HETEROCYCLIC SUBSTITUTED TRIFLUOROMETHYL PYRIMIDINONES AND THEIR USE

Information

  • Patent Application
  • 20160237059
  • Publication Number
    20160237059
  • Date Filed
    September 12, 2014
    9 years ago
  • Date Published
    August 18, 2016
    7 years ago
Abstract
The present application relates to novel heterocyclically substituted 6-(trifluoromethyl)pyrimidin-4(3H)-one derivatives, to processes for their preparation, to their use alone or in combinations for the treatment and/or prevention of diseases, and to their use for preparing medicaments for the treatment and/or prevention of diseases, in particular for treatment and/or prevention of cardiovascular, renal, inflammatory and fibrotic diseases.
Description

The present application relates to novel heterocyclically substituted 6-(trifluoromethyl)pyrimidin-4(3H)-one derivatives, to processes for their preparation, to their use alone or in combinations for the treatment and/or prevention of diseases, and to their use for preparing medicaments for the treatment and/or prevention of diseases, in particular for treatment and/or prevention of cardiovascular, renal, inflammatory and fibrotic diseases.


BACKGROUND OF THE INVENTION

Chemotactic cytokines or chemokines can be produced in most tissues, such as heart, kidney and lung, but also vessels, in the context of an immune response to tissue injury or inflammatory stimuli, for example bacterial toxins. They are essential for the recruitment of specific leukocyte subpopulations (such as neutrophiles, monocytes, basophiles, eosinophiles, effector-T-cells, dendritic cells) to the site of an inflammation [Mackay, Nature Immunol. 2 (2), 95-101 (2001)]. Binding to glycosaminoglycans of the extracellular matrix and the endothelium results in a local chemokine concentration gradient which allows chemotactic leukocyte migration to the inflammation or infection site in the body [Tanaka et al., Nature 361, 79-82 (1993); Luster, N. Engl. J. Med. 338 (7), 436-445 (1998)]. By virtue of the recruitment of inflammatory cells, chemokines therefore play a central role in the genesis and progression of numerous inflammatory disorders [Schall, Cytokine 3, 165-183 (1991); Schall et al., Curr. Opin. Immunol. 6, 865-873 (1994)]. In addition to the chemotactic action chemokines are also involved in the regulation of haematopoiesis, cell proliferation, angiogenesis or tumour growth, inter alia.


According to organization and position of conserved cysteine residues, the chemokines are classified into four different sub-groups (CXC, CC, C and CX3C) [Bacon et al., J. Interferon Cytokine Res. 22 (10), 1067-1068 (2002)]. The largest family are the CC chemokines, which also include the classic inflammatory chemokines such as the MCPs (monocyte chemoattractant proteins) whose expression is induced in most tissues in case of tissue damage or infection via proinflammatory cytokines such as IL-1, TNF-α or IFN-γ [Rollins, in: Cytokine Reference, Oppenheim et al., Ed., Academic Press, London, 1145-1160 (2000)]. The 48 chemokines hitherto identified in man bind to specific chemokine receptors which belong to the family of the G-protein-coupled receptors.


The CC chemokine receptor CCR2 is expressed inter alia on the surface of macrophages, monocytes, B cells, activated T cells, dendritic cells, epithelial cells and activated endothelial cells and binds the inflammatory chemokines MCP-1 (CCL2), MCP-2 (CCL8), MCP-3 (CCL7) and MCP-4 (CCL13). As the only ligand, MCP-1 appears to bind selectively to CCR2 [Struthers and Pasternak, Current Topics in Medicinal Chemistry 10 (13), 1278-1298 (2010)]. MCP-1 is expressed inter alia by cardiomyocytes, mesangial cells, alveolar cells, T lymphocytes, macrophages and monocytes [Deshmane et al., J. Interferon Cytokine Res. 29, 313-326 (2009)]. The CC chemokine receptor CCR2 is also the only high affinity receptor for MCP-1 characterized [Struthers and Pasternak, Current Topics in Medicinal Chemistry 10 (13), 1278-1298 (2010)]. In man, CCR2 is expressed on most blood monocytes [Tacke and Randolph, Immunobiology 211, 609-618 (2006)]. The activation of CCR2 by MCP-1 plays an important role in the infiltration and activation of monocytes [Dobaczewski and Frangogiannis, Frontiers in Bioscience Si, 391-405 (2009); Charo and Ransohoff, N. Engl. J. Med. 354 (6), 610-621 (2006)] in the context of the cellular immune response and in chronic inflammatory processes, for example in the heart and the kidney. This infiltration of monocytes and their differentiation in macrophages also represents a second source of pro-inflammatory modulators such as TNF-α, IL-8, IL-12 and matrix metalloproteases (MMPs), inter alia.


Furthermore, CCR2 mediates the migration of monocytes from the bone marrow and their subsequent invasion of inflammatory regions [Carter, Expert Opin. Ther. Patents 23 (5), 549-568 (2013)]. In addition, it appears that fibrocytes may also be formed from the population of the CCR2+ monocytes [Dobaczewski and Frangogiannis, Frontiers in Bioscience S1, 391-405 (2009)], which implies a role of CCR2 in fibrosis (for example of the lung or the liver). The CCR2-mediated invasion of monocytes is also one of the first steps of the formation of atherosclerosis [Gu et al., Mol. Cell 2 (2), 275-281 (1998)].


Experiments with animal models have shown that inhibition of the interaction of MCP-1 and CCR2—by inhibiting the activation of CCR2 using specific antagonists or MCP-1-selective antibodies or by genetic deletion (knock-out) of MCP-1 or CCR2-can reduce an inflammatory response in various disorders and monocyte-infiltration into inflamed lesions can be reduced (arthritis, asthma). CCR2/MCP-1-mediated cellular responses are involved in numerous disorders such as cardiomyopathies, myocardial infarction, myocarditis, chronic heart failure, diabetic renal disease, acute kidney damage, rheumatoid arthritis, multiple sclerosis, chronic-obstructive pulmonary disease (COPD), asthma, atherosclerosis, inflammatory bowel diseases (IBD), diabetes, neuropathic pain, macular degeneration, angiogenesis and cancer [Struthers and Pasternak, Current Topics in Medicinal Chemistry 10 (13), 1278-1298 (2010); Carter, Expert Opin. Ther. Pat. 23 (5), 549-568 (2013); Higgins et al., in: Chemokine Research, Basic Research and Clinical Application, Vol. II, Birkhiuser-Verlag, 115-123 (2007)].


CCR2 and Heart Failure/Cardioprotection:

In myocardial infarction, neutrophiles accumulate in the first hours after ischaemia, with maximum accumulation after one day. Various experimental studies on animals have confirmed that subsequently, in the first two weeks after infarction, monocytes and macrophages dominate the cell infiltrate [Nahrendorf et al., Circulation 121, 2437-2445 (2010)]. This is accompanied by upregulation of MCP-1 [Hayasaki et al., Circ. J. 70 (3), 342-351 (2006)]. Neutrophiles and also monocytes and macrophages produce local proteolytic enzymes and reactive oxygen species (ROS), thus damaging the cardiomyocytes which have survived the ischaemic period. Preclinical studies have shown that the infarct size can be reduced by anti-inflammatory treatment. It is expected that such a protection will also occur in patients suffering from acute myocardial infarction, which may reduce the infarct size and prevent a worsening of the cardiac function after the infarct.


CCR2-deficient mice show a reduction of the infarct size and reduced remodelling after myocardial infarction [Hayasaki et al., Circ. J. 70 (3), 342-351 (2006)]. Likewise, MCP-1-deficient mice have reduced remodelling after myocardial infarction [Dewald et al., Circ. Res. 96 (8), 881-889 (2005)]. In particular, ApoE−/− mice also show significantly improved infarct healing if the CCR2 receptor is blocked [Majmudar et al., Circulation 127, 2038-2046 (2013)]. In addition, it has been described that, compared to healthy controls, monocytes in patients suffering from heart failure release more MCP-1 [Aukrust et al., Circulation 97, 1136-1143 (1998); Aukrust et al., Arterioscler. Thromb. Vasc. Biol. 28, 1909-1919 (2008)], and increased MCP-1 plasma levels were also detected in patients with atrial fibrillation [Li et al., Heart Rhythm 7, 438-444 (2010)].


CCR2 and Kidney Function/Nephroprotection:

Immunological and inflammatory mechanisms play a crucial role in the development and progression of diabetic nephropathy. Here, monocytes and/or macrophages have a substantial effect in the pathogenesis [Chow et al., Kidney Int. 65, 116-128 (20014); Chow et al., Kidney Int. 69, 73-80 (2006)]. Deletion of CCR2 or blocking of the MCP-1 signal path reduces macrophage infiltration and reduces kidney damage both in Type 1 and in Type 2 diabetes in mice. In leptin receptor-deficient db/db mice, a murine model of Type 2 diabetes, treatment with CCR2-blocking substances leads to reduced albuminuria [Okamoto et al., Biol. Pharm. Bull. 35 (11), 2069-2074 (2012); Sayyed et al., Kidney Int. 80, 68-78 (2011)]. In humans, too, accumulation of macrophages can be observed in diabetic nephropathy, and this correlates strongly with the progression of renal dysfunction [Kelly et al., Am. J. Nephrol. 32, 469-475 (2010); Nguyen et al., Nephrology 11, 226-231 (2006)]. Furthermore, the urine and plasma concentrations of MCP-1 in patients correlate with renal function and the stage of the chronic kidney disease [Eardley et al., Kidney Int. 69, 1189-1197 (2006); Stinghen et al., Nephron Clin. Pract. 111, c117-c126 (2009)], which suggests a critical role of macrophages in the pathogenesis of diabetic nephropathy.


Experimental data additionally confirm a reduction of reperfusion damage after renal ischaemia/reperfusion and reduced fibrosis in the unilateral ureteral obstruction (UUO) model in CCR2 knock-out animals [Furuichi et al., J. Am. Soc. Nephrol. 14, 2503-2515 (2003); Kitagawa et al., Am. J. Pathol. 165 (1), 237-246 (2004)].


It was therefore an object of the present invention to identify and provide novel substances which act as potent antagonists of the CCR2 receptor and are suitable as such for treatment and/or prevention of disorders, in particular cardiovascular, renal, inflammatory and fibrotic disorders.


JP 54-115384-A [Chem. Abstr. 92:128952] and WO 2007/048734-A1 disclose 2-pyrazolylpyrimidines as fungicides, and WO 93/22311-A1 describes diazine-substituted pyrimidines as fungicides. DE 1 695 270-A discloses 2-amino-4-hydroxypyrimidine derivatives also having fungicidal action.


Heterocyclically substituted pyrimidine derivatives having pharmacological activity, which can be used for treating various disorders, are described, inter alia, in WO 95/11235-A1, WO 03/051906-A2, WO 03/072107-A1, WO 2004/111014-A1, WO 2005/026148-A1, WO 2005/099688-A2, WO 2006/066070-A2, WO 2008/009963-A2, WO 2009/019656-A1, WO 2010/144345-A1, WO 2011/022440-A2, WO 2011/026835-A1, WO 2011/092140-A1 and WO 2014/026039-A2. WO 2011/114148-A1 and WO 2012/041817-A1 recently disclosed bicyclic pyrimidine derivatives as antagonists of the CCR2 receptor.


The compound 5-(2-chloro-6-fluorobenzyl)-2-(pyridin-3-yl)-6-(trifluoromethyl)pyrimidin-4(1H)-one is indexed as “Chemical Library” substance [Chem. Abstr. Registry-No. 685113-32-0]. A therapeutic use of this compound has not been described.


