EP3452468A1 - Amide-substituted aryltriazole derivatives and uses thereof - Google Patents

Amide-substituted aryltriazole derivatives and uses thereof

Info

Publication number
EP3452468A1
EP3452468A1 EP17720501.0A EP17720501A EP3452468A1 EP 3452468 A1 EP3452468 A1 EP 3452468A1 EP 17720501 A EP17720501 A EP 17720501A EP 3452468 A1 EP3452468 A1 EP 3452468A1
Authority
EP
European Patent Office
Prior art keywords
methyl
compound
mmol
group
general formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP17720501.0A
Other languages
German (de)
French (fr)
Inventor
Marie-Pierre Collin-Kröpelin
Peter Kolkhof
Thomas Neubauer
Chantal FÜRSTNER
Elisabeth Pook
Matthias Beat WITTWER
Klemens Lustig
Anja BUCHMÜLLER
Hanna Tinel
Karoline DRÖBNER
Thomas MONDRITZKI
Heiko Schirmer
Carsten Schmeck
Pierre Wasnaire
Hana CERMECKA
Axel Kretschmer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bayer Pharma AG
Original Assignee
Bayer Pharma AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bayer Pharma AG filed Critical Bayer Pharma AG
Priority claimed from PCT/EP2017/060368 external-priority patent/WO2017191105A1/en
Publication of EP3452468A1 publication Critical patent/EP3452468A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Definitions

