100 mg, preferably between 0.1 mg and 50 mg, e.g. between 1 and 25 mg of the compound of the Formula I or a physiologically acceptable salt thereof calculated as the free base, the compound being usually administered 1 to 4 times per day, e.g. once daily or twice daily. The compounds of Formula I according to the invention can also be administered to children or juveniles while the individual dosage regimens in these cases will need to be particularly thoroughly adjusted by the physician and may usually comprise lower doses than will be administered to adults. In general, pharmaceutical compositions with an active substance content of 0.2 to 500 mg, in particular 10 to 200 mg, compound of Formula I per individual dose are suitable for administration to mammalian subjects, in particular to humans. The compounds of Formula I of the present invention may also be administered by intravenous infusion, at a dose which is likely to range from 0.001-10 mg/kg/hr. The above dosages are exemplary of the average case.
Beneficial effects may be anticipated if the hydroxyphenyl-substituted pyrrolo[2,3d] pyrimidine derivatives of Formula I are administered in combination with at least one other active agent useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases. The compounds of Formula I and the other active agents may e.g. be administered as a combination preparation. Within the context of this invention, the term "combination preparation" comprises both true combinations, meaning compounds of Formula I and other active agents physically combined in one preparation such as a tablet or injection fluid; as well as a "kit-of-parts", comprising compounds of Formula I and other active agents useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases in separate dosage forms, together with instructions for use, with or without further means for facilitating compliance with the administration of the component compounds, e.g. labels or drawings. With true combinations, the pharmacotherapy by definition is simultaneous. True combinations comprise fixed combinations of compounds of Formula I and at least one other active agent useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases. The contents of "kit-of-parts" can be administered either simultaneously or at different time intervals. The therapy, being either concomitant or sequential, will be dependant on the characteristics of the other medicaments used, characteristics such as onset and duration of action, plasma levels, clearance, etc., as well as on the disease, its stage, and characteristics of the individual patient.
Other pharmacologically active agents useful in the therapy of cardiovascular, renal, hepatic, and/or metabolic disorders or diseases which may be suitable for administration in combination with the compounds of Formula I as described herein comprise e.g. the following:
ACE-inhibitors, e.g. benazepril, captopril, cilazapril, enalapril, fosinopril, imidapril, lisino- pril, moexipril, quinapril, perindopril, ramipril, spirapril or trandolapril;
ACE/NEP mixed inhibitors, e.g. omapatrilat; adenosine; aldosterone receptor antagonists, e.g. eplerenone; alpha-adrenoceptor antagonists (non-selective), e.g. tolazoline or phenoxybenzamine; alpha-adrenoceptor antagonists (selective), e.g. doxazosin (mesylate), prazosin (hydrochloride and polythiazide), terazosin (hydrochloride) or urapidil; alpha2-adrenoceptor agonists (including centrally acting alpha2-adrenoceptor agonists), e.g. clonidine, guanfacine, guanabenz, methyldopa and moxonidine; anti-anginal drugs, e.g. bepridil, beta blockers, diltiazem, nicardipine, nifedipine, nitrates; anticoagulants, e.g. dalteparin, danaparoid, enoxaparin, heparin, tinzaparin, warfarin; antidiabetics, e.g. insulins, amylin, derivatives of GLP-1 and GLP-2 such as, for example, those disclosed in WO 98/08871 and orally active hypoglycemic active ingredients. The