The present invention provides compounds of the general formula (I)




embedded image


in which

  • A represents C—H, C—F or N,
  • E represents CH2, O or S,
  • R1 and R2 independently of one another represent hydrogen, fluorine, chlorine, methyl, trifluoromethyl or trifluoromethoxy,
    • where at least one of the two radicals R1 and R2 represents fluorine, chlorine, methyl, trifluoromethyl or trifluoromethoxy,
  • L represents a bond, CH2 or NH,
  • and
  • Het represents pyridyl, pyrimidinyl, pyrazinyl or pyridazinyl which may be (i) mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, bromine, trifluoromethyl, (C1-C4)-alkyl, phenyl, hydroxy, trifluoromethoxy, (C1-C4)-alkoxy, (C1-C4)-alkoxymethyl, (trifluoromethyl)sulphanyl, (C1-C4)-alkylsulphanyl, (C1-C4)-alkylsulphinyl, (C1-C4)-alkylsulphonyl, amino, mono-(C1-C4)-alkylamino, di-(C1-C4)-alkylamino and (C1-C4)-alkylcarbonylamino and which may be (ii) fused with a phenyl or pyridyl ring which for its part may be substituted by fluorine, chlorine, methyl, trifluoromethyl, methoxy, trifluoromethoxy, amino or acetylamino
    • or
    • represents 5-membered heteroaryl which contains one, two or three identical or different ring heteroatoms selected from the group consisting of N, O and S and which (i) may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, (C1-C4)-alkyl, cyclopropyl, phenyl, hydroxy, (C1-C4)-alkoxy, (C1-C4)-alkoxymethyl, (C1-C4)-alkylsulphanyl, (C1-C4)-alkylsulphinyl, (C1-C4)-alkylsulphonyl, amino, mono-(C1-C4)-alkylamino, di-(C1-C4)-alkylamino, (C1-C4)-alkylcarbonylamino, hydroxycarbonyl and (C1-C4)-alkoxycarbonyl and which (ii) may be fused with a phenyl or pyridyl ring which for its part may be substituted by fluorine, chlorine, methyl, trifluoromethyl, methoxy, trifluoromethoxy, amino or acetylamino,
    • or
    • represents 5- or 6-membered saturated or partially unsaturated heterocyclyl which contains one, two or three identical or different ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of (C1-C4)-alkyl, hydroxy, oxo, amino, imino, hydroxycarbonyl and (C1-C4)-alkoxycarbonyl,


      and the N-oxides, salts, solvates, salts of the N-oxides and solvates of the N-oxides or salts thereof.


Compounds according to the invention are the compounds of the formula (I) and their salts, solvates and solvates of the salts, the compounds, comprised by formula (I), of the formulae mentioned below and their salts, solvates and solvates of the salts and the compounds comprised by formula (I), mentioned below as working examples, and their salts, solvates and solvates of the salts, if the compounds, comprised by formula (I), mentioned below are not already salts, solvates and solvates of the salts.


Compounds according to the invention are likewise N-oxides of the compounds of the formula (I) and the salts, solvates and solvates of the salts thereof.


In the context of the present invention, preferred salts are physiologically acceptable salts of the compounds according to the invention. Also encompassed are salts which are not themselves suitable for pharmaceutical applications but can be used, for example, for the isolation, purification or storage of the compounds according to the invention.


Physiologically acceptable salts of the compounds according to the invention include acid addition salts of mineral acids, carboxylic acids and sulphonic acids, for example salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, ethanesulphonic acid, benzenesulphonic acid, toluenesulphonic acid, naphthalenedisulphonic acid, formic acid, acetic acid, trifluoroacetic acid, propionic acid, succinic acid, fumaric acid, maleic acid, lactic acid, tartaric acid, malic acid, citric acid, gluconic acid, benzoic acid and embonic acid.


Physiologically acceptable salts of the compounds according to the invention also include salts derived from conventional bases, by way of example and with preference alkali metal salts (e.g. sodium and potassium salts), alkaline earth metal salts (e.g. calcium and magnesium salts), zinc salts and ammonium salts derived from ammonia or organic amines having 1 to 16 carbon atoms, by way of example and with preference ethylamine, diethylamine, triethylamine, N,N-ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, tromethamine, dimethylaminoethanol, diethylaminoethanol, choline, procaine, dicyclohexylamine, dibenzylamine, N-methylmorpholine, N-methylpiperidine, arginine, lysine and 1,2-ethylenediamine.


In the context of the invention, solvates refer to those forms of the compounds according to the invention which, in the solid or liquid state, form a complex by coordination with solvent molecules. Hydrates are a specific form of solvates in which the coordination is with water. Preferred solvates in the context of the present invention are hydrates.


The compounds according to the invention may, depending on their structure, exist in different stereoisomeric forms, i.e. in the form of configurational isomers or else optionally as conformational isomers (enantiomers and/or diastereomers, including those in the case of atropisomers). The present invention therefore encompasses the enantiomers and diastereomers, and the respective mixtures thereof. The stereoisomerically homogeneous constituents can be isolated from such mixtures of enantiomers and/or diastereomers in a known manner; chromatography processes are preferably used for this purpose, especially HPLC chromatography on an achiral or chiral phase.


Where the compounds according to the invention can occur in tautomeric forms, the present invention encompasses all the tautomeric forms.


In particular, the 6-(trifluoromethyl)pyrimidin-4(3H)-one derivatives of the formula (I) according to the invention may also be present in the tautomeric pyrimidin-4(1H)-one form (I′) or 4-hydroxypyrimidine form (I″) (see Scheme 1 below); both tautomeric forms are expressly embraced by the present invention.




embedded image


The present invention also encompasses all suitable isotopic variants of the compounds according to the invention. An isotopic variant of a compound according to the invention is understood here to mean a compound in which at least one atom within the compound according to the invention has been exchanged for another atom of the same atomic number, but with a different atomic mass than the atomic mass which usually or predominantly occurs in nature. Examples of isotopes which can be incorporated into a compound according to the invention are those of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulphur, fluorine, chlorine, bromine and iodine, such as 2H (deuterium), 3H (tritium), 13C, 14C, 15N, 17O, 18O, 32P, 33P, 33S, 34S, 35S, 36S, 18F, 36Cl, 82Br, 123I, 124I, 129I and 131I. Particular isotopic variants of a compound according to the invention, especially those in which one or more radioactive isotopes have been incorporated, may be beneficial, for example, for the examination of the mechanism of action or of the active ingredient distribution in the body; due to comparatively easy preparability and detectability, especially compounds labelled with 3H or 14C isotopes are suitable for this purpose. In addition, the incorporation of isotopes, for example of deuterium, can lead to particular therapeutic benefits as a consequence of greater metabolic stability of the compound, for example to an extension of the half-life in the body or to a reduction in the active dose required; such modifications of the compounds according to the invention may therefore in some cases also constitute a preferred embodiment of the present invention. Isotopic variants of the compounds according to the invention can be prepared by generally customary processes known to those skilled in the art, for example by the methods described below and the procedures reported in the working examples, by using corresponding isotopic modifications of the particular reagents and/or starting compounds therein.


Moreover, the present invention also encompasses prodrugs of the compounds according to the invention. The term “prodrugs” refers here to compounds which may themselves be biologically active or inactive, but are converted while present in the body, for example by a metabolic or hydrolytic route, to compounds according to the invention.


In the context of the present invention, unless specified otherwise, the substituents are each defined as follows:


(C1-C4)-Alkyl and (C1-C3)-alkyl in the context of the invention represent a straight-chain or branched alkyl radical having 1 to 4 and 1 to 3 carbon atoms, respectively. Preferred examples include: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl and tert-butyl.


(C1-C4)-Alkylcarbonyl in the context of the invention represents a straight-chain or branched alkyl radical having 1 to 4 carbon atoms which is attached to the remainder of the molecule via a carbonyl group [—C(═O)—]. Preferred examples include: acetyl, propionyl, n-butyryl, isobutyryl, n-pentanoyl and pivaloyl.


(C1-C4)-Alkoxy in the context of the invention represents a straight-chain or branched alkoxy radical having 1 to 4 carbon atoms. Preferred examples include: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy and tert-butoxy.


(C1-C4)-Alkoxymethyl in the context of the invention represents a straight-chain or branched alkoxy radical having 1 to 4 carbon atoms which is attached to the remainder of the molecule via a methylene group [—CH2-] attached to the oxygen atom. Preferred examples include: methoxymethyl, ethoxymethyl, n-propoxymethyl, isopropoxymethyl, n-butoxymethyl and tert-butoxymethyl.


(C1-C4)-Alkoxycarbonyl in the context of the invention represents a straight-chain or branched alkoxy radical having 1 to 4 carbon atoms which is attached to the remainder of the molecule via a carbonyl group [—C(═O)-] attached to the oxygen atom. Preferred examples include: methoxycarbonyl, ethoxycarbonyl, n-propoxycarbonyl, isopropoxycarbonyl, n-butoxycarbonyl and tert-butoxycarbonyl.


(C1-C4)-Alkylsulphanyl [also referred to as (C1-C4)-alkylthio] in the context of the invention represents a straight-chain or branched alkyl radical having 1 to 4 carbon atoms which is attached to the remainder of the molecule via a sulphur atom. Preferred examples include: methylsulphanyl, ethylsulphanyl, n-propylsulphanyl, isopropylsulphanyl, n-butylsulphanyl, isobutylsulphanyl, sec-butylsulphanyl and tert-butylsulphanyl.


(C1-C4)-Alkylsulphinyl in the context of the invention represents a straight-chain or branched alkyl radical having 1 to 4 carbon atoms which is attached to the remainder of the molecule via a sulphinyl group [—S(═O)—]. Preferred examples include: methylsulphinyl, ethylsulphinyl, n-propylsulphinyl, isopropylsulphinyl, n-butylsulphinyl, isobutylsulphinyl, sec-butylsulphinyl and tert-butylsulphinyl.


(C1-C4)-Alkylsulphonyl in the context of the invention represents a straight-chain or branched alkyl radical having 1 to 4 carbon atoms which is attached to the remainder of the molecule via a sulphonyl group [—S(═O)2—]. Preferred examples include: methylsulphonyl, ethylsulphonyl, n-propylsulphonyl, isopropylsulphonyl, n-butylsulphonyl, isobutylsulphonyl, sec-butylsulphonyl and tert-butylsulphonyl.


Mono-(C1-C4)-alkylamino in the context of the invention represents an amino group having a straight-chain or branched alkyl substituent having 1 to 4 carbon atoms. Preferred examples include: methylamino, ethylamino, n-propylamino, isopropylamino, n-butylamino and tert-butylamino.


Di-(C1-C4)-alkylamino in the context of the invention represents an amino group having two identical or different straight-chain or branched alkyl substituents each having 1 to 4 carbon atoms. Preferred examples include: N,N-dimethylamino, N,N-diethylamino, N-ethyl-N-methylamino, N-methyl-N-n-propylamino, N-isopropyl-N-methylamino, N-isopropyl-N-n-propylamino, N,N-diisopropylamino, N-n-butyl-N-methylamino, N,N-di-n-butylamino and N-tert-butyl-N-methylamino.


(C1-C4)-Alkylcarbonylamino in the context of the invention represents an amino group having a straight-chain or branched alkylcarbonyl substituent which has 1 to 4 carbon atoms in the alkyl radical and is attached via the carbonyl group to the nitrogen atom. Preferred examples include: acetylamino, propionylamino, n-butyrylamino, isobutyrylamino, n-pentanoylamino and pivaloylamino.