  • the present invention relates to novel 5-(carboxamide)-l-aryl-l,2,4-triazole derivatives, to processes for the preparation of such compounds, to pharmaceutical compositions containing such compounds, and to the use of such compounds or compositions for the treatment and/or prevention of diseases, in particular for the treatment and/or prevention of renal and cardiovascular diseases.
  • the liquid content of the human body is subject to various physiological control mechanisms, the purpose of which is to keep it constant (volume homeostasis).
  • both the volume filling of the vascular system and also the osmolarity of the plasma are continuously recorded by appropriate sensors (baroreceptors and osmoreceptors).
  • the information which these sensors supply to the relevant centers in the brain regulates drinking behaviour and controls fluid excretion via the kidneys by means of humoral and neural signals.
  • the peptide hormone vasopressin is of central importance in this [Schrier R.W., Abraham W.T., New Engl. J. Med. 341, 577-585 (1999)].
  • Vasopressin is produced in specialized endocrine neurons in the Nucleus supraopticus and N. para- ventricularis in the wall of the third ventricle (hypothalamus) and is transported from there along the neural processes into the posterior lobes of the hypophysis (neurohypophysis). There the hormone is released into the bloodstream in response to stimulus. A loss of volume, e.g. as a result of acute bleeding, heavy sweating, prolonged thirst or diarrhoea, is a stimulus for intensified release of the hormone. Conversely, the secretion of vasopressin is inhibited by an increase in the intravascular volume, e.g. as a result of increased fluid intake.
  • Vasopressin exerts its action mainly via binding to three receptors, which are classified as Via, Vlb and V2 receptors and which belong to the family of G protein-coupled receptors.
  • Via receptors are mainly located on the cells of the vascular smooth musculature. Their activation gives rise to vasoconstriction, as a result of which the peripheral resistance and blood pressure rise. Apart from this, Via receptors are also detectable in the liver.
  • Vlb receptors also named V3 receptors
  • vasopressin regulates the basal and stress-induced secretion of adrenocorticotropic hormone (ACTH) via the Vlb receptor.
  • CSH corticotropin-releasing hormone
  • V2 receptors are located in the distal tubular epithelium and the epithelium of the collecting tubules in the kidney. Their activation renders these epithelia permeable to water. This phenomenon is due to the incorporation of aquaporins (special water channels) in the luminal membrane of the epithelial cells.
  • vasopressin for the reabsorption of water from the urine in the kidney becomes clear from the clinical picture of diabetes insipidus, which is caused by a deficiency of the hormone, e.g. owing to hypophysis damage. Patients who suffer from this disease excrete up to 20 liters of urine per 24 hours if they are not given replacement hormone. This volume corresponds to about 10% of the primary urine. Because of its great importance for the reabsorption of water from the urine, vasopressin is also synonymously referred to as antidiuretic hormone (ADH). Consequently, pharmacological inhibition of the action of vasopressin/ ADH on the V2 receptor results in increased urine excretion.
  • ADH antidiuretic hormone
  • V2 receptor antagonists cause increased water excretion, without substantially increasing the excretion of electrolytes. This means that with V2 antagonist drugs, volume homeostasis can be restored without affecting electrolyte homeostasis.
  • drugs with V2 antagonistic activity appear particularly suitable for the treatment of all disease conditions which are associated with an overloading of the body with water, without the electrolytes being adequately increased in parallel.
  • hyponatremia sodium concentration ⁇ 135 mmol/L
  • hyponatremia sodium concentration ⁇ 135 mmol/L
  • comatose states and death are imminent.
  • hypovolemic euvolemic
  • hypervolemic hyponatremia The forms of hypervolemia with edema formation are clinically significant. Typical examples of these are the syndrome of inappropriate ADH/vasopressin secretion (SIADH) (e.g.
  • the disturbed neurohormonal regulation essentially manifests itself in an elevation of the sympathetic tone and inappropriate activation of the renin-angiotensin-aldosterone system. While the inhibition of these components by beta-receptor blockers on the one hand and by ACE inhibitors or angiotensin-receptor blockers on the other is now an inherent part of the pharmacological treatment of heart failure, the inappropriate elevation of vasopressin secretion in advanced heart failure is at present still not adequately treatable. Apart from the retention of water mediated by V2 receptors and the unfavourable hemodynamic consequences associated therewith in terms of increased backload, the emptying of the left ventricle, the pressure in the pulmonary blood vessels and cardiac output are also adversely affected by V la-mediated vasoconstriction.
  • a direct hypertrophy-promoting action on the heart muscle is also attributed to vasopressin.
  • the direct action on the heart muscle is triggered by activation of VI a receptors.
  • vasopressin agents which inhibit the action of vasopressin on the V2 and/or the Via receptor appear suitable for the treatment of heart failure.
  • compounds with combined activity on both vasopressin receptors should have both desirable renal as well as hemodynamic effects and thus offer an especially ideal profile for the treatment of patients with heart failure.
  • the provision of such combined vasopressin antagonists also appears to make sense inasmuch as a volume diminution mediated solely via V2 receptor blockade can entail the stimulation of osmoreceptors and, as a result, may lead to a further compensatory increase in vasopressin release.
  • vasopressin such as for example vasoconstriction and heart muscle hypertrophy
  • Via receptors are mainly located on vascular smooth muscle cells (VSMC) but also on cardiomyocytes, fibroblasts and specialized renal cells like glomerular mesangial cells or cells of the macula densa which control the release of renin [Wasilewski MA, Myers VD, Recchia FA, Feldman AM, Tilley DG, Cell Signal., 28(3), 224-233, (2016)].
  • VSMC Via receptor by vasopressin gives rise to intracellular calcium release and according vasoconstriction. Therefore, stimulation of VSMC Via receptors causes increased vascular resistance and increased cardiac afterload. Cardiac output is adversely affected by V la-mediated vasoconstriction.
  • Via receptor is also expressed in the renal cortical and medullary vasculature, where it mediates vasoconstriction of renal vessels and affecting overall renal blood flow.
  • the activation of Via receptor can decrease renal medullary blood flow inducing further pathological processes as tissue hypoxia, reduced oxygen and according energy supply for tubular transport processes as well as direct damages of mesangial and macula densa cells. It has been demonstrated that mesangial Via receptor activation mediates TGFp signaling and causes an increase in production of collagen IV.
  • Via receptors are mainly expressed on VSMCs and thus participating in vascular function, a link to vascular diseases as peripheral arterial disease (PAD) including claudication and critical limb ischemia as well as coronary microvascular dysfunction (CMD) is conceivable. Apart from this, Via receptors are also expressed on human platelets and in the liver. The meaning of platelet Via receptors is not fully understood although vasopressin induces aggregation of human platelets via Via receptor at high concentrations ex vivo.
  • PAD peripheral arterial disease
  • CMD coronary microvascular dysfunction
  • vasopressin-induced platelet aggregation by Via receptor antagonists is a useful pharmacological ex vivo assay making use of human tissue endogenously expressing the Via receptor [Thibonnier M, Roberts JM, J Clin Invest.; 76:1857-1864, (1985)].
  • Vasopressin stimulates gluconeogenesis and glycogenolysis via activation of the hepatic Via receptor.
  • Animal studies have shown that vasopressin impairs glucose tolerance which could be inhibited by a Via receptor antagonist thereby providing a link of vasopressin receptor Via to diabetes mellitus.
  • a Via receptor antagonist thereby providing a link of vasopressin receptor Via to diabetes mellitus.
  • vasopressin also seems to play a causal role in the development of preeclampsia.
  • Chronic infusion of vasopressin during pregnancy in mice is sufficient to induce all of the major maternal and fetal phenotypes associated with human preeclampsia, including pregnancy-specific hypertension [Santillan MK, Santillan DA, Scroggins SM, Min JY, Sandgren JA, Pearson NA, Leslie KK, Hunter SK, Zamba GK, Gibson-Corley KN, Grobe JL.
  • Vasopressin in preeclampsia a novel very early human pregnancy biomarker and clinically relevant mouse model. Hypertension. 64(4), 852-859, (2014)].
  • Vasopressin levels can be elevated in women with dysmenorrhoea (a gynecological disorder which is characterised by cyclical cramping pelvic pain) during menstruation, which appear to increase myometrial smooth muscle contraction. It was found recently that a selective vasopressin Via receptor antagonist (relcovaptan/SR-49059) can reduce intrauterine contractions elicited by vasopressin.
  • agents which inhibit the action of vasopressin on the Via receptor appear suitable for the treatment of several cardiovascular diseases.
  • agents which inhibit the action of vasopressin selectively on the Via receptor offer an especially ideal profile for the treatment of otherwise normovolemic patients, i.e. those which are not eligible for decongestion by e.g. high doses of loop diuretics or V2 antagonists, and where induced aquaresis via V2 inhibition may be undesired.
  • Certain 4-phenyl-l,2,4-triazol-3-yl derivatives have been described in WO 2005/063754-A1 and WO 2005/105779-A1 to act as vasopressin Via receptor antagonists that are useful for the treatment of gynecological disorders, notably menstrual disorders such as dysmenorrhea.
  • WO 2011/104322-A1 a particular group of bis-aryl-bonded l,2,4-triazol-3-ones, including 5-phenyl-l,2,4-triazol-3-yl and 1 -phenyl- l,2,3-triazol-4-yl derivatives thereof, has been disclosed as antagonists of vasopressin Via and/or V2 receptors being useful for the treatment and/or prevention of cardiovascular diseases.
  • the compounds of the present invention have valuable pharmacological properties and can be used for the prevention and/or treatment of various diseases and disease-induced states in humans and other mammals.
  • the present invention relates to 5-(carboxamide)-l-aryl-l,2,4-triazole derivatives of the general formula (I)
  • R 1 represents a group of the formula
  • Ar represents a phenyl group or a 5- or 6-membered heteroaryl group attached via a ring carbon atom having one or two ring heteroatoms selected from a nitrogen atom and a sulfur atom, wherein any phenyl group and any 5- or 6-membered heteroaryl group are each optionally substituted, identically or differently, with one or two groups selected from a halogen atom, nitro, cyano, (Ci-C -alkyl, (Ci-C -alkoxy, (Ci-C -alkylsulfanyl, (Ci-
  • the compounds according to this invention can also be present in the form of their salts, solvates and/or solvates of the salts.
  • substituted means that one or more hydrogen atoms on the designated atom or group are replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded. Combinations of substituents and/or variables are permissible.
  • optionally substituted means that the number of substituents can be equal to or different from zero. Unless otherwise indicated, it is possible that optionally substituted groups are substituted with as many optional substituents as can be accommodated by replacing a hydrogen atom with a non-hydrogen substituent on any available carbon atom or heteroatom.
  • 5- to 6-membered heteroaryl means a monovalent, monocyclic aromatic ring having 5 or 6 ring atoms, which contains at least one ring heteroatom and optionally one, two or three further ring heteroatoms from a nitrogen atom and a sulfur atom, and which is bound via a ring carbon atom or optionally via a ring nitrogen atom (if allowed by valency).
  • Said heteroaryl group can be a 5-membered heteroaryl group, such as, for example, thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl or tetrazolyl; or a 6-membered heteroaryl group, such as, for example, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl.
  • a 5-membered heteroaryl group such as, for example, thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl
  • heteroaryl or heteroarylene groups include all possible isomeric forms thereof, e.g. : tautomers and positional isomers with respect to the point of linkage to the rest of the molecule.
  • pyridinyl includes pyridin-2-yl, pyridin-3-yl and pyridin-4-yl; or the term thienyl includes thien-2-yl and thien-3-yl.
  • the heteroaryl group in substituent Ar in the general formula (I), supra is a pyridinyl, a pyrimidyl, or a pyrazinyl group.
  • halogen atom means a fluorine, chlorine, bromine or iodine atom, particularly a fluorine, or chlorine atom.
  • C 1 -C4 as used in the present text, e.g. in the context of the definition of "Ci-C t-alkyl", “Ci-C t-alkoxy”, “ or "Ci-C t-alkylsulfanyl”, means an alkyl group having a finite number of carbon atoms of 1 to 4, i.e. 1, 2, 3, or 4 carbon atoms.
  • Ci-C t-alkyl means a linear or branched, saturated, monovalent hydrocarbon group having 1, 2, 3, or 4 carbon atoms, e.g. a methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, ieri-butyl, or an isomer thereof. Particularly, said group has 1, 2, 3 or 4 carbon atoms (“Ci-C t-alkyl”), e.g.
  • Ci-C t-alkylsulfanyl means a linear or branched, saturated, monovalent group of formula (Ci-C4-alkyl)-S-, in which the term "Ci-C4-alkyl” is as defined supra, e.g. a methylsulfanyl, ethylsulfanyl, propylsulfanyl, isopropylsulfanyl, butylsulfanyl, sec-butylsulfanyl, isobutylsulfanyl, ieri-butylsulfanyl group.
  • Ci-C4-alkoxy means a linear or branched, saturated, monovalent group of formula (Ci-C4-alkyl)-0-, in which the term "Ci-C4-alkyl” is as defined supra, e.g. a methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, isobutoxy, ieri-butoxy, or an isomer thereof.
  • the invention therefore includes one or more isotopic variant(s) of the compounds of general formula (I), particularly deuterium-containing compounds of general formula (I).
  • Isotopic variant of a compound or a reagent is defined as a compound exhibiting an unnatural proportion of one or more of the isotopes that constitute such a compound.
  • Isotopic variant of the compound of general formula (I) is defined as a compound of general formula (I) exhibiting an unnatural proportion of one or more of the isotopes that constitute such a compound.
  • unnatural proportion means a proportion of such isotope which is higher than its natural abundance.
  • the natural abundances of isotopes to be applied in this context are described in "Isotopic Compositions of the Elements 1997", Pure Appl. Chem., 70(1), 217-235, 1998.
  • isotopes examples include stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as 2H (deuterium), 3H (tritium), l lC, 13C, 14C, 15N, 170, 180, 32P, 33P, 33S, 34S, 35S, 36S, 18F, 36C1, 82Br, 1231, 1241, 1251, 1291 and 1311, respectively.
  • stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine such as 2H (deuterium), 3H (tritium), l lC, 13C, 14C, 15N, 170, 180, 32P, 33P, 33S, 34S, 35S, 36S, 18F, 36C1, 82Br, 1231, 1241, 1251, 1291 and 1311, respectively.
  • the isotopic variant(s) of the compounds of general formula (I) preferably contain deuterium ("deuterium- containing compounds of general formula (I)").
  • Isotopic variants of the compounds of general formula (I) in which one or more radioactive isotopes, such as 3H or 14C, are incorporated are useful e.g. in drug and/or substrate tissue distribution studies. These isotopes are particularly preferred for the ease of their incorporation and detectability.
  • Positron emitting isotopes such as 18F or 11C may be incorporated into a compound of general formula (I).
  • These isotopic variants of the compounds of general formula (I) are useful for in vivo imaging applications.
  • Deuterium- containing and 13C-containing compounds of general formula (I) can be used in mass spectrometry analyses in the context of preclinical or clinical studies.
  • Isotopic variants of the compounds of general formula (I) can generally be prepared by methods known to a person skilled in the art, such as those described in the schemes and/or examples herein, by substituting a reagent for an isotopic variant of said reagent, preferably for a deuterium- containing reagent.
  • a reagent for an isotopic variant of said reagent preferably for a deuterium- containing reagent.
  • deuterium from D20 can be incorporated either directly into the compounds or into reagents that are useful for synthesizing such compounds.
  • Deuterium gas is also a useful reagent for incorporating deuterium into molecules.
  • Catalytic deuteration of olefinic bonds and acetylenic bonds is a direct route for incorporation of deuterium.
  • Metal catalysts i.e.
  • deuterated reagents and synthetic building blocks are commercially available from companies such as for example C/D/N Isotopes, Quebec, Canada; Cambridge Isotope Laboratories Inc., Andover, MA, USA; and CombiPhos Catalysts, Inc., Princeton, NJ, USA.
  • deuterium-containing compound of general formula (I) is defined as a compound of general formula (I), in which one or more hydrogen atom(s) is/ are replaced by one or more deuterium atom(s) and in which the abundance of deuterium at each deuterated position of the compound of general formula (I) is higher than the natural abundance of deuterium, which is about 0.015%.
  • the abundance of deuterium at each deuterated position of the compound of general formula (I) is higher than 10%, 20%, 30%, 40%, 50%, 60%, 70% or 80%, preferably higher than 90%, 95%, 96% or 97%, even more preferably higher than 98% or 99% at said position(s). It is understood that the abundance of deuterium at each deuterated position is independent of the abundance of deuterium at other deuterated position(s).
  • the selective incorporation of one or more deuterium atom(s) into a compound of general formula (I) may alter the physicochemical properties (such as for example acidity [C. L. Perrin, et al., J. Am. Chem. Soc, 2007, 129, 4490], basicity [C. L. Perrin et al., J. Am. Chem. Soc, 2005, 127, 9641], lipophilicity [B. Testa et al., Int. J. Pharm., 1984, 19(3), 271]) and/or the metabolic profile of the molecule and may result in changes in the ratio of parent compound to metabolites or in the amounts of metabolites formed.
  • physicochemical properties such as for example acidity [C. L. Perrin, et al., J. Am. Chem. Soc, 2007, 129, 4490], basicity [C. L. Perrin et al., J. Am. Chem. Soc, 2005, 127,
  • Kassahun et al., WO2012/112363 are examples for this deuterium effect. Still other cases have been reported in which reduced rates of metabolism result in an increase in exposure of the drug without changing the rate of systemic clearance (e.g. Rofecoxib: F. Schneider et al., Arzneim. Forsch. / Drug. Res., 2006, 56, 295; Telaprevir: F. Maltais et al., J. Med. Chem., 2009, 52, 7993). Deuterated drugs showing this effect may have reduced dosing requirements (e.g. lower number of doses or lower dosage to achieve the desired effect) and/or may produce lower metabolite loads.
  • a compound of general formula (I) may have multiple potential sites of attack for metabolism.
  • deuterium-containirig compounds of general formula (I) having a certain pattern of one or more deuterium-hydrogen exchange(s) can be selected.
  • the deuterium atom(s) of deuterium- containing compound(s) of general formula (I) is/are attached to a carbon atom and/or is/are located at those positions of the compound of general formula (I), which are sites of attack for metabolizing enzymes such as e.g. cytochrome P450.
  • stable compound' or “stable structure” is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • the compounds of the present invention optionally contain one asymmetric centre, depending upon the location and nature of the various substituents desired. It is possible that one asymmetric carbon atom is present in the (R) or (S) configuration, which can result in racemic mixtures. In certain instances, it is possible that asymmetry also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds. Preferred compounds are those which produce the more desirable biological activity.
  • Separated, pure or partially purified isomers and stereoisomers or racemic mixtures of the compounds of the present invention are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques known in the art.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers.
  • appropriate acids are tartaric, diacetyltartaric, ditoluoyltartaric and camphorsulfonic acid.
  • Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallisation.
  • the optically active bases or acids are then liberated from the separated diastereomeric salts.
  • a different process for separation of optical isomers involves the use of chiral chromatography (e.g., HPLC columns using a chiral phase), with or without conventional derivatisation, optimally chosen to maximise the separation of the enantiomers.
  • Suitable HPLC columns using a chiral phase are commercially available, such as those manufactured by Daicel, e.g., Chiracel OD and Chiracel OJ, for example, among many others, which are all routinely selectable.
  • Enzymatic separations, with or without derivatisation are also useful.
  • the optically active compounds of the present invention can likewise be obtained by chiral syntheses utilizing optically active starting materials. In order to distinguish different types of isomers from each other reference is made to IUPAC Rules Section E (Pure Appl Chem 45, 11-30, 1976).
  • the present invention includes all possible stereoisomers of the compounds of the present invention as single stereoisomers, or as any mixture of said stereoisomers, e.g. (R)- or (S)- isomers, in any ratio.
  • Isolation of a single stereoisomer, e.g. a single enantiomer or a single diastereomer, of a compound of the present invention is achieved by any suitable state of the art method, such as chromatography, especially chiral chromatography, for example.
  • the compounds of the present invention can exist as tautomers.
  • the present invention includes all possible tautomers of the compounds of the present invention as single tautomers, or as any mixture of said tautomers, in any ratio.
  • the compounds of the present invention can exist as N-oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised.
  • the present invention includes all such possible N-oxides.
  • the present invention also covers useful forms of the compounds of the present invention, such as metabolites, hydrates, solvates, salts, in particular pharmaceutically acceptable salts, and/or co- precipitates.
  • the compounds of the present invention can exist as a hydrate, or as a solvate, wherein the compounds of the present invention contain polar solvents, in particular water, methanol or ethanol for example, as structural element of the crystal lattice of the compounds. It is possible for the amount of polar solvents, in particular water, to exist in a stoichiometric or non-stoichiometric ratio.
  • polar solvents in particular water
  • stoichiometric solvates e.g. a hydrate, hemi-, (semi-), mono-, sesqui-, di-, tri-, tetra-, penta- etc. solvates or hydrates, respectively, are possible.
  • the present invention includes all such hydrates or solvates. Hydrates are preferred solvates in the context of the present invention.
  • 3,3,3-trifluoro-2-oxopropyl derivatives of the formula (TB) according to the invention may also be present in the 3,3,3-trifluoro-2,2-dihydroxypropyl form (TB)' (hydrate form) (see Scheme 1 below); both forms are expressly embraced by the present invention.
  • the compounds of the present invention may exist in free form, e.g. as a free base, or as a free acid, or as a zwitterion, or to exist in the form of a salt.
  • Said salt may be any salt, either an organic or inorganic addition salt, particularly any pharmaceutically acceptable organic or inorganic addition salt, which is customarily used in pharmacy, or which is used, for example, for isolating or purifying the compounds of the present invention.
  • pharmaceutically acceptable salt refers to an inorganic or organic acid addition salt of a compound of the present invention.
  • pharmaceutically acceptable salt refers to an inorganic or organic acid addition salt of a compound of the present invention.
  • S. M. Berge, et al. “Pharmaceutical Salts,” J. Pharm. Sci. 1977, 66, 1-19.
  • a suitable pharmaceutically acceptable salt of the compounds of the present invention may be, for example, an acid-addition salt of a compound of the present invention bearing a nitrogen atom, in a chain or in a ring, for example, which is sufficiently basic, such as an acid-addition salt with an inorganic acid, or "mineral acid", such as hydrochloric, hydrobromic, hydroiodic, sulfuric, sulfamic, bisulfuric, phosphoric, or nitric acid, for example, or with an organic acid, such as formic, acetic, acetoacetic, pyruvic, trifluoroacetic, propionic, butyric, hexanoic, heptanoic, undecanoic, lauric, benzoic, salicylic, 2-(4-hydroxybenzoyl)-benzoic, camphoric, cinnamic, cyclopentanepropionic, digluconic, 3-hydroxy-2-naphthoic, nico
  • an alkali metal salt for example a sodium or potassium salt
  • an alkaline earth metal salt for example a calcium, magnesium or strontium salt, or an aluminium or a zinc salt
  • acid addition salts of the claimed compounds to be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods.
  • alkali and alkaline earth metal salts of acidic compounds of the present invention are prepared by reacting the compounds of the present invention with the appropriate base via a variety of known methods.
  • the present invention includes all possible salts of the compounds of the present invention as single salts, or as any mixture of said salts, in any ratio.
  • the present invention includes all possible crystalline forms, or polymorphs, of the compounds of the present invention, either as single polymorph, or as a mixture of more than one polymorph, in any ratio.
  • the present invention relates to compounds of formula (I), supra, wherein Ar represents a group of the formula
  • R 2A represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, methyl, ethyl, isopropyl, nitro, methoxy, difluoromethoxy and methylsulfanyl,
  • R 2B and R 2C represent, independently from each other, a group selected from a fluorine atom, a chlorine atom, methyl and ethyl,
  • R 2D , R 2E and R 2F represent, independently from each other, a group selected from a fluorine atom and a chlorine atom,
  • R 3A represents a group selected from a hydrogen atom, a fluorine atom, methyl, ethyl, methoxy and methoxycarbonyl,
  • R 3B represents a chlorine atom
  • R 3C represents a group selected from a chlorine atom and ethoxy
  • R 3D represents aminocarbonyl
  • R 3E represents a methyl group
  • R 3F represents a chlorine atom
  • R 3G represents a group selected from a fluorine atom, a chlorine atom and methyl
  • R 4A represents a methoxy group
  • the present invention relates to compounds according to formula (I), supra, wherein
  • R 1 represents a group of the formula
  • # l represents the point of attachment to the nitrogen atom
  • # l represents the point of attachment to the nitrogen atom
  • R 2A represents a group selected from a chlorine atom, a bromine atom, isopropyl, and nitro,
  • R 3G represents a group selected from a chlorine atom and methyl
  • the present invention covers compounds of general formula (I), supra, wherein
  • R 1 represents a group of the formula
  • Ar represents a group of the formula
  • # l represents the point of attachment to the nitrogen atom
  • R 2A represents a group selected from a hydrogen atom, a fluorine atom, and methylsulfanyl
  • R 2B and R 2C represent, independently from each other, a group selected from a fluorine atom, a chlorine atom, methyl and ethyl,
  • R 2F represents a group selected from a fluorine atom and a chlorine atom
  • R 3A represents a group selected from a hydrogen atom, a fluorine atom and ethyl
  • the present invention covers compounds of general formula (I), supra, wherein
  • R 1 represents a (2S)-3,3,3-trifluoro-2-hydroxypropyl group of the formula
  • # l represents the point of attachment to the nitrogen atom
  • R 1 represents a (2R)-3,3,3-trifluoro-2-hydroxypropyl group of the formula
  • # l represents the point of attachment to the nitrogen atom
  • the present invention covers combinations of two or more of the above mentioned embodiments under the heading "further embodiments of the first aspect of the present invention”.
  • the present invention covers any sub-combination within any embodiment or aspect of the present invention of compounds of general formula (I), supra.
  • the present invention covers any sub-combination within any embodiment or aspect of the present invention of intermediate compounds of general formula (II), (III), (IV), (V), (VI), (VII), (VIII), (X), (XIII), (XIV) and (XV).
  • the present invention covers the compounds of general formula (I) which are disclosed in the Example Section of this text, infra.
  • the present invention covers methods of preparing compounds of general formula (I) as defined supra, said methods comprising the step
  • R 1 is as defined for the compound of general formula (I) as defined supra, and R 5 represents a (Ci-C4)-alkyl group, in particular a methyl group,
  • R 6 represents a (Ci-C -alkyl group, in particular a methyl group
  • R 6 represents a (Ci-C -alkyl group, in particular a methyl group
  • each [A], [B] and [C] optionally followed, where appropriate, by (i) separating the compounds of formula (I) thus obtained into their respective enantiomers, and/or (ii) converting the compounds of formula (I) into their respective hydrates, solvates, salts and/or hydrates or solvates of the salts by treatment with the corresponding solvents and/or acids or bases.
  • the present invention covers methods of preparing compounds of the present invention of general formula (I), said methods comprising the steps as described in the Experimental Section herein.
  • the multicomponent cyclization (II)— (V) is carried out by first reacting imidate of formula (II) with an acid chloride of formula (III) in the presence of a base to form an intermediate which is in a subsequent step reacted with the aryl hydrazine compound of formula (IV). Typically the formed intermediate is not isolated and the reaction over the two steps is performed in one-pot.
  • the arylhydrazine compound for formula (I) may also be used in form of its salts, such as a hydrochloride salt or a tosylate salt. Under the alkaline reaction conditions, the hydrazine salt will be reconverted into the free base form. The amount of base added may then be adjusted in this respect.
  • a copper or zinc salt such as copper(II) sulfate, copper(II) chloride, zinc(II) sulfate and zinc(II) chloride.
  • copper(II) sulfate or zinc(II) sulfate are used.
  • Suitable bases for both steps are typically tertiary amine bases, such as /V,/V-diisopropylethylamine (DIPEA), triethylamine, triisopropylamine, -mefhylimidazole, -mefhylmorpholine, pyridine and 4-(A i ,/V-dimethylamino)pyridine.
  • DIPEA tertiary amine bases
  • the reaction is performed in an inert organic solvent, such as dichloromethane, 1,2-dichloroethane, methyl ieri-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxy ethane, toluene, pyridine, ethyl acetate, acetonitrile or A ⁇ -dimefhylformamide, or in a mixture of these solvents.
  • tetrahydrofuran or dioxane or a mixture thereof are used as solvents.
  • the first step is generally carried out at a temperature in the range of -10°C to +120°C, preferably at 0°C.
  • the second step is generally carried out at a temperature in the range of +20°C to +120°C, preferably at room temperature.
  • Concomitant microwave irradiation may have a beneficial effect in this reaction as well at a temperature in the range of +60°C to +150°C , preferably at +120°C.
  • the aminolysis reaction (V) — (I) is usually carried out in a solution of ammonia.
  • Suitable ammonia solutions for this step are saturated ammonia solutions, in particular a solution of ammonia in methanol, ethanol, isopropanol, tetrahydrofuran, dioxane or water or a mixture thereof.
  • a methanolic ammonia solution is used.
  • the reaction is preferably performed directly in the ammonia solution in the absence of any further reaction solvent.
  • This step is generally carried out at a temperature in the range of of +20°C to +120°C, preferably at room temperature. Concomitant microwave irradiation may have a beneficial effect in this reaction as well at a temperature in the range of +60°C to +150°C, preferably at +120°C.
  • the oxidation reaction (TA)— (TB) is carried out using customary oxidation methods known from the literature [e.g. JOC, 1983, 48, 4155 (Dess Martin oxidation); Tet Lett, 1994, 35,3485 (IBX oxidation); JOC, 1970, 35, 3589 (acid dichromate oxidation); Tet Lett, 1979, 399 (PDC oxidation); Tetrahedron, 1978, 34, 1651 (Swern oxidation)].
  • the alcohol group in the compounds of the general formula (I-A) is preferably oxidized using Dess-Martin periodinane (DMP). In a typical procedure the reaction is carried out in dichloromethane at a temperature of 0°C and subsequent warming up to room temperature.
  • DMP Dess-Martin periodinane
  • compounds of general formula (V) as defined supra may be prepared by a method comprising the step
  • R 1 is as defined for the compound of general formula (I) as defined supra.
  • R 6 represents a (Ci-C -alkyl group, in particular an ethyl group,
  • R ⁇ and R XB represent, independently from each other, a hydrogen atom, or
  • R ⁇ and R XB together form a -CH 2 CH 2 - or -C(CH 3 ) 2 C(CH 3 ) 2 - bridge,
  • R 1 and Ar are as defined for the compound of general formula (V):
  • R 6 represents a (Ci-C -alkyl group, in particular an ethyl group,
  • the coupling reaction (VI) + (VII)— > (V) is typically carried out with the aid of a copper catalyst and an amine base ["Chan-Lam coupling" conditions; see, for instance, D. M. T. Chan et al., Tetrahedron Lett. 44 (19), 3863-3865 (2003); J. X. Qiao and P. Y. S. Lam, Synthesis, 829-856 (2011); K. S. Rao and T.-S. Wu, Tetrahedron 68, 7735-7754 (2012)].
  • Copper catalysts suitable for this process are in particular copper(II) salts, such as copper(II) acetate, copper(II) trifluoromethane- sulfonate or copper(II) bromide.
  • Practical amine bases include, for example, triethylamine, N,N- diisopropylethylamine, pyridine and 4-(A r ,/V-dimethylamino)pyridine.
  • the reaction is performed in an inert organic solvent, such as dichloromethane, 1,2-dichloroethane, methyl ieri-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane, toluene, pyridine, ethyl acetate, acetonitrile or -dimethylformamide, or in a mixture of these solvents.
  • pyridine is used both as solvent and base.
  • the coupling is generally carried out at a temperature in the range of +20°C to +120°C, preferably at +20°C to +70°C. Concomitant microwave irradiation may have a beneficial effect in this reaction as well.
  • X represents a leaving group, such as chlorine, bromine, iodine, mesylate or tosylate, in particular chlorine or bromine,
  • R 1 is as defined for the compound of general formula (I) as defined supra,and
  • R 5 represents a (Ci-C -alkyl group, in particular a methyl group.
  • the /V-alkylation reaction (VIII) + (IX) -> (X) (step [a]) is typically carried out in the presence of a base.
  • Typical and exemplary bases include sodium carbonate, potassium carbonate, cesium carbonate, A ⁇ -diisopropylefhylamine, triethylamine, sodium ieri-butylate or potassium tert- butylate in acetonitrile, methylisobutylketone, dioxane, dimethylformamide, dimethylacetamide, N- methylpyrrolidinone, dimethylsulfoxide and sulfolane, preference is given to potassium carbonate in methylisobutylketone or acetonitrile.
  • the reaction may optionally be carried out in an advantageous manner with addition of an alkylation catalyst such as, for example, lithium bromide, sodium iodide, lithium iodide, tetra-n-butylammoniumbromide, tetra-n-butylammoniumiodide or benzyltriethylammoniumchloride.
  • an alkylation catalyst such as, for example, lithium bromide, sodium iodide, lithium iodide, tetra-n-butylammoniumbromide, tetra-n-butylammoniumiodide or benzyltriethylammoniumchloride.
  • the reactions are generally carried out in a temperature range of from +40°C to +120°C, preferably at from +60°C to +80°C.
  • the reactions can be carried out at atmospheric, at elevated or at reduced pressure (for example at from 0.5 to 5 bar); in general, the reactions are carried out at atmospheric
  • step [b] (X)— (II)).
  • the reaction is typically carried out under basic reactions conditions by reacting with a basic alcoholate.
  • Typical bases which may be used are sodium methanolate, sodium ethanolate, sodium propanolate, sodium isopropoxide, sodium ieri-butylate or potassium ieri-butylate in methanol, ethanol, n-propanol, isopropanol, n- butanol, isobutanol and tert-butanol.
  • Preference is given to sodium methanolate in methanol.
  • the reactions are generally carried out in a temperature range of from +20 to +80 °C, preferably at from +40 to +60 °C.
  • nitrile compounds of general formula (X) may optionally also be prepared as shown in the synthetic scheme 2 below:
  • the amide coupling (XI)— (XII) can be carried out directly with the help of a condensing agent or activating agent in the presence of a base or over two steps via an acyl chloride or carboxylic acid imidazolide.
  • Typical condensation and activating agents for the amide formation in process steps (XI)— (XII) include, for example, carbodiimides such as ⁇ , ⁇ '-diethyl-, N, N'-dipropyl-, ⁇ , ⁇ '- diisopropyl-N,N'-dicyclohexylcarbodiimide (DCC) or N-(3-dimethylaminoisopropyl)-N'-ethyl- carbodiimide hydrochloride (EDC), phosgene derivatives such as N, N'-carbonyldiimidazole (CDI), 1,2-oxazolium compounds such as 2-ethyl-5-phenyl-l,2-oxazolium-3-sulphate or 2-tert- butyl-5-methyl-isoxazolium perchlorate, acylamino compounds such as 2-ethoxy-l-ethoxy- carbonyl-l,2-dihydroquinoline, or iso
  • Typical and exemplary bases include sodium carbonate, potassium carbonate, cesium carbonate, ⁇ , ⁇ -diisopropylethylamine, triethylamine, sodium tert-butylate or potassium tert-butylate in acetonitrile, methylisobutylketone, dioxane, dimethylformamide, dimethylacetamide, N-methyl- pyrrolidinone, dimethylsulfoxide and sulfolane, preference is given to potassium carbonate in methylisobutylketone or acetonitrile.
  • the reaction may optionally be carried out in an advantageous manner with addition of an alkylation catalyst such as, for example, lithium bromide, sodium iodide, lithium iodide, tetra-n-butylammoniumbromide, tetra-n-butylammoniumiodide or benzyltriethylammoniumchloride.
  • an alkylation catalyst such as, for example, lithium bromide, sodium iodide, lithium iodide, tetra-n-butylammoniumbromide, tetra-n-butylammoniumiodide or benzyltriethylammoniumchloride.
  • Typical dehydrating agents include, for example trifluoroacetic acid anhydride, phosphorous pentoxide (P4O10), phosphoryl chloride (POCI3), phosphorous penta- chloride (PC ), CCI4-PPI13 (Appel reagent), hexamethylphosphoramide (HMPA); methyl N- (triethylammoniumsulfonyl)carbamate (Burgess reagent), (Chloromethylene)dimethyliminium chloride (Vilsmeier reagent), oxalyl chloride/DMSO and thionylchloride (SOCI2).
  • Typical and exemplary solvents for both steps (XI)— (XII) and (XII)— > (X) include for example, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane or mineral oil, fractions, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, 1,2-dichloroethane, trichloroethylene or chlorobenzene, or othersolvents such as acetone, ethyl acetate, acetonitrile, pyridine,dimethyl sulphoxide, ⁇ , ⁇ -dimethylformamide, ⁇ , ⁇ '- dimethylpropyleneurea (DMPU) or N-methylpyrrolidone (NMP
  • the carboxylic acid (XI) is first reacted with pivaloyl chloride in the presence of pyridine to form an intermediate which is in a subsequent step reacted with ammonia.
  • the formed intermediate is not isolated and the reaction over the two steps is performed in one-pot.
  • Suitable as bases for the first step are preferably, pyridine, 4-(N,N-dimethyl- amino)pyridine or ⁇ , ⁇ -diisopropylethylamine (DIPEA).
  • DIPEA ⁇ , ⁇ -diisopropylethylamine
  • the conversion of carboxamide (XII) into nitrile (X) is then typically performed by reaction with trifluoroacetic anhydride. Both reactions are conducted in an inert organic solvent, preferably tetrahydrofuran.
  • R 1 is as defined for the compound of general formula (I) as defined supra, which is then being condensed with a (thiooxo)amino compound of formula (XV)
  • R 6 represents a (Ci-C -alkyl group, in particular an ethyl group,
  • R 6 represents a (Ci-C -alkyl group, in particular an ethyl group,
  • transformation (XIII)— > (XIV) is carried out in the usual way by treating methyl ester (XIII) with hydrazine or hydrazine hydrate in an alcoholic solvent, such as methanol, ethanol, n-propanol, isopropanol or n-butanol, at a temperature in the range of +20°C to +100°C.
  • an alcoholic solvent such as methanol, ethanol, n-propanol, isopropanol or n-butanol
  • the condensation reaction (XIV) + (XV)— (VI) is usually carried out in an inert dipolar-aprotic solvent, such as -dimefhylformamide (DMF), /V-dimefhylacetamide (DMA), dimethylsulf- oxide (DMSO), /V-mefhylpyrrolidinone (NMP) or /V,/V'-dimefhylpropylene urea (DMPU), in the presence of a sufficiently strong base, such as sodium hydride or a sodium or potassium alkoxide, for example sodium or potassium methoxide, sodium or potassium ethoxide, or sodium or potassium ieri-butoxide.
  • a sufficiently strong base such as sodium hydride or a sodium or potassium alkoxide, for example sodium or potassium methoxide, sodium or potassium ethoxide, or sodium or potassium ieri-butoxide.
  • the (thiooxo) amine (XV) may be employed as such in this reaction or in salt form, e.g. as the hydrochloride salt. In the latter case, a proportional excess of base is used.
  • the reaction is generally performed at a temperature between +80°C and +150°C. Heating by means of a microwave reactor device may have a beneficial effect for this condensation reaction.
  • the 1,2,4-triazole derivative of formula (VI) produced by this reaction may also be present in other tautomeric forms, such as (VI-A) or (VI-B)
  • the compounds of the formulae (III), (IV) and (VII) are either commercially available, known from the literature, or can be prepared from readily available starting materials by adaptation of standard methods described in the literature. Detailed procedures and literature references for preparing the starting materials can also be found in the Experimental Part in the section on the preparation of the starting materials and intermediates.
  • the compounds of general formula (I) of the present invention can be converted to any salt, preferably pharmaceutically acceptable salts, as described herein, by any method which is known to the person skilled in the art.
  • any salt of a compound of general formula (I) of the present invention can be converted into the free compound, by any method which is known to the person skilled in the art.
  • the compounds of the present invention have valuable pharmacological properties and can be used for the prevention and/or treatment of various diseases and disease-induced states in humans and other mammals.
  • Compounds of general formula (I) of the present invention demonstrate a valuable pharmacological spectrum of action and pharmacokinetic profile, both of which could not have been predicted.
  • Compounds of the present invention have surprisingly been found to effectively inhibit the vasopressin Via receptor and it is possible therefore that said compounds be used for the treatment and/or prevention of diseases, preferably renal and cardiovascular diseases in humans and animals.
  • treatment includes inhibiting, delaying, relieving, mitigating, arresting, reducing, or causing the regression of a disease, disorder, condition, or state, the development and/or progression thereof, and/or the symptoms thereof.
  • prevention includes reducing the risk of having, contracting, or experiencing a disease, disorder, condition, or state, the development and/or progression thereof, and/or the symptoms thereof.
  • prevention includes prophylaxis. Treatment or prevention of a dis- order, disease, condition, or state may be partial or complete.
  • the compounds of the present invention are potent selective or dual antagonists of vasopressin Via and V2 receptors.
  • the compounds of the invention are therefore expected to be highly valuable as therapeutic agents for the treatment and/or prevention of diseases, in particular for the treatment and/or prevention of cardiovascular and renal diseases.
  • the compounds according to the invention are suitable for the treatment and/or prevention of renal diseases, in particular of acute and chronic kidney diseases, diabetic kidney diseases, and of acute and chronic renal failure.
  • the general terms 'renal disease' or 'kidney disease' describe a class of conditions in which the kidneys fail to filter and remove waste products from the blood.
  • kidney disease acute kidney disease (acute kidney injury, AKI) and chronic kidney disease (CKD).
  • the compounds according to the invention may further be used for the treatment and/or prevention of sequelae of acute kidney injury arising from multiple insults such as ischemia-reperfusion injury, radiocontrast administration, cardiopulmonary bypass surgery, shock and sepsis.
  • renal failure or renal insufficiency comprises both acute and chronic manifestations of renal insufficiency, as well as underlying or related kidney diseases such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathies, IgA nephropathy, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial diseases, nephropathic diseases such as primary and congenital kidney disease, nephritis, Alport syndrome, kidney inflammation, immunological kidney diseases such as kidney transplant rejection, immune complex-induced kidney diseases, nephropathy induced by toxic substances, contrast medium-induced nephropathy; minimal change glomerulonephritis (lipoid); Membranous glomerulonephritis; focal segmental glomerulosclerosis (FSGS); hemolytic uremic syndrome (HUS), amyloidosis, Goodpasture's syndrome, Wegener's gran
  • kidney diseases such as renal hypoper
  • the present invention also comprises the use of the compounds according to the invention for the treatment and/or prevention of sequelae of renal insufficiency, for example pulmonary edema, heart failure, uraemia, anaemia, electrolyte disturbances (e.g. hyperkalemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism.
  • the compounds according to the invention are also suitable for the treatment and/or prevention of polycystic kidney disease (PCKD) and of the syndrome of inadequate ADH secretion (SIADH).
  • PCKD polycystic kidney disease
  • SIADH syndrome of inadequate ADH secretion
  • Cardiovascular diseases in this context include, but are not limited to, the following: acute and chronic heart failure including worsening chronic heart failure (or hospitalization for heart failure) and including congestive heart failure, arterial hypertension, resistant hypertension, arterial pulmonary hypertension, coronary heart disease, stable and unstable angina pectoris, atrial and ventricular arrhythmias, disturbances of atrial and ventricular rhythm and conduction disturbances, for example atrioventricular blocks of degree I-III (AVB I-III), supraventricular tachyarrhythmia, atrial fibrillation, atrial flutter, ventricular fibrillation, ventricular flutter, ventricular tachyarrhythmia, torsade-de-pointes tachycardia, atrial and ventricular extrasystoles, AV-junction extrasystoles, sick-sinus syndrome, syncopes, AV- node re-entry tachycardia and Wolff-Parkinson
  • heart failure also includes more specific or related disease forms such as right heart failure, left heart failure, global insufficiency, ischemic cardiomyopathy, dilatative cardiomyopathy, congenital heart defects, heart valve defects, heart failure with heart valve defects, mitral valve stenosis, mitral valve insufficiency, aortic valve stenosis, aortic valve insufficiency, tricuspidal stenosis, tricuspidal insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined heart valve defects, heart muscle inflammation (myocarditis), chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcohol- toxic cardiomyopathy, cardiac storage diseases, heart failure with preserved ejection fraction (HFpEF or diastolic heart failure), and heart failure with reduced ejection fraction (HFrEF or systolic heart failure).
  • HFpEF preserved ejection fraction
  • HFrEF reduced
  • renal insufficiency comprises both acute and chronic manifestations of renal insufficiency, as well as underlying or related kidney diseases such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathies, IgA nephropathy, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial diseases, nephropathic diseases such as primary and congenital kidney disease, nephritis, immunological kidney diseases such as kidney transplant rejection, immune complex-induced kidney diseases, nephropathy induced by toxic substances, contrast medium-induced nephropathy, diabetic and non-diabetic nephropathy, pyelonephritis, renal cysts, nephrosclerosis, hyperten
  • the present invention also comprises the use of the compounds according to the invention for the treatment and/or prevention of sequelae of renal insufficiency, for example pulmonary edema, heart failure, uraemia, anaemia, electrolyte disturbances (e.g. hyperkalaemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism.
  • sequelae of renal insufficiency for example pulmonary edema, heart failure, uraemia, anaemia, electrolyte disturbances (e.g. hyperkalaemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism.
  • the compounds of the present invention may be particularly useful for the treatment and/or preven- tion of the cardiorenal syndrome (CRS) and its various subtypes.
  • CRS cardiorenal syndrome
  • This term embraces certain disorders of the heart and kidneys whereby acute or chronic dysfunction in one organ may induce acute or chronic dysfunction of the other.
  • CRS has been sub-classified into five types based upon the organ that initiated the insult as well as the acuity and chronicity of the disease (type 1 : development of renal insufficiency resulting from acute decompensated heart failure; type 2: chronic congestive heart failure resulting in progressive renal dysfunction; type 3: acute cardiac dysfunction resulting from an abrupt fall in renal function; type 4: chronic kidney disease leading to cardiac remodeling; type 5: systemic disease involving both the heart and the kidneys) [see, for example, M. R. Kahn et al., Nature Rev. Cardiol. 10, 261-273 (2013)].