orally active hypoglycemic active ingredients preferably comprise sulfonylureas, e.g tolbutamide, glibenclamide, glimepiride, glipizide, gliquidone, gli- soxepide, glibomuride or gliclazide; biguanides, e.g. metformin; meglitinides, e.g. repaglinide; beta3 adrenergic agonists; oxadiazolidinediones; glucosidase inhibitors e.g. alpha-glucosidase inhibitors such as miglitol or acarbose; glucagon receptor antagonists, GLP-1 agonists, potassium channel openers like diazoxide or those disclosed in WO 97/26265 or WO 99/03861 ; CB-1 (cannabinoid-1 receptor) antagonists/inverse agonists; insulin sensitizers like thiazolidinediones, e.g. troglita- zone, ciglitazone, pioglitazone, rosiglitazone or the compounds disclosed in WO 97/41097, in particular 5-[[4-[(3,4-dihydro-3-methyl-4-oxo-2- quinazolinylmethoxy]pheny-l]methyl]-2,4-thiazolidinedione; activators of insulin receptor kinase; inhibitors of liver enzymes involved in the stimulation of gluconeo- genesis and/or glycogenosis, for example inhibitors of glycogen phosphorylase; and modulators of glucose uptake and glucose excretion; antiplatelet drugs, e.g. abciximab, aspirin, aspirin and dipyridamole, cilostazol, clopido- grel, dipyridamole, eptifibatide, ticlodipine, tirofiban; antiarrhythmic drugs like class I antiarrhythmics, e.g. sodium channel blockers, disopyra- mide, flecainide, lidocaine, mexiletine, moricizine, procainamide, propafenone, quinidine, tocainide; or class Il antiarrhythmics, e.g. beta-adrenoceptor antagonists (see below); or class III antiarrhythmics, e.g. potassium channel blockers like amio- darone, azimilide, bepridil, dofetilide, ibutalide, sotalol, tedisamil; or class IV antiarrhythmics, e.g. calcium channel blockers (see below); ATi receptor antagonists ("sartans"), e.g. abitesartan, benzyllosartan, candesartan, elisartan, embusartan, enoltasosartan, eprosartan, fonsartan, forasartan, glycyl- losartan, irbesartan, isoteoline, losartan, milfasartan, olmesartan, opomisartan, pra- tosartan, ripisartan, saprisartan, saralasin, sarmesin, tasosartan, telmisartan, val- sartan, zolasartan; Kissei KRH-94, Lusofarmaco LR-B/057, Lusofarmaco LR-B/081 , Lusofarmaco LR B/087, Searle SC-52458, Sankyo CS-866, Takeda TAK-536, Uri- ach UR-7247, A-81282, A-81988, BIBR-363, BIBS39, BIBS-222, BMS-180560, BMS-184698, CGP-38560A, CGP-48369, CGP-49870, CGP-63170, CI-996, CV- 1 1 194, DA-2079, DE-3489, DMP-81 1 , DuP-167, DuP-532, GA-0056, E-4177, EMD- 66397, EMD-73495, EXP-063, EXP-929, EXP-3174, EXP-6155, EXP-6803, EXP- 771 1 , EXP-9270, FK-739, HN-65021 , HR-720, ICI-D6888, ICI-D7155, ICI-D8731 , KRI-1 177, KT3-671 , KW-3433, L-158809, L-158978, L-159282, L-159689, L- 159874, L-161177, L-162154, L-162234, L-162441 , L-163007, L-163017, LY- 235656, LY-285434, LY-301875, LY-302289, LY-315995, ME-3221 , PD-123177, PD-123319, PD-150304, RG-13647, RWJ-38970, RWJ-46458, S-8307, S-8308, SL-91.0102, U-96849, U-97018, UP-269-6, UP-275-22, WAY-126227, WK- 1492.2K, WK-1360, X-6803, XH-148, XR-510, YM-358, YM-31472, ZD-6888, ZD- 7155 and ZD-8731 , or any physiologically compatible salts, solvates, prodrugs or esters thereof; beta-adrenoceptor antagonists ("beta blockers") e.g. acebutolol, alprenolol, atenolol, be- taxolol, bisoprolol, bupranolol, carazolol, carteolol, celiprolol, mepindolol, metipranolol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, so- talol and/or timolol; beta- /alpha-adrenoceptor mixed antagonists, e.g. carvedilol or labetolol; calcium channel blocking agents ("calcium antagonists") e.g. amlodipine, bepridil, felodip- ine, isradipine, nicardipine, nifedipine, nilvadipine, nimodipine, nisoldipine, nitrendipine; gallopamil, verapamil; diltiazem and/or fendiline; digoxin; direct vasodilators, e.g. diazoxide, dihydralazine, hydralazine, nitrate or minoxidil;
ECE-inhbitors, e.g. FR-901533; PD-069185; CGS-26303; CGS-34043; CGS-35066; CGS-30084; CGS-35066; SM-19712; Ro0677447;