5-membered heteroaryl in the context of the invention represents a monocyclic aromatic heterocycle (heteroaromatic) having a total of 5 ring atoms which contains up to three identical or different ring heteroatoms from the group consisting of N, O and S and is attached via a ring carbon atom or optionally a ring nitrogen atom. Examples include: furyl, pyrrolyl, thienyl, pyrazolyl, imidazolyl, 1,2-oxazolyl (isoxazolyl), 1,3-oxazolyl, 1,2-thiazolyl (isothiazolyl), 1,3-thiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,4-thiadiazolyl and 1,3,4-thiadiazolyl. Preference is given to 5-membered heteroaryl which contains one ring nitrogen atom (“aza-heteroaryl”) and may additionally contain one or two further ring heteroatoms from the group consisting of N, O and S, such as pyrrolyl, pyrazolyl, imidazolyl, 1,2-oxazolyl, 1,3-oxazolyl, 1,2-thiazolyl, 1,3-thiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,4-oxadiazolyl and 1,3,4-oxadiazolyl.


5- or 6-membered heterocyclyl in the context of the invention represents a monocyclic saturated or partially unsaturated (i.e. non-aromatic) heterocycle having a total of 5 or 6 ring atoms which contains up to three identical or different ring heteroatoms from the group consisting of N, O and S and is attached via a ring carbon atom or optionally a ring nitrogen atom. Examples include: pyrrolidinyl, dihydropyrrolyl, tetrahydrofuranyl, thiolanyl, pyrazolidinyl, dihydropyrazolyl, imidazolidinyl, dihydroimidazolyl, 1,2-oxazolidinyl, dihydro-1,2-oxazolyl, 1,3-oxazolidinyl, dihydro-1,3-oxazolyl, 1,2-thiazolidinyl, 1,3-thiazolidinyl, 1,3-oxathiolanyl, 1,3-oxathiolyl, dihydro-1,2,3-triazolyl, dihydro-1,2,4-triazolyl, dihydro-1,2,4-oxadiazolyl, dihydro-1,3,4-oxadiazolyl, dihydro-1,2,4-thiadiazolyl, dihydro-1,3,4-thiadiazolyl, piperidinyl, tetrahydropyridyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperazinyl, tetrahydropyrimidinyl, hexahydropyrimidinyl, morpholinyl, 1,2-oxazinanyl, 1,3-oxazinanyl, dihydro-1,3-oxazinyl, 1,3-oxazinyl, 1,3-dioxanyl, 1,4-dioxanyl and thiomorpholinyl. Preference is given to 5-membered saturated or partially unsaturated heterocyclyl which contains one ring nitrogen atom (“aza heterocyclyl”) and may additionally contain one or two further ring heteroatoms from the group consisting of N, O and S, such as pyrrolidinyl, dihydropyrrolyl, pyrazolidinyl, dihydropyrazolyl, imidazolidinyl, dihydroimidazolyl, 1,2-oxazolidinyl, dihydro-1,2-oxazolyl, 1,3-oxazolidinyl, dihydro-1,3-oxazolyl, 1,2-thiazolidinyl, 1,3-thiazolidinyl, dihydro-1,2,3-triazolyl, dihydro-1,2,4-triazolyl, dihydro-1,2,4-oxadiazolyl, dihydro-1,3,4-oxadiazolyl, dihydro-1,2,4-thiadiazolyl and dihydro-1,3,4-thiadiazolyl.


An oxo substituent in the context of the invention represents an oxygen atom attached via a double bond to a carbon or sulphur atom.


An imino substituent in the context of the invention represents an NH group attached via a double bond to a carbon or sulphur atom.


In the context of the present invention, all radicals which occur more than once are defined independently of one another. When radicals in the compounds according to the invention are substituted, the radicals may be mono- or polysubstituted, unless specified otherwise. Substitution by one or two identical or different substituents is preferred. Particular preference is given to substitution by one substituent.


In the context of the present invention, preference is given to compounds of the formula (I) in which

  • A represents C—H,
  • E represents CH2 or O,
  • R1 represents fluorine, chlorine, methyl or trifluoromethyl,
  • R2 represents hydrogen, fluorine, chlorine, methyl or trifluoromethyl,
  • L represents a bond, CH2 or NH,
  • and
  • Het represents pyridyl, pyrimidinyl or pyrazinyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, bromine, trifluoromethyl, methyl, phenyl, hydroxy, trifluoromethoxy, methoxy, methylsulphanyl, methylsulphinyl, methylsulphonyl, amino, methylamino, dimethylamino and acetylamino
    • or
    • represents 5-membered heteroaryl which contains one ring nitrogen atom and may additionally contain one or two further ring heteroatoms from the group consisting of N, O and S and which (i) may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, (C1-C3)-alkyl, cyclopropyl, phenyl, hydroxy, methoxy, methylsulphanyl, methylsulphinyl, methylsulphonyl, amino, methylamino, dimethylamino and acetylamino and which (ii) may be fused with a phenyl or pyridyl ring which for its part may be substituted by fluorine, chlorine, methyl, trifluoromethyl, methoxy, trifluoromethoxy, amino or acetylamino
    • or
    • represents 5-membered saturated or partially unsaturated heterocyclyl which contains one ring nitrogen atom and may additionally contain one or two further ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of methyl, hydroxy, oxo and amino,
  • and their salts, solvates and solvates of the salts.


In the context of the present invention, particular preference is given to compounds of the formula (I) in which

  • A represents C—H,
  • E represents CH2 or O,
  • R1 represents fluorine, chlorine, methyl or trifluoromethyl,
  • R2 represents fluorine or chlorine,
  • L represents a bond
  • and
  • Het represents pyridyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, bromine, trifluoromethyl, methyl, hydroxy, methoxy, methylsulphanyl, methylsulphinyl, methylsulphonyl and amino
    • or
    • represents pyrazolyl, imidazolyl, 1,2-oxazolyl, 1,2-thiazolyl, 1,2,4-triazolyl or 1,2,4-oxadiazolyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, methoxy, methylsulphanyl, methylsulphinyl, methylsulphonyl and amino,
    • or
    • represents 2-oxoimidazolidin-1-yl or 2-oxo-1,3-oxazolidin-3-yl,
  • and their salts, solvates and solvates of the salts.


In a particular embodiment, the present invention encompasses compounds of the formula (I) in which

  • A represents C—H, C—F or N,
  • E represents CH2, O or S,
  • R1 and R2 independently of one another represent hydrogen, fluorine, chlorine, methyl or trifluoromethyl,
    • where at least one of the two radicals R1 and R2 represents fluorine, chlorine, methyl or trifluoromethyl,
  • L represents a bond, CH2 or NH,
  • and
  • Het represents pyridyl or pyrimidinyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, (C1-C4)-alkyl, hydroxy, trifluoromethoxy, (C1-C4)-alkoxy and amino
    • or
    • represents 5-membered heteroaryl which contains one, two or three identical or different ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, (C1-C4)-alkyl, cyclopropyl, hydroxy, (C1-C4)-alkoxy, (C1-C4)-alkoxymethyl, amino, hydroxycarbonyl and (C1-C4)-alkoxycarbonyl
    • or
    • represents 5- or 6-membered saturated or partially unsaturated heterocyclyl which contains one, two or three identical or different ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of (C1-C4)-alkyl, hydroxy, oxo, amino and imino,
  • and their salts, solvates and solvates of the salts.


In a further embodiment, the present invention encompasses compounds of the formula (I) in which

  • A represents C—H,
  • E represents CH2 or O,
  • R1 represents fluorine, chlorine, methyl or trifluoromethyl,
  • R2 represents hydrogen, fluorine, chlorine, methyl or trifluoromethyl,
  • L represents a bond, CH2 or NH,
  • and
  • Het represents pyridyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, trifluoromethoxy, methoxy and amino,
    • or
    • represents 5-membered heteroaryl which contains one ring nitrogen atom and may additionally contain one or two further ring heteroatoms selected from the group consisting of N and O and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, cyclopropyl, hydroxy, methoxy and amino
    • or
    • represents 5-membered saturated or partially unsaturated heterocyclyl which contains one ring nitrogen atom and may additionally contain one or two further ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of methyl, hydroxy, oxo and amino,
  • and their salts, solvates and solvates of the salts.


In a further embodiment, the present invention encompasses compounds of the formula (I) in which

  • A represents C—H,
  • E represents CH2 or O,
  • R1 represents fluorine, chlorine or trifluoromethyl,
  • R2 represents fluorine or chlorine,
  • L represents a bond
  • and
  • Het represents pyridyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, methoxy and amino
    • or
    • represents pyrazolyl, imidazolyl, 1,2,4-triazolyl or 1,2,4-oxadiazolyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, methoxy and amino,
    • or
    • represents 2-oxoimidazolidin-1-yl or 2-oxo-1,3-oxazolidin-3-yl,
  • and their salts, solvates and solvates of the salts.


A particular embodiment of the present invention relates to compounds of the formula (I) in which

  • A represents C—H,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • E represents CH2,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • E represents O,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • R1 and R2 each represent chlorine,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • R1 represents trifluoromethyl
  • and
  • R2 represents chlorine,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • R1 represents methyl
  • and
  • R2 represents chlorine,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • L represents a bond,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • Het represents pyridyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, trifluoromethoxy, methoxy and amino,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • Het represents pyrazolyl, 1,2-oxazolyl, imidazolyl, 1,2,4-triazolyl or 1,2,4-oxadiazolyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, methoxy and amino,
  • and their salts, solvates and solvates of the salts.


A further particular embodiment of the present invention relates to compounds of the formula (I) in which

  • Het represents pyridyl, pyrazolyl, imidazolyl or 1,2,4-triazolyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of chlorine, methyl, hydroxy, methoxy, methylsulphanyl and amino,
  • and their salts, solvates and solvates of the salts.


The individual radical definitions specified in the particular combinations or preferred combinations of radicals are, independently of the particular combinations of the radicals specified, also replaced as desired by radical definitions of other combinations.


Very particular preference is given to combinations of two or more of the preferred ranges mentioned above.


The invention further provides a process for preparing the compounds according to the invention of the formula (I), characterized in that


[A] a Compound of the Formula (II)



embedded image




    • in which A, R1 and R2 have the meanings given above,

    • E1 represents CH2 or O

    • and

    • T1 represents methyl, ethyl, n-propyl or n-butyl,

    • is condensed with a compound of the formula (III)







embedded image




    • in which Het and L have the meanings given above

    • or a salt thereof to give a compound of the formula (I-A) according to the invention







embedded image




    • in which A, E1, Het, L, R1 and R2 have the meanings given above


      or


      [B] a compound of the formula (IV)







embedded image




    • in which A, R1 and R2 have the meanings given above

    • and

    • E2 represents O or S

    • is reacted in the form of an alkali metal salt or in the presence of a base with a compound of the formula (V)







embedded image




    • in which Het and L have the meanings given above

    • to give a compound of the formula (I-B) according to the invention







embedded image




    • in which A, E2, Het, L, R1 and R2 have the meanings given above


      and the resulting compounds of the formulae (I-A) and (I-B) are optionally converted with the appropriate (i) solvents and/or (ii) acids or bases into their solvates, salts and/or solvates of the salts.





Suitable inert solvents for the process step (II)+(III)-(I-A) are, for example, alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, diisopropyl ether, methyl tert-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis-(2-methoxyethyl) ether, hydrocarbons or chlorinated hydrocarbons such as benzene, toluene, xylene or chlorobenzene, or dipolar aprotic solvents such as acetonitrile, butyronitrile, N,N-dimethylformamide (DMF), N,N-dimethylacetamide (DMA), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU) or N-methylpyrrolidinone (NMP). It is also possible to use mixtures of these solvents. Preference is given to using methanol, ethanol, 1,4-dioxane or N,N-dimethylformamide.


The compound of the formula (III) is preferably employed in the form of a salt, for example as hydrochloride, where in this case the reaction is carried out in the presence of an auxiliary base.


Bases suitable for this purpose are in particular alkali metal hydroxides such as lithium hydroxide, sodium hydroxide or potassium hydroxide, alkali metal bicarbonates such as sodium bicarbonate or potassium bicarbonate, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or sodium tert-butoxide or potassium tert-butoxide, or customary tertiary amine bases such as triethylamine, N-methylmorpholine, N-methylpiperidine, N,N-diisopropylethylamine, pyridine or 4-N,N-dimethylaminopyridine. The base used is preferably potassium carbonate, sodium methoxide or N,N-diisopropylethylamine.