  • the compounds according to the invention are also suitable for the treatment and/or prevention of polycystic kidney disease (PCKD) and of the syndrome of inadequate ADH secretion (SIADH). Furthermore, the compounds of the invention are suitable for use as a diuretic for the treatment of edemas and in electrolyte disorders, in particular in hypervolemic and euvolemic hyponatremia.
  • PCKD polycystic kidney disease
  • SIADH syndrome of inadequate ADH secretion
  • the compounds according to the invention may be used for the treatment and/or prevention of peripheral arterial disease (PAD) including claudication and including critical limb ischemia, coronary microvascular dysfunction (CMD) including CMD type 1-4, primary and secondary Raynaud's phenomenon, microcirculation disturbances, claudication, peripheral and autonomic neuropathies, diabetic microangiopathies, diabetic retinopathy, diabetic limb ulcers, gangrene, CREST syndrome, erythematous disorders, onychomycosis, rheumatic diseases and for promoting wound healing.
  • PAD peripheral arterial disease
  • CMD coronary microvascular dysfunction
  • CMD including CMD type 1-4
  • primary and secondary Raynaud's phenomenon microcirculation disturbances
  • claudication peripheral and autonomic neuropathies
  • diabetic microangiopathies diabetic retinopathy
  • diabetic limb ulcers diabetic limb ulcers
  • gangrene gangrene
  • CREST syndrome erythematous disorders
  • the compounds of the invention are suitable for treating urological diseases and diseases of the male and female urogenital system such as, for example, benign prostatic syndrome (BPS), benign prostatic hyperplasia (BPH), benign prostatic enlargement (BPE), bladder outlet obstruction (BOO), lower urinary tract syndromes (LUTS), neurogenic overactive bladder (OAB), interstitial cystitis (IC), urinary incontinence (UI), such as, for example, mixed, urge, stress and overflow incontinence (MUI, UUI, SUI, OUI), pelvic pains, erectile dysfunction, dysmenorrhea and endometriosis.
  • BPS benign prostatic syndrome
  • BPH benign prostatic hyperplasia
  • BPE benign prostatic enlargement
  • BOO bladder outlet obstruction
  • LUTS lower urinary tract syndromes
  • IC neurogenic overactive bladder
  • UI urinary incontinence
  • MUI mixed, urge, stress and overflow incontinence
  • UUI UUI
  • SUI
  • the compounds according to the invention may also be used for the treatment and/or prevention of inflammatory diseases, asthmatic diseases, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), acute lung injury (ALI), alpha- 1 -antitrypsin deficiency (AATD), pulmonary fibrosis, pulmonary emphysema (e.g. smoking-induced pulmonary emphysema) and cystic fibrosis (CF).
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress syndrome
  • ALI acute lung injury
  • AATD alpha- 1 -antitrypsin deficiency
  • pulmonary fibrosis pulmonary emphysema (e.g. smoking-induced pulmonary emphysema) and cystic fibrosis (CF).
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress syndrome
  • ALI acute lung injury
  • AATD alpha- 1 -
  • the compounds of the invention may be used for the treatment and/or prevention of pulmonary arterial hypertension (PAH) and other forms of pulmonary hypertension (PH), including pulmonary hypertension associated with left ventricular disease, HIV infection, sickle cell anaemia, thromboembolism (CTEPH), sarcoidosis, chronic obstructive pulmonary disease (COPD) or pulmonary fibrosis.
  • PAH pulmonary arterial hypertension
  • PH pulmonary hypertension associated with left ventricular disease
  • HIV infection sickle cell anaemia
  • thromboembolism CTEPH
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • the compounds according to the invention may be used for the treatment and/or prevention of liver cirrhosis, ascites, diabetes mellitus and diabetic complications such as, for example, neuropathy and nephropathy.
  • the compounds of the invention are suitable for the treatment and/or prevention of central nervous disorders such as anxiety states, depression, glaucoma, cancer, such as in particular pulmonary tumors, and circadian rhythm misalignment such as jet lag and shift work.
  • central nervous disorders such as anxiety states, depression, glaucoma, cancer, such as in particular pulmonary tumors, and circadian rhythm misalignment such as jet lag and shift work.
  • the compounds according to the invention may be useful for the treatment and/or prevention of pain conditions, diseases of the adrenals such as, for example, pheochromocytoma and adrenal apoplexy, diseases of the intestine such as, for example, Crohn's disease and diarrhea, menstrual disorders such as, for example, dysmenorrhea, endometriosis, preterm labor and tocolysis.
  • diseases of the adrenals such as, for example, pheochromocytoma and adrenal apoplexy
  • diseases of the intestine such as, for example, Crohn's disease and diarrhea
  • menstrual disorders such as, for example, dysmenorrhea, endometriosis, preterm labor and tocolysis.
  • the compounds of the present invention are believed to be particularly suitable for the treatment and/or prevention of acute and chronic kidney diseases including diabetic nephropathy, acute and chronic heart failure, preeclampsia, peripheral arterial disease (PAD), coronary microvascular dysfunction (CMD), Raynaud's syndrome, dysmenorrhea, cardiorenal syndrome, hypervolemic and euvolemic hyponatremia, liver cirrhosis, ascites, edema and the syndrome of inadequate ADH secretion (SIADH).
  • acute and chronic kidney diseases including diabetic nephropathy, acute and chronic heart failure, preeclampsia, peripheral arterial disease (PAD), coronary microvascular dysfunction (CMD), Raynaud's syndrome, dysmenorrhea, cardiorenal syndrome, hypervolemic and euvolemic hyponatremia, liver cirrhosis, ascites, edema and the syndrome of inadequate ADH secretion (SIADH).
  • the present invention further relates to the use of the compounds according to the invention for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
  • the present invention further relates to the use of the compounds according to the invention for preparing a pharmaceutical composition for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
  • the present invention further relates to the use of the compounds according to the invention in a method for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
  • the present invention further relates to a method for the treatment and/or prevention of diseases, especially of the aforementioned diseases, by using an effective amount of at least one of the compounds according to the invention.
  • the present invention covers pharmaceutical combinations, in particular medicaments, comprising at least one compound of general formula (I) of the present invention and at least one or more further active ingredients, in particular for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
  • the present invention covers a pharmaceutical combination, which comprises:
  • a "fixed combination” in the present invention is used as known to persons skilled in the art and is defined as a combination wherein, for example, a first active ingredient, such as one or more compounds of general formula (I) of the present invention, and a further active ingredient are present together in one unit dosage or in one single entity.
  • a “fixed combination” is a pharmaceutical composition wherein a first active ingredient and a further active ingredient are present in admixture for simultaneous administration, such as in a formulation.
  • Another example of a "fixed combination” is a pharmaceutical combination wherein a first active ingredient and a further active ingredient are present in one unit without being in admixture.
  • a non-fixed combination or "kit-of-parts" in the present invention is used as known to persons skilled in the art and is defined as a combination wherein a first active ingredient and a further active ingredient are present in more than one unit.
  • a non-fixed combination or kit- of-parts is a combination wherein the first active ingredient and the further active ingredient are present separately.
  • the components of the non-fixed combination or kit-of-parts can be administered separately, sequentially, simultaneously, concurrently or chronologically staggered.
  • the compounds of the present invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutically active ingredients where the combination causes no unacceptable adverse effects.
  • the present invention also covers such pharmaceutical combinations.
  • the compounds of the present invention can be combined with known agents for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
  • the compounds of the present invention may be used in fixed or separate combination with
  • antithrombotic agents for example and preferably from the group of platelet aggregation inhibitors, anticoagulants and profibrinolytic substances;
  • ⁇ blood pressure lowering agents for example and preferably from the group of calcium antagonists, angiotensin All antagonists, ACE inhibitors, NEP inhibitors, vasopeptidase inhibitors, endothelin antagonists, renin inhibitors, alpha-blockers, beta-blockers, mineralocorticoid receptor antagonists and diuretics;
  • antidiabetic agents hyperglycemic or antihyperglycemic agents
  • insulin and derivatives such as for example and preferably insulin and derivatives, sulfonylureas, biguanides, thiazolidinediones, acarbose,
  • DPP4 inhibitors DPP4 inhibitors, GLP-1 analogues, or SGLT inhibitors (gliflozins).
  • organic nitrates and NO-donors for example sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhalational NO;
  • cGMP cyclic guano sine monophosphate
  • PDE phosphodiesterases
  • sildenafil, vardenafil, tadalafil, udenafil, dasantafil, avanafil, mirodenafil, lodenafil CTP-499 or PF-00489791;
  • natriuretic peptides such as for example atrial natriuretic peptide (ANP, anaritide), B-type natriuretic peptide or brain natriuretic peptide (BNP, nesiritide), C-type natriuretic peptide (CNP) or urodilatin;
  • ARP atrial natriuretic peptide
  • BNP B-type natriuretic peptide or brain natriuretic peptide
  • CNP C-type natriuretic peptide
  • urodilatin urodilatin
  • sGC soluble guanylate cyclase
  • sGC guanylate cyclase
  • agents that stimulates the synthesis of cGMP, for example and with preference sGC modulators, for example and with preference riociguat, cinaciguat, vericiguat or BAY 1101042;
  • HNE human neutrophil elastase
  • ⁇ compounds inhibiting the signal transduction cascade in particular tyrosine and/or serine/threonine kinase inhibitors, such as for example nintedanib, dasatinib, nilotinib, bosutinib, regora- fenib, sorafenib, sunitinib, cediranib, axitinib, telatinib, imatinib, brivanib, pazopanib, vatalanib, gefitinib, erlotinib, lapatinib, canertinib, lestaurtinib, pelitinib, semaxanib or tandutinib;
  • tyrosine and/or serine/threonine kinase inhibitors such as for example nintedanib, dasatinib, nilotinib, bosutinib, regora- fen
  • cardiac myosin activators such as for example and preferably omecamtiv mecarbil (CK- 1827452);
  • anti-inflammatory drugs such as non-steroidal anti-inflammatory drugs (NSAIDs) including acetylsalicylic acid (aspirin), ibuprofen and naproxen, glucocorticoids, NEP inhibitors, 5- aminosalicylic acid derivatives, leukotriene antagonists, TNF-alpha inhibitors and chemokine receptor antagonists such as CCR1, 2 and/or 5 inhibitors;
  • NSAIDs non-steroidal anti-inflammatory drugs
  • acetylsalicylic acid aspirin
  • ibuprofen and naproxen glucocorticoids
  • NEP inhibitors 5- aminosalicylic acid derivatives
  • leukotriene antagonists such as TNF-alpha inhibitors
  • chemokine receptor antagonists such as CCR1, 2 and/or 5 inhibitors
  • ⁇ fat metabolism altering agents for example and preferably from the group of thyroid receptor agonists, cholesterol synthesis inhibitors, such as for example and preferably HMG-CoA- reductase or squalene synthesis inhibitors, ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors and lipopro- tein(a) antagonists.
  • cholesterol synthesis inhibitors such as for example and preferably HMG-CoA- reductase or squalene synthesis inhibitors, ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid adsorbers, bile acid
  • Antithrombotic agents are preferably to be understood as compounds from the group of platelet aggregation inhibitors, anticoagulants and profibrinolytic substances.
  • the compounds according to the invention are administered in combination with a platelet aggregation inhibitor, for example and preferably aspirin, clo- pidogrel, ticlopidine or dipyridamole.
  • a platelet aggregation inhibitor for example and preferably aspirin, clo- pidogrel, ticlopidine or dipyridamole.
  • the compounds according to the invention are administered in combination with a thrombin inhibitor, for example and preferably ximelagatran, dabiga- tran, melagatran, bivalirudin or enoxaparin.
  • a thrombin inhibitor for example and preferably ximelagatran, dabiga- tran, melagatran, bivalirudin or enoxaparin.
  • the compounds according to the invention are administered in combination with a GPIib/iIIa antagonist, for example and preferably tirofiban or abcixi- mab.
  • the compounds according to the invention are adminis- tered in combination with a factor Xa inhibitor, for example and preferably rivaroxaban, apixaban, otamixaban, fidexaban, razaxaban, fondaparinux, idraparinux, DU-176b, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
  • a factor Xa inhibitor for example and preferably rivaroxaban, apixaban, otamixaban, fidexaban, razaxaban, fondaparinux, idraparinux, DU-176b, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803,
  • the compounds according to the invention are adminis- tered in combination with heparin or a low molecular weight (LMW) heparin derivative.
  • LMW low molecular weight
  • the compounds according to the invention are administered in combination with a vitamin K antagonist, for example and preferably coumarin.
  • Blood pressure lowering agents are preferably to be understood as compounds from the group of calcium antagonists, angiotensin All antagonists, ACE inhibitors, NEP inhibitors, vasopeptidase inhibitors, endothelin antagonists, renin inhibitors, alpha-blockers, beta-blockers, mineralocorticoid receptor antagonists and diuretics.
  • the compounds according to the invention are administered in combination with a calcium antagonist, for example and preferably nifedipine, amlodipine, verapamil or diltiazem.
  • a calcium antagonist for example and preferably nifedipine, amlodipine, verapamil or diltiazem.
  • the compounds according to the invention are administered in combination with an alpha- 1 -receptor blocker, for example and preferably prazosin or tam- sulosin.
  • an alpha- 1 -receptor blocker for example and preferably prazosin or tam- sulosin.
  • the compounds according to the invention are administered in combination with a beta-blocker, for example and preferably propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipranolol, nadolol, mepindolol, carazolol, sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, carteolol, esmolol, labetalol, carve - dilol, adaprolol, landiolol, nebivolol, epanolol or bucindolol.
  • a beta-blocker for example and preferably propranolol, atenolol, timolol, pindolol, alpre
  • the compounds according to the invention are administered in combination with an angiotensin All receptor antagonist, for example and preferably losar- tan, candesartan, valsartan, telmisartan, irbesartan, olmesartan, eprosartan, embursartan or azilsartan.
  • angiotensin All receptor antagonist for example and preferably losar- tan, candesartan, valsartan, telmisartan, irbesartan, olmesartan, eprosartan, embursartan or azilsartan.
  • the compounds according to the invention are administered in combination with a vasopeptidase inhibitor or inhibitor of neutral endopeptidase (NEP), such as for example and preferably sacubitril, omapatrilat or AVE-7688.
  • NEP neutral endopeptidase
  • the compounds according to the invention are administered in combination with a dual angiotensin All receptor antagonist/NEP inhibitor (ARNI), for example and preferably LCZ696.
  • ARNI angiotensin All receptor antagonist/NEP inhibitor
  • the compounds according to the invention are adminis- tered in combination with an ACE inhibitor, for example and preferably enalapril, captopril, lisino- pril, ramipril, delapril, fosinopril, quinopril, perindopril, benazepril or trandopril.
  • an ACE inhibitor for example and preferably enalapril, captopril, lisino- pril, ramipril, delapril, fosinopril, quinopril, perindopril, benazepril or trandopril.
  • the compounds according to the invention are administered in combination with an endothelin antagonist, for example and preferably bosentan, darusen- tan, ambrisentan, tezosentan, sitaxsentan, avosentan, macitentan or atrasentan.
  • an endothelin antagonist for example and preferably bosentan, darusen- tan, ambrisentan, tezosentan, sitaxsentan, avosentan, macitentan or atrasentan.
  • the compounds according to the invention are administered in combination with a renin inhibitor, for example and preferably aliskiren, SPP-600 or SPP- 800.
  • a renin inhibitor for example and preferably aliskiren, SPP-600 or SPP- 800.
  • the compounds according to the invention are administered in combination with a mineralocorticoid receptor antagonist, for example and preferably fine- renone, spironolactone, canrenone, potassium canrenoate, eplerenone, esaxerenone (CS-3150), or apararenone (MT-3995), CS-3150, or MT-3995.
  • a mineralocorticoid receptor antagonist for example and preferably fine- renone, spironolactone, canrenone, potassium canrenoate, eplerenone, esaxerenone (CS-3150), or apararenone (MT-3995), CS-3150, or MT-3995.
  • the compounds according to the invention are administered in combination with a diuretic, such as for example and preferably furosemide, bumetanide, piretanide, torsemide, bendroflumethiazide, chlorothiazide, hydrochlorothiazide, xipamide, indapa- mide, hydroflumethiazide, methyclothiazide, polythiazide, trichloromethiazide, chlorothalidone, metolazone, quinethazone, acetazolamide, dichlorophenamide, methazolamide, glycerine, isosor- bide, mannitol, amiloride or triamterene.
  • a diuretic such as for example and preferably furosemide, bumetanide, piretanide, torsemide, bendroflumethiazide, chlorothiazide, hydrochlorothiazide, xipamide, ind
  • Fat metabolism altering agents are preferably to be understood as compounds from the group of CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA- reductase or squalene synthesis inhibitors, ACAT inhibitors, MTP inhibitors, PPAR-alpha, PPAR- gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors, lipase inhibitors and lipoprotein(a) antagonists.
  • cholesterol synthesis inhibitors such as HMG-CoA- reductase or squalene synthesis inhibitors
  • ACAT inhibitors MTP inhibitors
  • MTP inhibitors PPAR-alpha, PPAR- gamma and/or PPAR-delta agonists
  • cholesterol absorption inhibitors polymeric bile acid adsorbers
  • bile acid reabsorption inhibitors lipase inhibitors and lipoprotein(a)
  • the compounds according to the invention are administered in combination with a CETP inhibitor, for example and preferably dalcetrapib, anacetrapib, BAY 60-5521 or CETP-vaccine (Avant).
  • a CETP inhibitor for example and preferably dalcetrapib, anacetrapib, BAY 60-5521 or CETP-vaccine (Avant).
  • the compounds according to the invention are administered in combination with a thyroid receptor agonist, for example and preferably D-thyroxin, 3,5,3'- triiodothyronin (T3), COS 23425 or axitirome (COS 26214).
  • a thyroid receptor agonist for example and preferably D-thyroxin, 3,5,3'- triiodothyronin (T3), COS 23425 or axitirome (COS 26214).
  • the compounds according to the invention are administered in combination with an HMG-CoA-reductase inhibitor from the class of statins, for example and preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pravastatin.
  • the compounds according to the invention are administered in combination with a squalene synthesis inhibitor, for example and preferably BMS- 188494 or TAK-475.
  • a squalene synthesis inhibitor for example and preferably BMS- 188494 or TAK-475.
  • the compounds according to the invention are administered in combination with an ACAT inhibitor, for example and preferably avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
  • an ACAT inhibitor for example and preferably avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
  • the compounds according to the invention are administered in combination with an MTP inhibitor, for example and preferably implitapide, R- 103757, BMS-201038 or JTT-130.
  • an MTP inhibitor for example and preferably implitapide, R- 103757, BMS-201038 or JTT-130.
  • the compounds according to the invention are adminis- tered in combination with a PPAR-gamma agonist, for example and preferably pioglitazone or rosi- glitazone.
  • the compounds according to the invention are administered in combination with a PPAR-delta agonist, for example and preferably GW 501516 or BAY 68-5042.
  • the compounds according to the invention are administered in combination with a cholesterol absorption inhibitor, for example and preferably ezetimibe, tiqueside or pamaqueside.
  • a cholesterol absorption inhibitor for example and preferably ezetimibe, tiqueside or pamaqueside.
  • the compounds according to the invention are administered in combination with a lipase inhibitor, for example and preferably orlistat.
  • the compounds according to the invention are administered in combination with a polymeric bile acid adsorber, for example and preferably cholestyramine, colestipol, colesolvam, CholestaGel or colestimide.
  • a polymeric bile acid adsorber for example and preferably cholestyramine, colestipol, colesolvam, CholestaGel or colestimide.
  • ASBT IBAT
  • the compounds according to the invention are administered in combination with a lipoprotein(a) antagonist, for example and preferably gemcabene calcium (CI- 1027) or nicotinic acid.
  • a lipoprotein(a) antagonist for example and preferably gemcabene calcium (CI- 1027) or nicotinic acid.
  • the compounds according to the invention are administered in combination with a TGFbeta antagonist, by way of example and with preference pirfenidone or fresolimumab.
  • the compounds according to the invention are administered in combination with HIF-PH inhibitors, by way of example and with preference molidustat or roxadustat.
  • the compounds according to the invention are administered in combination with a CCR2 antagonist, by way of example and with preference CCX-140.
  • the compounds according to the invention are administered in combination with a TNFalpha antagonist, by way of example and with preference adalimumab.
  • the compounds according to the invention are administered in combination with a galectin-3 inhibitor, by way of example and with preference GCS-100.
  • the compounds according to the invention are administered in combination with a BMP-7 agonist, by way of example and with preference THR- 184.
  • the compounds according to the invention are administered in combination with a NOX1/4 inhibitor, by way of example and with preference GKT-137831.
  • the compounds according to the invention are administered in combination with a medicament which affects the vitamin D metabolism, by way of example and with preference cholecalciferol or paracalcitol.
  • the compounds according to the invention are administered in combination with a cytostatic agent, by way of example and with preference cyclophosphamide.
  • the compounds according to the invention are administered in combination with an immunosuppressive agent, by way of example and with preference ciclosporin.
  • the compounds according to the invention are administered in combination with a phosphate binder, by way of example and with preference sevelamer or lanthanum carbonate.
  • the compounds according to the invention are administered in combination with a calcimimetic for therapy of hyperparathyroidism.
  • the compounds according to the invention are administered in combination with agents for iron deficit therapy, by way of example and with preference iron products.
  • the compounds according to the invention are administered in combination with agents for the therapy of hyperurikaemia, by way of example and with preference allopurinol or rasburicase.
  • the compounds according to the invention are administered in combination with glycoprotein hormone for the therapy of anaemia, by way of example and with preference erythropoietin.
  • the compounds according to the invention are administered in combination with biologies for immune therapy, by way of example and with preference abatacept, rituximab, eculizumab or belimumab.
  • the compounds according to the invention are administered in combination with Jak inhibitors, by way of example and with preference ruxolitinib, tofacitinib, baricitinib, CYT387, GSK2586184, lestaurtinib, pacritinib (SB1518) or TG101348.
  • Jak inhibitors by way of example and with preference ruxolitinib, tofacitinib, baricitinib, CYT387, GSK2586184, lestaurtinib, pacritinib (SB1518) or TG101348.
  • the compounds according to the invention are administered in combination with prostacyclin analogs for therapy of microthrombi.
  • the compounds according to the invention are administered in combination with an alkali therapy, by way of example and with preference sodium bicarbonate.
  • the compounds according to the invention are administered in combination with an mTOR inhibitor, by way of example and with preference everolimus or rapamycin.
  • the compounds according to the invention are administered in combination with an NHE3 inhibitor, by way of example and with preference AZD1722.
  • the compounds according to the invention are administered in combination with an eNOS modulator, by way of example and with preference sapropterin.
  • the compounds according to the invention are administered in combination with a CTGF inhibitor, by way of example and with preference FG- 3019.
  • the compounds according to the invention are administered in combination with antidiabetics (hypoglycemic or antihyperglycemic agents), such as for example and preferably insulin and derivatives, sulfonylureas such as tolbutamide, carbutamide, acetohexamide, chlorpropamide, glipizide, gliclazide, glibenclamide, glyburide, glibornuride, gliquidone, glisoxepide, glyclopyramide, glimepiride, JB253 and JB558, meglitinides such as repaglinide and nateglinide, biguanides such as metformin and buformin, thiazolidinediones such as rosiglitazone and pioglitazone, alpha-glucosidase inhibitors such as miglitol, acarbose and voglibose, DPP4 inhibitors such as
  • the compounds of the present invention are administered in combination with one or more additional therapeutic agents selected from the group consisting of diuretics, angiotensin All antagonists, ACE inhibitors, beta-receptor blockers, mineralocorticoid receptor antagonists, antidiabetics, organic nitrates and NO donors, activators and stimulators of the soluble guanylate cyclase (sGC), and positive-inotropic agents.
  • additional therapeutic agents selected from the group consisting of diuretics, angiotensin All antagonists, ACE inhibitors, beta-receptor blockers, mineralocorticoid receptor antagonists, antidiabetics, organic nitrates and NO donors, activators and stimulators of the soluble guanylate cyclase (sGC), and positive-inotropic agents.
  • the compounds of the present invention are administered in combination with one or more additional therapeutic agents selected from the group consisting of diuretics, angiotensin All antagonists, ACE inhibitors, beta-receptor blockers, mineralocorticoid receptor antagonists, antidiabetics, organic nitrates and NO donors, activators and stimulators of the soluble guanylate cyclase (sGC), positive-inotropic agents, antiinflammatory agents, immunosuppressive agents, phosphate binders and/or compounds which modulate vitamin D metabolism.
  • additional therapeutic agents selected from the group consisting of diuretics, angiotensin All antagonists, ACE inhibitors, beta-receptor blockers, mineralocorticoid receptor antagonists, antidiabetics, organic nitrates and NO donors, activators and stimulators of the soluble guanylate cyclase (sGC), positive-inotropic agents, antiinflammatory agents, immunosuppressive agents, phosphate binders and/or compounds which modulate vitamin D metabolism.
  • the present invention relates to pharmaceutical compositions comprising at least one of the compounds according to the invention and one or more additional therapeutic agents for the treatment and/or prevention of diseases, especially of the aforementioned dis- eases.
  • the compounds of the present invention may be utilized, as such or in compositions, in research and diagnostics, or as analytical reference standards and the like, which are well known in the art.
  • the compounds of the present invention are administered as pharmaceuticals, to humans and other mammals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1% to 99.5% (more preferably, 0.5% to 90%) of active ingredient in combination with one or more pharmaceutically acceptable excipients.
  • the present invention relates to pharmaceutical compositions comprising at least one of the compounds according to the invention, conventionally together with one or more inert, non-toxic, pharmaceutically acceptable excipients, and to the use thereof for the treatment and/ or prevention of diseases, especially of the aforementioned diseases.
  • the compounds according to the invention can be administered in a suitable manner, such as, for example, via the oral, parenteral, pulmonary, nasal, sublingual, lingual, buccal, rectal, vaginal, dermal, transdermal, conjunctival, otic route or as an implant or stent.
  • the compounds according to the invention for oral administration, it is possible to formulate the compounds according to the invention to dosage forms known in the art that deliver the compounds of the invention rapidly and/or in a modified manner, such as, for example, tablets (uncoated or coated tablets, for example with enteric or controlled release coatings that dissolve with a delay or are insoluble), orally- disintegrating tablets, films/wafers, films/lyophylisates, capsules (for example hard or soft gelatine capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions. It is possible to incorporate the compounds according to the invention in crystalline and/or amorphised and/or dissolved form into said dosage forms.
  • Parenteral administration can be effected with avoidance of an absorption step (for example intravenous, intraarterial, intracardial, intraspinal or intralumbal) or with inclusion of absorption (for example intramuscular, subcutaneous, intracutaneous, percutaneous or intraperitoneal).
  • absorption step for example intravenous, intraarterial, intracardial, intraspinal or intralumbal
  • absorption for example intramuscular, subcutaneous, intracutaneous, percutaneous or intraperitoneal.
  • Administration forms which are suitable for parenteral administration are, inter alia, preparations for injection and infusion in the form of solutions, suspensions, emulsions, lyophylisates or sterile powders.
  • Examples which are suitable for other administration routes are pharmaceutical forms for inhalation [inter alia powder inhalers, nebulizers], nasal drops, nasal solutions, nasal sprays; tablets/films/wafers/capsules for lingual, sublingual or buccal administration; suppositories; eye drops, eye ointments, eye baths, ocular inserts, ear drops, ear sprays, ear powders, ear-rinses, ear tampons; vaginal capsules, aqueous suspensions (lotions, mixturae agitandae), lipophilic suspensions, emulsions, ointments, creams, transdermal therapeutic systems (such as, for example, patches), milk, pastes, foams, dusting powders, implants or stents.
  • the compounds according to the invention can be incorporated into the stated administration forms. This can be effected in a manner known per se by mixing with pharmaceutically suitable excipients.
  • Pharmaceutically suitable excipients include,
  • fillers and carriers for example cellulose, microcrystalline cellulose (such as, for example, Avicel ® ), lactose, mannitol, starch, calcium phosphate (such as, for example, Di-Cafos ® )),
  • ointment bases for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols
  • ointment bases for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols
  • bases for suppositories for example polyethylene glycols, cacao butter, hard fat
  • solvents for example water, ethanol, isopropanol, glycerol, propylene glycol, medium chain-length triglycerides fatty oils, liquid polyethylene glycols, paraffins
  • surfactants for example sodium dodecyl sulfate
  • lecithin for example sodium dodecyl sulfate
  • phospholipids for example sodium dodecyl sulfate
  • fatty alcohols such as, for example, Lanette ®
  • sorbitan fatty acid esters such as, for example, Span ®
  • polyoxyethylene sorbitan fatty acid esters such as, for example, Tween ®
  • polyoxyethylene fatty acid glycerides such as, for example,
  • Cremophor ® polyoxethylene fatty acid esters, polyoxyethylene fatty alcohol ethers, glycerol fatty acid esters, poloxamers (such as, for example, Pluronic ® ),
  • buffers for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine
  • acids and bases for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine
  • isotonicity agents for example glucose, sodium chloride
  • adsorbents for example highly-disperse silicas
  • viscosity-increasing agents for example, gel formers, thickeners and/or binders (for example
  • polyvinylpyrrolidone methylcellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose-sodium, starch, carbomers, polyacrylic acids (such as, for example, Carbopol ® ); alginates, gelatine),
  • disintegrants for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate (such as, for example, Explotab ® ), cross- linked polyvinylpyrrolidone, croscarmellose-sodium (such as, for example, AcDiSol ® )
  • disintegrants for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate (such as, for example, Explotab ® ), cross- linked polyvinylpyrrolidone, croscarmellose-sodium (such as, for example, AcDiSol ® )
  • lubricants for example magnesium stearate, stearic acid, talc, highly-disperse silicas (such as, for example, Aerosil ® )
  • mould release agents for example magnesium stearate, stearic acid, talc, highly-disperse silicas (such as, for example, Aerosil ® )
  • coating materials for example sugar, shellac
  • film formers for films or diffusion membranes which dissolve rapidly or in a modified manner
  • polyvinylpyrrolidones such as, for example, Kollidon ®
  • polyvinyl alcohol such as, for example, polyvinyl alcohol
  • hydroxypropylmethylcellulose hydroxypropylcellulose, ethylcellulose, hydroxypropylmethylcellulose phthalate, cellulose acetate, cellulose acetate phthalate, polyacrylates, polymethacrylates such as, for example, Eudragit ® )),
  • capsule materials for example gelatine, hydroxypropylmethylcellulose
  • polymethacrylates such as, for example, Eudragit ®
  • polyvinylpyrrolidones such as, for example, Kollidon ®
  • polyvinyl alcohols such as, for example, polyvinyl acetates, polyethylene oxides, polyethylene glycols and their copolymers and blockcopolymers
  • plasticizers for example polyethylene glycols, propylene glycol, glycerol, triacetine, triacetyl citrate, dibutyl phthalate
  • stabilisers for example antioxidants such as, for example, ascorbic acid, ascorbyl palmitate, sodium ascorbate, butylhydroxyanisole, butylhydroxytoluene, propyl gallate
  • antioxidants such as, for example, ascorbic acid, ascorbyl palmitate, sodium ascorbate, butylhydroxyanisole, butylhydroxytoluene, propyl gallate
  • preservatives for example parabens, sorbic acid, thiomersal, benzalkonium chloride, chlorhexidine acetate, sodium benzoate
  • ⁇ colourants for example inorganic pigments such as, for example, iron oxides, titanium dioxide
  • flavourings • flavourings, sweeteners, flavour- and/or odour-masking agents.
  • the present invention furthermore relates to a pharmaceutical composition which comprise at least one compound according to the invention, conventionally together with one or more pharmaceutically suitable excipient(s), and to their use according to the present invention.
  • the effective dosage of the compounds of the present invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
  • the total amount of the active ingredient to be administered will generally range from about 0.001 mg kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 20 mg/kg body weight per day.
  • Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing.
  • drug holidays in which a patient is not dosed with a drug for a certain period of time, to be beneficial to the overall balance between pharmacological effect and tolerability. It is possible for a unit dosage to contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day.
  • the average daily dosage for administration by injection will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the compound of the present invention may be administered parenterally at a dose of about 0.001 mg/kg to about 10 mg/kg, preferably of about 0.01 mg/kg to about 1 mg/kg of body weight.
  • an exemplary dose range is about 0.01 to 100 mg/kg, preferably about 0.01 to 20 mg/kg, and more preferably about 0.1 to 10 mg/kg of body weight. Ranges intermediate to the above-recited values are also intended to be part of the invention.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like.
  • the desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
  • ⁇ -NMR data are listed in the form of ⁇ -NMR peaklists. For each signal peak the ⁇ value in ppm is given, followed by the signal intensity, reported in round brackets. The ⁇ value-signal intensity pairs from different peaks are separated by commas. Therefore, a peaklist is described by the general form: ⁇ (intensityi), 82 (intensity2), ... , ⁇ (intensity;), ... , ⁇ ⁇ (intensity n ).
  • the intensity of a sharp signal correlates with the height (in cm) of the signal in a printed NMR spectrum. When compared with other signals, this data can be correlated to the real ratios of the signal intensities. In the case of broad signals, more than one peak, or the center of the signal along with their relative intensity, compared to the most intense signal displayed in the spectrum, are shown.
  • a ⁇ -NMR peaklist is similar to a classical ⁇ -NMR readout, and thus usually contains all the peaks listed in a classical NMR interpretation.
  • peaklists can show solvent signals, signals derived from stereoisomers of target compounds (also the subject of the invention), and/or peaks of impurities.
  • the peaks of stereoisomers, and/or peaks of impurities are typically displayed with a lower intensity compared to the peaks of the target compounds (e.g., with a purity of >90%).
  • Such stereoisomers and/or impurities may be typical for the particular manufacturing process, and therefore their peaks may help to identify the reproduction of our manufacturing process on the basis of "by-product fingerprints".
  • An expert who calculates the peaks of the target compounds by known methods can isolate the peaks of target compounds as required, optionally using additional intensity filters. Such an operation would be similar to peak-picking in classical ⁇ -NMR interpretation.
  • Chemical names were generated using the ACD/Name software from ACD/Labs. In some cases generally accepted names of commercially available reagents were used in place of ACD/Name generated names.
  • NMR nuclear magnetic resonance spectroscopy chemical shifts ( ⁇ ) are given in ppm. The chemical shifts were corrected by setting the DMSO signal to 2.50 ppm unless otherwise stated.
  • the compounds and intermediates produced according to the methods of the invention may require purification. Purification of organic compounds is well known to the person skilled in the art and there may be several ways of purifying the same compound. In some cases, no purification may be necessary. In some cases, the compounds may be purified by crystallization. In some cases, impurities may be stirred out using a suitable solvent. In some cases, the compounds may be purified by chromatography, particularly flash column chromatography, using for example prepacked silica gel cartridges, e.g.
  • the compounds may be purified by preparative HPLC using for example a Waters autopurifier equipped with a diode array detector and/or on-line electrospray ionization mass spectrometer in combination with a suitable prepacked reverse phase column and eluents such as gradients of water and acetonitrile which may contain additives such as trifluoroacetic acid, formic acid or aqueous ammonia.
  • purification methods as described above can provide those compounds of the present invention which possess a sufficiently basic or acidic functionality in the form of a salt, such as, in the case of a compound of the present invention which is sufficiently basic, a trifluoroacetate or formate salt for example, or, in the case of a compound of the present invention which is sufficiently acidic, an ammonium salt for example.
  • a salt of this type can either be transformed into its free base or free acid form, respectively, by various methods known to the person skilled in the art, or be used as salts in subsequent biological assays. It is to be understood that the specific form (e.g. salt, free base etc.) of a compound of the present invention as isolated and as described herein is not necessarily the only form in which said compound can be applied to a biological assay in order to quantify the specific biological activity.
  • Instrument MS Thermo Scientific FT-MS; Instrument type UHPLC+: Thermo Scientific UltiMate 3000; Column: Waters, HSST3, 2.1 x 75 mm, C18 1.8 ⁇ ; eluent A: 1 L water + 0.01% formic acid; eluent B: 1 L acetonitrile + 0.01% formic acid; gradient: 0.0 min 10% B ⁇ 2.5 min 95% B ⁇ 3.5 min 95% B; oven: 50°C; flow rate: 0.90 ml/min; UV detection: 210 nm/ optimum integration path 210-300 nm
  • the resulting mixture was refluxed for 3 h and cooled down to room temperature.
  • the reaction mixture was diluted with IN HCl (70 ml).
  • the organic phase was washed twice IN HCl.
  • the aqueous phase was extracted twice with ethyl acetate.
  • the combined organic phases were evaporated.
  • the residue was retaken in methanol (22.5 ml) and the resulting suspension was heated to 60°C until the solid was completely dissolved.
  • IN HCl (22.5 ml) were added and the resulting suspension was heated at 78°C for 10 min and cooled down to room temperature.
  • the solid was filtered off and dried under vacuum.
  • the solid was retaken in IN HCl (30 ml), heated at 35°C.
  • the resulting solution was treated at 5 °C with 52.8 g (438 mmol) of 2,2-dimethylpropanoylchloride over 15 minutes and the resulting mixture was stirred at room temperature for 2.5 hours.
  • 183 ml of 28% aqueous ammonia solution was added over 1 h while the solution temperature was kept between 10 °C and 20 °C and at the resulting mixture then stirred at 5 °C for an additional time period of 1 h.
  • 500 ml methyl tert-butylether and 300 ml 20% aqueous citric acid were then added while keeping the internal temperature between 10 °C and 20 °C.
  • the phases were the separated and the organic phase was washed with 300 ml of 20% aqueous citric acid followed by 300 ml saturated aqueous sodium hydrogencarbonate solution and finally with 300 ml of 10% aqueous sodium chloride solution.
  • the organic phase was evaporated at 60 °C under reduced pressure until an oily residue was obtained.
  • 300 ml THF was then added and the solution was evaporated again until an oily solution was obtained. This operation was repeated a second time.
  • the oil residue was retaken in 360 ml THF and treated with 172 g (820 mmol) trifluoroacetic acid anhydride over 20 min at a temperature between 10 °C and 20 °C. The resulting solution was then stirred at room temperature for 1 h.
  • the formed suspension was cooled to 20 °C and a solid formed which was filtered off and washed with 200 ml n-heptane and then dried under reduced pressure (50°C, 30 mbar) affording 88 g (93 % of th.) of ⁇ 3-(4-chlorophenyl)-5-oxo-4-[(2 l S')-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-l i- l,2,4-triazol-l-yl ⁇ acetonitrile as a solid.
  • Ethyl 2-chloropyridine-3-carboxylate (1.00 g, 5.39 mmol) was dissolved in hydrazine in THF (22 ml, 1M, 22 mmol) and stirred 16h at room temperature. Hydrazine in THF (11 ml, lM, 11 mmol) was added and the resulting mixture was stirred 24h at room temperature. The reaction mixture was evaporated. The residue was retaken in 10% MeOH in CHCI 3 and washed with an aqueous K 2 CO 3 solution. The aqueous phase was extracted with 10% MeOH in CHCI 3 .
  • Example 25A Methyl 3-( ⁇ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihyd] 1 ,2,4-triazol- 1 -yl ⁇ methyl)- l-(2-ethoxypyridin-3-yl)- 1H- 1 ,2,4-triazole-5-carboxylate
  • Example 33A Methyl l-(2-chloro-6-fluorophenyl)-3-( ⁇ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl ⁇ methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
  • Example 4A a solution of methyl 2- ⁇ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl ⁇ ethanimidate (Example 4A, 150 mg, 0.40 mmol) of Example 3A in 3 ml anhydrous THF was treated at 0°C with 75 ⁇ (0.44 mmol) N V- diisopropylethylamine and 40 ⁇ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min.
  • the title compound can also be synthesized via the procedure described in WO 2011/104322-A1 (Example 7A).
  • reaction mixture was stirred at 60 °C overnight, after which boronic acid (400 mg, 2.86 mmol) was added. After stirring for 6 additional days at room temperature, the reaction mixture was concentrated in vacuo and then diluted with ethyl acetate. The reaction mixture was then quenched with aqueous hydrochloric acid (0.5 M). After phase separation, the aqueous phase was extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • the crude product was first purified by flash chromatography (silica gel, eluent dichloromethane/methanol) then by preparative HPLC (Chromatorex CI 8, ⁇ , 125x30mm, water-acetonitrile-gradient 0.05% trifluoroacetic acid) affording 242 mg (9 % of th.)of the desired compound.
  • reaction mixture was stirred at 60 °C overnight, after which boronic acid (1.30 g, 8.30 mmol) was added due to incomplete conversion.
  • the reaction mixture was further stirred at 60 °C for 2 days. Over this time, additional portion of boronic acid (1.30 g, 8.30 mmol) was added.
  • the reaction mixture was concentrated in vacuo and then diluted with ethyl acetate. After this, the reaction mixture was then quenched with aqueous hydrochloric acid (0.5 M). After phase separation, the aqueous phase was extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • reaction mixture was stirred at 60 °C overnight, after which boronic acid (148 mg, 0.98 mmol) was added. After stirring for two additional days, the reaction mixture was diluted with ethyl acetate and then quenched with aqueous hydrochloric acid (1 M). After phase separation, the aqueous phase was extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated in vacuo.
  • Example 4A a solution of methyl 2- ⁇ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl ⁇ ethanimidate (Example 4A, 10 g, 26.40 mmol) of Example 3A in 200 ml anhydrous THF was treated at 0°C with 5.1 ml (29.1 mmol) N,N- diisopropylethylamine and 2.67 ml (29.0 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min.
  • Example 19A 50.0 mg, 83.9 ⁇
  • 7N NH 3 solution in MeOH 1.3 ml, 9.1 mmol
  • the residue was evaporated and purified by preparative HPLC (Method 6) affording 27.8 mg (57 % of th.) of the title compound.
  • Example 26A 100 mg, 90 % purity, 151 ⁇
  • 7N NH 3 solution in MeOH 9.0 ml, 63 mmol
  • the residue was evaporated and purified by preparative HPLC (Method 6) affording 44mg (52% of th.) of the title compound.
  • Demonstration of the activity of the compounds of the present invention may be accomplished through in vitro, ex vivo, and in vivo assays that are well known in the art. For example, to demonstrate the activity of the compounds of the present invention, the following assays may be used.
  • the identification of agonists and antagonists of the Via and V2 vasopressin receptors from humans, rats and dogs as well as the quantification of the activity of the compounds of the invention is carried out using recombinant cell lines. These cell lines originally derive from a hamster's ovary epithelial cell (Chinese Hamster Ovary, CHO Kl, ATCC: American Type Culture Collec- tion, Manassas, VA 20108, USA). The test cell lines constitutively express the human, rat or dog Via or V2 receptors.
  • cells are also stably transfected with a modified form of the calcium- sensitive photoproteins aequorin (human and rat Via) or obe- lin (dog Via), which, after reconstitution with the cof actor coelenterazine, emit light when there are increases in free calcium concentrations [Rizzuto R, Simpson AW, Brini M, Pozzan T, Nature 358, 325-327 (1992); Illarionov BA, Bondar VS, Illarionova VA, Vysotski ES, Gene 153 (2), 273- 274 (1995)] .
  • aequorin human and rat Via
  • obe- lin dog Via
  • the resulting vasopressin receptor cells react to stimulation of the recombinantly expressed Via receptors by intracellular release of calcium ions, which can be quantified by the resulting photoprotein luminescence.
  • the G s -coupled V2 receptors are stably transfected into cell lines expressing the gene for firefly luciferase under control of a CRE-responsible promoter. Acti- vation of V2 receptors induces the activation of the CRE-responsive promoter via cAMP increase, thereby inducing the expression of firefly luciferase.
  • the light emitted by photoproteins of Via cell lines as well as the light emitted by firefly luciferase of V2 cell lines corresponds to the activation or inhibition of the respective vasopressin receptor.
  • the bioluminescence of the cell lines is detected using a suitable luminometer [Milligan G, Marshall F, Rees S, Trends in Pharmacological Sciences 17, 235-237 (1996)] .
  • the cells are plated out in culture medium (DMEM/F12, 2% FCS, 2 mM glutamine, 10 mM HEPES, 5 ⁇ g/ml coelenterazine) in 384-well microtiter plates and kept in a cell incubator (96% humidity, 5% v/v CO 2 , 37°C).
  • culture medium DMEM/F12, 2% FCS, 2 mM glutamine, 10 mM HEPES, 5 ⁇ g/ml coelenterazine
  • test compounds in various concentrations are placed for 10 minutes in the wells of the microtiter plate before the agonist [Arg 8 ] -vasopressin at EC50 concentration is added. The resulting light signal is measured immediately in a luminometer.
  • Vasopressin V2 receptor cell lines are vascularlypressin V2 receptor cell lines.
  • the cells are plated out in culture medium (DMEM/F12, 2% FCS, 2 mM glutamine, 10 mM HEPES) in 384-well microtiter plates and kept in a cell incubator (96% humidity, 5% v/v CO 2 , 37°C).
  • test compounds in various concentrations and the agonist [Arg 8 ] -vasopressin at EC50 concentration are added together to the wells, and plates are incubated for 3 hours in a cell incubator.
  • TritonTM and the substrate luciferin luminescence of firefly luciferase is measured in a luminometer.
  • Table 1A lists individual IC 50 values for the compounds of the invention (including racemic mixtures as well as separated enantiomers) that were obtained from cell lines transfected with the human Via or V2 receptor:

Abstract

The present invention relates to novel 5-(carboxamide)-1-aryl-1,2,4-triazole derivatives, to processes for the preparation of such compounds, to pharmaceutical compositions containing such compounds, and to the use of such compounds or compositions for the treatment and/or prevention of diseases, in particular for the treatment and/or prevention of renal and cardiovascular diseases.

Description

AMIDE-SUBSTITUTED ARYLTRIAZOLE DERIVATIVES AND USES THEREOF
The present invention relates to novel 5-(carboxamide)-l-aryl-l,2,4-triazole derivatives, to processes for the preparation of such compounds, to pharmaceutical compositions containing such compounds, and to the use of such compounds or compositions for the treatment and/or prevention of diseases, in particular for the treatment and/or prevention of renal and cardiovascular diseases.
The liquid content of the human body is subject to various physiological control mechanisms, the purpose of which is to keep it constant (volume homeostasis). In the process, both the volume filling of the vascular system and also the osmolarity of the plasma are continuously recorded by appropriate sensors (baroreceptors and osmoreceptors). The information which these sensors supply to the relevant centers in the brain regulates drinking behaviour and controls fluid excretion via the kidneys by means of humoral and neural signals. The peptide hormone vasopressin is of central importance in this [Schrier R.W., Abraham W.T., New Engl. J. Med. 341, 577-585 (1999)].
Vasopressin is produced in specialized endocrine neurons in the Nucleus supraopticus and N. para- ventricularis in the wall of the third ventricle (hypothalamus) and is transported from there along the neural processes into the posterior lobes of the hypophysis (neurohypophysis). There the hormone is released into the bloodstream in response to stimulus. A loss of volume, e.g. as a result of acute bleeding, heavy sweating, prolonged thirst or diarrhoea, is a stimulus for intensified release of the hormone. Conversely, the secretion of vasopressin is inhibited by an increase in the intravascular volume, e.g. as a result of increased fluid intake.
Vasopressin exerts its action mainly via binding to three receptors, which are classified as Via, Vlb and V2 receptors and which belong to the family of G protein-coupled receptors. Via receptors are mainly located on the cells of the vascular smooth musculature. Their activation gives rise to vasoconstriction, as a result of which the peripheral resistance and blood pressure rise. Apart from this, Via receptors are also detectable in the liver. Vlb receptors (also named V3 receptors) are detectable in the central nervous system. Together with corticotropin-releasing hormone (CRH), vasopressin regulates the basal and stress-induced secretion of adrenocorticotropic hormone (ACTH) via the Vlb receptor. V2 receptors are located in the distal tubular epithelium and the epithelium of the collecting tubules in the kidney. Their activation renders these epithelia permeable to water. This phenomenon is due to the incorporation of aquaporins (special water channels) in the luminal membrane of the epithelial cells.
The importance of vasopressin for the reabsorption of water from the urine in the kidney becomes clear from the clinical picture of diabetes insipidus, which is caused by a deficiency of the hormone, e.g. owing to hypophysis damage. Patients who suffer from this disease excrete up to 20 liters of urine per 24 hours if they are not given replacement hormone. This volume corresponds to about 10% of the primary urine. Because of its great importance for the reabsorption of water from the urine, vasopressin is also synonymously referred to as antidiuretic hormone (ADH). Consequently, pharmacological inhibition of the action of vasopressin/ ADH on the V2 receptor results in increased urine excretion. In contrast to the action of other diuretics (thiazides and loop diuretics), however, V2 receptor antagonists cause increased water excretion, without substantially increasing the excretion of electrolytes. This means that with V2 antagonist drugs, volume homeostasis can be restored without affecting electrolyte homeostasis. Hence, drugs with V2 antagonistic activity appear particularly suitable for the treatment of all disease conditions which are associated with an overloading of the body with water, without the electrolytes being adequately increased in parallel.
A significant electrolyte abnormality is measurable in clinical chemistry as hyponatremia (sodium concentration < 135 mmol/L); it is the most important electrolyte abnormality in hospital patients, with an incidence of about 5% or 250 000 cases per year in the US alone. If the plasma sodium concentration falls below 115 mmol/L, comatose states and death are imminent. Depending on the underlying cause, a distinction is made between hypovolemic, euvolemic and hypervolemic hyponatremia. The forms of hypervolemia with edema formation are clinically significant. Typical examples of these are the syndrome of inappropriate ADH/vasopressin secretion (SIADH) (e.g. after craniocerebral trauma or as paraneoplasia in carcinomas) and hypervolemic hyponatremia in liver cirrhosis, various renal diseases and heart failure [De Luca L. et al., Am. J. Cardiol. 96 (suppl.), 19L-23L (2005)]. In particular, patients with heart failure, in spite of their relative hyponatremia and hypervolemia, often display elevated vasopressin levels, which are seen as the conse- quence of a generally disturbed neurohumoral regulation in heart failure [Francis G.S. et al., Circulation 82, 1724-1729 (1990)].
The disturbed neurohormonal regulation essentially manifests itself in an elevation of the sympathetic tone and inappropriate activation of the renin-angiotensin-aldosterone system. While the inhibition of these components by beta-receptor blockers on the one hand and by ACE inhibitors or angiotensin-receptor blockers on the other is now an inherent part of the pharmacological treatment of heart failure, the inappropriate elevation of vasopressin secretion in advanced heart failure is at present still not adequately treatable. Apart from the retention of water mediated by V2 receptors and the unfavourable hemodynamic consequences associated therewith in terms of increased backload, the emptying of the left ventricle, the pressure in the pulmonary blood vessels and cardiac output are also adversely affected by V la-mediated vasoconstriction. Furthermore, on the basis of experimental data in animals, a direct hypertrophy-promoting action on the heart muscle is also attributed to vasopressin. In contrast to the renal effect of volume expansion, which is mediated by activation of V2 receptors, the direct action on the heart muscle is triggered by activation of VI a receptors.
For these reasons, agents which inhibit the action of vasopressin on the V2 and/or the Via receptor appear suitable for the treatment of heart failure. In particular, compounds with combined activity on both vasopressin receptors (Via and V2) should have both desirable renal as well as hemodynamic effects and thus offer an especially ideal profile for the treatment of patients with heart failure. The provision of such combined vasopressin antagonists also appears to make sense inasmuch as a volume diminution mediated solely via V2 receptor blockade can entail the stimulation of osmoreceptors and, as a result, may lead to a further compensatory increase in vasopressin release. Through this, in the absence of a component simultaneously blocking the Via receptor, the harmful effects of vasopressin, such as for example vasoconstriction and heart muscle hypertrophy, could be further intensified [Saghi P. et al., Europ. Heart J. 26, 538-543 (2005)].
Via receptors are mainly located on vascular smooth muscle cells (VSMC) but also on cardiomyocytes, fibroblasts and specialized renal cells like glomerular mesangial cells or cells of the macula densa which control the release of renin [Wasilewski MA, Myers VD, Recchia FA, Feldman AM, Tilley DG, Cell Signal., 28(3), 224-233, (2016)]. The activation of VSMC Via receptor by vasopressin gives rise to intracellular calcium release and according vasoconstriction. Therefore, stimulation of VSMC Via receptors causes increased vascular resistance and increased cardiac afterload. Cardiac output is adversely affected by V la-mediated vasoconstriction. The increase in afterload and direct stimulation of Via receptors on cardiomyocytes can lead to cardiac hypertrophy and remodeling including fibrosis. Mice with cardiac-specific overexpression of Via receptor develop cardiac hypertrophy leading to dilation and left ventricular dysfunction, suggesting an essential role for Via receptor in the development of heart failure [Li X, Chan TO, Myers V, Chowdhury I, Zhang XQ, Song J, Zhang J, Andrei J, Funakoshi H, Robbins J, Koch WJ, Hyslop T, Cheung JY, Feldman AM, Circulation.; 124, 572-581 (2011)].
Via receptor is also expressed in the renal cortical and medullary vasculature, where it mediates vasoconstriction of renal vessels and affecting overall renal blood flow. Thus, the activation of Via receptor can decrease renal medullary blood flow inducing further pathological processes as tissue hypoxia, reduced oxygen and according energy supply for tubular transport processes as well as direct damages of mesangial and macula densa cells. It has been demonstrated that mesangial Via receptor activation mediates TGFp signaling and causes an increase in production of collagen IV. While this signaling contributes extracellular matrix accumulation and remodeling in the kidney, similar signaling pathways are believed to occur in cardiac cells especially after myocardial infarction, which emphasizes the central role of Via receptor in the development of hypertrophic and fibrotic processes in response to pathophysiological elevated vasopressin levels [Wasilewski MA, Myers VD, Recchia FA, Feldman AM, Tilley DG. Arginine vasopressin receptor signaling and functional outcomes in heart failure. Cell Signal., 28(3), 224-233 (2016)] .
Since Via receptors are mainly expressed on VSMCs and thus participating in vascular function, a link to vascular diseases as peripheral arterial disease (PAD) including claudication and critical limb ischemia as well as coronary microvascular dysfunction (CMD) is conceivable. Apart from this, Via receptors are also expressed on human platelets and in the liver. The meaning of platelet Via receptors is not fully understood although vasopressin induces aggregation of human platelets via Via receptor at high concentrations ex vivo. Therefore, inhibition of vasopressin-induced platelet aggregation by Via receptor antagonists is a useful pharmacological ex vivo assay making use of human tissue endogenously expressing the Via receptor [Thibonnier M, Roberts JM, J Clin Invest.; 76:1857-1864, (1985)].
Vasopressin stimulates gluconeogenesis and glycogenolysis via activation of the hepatic Via receptor. Animal studies have shown that vasopressin impairs glucose tolerance which could be inhibited by a Via receptor antagonist thereby providing a link of vasopressin receptor Via to diabetes mellitus. [Taveau C, Chollet C, Waeckel L, Desposito D, Bichet DG, Arthus MF, Magnan C, Philippe E, Paradis V, Foufelle F, Hainault I, Enhorning S, Velho G, Roussel R, Bankir L, Melander O, Bouby N. Vasopressin and hydration play a major role in the development of glucose intolerance and hepatic steatosis in obese rats. Diabetologia., 58(5), 1081-1090, (2015)]. Vasopressin was shown to contribute to the development of albuminuria and to diabetes-induced nephropathy in animal models which is consistent with epidemiological findings in humans.
It was found recently that vasopressin also seems to play a causal role in the development of preeclampsia. Chronic infusion of vasopressin during pregnancy in mice is sufficient to induce all of the major maternal and fetal phenotypes associated with human preeclampsia, including pregnancy-specific hypertension [Santillan MK, Santillan DA, Scroggins SM, Min JY, Sandgren JA, Pearson NA, Leslie KK, Hunter SK, Zamba GK, Gibson-Corley KN, Grobe JL. Vasopressin in preeclampsia: a novel very early human pregnancy biomarker and clinically relevant mouse model. Hypertension. 64(4), 852-859, (2014)].
Vasopressin levels can be elevated in women with dysmenorrhoea (a gynecological disorder which is characterised by cyclical cramping pelvic pain) during menstruation, which appear to increase myometrial smooth muscle contraction. It was found recently that a selective vasopressin Via receptor antagonist (relcovaptan/SR-49059) can reduce intrauterine contractions elicited by vasopressin.
For these reasons, agents which inhibit the action of vasopressin on the Via receptor appear suitable for the treatment of several cardiovascular diseases. In particular, agents which inhibit the action of vasopressin selectively on the Via receptor offer an especially ideal profile for the treatment of otherwise normovolemic patients, i.e. those which are not eligible for decongestion by e.g. high doses of loop diuretics or V2 antagonists, and where induced aquaresis via V2 inhibition may be undesired. Certain 4-phenyl-l,2,4-triazol-3-yl derivatives have been described in WO 2005/063754-A1 and WO 2005/105779-A1 to act as vasopressin Via receptor antagonists that are useful for the treatment of gynecological disorders, notably menstrual disorders such as dysmenorrhea.
In WO 2011/104322-A1, a particular group of bis-aryl-bonded l,2,4-triazol-3-ones, including 5-phenyl-l,2,4-triazol-3-yl and 1 -phenyl- l,2,3-triazol-4-yl derivatives thereof, has been disclosed as antagonists of vasopressin Via and/or V2 receptors being useful for the treatment and/or prevention of cardiovascular diseases.
In WO 2016/071212-A1 certain 5-(hydroxyalkyl)-l-phenyl-l,2,4-triazole derivatives have been disclosed, which act as potent antagonists of both vasopressin Via and V2 receptors and, in addition, exhibit significantly enhanced aquaretic potency in vivo after oral application.
It was an object of the present invention to provide novel compounds which act as potent selective or dual Vla/V2 receptor antagonists and as such are suitable for the treatment and/or prevention of diseases, more particularly for the treatment and/or prevention of renal and cardiovascular disorders.
The compounds of the present invention have valuable pharmacological properties and can be used for the prevention and/or treatment of various diseases and disease-induced states in humans and other mammals.
In one aspect, the present invention relates to 5-(carboxamide)-l-aryl-l,2,4-triazole derivatives of the general formula (I)
(I),
in which
R1 represents a group of the formula
in which
represents the point of attachment to the nitrogen atom, Ar represents a phenyl group or a 5- or 6-membered heteroaryl group attached via a ring carbon atom having one or two ring heteroatoms selected from a nitrogen atom and a sulfur atom, wherein any phenyl group and any 5- or 6-membered heteroaryl group are each optionally substituted, identically or differently, with one or two groups selected from a halogen atom, nitro, cyano, (Ci-C -alkyl, (Ci-C -alkoxy, (Ci-C -alkylsulfanyl, (Ci-
C -alkoxycarbonyl, aminocarbonyl and
wherein said (Ci-C -alkyl group, said (Ci-C -alkoxy group and said (C1-C4)- alkylsulfanyl group are each optionally substituted with up to three fluorine atoms.
The compounds according to this invention can also be present in the form of their salts, solvates and/or solvates of the salts.
The term "substituted" means that one or more hydrogen atoms on the designated atom or group are replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded. Combinations of substituents and/or variables are permissible.
The term "optionally substituted" means that the number of substituents can be equal to or different from zero. Unless otherwise indicated, it is possible that optionally substituted groups are substituted with as many optional substituents as can be accommodated by replacing a hydrogen atom with a non-hydrogen substituent on any available carbon atom or heteroatom.
When groups in the compounds according to the invention are substituted, it is possible for said groups to be mono-substituted or poly-substituted with substituent(s), unless otherwise specified. Within the scope of the present invention, the meanings of all groups which occur repeatedly are independent from one another. It is possible that groups in the compounds according to the invention are substituted with one, two or three identical or different substituents.
The term "comprising" when used in the specification includes "consisting of.
If within the present text any item is referred to as "as mentioned herein", it means that it may be mentioned anywhere in the present text.
The terms as mentioned in the present text have the following meanings:
The term "5- to 6-membered heteroaryl" means a monovalent, monocyclic aromatic ring having 5 or 6 ring atoms, which contains at least one ring heteroatom and optionally one, two or three further ring heteroatoms from a nitrogen atom and a sulfur atom, and which is bound via a ring carbon atom or optionally via a ring nitrogen atom (if allowed by valency).
Said heteroaryl group can be a 5-membered heteroaryl group, such as, for example, thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl or tetrazolyl; or a 6-membered heteroaryl group, such as, for example, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl.
In general, and unless otherwise mentioned, the heteroaryl or heteroarylene groups include all possible isomeric forms thereof, e.g. : tautomers and positional isomers with respect to the point of linkage to the rest of the molecule. Thus, for some illustrative non-restricting examples, the term pyridinyl includes pyridin-2-yl, pyridin-3-yl and pyridin-4-yl; or the term thienyl includes thien-2-yl and thien-3-yl.
Particularly, the heteroaryl group in substituent Ar in the general formula (I), supra, is a pyridinyl, a pyrimidyl, or a pyrazinyl group.
The term "halogen atom" means a fluorine, chlorine, bromine or iodine atom, particularly a fluorine, or chlorine atom.
The term "C1-C4", as used in the present text, e.g. in the context of the definition of "Ci-C t-alkyl", "Ci-C t-alkoxy", " or "Ci-C t-alkylsulfanyl", means an alkyl group having a finite number of carbon atoms of 1 to 4, i.e. 1, 2, 3, or 4 carbon atoms.
The term "Ci-C t-alkyl" means a linear or branched, saturated, monovalent hydrocarbon group having 1, 2, 3, or 4 carbon atoms, e.g. a methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, isobutyl, ieri-butyl, or an isomer thereof. Particularly, said group has 1, 2, 3 or 4 carbon atoms ("Ci-C t-alkyl"), e.g. a methyl, ethyl, propyl, isopropyl, butyl, sec-butyl isobutyl, or ieri-butyl group, more particularly 1, 2 or 3 carbon atoms ("Ci-C3-alkyl"), e.g. a methyl, ethyl, n-propyl or isopropyl group, even more particularly a methyl group.
The term "Ci-C t-alkylsulfanyl" means a linear or branched, saturated, monovalent group of formula (Ci-C4-alkyl)-S-, in which the term "Ci-C4-alkyl" is as defined supra, e.g. a methylsulfanyl, ethylsulfanyl, propylsulfanyl, isopropylsulfanyl, butylsulfanyl, sec-butylsulfanyl, isobutylsulfanyl, ieri-butylsulfanyl group.
The term "Ci-C4-alkoxy" means a linear or branched, saturated, monovalent group of formula (Ci-C4-alkyl)-0-, in which the term "Ci-C4-alkyl" is as defined supra, e.g. a methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, isobutoxy, ieri-butoxy, or an isomer thereof.
It is possible for the compounds of general formula (I) to exist as isotopic variants. The invention therefore includes one or more isotopic variant(s) of the compounds of general formula (I), particularly deuterium-containing compounds of general formula (I).
The term "Isotopic variant" of a compound or a reagent is defined as a compound exhibiting an unnatural proportion of one or more of the isotopes that constitute such a compound. The term "Isotopic variant of the compound of general formula (I)" is defined as a compound of general formula (I) exhibiting an unnatural proportion of one or more of the isotopes that constitute such a compound.
The expression "unnatural proportion" means a proportion of such isotope which is higher than its natural abundance. The natural abundances of isotopes to be applied in this context are described in "Isotopic Compositions of the Elements 1997", Pure Appl. Chem., 70(1), 217-235, 1998.
Examples of such isotopes include stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as 2H (deuterium), 3H (tritium), l lC, 13C, 14C, 15N, 170, 180, 32P, 33P, 33S, 34S, 35S, 36S, 18F, 36C1, 82Br, 1231, 1241, 1251, 1291 and 1311, respectively.
With respect to the treatment and/or prevention of the disorders specified herein the isotopic variant(s) of the compounds of general formula (I) preferably contain deuterium ("deuterium- containing compounds of general formula (I)"). Isotopic variants of the compounds of general formula (I) in which one or more radioactive isotopes, such as 3H or 14C, are incorporated are useful e.g. in drug and/or substrate tissue distribution studies. These isotopes are particularly preferred for the ease of their incorporation and detectability. Positron emitting isotopes such as 18F or 11C may be incorporated into a compound of general formula (I). These isotopic variants of the compounds of general formula (I) are useful for in vivo imaging applications. Deuterium- containing and 13C-containing compounds of general formula (I) can be used in mass spectrometry analyses in the context of preclinical or clinical studies.
Isotopic variants of the compounds of general formula (I) can generally be prepared by methods known to a person skilled in the art, such as those described in the schemes and/or examples herein, by substituting a reagent for an isotopic variant of said reagent, preferably for a deuterium- containing reagent. Depending on the desired sites of deuteration, in some cases deuterium from D20 can be incorporated either directly into the compounds or into reagents that are useful for synthesizing such compounds. Deuterium gas is also a useful reagent for incorporating deuterium into molecules. Catalytic deuteration of olefinic bonds and acetylenic bonds is a direct route for incorporation of deuterium. Metal catalysts (i.e. Pd, Pt, and Rh) in the presence of deuterium gas can be used to directly exchange deuterium for hydrogen in functional groups containing hydrocarbons. A variety of deuterated reagents and synthetic building blocks are commercially available from companies such as for example C/D/N Isotopes, Quebec, Canada; Cambridge Isotope Laboratories Inc., Andover, MA, USA; and CombiPhos Catalysts, Inc., Princeton, NJ, USA.
The term "deuterium-containing compound of general formula (I)" is defined as a compound of general formula (I), in which one or more hydrogen atom(s) is/ are replaced by one or more deuterium atom(s) and in which the abundance of deuterium at each deuterated position of the compound of general formula (I) is higher than the natural abundance of deuterium, which is about 0.015%. Particularly, in a deuterium-containing compound of general formula (I) the abundance of deuterium at each deuterated position of the compound of general formula (I) is higher than 10%, 20%, 30%, 40%, 50%, 60%, 70% or 80%, preferably higher than 90%, 95%, 96% or 97%, even more preferably higher than 98% or 99% at said position(s). It is understood that the abundance of deuterium at each deuterated position is independent of the abundance of deuterium at other deuterated position(s).
The selective incorporation of one or more deuterium atom(s) into a compound of general formula (I) may alter the physicochemical properties (such as for example acidity [C. L. Perrin, et al., J. Am. Chem. Soc, 2007, 129, 4490], basicity [C. L. Perrin et al., J. Am. Chem. Soc, 2005, 127, 9641], lipophilicity [B. Testa et al., Int. J. Pharm., 1984, 19(3), 271]) and/or the metabolic profile of the molecule and may result in changes in the ratio of parent compound to metabolites or in the amounts of metabolites formed. Such changes may result in certain therapeutic advantages and hence may be preferred in some circumstances. Reduced rates of metabolism and metabolic switching, where the ratio of metabolites is changed, have been reported (A. E. Mutlib et al., Toxicol. Appl. Pharmacol., 2000, 169, 102). These changes in the exposure to parent drug and metabolites can have important consequences with respect to the pharmacodynamics, tolerability and efficacy of a deuterium-containing compound of general formula (I). In some cases deuterium substitution reduces or eliminates the formation of an undesired or toxic metabolite and enhances the formation of a desired metabolite (e.g. Nevirapine: A. M. Sharma et al., Chem. Res. Toxicol., 2013, 26, 410; Efavirenz: A. E. Mutlib et al., Toxicol. Appl. Pharmacol., 2000, 169, 102). In other cases the major effect of deuteration is to reduce the rate of systemic clearance. As a result, the biological half-life of the compound is increased. The potential clinical benefits would include the ability to maintain similar systemic exposure with decreased peak levels and increased trough levels. This could result in lower side effects and enhanced efficacy, depending on the particular compound's pharmacokinetic/ pharmacodynamic relationship. ML-337 (C. J. Wenthur et al., J. Med. Chem., 2013, 56, 5208) and Odanacatib (K. Kassahun et al., WO2012/112363) are examples for this deuterium effect. Still other cases have been reported in which reduced rates of metabolism result in an increase in exposure of the drug without changing the rate of systemic clearance (e.g. Rofecoxib: F. Schneider et al., Arzneim. Forsch. / Drug. Res., 2006, 56, 295; Telaprevir: F. Maltais et al., J. Med. Chem., 2009, 52, 7993). Deuterated drugs showing this effect may have reduced dosing requirements (e.g. lower number of doses or lower dosage to achieve the desired effect) and/or may produce lower metabolite loads.
A compound of general formula (I) may have multiple potential sites of attack for metabolism. To optimize the above-described effects on physicochemical properties and metabolic profile, deuterium-containirig compounds of general formula (I) having a certain pattern of one or more deuterium-hydrogen exchange(s) can be selected. Particularly, the deuterium atom(s) of deuterium- containing compound(s) of general formula (I) is/are attached to a carbon atom and/or is/are located at those positions of the compound of general formula (I), which are sites of attack for metabolizing enzymes such as e.g. cytochrome P450.
Where the plural form of the word compounds, salts, polymorphs, hydrates, solvates and the like, is used herein, this is taken to mean also a single compound, salt, polymorph, isomer, hydrate, solvate or the like.
By "stable compound' or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
The compounds of the present invention optionally contain one asymmetric centre, depending upon the location and nature of the various substituents desired. It is possible that one asymmetric carbon atom is present in the (R) or (S) configuration, which can result in racemic mixtures. In certain instances, it is possible that asymmetry also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds. Preferred compounds are those which produce the more desirable biological activity. Separated, pure or partially purified isomers and stereoisomers or racemic mixtures of the compounds of the present invention are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques known in the art.
The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers. Examples of appropriate acids are tartaric, diacetyltartaric, ditoluoyltartaric and camphorsulfonic acid. Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallisation. The optically active bases or acids are then liberated from the separated diastereomeric salts. A different process for separation of optical isomers involves the use of chiral chromatography (e.g., HPLC columns using a chiral phase), with or without conventional derivatisation, optimally chosen to maximise the separation of the enantiomers. Suitable HPLC columns using a chiral phase are commercially available, such as those manufactured by Daicel, e.g., Chiracel OD and Chiracel OJ, for example, among many others, which are all routinely selectable. Enzymatic separations, with or without derivatisation, are also useful. The optically active compounds of the present invention can likewise be obtained by chiral syntheses utilizing optically active starting materials. In order to distinguish different types of isomers from each other reference is made to IUPAC Rules Section E (Pure Appl Chem 45, 11-30, 1976).
The present invention includes all possible stereoisomers of the compounds of the present invention as single stereoisomers, or as any mixture of said stereoisomers, e.g. (R)- or (S)- isomers, in any ratio. Isolation of a single stereoisomer, e.g. a single enantiomer or a single diastereomer, of a compound of the present invention is achieved by any suitable state of the art method, such as chromatography, especially chiral chromatography, for example.
Further, it is possible for the compounds of the present invention to exist as tautomers. The present invention includes all possible tautomers of the compounds of the present invention as single tautomers, or as any mixture of said tautomers, in any ratio.
Further, the compounds of the present invention can exist as N-oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised. The present invention includes all such possible N-oxides.
The present invention also covers useful forms of the compounds of the present invention, such as metabolites, hydrates, solvates, salts, in particular pharmaceutically acceptable salts, and/or co- precipitates.
The compounds of the present invention can exist as a hydrate, or as a solvate, wherein the compounds of the present invention contain polar solvents, in particular water, methanol or ethanol for example, as structural element of the crystal lattice of the compounds. It is possible for the amount of polar solvents, in particular water, to exist in a stoichiometric or non-stoichiometric ratio. In the case of stoichiometric solvates, e.g. a hydrate, hemi-, (semi-), mono-, sesqui-, di-, tri-, tetra-, penta- etc. solvates or hydrates, respectively, are possible. The present invention includes all such hydrates or solvates. Hydrates are preferred solvates in the context of the present invention.
In particular, the 3,3,3-trifluoro-2-oxopropyl derivatives of the formula (TB) according to the invention (ketone form) may also be present in the 3,3,3-trifluoro-2,2-dihydroxypropyl form (TB)' (hydrate form) (see Scheme 1 below); both forms are expressly embraced by the present invention.
Scheme 1
Further, it is possible for the compounds of the present invention to exist in free form, e.g. as a free base, or as a free acid, or as a zwitterion, or to exist in the form of a salt. Said salt may be any salt, either an organic or inorganic addition salt, particularly any pharmaceutically acceptable organic or inorganic addition salt, which is customarily used in pharmacy, or which is used, for example, for isolating or purifying the compounds of the present invention.
The term "pharmaceutically acceptable salt" refers to an inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. "Pharmaceutical Salts," J. Pharm. Sci. 1977, 66, 1-19.
A suitable pharmaceutically acceptable salt of the compounds of the present invention may be, for example, an acid-addition salt of a compound of the present invention bearing a nitrogen atom, in a chain or in a ring, for example, which is sufficiently basic, such as an acid-addition salt with an inorganic acid, or "mineral acid", such as hydrochloric, hydrobromic, hydroiodic, sulfuric, sulfamic, bisulfuric, phosphoric, or nitric acid, for example, or with an organic acid, such as formic, acetic, acetoacetic, pyruvic, trifluoroacetic, propionic, butyric, hexanoic, heptanoic, undecanoic, lauric, benzoic, salicylic, 2-(4-hydroxybenzoyl)-benzoic, camphoric, cinnamic, cyclopentanepropionic, digluconic, 3-hydroxy-2-naphthoic, nicotinic, pamoic, pectinic, 3- phenylpropionic, pivalic, 2-hydroxyethanesulfonic, itaconic, trifluoromethanesulfonic, dodecylsulfuric, ethanesulfonic, benzenesulfonic, para-toluenesulfonic, methanesulfonic, 2-naphthalenesulfonic, naphthalinedisulfonic, camphorsulfonic acid, citric, tartaric, stearic, lactic, oxalic, malonic, succinic, malic, adipic, alginic, maleic, fumaric, D-gluconic, mandelic, ascorbic, glucoheptanoic, glycerophosphoric, aspartic, sulfosalicylic, or thiocyanic acid, for example.
Further, another suitably pharmaceutically acceptable salt of a compound of the present invention which is sufficiently acidic, is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium, magnesium or strontium salt, or an aluminium or a zinc salt, or an ammonium salt derived from ammonia or from an organic primary, secondary or tertiary amine having 1 to 20 carbon atoms, such as ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, diethylaminoethanol, tris(hydroxymethyl)aminomethane, procaine, dibenzylamine, -mefhylmorpholine, arginine, lysine, 1,2-ethylenediamine, /V-methylpiperidine, -mefhyl-glucamine, -dimethyl-glucamine, -efhyl-glucamine, 1,6-hexanediamine, glucosamine, sarcosine, serinol, 2-amino-l,3-propanediol, 3-amino-l,2-propanediol, 4-amino- 1,2,3-butanetriol, or a salt with a quarternary ammonium ion having 1 to 20 carbon atoms, such as tetramethylammonium, tetraethylammonium, tetra(«-propyl)ammonium, tetra(n-butyl) ammonium, /V-benzyl-/VN,/V-trimethylammonium, choline or benzalkonium. Those skilled in the art will further recognise that it is possible for acid addition salts of the claimed compounds to be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. Alternatively, alkali and alkaline earth metal salts of acidic compounds of the present invention are prepared by reacting the compounds of the present invention with the appropriate base via a variety of known methods.
The present invention includes all possible salts of the compounds of the present invention as single salts, or as any mixture of said salts, in any ratio.
In the present text, in particular in the Experimental Section, for the synthesis of intermediates and of examples of the present invention, when a compound is mentioned as a salt form with the corresponding base or acid, the exact stoichiometric composition of said salt form, as obtained by the respective preparation and/or purification process, is, in most cases, unknown.
Unless specified otherwise, suffixes to chemical names or structural formulae relating to salts, such as "hydrochloride", "trifluoroacetate", "sodium salt", or "x HCl", "x CF3COOH", "x Na+", for example, mean a salt form, the stoichiometry of which salt form not being specified.
This applies analogously to cases in which synthesis intermediates or example compounds or salts thereof have been obtained, by the preparation and/or purification processes described, as solvates, such as hydrates, with (if defined) unknown stoichiometric composition.
Furthermore, the present invention includes all possible crystalline forms, or polymorphs, of the compounds of the present invention, either as single polymorph, or as a mixture of more than one polymorph, in any ratio.
In a distinct embodiment, the present invention relates to compounds of formula (I), supra, wherein Ar represents a group of the formula
in which
represents the point of attachment to the nitrogen atom,
R2A represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, methyl, ethyl, isopropyl, nitro, methoxy, difluoromethoxy and methylsulfanyl,
R2B and R2C represent, independently from each other, a group selected from a fluorine atom, a chlorine atom, methyl and ethyl,
R2D, R2E and R2F represent, independently from each other, a group selected from a fluorine atom and a chlorine atom,
R3A represents a group selected from a hydrogen atom, a fluorine atom, methyl, ethyl, methoxy and methoxycarbonyl,
R3B represents a chlorine atom,
R3C represents a group selected from a chlorine atom and ethoxy,
R3D represents aminocarbonyl,
R3E represents a methyl group,
R3F represents a chlorine atom,
R3G represents a group selected from a fluorine atom, a chlorine atom and methyl,
R4A represents a methoxy group,
armaceutically acceptable salt, hydrate and/or solvate thereof. In a preferred embodiment, the present invention relates to compounds according to formula (I), supra, wherein
R1 represents a group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
represents a group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
R2A represents a group selected from a chlorine atom, a bromine atom, isopropyl, and nitro,
R3G represents a group selected from a chlorine atom and methyl,
or a pharmaceutically acceptable salt, hydrate and/or solvate thereof.
In accordance with a further preferred embodiment, the present invention covers compounds of general formula (I), supra, wherein
R1 represents a group of the formula
in which
represents the point of attachment to the nitrogen atom,
Ar represents a group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
R2A represents a group selected from a hydrogen atom, a fluorine atom, and methylsulfanyl,
R2B and R2C represent, independently from each other, a group selected from a fluorine atom, a chlorine atom, methyl and ethyl,
R2F represents a group selected from a fluorine atom and a chlorine atom,
R3A represents a group selected from a hydrogen atom, a fluorine atom and ethyl,
In accordance with a further preferred embodiment, the present invention covers compounds of general formula (I), supra, wherein
R1 represents a (2S)-3,3,3-trifluoro-2-hydroxypropyl group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
or a pharmaceutically acceptable salt, hydrate and/or solvate thereof.
R1 represents a (2R)-3,3,3-trifluoro-2-hydroxypropyl group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
or a pharmaceutically acceptable salt, hydrate and/or solvate thereof. In a particular further embodiment of the first aspect, the present invention covers combinations of two or more of the above mentioned embodiments under the heading "further embodiments of the first aspect of the present invention".
The present invention covers any sub-combination within any embodiment or aspect of the present invention of compounds of general formula (I), supra.
The present invention covers any sub-combination within any embodiment or aspect of the present invention of intermediate compounds of general formula (II), (III), (IV), (V), (VI), (VII), (VIII), (X), (XIII), (XIV) and (XV). The present invention covers the compounds of general formula (I) which are disclosed in the Example Section of this text, infra.
In accordance with a second aspect, the present invention covers methods of preparing compounds of general formula (I) as defined supra, said methods comprising the step
[A] of allowing an intermediate compound of formula (II):
in which R1 is as defined for the compound of general formula (I) as defined supra, and R5 represents a (Ci-C4)-alkyl group, in particular a methyl group,
to react in a first step in the presence of a base with a compound of general formula (III):
in which
R6 represents a (Ci-C -alkyl group, in particular a methyl group,
to give an intermediate compound, which is then allowed to react in the presence of a base, and optionally a copper salt, in a second step with a hydrazine compound of general formula (IV) or a respective salt thereof
H N 2
Ar (IV) in which Ar is as defined for the compound of general formula (I) as defined supra,
thereby giving a compound of general formula (V) :
in which R and Ar are as defined for the compound of general formula (I) as defined supra, and
R6 represents a (Ci-C -alkyl group, in particular a methyl group,
followed by a subsequent step
[B] of allowing the compound of formula (V) obtained in step [A] to react with ammonia thereby giving a compound of general formula (I):
in which R and Ar are as defined for the compound of general formula (I) as defined supra,
optionally followed by step
[C] conversion of the alcohols of general formula (I- A):
in which Ar is as defined for the compound of general formula (I) as defined supra, to the ketones of general formula (I-B)
in which Ar is as defined for the compound of general formula (I) as defined supra, using known oxidation methods,
each [A], [B] and [C] optionally followed, where appropriate, by (i) separating the compounds of formula (I) thus obtained into their respective enantiomers, and/or (ii) converting the compounds of formula (I) into their respective hydrates, solvates, salts and/or hydrates or solvates of the salts by treatment with the corresponding solvents and/or acids or bases.
The present invention covers methods of preparing compounds of the present invention of general formula (I), said methods comprising the steps as described in the Experimental Section herein.
The schemes and procedures described below illustrate synthetic routes to the compounds of general formula (I) of the invention and are not intended to be limiting. It is clear to the person skilled in the art that the order of transformations as exemplified in schemes 2, 3, 4, 5, 6 and 7 can be modified in various ways. The order of transformations exemplified in these schemes is therefore not intended to be limiting. In addition, interconversion of any of the substituents, R1, R2, R3, R4, R5 and R6 and Ar can be achieved before and/or after the exemplified transformations. These modifications can be such as the introduction of protecting groups, cleavage of protecting groups, reduction or oxidation of functional groups, halogenation, metallation, substitution or other reactions known to the person skilled in the art. These transformations include those which introduce a functionality which allows for further interconversion of substituents. Appropriate protecting groups and their introduction and cleavage are well-known to the person skilled in the art (see for example T.W. Greene and P.G.M. Wuts in Protective Groups in Organic Synthesis, 3rd edition, Wiley 1999). Specific examples are described in the subsequent paragraphs.
The multicomponent cyclization (II)— (V) is carried out by first reacting imidate of formula (II) with an acid chloride of formula (III) in the presence of a base to form an intermediate which is in a subsequent step reacted with the aryl hydrazine compound of formula (IV). Typically the formed intermediate is not isolated and the reaction over the two steps is performed in one-pot. The arylhydrazine compound for formula (I) may also be used in form of its salts, such as a hydrochloride salt or a tosylate salt. Under the alkaline reaction conditions, the hydrazine salt will be reconverted into the free base form. The amount of base added may then be adjusted in this respect.
For the preparation of compounds of general formula (I) in which Ar constitutes a pyridine group it may be beneficial in the second step to add a copper or zinc salt, such as copper(II) sulfate, copper(II) chloride, zinc(II) sulfate and zinc(II) chloride. Typically and preferably copper(II) sulfate or zinc(II) sulfate are used.
Suitable bases for both steps are typically tertiary amine bases, such as /V,/V-diisopropylethylamine (DIPEA), triethylamine, triisopropylamine, -mefhylimidazole, -mefhylmorpholine, pyridine and 4-(Ai,/V-dimethylamino)pyridine. Preferably, /V,/V-diisopropylethylamine (DIPEA) is used as base. The reaction is performed in an inert organic solvent, such as dichloromethane, 1,2-dichloroethane, methyl ieri-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxy ethane, toluene, pyridine, ethyl acetate, acetonitrile or A^ -dimefhylformamide, or in a mixture of these solvents. Preferably tetrahydrofuran or dioxane or a mixture thereof are used as solvents. The first step is generally carried out at a temperature in the range of -10°C to +120°C, preferably at 0°C. The second step is generally carried out at a temperature in the range of +20°C to +120°C, preferably at room temperature. Concomitant microwave irradiation may have a beneficial effect in this reaction as well at a temperature in the range of +60°C to +150°C , preferably at +120°C.
The aminolysis reaction (V) — (I) is usually carried out in a solution of ammonia. Suitable ammonia solutions for this step are saturated ammonia solutions, in particular a solution of ammonia in methanol, ethanol, isopropanol, tetrahydrofuran, dioxane or water or a mixture thereof. Preferably, a methanolic ammonia solution is used. The reaction is preferably performed directly in the ammonia solution in the absence of any further reaction solvent. This step is generally carried out at a temperature in the range of of +20°C to +120°C, preferably at room temperature. Concomitant microwave irradiation may have a beneficial effect in this reaction as well at a temperature in the range of +60°C to +150°C, preferably at +120°C.
The oxidation reaction (TA)— (TB) is carried out using customary oxidation methods known from the literature [e.g. JOC, 1983, 48, 4155 (Dess Martin oxidation); Tet Lett, 1994, 35,3485 (IBX oxidation); JOC, 1970, 35, 3589 (acid dichromate oxidation); Tet Lett, 1979, 399 (PDC oxidation); Tetrahedron, 1978, 34, 1651 (Swern oxidation)]. Thus, the alcohol group in the compounds of the general formula (I-A) is preferably oxidized using Dess-Martin periodinane (DMP). In a typical procedure the reaction is carried out in dichloromethane at a temperature of 0°C and subsequent warming up to room temperature.
Compounds of general formula (I) in which Ar comprises a sulfone substituent of the formula -S(=0)2CH3 may be prepared from the respective corresponding methylsulfanyl precursors by customary oxidation methods known in the art, such as oxidation with 3-chloroperoxybenzoic acid (mCPBA).
Alternatively, compounds of general formula (V) as defined supra, may be prepared by a method comprising the step
[D] of allowing an intermediate compound of formula (VI):
in which
R1 is as defined for the compound of general formula (I) as defined supra, and
R6 represents a (Ci-C -alkyl group, in particular an ethyl group,
and/or a tautomer thereof to react with a compound of general formula (VII):
Ar-B'
°-R (VII),
in which
Ar is as defined for the compound of general formula (I) as defined supra,
R^ and RXB represent, independently from each other, a hydrogen atom, or
R^ and RXB together form a -CH2CH2- or -C(CH3)2 C(CH3)2- bridge,
in the presence of a copper catalyst and an amine base thereby giving a compound of general formula (V) in which
R1 and Ar are as defined for the compound of general formula (V):
Cl (V), in which R and Ar are as defined for the compound of general formula (I) as defined supra, and
R6 represents a (Ci-C -alkyl group, in particular an ethyl group,
The coupling reaction (VI) + (VII)— > (V) is typically carried out with the aid of a copper catalyst and an amine base ["Chan-Lam coupling" conditions; see, for instance, D. M. T. Chan et al., Tetrahedron Lett. 44 (19), 3863-3865 (2003); J. X. Qiao and P. Y. S. Lam, Synthesis, 829-856 (2011); K. S. Rao and T.-S. Wu, Tetrahedron 68, 7735-7754 (2012)]. Copper catalysts suitable for this process are in particular copper(II) salts, such as copper(II) acetate, copper(II) trifluoromethane- sulfonate or copper(II) bromide. Practical amine bases include, for example, triethylamine, N,N- diisopropylethylamine, pyridine and 4-(Ar,/V-dimethylamino)pyridine. The reaction is performed in an inert organic solvent, such as dichloromethane, 1,2-dichloroethane, methyl ieri-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane, toluene, pyridine, ethyl acetate, acetonitrile or -dimethylformamide, or in a mixture of these solvents. Preferably, pyridine is used both as solvent and base. The coupling is generally carried out at a temperature in the range of +20°C to +120°C, preferably at +20°C to +70°C. Concomitant microwave irradiation may have a beneficial effect in this reaction as well.
Compounds of general formula (II) as defined supra, can be prepared by a method comprising the step
[a] of allowing an intermediate compound of formula (VIII):
in which R1 is as defined for the compound of general formula (I) as defined supra, to react with a nitrile compound of general formula (IX),
in which X represents a leaving group, such as chlorine, bromine, iodine, mesylate or tosylate, in particular chlorine or bromine,
thereby giving a compound of general formula (X)
in which R is as defined for the compound of general formula (I) as defined supra, followed by a subsequent step
[b] of allowing the compound of formula (X) obtained in step [a] to react with a basic alcoholate, preferably sodium methanolate, thereby giving a compound of general formula (II),
in which
R1 is as defined for the compound of general formula (I) as defined supra,and
R5 represents a (Ci-C -alkyl group, in particular a methyl group.
The /V-alkylation reaction (VIII) + (IX) -> (X) (step [a]) is typically carried out in the presence of a base. Typical and exemplary bases include sodium carbonate, potassium carbonate, cesium carbonate, A^ -diisopropylefhylamine, triethylamine, sodium ieri-butylate or potassium tert- butylate in acetonitrile, methylisobutylketone, dioxane, dimethylformamide, dimethylacetamide, N- methylpyrrolidinone, dimethylsulfoxide and sulfolane, preference is given to potassium carbonate in methylisobutylketone or acetonitrile. The reaction may optionally be carried out in an advantageous manner with addition of an alkylation catalyst such as, for example, lithium bromide, sodium iodide, lithium iodide, tetra-n-butylammoniumbromide, tetra-n-butylammoniumiodide or benzyltriethylammoniumchloride. The reactions are generally carried out in a temperature range of from +40°C to +120°C, preferably at from +60°C to +80°C. The reactions can be carried out at atmospheric, at elevated or at reduced pressure (for example at from 0.5 to 5 bar); in general, the reactions are carried out at atmospheric pressure. It may be advantageous to slowly perform the addition of the alkylation agent (VII) over a longer timespan. Conversion to the imidate of general formula (II) can be achieved via standard reaction protocols known to the person skilled in the art (step [b] : (X)— (II)). The reaction is typically carried out under basic reactions conditions by reacting with a basic alcoholate. Typical bases, which may be used are sodium methanolate, sodium ethanolate, sodium propanolate, sodium isopropoxide, sodium ieri-butylate or potassium ieri-butylate in methanol, ethanol, n-propanol, isopropanol, n- butanol, isobutanol and tert-butanol. Preference is given to sodium methanolate in methanol. The reactions are generally carried out in a temperature range of from +20 to +80 °C, preferably at from +40 to +60 °C.
Alternatively, the nitrile compounds of general formula (X) may optionally also be prepared as shown in the synthetic scheme 2 below:
Scheme 2
TFAA = trifluoroacetic acid anhydride