ECE/NEP-mixed inhibitors, e.g. daglutril and the compounds disclosed in European Patent Application EP 733642 or in International Patent Application WO 2005/030795; endothelin receptor antagonists (mixed or selective) e.g. atrasentan, bosentan, clazosen- tan, darusentan, sitaxsentan, tezosentan, BMS-193884 or J-104132; ganglion blockers, e.g. reserpine or guanethidine;
HMG CoA reductase inhibitors ("statins") e.g. atorvastatin, berivastatin, cerivastatin, cril- vastatin, fluvastatin, glenvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin, tempostatin or any physiologically compatible salts, solvates, prodrugs or esters thereof; natriuretic peptides, e.g. ANP, nesiritide and/or ularitide; NEP-inhibitors (selective); non-adenosine modifying diuretics, e.g. thiazide diuretics like althiazide, bemetizide, ben- droflumethiazide, benzylhydrochlorothiazide, benzthiazide, buthiazide, chlorothiazide, cyclothiazide, cyclopenthiazide, ethiazide, hydrochlorothiazide, hydroflumethiazide, methylclothiazide, paraflutizide, polythiazide, teclothiazide, trichlormethi- azide or any physiologically compatible tautomers, salts, solvates, prodrugs or esters thereof; thiazide analogues like chloraminofenamide, chlortalidone, clofena- mide, clopamide, clorexolone, fenquizone, indapamide, mefruside, metolazone, quinethazone, tripamide and xipamide; loop diuretics like azosemide, bumetanide, furosemide, piretanide, torsemide or any physiologically compatible tautomers, salts, solvates, prodrugs or esters thereof; potassium sparing diuretics like amilo- ride, potassium canrenoate, spironolactone, triamterene or any physiologically compatible tautomers, salts, solvates, prodrugs or esters thereof; carbonic anhy- drase inhibitors like acetazolamide, brinzolamide, dichlorophenamide, dorzolamide, ethoxzolamide, indisulam, methazolamide, zonisamide or any physiologically compatible tautomers, salts, solvates, prodrugs or esters thereof; and/or ethacrynic acid; renin-inhibitors, e.g. alskiren; urotensin Il receptor antagonists; vasopressin receptor (VP1 and/or VP2) antagonists.
Example I: solid pharmaceutical composition of the test substances
Capsules containing 4-[4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrimidin- 2-yl]-phenol:
Capsules with the following composition per capsule are produced: 4-[4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol 20 mg
Corn starch 60 mg
Lactose 300 mg
Ethyl acetate q.s.
The active substance, the corn starch and the lactose are processed into a homogeneous pasty mixture using ethyl acetate. The paste is ground and the resulting granules are placed on a suitable tray and dried at 45°C in order to remove the solvent. The dried granules are passed through a crusher and mixed in a mixer with the further following auxiliaries:
Talcum 5 mg
Magnesium stearate 5 mg
Corn starch 9 mg and then poured into standard 400 mg capsules (= capsule size 0).
Example II: liquid pharmaceutical composition of the test substances
Solution containing 4-[4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrimidin-2- yl]-phenol:
A solution suitable for pharmaceutical use is produced with the following amounts per dose:
4-[4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol 10.00 mg 0.1 M methanesulfonic acid (ζ = 1.002 g/cm3) 326.98 mg
5% dextrose solution for injection (ζ = 1.018 g/cm3) 664.23 mg
The active substance is weighed on an analytical balance into a beaker, the 0.1 M methanesulfonic acid solution and the 5% dextrose solution are added to the active substance and the mixture is then homogenized by ultrasound treatment for approx. 10 min. The resulting yellow, clear solution (pH approximately 2.3) may then be filled into suitable administration forms.
The following examples of compounds of Formula I are intended to explain the invention further, without limiting its scope.
NMR Spectra were recorded in DMSO-dβ on a Bruker DPX 200 spectrometer.