The reaction (II)+(III)→(I-A) is generally carried out in a temperature range of from +20° C. to +150° C., preferably at from +60° C. to +120° C.


The process step (IV)+(V)→(I-B) is generally carried out in a temperature range of from +80° C. to +150° C. in a corresponding high-boiling inert solvent such as ethylene glycol, bis(2-methoxyethyl) ether, N,N-dimethylformamide (DMF), N,N-dimethylacetamide (DMA), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU) or N-methylpyrrolidinone (NMP). Preference is given to using ethylene glycol.


Suitable bases for this reaction are in particular alkali metal hydroxides such as lithium hydroxide, sodium hydroxide or potassium hydroxide, alkali metal carbonates such as lithium carbonate, sodium carbonate, potassium carbonate or caesium carbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or sodium tert-butoxide or potassium tert-butoxide, or alkali metal hydrides such as sodium hydride or potassium hydride. Preference is given to using caesium carbonate.


The process steps described above can be carried out at atmospheric, elevated or reduced pressure (for example in the range from 0.5 to 5 bar); in general, the reactions are each carried out at atmospheric pressure.


For their part, the compounds of the formula (II) can be prepared by


[A-1] Alkylating a Trifluoroacetoacetic Ester of the Formula (VI)



embedded image




    • in which T1 has the meaning given above

    • in the presence of a base with a compound of the formula (VII)







embedded image




    • in which A, R1 and R2 have the meanings given above

    • and

    • X represents a leaving group, for example chlorine, bromine, iodine, mesylate, triflate or tosylate,

    • to give a compound of the formula (II-A)







embedded image




    • in which A, T1, R1 and R2 have the meanings given above


      or by





[A-2] Acylating an Aryloxyacetic Ester of the Formula (VIII)



embedded image




    • in which A, T1, R1 and R2 have the meanings given above

    • in the presence of a base with a trifluoroacetic ester of the formula (IX)







embedded image




    • in which

    • T2 represents methyl or ethyl

    • to give a compound of the formula (II-B)







embedded image




    • in which A, T1, R1 and R2 have the meanings given above.





Inert solvents for the process step (VI)+(VII)→(II-A) are, for example, ethers such as diethyl ether, diisopropyl ether, methyl tert-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis(2-methoxyethyl) ether, or dipolar aprotic solvents such as acetone, methyl ethyl ketone, ethyl acetate, acetonitrile, butyronitrile, N,N-dimethylformamide (DMF), N,N-dimethylacetamide (DMA), dimethyl sulphoxide (DMSO), N-methylpyrrolidinone (NMP) or N,N′-dimethylpropyleneurea (DMPU). It is also possible to use mixtures of such solvents. Preference is given to using tetrahydrofuran.


Suitable bases for this reaction are in particular alkali metal carbonates such as sodium carbonate, potassium carbonate or caesium carbonate, alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or sodium tert-butoxide or potassium tert-butoxide, alkali metal hydrides such as sodium hydride or potassium hydride, amides such as lithium bis(trimethylsilyl)amide or potassium bis(trimethylsilyl)amide or lithium diisopropylamide, or tertiary amine bases such as triethylamine, N-methylmorpholine, N-methylpiperidine, N,N-diisopropylethylamine, pyridine or 4-N,N-dimethylaminopyridine. The base used is preferably N,N-diisopropylethylamine.


The reaction (VI)+(VII)→(II-A) is generally carried out in a temperature range of from 0° C. to +150° C., preferably from +20° C. to +100° C. Addition of an alkylation catalyst such as lithium chloride or lithium bromide, sodium iodide or potassium iodide, tetra-n-butylammonium bromide or benzyltriethylammonium chloride may optionally be advantageous. Suitable inert solvents for the process step (VIII)+(IX)→(II-B) are, for example, alcohols such as methanol, ethanol, n-propanol, isopropanol, n-butanol or tert-butanol, ethers such as diethyl ether, diisopropyl ether, methyl tert-butyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane or bis-(2-methoxyethyl) ether, hydrocarbons or chlorinated hydrocarbons such as benzene, toluene, xylene or chlorobenzene, or dipolar aprotic solvents such as acetonitrile, butyronitrile, N,N-dimethylformamide (DMF), N,N-dimethylacetamide (DMA), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU) or N-methylpyrrolidinone (NMP). It is also possible to use mixtures of such solvents. Here, preference is given to using toluene.


Preferred bases for this reaction are alkali metal alkoxides such as sodium methoxide or potassium methoxide, sodium ethoxide or potassium ethoxide or sodium or potassium tert-butoxide, alkali metal hydrides such as sodium hydride or potassium hydride, or amides such as lithium bis(trimethylsilyl)amide or potassium bis(trimethylsilyl)amide or lithium diisopropylamide. Preference is given to using sodium hydride.


The reaction (VIII)+(IX)→(II-B) is generally carried out in a temperature range of from 0° C. to +120° C.


The compounds of the formula (V) can be prepared by condensing, analogously to process [A], a trifluoroacetoacetic ester of the formula (VI)




embedded image


in which T1 has the meaning given above


with a compound of the formula (III)




embedded image


in which Het and L have the meanings given above


or a salt thereof to give a compound of the formula (X)




embedded image


in which Het and L have the meanings given above


and then brominating the latter to give the compound of the formula (V).


The condensation reaction (VI)+(III)→(X) is carried out in a manner analogous to the reaction (II)+(III)→(I-A) described above in process [A]. Subsequent bromination of (X) to the compound (V) is preferably carried out with the aid of elemental bromine, N-bromosuccinimide (NBS) or 1,3-dibromo-5,5-dimethylhydantoin in an inert solvent such as dichloromethane, chloroform, tetrahydrofuran, acetonitrile, N,N-dimethylformamide (DMF) or acetic acid, within a temperature range of from −78° C. to +50° C.


Compounds of the Formula (I-C) According to the Invention




embedded image


in which A, E1, Het, R1 and R2 have the meanings given above


can alternatively also be prepared by condensing a compound of the formula (II)




embedded image


in which A, E1, R1, R2 and T1 have the meanings given above


initially analogously to process [A] with S-methylisothiourea or a salt thereof to give a compound of the formula (XI)




embedded image


in which A, E1, R1 and R2 have the meanings given above,


then oxidizing to give the compound of the formula (XII)




embedded image


in which A, E1, R1 and R2 have the meanings given above


and


n represents the number 1 or 2


and then reacting this compound, optionally in the presence of a base, with a compound of the formula (XIII)




embedded image


in which Het has the meaning given above.


In an analogous manner, starting with a compound of the formula (XIV)




embedded image


in which

  • Het*, within the above scope of the meaning of Het, represents a 5-membered heteroaryl or 5- or 6-membered heterocyclyl ring containing an NH grouping, as shown, by reaction with the compound (XII) it is also possible to obtain compounds of the formula (I-D)




embedded image


  • in which A, E1, R1 and R2 have the meanings given above

  • and

  • Het*, within the above scope of the meaning of Het, represents a 5-membered heteroaryl or 5- or 6-membered heterocyclyl ring attached via a ring nitrogen atom, as shown.



The conversion of compound (II) with S-methylisothiourea or a salt thereof into the compound (XI) is carried out under conditions analogous to those described above in process [A] for the reaction (II)+(III)→(I-A). The subsequent oxidation to the sulphoxide [n=1] or sulphone [n=2] of the formula (XII) is carried out by customary methods using appropriate amounts of a peroxide or a peracid such as hydrogen peroxide, potassium permanganate, potassium monopersulphate, peracetic acid or meta-chloroperbenzoic acid.


The reaction (XII)+(XIII)→(I-C) or (XII)+(XIV)→(I-D) is generally carried out in a temperature range of from +100° C. to +200° C. in a high-boiling inert solvent such as toluene, N,N-dimethylformamide (DMF), N,N-dimethylacetamide (DMA), dimethyl sulphoxide (DMSO), N,N′-dimethylpropyleneurea (DMPU) or N-methylpyrrolidinone (NMP). It may be advantageous to carry out the reaction in the presence of a base such as potassium carbonate, sodium tert-butoxide or potassium tert-butoxide or sodium hydride. If a plurality of possible reaction centres are present in (XIII) or (XIV), the presence or absence of such a base may optionally have a favourable effect on the chemoselectivity of the reaction [cf. Reaction Scheme 4 below].


Compounds of the formula (I) according to the invention may also be obtained by initially preparing, analogously to the processes described above, compounds of the formula (XV)




embedded image


  • in which A, E, R1 and R2 have the meanings given above

  • and

  • Z generally represents a functional group which allows the construction of the L-Het grouping defined above by subsequent chemical transformation

  • and then converting these compounds by methods known from the literature in the couse of a de novo heteroaryl or heterocyclyl synthesis into compounds of the formula (I).



Examples of functional groups Z in formula (XV) suitable for such purposes are in particular amines [—NH2], nitriles [—CN, —CH2—CN], carboxylic esters, carboxamides, carboxamidines, carboxamidoximes and carbohydrazides [—CH2—C(═O)—OCH3, —CH2—C(═O)—NH2, —CH2—C(═NH)—NH2, —C(═N—OH)—NH2, —CH2—C(═N—OH)—NH2, —CH2—C(═O)—NH—NH2] and also aldehydes and their derivatives such as acetals and oximes [—CH═O, —CH(OCH3)2, —CH(OC2H5)2, —CH═N—OH]. Further conversions of these functional groups in order to construct the respective L-Het grouping, as defined above, are carried out by known methods familiar to the person skilled in the art and include in particular acylation reactions with activated carbonic acid and carboxylic acid derivatives and subsequent condensation and ring closure reactions (“heterocyclizations”) [see also the Reaction Schemes 5-7 shown below and the preparation, described in detail in the Experimental Part, of intermediates and working examples].


The compounds of the formulae (III), (IV), (VI), (VII), (VIII), (IX), (XIII) and (XIV) are either commercially available or described as such in the literature, or they can be prepared from other commercially available compounds by customary methods known from the literature. Numerous detailed procedures and further literature references can also be found in the Experimental Part, in the section on the preparation of the starting compounds and intermediates.


The preparation of the compounds according to the invention can be illustrated in an exemplary manner by the Reaction Schemes 2-7 below:




embedded image




embedded image




embedded image




embedded image




embedded image




embedded image


The compounds according to the invention have valuable pharmacological properties and can be used for prevention and treatment of diseases in humans and animals.


The compounds according to the invention are potent antagonists of the CCR2 receptor and are therefore particularly suitable for the treatment and/or prevention of disorders, in particular cardiovascular, renal, inflammatory, allergic and/or fibrotic disorders.


In the context of the present invention, cardiovascular disorders are understood to mean, for example, the following disorders: acute and chronic heart failure, arterial hypertension, coronary heart disease, acute coronary syndrome, myocardial infarction (STEMI, NSTEMI), acute myocardial infarction, stable and unstable angina pectoris, myocardial ischaemia, autoimmune heart disorders (pericarditis, endocarditis, valvolitis, aortitis, cardiomyopathies), shock, atherosclerosis, cardiac hypertrophy, cardiac fibrosis, atrial and ventricular arrhythmias, transitory and ischaemic attacks, stroke, pre-eclampsia, inflammatory cardiovascular disorders, peripheral and cardiac vascular disorders, peripheral perfusion disorders, arterial pulmonary hypertension, spasms of the coronary arteries and peripheral arteries, arterial and venous thromboses, thromboembolic disorders, oedema development, for example pulmonary oedema, cerebral oedema, renal oedema or heart failure-related oedema, restenoses, for example after thrombolysis treatments, percutaneous transluminal angioplasty (PTA), transluminal coronary angioplasty (PTCA), heart transplants and bypass operations, micro- and macrovascular damage (vasculitis), reperfusion damage, microalbuminuria, myocardial insufficiency, endothelial dysfunction, and also for the reduction in size of the myocardial region affected by myocardial infarction, and for the prevention of secondary infarctions.