The amide coupling (XI)— (XII) can be carried out directly with the help of a condensing agent or activating agent in the presence of a base or over two steps via an acyl chloride or carboxylic acid imidazolide. Typical condensation and activating agents for the amide formation in process steps (XI)— (XII) include, for example, carbodiimides such as Ν,Ν'-diethyl-, N, N'-dipropyl-, Ν,Ν'- diisopropyl-N,N'-dicyclohexylcarbodiimide (DCC) or N-(3-dimethylaminoisopropyl)-N'-ethyl- carbodiimide hydrochloride (EDC), phosgene derivatives such as N, N'-carbonyldiimidazole (CDI), 1,2-oxazolium compounds such as 2-ethyl-5-phenyl-l,2-oxazolium-3-sulphate or 2-tert- butyl-5-methyl-isoxazolium perchlorate, acylamino compounds such as 2-ethoxy-l-ethoxy- carbonyl-l,2-dihydroquinoline, or isobutyl chloroformate, propanephosphonic anhydride, diethyl cyanophosphonate, bis(2-oxo-3-oxazo-lidinyl)phosphoryl chloride, benzotriazol-l-yloxytris- (dimethylamino)phosphonium hexafluorophosphate, benzotriazol-l-yloxytris(pyrrolidino)- phosphonium hexafluorophosphate (PyBOP), 0-(benzotriazol-l-yl)-N, N, Ν',Ν'-tetramethyl- uronium tetrafluoroborate (TBTU), 0-(benzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HBTU), 2-(2-oxo-l-(2H)-pyridyl)-l,l,3,3-tetramethyluronium tetrafluoroborate (TPTU), 0-(7-azabenzotriazol- 1 -yl)-N,N,N',N'-tetramethyluroniumhexafluorophosphate (HATU) or 0-(lH-6-chloro-benzotriazol-l-yl)-l,l,3,3-tetramethyluronium tetrafluoroborate (TCTU), optionally in combination with other additives such as 1-hydroxybenzotriazole (HOBt) or N- hydroxysuccinimide (HOSu). The acyl chlorides are typically prepared with thionyl chloride or oxalyl chloride in an inert solvent like dichloromethane or Ν,Ν-dimethylformamide. It is also possible to use mixtures of the solvents mentioned
Typical and exemplary bases include sodium carbonate, potassium carbonate, cesium carbonate, Ν,Ν-diisopropylethylamine, triethylamine, sodium tert-butylate or potassium tert-butylate in acetonitrile, methylisobutylketone, dioxane, dimethylformamide, dimethylacetamide, N-methyl- pyrrolidinone, dimethylsulfoxide and sulfolane, preference is given to potassium carbonate in methylisobutylketone or acetonitrile. The reaction may optionally be carried out in an advantageous manner with addition of an alkylation catalyst such as, for example, lithium bromide, sodium iodide, lithium iodide, tetra-n-butylammoniumbromide, tetra-n-butylammoniumiodide or benzyltriethylammoniumchloride.The conversion to the nitrile (XII)— > (X) can be carried out with the help of dehydrating agent. Typical dehydrating agents include, for example trifluoroacetic acid anhydride, phosphorous pentoxide (P4O10), phosphoryl chloride (POCI3), phosphorous penta- chloride (PC ), CCI4-PPI13 (Appel reagent), hexamethylphosphoramide (HMPA); methyl N- (triethylammoniumsulfonyl)carbamate (Burgess reagent), (Chloromethylene)dimethyliminium chloride (Vilsmeier reagent), oxalyl chloride/DMSO and thionylchloride (SOCI2).
Typical and exemplary solvents for both steps (XI)— (XII) and (XII)— > (X) include for example, ethers such as diethyl ether, dioxane, tetrahydrofuran, glycol dimethyl ether or diethylene glycol dimethyl ether, hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane or mineral oil, fractions, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride, 1,2-dichloroethane, trichloroethylene or chlorobenzene, or othersolvents such as acetone, ethyl acetate, acetonitrile, pyridine,dimethyl sulphoxide, Ν,Ν-dimethylformamide, Ν,Ν'- dimethylpropyleneurea (DMPU) or N-methylpyrrolidone (NMP). It is also possible to use mixtures of the solvents mentioned.
In a typical and preferred procedure the carboxylic acid (XI) is first reacted with pivaloyl chloride in the presence of pyridine to form an intermediate which is in a subsequent step reacted with ammonia. Typically the formed intermediate is not isolated and the reaction over the two steps is performed in one-pot. Suitable as bases for the first step are preferably, pyridine, 4-(N,N-dimethyl- amino)pyridine or Ν,Ν-diisopropylethylamine (DIPEA). The conversion of carboxamide (XII) into nitrile (X) is then typically performed by reaction with trifluoroacetic anhydride. Both reactions are conducted in an inert organic solvent, preferably tetrahydrofuran.
Compounds of general formula (VI) as defined supra, can be prepared by a method comprising the step
[a] of allowing a compound of formula (XIII):
in which R is as defined for the compound of general formula (I) as defined supra,to first react with hydrazine to give the hydrazine of formula (XIV)
in which R1 is as defined for the compound of general formula (I) as defined supra, which is then being condensed with a (thiooxo)amino compound of formula (XV)
in which
R6 represents a (Ci-C -alkyl group, in particular an ethyl group,
or a salt thereof,
the presence of a base to give a 1,2,4-triazole derivative of formula (VI)
and/or a tautomer thereof,
in which R is as defined for the compound of general formula (I) as defined supra, and
R6 represents a (Ci-C -alkyl group, in particular an ethyl group,
The transformation (XIII)— > (XIV) is carried out in the usual way by treating methyl ester (XIII) with hydrazine or hydrazine hydrate in an alcoholic solvent, such as methanol, ethanol, n-propanol, isopropanol or n-butanol, at a temperature in the range of +20°C to +100°C.
The condensation reaction (XIV) + (XV)— (VI) is usually carried out in an inert dipolar-aprotic solvent, such as -dimefhylformamide (DMF), /V-dimefhylacetamide (DMA), dimethylsulf- oxide (DMSO), /V-mefhylpyrrolidinone (NMP) or /V,/V'-dimefhylpropylene urea (DMPU), in the presence of a sufficiently strong base, such as sodium hydride or a sodium or potassium alkoxide, for example sodium or potassium methoxide, sodium or potassium ethoxide, or sodium or potassium ieri-butoxide. The (thiooxo) amine (XV) may be employed as such in this reaction or in salt form, e.g. as the hydrochloride salt. In the latter case, a proportional excess of base is used. The reaction is generally performed at a temperature between +80°C and +150°C. Heating by means of a microwave reactor device may have a beneficial effect for this condensation reaction.
The 1,2,4-triazole derivative of formula (VI) produced by this reaction may also be present in other tautomeric forms, such as (VI-A) or (VI-B)
or as a mixture of tautomers.
The compounds of formula (VIII) and (XI) can be synthesized by the procedures described in Int. Pat. Appl. WO 2010/105770 and WO 201 1/104322 (see also synthesis schemes 3 and 4 below).
The compounds of the formulae (III), (IV) and (VII) are either commercially available, known from the literature, or can be prepared from readily available starting materials by adaptation of standard methods described in the literature. Detailed procedures and literature references for preparing the starting materials can also be found in the Experimental Part in the section on the preparation of the starting materials and intermediates.
The preparation of the compounds of the invention may be illustrated by means of the following synthesis schemes:
[cf. Int. Pat. Appl. WO 2011/104322-Al].
Scheme 4
[cf. Int. Pat. Appl. WO 201 1/104322-Al] . Scheme 5
TFAA: trifluoro acetic acid anhydride
Scheme 6
The compounds of general formula (I) of the present invention can be converted to any salt, preferably pharmaceutically acceptable salts, as described herein, by any method which is known to the person skilled in the art. Similarly, any salt of a compound of general formula (I) of the present invention can be converted into the free compound, by any method which is known to the person skilled in the art.
The compounds of the present invention have valuable pharmacological properties and can be used for the prevention and/or treatment of various diseases and disease-induced states in humans and other mammals. Compounds of general formula (I) of the present invention demonstrate a valuable pharmacological spectrum of action and pharmacokinetic profile, both of which could not have been predicted. Compounds of the present invention have surprisingly been found to effectively inhibit the vasopressin Via receptor and it is possible therefore that said compounds be used for the treatment and/or prevention of diseases, preferably renal and cardiovascular diseases in humans and animals.
In the context of the present invention, the term "treatment" or "treating" includes inhibiting, delaying, relieving, mitigating, arresting, reducing, or causing the regression of a disease, disorder, condition, or state, the development and/or progression thereof, and/or the symptoms thereof. The term "prevention" or "preventing" includes reducing the risk of having, contracting, or experiencing a disease, disorder, condition, or state, the development and/or progression thereof, and/or the symptoms thereof. The term prevention includes prophylaxis. Treatment or prevention of a dis- order, disease, condition, or state may be partial or complete.
Throughout this document, for the sake of simplicity, the use of singular language is given preference over plural language, but is generally meant to include the plural language if not otherwise stated. For example, the expression "A method of treating a disease in a patient, comprising administering to a patient an effective amount of a compound of formula (I)" is meant to include the simultaneous treatment of more than one disease as well as the administration of more than one compound of formula (I).
The compounds of the present invention are potent selective or dual antagonists of vasopressin Via and V2 receptors. The compounds of the invention are therefore expected to be highly valuable as therapeutic agents for the treatment and/or prevention of diseases, in particular for the treatment and/or prevention of cardiovascular and renal diseases.
The compounds according to the invention are suitable for the treatment and/or prevention of renal diseases, in particular of acute and chronic kidney diseases, diabetic kidney diseases, and of acute and chronic renal failure. The general terms 'renal disease' or 'kidney disease' describe a class of conditions in which the kidneys fail to filter and remove waste products from the blood. There are two major forms of kidney disease: acute kidney disease (acute kidney injury, AKI) and chronic kidney disease (CKD). The compounds according to the invention may further be used for the treatment and/or prevention of sequelae of acute kidney injury arising from multiple insults such as ischemia-reperfusion injury, radiocontrast administration, cardiopulmonary bypass surgery, shock and sepsis. In the sense of the present invention, the term renal failure or renal insufficiency comprises both acute and chronic manifestations of renal insufficiency, as well as underlying or related kidney diseases such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathies, IgA nephropathy, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial diseases, nephropathic diseases such as primary and congenital kidney disease, nephritis, Alport syndrome, kidney inflammation, immunological kidney diseases such as kidney transplant rejection, immune complex-induced kidney diseases, nephropathy induced by toxic substances, contrast medium-induced nephropathy; minimal change glomerulonephritis (lipoid); Membranous glomerulonephritis; focal segmental glomerulosclerosis (FSGS); hemolytic uremic syndrome (HUS), amyloidosis, Goodpasture's syndrome, Wegener's granulomatosis, Purpura Schonlein-Henoch, diabetic and non-diabetic nephropathy, pyelonephritis, renal cysts, nephrosclerosis, hypertensive nephrosclerosis and nephrotic syndrome, which can be characterized diagnostically, for example, by abnormally reduced creatinine and/or water excretion, abnormally increased blood concentrations of urea, nitrogen, potassium and/or creatinine, altered activity of renal enzymes such as, for example, glutamyl synthetase, altered urine osmolarity or urine volume, increased microalbuminuria, macroalbuminuria, lesions of glomeruli and arterioles, tubular dilatation, hyperphosphataemia and/or the need for dialysis. The present invention also comprises the use of the compounds according to the invention for the treatment and/or prevention of sequelae of renal insufficiency, for example pulmonary edema, heart failure, uraemia, anaemia, electrolyte disturbances (e.g. hyperkalemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism. The compounds according to the invention are also suitable for the treatment and/or prevention of polycystic kidney disease (PCKD) and of the syndrome of inadequate ADH secretion (SIADH).
Cardiovascular diseases in this context that may be treated and/or prevented with the compounds of the invention include, but are not limited to, the following: acute and chronic heart failure including worsening chronic heart failure (or hospitalization for heart failure) and including congestive heart failure, arterial hypertension, resistant hypertension, arterial pulmonary hypertension, coronary heart disease, stable and unstable angina pectoris, atrial and ventricular arrhythmias, disturbances of atrial and ventricular rhythm and conduction disturbances, for example atrioventricular blocks of degree I-III (AVB I-III), supraventricular tachyarrhythmia, atrial fibrillation, atrial flutter, ventricular fibrillation, ventricular flutter, ventricular tachyarrhythmia, torsade-de-pointes tachycardia, atrial and ventricular extrasystoles, AV-junction extrasystoles, sick-sinus syndrome, syncopes, AV- node re-entry tachycardia and Wolff-Parkinson-White syndrome, acute coronary syndrome (ACS), autoimmune heart diseases (pericarditis, endocarditis, valvulitis, aortitis, cardiomyopathies), shock such as cardiogenic shock, septic shock and anaphylactic shock, aneurysms, Boxer cardiomyopathy (premature ventricular contraction), furthermore thromboembolic diseases and ischaemias such as peripheral perfusion disturbances, reperfusion injury, arterial and venous thromboses, myocardial insufficiency, endothelial dysfunction, micro- and macrovascular damage (vasculitis) and for preventing restenoses such as after thrombolysis therapies, percutaneous transluminal angioplasty (PTA), percutaneous transluminal coronary angioplasty (PTCA), heart transplantation and bypass operations, arteriosclerosis, disturbances of lipid metabolism, hypolipoproteinaemias, dyslipidemias, hypertriglyceridemias, hyperlipidemias and combined hyperlipidemias, hyper- cholesterolaemias, abetalipoproteinaemia, sitosterolemia, xanthomatosis, Tangier disease, adipositas, obesity, metabolic syndrome, transitory and ischemic attacks, stroke, inflammatory cardiovascular diseases, peripheral and cardiac vascular diseases, peripheral circulation disorders, spasms of the coronary arteries and peripheral arteries, and edema such as, for example, pulmonary edema, cerebral edema, renal edema and heart failure-related edema.
In the sense of the present invention, the term heart failure also includes more specific or related disease forms such as right heart failure, left heart failure, global insufficiency, ischemic cardiomyopathy, dilatative cardiomyopathy, congenital heart defects, heart valve defects, heart failure with heart valve defects, mitral valve stenosis, mitral valve insufficiency, aortic valve stenosis, aortic valve insufficiency, tricuspidal stenosis, tricuspidal insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined heart valve defects, heart muscle inflammation (myocarditis), chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcohol- toxic cardiomyopathy, cardiac storage diseases, heart failure with preserved ejection fraction (HFpEF or diastolic heart failure), and heart failure with reduced ejection fraction (HFrEF or systolic heart failure).
The compounds according to the invention are also suitable for the treatment and/or prevention of renal diseases, in particular of acute and chronic renal insufficiency, and of acute and chronic renal failure. In the sense of the present invention, the term renal insufficiency comprises both acute and chronic manifestations of renal insufficiency, as well as underlying or related kidney diseases such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathies, IgA nephropathy, glomerulonephritis, acute glomerulonephritis, glomerulosclerosis, tubulointerstitial diseases, nephropathic diseases such as primary and congenital kidney disease, nephritis, immunological kidney diseases such as kidney transplant rejection, immune complex-induced kidney diseases, nephropathy induced by toxic substances, contrast medium-induced nephropathy, diabetic and non-diabetic nephropathy, pyelonephritis, renal cysts, nephrosclerosis, hypertensive nephrosclerosis and nephrotic syndrome, which can be characterized diagnostically, for example, by abnormally reduced creatinine and/or water excretion, abnormally increased blood concentrations of urea, nitrogen, potassium and/or creatinine, altered activity of renal enzymes such as, for example, glutamyl synthetase, altered urine osmolarity or urine volume, increased microalbuminuria, macroalbuminuria, lesions of glomeruli and arterioles, tubular dilatation, hyperphosphataemia and/or the need for dialysis. The present invention also comprises the use of the compounds according to the invention for the treatment and/or prevention of sequelae of renal insufficiency, for example pulmonary edema, heart failure, uraemia, anaemia, electrolyte disturbances (e.g. hyperkalaemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism.
The compounds of the present invention may be particularly useful for the treatment and/or preven- tion of the cardiorenal syndrome (CRS) and its various subtypes. This term embraces certain disorders of the heart and kidneys whereby acute or chronic dysfunction in one organ may induce acute or chronic dysfunction of the other. CRS has been sub-classified into five types based upon the organ that initiated the insult as well as the acuity and chronicity of the disease (type 1 : development of renal insufficiency resulting from acute decompensated heart failure; type 2: chronic congestive heart failure resulting in progressive renal dysfunction; type 3: acute cardiac dysfunction resulting from an abrupt fall in renal function; type 4: chronic kidney disease leading to cardiac remodeling; type 5: systemic disease involving both the heart and the kidneys) [see, for example, M. R. Kahn et al., Nature Rev. Cardiol. 10, 261-273 (2013)].
The compounds according to the invention are also suitable for the treatment and/or prevention of polycystic kidney disease (PCKD) and of the syndrome of inadequate ADH secretion (SIADH). Furthermore, the compounds of the invention are suitable for use as a diuretic for the treatment of edemas and in electrolyte disorders, in particular in hypervolemic and euvolemic hyponatremia.
Moreover, the compounds according to the invention may be used for the treatment and/or prevention of peripheral arterial disease (PAD) including claudication and including critical limb ischemia, coronary microvascular dysfunction (CMD) including CMD type 1-4, primary and secondary Raynaud's phenomenon, microcirculation disturbances, claudication, peripheral and autonomic neuropathies, diabetic microangiopathies, diabetic retinopathy, diabetic limb ulcers, gangrene, CREST syndrome, erythematous disorders, onychomycosis, rheumatic diseases and for promoting wound healing.
Furthermore, the compounds of the invention are suitable for treating urological diseases and diseases of the male and female urogenital system such as, for example, benign prostatic syndrome (BPS), benign prostatic hyperplasia (BPH), benign prostatic enlargement (BPE), bladder outlet obstruction (BOO), lower urinary tract syndromes (LUTS), neurogenic overactive bladder (OAB), interstitial cystitis (IC), urinary incontinence (UI), such as, for example, mixed, urge, stress and overflow incontinence (MUI, UUI, SUI, OUI), pelvic pains, erectile dysfunction, dysmenorrhea and endometriosis.
The compounds according to the invention may also be used for the treatment and/or prevention of inflammatory diseases, asthmatic diseases, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), acute lung injury (ALI), alpha- 1 -antitrypsin deficiency (AATD), pulmonary fibrosis, pulmonary emphysema (e.g. smoking-induced pulmonary emphysema) and cystic fibrosis (CF). In addition, the compounds of the invention may be used for the treatment and/or prevention of pulmonary arterial hypertension (PAH) and other forms of pulmonary hypertension (PH), including pulmonary hypertension associated with left ventricular disease, HIV infection, sickle cell anaemia, thromboembolism (CTEPH), sarcoidosis, chronic obstructive pulmonary disease (COPD) or pulmonary fibrosis.
Additionally, the compounds according to the invention may be used for the treatment and/or prevention of liver cirrhosis, ascites, diabetes mellitus and diabetic complications such as, for example, neuropathy and nephropathy.
Further, the compounds of the invention are suitable for the treatment and/or prevention of central nervous disorders such as anxiety states, depression, glaucoma, cancer, such as in particular pulmonary tumors, and circadian rhythm misalignment such as jet lag and shift work.
Furthermore, the compounds according to the invention may be useful for the treatment and/or prevention of pain conditions, diseases of the adrenals such as, for example, pheochromocytoma and adrenal apoplexy, diseases of the intestine such as, for example, Crohn's disease and diarrhea, menstrual disorders such as, for example, dysmenorrhea, endometriosis, preterm labor and tocolysis.
Due to their activity and selectivity profile, the compounds of the present invention are believed to be particularly suitable for the treatment and/or prevention of acute and chronic kidney diseases including diabetic nephropathy, acute and chronic heart failure, preeclampsia, peripheral arterial disease (PAD), coronary microvascular dysfunction (CMD), Raynaud's syndrome, dysmenorrhea, cardiorenal syndrome, hypervolemic and euvolemic hyponatremia, liver cirrhosis, ascites, edema and the syndrome of inadequate ADH secretion (SIADH).
The diseases mentioned above have been well characterized in humans, but also exist with a com- parable etiology in other mammals, and may be treated in those with the compounds and methods of the present invention.
Thus, the present invention further relates to the use of the compounds according to the invention for the treatment and/or prevention of diseases, especially of the aforementioned diseases. The present invention further relates to the use of the compounds according to the invention for preparing a pharmaceutical composition for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
The present invention further relates to the use of the compounds according to the invention in a method for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
The present invention further relates to a method for the treatment and/or prevention of diseases, especially of the aforementioned diseases, by using an effective amount of at least one of the compounds according to the invention.
In accordance with another aspect, the present invention covers pharmaceutical combinations, in particular medicaments, comprising at least one compound of general formula (I) of the present invention and at least one or more further active ingredients, in particular for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
Particularly, the present invention covers a pharmaceutical combination, which comprises:
• one or more first active ingredients, in particular compounds of general formula (I) as defined supra, and
• one or more further active ingredients, in particular for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
The term "combination" in the present invention is used as known to persons skilled in the art, it being possible for said combination to be a fixed combination, a non-fixed combination or a kit-of- parts.
A "fixed combination" in the present invention is used as known to persons skilled in the art and is defined as a combination wherein, for example, a first active ingredient, such as one or more compounds of general formula (I) of the present invention, and a further active ingredient are present together in one unit dosage or in one single entity. One example of a "fixed combination" is a pharmaceutical composition wherein a first active ingredient and a further active ingredient are present in admixture for simultaneous administration, such as in a formulation. Another example of a "fixed combination" is a pharmaceutical combination wherein a first active ingredient and a further active ingredient are present in one unit without being in admixture.
A non-fixed combination or "kit-of-parts" in the present invention is used as known to persons skilled in the art and is defined as a combination wherein a first active ingredient and a further active ingredient are present in more than one unit. One example of a non-fixed combination or kit- of-parts is a combination wherein the first active ingredient and the further active ingredient are present separately. It is possible for the components of the non-fixed combination or kit-of-parts to be administered separately, sequentially, simultaneously, concurrently or chronologically staggered. The compounds of the present invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutically active ingredients where the combination causes no unacceptable adverse effects. The present invention also covers such pharmaceutical combinations. For example, the compounds of the present invention can be combined with known agents for the treatment and/or prevention of diseases, especially of the aforementioned diseases.
In particular, the compounds of the present invention may be used in fixed or separate combination with
• antithrombotic agents, for example and preferably from the group of platelet aggregation inhibitors, anticoagulants and profibrinolytic substances;
· blood pressure lowering agents, for example and preferably from the group of calcium antagonists, angiotensin All antagonists, ACE inhibitors, NEP inhibitors, vasopeptidase inhibitors, endothelin antagonists, renin inhibitors, alpha-blockers, beta-blockers, mineralocorticoid receptor antagonists and diuretics;
• antidiabetic agents (hypoglycemic or antihyperglycemic agents), such as for example and preferably insulin and derivatives, sulfonylureas, biguanides, thiazolidinediones, acarbose,
DPP4 inhibitors, GLP-1 analogues, or SGLT inhibitors (gliflozins).
• organic nitrates and NO-donors, for example sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhalational NO;
• compounds that inhibit the degradation of cyclic guano sine monophosphate (cGMP), for example inhibitors of phosphodiesterases (PDE) 1, 2, 5 and/or 9, in particular PDE-5 inhibitors such as sildenafil, vardenafil, tadalafil, udenafil, dasantafil, avanafil, mirodenafil, lodenafil, CTP-499 or PF-00489791;
• natriuretic peptides, such as for example atrial natriuretic peptide (ANP, anaritide), B-type natriuretic peptide or brain natriuretic peptide (BNP, nesiritide), C-type natriuretic peptide (CNP) or urodilatin;
• calcium sensitizers, such as for example and preferably levosimendan;
• NO- and heme-independent activators of soluble guanylate cyclase (sGC), for example and with preference the compounds described in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 and WO 02/070510;
· NO-independent, but heme-dependent stimulators of guanylate cyclase (sGC), for example and with preference the compounds described in WO 00/06568, WO 00/06569, WO 02/42301, WO 03/095451, WO 2011/147809, WO 2012/004258, WO 2012/028647 and WO 2012/059549; • agents, that stimulates the synthesis of cGMP, for example and with preference sGC modulators, for example and with preference riociguat, cinaciguat, vericiguat or BAY 1101042;
• inhibitors of human neutrophil elastase (HNE), such as for example sivelestat or DX-890 (reltran);
· compounds inhibiting the signal transduction cascade, in particular tyrosine and/or serine/threonine kinase inhibitors, such as for example nintedanib, dasatinib, nilotinib, bosutinib, regora- fenib, sorafenib, sunitinib, cediranib, axitinib, telatinib, imatinib, brivanib, pazopanib, vatalanib, gefitinib, erlotinib, lapatinib, canertinib, lestaurtinib, pelitinib, semaxanib or tandutinib;
• compounds influencing the energy metabolism of the heart, such as for example and preferably etomoxir, dichloroacetate, ranolazine or trimetazidine, or full or partial adenosine Al receptor agonists as GS-9667 (previously known as CVT-3619), capadenoson and neladenoson bialanate (BAY 1067197);
• compounds influencing the heart rate, such as for example and preferably ivabradine;
• cardiac myosin activators, such as for example and preferably omecamtiv mecarbil (CK- 1827452);
• anti-inflammatory drugs such as non-steroidal anti-inflammatory drugs (NSAIDs) including acetylsalicylic acid (aspirin), ibuprofen and naproxen, glucocorticoids, NEP inhibitors, 5- aminosalicylic acid derivatives, leukotriene antagonists, TNF-alpha inhibitors and chemokine receptor antagonists such as CCR1, 2 and/or 5 inhibitors;
· fat metabolism altering agents, for example and preferably from the group of thyroid receptor agonists, cholesterol synthesis inhibitors, such as for example and preferably HMG-CoA- reductase or squalene synthesis inhibitors, ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors and lipopro- tein(a) antagonists.
Antithrombotic agents are preferably to be understood as compounds from the group of platelet aggregation inhibitors, anticoagulants and profibrinolytic substances.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a platelet aggregation inhibitor, for example and preferably aspirin, clo- pidogrel, ticlopidine or dipyridamole.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thrombin inhibitor, for example and preferably ximelagatran, dabiga- tran, melagatran, bivalirudin or enoxaparin. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a GPIib/iIIa antagonist, for example and preferably tirofiban or abcixi- mab.
In a preferred embodiment of the invention, the compounds according to the invention are adminis- tered in combination with a factor Xa inhibitor, for example and preferably rivaroxaban, apixaban, otamixaban, fidexaban, razaxaban, fondaparinux, idraparinux, DU-176b, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
In a preferred embodiment of the invention, the compounds according to the invention are adminis- tered in combination with heparin or a low molecular weight (LMW) heparin derivative.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a vitamin K antagonist, for example and preferably coumarin.
Blood pressure lowering agents are preferably to be understood as compounds from the group of calcium antagonists, angiotensin All antagonists, ACE inhibitors, NEP inhibitors, vasopeptidase inhibitors, endothelin antagonists, renin inhibitors, alpha-blockers, beta-blockers, mineralocorticoid receptor antagonists and diuretics.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a calcium antagonist, for example and preferably nifedipine, amlodipine, verapamil or diltiazem.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an alpha- 1 -receptor blocker, for example and preferably prazosin or tam- sulosin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a beta-blocker, for example and preferably propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipranolol, nadolol, mepindolol, carazolol, sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, carteolol, esmolol, labetalol, carve - dilol, adaprolol, landiolol, nebivolol, epanolol or bucindolol.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an angiotensin All receptor antagonist, for example and preferably losar- tan, candesartan, valsartan, telmisartan, irbesartan, olmesartan, eprosartan, embursartan or azilsartan.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a vasopeptidase inhibitor or inhibitor of neutral endopeptidase (NEP), such as for example and preferably sacubitril, omapatrilat or AVE-7688. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a dual angiotensin All receptor antagonist/NEP inhibitor (ARNI), for example and preferably LCZ696.
In a preferred embodiment of the invention, the compounds according to the invention are adminis- tered in combination with an ACE inhibitor, for example and preferably enalapril, captopril, lisino- pril, ramipril, delapril, fosinopril, quinopril, perindopril, benazepril or trandopril.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an endothelin antagonist, for example and preferably bosentan, darusen- tan, ambrisentan, tezosentan, sitaxsentan, avosentan, macitentan or atrasentan.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a renin inhibitor, for example and preferably aliskiren, SPP-600 or SPP- 800.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a mineralocorticoid receptor antagonist, for example and preferably fine- renone, spironolactone, canrenone, potassium canrenoate, eplerenone, esaxerenone (CS-3150), or apararenone (MT-3995), CS-3150, or MT-3995.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a diuretic, such as for example and preferably furosemide, bumetanide, piretanide, torsemide, bendroflumethiazide, chlorothiazide, hydrochlorothiazide, xipamide, indapa- mide, hydroflumethiazide, methyclothiazide, polythiazide, trichloromethiazide, chlorothalidone, metolazone, quinethazone, acetazolamide, dichlorophenamide, methazolamide, glycerine, isosor- bide, mannitol, amiloride or triamterene.
Fat metabolism altering agents are preferably to be understood as compounds from the group of CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA- reductase or squalene synthesis inhibitors, ACAT inhibitors, MTP inhibitors, PPAR-alpha, PPAR- gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors, lipase inhibitors and lipoprotein(a) antagonists.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a CETP inhibitor, for example and preferably dalcetrapib, anacetrapib, BAY 60-5521 or CETP-vaccine (Avant).
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thyroid receptor agonist, for example and preferably D-thyroxin, 3,5,3'- triiodothyronin (T3), COS 23425 or axitirome (COS 26214). In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an HMG-CoA-reductase inhibitor from the class of statins, for example and preferably lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pravastatin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a squalene synthesis inhibitor, for example and preferably BMS- 188494 or TAK-475.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an ACAT inhibitor, for example and preferably avasimibe, melinamide, pactimibe, eflucimibe or SMP-797.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an MTP inhibitor, for example and preferably implitapide, R- 103757, BMS-201038 or JTT-130.
In a preferred embodiment of the invention, the compounds according to the invention are adminis- tered in combination with a PPAR-gamma agonist, for example and preferably pioglitazone or rosi- glitazone.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a PPAR-delta agonist, for example and preferably GW 501516 or BAY 68-5042.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a cholesterol absorption inhibitor, for example and preferably ezetimibe, tiqueside or pamaqueside.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a lipase inhibitor, for example and preferably orlistat.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a polymeric bile acid adsorber, for example and preferably cholestyramine, colestipol, colesolvam, CholestaGel or colestimide.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a bile acid reabsorption inhibitor, for example and preferably ASBT (= IBAT) inhibitors such as AZD-7806, S-8921, AK-105, BARI-1741, SC-435 or SC-635.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a lipoprotein(a) antagonist, for example and preferably gemcabene calcium (CI- 1027) or nicotinic acid. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a TGFbeta antagonist, by way of example and with preference pirfenidone or fresolimumab.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with HIF-PH inhibitors, by way of example and with preference molidustat or roxadustat.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a CCR2 antagonist, by way of example and with preference CCX-140.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a TNFalpha antagonist, by way of example and with preference adalimumab.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a galectin-3 inhibitor, by way of example and with preference GCS-100.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a BMP-7 agonist, by way of example and with preference THR- 184.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a NOX1/4 inhibitor, by way of example and with preference GKT-137831.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a medicament which affects the vitamin D metabolism, by way of example and with preference cholecalciferol or paracalcitol.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a cytostatic agent, by way of example and with preference cyclophosphamide.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an immunosuppressive agent, by way of example and with preference ciclosporin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a phosphate binder, by way of example and with preference sevelamer or lanthanum carbonate. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a calcimimetic for therapy of hyperparathyroidism.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with agents for iron deficit therapy, by way of example and with preference iron products.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with agents for the therapy of hyperurikaemia, by way of example and with preference allopurinol or rasburicase.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with glycoprotein hormone for the therapy of anaemia, by way of example and with preference erythropoietin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with biologies for immune therapy, by way of example and with preference abatacept, rituximab, eculizumab or belimumab.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with Jak inhibitors, by way of example and with preference ruxolitinib, tofacitinib, baricitinib, CYT387, GSK2586184, lestaurtinib, pacritinib (SB1518) or TG101348.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with prostacyclin analogs for therapy of microthrombi.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an alkali therapy, by way of example and with preference sodium bicarbonate.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an mTOR inhibitor, by way of example and with preference everolimus or rapamycin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an NHE3 inhibitor, by way of example and with preference AZD1722.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an eNOS modulator, by way of example and with preference sapropterin. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a CTGF inhibitor, by way of example and with preference FG- 3019.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with antidiabetics (hypoglycemic or antihyperglycemic agents), such as for example and preferably insulin and derivatives, sulfonylureas such as tolbutamide, carbutamide, acetohexamide, chlorpropamide, glipizide, gliclazide, glibenclamide, glyburide, glibornuride, gliquidone, glisoxepide, glyclopyramide, glimepiride, JB253 and JB558, meglitinides such as repaglinide and nateglinide, biguanides such as metformin and buformin, thiazolidinediones such as rosiglitazone and pioglitazone, alpha-glucosidase inhibitors such as miglitol, acarbose and voglibose, DPP4 inhibitors such as vildagliptin, sitagliptin, saxagliptin, linagliptin, alogliptin, septagliptin and teneligliptin, GLP-1 analogues such as exenatide (also exendin-4, liraglutide, lixisenatide and taspoglutide, or SGLT inhibitors (gliflozins) such as canagliflozin, dapagliflozin and empagliflozin.
In a particularly preferred embodiment, the compounds of the present invention are administered in combination with one or more additional therapeutic agents selected from the group consisting of diuretics, angiotensin All antagonists, ACE inhibitors, beta-receptor blockers, mineralocorticoid receptor antagonists, antidiabetics, organic nitrates and NO donors, activators and stimulators of the soluble guanylate cyclase (sGC), and positive-inotropic agents.
In a further particularly preferred embodiment, the compounds of the present invention are administered in combination with one or more additional therapeutic agents selected from the group consisting of diuretics, angiotensin All antagonists, ACE inhibitors, beta-receptor blockers, mineralocorticoid receptor antagonists, antidiabetics, organic nitrates and NO donors, activators and stimulators of the soluble guanylate cyclase (sGC), positive-inotropic agents, antiinflammatory agents, immunosuppressive agents, phosphate binders and/or compounds which modulate vitamin D metabolism.
Thus, in a further embodiment, the present invention relates to pharmaceutical compositions comprising at least one of the compounds according to the invention and one or more additional therapeutic agents for the treatment and/or prevention of diseases, especially of the aforementioned dis- eases.
Furthermore, the compounds of the present invention may be utilized, as such or in compositions, in research and diagnostics, or as analytical reference standards and the like, which are well known in the art.
When the compounds of the present invention are administered as pharmaceuticals, to humans and other mammals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1% to 99.5% (more preferably, 0.5% to 90%) of active ingredient in combination with one or more pharmaceutically acceptable excipients.
Thus, in another aspect, the present invention relates to pharmaceutical compositions comprising at least one of the compounds according to the invention, conventionally together with one or more inert, non-toxic, pharmaceutically acceptable excipients, and to the use thereof for the treatment and/ or prevention of diseases, especially of the aforementioned diseases.
It is possible for the compounds according to the invention to have systemic and/or local activity. For this purpose, they can be administered in a suitable manner, such as, for example, via the oral, parenteral, pulmonary, nasal, sublingual, lingual, buccal, rectal, vaginal, dermal, transdermal, conjunctival, otic route or as an implant or stent.
For these administration routes, it is possible for the compounds according to the invention to be administered in suitable administration forms.
For oral administration, it is possible to formulate the compounds according to the invention to dosage forms known in the art that deliver the compounds of the invention rapidly and/or in a modified manner, such as, for example, tablets (uncoated or coated tablets, for example with enteric or controlled release coatings that dissolve with a delay or are insoluble), orally- disintegrating tablets, films/wafers, films/lyophylisates, capsules (for example hard or soft gelatine capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions. It is possible to incorporate the compounds according to the invention in crystalline and/or amorphised and/or dissolved form into said dosage forms.
Parenteral administration can be effected with avoidance of an absorption step (for example intravenous, intraarterial, intracardial, intraspinal or intralumbal) or with inclusion of absorption (for example intramuscular, subcutaneous, intracutaneous, percutaneous or intraperitoneal). Administration forms which are suitable for parenteral administration are, inter alia, preparations for injection and infusion in the form of solutions, suspensions, emulsions, lyophylisates or sterile powders.
Examples which are suitable for other administration routes are pharmaceutical forms for inhalation [inter alia powder inhalers, nebulizers], nasal drops, nasal solutions, nasal sprays; tablets/films/wafers/capsules for lingual, sublingual or buccal administration; suppositories; eye drops, eye ointments, eye baths, ocular inserts, ear drops, ear sprays, ear powders, ear-rinses, ear tampons; vaginal capsules, aqueous suspensions (lotions, mixturae agitandae), lipophilic suspensions, emulsions, ointments, creams, transdermal therapeutic systems (such as, for example, patches), milk, pastes, foams, dusting powders, implants or stents. The compounds according to the invention can be incorporated into the stated administration forms. This can be effected in a manner known per se by mixing with pharmaceutically suitable excipients. Pharmaceutically suitable excipients include, inter alia,
• fillers and carriers (for example cellulose, microcrystalline cellulose (such as, for example, Avicel®), lactose, mannitol, starch, calcium phosphate (such as, for example, Di-Cafos®)),
• ointment bases (for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols),
• bases for suppositories (for example polyethylene glycols, cacao butter, hard fat),
• solvents (for example water, ethanol, isopropanol, glycerol, propylene glycol, medium chain-length triglycerides fatty oils, liquid polyethylene glycols, paraffins),
• surfactants, emulsifiers, dispersants or wetters (for example sodium dodecyl sulfate), lecithin, phospholipids, fatty alcohols (such as, for example, Lanette®), sorbitan fatty acid esters (such as, for example, Span®), polyoxyethylene sorbitan fatty acid esters (such as, for example, Tween®), polyoxyethylene fatty acid glycerides (such as, for example,
Cremophor®), polyoxethylene fatty acid esters, polyoxyethylene fatty alcohol ethers, glycerol fatty acid esters, poloxamers (such as, for example, Pluronic®),
• buffers, acids and bases (for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine),
• isotonicity agents (for example glucose, sodium chloride),
• adsorbents (for example highly-disperse silicas),
• viscosity-increasing agents, gel formers, thickeners and/or binders (for example
polyvinylpyrrolidone, methylcellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose-sodium, starch, carbomers, polyacrylic acids (such as, for example, Carbopol®); alginates, gelatine),
• disintegrants (for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate (such as, for example, Explotab®), cross- linked polyvinylpyrrolidone, croscarmellose-sodium (such as, for example, AcDiSol®)),
• flow regulators, lubricants, glidants and mould release agents (for example magnesium stearate, stearic acid, talc, highly-disperse silicas (such as, for example, Aerosil®)),
• coating materials (for example sugar, shellac) and film formers for films or diffusion membranes which dissolve rapidly or in a modified manner (for example
polyvinylpyrrolidones (such as, for example, Kollidon®), polyvinyl alcohol,
hydroxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, hydroxypropylmethylcellulose phthalate, cellulose acetate, cellulose acetate phthalate, polyacrylates, polymethacrylates such as, for example, Eudragit®)),
• capsule materials (for example gelatine, hydroxypropylmethylcellulose),
• synthetic polymers (for example polylactides, polyglycolides, polyacrylates,
polymethacrylates (such as, for example, Eudragit®), polyvinylpyrrolidones (such as, for example, Kollidon®), polyvinyl alcohols, polyvinyl acetates, polyethylene oxides, polyethylene glycols and their copolymers and blockcopolymers),
• plasticizers (for example polyethylene glycols, propylene glycol, glycerol, triacetine, triacetyl citrate, dibutyl phthalate),
• penetration enhancers, • stabilisers (for example antioxidants such as, for example, ascorbic acid, ascorbyl palmitate, sodium ascorbate, butylhydroxyanisole, butylhydroxytoluene, propyl gallate),
• preservatives (for example parabens, sorbic acid, thiomersal, benzalkonium chloride, chlorhexidine acetate, sodium benzoate),
· colourants (for example inorganic pigments such as, for example, iron oxides, titanium dioxide),
• flavourings, sweeteners, flavour- and/or odour-masking agents.
The present invention furthermore relates to a pharmaceutical composition which comprise at least one compound according to the invention, conventionally together with one or more pharmaceutically suitable excipient(s), and to their use according to the present invention.
Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of cardiovascular and renal disorders, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions identified above in mammals, and by comparison of these results with the results of known active ingredients or medicaments that are used to treat these conditions, the effective dosage of the compounds of the present invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
The total amount of the active ingredient to be administered will generally range from about 0.001 mg kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 20 mg/kg body weight per day. Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing. In addition, it is possible for "drug holidays", in which a patient is not dosed with a drug for a certain period of time, to be beneficial to the overall balance between pharmacological effect and tolerability. It is possible for a unit dosage to contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day. The average daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/kg of total body weight. Illustratively, the compound of the present invention may be administered parenterally at a dose of about 0.001 mg/kg to about 10 mg/kg, preferably of about 0.01 mg/kg to about 1 mg/kg of body weight. In oral administration, an exemplary dose range is about 0.01 to 100 mg/kg, preferably about 0.01 to 20 mg/kg, and more preferably about 0.1 to 10 mg/kg of body weight. Ranges intermediate to the above-recited values are also intended to be part of the invention. Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
The following exemplary embodiments illustrate the invention. The invention is not restricted to the examples.
The percentages in the following tests and examples are, unless stated otherwise, by weight; parts are by weight. Solvent ratios, dilution ratios and concentrations reported for liquid/liquid solutions are each based on volume.
EXPERIMENTAL SECTION
The ^-NMR data are listed in the form of ^-NMR peaklists. For each signal peak the δ value in ppm is given, followed by the signal intensity, reported in round brackets. The δ value-signal intensity pairs from different peaks are separated by commas. Therefore, a peaklist is described by the general form: δι (intensityi), 82 (intensity2), ... , δί (intensity;), ... , δη (intensityn).
The intensity of a sharp signal correlates with the height (in cm) of the signal in a printed NMR spectrum. When compared with other signals, this data can be correlated to the real ratios of the signal intensities. In the case of broad signals, more than one peak, or the center of the signal along with their relative intensity, compared to the most intense signal displayed in the spectrum, are shown. A ^-NMR peaklist is similar to a classical ^-NMR readout, and thus usually contains all the peaks listed in a classical NMR interpretation. Moreover, similar to classical ^-NMR printouts, peaklists can show solvent signals, signals derived from stereoisomers of target compounds (also the subject of the invention), and/or peaks of impurities. The peaks of stereoisomers, and/or peaks of impurities are typically displayed with a lower intensity compared to the peaks of the target compounds (e.g., with a purity of >90%). Such stereoisomers and/or impurities may be typical for the particular manufacturing process, and therefore their peaks may help to identify the reproduction of our manufacturing process on the basis of "by-product fingerprints". An expert who calculates the peaks of the target compounds by known methods (MestReC, ACD simulation, or by use of empirically evaluated expectation values), can isolate the peaks of target compounds as required, optionally using additional intensity filters. Such an operation would be similar to peak-picking in classical ^-NMR interpretation. A detailed description of the reporting of NMR data in the form of peaklists can be found in the publication "Citation of NMR Peaklist Data within Patent Applications" (cf. Research Disclosure Database Number 605005, 2014, 01 Aug 2014, or http://www.researchdisclosure.com/searching- disclosures). In the peak picking routine, as described in the Research Disclosure Database Number 605005, the parameter "MinimumHeight" can be adjusted between 1% and 4%. Depending on the chemical structure and/or depending on the concentration of the measured compound it may be reasonable to set the parameter "MinimumHeight" <1%.