Mass spectra ("MS") were recorded as high performance liquid chromatography mass spectra ("HPLC-MS") on an Agilent Technologies 1 100 HPLC apparatus with a Zorbax Extend-C18 column (150 x 4.6 mm, 5 μm) at 30 0C. The mobile phases were in each case A: 0.02 % trifluoroacetic acid in water, and B: acetonitrile. Gradient: B: 0 min. 10 %
→ 10 min. 90 % →16 min. 90 %. Flow rate: 1.0 mL/min. Detection: 210 nm. Injection: 10 μl. Sample preparation: at approximate concentration of 0.04-1.0 mg/mL.
Example 1 : 4-[4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol
Figure imgf000028_0001
A) 4-Hydroxybenzamidine hydrochloride (15.2 g) was dissolved in absolute ethanol (520 ml.) and a part of the ethanol (370 ml.) was subsequently distilled off again. More absolute ethanol (370 ml.) was added to this receiving solution, together with DBU (30 ml_), 2-cyano-4,4-diethoxy-butyric acid ethyl ester (40.0 g) and triethyl amine (26 ml_). The resulting mixture was heated until reflux and further refluxed for 45 hours ("hrs"). Most of the solvent (430 ml.) was then distilled off while the remainder was allowed to cool to 50 0C. An aqueous solution of hydrochloric acid (2.3 M, 229 ml.) was added dropwise, then the reaction mixture was allowed to cool to ambient temperature and stirring was continued for 1 hr. The resulting precipitate was filtered off, washed with water (3 x 100 ml.) and dried in vacuo at 40 0C to yield 16.0 g 2-(4-hydroxy-phenyl)-7/-/-pyrrolo[2,3-d]pyrimidin-4-ol.
B) 9.0 g of 2-(4-hydroxy-phenyl)-7/-/-pyrrolo[2,3-d]pyrimidin-4-ol as obtained above and 45 ml. of DMF were stirred at RT for 10 min. Phosphorus oxychloride (13.7 ml.) was added and the resulting mixture was heated to 100 0C. After 1.2 hrs, the mixture was first allowed to cool to 20 0C and subsequently was cooled to 0 0C. Water (450 mL) was added slowly and the resulting mixture was filtered. The remaining precipitate was washed with water (2 x 90 mL) and dried in vacuo at 40 0C to yield 8.5 g 4-(4-chloro-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl)-phenol.
C) A mixture of 4-(4-chloro-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl)-phenol (2.0 g) as obtained above, frans-4-aminocyclohexanol hydrochloride (2.4 g) and sodium carbonate (3.4 g) in 1-pentanol (24 mL) was stirred at 138 0C for 17 hrs. Then, most of the solvent was distilled off and water (55ml_) was added. Distillation was continued to remove more 1-pentanol and the residue was allowed to cool to room temperature. Again water (350 ml.) was added and the pH was adjusted to 7-8 with acetic acid. The aqueous phase was extracted with a mixture of ethyl acetate and methyl ethyl ketone and the combined organic layers were dried over Na2SO4 and concentrated in vacuo. The residue was purified by chromatography on silica gel (eluent: n- heptane/ ethyl acetate). The combined product fractions yielded approximately 0.6 g of the title compound as an off-white powder, HPLC-MS: retention time ("Rt") approximately 4.3 min., m/z 325 = M(MIe compound) + H; 1H-NMR (DMSOd6): see table 3 below.
Table 3: 1H-NMR data of the test substance of Example 1
Figure imgf000029_0001
= solvent, ** = standard Example 2: 4-[4-(frans-4-hydroxy-cyclohexylamino)-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl]-benzene-1 ,2-diol
Figure imgf000030_0001
A) Hydrogen chloride gas was bubbled into a solution of 3,4-dihydroxybenzonitrile (200.0 g) in absolute ethanol (1850 ml.) for 45 min. The reaction apparatus was closed and the mixture was stirred at room temperature ("RT") for 56 hrs. The formed precipitate was filtered off and washed with a small amount of cold ethanol. Anhydrous ammonium acetate (128.9 g) and absolute ethanol (3500 ml.) were added to the precipitate and the mixture was stirred at 60 0C for 24 hrs. The resulting mixture was concentrated in vacuo to approximately 1/6 of its prior volume and the concentrate was treated with diethyl ether (3500 ml_). The resulting precipitate was filtered off and dried to yield 196.8 g of 3,4-dihydroxybenzamidine hydrochloride as a white powder.