In the context of the present invention, the term “heart failure” encompasses both acute and chronic forms of heart failure, and also more specific or related disease types thereof, such as acute decompensated heart failure, right heart failure, left heart failure, global failure, ischaemic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, idiopathic cardiomyopathy, congenital heart defects, heart valve defects, heart failure associated with heart valve defects, mitral valve stenosis, mitral valve insufficiency, aortic valve stenosis, aortic valve insufficiency, tricuspid valve stenosis, tricuspid valve insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined heart valve defects, myocardial inflammation (myocarditis), chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic cardiomyopathy, cardiac storage disorders, diastolic heart failure, systolic heart failure, and acute phases of worsening of existing heart failure (worsening heart failure).


In addition, the compounds according to the invention are suitable for treatment and/or prevention of renal disorders, especially of acute and chronic renal insufficiency, and of acute and chronic kidney failure.


In the context of the present invention, the term “acute renal insufficiency” encompasses acute manifestations of kidney disease, of kidney failure and/or renal insufficiency with and without the need for dialysis, and also underlying or related renal disorders such as renal hypoperfusion, ischaemic kidney disorders (AKI), intradialytic hypotension, volume deficiency (e.g. owing to dehydration or blood loss), shock, acute glomerulonephritis, haemolytic-uraemic syndrome (HUS), vascular catastrophe (arterial or venous thrombosis or embolism), cholesterol embolism, acute Bence-Jones kidney in the event of plasmacytoma, acute supravesicular or subvesicular efflux obstructions, immunological renal disorders such as kidney transplant rejection and immune complex-induced renal disorders, tubular dilatation, hyperphosphataemia, furthermore acute renal disorders characterized by the need for dialysis, including in the case of partial resections of the kidney, dehydration through forced diuresis, uncontrolled blood pressure rise with malignant hypertension, urinary tract obstruction, urinary tract infection and amyloidosis, moreover systemic disorders with glomerular factors, such as rheumatological-immunological systemic disorders (e.g. lupus erythematodes), renal artery thrombosis, renal vein thrombosis, analgesic nephropathy and renal tubular acidosis, and X-ray contrast agent- or medicament-induced acute interstitial renal disorders.


In the context of the present invention, the term “chronic renal insufficiency” (CKD) encompasses chronic manifestations of kidney disease, of kidney failure and/or renal insufficiency with and without the need for dialysis, and also underlying or related renal disorders such as renal hypoperfusion, intradialytic hypotension, obstructive uropathy, glomerulopathy, glomerular and tubular proteinuria, renal oedema, haematuria, primary, secondary and chronic glomerulonephritis, membranous and membranoproliferative glomerulonephritis, Alport syndrome, glomerulosclerosis, tubulointerstitial disorders, nephropathic disorders such as primary and congenital kidney disease, renal inflammation, immunological renal disorders such as kidney transplant rejection, immune complex-induced renal disorders, 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 elevated blood concentrations of urea, nitrogen, potassium and/or creatinine, altered activity of renal enzymes, for example glutamyl synthetase, altered urine osmolarity or urine volume, elevated microalbuminuria, macroalbuminuria, glomerular and arteriolar lesions, tubular dilatation, hyperphosphataemia and/or the need for dialysis, and chronic renal disorders in the event of renal cell carcinoma, after partial resections of the kidney, in cases of dehydration through forced diuresis, uncontrolled blood pressure rise with malignant hypertension, urinary tract obstruction, urinary tract infection and amyloidosis, furthermore systemic disorders with glomerular factors, such as rheumatological-immunological systemic disorders (e.g. lupus erythematodes), renal artery stenosis, renal artery thrombosis, renal vein thrombosis, analgesic nephropathy, renal tubular acidosis, X-ray contrast agent- or medicament-induced chronic interstitial renal disorders and also in metabolic syndrome.


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 oedema, heart failure, uraemia, anaemia, electrolyte disturbances (for example hyperkalaemia, hyponatraemia) and disturbances in bone and carbohydrate metabolism.


The compounds according to the invention are further suitable for the treatment and/or prevention of polycystic kidney disease (PCKD) and of the syndrome of inappropriate ADH secretion (SIADH).


In addition, the compounds according to the invention are also suitable for treatment and/or prevention of pulmonary arterial hypertension (PAH) and other forms of pulmonary hypertension (PH), of chronic obstructive pulmonary disease (COPD), of acute respiratory distress syndrome (ARDS), of acute lung injury (ALI), pulmonary fibrosis, pulmonary emphysema (for example pulmonary emphysema caused by cigarette smoke), cystic fibrosis (CF), cardiogenic shock, aneurysms, sepsis (SIRS), multiple organ failure (MODS, MOF), inflammatory disorders of the kidney, chronic intestinal disorders (IBD, Crohn's Disease, ulcerative colitis), pancreatitis, peritonitis, rheumatoid disorders, inflammatory skin disorders and inflammatory eye disorders.


The compounds according to the invention can additionally be used for treatment and/or prevention of asthmatic disorders of varying severity with intermittent or persistent characteristics (refractive asthma, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, medicament- or dust-induced asthma), of various forms of bronchitis (chronic bronchitis, infectious bronchitis, eosinophilic bronchitis), of Bronchiolitis obliterans, bronchiectasis, pneumonia, idiopathic interstitial pneumonia, farmer's lung and related disorders, of coughs and colds (chronic inflammatory cough, iatrogenic cough), inflammation of the nasal mucosa (including medicament-related rhinitis, vasomotoric rhinitis and seasonal allergic rhinitis, for example hay fever) and of polyps.


Furthermore, the compounds according to the invention are suitable for treatment and/or prevention of fibrotic disorders of the internal organs, for example the lung, the heart, the kidney, the bone marrow and in particular the liver, and also dermatological fibroses and fibrotic eye disorders. In the context of the present invention, the term “fibrotic disorders” encompasses particularly the following disorders: hepatic fibrosis, cirrhosis of the liver, pulmonary fibrosis, endomyocardial fibrosis, cardiomyopathy, nephropathy, glomerulonephritis, interstitial renal fibrosis, fibrotic damage resulting from diabetes, bone marrow fibrosis, peritoneal fibrosis and similar fibrotic disorders, scleroderma, amyotrophic lateral sclerosis (ALS), morphoea, keloids, hypertrophic scarring (also following surgical procedures), diabetic retinopathy and proliferative vitroretinopathy.


The compounds according to the invention can also be used for the treatment and/or prevention of metabolic disorders such as obesity and Type 2 diabetes, which are also accompanied by chronic inflammation, furthermore for the treatment and/or prevention of neurodegenerative disorders including Alzheimer's disease, multiple sclerosis and ischaemic brain damage, and also for pain, in particular neuropathic pain.


In addition, the compounds according to the invention can also be used for treatment and/or prevention of cancers (skin cancer, brain tumours, breast cancer, bone marrow tumours, leukaemias, liposarcomas, carcinoma of the gastrointestinal tract, of the liver, pancreas, lung, kidney, urinary tract, prostate and genital tract, and also malignant tumours in the lymphoproliferative system, for example Hodgkin's and non-Hodgkin's lymphoma), of disorders of the gastrointestinal tract and of the abdomen (glossitis, gingivitis, periodontitis, oesophagitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease, colitis, proctitis, pruritus ani, diarrhoea, coeliac disease, hepatitis, chronic hepatitis, hepatic fibrosis, cirrhosis of the liver, pancreatitis and cholecystitis), of skin disorders (allergic skin disorders, psoriasis, acne, eczema, neurodermitis, various forms of dermatitis, and also keratitis, bullosis, vasculitis, cellulitis, panniculitis, lupus erythematodes, erythema, lymphoma, skin cancer), of disorders of the skeletal bone and of the joints, and also of the skeletal muscle (various forms of arthritis and of arthropathies), and of further disorders with an inflammatory or immunological component, for example paraneoplastic syndrome, in the event of rejection reactions after organ transplants and for wound healing and angiogenesis, especially in the case of impaired wound healing and chronic wounds, for example diabetic foot ulcers and chronic venous leg ulcers.


The compounds according to the invention are additionally suitable for treatment and/or prevention of ophthalmologic disorders, for example glaucoma, age-related macular degeneration (AMD), of dry (non-exudative) AMD, wet (exudative, neovascular) AMD, choroidal neovascularization (CNV), diabetic retinopathy, atrophic changes to the retinal pigment epithelium (RPE), hypertrophic changes to the retinal pigment epithelium, macular oedema, diabetic macular oedema, retinal vein occlusion, choroidal retinal vein occlusion, macular oedema due to retinal vein occlusion, angiogenesis at the front of the eye, for example corneal angiogenesis, for example following keratitis, cornea transplant or keratoplasty, corneal angiogenesis due to hypoxia (as a result of extensive wearing of contact lenses), pterygium conjunctiva, subretinal oedema and intraretinal oedema. The compounds according to the invention are furthermore suitable for the treatment and/or prevention of elevated and high intraocular pressure as a result of traumatic hyphaema, periorbital oedema, postoperative viscoelastic retention or intraocular inflammation.


By virtue of their property profile, the compounds according to the invention are suitable in particular for the treatment and/or prevention of acute coronary syndrome, myocardial infarction, acute and chronic heart failure, acute and chronic kidney failure and acute lung damage.


The above-mentioned, well-characterized diseases in humans can also occur with a comparable aetiology in other mammals and can likewise be treated there with the compounds of the present invention.


In the context of the present invention, the term “treatment” or “treating” includes inhibition, retardation, checking, alleviating, attenuating, restricting, reducing, suppressing, repelling or healing of a disease, a condition, a disorder, an injury or a health problem, or the development, the course or the progression of such states and/or the symptoms of such states. The term “therapy” is understood here to be synonymous with the term “treatment”.


The terms “prevention”, “prophylaxis” or “preclusion” are used synonymously in the context of the present invention and refer to the avoidance or reduction of the risk of contracting, experiencing, suffering from or having a disease, a condition, a disorder, an injury or a health problem, or a development or progression of such states and/or the symptoms of such states.


The treatment or prevention of a disease, a condition, a disorder, an injury or a health problem may be partial or complete.


The present invention thus further provides for the use of the compounds according to the invention for the treatment and/or prevention of disorders, in particular the disorders mentioned above.


The present invention further provides for the use of the compounds according to the invention for producing a medicament for the treatment and/or prevention of disorders, in particular the disorders mentioned above.


The present invention further provides a medicament comprising at least one of the compounds according to the invention, for the treatment and/or prevention of disorders, in particular the disorders mentioned above.


The present invention furthermore provides for the use of the compounds according to the invention in a method for treatment and/or prevention of disorders, in particular the disorders mentioned above.


The present invention further provides a method for treatment and/or prevention of disorders, in particular the disorders mentioned above, using an effective amount of at least one of the compounds according to the invention.