Chemical names were generated using the ACD/Name software from ACD/Labs. In some cases generally accepted names of commercially available reagents were used in place of ACD/Name generated names.
The following table 1 lists the abbreviations used in this paragraph and in the Examples section as far as they are not explained within the text body. Other abbreviations have their meanings customary per se to the skilled person. Table 1: Abbreviations
The following table lists the abbreviations used herein.
Meaning
broad OH-NMR signal)
concentrated
chemical ionisation
doublet
double-doublet
dimethylsulfoxide
double-triplet
Evaporative Light Scattering Detector
electrospray (ES) ionisation
hour(s)
hydrochloric acid
high performance liquid chromatography
liquid chromatography mass spectrometry
multiplet
minute(s)
MS mass spectrometry
NMR nuclear magnetic resonance spectroscopy: chemical shifts (δ) are given in ppm. The chemical shifts were corrected by setting the DMSO signal to 2.50 ppm unless otherwise stated.
PDA Photo Diode Array q quartet
Rt retention time (as measured either with HPLC or UPLC) in minutes s singlet
SQD Single-Quadrupole-Detector
triplet triple-doublet
tetrahydrofuran
ultra performance liquid chromatography
The various aspects of the invention described in this application are illustrated by the following examples which are not meant to limit the invention in any way.
The example testing experiments described herein serve to illustrate the present invention and the invention is not limited to the examples given.
EXPERIMENTAL SECTION - GENERAL PART
All reagents, for which the synthesis is not described in the experimental part, are either commercially available, or are known compounds or may be formed from known compounds by known methods by a person skilled in the art.
The compounds and intermediates produced according to the methods of the invention may require purification. Purification of organic compounds is well known to the person skilled in the art and there may be several ways of purifying the same compound. In some cases, no purification may be necessary. In some cases, the compounds may be purified by crystallization. In some cases, impurities may be stirred out using a suitable solvent. In some cases, the compounds may be purified by chromatography, particularly flash column chromatography, using for example prepacked silica gel cartridges, e.g. Biotage SNAP cartidges KP-Sil® or KP-NH® in combination with a Biotage autopurifier system (SP4® or Isolera One®) and eluents such as gradients of hexane/ethyl acetate or DCM/methanol. In some cases, the compounds may be purified by preparative HPLC using for example a Waters autopurifier equipped with a diode array detector and/or on-line electrospray ionization mass spectrometer in combination with a suitable prepacked reverse phase column and eluents such as gradients of water and acetonitrile which may contain additives such as trifluoroacetic acid, formic acid or aqueous ammonia.
In some cases, purification methods as described above can provide those compounds of the present invention which possess a sufficiently basic or acidic functionality in the form of a salt, such as, in the case of a compound of the present invention which is sufficiently basic, a trifluoroacetate or formate salt for example, or, in the case of a compound of the present invention which is sufficiently acidic, an ammonium salt for example. A salt of this type can either be transformed into its free base or free acid form, respectively, by various methods known to the person skilled in the art, or be used as salts in subsequent biological assays. It is to be understood that the specific form (e.g. salt, free base etc.) of a compound of the present invention as isolated and as described herein is not necessarily the only form in which said compound can be applied to a biological assay in order to quantify the specific biological activity.
UPLC-MS Standard Procedures
Method 1:
Instrument: Waters ACQUITY SQD UPLC System; Column: Waters Acquity UPLC HSS T3 1.8 μ 50 x 1 mm; eluent A: 1 L water + 0.25 ml 99% formic acid , eluent B: 1 L acetonitrile + 0.25 ml 99% formic acid; gradient: 0.0 min 90% A→ 1.2 min 5% A→ 2.0 min 5% A; oven: 50°C; fow rate: 0.40 ml/min; UV detection: 208 - 400 nm.
Method 2:
Instrument MS: Thermo Scientific FT-MS; Instrument type UHPLC+: Thermo Scientific UltiMate 3000; Column: Waters, HSST3, 2.1 x 75 mm, C18 1.8 μιη; eluent A: 1 L water + 0.01% formic acid; eluent B: 1 L acetonitrile + 0.01% formic acid; gradient: 0.0 min 10% B→ 2.5 min 95% B→ 3.5 min 95% B; oven: 50°C; flow rate: 0.90 ml/min; UV detection: 210 nm/ optimum integration path 210-300 nm
Method 3:
Instrument: Agilent MS Quad 6150; HPLC: Agilent 1290; Column: Waters Acquity UPLC HSS T3 1.8 μ 50 x 2.1 mm; eluent A: 1 L water + 0.25 ml 99% formic acid , eluent B: 1 L acetonitrile + 0.25 ml 99% formic acid; gradient: 0.0 min 90% A→ 0.3 min 90% A→ 1.7 min 5% A→ 3.0 min 5% A; oven: 50°C; flow rate: 1,20 ml/min; UV detection: 205 - 305 nm.
Method 4:
Instrument MS: Waters (Micromass) Quattro Micro; Instrument Waters UPLC Acquity; Column : Waters BEH C18 1.7 μ 50 x 2.1 mm; eluent A: 1 L water + 0.01 mol ammonium formiate, eluent B: 1 L acetonitrile; gadient: 0.0 min 95% A→ 0.1 min 95% A→ 2.0 min 15% A→ 2.5 min 15% A→ 2.51 min 10% A→ 3.0 min 10% A; oven: 40°C; flow rate: 0.5 ml/min; UV detection: 210 nm Method 5:
Instrument: Waters ACQUITY SQD UPLC System; Column: Waters Acquity UPLC HSS T3 1.8 μ 50 x 1 mm; eluent A: 1 L water + 0.25 ml 99% formic acid, eluent B: 1 L acetonitrile + 0.25 ml 99% formic acid; gradient: 0.0 min 95% A→ 6.0 min 5% A→ 7.5 min 5% A; oven: 50°C; flow rate: 0.35 ml/min; UV detection: 210 - 400 nm.
Method 6
Column: Chromatorex or Reprosil; C18; 10 μιη; 125 x 30 mm; Eluent A: 10 L Water + 20 ml 50% Formic acid, Eluent B: Acetonitrile; Gradient: 0-5.50 min 10% B : 90% A; Sample injection: at 3.00 min; 5.50 min 90% A : 10% B→ 17.65 min 5% A : 95% B; 17.65-19.48 min 5% A : 95% B; Flow: 75 ml/min; UV- Detection: 210 nm.
EXPERIMENTAL SECTION - STARTING MATERIALS AND INTERMEDIATES
Example 1A
5-(4-Chlorophenyl)-4-[(2R)-33,3-trifluoro-2-hydroxypropyl]-2,4-dihydro-3H-l,2,4-triazol-3-one
A solution of 5-(4-chlorophenyl)-4-(3,3,3-trifluoro-2,2-dihydroxypropyl)-2,4-dihydro-3H- 1,2,4- triazol-3-one (synthesis described as Example 3A in WO 2010/105770-A1) (10.0 g, 30.9 mmol), N-[(lR,2R)-2-amino-l,2-diphenylethyl]-4-methylbenzenesulfonamide (56.6 mg, 154 μιηοΐ) and 1- methyl-4-(propan-2-yl)benzene - dichlororuthenium (1 : 1) (47.3 mg, 77.2 μιηοΐ) in ethyl acetate was treated with triethylamine (8.6 ml, 62 mmol) followed by formic acid (5.8 ml, 150 mmol). The resulting mixture was refluxed for 3 h and cooled down to room temperature. The reaction mixture was diluted with IN HCl (70 ml). The organic phase was washed twice IN HCl. The aqueous phase was extracted twice with ethyl acetate. The combined organic phases were evaporated. The residue was retaken in methanol (22.5 ml) and the resulting suspension was heated to 60°C until the solid was completely dissolved. IN HCl (22.5 ml) were added and the resulting suspension was heated at 78°C for 10 min and cooled down to room temperature. The solid was filtered off and dried under vacuum. The solid was retaken in IN HCl (30 ml), heated at 35°C. The resulting suspension was treated with methanol (30 ml), heated 4 h at 35°C and filtered off at 35°C. The mulat was evaporated affording 4.9 g (ee = 99.6%, 51% th.)of 5-(4-Chlorophenyl)-4-[(2R)-3,3,3-trifluoro-2- hydroxypropyl]-2,4-dihydro-3H-l,2,4-triazol-3-one.
LC-MS (Method 2): Rt = 1.40 min; MS (ESIpos): m/z = 308 [M+H]+
¾-NMR (400MHz, DMSO): δ [ppm] = 12.10 (s, 1H), 7.52 - 7.79 (m, 4H), 6.84 (d, 1H), 3.54 - 4.52 (m, 3H).
Example 2A
{3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol- l-yl}acetonitrile
In a 2 L reaction vessel, 100 g (273 mmol) of {3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-l/ ,2,4-triazol-l-yl}acetic acid (synthesis described as Example 8A in WO 2010/105770-A1), 43.3 g (547 mmol) of pyridine and 33 mg (0.3 mmol) of 4- dimethylaminopyridine were dissolved in 300 ml THF. The resulting solution was treated at 5 °C with 52.8 g (438 mmol) of 2,2-dimethylpropanoylchloride over 15 minutes and the resulting mixture was stirred at room temperature for 2.5 hours. After cooling to 0 °C, 183 ml of 28% aqueous ammonia solution was added over 1 h while the solution temperature was kept between 10 °C and 20 °C and at the resulting mixture then stirred at 5 °C for an additional time period of 1 h. 500 ml methyl tert-butylether and 300 ml 20% aqueous citric acid were then added while keeping the internal temperature between 10 °C and 20 °C. The phases were the separated and the organic phase was washed with 300 ml of 20% aqueous citric acid followed by 300 ml saturated aqueous sodium hydrogencarbonate solution and finally with 300 ml of 10% aqueous sodium chloride solution. The organic phase was evaporated at 60 °C under reduced pressure until an oily residue was obtained. 300 ml THF was then added and the solution was evaporated again until an oily solution was obtained. This operation was repeated a second time. The oil residue was retaken in 360 ml THF and treated with 172 g (820 mmol) trifluoroacetic acid anhydride over 20 min at a temperature between 10 °C and 20 °C. The resulting solution was then stirred at room temperature for 1 h. 720 ml 4-methyl-2-pentanone and 650 ml 7.5% aqueous sodium hydroxide solution were added at a temperature between 10 °C and 20 °C. Finally the pH-value was adjusted to pH = 9.5 using 7.5% aqueous sodium hydroxide solution. After phase separation, the organic phase was washed twice with 450 ml 10% aqueous sodium chloride solution. The organic phase was evaporated at a temperature of 80 °C under reduced pressure while 1200 ml n-heptane was added. The formed suspension was cooled to 20 °C and a solid formed which was filtered off and washed with 200 ml n-heptane and then dried under reduced pressure (50°C, 30 mbar) affording 88 g (93 % of th.) of {3-(4-chlorophenyl)-5-oxo-4-[(2lS')-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-l i- l,2,4-triazol-l-yl}acetonitrile as a solid.
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 7.78 (d, 2H), 7.55 (d, 2H), 6.91 (d, 1H), 5.17 (s, 2 H), 4.34-4.23 (m, 1 H), 3.98 (dd, 1H), 3.81 (dd, 1H). Example 3A
{3-(4-Chlorophenyl)-5-oxo-4-[(2R)-333-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol- l-yl}acetonitrile
A solution of 40 g (130 mmol) 5-(4-Chlorophenyl)-4-[(2R)-3,3,3-trifluor-2-hydroxypropyl]-2,4- dihydro-3H-l,2,4-triazol-3-one (Example 1A) in 400 ml methylisobutyl ketone was treated with 17.9 g (143 mmol) bromoacetonitrile and 53.9 g (390 mmol) potassium carbonate and stirred for 4 hours at 60°C. After cooling to 20 °C, 200 ml water was added and the mixture was stirred for 10 min. After phase separation, the organic phase was washed with 200 ml water. The organic phase was evaporated at 80 °C under reduced pressure while 300 ml n-heptane was added. The formed suspension was cooled to 20 °C and a solid formed which was filtered off and and washed with 50 ml n-heptane and then dried under reduced pressure (50°C, 30 mbar) affording 25.2 g (56 % of th.) of {3-(4-chlorophenyl)-5-oxo-4-[(2R)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lii-l,2,4- triazol- 1 -yl } acetonitrile.
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 7.78 (d, 2H), 7.55 (d, 2H), 6.91 (d, 1H), 5.17 (s, 2 H), 4.34-4.23 (m, 1 H), 3.98 (dd, 1H), 3.81 (dd, 1H).
Example 4A
Methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl Jethanimidate
In a 4 L reaction vessel, 200 g (576.9 mmol) of {3-(4-chlorophenyl)-5-oxo-4-[(2lS)-3,3,3-trifluoro- 2-hydroxypropyl]-4,5-dihydro-lii-l,2,4-triazol-l-yl}acetonitrile (Example 2A) in 1600 ml methanol was treated with 5.2 g (28 mmol) sodium methanolate (30% in methanol) and the resulting mixture was stirred at 50 °C for 2.5 hours. The solution was then evaporated at 50 °C under reduced pressure until an oily solution was obtained. 2000 ml methyl tert-butylether was added and the solution was concentrated until a volume of 800 ml was achieved. 3000 ml n- heptane was then added and a suspension was formed. After cooling at 20 °C, the solid was filtered and washed with 500 ml n-heptane and then dried under reduced pressure (50°C, 30 mbar) affording 175 g (80 % of th.) of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } ethanimidateas a solid.
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 3.67 (s, 3 H), 3.81 (dd, 1H), 3.96 (dd, 1H), 4.23-4.35 (m, 1 H), 4.50 (s, 2 H), 6.93 (br. s, 1H), 7.62 (d, 2H), 7.78 (d, 2H), 8.01 (s, 1H).
Example 5A
Methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl Jethanimidate
In a 4 L reaction vessel, 200 g (576.9 mmol) of {3-(4-chlorophenyl)-5-oxo-4-[(2,S,)-3,3,3-trifluoro-
2- hydroxypropyl]-4,5-dihydro-lii-l,2,4-triazol-l-yl}acetonitrile (Example 3A) in 1600 ml methanol was treated with 5.2 g (28 mmol) sodium methanolate (30% in methanol) and the resulting mixture was stirred at 50 °C for 2.5 hours. The solution was then evaporated at 50 °C under reduced pressure until an oily solution was obtained. 2000 ml methyl tert-butylether was added and the solution was concentrated until a volume of 800 ml was achieved. 3000 ml n- heptane was then added and a suspension was formed. After cooling at 20 °C, the solid was filtered and washed with 500 ml n-heptane and then dried under reduced pressure (50°C, 30 mbar) affording 175 g (80 % of th.) of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } ethanimidateas a solid.
1H-NMR (400 MHz, DMSO-d6): δ [ppm] = 3.67 (s, 3 H), 3.81 (dd, 1H), 3.96 (dd, 1H), 4.23-4.35 (m, 1 H), 4.50 (s, 2 H), 6.93 (br. s, 1H), 7.62 (d, 2H), 7.78 (d, 2H), 8.01 (s, 1H).
Example 6A
3- Ethyl-2-hydrazinylpyridine
2-Bromo-3-ethylpyridine (950 mg, 5.11 mmol) was dissolved in hydrazine hydrate (1 : 1) (7.5 ml) and stirred at reflux for 48 h. The reaction mixture was stored at 4°C for 72 h and the precipitate was collected by filtration and washed with water. The solid was dried under high vacuum affording 273 mg (39 % of th.) of the title compound.
LC-MS (Method 4): Rt = 0.86 min; MS (ESIpos): m/z = 138 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 7.94 (d, 1H), 7.35-7.06 (m, 2H), 6.57 (t, 1H), 4.10 (br. s, 2H), 2.41 (q, 2H), 1.11 (t, 3H).
Example 7A
2-Hydrazinyl-3-methylpyridine
2-Bromo-3-methylpyridine (1.46 g, 8.50 mmol) was dissolved in hydrazine hydrate (1 : 1) (10 ml) and stirred at reflux overnight. The reaction mixture was cooled to room temperature and diluted with water. The precipitate was collected by filtration and washed with water. The solid was dried under high vacuum affording 360 mg (34 % of th.).
LC-MS (Method 4): Rt = 0.74 min; MS (ESIpos): m/z = 124 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 7.94 (br. d, 1H), 7.27-6.97 (m, 2H), 6.53 (dd, 1H), 4.12 (br. s, 2H), 2.02 (s, 3H).
Example 8A
4-Hydrazinyl-3-methylpyridine
4-Chloro-3-methylpyridine hydrochloride (1 : 1) (1.00 g, 6.10 mmol) was dissolved in hydrazine hydrate (1 : 1) (10 ml) and stirred at reflux overnight. The reaction mixture was cooled to 4°C for 6 h. The precipitate was collected by filtration and washed with water. The solid was dried under high vacuum affording 220 mg (29 % of th.).
LC-MS (Method 4): Rt = 0.32 min; MS (ESIpos): m/z = 124 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 8.01 (d, 1H), 7.83 (s, 1H), 7.04 (br s, 1H), 6.87 (d, 1H), 4.12 (br s, 2H), 1.97 (s, 3H).
Example 9A
2-Hydrazinyl-3-methoxypyridine
2-Chloro-3-methoxypyridine (1.22 g, 8.46 mmol) was dissolved in hydrazine hydrate (1 : 1) (10 ml) and stirred at reflux for 4h. The reaction mixture was cooled to room temperature and evaporated. The residue was retaken in 10% MeOH in CHCI3 and washed with an aqueous K2CO3 solution. The aqueous phase was extracted with 10% MeOH in CHCI3 . The combined organic layers were dried over magnesium sulfate and evaporated affording 781 mg (59% of th.) of the title compound.
LC-MS (Method 4): Rt = 0.76 min; MS (ESIpos): m/z = 140 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 7.65 (dd, 1H), 7.09-6.87 (m, 2H), 6.56 (dd, 1H), 4.37- 3.85 (br. s, 2H), 3.76 (s, 3H)
Example 10A
Ethyl 2-hydrazinylpyridine-3-carboxylate
Ethyl 2-chloropyridine-3-carboxylate (1.00 g, 5.39 mmol) was dissolved in hydrazine in THF (22 ml, 1M, 22 mmol) and stirred 16h at room temperature. Hydrazine in THF (11 ml, lM, 11 mmol) was added and the resulting mixture was stirred 24h at room temperature. The reaction mixture was evaporated. The residue was retaken in 10% MeOH in CHCI3 and washed with an aqueous K2CO3 solution. The aqueous phase was extracted with 10% MeOH in CHCI3. The combined organic layers were dried over magnesium sulfate and evaporated affording 1.87 g (purity 50%, 59% of th.) of the title compound. LC-MS (Method 4): Rt = 1.21 min; MS (ESIpos): m/z = 182 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 8.54 (br. s, 1H), 8.34 (dd, 1H), 8.08 (dt, 1H), 6.67 (dd, 1H), 4.44 (br. s, 2H), 4.28 (q, 2H), 1.30 (t, 3H).
Example 11A
3-Fluoro-4-hydrazinylpyridine
4-Chloro-3-fluoropyridine (1.00 g, 7.60 mmol) was dissolved in hydrazine hydrate (1 : 1) (13 ml) and stirred at reflux for 1 h. The reaction mixture was cooled to 4°C overnight. The precipitate was collected by filtration and washed with water. The solid was dried under high vacuum affording 105 mg ( 10% of th.) of the title compound.
LC-MS (Method 4): Rt = 0.42 min; MS (ESIpos): m/z = 128 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 8.00 (m, 2H), 7.68 (br s, 1H), 7.03 (dd, 1H), 4.24 (br. d, 2H).
Example 12A
2-Chloro-3-hydrazinyl-4-methylpyridine
At -20°C, solution of 2-chloro-4-methylpyridin-3-amine (2.64 g, 18.5 mmol) in cone. HC1 (15.5 ml) was treated with a solution of NaNC>2 (1.40 g, 20.4 mmol) in water (15 ml) and stirred 1 h. A solution of SnC (12.5g, 55.5 mmol) in cone. HC1 (7.5 ml) was then added dropwise and the resulting mixture was stirred 1 h while the temperature is maintained between -20°C and -15°C. The reaction mixture was adjusted to pH 10-11 with an 40% aqueous KOH solution and extracted twice with ethyl acetate. The tin salts where filtered off and the combined organic phase where washed with brine and dried over magnesium sulfate affording 1.85g (60% of th.) of the title compound.
LC-MS (Method 4): Rt = 0.81 min; MS (ESIpos): m/z = 158 [M+H]+ Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 7.74 (d, 1H), 7.09 (d, 1H), 5.95 (s, 1H), 4.30 (s, 2H), 2.41 (s, 3H).
Example 13A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-ethylpyridin-2-yl)-lH-l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 610 mg, 1.61 mmol) in dioxane (12 ml) was treated at 0°C with N,N-diisopropylethylamine (340 μΐ, 1.9 mmol) and methyl chloro(oxo) acetate (180 μΐ, 1.9 mmol) and stirred at 0°C for 30 min. A suspension of 3- ethyl-2-hydrazinylpyridine (Example 6A, 265 mg, 1.93 mmol) and copper sulfate (308 mg, 1.93 mmol) in dioxane (6 ml) were added dropwise. The resulting mixture was stirred overnight at room temperature, diluted with ethyl acetate and washed with water. The organic phase was extracted with ethyl acetate. The combined organic phases were washed with brine, dried over magnesium sulfate and evaporated. The residue was purified by flash chromatography (silica gel, CH2Cl2/MeOH gradient) affording 267 mg (25 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.87 min; MS (ESIpos): m/z = 552 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.41 (dd, 1H), 8.02 (dd, 1H), 7.83-7.54 (m, 5H), 6.91 (d, 1H), 5.20 (s, 2H), 4.31-3.57 (m, 6H), 2.42 (q, 2H), 1.02 (t, 3H).
Example 14A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-methylpyridin-2-yl)-lH-l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 897 mg, 2.37 mmol) in dioxane (18 ml) was treated at 0°C with N,N-diisopropylethylamine (500 μΐ, 2.8 mmol) and methyl chloro(oxo) acetate (260 μΐ, 2.8 mmol) and stirred at 0°C for 30 min. A suspension of 2- hydrazinyl-3-methylpyridine (Example 7A, 350 mg, 2.84 mmol) and copper sulfate (454 mg, 2.84 mmol) of dioxane (6 ml) were added dropwise. The resulting mixture was stirred overnight at room temperature, diluted with ethyl acetate and washed with water. The organic phase was extracted with ethyl acetate. The combined organice phase were washed with brine, dried over magnesium sulfate and evaporated. The residue was purified by flash chromatography (silica gel, CH2Cl2/MeOH gradient) followed by preparative HPLC (Method 6) affording 530 mg (41 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.77 min; MS (ESIpos): m/z = 538 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.40 (d, 1H), 7.98 (d, 1H), 7.80-7.51 (m, 5H), 6.91 (d, 1H), 5.19 (s, 2H), 4.44-3.65 (m, 6H), 2.11 (s, 3H).
Example 15A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-methylpyridin-4-yl)-lH-l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 396 μιηοΐ) in THF (3.0 ml) was treated at 0°C with N,N-diisopropylethylamine (76 μΐ, 440 μιηοΐ) and methyl chloro(oxo)acetate (40 μΐ, 440 μmol) and stirred at 0°C for 30 min. 4-hydrazinyl-3-methylpyridine (Exampe 8A, 53.7 mg, 436 μιηοΐ) was added dropwise and the resulting mixture was stirred 1 h at room temperature and 1 h at 120°C under microwave irradiation. Purification by preparative HPLC (R Method 6) afforded 28.3 mg (11 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.68 min; MS (ESIpos): m/z = 538 [M+H]+
Example 16A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-methoxypyridin-2-yl)-lH-l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4 A, 1.00 g, 2.64 mmol) in dioxane (20 ml) was treated at 0°C with N,N-diisopropylethylamine (550 μΐ, 3.2 mmol) and methyl chloro(oxo)acetate (290 μΐ, 3.2 mmol) and stirred at 0°C for 30 min. A suspension of 2- hydrazinyl-3-methoxypyridine (Example 9A, 848 mg, 52 % purity, 3.17 mmol) and copper sulfate (506 mg, 3.17 mmol) in dioxane (10 ml) were added dropwise. The resulting mixture was stirred overnight at room temperature, diluted with ethyl acetate and washed with water. The organic phase was extracted with ethyl acetate. The combined organice phase were washed with brine, dried over magnesium sulfate and evaporated. The residue was purified by flash chromatography (silica gel, CEbCVMeOH gradient) followed by preparative HPLC (Method 6) affording 604 mg (38 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.71 min; MS (ESIpos): m/z = 554 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.16 (dd, 1H), 7.92-7.54 (m, 6H), 6.92 (d, 1H), 5.18 (s, 2H), 4.50-3.66 (m, 9H).
Example 17A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(3-fluoropyridin-2-yl)- 1H- l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4 A, 1.00 g, 2.64 mmol) in THF (20 ml) was treated at 0°C with N,N-diisopropylethylamine (510 μΐ, 2.9 mmol) and methyl chloro(oxo)acetate (270 μΐ, 2.9 mmol) and stirred at 0°C for 30 min. 3-fluoro-2-hydrazinylpyridine (369 mg, 2.90 mmol) was added dropwise and the resulting mixture was stirred 1 h at room temperature and 3 h at 120°C under microwave irradiation. Purification by preparative HPLC (Method 6) afforded 700 mg (38 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.81 min; MS (ESIpos): m/z = 542 [M+H]+
Example 18A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-chloropyridin-4-yl)-lH-l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 396 μιηοΐ) in THF (3.0 ml) was treated at 0°C with N,N-diisopropylethylamine (75 μΐ, 435 μιηοΐ) and methyl chloro(oxo)acetate (40 μΐ, 440 μιηοΐ) and stirred at 0°C for 30 min. 3-chloro-4-hydrazinylpyridine (62.5 mg, 436 μιηοΐ) was added followed by N,N-diisopropylethylamine (75 μΐ, 435 μιηοΐ) and the resulting mixture was stirred 1 h at room temperature and 1 h at 120°C under microwave irradiation. Purification by preparative HPLC (Method 6) afforded 38.3 mg (16 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.25 min; MS (ESIpos): m/z = 558 [M+H]+ Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 9.02-8.63 (m, 2H), 7.93-7.50 (m, 5H), 6.91 (d, 1H), 5.21 (s, 2H), 4.51-3.63 (m, 6H).
Example 19A
Ethyl 2-[3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl }methyl)-5-(methoxycarboriyl)- 1 H- 1 ,2,4-triazol- 1 -yl]pyridine-3-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 2.35 g, 6.19 mmol) in dioxane (47 ml) was treated at 0°C with N,N-diisopropylethylamine (1.3 ml, 7.4 mmol) and methyl chloro(oxo)acetate (680 μΐ, 7.4 mmol) and stirred at 0°C for 30 min. A suspension of ethyl 2-hydrazinylpyridine-3-carboxylate (Example 10A, 1.87 g, 72 % purity, 7.43 mmol) and copper sulfate (1.45 g, 9.08 mmol) in dioxane (23 ml) were added dropwise. The resulting mixture was stirred 72 h at room temperature. Purification by flash chromatography (silica gel, CE CVMeOH gradient) followed by preparative HPLC (Method 6) affording 833 mg (23 % of th.) of the title compound.
LC-MS (Method 1): Rt = 0.98 min; MS (ESIpos): m/z = 596 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.94-8.36 (m, 2H), 8.11-7.44 (m, 5H), 6.91 (d, 1H), 5.17 (s, 2H), 4.58-3.66 (m, 8H), 0.97 (t, 3H).
Example 20A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(3-fluoropyridin-4-yl)- 1H- l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (250 mg, 660 μιηοΐ) in THF (5.0 ml) was treated at 0°C with N,N-diisopropylethylamine (130 μΐ, 730 μmol) and methyl chloro(oxo)acetate (67 μΐ, 730 μιηοΐ) and stirred at 0°C for 30 min. 3-fluoro-4-hydrazinylpyridine (Example 11 A, 92.3 mg, 726 μιηοΐ) was added and the resulting mixture was stirred 1 h at room temperature and 1 h at 120°C under microwave irradiation. Purification by preparative HPLC (Method 6) afforded 78.0 mg (21 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.73 min; MS (ESIpos): m/z = 542 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.91 (d, 1H), 8.68 (d, 1H), 8.01-7.48 (m, 5H), 6.91 (br. s, 1H), 5.21 (s, 2H), 4.49-3.71 (m, 6H).
Example 21 A
3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(3-methoxypyrazin-2-yl)-lH-l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 242 mg, 640 μιηοΐ) in THF (5.0 ml, 62 mmol) was treated at 0°C with N,N-diisopropylethylamine (123 μΐ, 0.70 mmol) and methyl chloro(oxo) acetate (65 μΐ, 700 μιηοΐ) and stirred at 0°C for 30 min. 2-hydrazinyl-3- methoxypyrazine dihydrochloride (150 mg, 704 μιηοΐ) followed by N,N-diisopropylethylamine (437 μΐ, 2.5 mmol) were added and the resulting mixture was stirred 1 h at room temperature and 1 h at 120°C under microwave irradiation. Purification by preparative HPLC (Method 6) afforded 49 mg (85% of th.) of the title compound.
LC-MS (Method 3): Rt = 1.27 min; MS (ESIpos): m/z = 555 [M+H]+
Example 22A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(2-chloropyridin-3-yl)-lH-l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 300 mg, 792 μιηοΐ) in THF (6.0 ml) was treated at 0°C with N,N-diisopropylethylamine (150 μΐ, 0.85 mmol) and methyl chloro(oxo) acetate (80 μΐ, 870 μιηοΐ) and stirred at 0°C for 30 min. 2-chloro-3- hydrazinylpyridine hydrochloride (1: 1) (157 mg, 871 μιηοΐ) followed by N,N- diisopropylethylamine (150 μΐ, 0.85 mmol) were added and the resulting mixture was stirred overnight at room temperature and 30 min at 120°C under microwave irradiation. Purification by preparative HPLC (Method 6) afforded 145 mg (33 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.80 min; MS (ESIpos): m/z = 558 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.64 (dd, 1H), 8.24 (dd, 1H), 7.88-7.53 (m, 5H), 6.91 (d, 1H), 5.20 (s, 2H), 4.50-3.70 (m, 6H).
Example 23A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(pyridin-2-yl)- 1H- l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4 A, 1.00 g, 2.64 mmol) in THF (20 ml) was treated at 0°C with N,N-diisopropylethylamine (0.5 ml, 2.9 mmol) and methyl chloro(oxo)acetate (270 μΐ, 2.9 mmol) and stirred at 0°C for 30 min. 2-hydrazinylpyridine hydrochloride (1: 1) (423 mg, 2.90 mmol) followed by N,N-diisopropylethylamine (0.5 ml, 2.9 mmol) were added and the resulting mixture was stirred 1 h at room temperature and 1 h at 120°C under microwave irradiation. Purification by preparative HPLC (Method 6) afforded 483 mg (33 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.79 min; MS (ESIpos): m/z = 524
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 8.61-8.41 (m, 1H), 8.12 (td, 1H), 7.93-7.50 (m, 6H), 6.91 (d, 1H), 5.19 (s, 2H), 4.49-3.75 (m, 6H).
Example 24A
Methyl l-(2-chloro-4-methylpyridin-3-yl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 396 μιηοΐ) in THF (3.0 ml) was treated at 0°C with N,N-diisopropylethylamine (76 μΐ, 440 μιηοΐ) and methyl chloro(oxo)acetate (40 μΐ, 440 μιηοΐ) and stirred at 0°C for 30 min. 2-chloro-3-hydrazinyl-4- methylpyridine (Example 12A, 68.7 mg, 436 μιηοΐ) was added and the resulting mixture was stirred overnight at room temperature and 1 h at 120°C under microwave irradiation. Purification by preparative HPLC (Method 6) afforded 61.0 mg (26 % of th.) of the title compound.
LC-MS (Method 1): Rt = 0.98 min; MS (ESIpos): m/z = 572 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.49 (d, 1H), 7.89-7.50 (m, 5H), 6.91 (d, 1H), 5.22 (m, 2H), 4.30-3.70 (m, 6H), 2.06 (s, 3H).
Example 25A Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihyd] 1 ,2,4-triazol- 1 -yl } methyl)- l-(2-ethoxypyridin-3-yl)- 1H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 396 μιηοΐ) in THF (3.0 ml) was treated at 0°C with N,N-diisopropylethylamine (76 μΐ, 440 μmol) and methyl chloro(oxo)acetate (40 μΐ, 440 μιηοΐ) and stirred at 0°C for 30 min. 2-ethoxy-3-hydrazinylpyridine (70.2 mg, 436 μιηοΐ) was added and the resulting mixture was stirred 1.5 h at room temperature. Purification by preparative HPLC (Method 6) afforded 170 mg (73 % of th.) of the title compound. LC-MS (Method 2): Rt = 1.92 min; MS (ESIpos): m/z = 568 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.32 (dd, 1H), 7.97 (dd, 1H), 7.86-7.54 (m, 4H), 7.31- 6.78 (m, 2H), 5.17 (s, 2H), 4.43-4.20 (m, 3H), 4.30-3.70 (m, 5H), 1.16 (t, 3H).
Example 26A
Ethyl 3-[3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl }methyl)-5-(methoxycarbonyl)- 1 H- 1 ,2,4-triazol- 1 -yl]pyridine-4-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 500 mg, 1.32 mmol) in THF (10 ml) was treated at 0°C with N,N-diisopropylethylamine (250 μΐ, 1.5 mmol) and methyl chloro(oxo)acetate (130 μΐ, 1.5 mmol) and stirred at 0°C for 30 min. Ethyl 3- hydrazinylpyridine-4-carboxylate (310 mg, 1.45 mmol) was added and the resulting mixture was stirred overnight at room temperature and 10 h at reflux temperature. Purification by preparative HPLC (Method 6) afforded 417 mg (48 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.83 min; MS (ESIpos): m/z = 596 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 9.00-8.87 (m, 2H), 7.96 (d, 1H), 7.75 (d, 2H), 7.63 (d, 2H), 6.91 (d, 1H), 5.17 (s, 2H), 4.41-3.72 (m, 8H), 0.93 (t, 3H).
Example 27A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl Jmethyl)- l-(4-chloropyridin-2-yl)- 1H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 631 mg, 1.67 mmol) in dioxane (12.7 ml) was treated at 0°C with N,N-diisopropylethylamine (350 μΐ, 2.0 mmol) and methyl chloro(oxo) acetate (180 μΐ, 2.0 mmol) and stirred at 0°C for 30 min. A suspension of 4- chloro-2-hydrazinylpyridine (287 mg, 2.00 mmol) and copper sulfate (319 mg, 2.00 mmol) in dioxane (6.3 ml) were added dropwise. The resulting mixture was stirred overnight at room temperature, diluted with ethyl acetate and washed with water. The organic phase was extracted with ethyl acetate. The combined organice phase were washed with brine, dried over magnesium sulfate and evaporated. The residue was purified by flash chromatography (silica gel, cyclohexane/ethyl acetate gradient) affording 683 mg (73 % of th.).
LC-MS (Method 3): Rt = 1.30 min; MS (ESIpos): m/z = 558 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.62 (d, 1H), 7.96 (d, 1H), 7.84-7.57 (m, 5H), 6.91 (d, 1H), 5.18 (s, 2H), 4.41-3.76 (m, 6H).
Example 28A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(pyrimidin-2-yl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 396 μιηοΐ) in THF (3.0 ml) was treated at 0°C with N,N-diisopropylethylamine (76 μΐ, 440 μmol) and methyl chloro(oxo)acetate (40 μΐ, 440 μmol) and stirred at 0°C for 30 min. 2-hydrazinylpyrimidine (48.0 mg, 436 μmol) was added and the resulting mixture was stirred overnight at room temperature. Purification by preparative HPLC (Method 6) afforded 115 mg (53 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.67 min; MS (ESIpos): m/z = 525
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 9.01 (d, 2H), 7.82-7.58 (m, 5H), 6.92 (d, 1H), 5.20 (s, 2H), 4.41-3.74 (m, 6H).
Example 29A
Methyl l-(2-bromophenyl)-3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxy- propyl] -4,5-dihydro- 1H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 97 mg (0.44 mmol) of (2-bromophenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of N-diisopropylethylamine. The resulting mixture was stirred 2 h at RT, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 121 mg (48 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.99 min; MS(ESIpos): m/z = 601.02 (M+H)+ H NMR (DMSO-de, 400 MHz): δ = 7.96-7.45 (m, 8H), 6.92 (d, 1H), 5.18 (s, 2H), 4.38-4.22 (m, 1H), 4.08-3.81 (m, 2H), 3.76 (s, 3H).
Example 30A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 - [2-(mefhylsulf anyl)phenyl] - 1 H- 1 ,2,4-triazole-5 -carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 300 mg, 0.79 mmol) in 6 ml anhydrous THF was treated at 0°C with 151 μΐ (0.44 mmol) N V- diisopropylethylamine and 80 μΐ (0.87 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 134 mg (0.87 mmol) of [2-(methylsulfanyl)phenyl]hydrazine was added, followed by 289 μΐ (1.66 mmol) of A^ -diisopropylefhylamine. The resulting mixture was stirred 1.5 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 325 mg (68 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.97 min; MS(ESIpos): m/z = 569.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.88-7.25 (m, 8H), 6.92 (d, 1H), 5.16 (s, 2H), 4.41-4.15 (m, 1H), 4.09-3.79 (m, 2H), 3.74 (s, 3H), 2.37 (s, 3H).
Example 31 A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(2-methylphenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 53 mg (0.44 mmol) of (2-methylphenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of N-diisopropylefhylamine. The resulting mixture was stirred 2 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 84 mg (39 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.95 min; MS(ESIpos): m/z = 537.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.88-7.22 (m, 8H), 6.91 (d, 1H), 5.17 (s, 2H), 4.29 (br s, 1H), 4.07-3.79 (m, 2H), 3.74 (s, 3H), 1.95 (s, 3H).
Example 32A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(2-ethylphenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 75 mg (0.44 mmol) of (2-ethylphenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of /V,/V-diisopropylefhylamine. The resulting mixture was stirred 2 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 135 mg (57 % of th.) of the title compound.
LC-MS (Method 2): Rt = 2.04 min; MS(ESIpos): m/z = 551.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.85-7.25 (m, 8H), 6.91 (d, 1H), 5.17 (s, 2H), 4.39 - 4.20 (br m, 1H), 4.10-3.78 (m, 2H), 3.74 (s, 3H), 2.24 (q, 2H), 0.95 (t, 3H).
Example 33A Methyl l-(2-chloro-6-fluorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 69 mg (0.44 mmol) of (2-chloro-6-fluorophenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of A^ -diisopropylefhylamine. The resulting mixture was stirred 2 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 48 mg (21 % of th.) of the title compound.
LC-MS (Method 2): Rt = 2.00 min; MS(ESIpos): m/z = 575.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.98-7.48 (m, 7H), 6.91 (d, 1H), 5.22 (s, 2H), 4.48-3.67 (m, 6H).
Example 34A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(2,6-dichlorophenyl)- 1H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 93 mg (0.44 mmol) of (2,6-dichlorophenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of N-diisopropylemylamine. The resulting mixture was stirred 2 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 43 mg (18 % of th.) of the title compound.
LC-MS (Method 1): Rt = 1.12 min; MS(ESIpos): m/z = 591.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.80-7.59 (m, 7H), 6.91 (d, 1H), 5.23 (d, 2H), 4.38-4.22 (m, 1H), 4.11-3.75 (m, 5H).
Example 35A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(2,6-dimethylphenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 75 mg (0.44 mmol) of (2,6-dimethylphenyl)hydrazine hydrochloride was added, followed by 145 μΐ (0.83 mmol) of N-diisopropylethylamine. The resulting mixture was stirred 2 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 85 mg (37 % of th.) of the title compound.
LC-MS (Method 2): Rt = 2.05 min; MS(ESIpos): m/z = 551.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.80-7.58 (m, 4H), 7.44-7.18 (m, 3H), 6.91 (d, 1H), 5.19 (d, 2H), 4.38-4.21 (m, 1H), 4.10-3.79 (m, 2H), 3.76 (s, 3H); 1.86 (s, 6H).
Example 36A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(2-nitrophenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 66 mg (0.44 mmol) of 2-nitrophenylhydrazine was added, followed by 145 μΐ (0.83 mmol) of N-diisopropylethylamine. The resulting mixture was stirred 1 h at room temperature. 30 mg (0.20 mmol) of (2-nitrophenyl)hydrazine was added, followed by 34 μΐ (0.20 mmol) of N V- diisopropylethylamine were added and stirred overnight at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 40 mg with a (purity 63 %, 11 % of th.) of the title compound.
LC-MS (Method 3): Rt = 3.15 min; MS(ESIpos): m/z = 568.2 (M+H)+
Example 37A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(2,6-difluorophenyl)- IH- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 78 mg (0.44 mmol) of (2,6-difluorophenyl)hydrazine hydrochloride was added, followed by 145 μΐ (0.83 mmol) of A^ -diisopropylefhylamine. The resulting mixture was stirred 1.5 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 70 mg (31 % of th.) of the title compound. LC-MS (Method 2): Rt = 1.95 min; MS(ESIpos): m/z = 559.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.86-7.35 (m, 7H), 6.91 (d, 1H), 5.21 (s, 2H), 4.37-4.22 (m, 1H), 4.06-3.96 (m, 1H), 3.91-3.78 (m, 4H).
Example 38A
Methyl l-(2-chloro-4-fluorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min.85 mg (0.44 mmol) of (2-chloro-4-fluorophenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of N-diisopropylefhylamine. The resulting mixture was stirred overnight at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 109 mg (47 % of th.) of the title compound.
LC-MS (Method 2): Rt = 2.00 min; MS(ESIpos): m/z = 575.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.94-7.32 (m, 7H), 6.91 (d, 1H), 5.18 (s, 2H), 4.50-3.64 (m, 6H).
Example 39A
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 - [2-(difluoromethoxy)phenyl] - 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 91 mg (0.44 mmol) of [2-(difluoromethoxy)phenyl]hydrazine was added, followed by 145 μΐ (0.83 mmol) of A^ -diisopropylefhylamine. The resulting mixture was stirred 2 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 115 mg (47 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.93 min; MS(ESIpos): m/z = 589.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.84-6.83 (m, 10H), 5.17 (s, 2H), 4.41-4.21 (m, 1H), 3.77 (s, 5H).
Example 40A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl }methyl)- l-(2-isopropylphenyl)-lH- l,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 81 mg (0.44 mmol) of (2-isopropylphenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of N-diisopropylefhylamine. The resulting mixture was stirred 2 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 127 mg (56 % of th.) of the title compound.
LC-MS (Method 2): Rt = 2.15 min; MS(ESIpos): m/z = 565.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.82-7.20 (m, 8H), 6.97-6.82 (m, IH), 5.17 (s, 2H), 4.39 - 4.20 (br m, IH), 4.06-3.80 (m, 2H), 3.73 (s, 3H), 2.33 (d, IH), 1.03 (d, 6H).
Example 41 A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(2-fluorophenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl} ethanimidate (Example 4A, 150 mg, 0.40 mmol) of Example 3A in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N V- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 70 mg (0.44 mmol) of (2-fluorophenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of /V,/V-diisopropylefhylamine. The resulting mixture was stirred 1.5 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 126 mg (52 % of th.) of the title compound.
LC-MS (Method 1): Rt = 1.02 min; MS(ESIpos): m/z = 541.2 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.86-7.30 (m, 8H), 6.92 (d, IH), 5.18 (s, 2H), 4.39 - 4.21 (br m, IH), 4.11-3.73 (m, 5H).
Example 42A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -phenyl- IH- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 300 mg, 0.79 mmol) in 6 ml anhydrous THF was treated at 0°C with 150 μΐ (0.74 mmol) N,N- diisopropylethylamine and 80 μΐ (0.87 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 94 mg (0.88 mmol) of phenylhydrazine was added. The resulting mixture was stirred 10 min at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 347 mg (83 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.31 min; MS(ESIpos): m/z = 523.0 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.91-7.34 (m, 9H), 6.91 (d, IH), 5.15 (s, 2H), 4.58-3.62 (m, 6H).
Example 43A
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl Jmethyl)- l-(2-ethyl-6-methylphenyl)- IH- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl]-4,5-dihydro-lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 150 mg, 0.40 mmol) in 3 ml anhydrous THF was treated at 0°C with 75 μΐ (0.44 mmol) N,N- diisopropylethylamine and 40 μΐ (0.44 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 81 mg (0.44 mmol) of (2-ethyl-6-methylphenyl)hydrazine was added, followed by 145 μΐ (0.83 mmol) of A^ -diisopropylefhylamine. The resulting mixture was stirred 1.5 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 79 mg (35 % of th.) of the title compound. LC-MS (Method 1): Rt = 1.12 min; MS(ESIpos): m/z = 565.3 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 7.84-7.18 (m, 7H), 6.91 (d, 1H), 5.19 (dd, 2H), 4.39-4.19 (m, 1H), 4.07-3.80 (m, 2H), 3.75 (s, 3H), 2.24-2.01 (m, 2H), 1.85 (s, 3H), 0.94 (t, 3H).
Example 44A
Methyl {3-(4-chlorophenyl)-5-oxo-4-[(2,S,)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-m- 1,2,4- triazol- 1 -yl } acetate
Under argon, potassium ieri-butoxide (9.1 g, 81.3 mmol) was added portionwise at room temperature to a solution of 5-(4-chlorophenyl)-4-[(2lS)-3,3,3-trifluoro-2-hydroxypropyl]-2,4- dihydro-3/M,2,4-triazol-3-one (Example 5A in WO 2011/104322-A1; 20 g, 65.01 mmol) in THF (40 ml). To this solution was added methyl bromoacetate (10.94 g, 71.51 mmol), and the mixture was stirred at room temperature overnight. The reaction mixture was then diluted with water and extracted with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated in vacuo affording 16.4 g (46 % of th.) of the title compound.
LC/MS (Method 1): Rt = 0.90 min; MS [ESIpos] : m/z = 380 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 3.70 (s, 3H), 3.85 (dd, 1H), 4.00 (dd, 1H), 4.19-4.33 (m, 1H), 4.72 (s, 2H), 6.92 (d, 1H), 7.60-7.69 (m, 2H), 7.73-7.81 (m, 2H).
The title compound can also be synthesized via the procedure described in WO 2011/104322-A1 (Example 7A).
Example 45A
2-{ 3-(4-Chlorophenyl)-5-oxo-4-[(2,S,)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-m-l,2,4- triazol- 1 -yl } acetohydrazide
Methyl {3-(4-chlorophenyl)-5-oxo-4-[(2,S,)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-m- l,2,4- triazol- l-yl} acetate (Example 44A, 7.2 g, 19.0 mmol) ws dissolved in 60 ml of absolute ethanol. To this solution was added 2.1 g (41.7 mmol) of hydrazine hydrate, and the mixture was stirred under reflux for 5 h and then at room temperature overnight. The resulting mixture was partially concentrated in vacuo and then diluted with water and extracted with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was dissolved in dichloromethane, and after crystallization the white solid was filtered off and dried under high vacuum affording 7.0 g (97% of th.) of the title compound.
LC/MS (Method 1): Rt = 0.73 min; MS [ESIpos] : m/z = 380 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 3.82 (dd, 1H), 3.96 (dd, 1H), 4.24-4.34 (m, 3H), 4.38 (d, 2H), 6.90 (d, 1H), 7.61-7.66 (m, 2H), 7.73-7.78 (m, 2H), 9.23 (t, 1H).
Example 46A
Ethyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2lS')-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
2-{ 3-(4-Chlorophenyl)-5-oxo-4-[(2lS,)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro- lii-l,2,4- triazol- l-yl}acetohydrazide (Example 45 A, 6.0 g, 15.8 mmol) was dissolved in a mixture of toluene (64 ml) and ethyl acetate (12.8 ml) at room temperature. To this solution was added 2.84 g (21.3 mmol) of ethyl amino(thioxo)acetate, and the mixture was stirred at 100 °C overnight. After cooling, the reaction mixture was concentrated in vacuo and then diluted with ethyl acetate. The resulting mixture was washed with water, and after phase separation, the aqueous phase was extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by flash chromatography (silica gel, eluent cyclohexane/ethyl acetate) affording 2.6 g (33% of th.) of the desired compound.
LC-MS (Method 3): Rt = 1.14 min; MS (ESIpos): m/z = 461 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 1.30 (t, 3 H), 3.83 (dd, 1 H), 3.99 (dd, 1 H), 4.25 - 4.41 (m, 3 H), 5.01 - 5.29 (m, 2 H), 6.92 (d, 1 H), 7.60 - 7.66 (m, 2 H), 7.74 - 7.78 (m, 2 H), 15.02 (br. d, 1 H).
Example 47A
Ethyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(3-fluorophenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
To a solution of ethyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2lS')-3,3,3-trifluoro-2-hydroxypropyl]-4,5- dihydro-lH-l,2,4-triazol-l-yl}methyl)-lH-l,2,4-triazole-5-carboxylate (Exampe 46A, 2.1 g, 4.56 mmol) in pyridine (61 ml) was added (3-fluorophenyl)boronic acid (765 mg, 5.47 mmol) and copper(II) acetate (1.24 g, 6.84 mmol). The reaction mixture was stirred at 60 °C overnight, after which boronic acid (400 mg, 2.86 mmol) was added. After stirring for 6 additional days at room temperature, the reaction mixture was concentrated in vacuo and then diluted with ethyl acetate. The reaction mixture was then quenched with aqueous hydrochloric acid (0.5 M). After phase separation, the aqueous phase was extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated in vacuo. The crude product was first purified by flash chromatography (silica gel, eluent dichloromethane/methanol) then by preparative HPLC (Chromatorex CI 8, ΙΟμιη, 125x30mm, water-acetonitrile-gradient 0.05% trifluoroacetic acid) affording 242 mg (9 % of th.)of the desired compound.
LC-MS (Method 5): Rt = 3.46 min; MS (ESIpos): m/z = 555 (M+H)+ H NMR (DMSO-de, 400 MHz): δ = 1.16 (t, 3 H), 3.85 (dd, 1 H), 3.97 - 4.05 (m, 1 H), 4.24 (q, 2 H), 4.25 - 4.38 (m, 1 H), 5.15 (s, 2 H), 6.91 (d, 1 H), 7.38 -7.46 (m, 2 H), 7.54 - 7.65 (m, 4 H), 7.76 (d, 2 H).
Example 48A
Ethyl l-(3-chlorophenyl)-3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]- 4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
To a solution of ethyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2lS,)-3,3,3-trifluoro-2-hydroxypropyl]-4,5- dihydro-lH-l,2,4-triazol-l-yl}methyl)-lH-l,2,4-triazole-5-carboxylate (Example 46A, 2.55 g, 5.53 mmol) in pyridine (75 ml) was added (3-chlorophenyl)boronic acid (1.731 g, 11.07 mmol) and copper(II) acetate (1.508 g, 8.30 mmol). The reaction mixture was stirred at 60 °C overnight, after which boronic acid (1.30 g, 8.30 mmol) was added due to incomplete conversion. The reaction mixture was further stirred at 60 °C for 2 days. Over this time, additional portion of boronic acid (1.30 g, 8.30 mmol) was added. After cooling, the reaction mixture was concentrated in vacuo and then diluted with ethyl acetate. After this, the reaction mixture was then quenched with aqueous hydrochloric acid (0.5 M). After phase separation, the aqueous phase was extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by preparative HPLC (Chromatorex CI 8, ΙΟμιη, 125x30mm, water-acetonitrile-gradient 0.05% trifluoro acetic acid), and the desired compound (203.4 mg, 0.36 mmol) was obtained (6.4% of th.).
LC-MS (Method 3): Rt = 1.44 min; MS (ESIpos): m/z = 571 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 1.17 (t, 3 H), 3.85 (dd, 1 H), 4.01 (dd, 1 H), 4.24 (q, 2 H), 4.31 (br. s., 1 H), 5.15 (s, 2 H), 6.91 (br. d, 1 H), 7.54 - 7.58 (m, 2 H), 7.60 - 7.65 (m, 3 H), 7.73 - 7.80 (m, 3 H)
Example 49 A
Ethyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(2-methoxyphenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
To a solution of ethyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2lS)-3,33-trifluoro-2-hydroxypropyl]-4,5- dihydro-lH-l,2,4-triazol-l-yl}methyl)-lH-l,2,4-triazole-5-carboxylate (Example 46A, 450 mg, 0.98 mmol) in pyridine (10 ml) was added (2-methoxyphenyl)boronic acid (297 mg, 1.95 mmol) and copper(II) acetate (266 mg, 1.47 mmol). The reaction mixture was stirred at 60 °C overnight, after which boronic acid (148 mg, 0.98 mmol) was added. After stirring for two additional days, the reaction mixture was diluted with ethyl acetate and then quenched with aqueous hydrochloric acid (1 M). After phase separation, the aqueous phase was extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash chromatography (silica gel, eluent cyclohexane/ethyl acetate) followed by preparative SFC (column OX-H, 250 x 20 mm, 30% methanol) affording 72.2 mg (13 % of th.) of the title compound.
LC-MS (Method 1): Rt = 1.10 min; MS (ESIpos): m/z = 567 (M+H)+
¾ NMR (DMSO-de, 400 MHz): 5 = 1.11 (t, 3 H), 3.72 (s, 3 H), 3.85 (dd, 1 H), 4.02 (dd, 1 H), 4.21 (q, 2 H), 4.25 - 4.36 (m, 1 H), 5.14 (s, 2 H), 6.91 (d, 1 H), 7.11 (td, 1 H), 7.23 (dd, 1 H), 7.47 (dd, 1 H), 7.52 (td, 1 H), 7.60 - 7.65 (m, 2 H), 7.74 - 7.79 (m, 2 H).
Example 50A
Methyl l-(2-chlorophenyl)-3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxy- propyl] -4,5-dihydro- 1H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl}ethanimidate (Example 4A, 10 g, 26.40 mmol) of Example 3A in 200 ml anhydrous THF was treated at 0°C with 5.1 ml (29.1 mmol) N,N- diisopropylethylamine and 2.67 ml (29.0 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 5.2 g (29.1 mmol) of (2-chlorophenyl)hydrazine hydrochloride was added, followed by 5.1 ml (29.1 mmol) of A^/V-diisopropylefhylamine. The resulting mixture was stirred 1 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was retaken in ethyl acetate and washed with water. The aqueous phase was extracted with ethyl acetate. The combined organic layers were washed with a saturated sodium chloride solution, dried over magnesium sulfate and purified by flash chromatography (silica gel, cyclohexane/ethyl acetate eluent) affording 10.6 g (70 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.95 min; MS(ESIpos): m/z = 557.1 (M+H)+
Example 51 A
Methyl l-(2-chlorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2R)-3,3,3-trifluoro-2-hydroxy- propyl] -4,5-dihydro- IH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate
Under argon, a solution of methyl 2-{3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- lH-l,2,4-triazol-l-yl}ethanimidate (Example 5 A, 744 mg, 1.97 mmol) in 15 ml anhydrous THF was treated at 0°C with 0.75 ml (4.3 mmol) N V- diisopropylethylamine and 0.38 ml (1.1 mmol) methyl chlorooxoacetate and stirred at 0°C for 30 min. 387 mg (2.2 mmol) of (2-chlorophenyl)hydrazine hydrochloride was added, followed by 0.38 ml (1.1 mmol) of A^ -diisopropylefhylamine. The resulting mixture was stirred 1 h at room temperature, followed by lh at 120°C in the microwave and evaporated. The residue was retaken in ethyl acetate and washed with water. The aqueous phase was extracted with ethyl acetate. The combined organic layers were washed with a saturated sodium chloride solution, dried over magnesium sulfate and purified by flash chromatography (silica gel, cyclohexane/ethyl acetate eluent) affording 734 mg (60 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.36 min; MS(ESIpos): m/z = 557.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ [ppm] = 7.87-7.44 (m, 8H), 6.91 (d, IH), 5.18 (s, 2H), 4.43-3.66 (m, 6H). EXPERIMENTAL SECTION - EXAMPLES
Example 1
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(3-ethylpyridin-2-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(3-ethylpyridin-2-yl)- 1H- 1 ,2,4-triazole-5-carboxylate (Example 13A, 100 mg, 85 % purity, 154 μιηοΐ) in 7N NH3 solution in MeOH (10 ml, 70 mmol) was stirred 1 h at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 58.2 mg (70 % of th.) of the title compound.
LC-MS (Method 1): Rt = 0.88 min; MS (ESIpos): m/z = 537 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.50-8.19 (m, 2H), 8.08-7.43 (m, 7H), 6.91 (d, 1H), 5.16 (m, 2H), 4.44-4.37 (br. m, 1H), 4.44-3.79 (m, 2H), 2.34 (q, 2H), 1.01 (t, 3H).
Example 2
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(3-methylpyridin-2-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-methylpyridin-2-yl)-lH-l,2,4-triazole-5-carboxylate (Example 14A, 150 mg, 279 μιηοΐ) in 7N NH3 solution in MeOH (15 ml, 105 mmol) was stirred 1 h at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 123 mg (84 % of th.) of the title compound. LC-MS (Method 1): Rt = 0.84 min; MS (ESIpos): m/z = 523 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] 8.46-8.20 (m, 2H), 8.04-7.41 (m, 7H), 6.91 (d, IH), 5.16 (m, 2H), 4.47-3.70 (m, 3H), 2.04 (s, 3H).
Example 3
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(3-methylpyridin-4-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-methylpyridin-4-yl)-lH-l,2,4-triazole-5-carboxylate (Example 15A, 28.0 mg, 80 % purity, 41.6 μιηοΐ) in 7N NH3 solution in MeOH (2.5 ml, 17 mmol) was stirred 1 h at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 14.8 mg (62 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.47 min; MS (ESIpos): m/z = 523 [M+H]+
Ή-NMR (500 MHz, DMSO-d6) δ [ppm] :8.70-8.51 (m, 2H), 8.33 (br. s, IH), 7.99 (br. s, IH), 7.84-7.59 (m, 4H), 7.42 (d, IH), 6.91 (br d, IH), 5.15 (m, 2H), 4.29 (br. m, IH), 4.10-3.77 (m, 2H), 2.00 (s, 3H).
Example 4
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(3-methoxypyridin-2-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-methoxypyridiri-2-yl)-lH-l,2,4-triazole-5-carboxylate (Example 16 A, 200 mg, 361 μιηοΐ) in 7N NH3 solution in MeOH (20 ml, 140 mmol) was stirred 1 h at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 144 mg (73 % of th.) of the title compound.
LC-MS (Method 1): Rt = 0.86 min; MS (ESIpos): m/z = 539 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.22 (br. s, 1H), 8.10 (dd, 1H), 7.86 (br. s, 1H), 7.80- 7.55 (m, 6H), 6.91 (d, 1H), 5.14 (s, 2H), 4.41-3.70 (m, 6H).
Example 5
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(3-fluoropyridin-2-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-fluoropyridin-2-yl)-lH-l,2,4-triazole-5-carboxylate (Example 17A, 40.0 mg, 73.8 μιηοΐ) in 7N NH3 solution in MeOH (500 μΐ, 3.5 mmol) was stirred 1 h at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 38.7 mg (96 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.12 min; MS (ESIpos): m/z = 527 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.43 (d, 2H), 8.05 (m, 2H), 7.83-7.54 (m, 5H), 6.90 (d, 1H), 5.18 (s, 2H), 4.40-4.19 (br. m, 1H), 4.09-3.76 (m, 2H).
Example 6
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1 -yl } methyl)- 1 -(3-chloropyridin-4-yl)- 1 H- 1 ,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-chloropyridiri-4-yl)-lH-l,2,4-triazole-5-carboxylate (Example 18A, 30.0 mg, 53.7 μιηοΐ) in 7N NH3 solution in MeOH (954 mg, 56.0 mmol) was stirred 45 min at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 24.0 mg (82 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.14 min; MS (ESIpos): m/z = 543 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.89 (s, 1H), 8.73 (d, 1H), 8.39 (br. s, 1H), 8.04 (br. s, 1H), 7.82-7.56 (m, 5H), 6.90 (d, 1H), 5.17 (m, 2H), 4.40-4.20 (br. m, 1H), 4.13-3.75 (m, 2H).
Example 7
Methyl 2-[5-carbamoyl-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]- 4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazol- 1 -yl]pyridine-3-carboxylate
Ethyl 2-[3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl }methyl)-5-(methoxycarbonyl)- 1 H- 1 ,2,4-triazol- 1 -yl]pyridine-3-carboxylate
(Example 19A, 50.0 mg, 83.9 μιηοΐ) in 7N NH3 solution in MeOH (1.3 ml, 9.1 mmol) was stirred 1 h at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 27.8 mg (57 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.12 min; MS (ESIpos): m/z = 567 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 8.77 (dd, 1H), 8.58-8.21 (m, 2H), 8.03-7.54 (m, 6H), 6.91 (d, 1H), 5.21 - 5.04 (m, 2H), 4.50-3.50 (s, 6H). Example 8
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,33-trifluoro-2-hydroxypropyl]-4,5-dihydro
triazol-l-yl}methyl)-l-(3-fluoropyridin-4-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(3-fluoropyridin-4-yl)-lH-l,2,4-triazole-5-carboxylate (Example 20A, 100 mg, 185 μιηοΐ) in 7N N¾ solution in MeOH (10 ml, 70 mmol) was stirred 1 h at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 66.6 mg (66 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.52 min; MS (ESIpos): m/z = 527 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.84 (d, 1H), 8.63 (d, 1H), 8.45 (br. s, 1H), 8.10 (br. s, 1H), 7.86-7.55 (m, 5H), 6.90 (d, 1H), 5.18 (m, 2H), 4.47-3.72 (m, 3H).
Example 9
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(3-methoxypyrazin-2-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(3-methoxypyrazin-2-yl)- 1 H- 1 ,2,4-triazole-5-carboxylate (Example 21A, 49.0 mg, 88.3 μιηοΐ) in 7N NH3 solution in MeOH (4.0 ml, 28 mmol) was stirred 45 min at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 24.8 mg (52 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.57 min; MS (ESIpos): m/z = 540 [M+H]+ Ή-NMR (500 MHz, DMSO-d6) δ [ppm]: 8.46 (d, 1H), 8.39 (br. s, 1H), 8.24 (d, 1H), 8.01 (br. s, 1H), 7.83-7.58 (m, 4H), 6.91 (d, 1H), 5.17 (s, 2H), 4.39-4.22 (br. m, 1H), 4.09-3.79 (m, 5H).
Example 10
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1 -yl } methyl)- 1 -(2-chloropyridin-3-yl)- 1 H- 1 ,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(2-chloropyridin-3-yl)-lH-l,2,4-triazole-5-carboxylate (Example 22A, 116 mg, 208 μιηοΐ) in 7N N¾ solution in MeOH (8.0 ml, 56 mmol) was stirred 45 min at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 100 mg (89 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.17 min; MS (ESIpos): m/z = 543 [M+H]+
Ή-NMR (500 MHz, DMSO-d6) δ [ppm] : 8.58 (dd, 1H), 8.34 (br. s, 1H), 8.15 (dd, 1H), 8.00 (br. s, 1H), 7.83-7.56 (m, 5H), 6.92 (br d, 1H), 5.23-5.09 (m, 2H), 4.34-4.20 (br. m, 1H), 4.07-3.80 (m, 2H).
Example 11
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1 -yl } methyl) - 1 - (pyridin-2-y 1)- 1 H- 1 ,2,4-triazole-5 -carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(pyridin-2-yl)-lH-l,2,4-triazole-5-carboxylate (Example 23A, 40.0 mg, 76.4 μιηοΐ) in 7N NH3 solution in MeOH (500 μΐ, 3.5 mmol) was stirred 1 h at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 29.3 mg (75 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.09 min; MS (ESIpos): m/z = 509 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.51 (d, 1H), 8.32 (br. s, 1H), 8.06 (td, 1H), 7.95 (br. s, 1H), 7.83-7.47 (m, 6H), 6.92 (d, 1H), 5.14 (s, 2H), 4.41-4.21 (br. m, 1H), 4.12-3.77 (m, 2H).
Example 12
l-(2-Chloro-4-methylpyridin-3-yl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxamide
Methyl l-(2-chloro-4-methylpyridin-3-yl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate (Example 24A, 50.0 mg, 87.4 μιηοΐ) in 7N NH3 solution in MeOH (6.0 ml, 42 mmol) was stirred 1 h at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 33.2 mg (67 % of th.) of the title compound.
LC-MS (Method 1): Rt = 0.88 min; MS (ESIpos): m/z = 557 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 8.47-8.34 (m, 2H), 8.01 (br. s, 1H), 7.80-7.47 (m, 5H), 6.91 (br d, 1H), 5.30-5.07 (m, 2H), 4.38 - 4.18 (br m, 1H), 4.10-3.79 (m, 2H), 2.03 (s, 3H).
Example 13
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(2-ethoxypyridin-3-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(2-ethoxypyridin-3-yl)- 1H- 1 ,2,4-triazole-5-carboxylate (Example 25A, 75.0 mg, 132 μιηοΐ) in 7N NH3 solution in MeOH (8.0 ml, 56 mmol) was stirred 1 h at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 61.0 mg (84 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.67 min; MS (ESIpos): m/z = 553 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.31-8.16 (m, 2H), 8.00-7.82 (m, 2H), 7.81-7.56 (m, 4H), 7.13 (dd, 1H), 6.92 (d, 1H), 5.13 (m, 2H), 4.29 (m, 3H), 4.09-3.80 (m, 2H), 1.18 (t, 3H).
Example 14
3-[5-Carbamoyl-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5- dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1H- 1 ,2,4-triazol- 1 -yl]pyridine-4-carboxamide
Ethyl 3-[3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl }methyl)-5-(methoxycarbonyl)- 1 H- 1 ,2,4-triazol- 1 -yl]pyridine-4-carboxylate
(Example 26A, 100 mg, 90 % purity, 151 μιηοΐ) in 7N NH3 solution in MeOH (9.0 ml, 63 mmol) was stirred overnight at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 44mg (52% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.28 min; MS (ESIpos): m/z = 552 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm]: 8.83-8.64 (m, 2H), 8.28-7.52 (m, 9H), 6.93 (d, 1H), 5.20-5.02 (m, 2H), 4.46-4.21 (br. m, 1H), 4.08-3.76 (m, 2H) Example 15
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,33-trifluoro-2-hydroxypropyl]-4,5-dihydro
triazol- 1 -yl } methyl)- 1 -(4-chloropyridin-2-yl)- 1 H- 1 ,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(4-chloropyridin-2-yl)-lH-l,2,4-triazole-5-carboxylate (Example 27 A, 182 mg, 326 μιηοΐ) in 7N NH3 solution in MeOH (18 ml, 126 mmol) was stirred 30 min at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 136 mg (77 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.65 min; MS (ESIpos): m/z = 543 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 8.58 (d, 1H), 8.42 (br. s, 1H), 8.16 (br. s, 1H), 7.89-7.57 (m, 6H), 6.90 (d, 1H), 5.15 (s, 2H), 4.36-4.22 (br. m, 1H), 4.10-3.77 (m, 2H).
Example 16
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- l-yl}methyl)-l-(pyrimidin-2-yl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(pyrimidin-2-yl)-lH-l,2,4-triazole-5-carboxylate (Example 28A, 75.0 mg, 143 μιηοΐ) in 7N NH3 solution in MeOH (12 ml, 84 mmol) was stirred 1 h at room temperature and evaporated. The residue was evaporated and purified by preparative HPLC (Method 6) affording 58.0 mg (80 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.40 min; MS (ESIpos): m/z = 510 [M+H]+ Ή-NMR (400 MHz, DMSO-d6) δ [ppm] : 8.97 (d, 2H), 8.42 (br. s, 1H), 8.00 (br. s, 1H), 7.85-7.55 (m, 5H), 6.92 (d, 1H), 5.16 (s, 2H), 4.42-4.23 (br. m, 1H), 4.13-3.78 (m, 2H).
Example 17
l-(2-Bromophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5- dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1H- 1 ,2,4-triazole-5-carboxamide
Methyl l-(2-bromophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxy- propyl] -4,5-dihydro- 1H- 1 ,2,4-triazol- 1 -yl } methyl)- 1H- 1 ,2,4-triazole-5-carboxylate (Example 29A, 121 mg, 0.20 mmol) in 7N N¾ solution in MeOH (2 ml, 14 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 85 mg (72% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.73 min; MS(ESIpos): m/z = 586.0 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.23 (s, 1H), 7.89 (s, 1H), 7.83-7.39 (m, 8H), 6.90 (d, 1H), 5.24-5.05 (m, 2H), 4.30 (br d, 1H), 4.09-3.76 (m, 2H).
Example 18
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1 -yl } methyl)- 1 - [2-(methylsulfanyl)phenyl] - 1 H- 1 ,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 - [2-(methylsulf anyl)phenyl] - 1 H- 1 ,2,4-triazole-5 -carboxylate (Example 30A, 142 mg, 0.25 mmol) in 7N N¾ solution in MeOH (2 ml, 14 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 107 mg (74% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.72 min; MS(ESIpos): m/z = 554.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.15 (s, 1H), 7.88-7.16 (m, 9H), 6.90 (d, 1H), 5.13 (d, 2H), 4.40-4.19 (m, 1H), 4.08-3.75 (m, 2H), 2.35 (s, 3H).
Example 19
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(2-methylphenyl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(2-methylphenyl)-lH-l,2,4-triazole-5-carboxylate (Example 31A, 137 mg, 0.25 mmol) in 7N Ν¾ solution in MeOH (2.2 ml, 15.4 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 64 mg (47% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.74 min; MS(ESIpos): m/z = 522.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.21 (s, 1H), 7.87 (s, 1H), 7.80-7.54 (m, 4H), 7.50-7.21 (m, 4H), 6.90 (d, 1H), 5.13 (d, 2H), 4.40-4.18 (m, 1H), 4.08-3.76 (m, 2H), 1.95 (s, 3H).
Example 20
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1 -yl } methyl) - 1 - (2-ethylphenyl) - 1 H- 1 ,2,4-triazole-5 -carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(2-ethylphenyl)-lH-l,2,4-triazole-5-carboxylate (Example 32A, 130 mg, 0.24 mmol) in 7N N¾ solution in MeOH (2 ml, 14 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 101 mg (80% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.83 min; MS(ESIpos): m/z = 536.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.20 (s, 1H), 7.86 (s, 1H), 7.77-7.58 (m, 4H), 7.50-7.22 (m, 4H), 6.89 (d, 1H), 5.13 (d, 2H), 4.43-4.19 (m, 1H), 4.09-3.73 (m, 2H), 2.24 (q, 2H), 0.95 (t, 3H).
Example 21
l-(2-Chloro-6-fluorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxamide
Methyl l-(2-chloro-6-fluorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate (Example 33A, 48 mg, 85 μιηοΐ) in 7N NH3 solution in MeOH (1.4 ml, 9.8 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 36 mg (76% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.76 min; MS(ESIpos): m/z = 560.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.39 (s, 1H), 8.02 (s, 1H), 7.84-7.41 (m, 7H), 6.90 (d, 1H), 5.31-5.04 (m, 2H), 4.39 - 4.20 (br m, 1H), 4.09-3.74 (m, 2H).
Example 22
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1-yl } methyl)- l-(2,6-dichlorophenyl)- 1H- 1 ,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(2,6-dichlorophenyl)-lH-l,2,4-triazole-5-carboxylate (Example 34 A, 35 mg, 0.20 mmol) in 7N N¾ solution in MeOH (2.5 ml, 17.5 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 29 mg (34% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.82 min; MS(ESIpos): m/z = 576.0 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.35 (s, 1H), 7.98 (s, 1H), 7.86-7.52 (m, 7H), 6.91 (d, 1H), 5.35-5.00 (m, 2H), 4.44-4.16 (m, 1H), 4.07-3.69 (m, 2H).
Example 23
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(2,6-dimethylphenyl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(2,6-dimethylphenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate (Example 35 A, 85 mg, 0.16 mmol) in 7N N¾ solution in MeOH (1.3 ml, 9.1 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 59 mg (71 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.79 min; MS(ESIpos): m/z = 536.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.23 (s, 1H), 7.86 (s, 1H), 7.78-7.56 (m, 4H), 7.35-6.84 (m, 4H), 5.24-5.08 (m, 2H), 4.36-4.22 (m, 1H), 4.07-3.78 (m, 2H), 1.86 (s, 6H).
Example 24 3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(2-nitrophenyl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(2-nitrophenyl)-lH-l,2,4-triazole-5-carboxylate (Example 36A, 40 mg, 0.07 mmol) in 7N N¾ solution in MeOH (2 ml, 14 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 15 mg (40% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.68 min; MS(ESIpos): m/z = 553.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.41-8.16 (m, 2H), 8.05-7.55 (m, 8H), 6.91 (d, 1H), 5.31-5.01 (m, 2H), 4.39 -4.20 (br m, 1H), 4.09-3.77 (m, 2H).
Example 25
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(2,6-difluorophenyl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(2,6-difluorophenyl)- 1H- 1 ,2,4-triazole-5-carboxylate (Example 37A, 70 mg, 0.13 mmol) in 7N N¾ solution in MeOH (1.6 ml, 11.2 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 47 mg (69% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.72 min; MS(ESIpos): m/z = 544.1 (M+H)+ H NMR (DMSO-de, 400 MHz): δ = 8.41 (s, 1H), 8.05 (s, 1H), 7.85-7.29 (m, 7H), 6.89 (d, 1H), 5.18 (d, 2H), 4.39 - 4.20 (br m, 1H), 4.11-3.76 (m, 2H).
Example 26
l-(2-Chloro-4-fluorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxamide
Methyl l-(2-chloro-4-fluorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2- hydroxypropyl] -4,5-dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxylate (Example 38A, 70 mg, 0.120 mmol) in 7N NH3 solution in MeOH (10 ml, 70 mmol) was stirred 1 h at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 50 mg (73% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.78 min; MS(ESIpos): m/z = 560.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.36-7.25 (m, 9H), 6.91 (d, 1H), 5.15 (d, 2H), 4.39 - 4.20 (br m, 1H), 4.10-3.74 (m, 2H).
Example 27
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1 -yl } methyl)- 1 - [2-(difluoromethoxy)phenyl] - 1 H- 1 ,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 - [2-(difluoromethoxy)phenyl] - 1 H- 1 ,2,4-triazole-5-carboxylate (Example 39A, 137 mg, 0.23 mmol) in 7N N¾ solution in MeOH (2 ml, 14 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 80 mg (60% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.77 min; MS(ESIpos): m/z = 574.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.22 (s, 1H), 7.90 (s, 1H), 7.82-6.84 (m, 10H), 5.14 (d, 2H), 4.39 - 4.20 (br m, 1H), 4.11-3.75 (m, 2H).
Example 28
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(2-isopropylphenyl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(2-isopropylphenyl)- 1H- 1 ,2,4-triazole-5-carboxylate (Example 40 A, 127 mg, 0.22 mmol) in 7N Ν¾ solution in MeOH (2 ml, 14 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 100 mg (81 % of th.) of the title compound.
LC-MS (Method 2): Rt = 1.89 min; MS(ESIpos): m/z = 550.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.20 (s, 1H), 7.94-7.15 (m, 9H), 6.89 (d, 1H), 5.14 (d, 2H), 4.39 - 4.20 (br m, 1H), 4.10-3.71 (m, 2H), 2.44-2.25 (m, 1H), 1.03 (d, 6H).
Example 29
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1-yl Jmethyl)- l-(2-fluorophenyl)- 1H- 1 ,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-(2-fluorophenyl)-lH-l,2,4-triazole-5-carboxylate (Example 41A, 120 mg, 0.22 mmol) in 7N N¾ solution in MeOH (3 ml, 21 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 96 mg (82% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.68 min; MS(ESIpos): m/z = 526.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.29 (s, 1H), 7.96 (s, 1H), 7.83-7.29 (m, 8H), 6.90 (d, 1H), 5.15 (d, 2H), 4.39 - 4.20 (br m, 1H), 4.13-3.73 (m, 2H).
Example 30
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1 -yl } methyl)- 1 -phenyl- 1H- 1 ,2,4-triazole-5-carboxamide
Methyl 3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl}methyl)-l-phenyl-lH-l,2,4-triazole-5-carboxylate (Example 42A, 38 mg, 0.07 mmol) in 7N N¾ solution in MeOH (0.5 ml, 3.0 mmol) was stirred 45 min at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 31 mg (77% of th.) of the title compound.
LC-MS (Method 3): Rt = 1.18 min; MS(ESIpos): m/z = 508.0 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.38-7.38 (m, 11H), 6.90 (d, 1H), 5.13 (s, 2H), 4.41-3.74 (m, 3H).
Example 31
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol-l-yl}methyl)-l-(2-ethyl-6-methylphenyl)-lH-l,2,4-triazole-5-carboxamide
Methyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- l-(2-ethyl-6-methylpheriyl)- 1H- 1 ,2,4-triazole-5-carboxylate (Example 43A, 70 mg, 0.12 mmol) in 7N N¾ solution in MeOH (3 ml, 21 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 59 mg (87% of th.) of the title compound.
LC-MS (Method 2): Rt = 1.88 min; MS(ESIpos): m/z = 550.1 (M+H)+
¾ NMR (DMSO-de, 400 MHz): δ = 8.23 (s, 1H), 7.85 (s, 1H), 7.79-7.57 (m, 4H), 7.44-6.79 (m, 4H), 5.30-4.99 (m, 2H), 4.38 - 4.18 (br m, 1H), 4.09-3.74 (m, 2H), 2.25-2.01 (m, 2H), 1.85 (s, 3H), 0.94 (td, 3H).
Example 32
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- 1-yl } methyl)- 1- (3 -fluorophenyl)- 1H- 1 ,2,4-triazole-5-carboxamide
Ethyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2lS')-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- l,2,4-triazol-l-yl }methyl)- l-(3-fluorophenyl)-lH-l,2,4-triazole-5-carboxylate (Example 47A, 15 mg, 0.03 mmol) was dissolved in a 2 M solution of ammonia in ethanol (0.5 ml, 1.0 mmol). The reaction mixture was stirred at room temperature overnight and then concentrated in vacuo. The crude product was purified by preparative HPLC (Method 6) affording 8 mg (56% of th.) of the title compound.
LC-MS (Method 5): Rt = 2.86 min; MS (ESIpos): m/z = 526 (M+H)+ H NMR (500 MHz, DMSO-d6) δ ppm 3.85 (dd, 1 H), 4.00 (dd, 1 H), 4.25 - 4.35 (m, 1 H), 5.09 - 5.17 (m, 2 H), 6.90 (d, 1 H), 7.34 - 7.40 (m, 2 H), 7.46 (dt, 1 H), 7.56 (td, 1 H), 7.60 - 7.65 (m, 2 H), 7.73 - 7.79 (m, 2 H), 8.02 (br. s, 1 H), 8.30 (br. s, 1 H).
Example 33
l-(3-Chlorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5- dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1H- 1 ,2,4-triazole-5-carboxamide
Ethyl l-(3-chlorophenyl)-3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2lS')-3,3,3-trifluoro-2-hydroxypropyl]- 4,5-dihydro-lH-l,2,4-triazol-l-yl}methyl)-lH-l,2,4-triazole-5-carboxylate (Example 48A, 37 mg (0.06 mmol) was dissolved in a 2 M solution of ammonia in ethanol (0.6 ml, 1.2 mmol). After 2 h of stirring at room temperature, the reaction mixture was concentrated in vacuo. The crude product was purified by preparative HPLC (Method 6) affording 23.2 mg (67% of th.) of the title compound.
LC-MS (Method 5): Rt = 3.04 min; MS (ESIpos): m/z = 542 (M+H)+
¾ NMR (DMSO-A, 500 MHz): δ [ppm] 3.85 (dd, 1 H), 4.00 (dd, 1 H), 4.20 - 4.39 (br.m, 1 H), 5.09 - 5.17 (m, 2 H), 6.86 - 6.93 (m, 1 H), 7.47 - 7.51 (m, 1 H), 7.54 (t, 1 H), 7.56 - 7.60 (m, 1 H), 7.60 - 7.66 (m, 3 H), 7.73 - 7.78 (m, 2 H), 8.02 (br. s, 1 H), 8.29 (br. s, 1 H).
Example 34
3-({3-(4-Chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH-l,2,4- triazol- l-yl}methyl)-l-(2-methoxyphenyl)-lH-l,2,4-triazole-5-carboxamide
Ethyl 3-({ 3-(4-chlorophenyl)-5-oxo-4-[(25)-333-trifluoro-2-hydroxypropyl]-4,5-dihydro-lH- 1 ,2,4-triazol- 1 -yl } methyl)- 1 -(2-methoxyphenyl)- 1 H- 1 ,2,4-triazole-5-carboxylate (Example 49A, 35 mg, 0.06 mmol) was dissolved in a 2 M solution of ammonia in ethanol (0.6 ml, 1.2 mmol). The reaction mixture was stirred at room temperature followed by 3 h at 100 °C under microwave irradiation. After cooling, the reaction mixture was concentrated in vacuo. The crude product was purified by preparative HPLC (Method 6) affording 19.2 mg (57 % of th.) of the title compound.
LC-MS (Method 5): Rt = 2.74 min; MS (ESIpos): m/z = 538 (M+H)+
¾ NMR (500 MHz, DMSO-d6): δ [ppm] 3.71 (s, 3 H), 3.85 (dd, 1 H), 4.00 (dd, 1 H), 4.25 - 4.35 (m, 1 H), 5.06 - 5.15 (m, 2 H), 6.90 (br. d, 1 H), 7.06 (td, 1 H), 7.16 - 7.20 (m, 1 H), 7.37 (dd, 1 H), 7.46 (td, 1 H), 7.60 - 7.65 (m, 2 H), 7.73 - 7.81 (m, 3 H), 8.09 (br. s, 1 H).
Example 35
l-(2-Chlorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxypropyl]-4,5- dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1H- 1 ,2,4-triazole-5-carboxamide
Methyl l-(2-chlorophenyl)-3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxy- propyl] -4,5-dihydro- 1H- 1 ,2,4-triazol- 1 -yl } methyl)- 1H- 1 ,2,4-triazole-5-carboxylate (Example 50A, 100 mg, 0.17 mmol) in 2N N¾ solution in EtOH (1 ml, 2 mmol) was stirred overnight at 120°C in the microwave and evaporated. The residue was purified by preparative HPLC (Method 6) affording 30 mg (31 % of th.) of the title compound.
LC-MS (Method 3): Rt = 1.21 min; MS(ESIpos): m/z = 542.0 (M+H)
¾ NMR (DMSO-de, 400 MHz): δ [ppm] = 8.36-7.37 (m, 10H), 6.91 (d, 1H), 5.15 (d, 2H), 4.48- 4.15 (m, 1H), 4.09-3.74 (m, 2H).
Example 36
l-(2-Chlorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2R)-3,3,3-trifluoro-2-hydroxypropyl]-4,5- dihydro- 1 H- 1 ,2,4-triazol- 1 -yl } methyl)- 1H- 1 ,2,4-triazole-5-carboxamide
Methyl l-(2-chlorophenyl)-3-({3-(4-chlorophenyl)-5-oxo-4-[(2R)-3,3,3-trifluoro-2-hydroxy- propyl] -4,5-dihydro- 1H- 1 ,2,4-triazol- 1 -yl } methyl)- 1H- 1 ,2,4-triazole-5-carboxylate (Example 51 A, 367 mg, 0.66 mmol) in 7N N¾ solution in MeOH (1 ml, 7 mmol) was stirred overnight at room temperature and evaporated. The residue was purified by preparative HPLC (Method 6) affording 215 mg (60% of th.) of the title compound.
LC-MS (Method 1): Rt = 0.94 min; MS(ESIpos): m/z = 542.2 (M+H)
¾ NMR (DMSO-de, 400 MHz): δ [ppm] = 8.25 (s, 1H), 7.91 (s, 1H), 7.81-7.44 (m, 8H), 6.90 (d, 1H), 5.15 (d, 2H), 4.39 - 4.20 (br m, 1H), 4.08-3.75 (m, 2H).
Example 37
l-(2-Chlorophenyl)-3-{ [3-(4-chlorophenyl)-5-oxo-4-(3,3,3-trifluoro-2-oxopropyl)-4,5-dihydro-lH- l,2,4-triazol-l-yl]methyl }- lH-l,2,4-triazole-5-carboxamide (ketone form) or l-(2-Chlorophenyl)- 3-{ [3-(4-Chlorophenyl)-5-oxo-4-(3,3,3-trifluoro-2,2-dihydroxypropyl)-4,5-dihydro-lH-l,2,4- triazol- 1 -yl] methyl } - 1H- 1 ,2,4-triazole-5-carboxamide (hydrate form)
A solution of l-(2-Chlorophenyl)-3-({ 3-(4-chlorophenyl)-5-oxo-4-[(2S)-3,3,3-trifluoro-2-hydroxy- propyl] -4,5-dihydro- 1H- 1 ,2,4-triazol- 1 -yl } methyl)- 1 H- 1 ,2,4-triazole-5-carboxamide (Example 35, 500 mg, 0.92 mmol) in 10 ml of THF was treated with 782 mg (1.84 mmol) of Dess-Martin periodinane and stirred 2 h at room temperature. 10 ml of a saturated sodium thiosulfate solution and 10 ml of a saturated hydrogenocarbonate were added and the resulting mixture was stirred 5 min at room temperature. The aqueous layer was extracted with ethyl acetate (x 3). The combined organic layers were washed with a saturated NaCl solution, dried over magnesium sulfate and evaporated. Purification by flash chromatography (silica gel, eluent cyclohexane/ethyl acetate) followed by preparative HPLC (Method 6) afforded 6 mg (1% of th.) of the title compound
LC-MS (Method 4): Rt = 2.80 min; MS(ESIpos): m/z = 540.2 (M+H)
¾ NMR (DMSO-de, 400 MHz): δ [ppm] = 8.26 (s, 1H), 7.90 (s, 1H), 7.76-7.36 (m, 10H), 5.16 (s, 2H), 4.06 (s, 2H), as monohydrate.
EXPERIMENTAL SECTION - BIOLOGICAL ASSAYS
Abbreviations and Acronyms:
Acc. No. accession number
AVP arginine vasopressin
Bmax maximal ligand binding capacity
BSA bovine serum albumin
cAMP cyclic adenosine monophosphate
Cat. No. catalogue number
cDNA complementary deoxyribonucleic acid
CHO Chinese hamster ovary
CRE cAMP response element
Ct cycle threshold
DMEM/F12 Dulbecco's modified Eagle's medium / Ham's F12 medium (1 : 1)
DNA deoxyribonucleic acid
DMSO dimethylsulfoxide
DTT dithiothreitol
EC50 half-maximal effective concentration
EDTA ethylenediamine-tetraacetic acid
FAM carboxyfluorescein succinimidyl ester
f.c. final concentration
FCS fetal calf serum
HEPES 4-(2 -hydro xyethyl)piperazine- 1 -ethanesulfonic acid IC50 half-maximal inhibitory concentration
Kd dissociation constant
¾ dissociation constant of an inhibitor
mRNA messenger ribonucleic acid
PBS phosphate buffered saline
PEG polyethylene glycol
p.o. per os, peroral
RNA ribonucleic acid
RTPCR real-time polymerase chain reaction
SPA scintillation proximity assay
TAMRA carboxytetramethylrhodamine
TRIS; Tris 2-amino-2-hydroxymethylpropane-l,3-diol
Demonstration of the activity of the compounds of the present invention may be accomplished through in vitro, ex vivo, and in vivo assays that are well known in the art. For example, to demonstrate the activity of the compounds of the present invention, the following assays may be used. B-l. Cellular in vitro assay for determining vasopressin receptor activity
The identification of agonists and antagonists of the Via and V2 vasopressin receptors from humans, rats and dogs as well as the quantification of the activity of the compounds of the invention is carried out using recombinant cell lines. These cell lines originally derive from a hamster's ovary epithelial cell (Chinese Hamster Ovary, CHO Kl, ATCC: American Type Culture Collec- tion, Manassas, VA 20108, USA). The test cell lines constitutively express the human, rat or dog Via or V2 receptors. In case of the G q-coupled Via receptors, cells are also stably transfected with a modified form of the calcium- sensitive photoproteins aequorin (human and rat Via) or obe- lin (dog Via), which, after reconstitution with the cof actor coelenterazine, emit light when there are increases in free calcium concentrations [Rizzuto R, Simpson AW, Brini M, Pozzan T, Nature 358, 325-327 (1992); Illarionov BA, Bondar VS, Illarionova VA, Vysotski ES, Gene 153 (2), 273- 274 (1995)] . The resulting vasopressin receptor cells react to stimulation of the recombinantly expressed Via receptors by intracellular release of calcium ions, which can be quantified by the resulting photoprotein luminescence. The Gs-coupled V2 receptors are stably transfected into cell lines expressing the gene for firefly luciferase under control of a CRE-responsible promoter. Acti- vation of V2 receptors induces the activation of the CRE-responsive promoter via cAMP increase, thereby inducing the expression of firefly luciferase. The light emitted by photoproteins of Via cell lines as well as the light emitted by firefly luciferase of V2 cell lines corresponds to the activation or inhibition of the respective vasopressin receptor. The bioluminescence of the cell lines is detected using a suitable luminometer [Milligan G, Marshall F, Rees S, Trends in Pharmacological Sciences 17, 235-237 (1996)] .
Test procedure:
Vasopressin Via receptor cell lines:
On the day before the assay, the cells are plated out in culture medium (DMEM/F12, 2% FCS, 2 mM glutamine, 10 mM HEPES, 5 μg/ml coelenterazine) in 384-well microtiter plates and kept in a cell incubator (96% humidity, 5% v/v CO2, 37°C). On the day of the assay, test compounds in various concentrations are placed for 10 minutes in the wells of the microtiter plate before the agonist [Arg8] -vasopressin at EC50 concentration is added. The resulting light signal is measured immediately in a luminometer.
Vasopressin V2 receptor cell lines:
On the day before the assay, the cells are plated out in culture medium (DMEM/F12, 2% FCS, 2 mM glutamine, 10 mM HEPES) in 384-well microtiter plates and kept in a cell incubator (96% humidity, 5% v/v CO2, 37°C). On the day of the assay, test compounds in various concentrations and the agonist [Arg8] -vasopressin at EC50 concentration are added together to the wells, and plates are incubated for 3 hours in a cell incubator. Upon addition of the cell lysis reagent Triton™ and the substrate luciferin, luminescence of firefly luciferase is measured in a luminometer.
Table 1A below lists individual IC50 values for the compounds of the invention (including racemic mixtures as well as separated enantiomers) that were obtained from cell lines transfected with the human Via or V2 receptor:
Table 1A
Example ICso hVla ICso hV2 ratio ICso
No. [μΜ] [μΜ] hV2/hVla
1 0.00510 0.00873 1.7
2 0.00515 0.02325 4.5
3 0.00670 0.09400 14.0
4 0.00720 0.02700 3.8 0.01750 0.04600 2.6
0.02700 0.39000 14.4
0.05400 0.33000 6.1
0.07000 0.24750 3.5
0.09350 0.24500 2.6
0.09750 0.15250 1.6
0.11500 0.15000 1.3
0.12500 0.15000 1.2
0.13500 0.52500 3.9
0.15200 0.63750 4.2
0.17500 0.03500 0.2
0.69000 2.25000 3.3
0.00045 0.00700 15.6
0.00054 0.00165 3.1
0.00065 0.00491 7.5
0.00075 0.00273 3.7
0.00075 0.00525 7.0
0.00165 0.00363 2.2
0.00245 0.00253 1.0
0.00260 0.03050 11.7
0.00295 0.00645 2.2
0.00350 0.01700 4.9 - I l l -
B-2. Radioactive binding assay
IC50 and Kj values can be determined in radioactive binding assays using membrane fractions of recombinant human embryonic kidney cell line 293 (HEK293) or CHO-K1 cell lines expressing the respective human vasopressin Via and V2 receptors.
Human recombinant vasopressin Via receptors expressed in HEK293 cells are used in 50 mM Tris-HCl buffer, pH 7.4, 5 mM MgCh, 0.1% BSA using standard techniques. Aliquots of prepared membranes are incubated with test compounds in various concentrations in duplicates and 0.03nM [125i]Phenylacetyl-D-Tyr(Me)-Phe-Gln-Asn-Arg-Pro-Arg-Tyr-NH2 for 120 minutes at 25°C. Nonspecific binding is estimated in the presence of 1 μΜ [Arg8]Vasopressin. Receptors are filtered and washed, the filters are then counted to determine [125I]Phenylacetyl-D-Tyr(Me)-Phe-Gln-Asn-Arg- Pro-Arg-Tyr-NH2 specifically bound.
CHO-K1 cells stably transfected with a plasmid encoding human vasopressin V2 receptor are used to prepare membranes in 50 mM Tris-HCl buffer, pH 7.4, 10 mM MgCl2, 0.1% BSA using standard techniques. Aliquots of prepared membrane are incubated with test compounds in various concentrations in duplicates and 4 nM [3H](Arg8)-Vasopressin for 120 minutes at 25°C. Nonspecific binding is estimated in the presence of 1 mM (Arg8)-vasopressin. Membranes are filtered and washed 3 times and the filters are counted to determine [3H](Args)- Vasopressin specifically bound.
IC50 values are determined by a non-linear, least squares regression analysis using MathlQTM (ID Business Solutions Ltd., UK). The inhibition constant ¾ is calculated using the equation of Cheng and Prusoff (Cheng, Y., Prusoff, W.H., Biochem. Pharmacol. 22:3099-3108, 1973).
B-3. Cellular in vitro assay for detecting the action of vasopressin Via receptor antagonists on the regulation of pro-fibrotic genes
The cell line H9C2 (American Type Culture Collection ATCC No. CRL-1446), described as a cardiomyocyte type isolated from rat cardiac tissue, endogenously expresses the vasopressin Via receptor AVPR1 A in high copy number, whereas AVPR2 expression cannot be detected. Likewise, the cell line NRK49F (ATCC No. CRL1570) isolated from rat kidney tissue, shows similar expression pattern of high AVPR1A mRNA expression and diminishing AVPR2 expression. For cell assays detecting the inhibition of AVPR1A receptor-dependent regulation of gene expression by receptor antagonists, the procedure is as follows:
H9C2 cells or NRK49F cells are seeded in 6-well microtiter plates for cell culture at a cell density of 50 000 cells/ well in 2.0 ml of Opti-MEM medium (Invitrogen Corp., Carlsbad, CA, USA, Cat. No. 11058-021) and held in a cell incubator (96% humidity, 8% v/v C02, 37°C). After 24 hours, sets of three wells (triplicate) are charged with vehicle solution (negative control) and vasopressin solution ([Arg8] -vasopressin acetate, Sigma, Cat. No. V9879), or test compound (dissolved in vehicle: water with 20% v/v ethanol) and vasopressin solution. In the cell culture, the final vasopressin concentration is 1 nM. The test compound solution is added to the cell culture in small volumes, so that a final concentration of 0.03% of ethanol in the cell assay is not exceeded. After an incubation time of 5 hours, the culture supernatant is drawn off under suction, the adherent cells are lysed in 350 μΐ of RLT buffer (Qiagen, Cat. No. 79216), and the RNA is isolated from the lysate using the RNeasy kit (Qiagen, Cat. No. 74104). This is followed by DNAse digestion (Invitrogen, Cat. No. 18068-015), cDNA synthesis (Promaga, ImProm-II Reverse Transcription System, Cat. No. A3800) and Reverse Transcription Polymerase Chain Reaction (RTPCR) (pPCR MasterMix RT-QP2X-03-075, Eurogentec, Seraing, Belgium). All procedures take place in accordance with the working protocols of the test reagents' manufacturers. The primer sets for the RTPCR are selected on the basis of the mRNA gene sequences (NCBI GenBank Entrez Nucleotide Data Base) using the Primer3Plus program with 6-FAM TAMRA-labelled probes. The RTPCR for determining the relative mRNA expression in the cells of the various assay batches is carried out using the Applied Biosystems ABI Prism 7700 Sequence Detector in 384-well microtiter plate format in accordance with the instrument operating instructions. The relative gene expression is represented by the delta-delta Ct value [Applied Biosystems, User Bulletin No. 2 ABI Prism 7700 SDS, December 11, 1997 (updated 10/2001)] with reference to the level of expression of the ribosomal protein L-32 gene (GenBank Acc. No. NM_013226) and the threshold Ct value of Ct = 35.
B-4. Inhibition of vasopressin induced aggregation of human platelets
Human platelets endogenously express the Via receptor. It was found that relatively high vasopressin concentrations (ca. 50-100 nM) stimulate platelet aggregation ex vivo. Therefore, platelets enriched from human blood may serve as a Via expressing tissue for pharmacological studies with corresponding high concentrations of vasopressin antagonists.
Human blood is collected in a 10 mM trisodium citrate solution by venous puncture from nonsmoking healthy volunteers (n=4-8) who were drug free for at least 1 week. Platelet-rich plasma (PRP) is obtained by centrifuging the blood sample at 140 g for 20 min at 4°C. The resulting pellet is further centrifuged (15.000 rpm, 2 min) to produce platelet-poor plasma (PPP). Platelet aggregation is measured turbidimetrically using an aggregometer (APACT 4). The reaction is followed by monitoring changes in light transmission on 178 μΐ^ PRP aliquots, under continuous stirring at 37°C, against PPP control. Various concentrations of vasopressin antagonists (in 2 μΐ^) are added to PRP 5 min before the addition of 20 μΐ^ Arg-vasopressin (final concentration 100 nM. The inhibitory effects of the compounds are determined by measuring the height of the aggregation wave from the bottom of the shape change compared with the control response. IC50 values are calculated a dose-response inhibition curve by an iterative nonlinear regression program
B-5. Effects on the contraction of isolated rat vessel rings
Isolated aorta
Test compounds can be investigated on isolated aortic rings from male Wistar rats endogenously expressing the Via receptor. Male Wistar rats are euthanized using carbon dioxide. The aorta is removed and placed in ice-cold Krebs-Henseleit buffer of following composition (in mmol/1): NaCl 112, KC1 5.9, CaCl2 2.0, MgCl2 1.2, NaH2P04 1.2, NaHC03 25, glucose 11.5. The aorta is cut into 3 mm rings and transferred to 20 ml organ baths containing Krebs-Henseleit solution equilibrated with 95% 02, 5% CO2 at 37°C. For recording of isometric tension the rings are mounted between two hooks. The resting tension is adjusted to 3 g. After an equilibration period, each experiment is started by exposing the preparation to K+ (50 mM) Krebs-Henseleit solution. The aortic rings are than pre-contracted using 1 nmol/1 Arg-vasopressin. After a stable contraction is established, a cumulative dose response curve of the test compound is constructed. The stabilized contraction induced by Arg-vasopressin is defined as 100% tension. The relaxation is expressed as percentage tension.
Isolated A. renalis Male Wistar rats (200-250 g) are euthanized using carbon dioxide. The A. renalis is removed and placed in ice-cold Krebs-Henseleit buffer of following composition (in mmol/1): NaCl 112, KCl 5.9, CaCl2 2.0, MgCl2 1.2, NaH2P04 1.2, NaHCOs 25, glucose 11.5. For measurement of isometric tension, ring segments, 2 mm in length, are mounted in a small vessel chamber myograph (Danish Myo Technology A/S, Denmark) using two tungsten wires fixed to mounting jaws. One mounting jaw is attached to a micrometer, allowing control of vessel circumference. The other mounting jaw is attached to a force transducer for measurement of tension development. The whole preparation is kept in a chamber with physiological salt solution at 37°C, bubbled with oxygen. After a 30 min equilibration period, the vessels are stretched to their optimal lumen diameter for active tension development which is determined based on the internal circumference-wall tension ratio. The internal circumference is set to 90% of what the vessels would have if they are exposed to a passive tension equivalent to that produced by a transmural pressure of 100 mmHg.
Afterwards, the vessels are washed three times with Krebs-Henseleit buffer and left to equilibrate for 30 min. The contractility is then tested by a twofold exposure to a high K+ solution (50 mmol/1 KCl). After washing with Krebs-Henseleit buffer the vessels are then pre-contracted using 1 nmol/1 Arg- vasopressin. After a stable contraction is established, a cumulative dose response curve of the test compound is constructed. The stabilized contraction induced by Arg-vasopressin is defined as 100% tension. The relaxation is expressed as percentage tension.
B-6. In vivo assay for detecting cardiovascular effects: blood pressure measurement in anaesthetized rats (vasopressin 'challenge' model)
Male Sprague-Dawley rats (250-350 g body weight) are used under ketamine/ xylazine/ pentobarbital injection anaesthesia. Polyethylene tubes (PE-50, Intramedic®), prefilled with heparin-containing (500 IU/ml) isotonic sodium chloride solution, are introduced into the jugular vein and the femoral vein and then tied in. Arg-vasopressin (SIGMA) is injected via one venous access, with the aid of a syringe; the test substance is administered via the second venous access. For determination of the systolic blood pressure, a pressure catheter (Millar SPR-320 2F) is tied into the carotid artery. The arterial catheter is connected to a pressure transducer which feeds its signals to a recording computer equipped with suitable recording software. In a typical experiment, the experimental animal is administered 3-4 successive bolus injections at intervals of 10-15 min with a defined amount of Arg-vasopressin (30 ng/kg) in isotonic sodium chloride solution. When the blood pressure has reached initial levels again, the test substance is administered as a bolus, with subsequent continuous infusion, in a suitable solvent. After this, at defined intervals (10-15 min), the same amount of Arg-vasopressin as at the start is administered again. On the basis of the blood pressure values, a determination is made of the extent to which the test substance counteracts the hypertensive effect of Arg-vasopressin. Control animals only receive solvent instead of the test substance. Following intravenous administration, the compounds of the invention, in comparison to the solvent controls, bring about an inhibition of the blood pressure increase caused by Arg-vasopressin.
B-7. In vivo assay for detecting cardiovascular effects: diuresis investigations in conscious rats kept in metabolism cages
Wistar rats (220-450 g body weight) are kept with free access to feed (Altromin) and drinking water. During the experiment, the animals are kept with free access to drinking water for 4 to 8 or up to 24 hours individually in metabolism cages suitable for rats of this weight class (Tecniplast Deutschland GmbH, D-82383 HohenpeiBenberg). At the beginning of the experiment, the animals are administered the test substance in a volume of 1 to 3 ml/kg body weight of a suitable solvent by means of gavage into the stomach. Control animals only receive solvent. Controls and substance tests are carried out in parallel on the same day. Control groups and substance-dose groups each consist of 4 to 8 animals. During the experiment, the urine excreted by the animals is collected continuously in a receiver at the base of the cage. The volume of urine per time unit is determined separately for each animal, and the concentration of urinary electrolytes is measured by standard methods of flame photometry. Before the beginning of the experiment, the body weight of the individual animals is determined.
B-8. In vivo assay for detecting protective renal effects: Acute ischemia/reperfusion injury model in rodents
Laboratory bred male C57B1/6J mice 6-8 weeks old are obtained from Taconic Biosciences, male 6-8 weeks old Sprague Dawley® rat are obtained from Charles River. Both rats and mice are maintained under standard laboratory conditions, 12 hour light-dark cycles with access to normal chow and drinking water at libitum. For the ischemia reperfusion injury model a total of 10-12 rats or mice is used in each control and experimental group.
Animals are anesthetized with continuous inhaled isoflurane. A right nephrectomy is performed through a right flank incision 7 days before the ischemic procedures in the contralateral kidneys. For renal ischemia a left flank incision is made. Renal vessels are exposed by dissection of the left renal pedicle. Non-traumatic vascular clamps are used to stop blood flow (artery and vein) during 45 min (rats) or 25 min (mice) of ischemia. Reperfusion is established by removing the clamps. The abdominal wall (muscular layer and skin) is closed with 5.0 polypropylene sutures. Temgesic® (Buprenorphin, 0.025 mg/kg s.c.) is applied as an analgesic.
Urine of each animal is collected in metabolic cages over night before sacrifice at 24h post ischemia. Upon sacrifice, blood samples are obtained under terminal anesthesia. After centrifugation of the blood samples, serum is isolated. Both serum creatinine and serum urea are measured via clinical biochemistry analyzer (Pentra 400). For the assessment of serum and urinary kidney injury biomarkers (Neutrophil gelatinase-associated lipocalin [NGAL], kidney injury molecule- 1 [KIM-1] and Osteopontin) ELlSA's are performed according to the manufacturers protocol. Both urinary creatinine and albumin are measured to determine the albumin/creatinine ratio.
Total RNA is isolated from kidneys. Left kidneys are snap-frozen in liquid nitrogen at sacrifice. Kidney tissue is then homogenized and RNA is obtained. Total RNA is transcribed to cDNA. Using TaqMan real-time PCR renal NGAL, Osteopontin, KIM-1, Nephrin and Podocin mRNA expression is analyzed in whole kidney tissue.
Differences between groups are analyzed by one-way ANOVA with Dunnett's corrections for multiple comparisons. Statistical significance is defined as p < 0.05. All statistical analyses are done using GraphPad Prism 6.
B-9. In vivo assay for detecting cardiovascular effects: hemodynamic investigations in anaesthetized dogs
Male beagle dogs (Beagle, Marshall BioResources, USA) with a weight of between 10 and 15 kg are anesthetized with pentobarbital (30 mg/kg iv, Narcoren®, Merial, Germany) for the surgical interventions and the hemodynamic and functional investigation termini. Pancuroniumbromide (Pancuronium Inresa, Inresa, Germany, 2-4 mg/animal i.v.) serves additionally as a muscle relaxant. The dogs are intubated and ventilated with an oxygen/ambient air mixture (30/70%), about 2,5-4 L/min. Ventilation takes place using a ventilator from GE Healthcare (Avance, Germany) and is monitored using a carbon dioxide analyzer (-Datex Ohmeda). The anesthesia is maintained by continual infusion of pentobarbital (50 μg/kg/min); fentanyl is used as an analgesic (10 g/kg/h).
In preparatory interventions, the dogs are fitted with a cardiac pacemaker. At start of experiment, a cardiac pacemaker from Biotronik (Logos®, Germany) is implanted into a subcutaneous skin pocket and is contacted with the heart via a pacemaker electrode (Siello S60®, Biotronik, Germany) which is advanced through the external jugular vein, with illumination, into the right ventricle.
Thereafter accesses are removed and the dog wakes spontaneously from the anesthesia. After a further 7 days, the above-described pacemaker is activated and the heart is stimulated at a frequency of 220 beats per minute.
The actual drug testing experiments take place 28 days after the beginning of pacemaker stimulation, using the following instrumentation:
• Introduction of a bladder catheter for bladder relief and for measuring the flow of urine
• Attachment of electrocardiography (ECG) leads to the extremities for ECG measurement • Introduction of a sheath introducer filled with sodium chloride solution into the femoral artery. This tube is connected to a pressure sensor (Braun Melsungen, Melsungen, Germany) for measuring the systemic blood pressure
• Introduction of a Millar Tip catheter (type 350 PC, Millar Instruments, Houston, USA) through a port secured in the carotid artery, for measuring cardiac hemodynamics .
• Introduction of a Swan-Ganz catheter (CCOmbo 7.5F, Edwards, Irvine, USA) via the jugular vein into the pulmonary artery, for measuring the cardiac output, oxygen saturation, pulmonary arterial pressures and central venous pressure
• Siting of a venous catheter in the cephalic vein, for infusing pentobarbital, for liquid replacement and for blood sampling (determination of the plasma levels of substance or other clinical blood values)
• Siting of a venous catheter in the saphenous vein, for infusing fentanyl and for administration of substance
• Infusion of vasopressin (Sigma) in increasing dosage, up to a dose of 4 mU kg/min. The pharmacological substances are then tested with this dosage.
The primary signals are amplified if necessary (ACQ7700, Data Sciences International, USA or Edwards-Vigilance-Monitor, Edwards, Irvine, USA) and subsequently fed into the Ponemah system (Data Sciences International, USA) for evaluation. The signals are recorded continuously throughout the experimental period, and are further processed digitally by said software, and averaged over 30 seconds.
C) Working examples of pharmaceutical compositions
The substances according to the invention can be converted to pharmaceutical preparations as follows:
Tablet:
Composition:
100 mg of the compound of Example 1, 50 mg of lactose (monohydrate), 50 mg of maize starch, 10 mg of polyvinylpyrrolidone (PVP 25) (from BASF, Germany) and 2 mg of magnesium stearate.
Tablet weight 212 mg. Diameter 8 mm, radius of curvature 12 mm.
Production:
The mixture of the compound of Example 1, lactose and starch is granulated with a 5% strength solution (m/m) of the PVP in water. After drying, the granules are mixed with the magnesium stearate for 5 min. This mixture is compressed in a conventional tabletting press (see above for format of the tablet).
Oral suspension:
Composition:
1000 mg of the compound of Example 1, 1000 mg of ethanol (96%), 400 mg of Rhodigel (xanthan gum) (from FMC, USA) and 99 g of water.
10 ml of oral suspension correspond to a single dose of 100 mg of the compound of the invention. Production:
The Rhodigel is suspended in ethanol, and the compound of Example 1 is added to the suspension. The water is added while stirring. The mixture is stirred for about 6 h until swelling of the Rhodigel is complete.
Sterile i.v. solution:
The compound according to the invention is dissolved at a concentration below saturation solubility in a physiologically acceptable solvent (for example isotonic sodium chloride solution, glucose solution 5% and/or PEG 400 solution 30%). The solution is sterilized by filtration and filled into sterile and pyrogen-free injection containers.
Although the invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of the invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The claims are intended to be construed to include all such embodiments and equivalent variations.