B) 3,4-Dihydroxybenzamidine hydrochloride as obtained above (61.7 g) was dissolved in absolute ethanol (2000 ml.) and a part of the ethanol (1400 ml.) was subsequently distilled off again. More absolute ethanol (1400 mL) was added to this receiving solution, together with DBU (1 13 mL), 2-cyano-4,4-diethoxy-butyric acid ethyl ester (148.0 g) and triethyl amine (96 mL). The resulting mixture was heated until reflux and further refluxed for 57 hrs. Most of the solvent (1700 mL) was then distilled off while the remainder was allowed to cool to 50 0C. An aqueous solution of hydrochloric acid (2.3 M, 930 mL) was added slowly, then the reaction mixture was allowed to cool to ambient temperature and stirring was continued for 25 hrs. The resulting precipitate was filtered off, washed with water (3 x 150 mL) and dried in vacuo at 40 0C to yield 31.5 g 4-(4-hydroxy-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl)- benzene-1 ,2-diol.
C) 15.0 g of 4-(4-hydroxy-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl)-benzene-1 ,2-diol as obtained above and 195 mL of DMF were stirred at RT for 10 min. under nitrogen atmosphere. The mixture was heated to 50 0C, phosphorus oxychloride (35.0 g) was added and the resulting mixture was heated to 90 0C. After 2 hrs, the mixture was first allowed to cool to 30 0C and subsequently was cooled to 0 0C in an ice bath. Water (30 ml.) was added slowly while keeping the temperature below 40 0C. The resulting mixture was poured into ice water (2100 ml.) and the aqueous phase was extracted with ethyl acetate (2 x 200 ml_, 1 x 300 ml_). The combined organic layers were filtered and washed with water (2 x 300 ml.) and brine (200 ml_). Drying over Na2SO4 and concentration in vacuo finally yielded 12.6 g 4-(4-chloro-7/-/- pyrrolo[2,3-d]pyrimidin-2-yl)-benzene-1 ,2-diol.
A solution of 4-(4-chloro-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl)-benzene-1 ,2-diol (5.0 g) as obtained above in 1-pentanol (95 ml.) was heated to reflux and part of the solvent (40 ml.) was distilled off. Trans-4-aminocyclohexanol hydrochloride (4.1 g) and sodium carbonate (8.1 g) were added to the mixture and distillation was continued to remove more water. Heating was continued and more frans-4-aminocyclohexanol hydrochloride (1.8 g) was added after 5 hrs. Heating was continued for another 5.5 hrs and the mixture was then allowed to cool to RT. Water (200 ml.) was added and the pH of the mixture was adjusted to 7 by addition of acetic acid. Methyl ethyl ketone (250 ml.) was added and the layers were separated. The aqueous layer was extracted with methyl ethyl ketone (2 x 50 ml_), the combined organic layers were washed with brine (250 ml.) and dried over Na2SO4- The dried combined organic layers were partially concentrated and n-heptane (250 ml.) was added. The resulting slurry was stirred at RT for 15 min., the precipitate was filtered off, washed with cold n-heptane (40 ml.) and dried at 30 0C in vacuo. The solid material was dissolved in hot methanol and precipitated with dichloromethane. Crude product from a previous batch was added and the before treatment was repeated. The remaining solid was re-crystallized from tetrahydrofuran ("THF") to yield 1.7 g of the title compound as an off-white powder, HPLC-MS: Rt approx. 3.5 min., m/z 341 = M(title compound) + H; 1H-NMR (DMSO-d6): see table 4 below.
Table 4: 1H-NMR data of the test substance of Example 2
Figure imgf000032_0001
= solvent; ** = standard
Example 3:
3-[4-(frans-4-hydroxy-cyclohexylamino)-7H-pyrrolo[2,3-d]pyrinniclin-2-yl]-phenol
Figure imgf000033_0001