The compounds according to the invention can be employed by themselves or, if required, in combination with one or more other pharmacologically active substances, as long as this combination does not lead to undesirable and unacceptable side effects. The present invention furthermore therefore provides medicaments containing at least one of the compounds according to the invention and one or more further active compounds, in particular for treatment and/or prevention of the abovementioned disorders. Preferred examples of active compounds suitable for combinations include:

    • compounds which inhibit the signal transduction cascade, by way of example and with preference from the group of the kinase inhibitors, especially from the group of the tyrosine kinase and/or serine/threonine kinase inhibitors;
    • compounds which inhibit the degradation and alteration of the extracellular matrix, by way of example and with preference inhibitors of the matrix metalloproteases (MMPs), especially inhibitors of stromelysin, collagenases, gelatinases and aggrecanases (in this context particularly of MMP-1, MMP-3, MMP-8, MMP-9, MMP-10, MMP-11 and MMP-13) and of metalloelastase (MMP-12);
    • compounds which block the binding of serotonin to its receptors, by way of example and with preference antagonists of the 5-HT2B receptor such as PRX-08066;
    • organic nitrates and NO donors, for example sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhaled NO;
    • NO-independent but haem-dependent stimulators of guanylate cyclase, such as especially riociguat and 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;
    • NO— and haem-independent activators of soluble guanylate cyclase, such as especially the compounds described in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 and WO 02/070510;
    • compounds which inhibit the degradation of cyclic guanosine monophosphate (cGMP) and/or cyclic adenosine monophosphate (cAMP), for example inhibitors of phosphodiesterases (PDE) 1, 2, 3, 4 and/or 5, especially PDE 5 inhibitors such as sildenafil, vardenafil, tadalafil, udenafil, dasantafil, avanafil, mirodenafil or lodenafil;
    • prostacyclin analogues and IP receptor agonists, by way of example and with preference iloprost, beraprost, treprostinil, epoprostenol or NS-304;
    • bronchodilatory agents, by way of example and with preference from the group of the beta-adrenergic receptor agonists, such as especially albuterol, isoproterenol, metaproterenol, terbutalin, fenoterol, formoterol, reproterol, salbutamol or salmeterol, and from the group of the anticholinergics, such as especially ipratropium bromide, tiotropium bromide or oxitropium bromide;
    • anti-inflammatory agents, by way of example and with preference from the group of the glucocorticoids, such as especially prednisone, prednisolone, methylprednisolone, triamcinolone, dexamethasone, beclomethasone, betamethasone, flunisolide, budesonide or fluticasone;
    • compounds which inhibit soluble epoxide hydrolase (sEH), for example N,N′-dicyclohexylurea, 12-(3-adamantan-1-ylureido)dodecanoic acid or 1-adamantan-1-yl-3-{5-[2-(2-ethoxyethoxy)ethoxy]pentyl}urea;
    • compounds which influence the energy metabolism of the heart, by way of example and with preference etomoxir, dichloroacetate, ranolazine or trimetazidine;
    • vasopressin receptor antagonists, for example and with preference conivaptan, tolvaptan, lixivaptan, mozavaptan, satavaptan, SR-121463, RWJ-676070 or BAY 86-8050;
    • antihyperglycaemic agents (antidiabetics), by way of example and with preference from the group of the biguanides such as metformin, of the sulphonylureas, such as glibenclamide or glimepiride, of the glinides, such as repaglinide or nateglinide, of the DPP IV inhibitors, such as sitagliptin, vildagliptin or saxagliptin, of the glucosidase inhibitors, such as acarbose or miglitol, and of the amyline analogues, such as pramlintide;
    • hypotensive active ingredients, for example and with preference from the group of calcium antagonists, angiotensin AII antagonists, ACE inhibitors, vasopeptidase inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, and rho kinase inhibitors and the diuretics;
    • agents having antithrombotic activity, for example and with preference from the group of the platelet aggregation inhibitors, the anticoagulants and the profibrinolytic substances; and/or
    • active compounds altering lipid metabolism, for example and with preference from the group of the thyroid receptor agonists, cholesterol synthesis inhibitors such as, by way of example and preferably, HMG-CoA reductase inhibitors or squalene synthesis inhibitors, the ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid adsorbents, bile acid reabsorption inhibitors and lipoprotein(a) antagonists.


In a preferred embodiment of the invention, the compounds according to the invention are employed in combination with a kinase inhibitor, by way of example and with preference nintedanib, dasatinib, nilotinib, bosutinib, regorafenib, sorafenib, sunitinib, cediranib, axitinib, telatinib, imatinib, brivanib, pazopanib, vatalanib, gefitinib, erlotinib, lapatinib, canertinib, lestaurtinib, lonafarnib, pelitinib, semaxanib, tandutinib or tipifarnib.


Hypotensive agents are preferably understood to mean compounds from the group of calcium antagonists, angiotensin AII antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, rho kinase inhibitors, and the diuretics.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a calcium antagonist, by way of example and with preference 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, by way of example and with preference prazosin.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a beta-receptor blocker, by way of example and with preference propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipranolol, nadolol, mepindolol, carazalol, sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, carteolol, esmolol, labetalol, carvedilol, adaprolol, landiolol, nebivolol, epanolol or bucindolol.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an angiotensin AII antagonist, by way of example and with preference losartan, candesartan, valsartan, telmisartan or embusartan.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an ACE inhibitor, by way of example and with preference enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an endothelin antagonist, by way of example and with preference bosentan, darusentan, ambrisentan or sitaxsentan.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a renin inhibitor, by way of example and with preference 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, by way of example and with preference spironolactone or eplerenone.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a rho kinase inhibitor, by way of example and with preference fasudil, Y-27632, SLx-2119, BF-66851, BF-66852, BF-66853, KI-23095, SB-772077, GSK-269962A or BA-1049.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a diuretic, preferred examples being furosemide, bumetanide, torsemide, bendroflumethiazide, chlorthiazide, hydrochlorthiazide, hydroflumethiazide, methyclothiazide, polythiazide, trichlormethiazide, chlorthalidone, indapamide, metolazone, quinethazone, acetazolamide, dichlorophenamide, methazolamide, glycerol, isosorbide, mannitol, amiloride or triamterene.


Antithrombotic agents are preferably understood to mean compounds from the group of the platelet aggregation inhibitors, the anticoagulants and the profibrinolytic substances.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a platelet aggregation inhibitor, by way of example and with preference aspirin, clopidogrel, ticlopidin or dipyridamole.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a thrombin inhibitor, by way of example and with preference ximelagatran, melagatran, dabigatran, bivalirudin or clexane.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a GPIIb/IIIa antagonist such as, by way of example and with preference, tirofiban or abciximab.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a factor Xa inhibitor, by way of example and with preference rivaroxaban, apixaban, edoxaban, razaxaban, fondaparinux, idraparinux, DU-176b, PMD-3112, YM-150, KFA-1982, EMD-503982, MCM-17, MLN-1021, DPC 906, JTV 803, SSR-126512 or SSR-128428.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with heparin or with 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, by way of example and with preference coumarin.


Agents which modify lipid metabolism are preferably understood to mean compounds from the group of CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA reductase inhibitors or squalene synthesis inhibitors, of ACAT inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, polymeric bile acid adsorbents, 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, by way of example and with preference torcetrapib (CP-529 414), JJT-705 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, by way of example and with preference D-thyroxin, 3,5,3′-triiodothyronin (T3), CGS 23425 or axitirome (CGS 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, by way of example and with preference lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rosuvastatin or pitavastatin.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a squalene synthesis inhibitor, by way of example and with preference 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, by way of example and with preference 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, by way of example and with preference implitapide, BMS-201038, R-103757 or JTT-130.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a PPAR-gamma agonist, by way of example and with preference pioglitazone or rosiglitazone.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a PPAR-delta agonist, by way of example and with preference 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, by way of example and with preference 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, by way of example and with preference orlistat.


In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a polymeric bile acid adsorbent, by way of example and with preference 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, by way of example and with preference ASBT (=IBAT) inhibitors, for example 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, by way of example and with preference gemcabene calcium (CI-1027) or nicotinic acid.


Particular preference is given to combinations of the compounds according to the invention with one or more further active compounds selected from the group of the antihyperglycaemic agents (antidiabetics), the hypotensive agents, the platelet aggregation inhibitors, the anticoagulants and the HMG-CoA reductase inhibitors (statins).


The present invention further provides medicaments which comprise at least one compound according to the invention, typically together with one or more inert, non-toxic, pharmaceutically suitable excipients, and for the use thereof for the aforementioned purposes.


The compounds according to the invention may act systemically and/or locally. For this purpose, they can be administered in a suitable manner, for example by the oral, parenteral, pulmonal, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival or otic route, or as an implant or stent.


The compounds according to the invention can be administered in suitable administration forms for these administration routes.


Suitable administration forms for oral administration are those which work according to the prior art and release the compounds according to the invention rapidly and/or in a modified manner and which contain the compounds according to the invention in crystalline and/or amorphized and/or dissolved form, for example tablets (uncoated or coated tablets, for example with gastric juice-resistant or retarded-dissolution or insoluble coatings which control the release of the compound according to the invention), tablets or films/oblates which disintegrate rapidly in the oral cavity, films/lyophilizates or capsules (for example hard or soft gelatin capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.


Parenteral administration can bypass an absorption step (e.g. intravenously, intraarterially, intracardially, intraspinally or intralumbally) or include an absorption (e.g. intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally). Suitable administration forms for parenteral administration include injection and infusion formulations in the form of solutions, suspensions, emulsions, lyophilizates or sterile powders.


For the other administration routes, suitable examples are inhalable medicament forms (including powder inhalers, nebulizers, metered aerosols), nasal drops, solutions or sprays, tablets, films/oblates or capsules for lingual, sublingual or buccal administration, suppositories, ear or eye preparations, vaginal capsules, aqueous suspensions (lotions, shaking mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (e.g. patches), milk, pastes, foams, sprinkling powders, implants or stents.


Preference is given to oral and intravenous administration.


The compounds according to the invention can be converted to the administration forms mentioned. This can be done in a manner known per se, by mixing with inert, nontoxic, pharmaceutically suitable excipients. These excipients include carriers (for example microcrystalline cellulose, lactose, mannitol), solvents (e.g. liquid polyethylene glycols), emulsifiers and dispersing or wetting agents (for example sodium dodecylsulphate, polyoxysorbitan oleate), binders (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), stabilizers (e.g. antioxidants, for example ascorbic acid), colorants (e.g. inorganic pigments, for example iron oxides) and flavour and/or odour correctants.


In general, it has been found to be advantageous in the case of parenteral administration to administer amounts of about 0.001 to 5 mg/kg, preferably about 0.01 to 3 mg/kg, of body weight to achieve effective results. In the case of oral administration, the dosage is about 0.01 to 100 mg/kg, preferably about 0.01 to 50 mg/kg and most preferably 0.1 to 30 mg/kg of body weight. In the case of intrapulmonary administration, the amount is generally about 0.1 to 50 mg per inhalation.


It may nevertheless be necessary in some cases to deviate from the stated amounts, specifically as a function of the body weight, route of administration, individual response to the active ingredient, nature of the preparation and time or interval over which administration takes place. Thus, in some cases less than the abovementioned minimum amount may be sufficient, while in other cases the upper limit mentioned must be exceeded. In the case of administration of relatively large amounts, it may be advisable to divide these into several individual doses over the course of the day.


The working examples which follow illustrate the invention. The invention is not limited to the examples.