Claims

CLAIMS:
1. A compound of general formula (I)
in which
R1 represents a group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
Ar represents a phenyl group or a 5- or 6-membered heteroaryl group attached via a ring carbon atom having one or two ring heteroatoms selected from a nitrogen atom and a sulfur atom,
wherein any phenyl group and any 5- or 6-membered heteroaryl group are each optionally substituted, identically or differently, with one or two groups selected from a halogen atom, nitro, cyano, (Ci-C -alkyl, (Ci-C -alkoxy, (Ci-C -alkylsulfanyl, (Ci- C -alkoxycarbonyl, aminocarbonyl and
wherein said (Ci-C -alkyl group, said (Ci-C -alkoxy group and said (C1-C4)- alkylsulfanyl group are each optionally substituted with up to three fluorine atoms,
or a pharmaceutically acceptable salt, hydrate and/or solvate thereof.
2. A compound of general formula (I) according to Claim 1, wherein
Ar represents a group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
R2A represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, methyl, ethyl, isopropyl, nitro, methoxy, difluoromethoxy and methylsulfanyl,
R2B and R2C represent, independently from each other, a group selected from a fluorine atom, a chlorine atom, methyl and ethyl,
R2D, R2E and R2F represent, independently from each other, a group selected from a fluorine atom and a chlorine atom,
R3A represents a group selected from a hydrogen atom, a fluorine atom, methyl, ethyl, methoxy and methoxycarbonyl,
R3B represents a chlorine atom,
R3C represents a group selected from a chlorine atom and ethoxy,
R3D represents aminocarbonyl,
R3E represents a methyl group,
R3F represents a chlorine atom,
R3G represents a group selected from a fluorine atom, a chlorine atom and methyl,
R4A represents a methoxy group,
a pharmaceutically acceptable salt, hydrate and/or solvate thereof.
3. A compound of general formula (I) according to Claim 1 or 2, wherein
R1 represents a group of the formula
in which
represents the point of attachment to the nitrogen atom,
Ar represents a group of the formula
in which
represents the point of attachment to the nitrogen atom,
R2A represents a group selected from a chlorine atom, a bromine atom, isopropyl, and nitro,
R3G represents a group selected from a chlorine atom and methyl,
or a pharmaceutically acceptable salt, hydrate and/or solvate thereof.
4. A compound of general formula (I) according to Claim 1 or 2, wherein
R1 represents a group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
Ar represents a group of the formula
in which
#l represents the point of attachment to the nitrogen atom,
R2A represents a group selected from a hydrogen atom, a fluorine atom, and methylsulfanyl,
R2B and R2C represent, independently from each other, a group selected from a fluorine atom, a chlorine atom, methyl and ethyl,
R2F represents a group selected from a fluorine atom and a chlorine atom,
R3A represents a group selected from a hydrogen atom, a fluorine atom and ethyl, or a pharmaceutically acceptable salt, hydrate and/or solvate thereof.
A method of preparing a compound of general formula (I) according to any one of claims 1 to 4 said method comprising the step
[A] of allowing an intermediate compound of formula (II):
in which R1 is as defined for the compound of general formula (I) according to any one of claims 1 to 4,
R5 represents a (Ci-C -alkyl group, in particular a methyl group,
to react in a first step in the presence of a base, and optionally a copper salt, with a compound of general formula (III):
in which
R6 represents a (Ci-C -alkyl group, in particular a methyl group,
to give an intermediate compound, which is then allowed to react in the presence of a base in a second step with a hydrazine compound of general formula (IV) or a respective salt thereof HN 2
I
Ar (IV)
in which Ar is as defined for the compound of general formula (I) according to any claims 1 to 4,
thereby giving a compound of general formula (V) :
in which R and Ar are as defined for the compound of general formula (I) according to any one of claims 1 to 4, and
R6 represents a (Ci-C -alkyl group, in particular a methyl group,
followed by a subsequent step
[B] of allowing the compound of formula (V) obtained in step [A] to react with ammonia thereby giving a compound of general formula (I):
in which R1 and Ar are as defined for the compound of general formula (I) according to any one of claims 1 to 4,
optionally followed by step
[C] conversion of the alcohols of general formula (I- A):
in which Ar is as defined for the compound of general formula (I) according to any one of claims 1 to 4,
to the ketones of general formula (I-B):
in which Ar is as defined for the compound of general formula (I) according to any one of claims 1 to 4,
using known oxidation methods,
each [B] and [C] optionally followed, where appropriate, by (i) separating the compounds of formula (I) thus obtained into their respective enantiomers, and/or (ii) converting the compounds of formula (I) into their respective hydrates, solvates, salts and/or hydrates or solvates of the salts by treatment with the corresponding solvents and/or acids or bases.
6. Compound as defined in any of Claims 1 to 4 for use in the treatment and/or prevention of diseases.
7. Compound as defined in any of Claims 1 to 4 for use in a method for the treatment and/or prevention of acute and chronic kidney diseases including diabetic nephropathy, acute and chronic heart failure, preeclampsia, peripheral arterial disease (PAD), coronary microvascular dysfunction (CMD), Raynaud's syndrome, dysmenorrhea, cardiorenal syndrome, hypervolemic and euvolemic hyponatremia, liver cirrhosis, ascites, edema and the syndrome of inadequate ADH secretion (SIADH). Use of a compound as defined in any of Claims 1 to 4 for the manufacture of a pharmaceutical composition for the treatment and/or prevention of acute and chronic kidney diseases including diabetic nephropathy, acute and chronic heart failure, preeclampsia, peripheral arterial disease (PAD), coronary microvascular dysfunction (CMD), Raynaud's syndrome, dysmenorrhea, cardiorenal syndrome, hypervolemic and euvolemic hyponatremia, liver cirrhosis, ascites, edema and the syndrome of inadequate ADH secretion (SIADH).
Pharmaceutical composition comprising a compound as defined in any of Claims 1 to 4 and one or more pharmaceutically acceptable excipients.
Pharmaceutical composition comprising one or more first active ingredients, in particular compounds of general formula (I) according to any one of claims 1 to 4, and one or more further active ingredients, in particular one or more additional therapeutic agents selected from the group consisting of diuretics, angiotensin All antagonists, ACE inhibitors, beta- receptor blockers, mineralocorticoid receptor antagonists, organic nitrates, NO donors, activators and stimulators of the soluble guanylate cyclase, and positive-inotropic agents, antiinflammatory agents, immunosuppressive agents, phosphate binders and/or compounds which modulate vitamin D metabolism.
The pharmaceutical composition as defined in Claim 9 or 10 for the treatment and/or prevention of acute and chronic kidney diseases including diabetic nephropathy, acute and chronic heart failure, preeclampsia, peripheral arterial disease (PAD) and coronary microvascular dysfunction (CMD), Raynaud's syndrome, dysmenorrhea, cardiorenal syndrome, hypervolemic and euvolemic hyponatremia, liver cirrhosis, ascites, edema and the syndrome of inadequate ADH secretion (SIADH).
Method for the treatment and/or prevention of acute and chronic kidney diseases including diabetic nephropathy, acute and chronic heart failure, preeclampsia peripheral arterial disease (PAD) and coronary microvascular dysfunction (CMD), Raynaud's syndrome, dysmenorrhea, cardiorenal syndrome, hypervolemic and euvolemic hyponatremia, liver cirrhosis, ascites, edema and the syndrome of inadequate ADH secretion (SIADH) in a human or other mammal, comprising administering to a human or other mammal in need thereof a therapeutically effective amount of one or more compounds as defined in any of Claims 1 to 4, or of a pharmaceutical composition as defined in any of Claims 9 to 11.
EP17720501.0A 2016-05-03 2017-05-02 Amide-substituted aryltriazole derivatives and uses thereof Withdrawn EP3452468A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP2016168169 2016-05-03
PCT/EP2017/060368 WO2017191105A1 (en) 2016-05-03 2017-05-02 Amide-substituted aryltriazole derivatives and uses thereof

Publications (1)

Publication Number Publication Date
EP3452468A1 true EP3452468A1 (en) 2019-03-13

Family

ID=65238432

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17720501.0A Withdrawn EP3452468A1 (en) 2016-05-03 2017-05-02 Amide-substituted aryltriazole derivatives and uses thereof

Country Status (1)

Country Link
EP (1) EP3452468A1 (en)

Similar Documents

Publication Publication Date Title
AU2017259870B2 (en) Amide-substituted pyridinyltriazole derivatives and uses thereof
US10525041B2 (en) Fluoroalkyl-substituted aryltriazole derivatives and uses thereof
US10526314B2 (en) Hydroxyalkyl-substituted heteroaryltriazole derivatives and uses thereof
EP3452470B1 (en) Amide-substituted phenyltriazole derivatives and uses thereof
US10927098B2 (en) Hydroxyalkyl-substituted triazole derivatives and uses thereof
WO2017191105A1 (en) Amide-substituted aryltriazole derivatives and uses thereof
WO2019081302A1 (en) Substituted triazole derivatives and uses thereof
EP3700900A1 (en) Substituted triazole derivatives and uses thereof
US11230540B2 (en) Substituted triazole derivatives and uses thereof
EP3700898A1 (en) Substituted triazole derivatives and uses thereof
EP3700896A1 (en) Amine substituted triazole derivatives and uses thereof
EP3452468A1 (en) Amide-substituted aryltriazole derivatives and uses thereof

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

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

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

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

Free format text: ORIGINAL CODE: 0009012

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

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20181203

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20190920

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

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

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

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20210303

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

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

18D Application deemed to be withdrawn

Effective date: 20210714