A) Hydrogen chloride gas was bubbled into an ice-cooled solution of 3-hydroxy- benzonitrile (200.0 g) in absolute ethanol (2000 ml.) until a saturated or nearly saturated solution was obtained. The formed precipitate was filtered off and washed with cold ethanol (300 ml_). Anhydrous ammonium acetate (146.2 g) and absolute ethanol (4000 mL) were added to the precipitate and the mixture was stirred overnight at 60 0C. The resulting mixture was concentrated in vacuo to approximately 1/6 of its prior volume and the concentrate was treated with diethyl ether (2000 mL). The resulting precipitate was filtered off with suction, washed with diethyl ether and dried in vacuo to yield 272.5 g 3-hydroxybenzamidine hydrochloride as a white powder.
B) 3-Dihydroxybenzamidine hydrochloride as obtained above (89.2 g) was dissolved in absolute ethanol (3080 mL) and a part of the ethanol (2200 mL) was subsequently distilled off again. More absolute ethanol (2200 mL) was added to this receiving solution, together with DBU (175 g), 2-cyano-4,4-diethoxy-butyric acid ethyl ester (237.0 g) and triethyl amine (1 1 1 g). The resulting mixture was heated until reflux and further refluxed for 22 hrs. Most of the solvent (2950 mL) was then distilled off while the remainder was allowed to cool to 50 0C. An aqueous solution of hydrochloric acid (2.3 M, 1360 mL) was added slowly, the reaction mixture was allowed to cool to ambient temperature and stirring was continued for 1 hr. The resulting precipitate was filtered off, washed with water (3 x 100 mL) and dried in vacuo at 40 0C for 2 days to yield 82.8 g 2-(3-hydroxyphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-ol.
C) 35.0 g 2-(3-hydroxyphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-ol as obtained above and 385 mL of anhydrous DMF were stirred at RT for 10 min. under nitrogen atmosphere. Phosphorus oxychloride (87.4 g) was added in small portions while the internal temperature was kept below 90 0C. After 2 hrs at 85-90 0C, the mixture was cooled below 20 0C in an ice bath. Under vigorous stirring, water (2100 ml.) was added slowly while the temperature was kept below 30 0C. The precipitated crude product was filtered, washed with water (2 x 420 ml.) and dried in vacuo overnight to yield 28.8 g 3-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-2-yl)-phenol.
A mixture of 3-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-2-yl)-phenol (6.7 g) as obtained above, frans-4-aminocyclohexanol hydrochloride (8.27 g) and sodium carbonate (1 1.6 g) in 1-pentanol (136 ml.) was heated to reflux and 35 ml. of 1-pentanol was distilled off. The reaction mixture was then heated to reflux for 10 hrs and subsequently allowed to cool to RT. Water (200 ml.) was added and the pH of the mixture was neutralised with acetic acid. Methyl ethyl ketone (100 ml.) was added and the layers were separated. The aqueous layer was further extracted with methyl ethyl ketone (2 x 50 ml_), the combined organic layers were washed with water (200 ml.) brine (100 ml.) and dried over Na2SO4. The dried combined organic layers were partially concentrated and n-heptane (200 ml.) was added. The precipitated crude product was filtered and washed with n-heptane (20 ml_). After drying in vacuo at 40 0C, the crude product was re-crystallized from n-butanol/methyl ethyl ketone (1 :1 v/v) and n-heptane. The obtained precipitate from re-crystallization was isolated by filtration and dried in vacuo at 40 0C before it was dissolved in methanol (50 ml.) and methyl ethyl ketone (450 ml_). The resulting solution was passed through a column filled with silica gel and the column was eluted with methyl ethyl ketone until the entire product had been recovered from the column. The combined product fractions were evaporated to dryness to yield 4.4 g of the title compound as a pale orange solid, HPLC-MS: Rt approx. 4.0 min., m/z 325 = M(title compound) + H; 1H-NMR (DMSOd6): see table 5 below.
Table 5: 1H-NMR data of the test substance of Example 3
Figure imgf000035_0001
= solvent; ** = standard