A. EXAMPLES
Abbreviations and Acronyms















abs.
absolute


Ac
acetyl


Ac2O
acetic anhydride


aq.
aqueous, aqueous solution


Boc
tert-butoxycarbonyl


br.
broad (in NMR signal)


Ex.
Example


Bu
butyl


c
concentration


cat.
catalytic


CI
chemical ionization (in MS)


d
doublet (in NMR)


d
day(s)


TLC
thin-layer chromatography


DCI
direct chemical ionization (in MS)


dd
doublet of doublets (in NMR)


DIPEA
N,N-diisopropylethylamine


DMAP
4-N,N-dimethylaminopyridine


DME
1,2-dimethoxyethane


DMF
N,N-dimethylformamide


DMSO
dimethyl sulphoxide


dt
doublet of triplets (in NMR)


ee
enantiomeric excess


EI
electron impact ionization (in MS)


ent
enantiomerically pure, enantiomer


eq.
equivalent(s)


ES
electrospray ionization (in MS)


Et
ethyl


GC
gas chromatography


GC-MS
gas chromatography-coupled mass spectrometry


h
hour(s)


HPLC
high-pressure, high-performance liquid chromatography


iPr
isopropyl


conc.
concentrated (in case of a solution)


LC
liquid chromatography


LC-MS
liquid chromatography-coupled mass spectrometry


LDA
lithium diisopropylamide


lit.
literature (reference)


m
multiplet (in NMR)


Me
methyl


min
minute(s)


MPLC
medium-pressure liquid chromatography (on silica gel;



also referred to as flash chromatography)


Ms
methanesulphonyl (mesyl)


MS
mass spectrometry


NMP
N-methyl-2-pyrrolidinone


NMR
nuclear magnetic resonance spectrometry


Pd/C
palladium on activated carbon


PEG
polyethylene glycol


Pr
propyl


prep.
preparative


q (or quart)
quartet (in NMR)


qd
quartet of doublets (in NMR)


quant.
quantitative (in chemical yield)


quint
quintet (in NMR)


rac
racemic, racemate


Rf
retention index (in TLC)


RP
reversed phase (in HPLC)


RT
room temperature


Rt
retention time (in HPLC, LC/MS)


s
singlet (in NMR)


sept
septet (in NMR)


t
triplet (in NMR)


tBu
tert-butyl


td
triplet of doublets (in NMR)


Tf
trifluoromethylsulphonyl (triflyl)


TFA
trifluoroacetic acid


THF
tetrahydrofuran


Ts
para-tolylsulphonyl (tosyl)


UV
ultraviolet spectrometry


v/v
ratio by volume (of a solution)


tog.
together









LC-MS Methods:
Method 1 (LC-MS):

Instrument: Waters ACQUITY SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8μ50×1 mm; mobile phase A: 11 of water+0.25 ml of 99% strength formic acid, mobile phase B: 1 l of acetonitrile+0.25 ml of 99% formic acid; gradient: 0.0 min 90% A÷1.2 min 5% A→2.0 min 5% A; oven: 50° C.; flow rate: 0.40 ml/min; UV detection: 208-400 nm.


Method 2 (LC-MS):

Instrument: Waters ACQUITY SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8μ 50×1 mm; mobile phase A: 1 l of water+0.25 ml of 99% strength formic acid, mobile phase B: 1 l of acetonitrile+0.25 ml of 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 3 (LC-MS):

Instrument: Micromass Quattro Premier with Waters UPLC Acquity; column: Thermo Hypersil GOLD 1.9 μ50×1 mm; mobile phase A: 1 l of water+0.5 ml of 50% strength formic acid, mobile phase B: 1 l of acetonitrile+0.5 ml of 50% strength formic acid; gradient: 0.0 min 97% A→0.5 min 97% A→3.2 min 5% A→4.0 min 5% A; oven: 50° C.; flow rate: 0.3 ml/min; UV detection: 210 nm.


Method 4 (LC-MS):

MS instrument: Waters Micromass QM; HPLC instrument: Agilent 1100 series; column: Agilent ZORBAX Extend-C18 3.5μ, 3.0×50 mm; mobile phase A: 1 l of water+0.01 mol of ammonium carbonate, mobile phase B: 1 l of acetonitrile; gradient: 0.0 min 98% A→0.2 min 98% A→3.0 min 5% A→4.5 min 5% A; oven: 40° C.; flow rate: 1.75 ml/min; UV detection: 210 nm.


Method 5 (LC-MS):

MS instrument: Waters Micromass ZQ; HPLC instrument: Agilent 1100 series; column: Agilent ZORBAX Extend-C18 3.5μ, 3.0×50 mm; mobile phase A: 1 l of water+0.01 mol of ammonium carbonate, mobile phase B: 1 l of acetonitrile; gradient: 0.0 min 98% A→0.2 min 98% A→3.0 min 5% A→4.5 min 5% A; oven: 40° C.; flow rate: 1.75 ml/min; UV detection: 210 nm.


Method 6 (LC-MS):

Instrument: Agilent MS Quad 6150; HPLC: Agilent 1290; column: Waters Acquity UPLC HSS T3 1.8μ50×2.1 mm; mobile phase A: 1 l of water+0.25 ml of 99% strength formic acid, mobile phase B: 1 l of acetonitrile+0.25 ml of 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.


Further Details:

The percentages in the example and test descriptions which follow are, unless indicated otherwise, percentages by weight; parts are parts by weight. Solvent ratios, dilution ratios and concentration figures for liquid/liquid solutions are each based on volume.


Purities are generally based on corresponding peak integrations in the LC/MS chromatogram, but they may additionally have been determined with the aid of the 1H-NMR spectrum. If no purity is indicated, the purity is generally 100% according to automated peak integration in the LC/MS chromatogram, or the purity has not been determined explicitly.


Stated yield in % of theory are generally corrected for purity if a purity of <100% is indicated. In solvent-containing or impure batches, the formal yield may be “>100%”; in these cases the yield is not corrected for solvent or purity.


When compounds according to the invention are purified by preparative HPLC where the mobile phases contain additives such as, for example, trifluoroacetic acid, formic acid or ammonia, the compounds according to the invention may be obtained in salt form, for example as trifluoroacetate, formate or ammonium salt, if the compounds according to the invention have a sufficiently basic or acidic functionality. Such a salt can be converted to the free base or acid by various methods known to the person skilled in the art.


Some of the descriptions below of the coupling patterns of 1H-NMR signals were taken directly from the suggestions of the ACD SpecManager (ACD/Labs Release 12.00, Product version 12.5) and have not necessarily been rigorously checked. In some cases, the suggestions of the SpecManager were adjusted manually. Manually adjusted or assigned descriptions are generally based on the optical appearance of the signals in question and do not necessarily correspond to a strict, physically correct interpretation. In general, the stated chemical shift refers to the centre of the signal in question. In the case of broad multiplets, an interval is given. Signals obscured by solvent or water were either tentatively assigned or have not been listed.


Melting points and melting points ranges, if stated, are uncorrected.


For all the reactants or reagents for which the preparation is not described explicitly in the following, they were obtained commercially from generally accessible sources. For all the other reactants or reagents for which the preparation likewise is not described in the following and which were not commercially obtainable or were obtained from sources which are not generally accessible, reference is made to the published literature in which their preparation is described.







Starting Materials and Intermediates
EXAMPLE 1A
Ethyl 2-[4-chloro-3-(trifluoromethyl)phenoxy]acetate



embedded image


At 23° C. (cooling !), 25 g (127.2 mmol) of 4-chloro-3-(trifluoromethyl)phenol in 50 ml of THF were added dropwise to a suspension of 5.6 g (140 mmol) of sodium hydride (60% in paraffin) in 125 ml of THF, with evolution of hydrogen in an exothermic reaction. After 30 min of stirring, 23.4 g (140 mmol) of ethyl bromoacetate in 50 ml of THF were added dropwise, and the mixture was stirred at 23° C. for 2 h. Another 2.34 g of ethyl bromoacetate were added, and the mixture was stirred at 23° C. for a further 2 h. The mixture was then diluted was ethyl acetate and washed with water, and the aqueous phase was re-extracted with ethyl acetate. The combined organic phases were washed with water and dried over sodium sulphate. After removal of the drying agent by filtration, the mixture was concentrated under reduced pressure. Drying under high vacuum gave 38.3 g (96% of theory, purity 90%) of the target compound. The product could be converted further without further purification.


LC-MS (Method 1): Rt=1.15 min; MS (ESneg): not ionizable



1H-NMR (400 MHz, DMSO-d6): □=1.21 (t, 3H), 4.17 (q, 2H), 4.94 (s, 2H), 7.29 (dd, 1H), 7.37 (d, 1H), 7.64 (d, 1H).


EXAMPLE 2A
Ethyl 2-[4-fluoro-3-(trifluoromethyl)phenoxy]acetate



embedded image


At 23° C., 2 g (11.1 mmol) of 4-fluoro-3-(trifluoromethyl)phenol were added dropwise to a suspension of 0.49 g (12.2 mmol) of sodium hydride (60% in paraffin) in 25 ml of THF, with evolution of hydrogen in an exothermic reaction. After 30 min of stirring, 1.86 g (11.1 mmol) of ethyl bromoacetate were added, and the mixture was stirred at 23° C. for 18 h. The mixture was then diluted was ethyl acetate and washed with water, and the organic phase was dried over magnesium sulphate. After removal of the drying agent by filtration, the mixture was concentrated under reduced pressure. Drying under high vacuum gave 2.43 g (78% of theory, purity 95%) of the target compound.


LC-MS (Method 3): Rt=2.42 min; MS (ESpos): m/z=267 (M+H)+.


The following compounds are known from the literature, commercially available or can be prepared analogously to Example 2A:











TABLE 1





Example




No.
IUPAC name/structure
CAS number; literature







3A
ethyl (4-chloro-3-fluorophenoxy)acetate   embedded image
CAS 1096703-33-1; preparation described in WO 2012/041817 (Intermediate 87)





4A
ethyl (3-chloro-4-fluorophenoxy)acetate   embedded image
CAS 667437-18-5; preparation described in Tetrahedron 2004, 60 (52), 12231- 12237





5A
ethyl (3,4-difluorophenoxy)acetate   embedded image
CAS 1094524-83-0





6A
ethyl (3-chlorophenoxy)acetate   embedded image
CAS 52094-98-1; commercially available





7A
ethyl 2-[(5-chloropyridin-3-yl)oxy]acetate   embedded image
CAS 53233-36-6; commercially available





8A
ethyl (3,4-dichlorophenoxy)acetate   embedded image
CAS 62855-72-5; preparation described in WO 2012/041817 (Intermediate 88)









EXAMPLE 9A
Ethyl 2-[4-chloro-3-(trifluoromethyl)phenoxy]-4,4,4-trifluoro-3-oxobutanoate



embedded image


Initially 26 g (182.8 mmol) of ethyl trifluoroacetate and then 38.3 g (121.9 mmol, purity 90%) of ethyl [4-chloro-3-(trifluoromethyl)phenoxy]acetate were added dropwise to a suspension of 12.19 g (304.7 mmol) of sodium hydride (60% in paraffin) in 150 ml of toluene. The mixture was heated to reflux, resulting in a noticeable evolution of gas, and boiled for one hour. The cooled reaction was then acidified with 1 N hydrochloric acid. The organic phase was separated off, washed with dilute brine, dried over sodium sulphate and filtered, and the filtrate was concentrated. Drying under high vacuum gave 50.6 g (76% of theory, purity 69%) of the target compound. The product was converted further without further purification.


LC-MS (Method 3): Rt=2.51 min; MS (ESneg): m/z=377 (M−H).