Claims

Claims
1. A compound of the general Formula I,
Figure imgf000036_0001
wherein
R1 is hydrogen or hydroxy, and physiologically compatible salts, tautomers, isotopically-labelled analogues, solvates and/or esters thereof.
2. A compound according to Claim 1 wherein R1 is hydrogen
3. A compound according to Claim 2 wherein the hydroxy substituent at the phenyl group is in the para-position.
4. A compound according to Claim 1 wherein the substituents at the cyclohexyl group are in the frans-position to each other.
5. A compound according to Claim 1 which is 4-[4-(frans-4-hydroxy-cyclohexyl- amino)-7/-/-pyrrolo[2,3-d]pyrimidin-2-yl]-phenol.
6. A compound of general formula I according to Claim 1 , for the use as medicament.
7. A compound of general formula I according to Claim 1 , for the prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases.
8. The use of compounds of Formula I according to claim 1 for the preparation of medicaments for the prophylaxis and/or treatment of cardiovascular, renal and/or hepatic disorders or diseases.
9. A use according to Claim 8 wherein the cardiovascular, renal and/or hepatic disorder or disease is selected from the group consisting of acute decompensated heart failure; acute heart failure; acute renal failure; compensated chronic heart failure; congestive heart failure; contrast media-induced renal failure; decompensated chronic heart failure; diabetic nephropathy; edema; forward heart failure; heart failure; hepatorenal syndrome; high-output heart failure; hypertension; hypotension on hemodyalisis; ischemia reperfusion injury; ischemic renal failure; left-sided heart failure; liver cirrhosis; liver cirrhosis with ascites; nephritis; renal disease; renal dysfunction; renal failure; renal hypertension; renal impairment; right-sided heart failure, or combinations of any of the foregoing.
10. A pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I according to Claim 1 and conventional pharmaceutically acceptable auxiliaries and/or carriers.
1 1. A process for the preparation of compounds of Formula I,
Figure imgf000037_0001
wherein
R1 is hydrogen or hydroxy, and physiologically acceptable salts, tautomers, isotopically-labelled analogues, solvates and/or esters thereof, characterised in that a compound of the general Formula II,
Figure imgf000038_0001
wherein R1 has the above meaning and X stands for a cleavable leaving group, is reacted with a compound of the general Formula III,
Figure imgf000038_0002
wherein R2 is hydrogen or a protective group for the hydroxy group; where R2 does not represent a desired physiologically acceptable ester, the protective group for the hydroxy group R2 is cleaved off in the resulting compounds, and if desired, resulting compounds of Formula I are converted into their physiologically acceptable salts, solvates and/or esters, or salts, solvates and/or esters of the compounds of Formula I are converted into the free compounds of formula I.
12. Compounds of the general Formula II,
Figure imgf000038_0003
wherein
R1 is hydrogen or hydroxy, and
X is a cleavable leaving group, and salts, tautomers, solvates and/or esters thereof.
PCT/EP2009/057473 2008-06-18 2009-06-16 HYDROXYPHENYL-SUBSTITUTED PYRROLO[2,3d]PYRIMIDINE DERIVATIVES, PROCESSES AND INTERMEDIATE PRODUCTS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THESE COMPOUNDS WO2009153261A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US7350508P 2008-06-18 2008-06-18
US61/073,505 2008-06-18
EP08104458 2008-06-18
EP08104458.8 2008-06-18

Publications (1)

Publication Number Publication Date
WO2009153261A1 true WO2009153261A1 (en) 2009-12-23

Family

ID=40352308

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2009/057473 WO2009153261A1 (en) 2008-06-18 2009-06-16 HYDROXYPHENYL-SUBSTITUTED PYRROLO[2,3d]PYRIMIDINE DERIVATIVES, PROCESSES AND INTERMEDIATE PRODUCTS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THESE COMPOUNDS

Country Status (1)

Country Link
WO (1) WO2009153261A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2567959A1 (en) 2011-09-12 2013-03-13 Sanofi 6-(4-Hydroxy-phenyl)-3-styryl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013182580A1 (en) * 2012-06-07 2013-12-12 F. Hoffmann-La Roche Ag Pyrrolopyrimidone and pyrrolopyridone inhibitors of tankyrase
WO2015078417A1 (en) * 2013-11-29 2015-06-04 四川好医生药业集团有限公司 Pyrrolopyrimidine compound and use thereof in preparation of hypoglaecemic drug
US20150329542A1 (en) * 2014-05-14 2015-11-19 Pfizer Inc. Pyrazolopyridines and pyrazolopyrimidines

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999062518A1 (en) * 1998-06-02 1999-12-09 Osi Pharmaceuticals, Inc. PYRROLO[2,3d]PYRIMIDINE COMPOSITIONS AND THEIR USE

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999062518A1 (en) * 1998-06-02 1999-12-09 Osi Pharmaceuticals, Inc. PYRROLO[2,3d]PYRIMIDINE COMPOSITIONS AND THEIR USE

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
P KALK, B EGGERT, K RELLE, M GODES, S HEIDEN, Y SHARKOVSKA, Y FISHER, D TIEGLER, G-W BIELENBERG AND B HOCHER: "The Adenosine A1 Receptor Antagonist SLV320 reduces Myocardial Fibrosis in Rats with 5/6 Nephrectomy without affecting Blood Pressure", BRITISH JOURNAL OF PHARMACOLOGY, no. 151, 11 June 2007 (2007-06-11), pages 1025 - 1032, XP002516507 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2567959A1 (en) 2011-09-12 2013-03-13 Sanofi 6-(4-Hydroxy-phenyl)-3-styryl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013182580A1 (en) * 2012-06-07 2013-12-12 F. Hoffmann-La Roche Ag Pyrrolopyrimidone and pyrrolopyridone inhibitors of tankyrase
US8722661B2 (en) 2012-06-07 2014-05-13 Hoffmann-La Roche Inc. Pyrrolopyrimidone and pyrrolopyridone inhibitors of tankyrase
CN104350056A (en) * 2012-06-07 2015-02-11 霍夫曼-拉罗奇有限公司 Pyrrolopyrimidone and pyrrolopyridone inhibitors of tankyrase
JP2015522557A (en) * 2012-06-07 2015-08-06 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Pyrrolopyrimidone and pyrrolopyridone inhibitors of tankyrase
WO2015078417A1 (en) * 2013-11-29 2015-06-04 四川好医生药业集团有限公司 Pyrrolopyrimidine compound and use thereof in preparation of hypoglaecemic drug
US20150329542A1 (en) * 2014-05-14 2015-11-19 Pfizer Inc. Pyrazolopyridines and pyrazolopyrimidines
US9518052B2 (en) * 2014-05-14 2016-12-13 Pfizer Inc. Pyrazolopyridines and pyrazolopyrimidines
US10022376B2 (en) 2014-05-14 2018-07-17 Pfizer Inc. Pyrazolopyridines and pyrazolopyrimidines

Similar Documents

Publication Publication Date Title
AU2005244450B2 (en) Medicaments containing N-sulfamoyl-N&#39;-arylpiperazines for the prophylaxis or treatment of obesity and related conditions
KR20080074217A (en) Novel n-sulfamoyl-piperidineamides for the prophylaxis or treatment of obesity and related conditions
JP7447110B2 (en) Tetrahydropyran (THP)-substituted bicyclic-pyrimidinedione compounds
US7772225B2 (en) N-sulfamoyl-piperidineamides for the treatment or inhibition of obesity and related conditions
US7547708B2 (en) N-sulfamoyl-N′-benzopyranpiperidine compounds and uses thereof
WO2009153261A1 (en) HYDROXYPHENYL-SUBSTITUTED PYRROLO[2,3d]PYRIMIDINE DERIVATIVES, PROCESSES AND INTERMEDIATE PRODUCTS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THESE COMPOUNDS
EP1951725B1 (en) N-sulfamoyl-n -benzopyranpiperidines as inhbitors of carbonic anhydrases
US20050261292A1 (en) Pharmaceutical composition containing N-sulfamoyl-N&#39;-arylpiperazines for the treatment or inhibition of obesity and related conditions
TWI837202B (en) Tetrahydropyran (thp)-substituted bicyclic-pyrimidinedione compounds
KR20070022322A (en) Medicaments containing n­sulfamoyl­n?­arylpiperazines for the prophylaxis or treatment of obesity and related conditions
WO2011134019A1 (en) Novel biphenyl sartans

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: 09765841

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: 09765841

Country of ref document: EP

Kind code of ref document: A1