The following synthesis intermediates were prepared analogously to Example 9A:











TABLE 2





Example
IUPAC name/structure



No.
(yield; reaction time)
Analytical data







10A
ethyl 2-(4-chloro-3-fluorophenoxy)-4,4,4- trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.01 min; MS (ESneg): m/z = 326.9 (M − H)





11A
ethyl 2-(3-chloro-4-fluorophenoxy)-4,4,4- trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.00 min; MS (ESneg): m/z = 326.9 (M − H)





12A
ethyl 4,4,4-trifluoro-2-[4-fluoro-3- (trifluoromethyl)phenoxy]-3-oxobutanoate   embedded image
LC-MS (Method 3): Rt = 2.35 min; MS (ESneg): m/z = 361.0 (M − H)





13A
ethyl 2-(3-chlorophenoxy)-4,4,4-trifluoro-3- oxobutanoate   embedded image
LC-MS (Method 1): Rt = 0.98-1.00 min; MS (ESneg): m/z = 309.0 (M − H)





14A
ethyl 2-[(5-chloropyridin-3-yl)oxy]-4,4,4- trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 0.83-0.86 min; MS (ESneg): m/z = 309.9 (M − H)





15A
ethyl 2-(3,4-difluorophenoxy)-4,4,4-trifluoro- 3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 0.95 min; MS (ESneg): m/z = 311.0 (M − H)





16A
ethyl 2-(3,4-dichlorophenoxy)-4,4,4-trifluoro- 3-oxobutanoate   embedded image
LC-MS (Method 3): Rt = 2.31 min; MS (ESneg): m/z = 343.0 (M − H)









EXAMPLE 17A
ethyl 4,4,4-trifluoro-3-oxo-2-[3-(trifluoromethyl)benzyl]butanoate



embedded image


10.8 g (83.7 mmol) of N,N-diisopropylethylamine and 1.77 g (41.8 mmol) of lithium chloride were added to 10 g (41.8 mmol) of 3-(bromomethyl)benzotrifluoride and 11.6 g (62.75 mmol) of ethyl trifluoroacetate in 51.6 ml of THF. The mixture was stirred at 67° C. for 18 h. The reaction was then concentrated under reduced pressure and the residue was taken up in ethyl acetate. The solution was washed with 1 N hydrochloric acid and the organic phase was dried over sodium sulphate, filtered and concentrated. The yellow oil (9.56μ, 27% of theory), which was obtained in a purity of 40% (HPLC), was used without further purification for the next step.


LC-MS (Method 1): Rt=1.12 min; MS (ESneg): m/z=341 (M−H).


Analogously to Example 17A, the following compound was prepared from the corresponding benzyl halide:











TABLE 3





Example
IUPAC name/structure



No.
(yield)
Analytical data







18A
ethyl 2-(4-chlorobenzyl)-4,4,4-trifluoro-3- oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.09 min; MS (ESneg): m/z = 307.1 (M − H)









The following synthesis intermediates were prepared analogously to the method described in WO 2011/114148 (Methode XX) from the corresponding benzyl halides:











TABLE 4





Example
IUPAC name/structure



No.
(yield)
Analytical data







19A
ethyl 4,4,4-trifluoro-2-[3-fluoro-5- (trifluoromethyl)benzyl]-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.10 min and 1.37 min; MS (ESneg): m/z = 359.1 (M − H)





20A
ethyl 2-(4-chloro-3-fluorobenzyl)-4,4,4- trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.07 min; MS (ESneg): m/z = 325.0 (M − H)





21A
ethyl 2-[4-chloro-3-(trifluoromethyl)benzyl]- 4,4,4-trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.14 min; MS (ESneg): m/z = 374.9 (M − H)





22A
ethyl 2-(3-chloro-4-methylbenzyl)-4,4,4- trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.12 min; MS (ESneg): m/z = 321.1 (M − H)





23A
ethyl 2-(3-chloro-4-fluorobenzyl)-4,4,4- trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.06 min; MS (ESneg): m/z = 325.1 (M − H)





24A
ethyl 2-[3-chloro-4-(trifluoromethyl)benzyl]- 4,4,4-trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.14 min; MS (ESneg): m/z = 375.1 (M − H)









EXAMPLE 25A
1H-Pyrazole-3-carboximidamide hydrochloride



embedded image


Step 1:


5 g (53.7 mmol) of 1H-pyrazole-3-carbonitrile were dissolved in 12.5 ml of ethanol and 50 ml of chloroform. With cooling using an ice/acetone bath, gaseous hydrogen chloride was introduced for 50 minutes. The cooling bath was then removed and the mixture was stirred for 3 h. During this time, a solid precipitated out, and this was filtered off and washed with chloroform. Drying under high vacuum gave 7.7 g (81% of theory) of the intermediate ethyl 1H-pyrazole-3-carboximidoate hydrochloride.


Step 2:


90 ml of a 7 N solution of ammonia in methanol were initially charged, and 9.0 g (51 mmol) of ethyl 1H-pyrazole-3-carboximidoate hydrochloride were added with ice cooling. The ice bath was then removed and the mixture was stirred for 16 h. The mixture was then concentrated to dryness and the residue that remained was dried under high vacuum. This gave 7.9 g (quant.) of the title compound.


LC-MS (Method 4): Rt=0.28 min; MS (ESpos): m/z=109 (M+H)+



1H NMR (400 MHz, DMSO-d6): □□=7.14 (d, 1H), 8.05 (d, 1H).


Analogously to Example 25A, the compounds listed in Table 5 were prepared from the corresponding nitriles:











TABLE 5





Example
IUPAC name/structure



No.
(yield)
Analytical data







26A
5-amino-1H-pyrazole-4-carboximidamide hydrochloride   embedded image
LC-MS (Method 4): Rt = 0.21 min; MS (ESpos): m/z = 126 (M + H)+





27A
1-methyl-1H-imidazole-4-carboximidamide hydrochloride   embedded image

1H NMR (400 MHz, DMSO-d6): δ = 3.77 (s, 3H), 7.94 (s, 1H), 8.27 (d, 1H), 8.63-8.90 (m, 1H), 9.04 (br. s, 2H).






28A
5-amino-1-methyl-1H-pyrazole-4- carboximidamide hydrochloride   embedded image
LC-MS (Method 5): Rt = 0.26 min; MS (ESpos): m/z = 140 (M + H)+1H NMR (400 MHz, DMSO-d6): δ = 3.55-3.63 (m, 3H), 6.68 (br. s, 2H), 7.79 (s, 1H), 8.38 (br. s, 3H).





29A
5-amino-1H-imidazole-4-carboximidamide hydrochloride   embedded image
LC-MS (Method 5): Rt = 0.22 min; MS (ESpos): m/z = 126 (M + H)+





30A
1H-pyrazole-4-carboximidamide hydrochloride   embedded image
LC-MS (Method 4): Rt = 0.26 min; MS (ESpos): m/z = 110.9 (M + H)+





31A
4-chloro-1H-pyrazole-3-carboximidamide hydrochloride   embedded image
LC-MS (Method 1): Rt = 0.17 min; MS (ESpos): m/z = 145.1 (M + H)+





32A
1,5-dimethyl-1H-pyrazole-3-carboximidamide hydrochloride   embedded image
LC-MS (Method 4): Rt = 0.41 min; MS (ESpos): m/z = 139.0 (M + H)+









EXAMPLE 33A
1-(6-Methoxypyridin-2-yl)guanidine



embedded image


The title compound was prepared analogously to the preparation of 1-(3-methoxypyridin-2-yl)guanidine [Bioorg. Med. Chem. Lett. 2002, 12 (2), 181-184] using N,N′-di-Boc-protected S-methylisothiourea.


LC-MS (Method 4): Rt=1.33 min; MS (ESneg): m/z=167.1 (M−H).


EXAMPLE 34A
5-(3,4-Dichlorophenoxy)-2-(methylsulphanyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 8.65 g (63 mmol) of potassium carbonate, 6.77 g (75 mmol) of S-methylisothiourea hemisulphate and 8 g (12.5 mmol; purity 54%) of ethyl 2-(3,4-dichlorophenoxy)-4,4,4-trifluoro-3-oxobutanoate (Example 16A) in 101 ml of dioxane was stirred at 95° C. for 2 h. 1 ml of 1 N hydrochloric acid was then added, the mixture was concentrated under reduced pressure and 300 ml of water were added to the residue. The precipitated solid was filtered off with suction and washed successively with water, petroleum ether and diethyl ether. Drying under high vacuum gave 5.85 g (91% of theory) of the title compound in a purity of 72% (HPLC).


LC-MS (Method 1): Rt=1.13 min; MS (ESpos): m/z=371.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=2.56 (s, 3H), 7.13 (dd, 1H), 7.48 (d, 1H), 7.56 (d, 1H), 13.72 (br. s, 1H).


The compounds listed in Table 6 were prepared analogously to Example 34A by reacting S-methylisothiourea hemisulphate with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 6





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







35A
5-(3,4-dichlorobenzyl)-2-(methylsulphanyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 2): Rt = 3.85 min; MS (ESpos): m/z = 368.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.86 (s, 2H), 7.10-7.19 (m, 1H), 7.44 (d, J = 1.3 Hz, 1H), 7.52 (d, J = 8.3 Hz, 1H), 13.47 (br. s, 1H).





36A
5-[4-chloro-3-(trifluoromethyl)benzyl]-2- (methylsulphanyl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.24 min; MS (ESpos): m/z = 403.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.52 (s, partially obscured by DMSO signal), 3.94 (s, 2H), 7.41-7.46 (m, 1H), 7.60 (d, 1H), 7.77 (m, 1H), 13.49 (br. s, 1H).





37A
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2- (methylsulphanyl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.23 min; MS (ESpos): m/z = 405.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.56 (s, 3H), 7.43 (dd, 1H), 7.57 (d, 1H), 7.65 (d, 1H), 13.76 (br. s, 1H).









EXAMPLE 38A
5-(3,4-Dichlorophenoxy)-2-(methylsulphonyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 4 g (7.8 mmol) of 5-(3,4-dichlorophenoxy)-2-(methylsulphanyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 34A; purity 72%), 14.37 g (23.4 mmol) of Oxone™ and 4.07 g (23.4 mmol) of dipotassium phosphate was stirred in 68 ml of dioxane and 32 ml of water at 22° C. for 18 h. The reaction mixture was subsequently stirred with 1 litre of water and the resulting white crystals were filtered off with suction. After washing with 100 ml of water and 50 ml of petroleum ether, the solid was dried under high vacuum. This gave 2.46 g (75% of theory) of the title compound.


LC-MS (Method 1): Rt=0.95 min; MS (ESneg): m/z=400.9 (M−H).


The following synthesis intermediates were prepared analogously to Example 38A.











TABLE 7





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







39A
5-[4-chloro-3-(trifluoromethyl)benzyl]-2- (methylsulphonyl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.10 min; MS (ESpos): m/z = 434.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.36 (s, 3H), 4.12 (s, 2H), 7.42 (d, 1H), 7.63 (d, 1H), 7.74 (d, 1H).





40A
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2- (methylsulphonyl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 4): Rt = 2.17 min; MS (ESneg): m/z = 435 (M − H)1H-NMR (400 MHz, DMSO-d6): δ = 3.25 (s, 3H), 7.20 (dd, 1H), 7.40 (d, 1H), 7.63 (d, 1H).









EXAMPLE 41A
2-Amino-5-(3,4-dichlorobenzyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 110 mg (0.8 mmol) of potassium carbonate, 76 mg (0.8 mmol) of guanidine hydrochloride and 400 mg (0.8 mmol) of ethyl 2-(3,4-dichlorobenzyl)-4,4,4-trifluoro-3-oxobutanoate (purity 68%; CAS 179110-12-4; WO 2012/041817, Intermediate 56) in 4 ml of ethanol were heated under reflux for 6 h. The solution was then concentrated under reduced pressure and the residue was purified by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% of formic acid). This gave 78 mg (29% of theory) of the title compound.


LC-MS (Method 1): Rt=1.04 min; MS (ESpos): m/z=338.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.74 (s, 2H), 6.98 (br. s, 2H), 7.11 (dd, 1H), 7.38 (d, 1H), 7.51 (d, 1H), 11.53 (br. s, 1H).


EXAMPLE 42A
2-Amino-5-[4-chloro-3-(trifluoromethyl)phenoxy]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 20.15 g (146 mmol) of potassium carbonate, 10.5 g (109 mmol) of guanidine hydrochloride and 20 g (36.5 mmol, purity 69%) of ethyl 2-[4-chloro-3-(trifluoromethyl)phenoxy]-4,4,4-trifluoro-3-oxobutanoate (Example 9A) in 150 ml of dioxane was heated under reflux for 1 h.


The reaction mixture was then added to 1.8 litres of water and neutralized with 1 N hydrochloric acid. The precipitated solid was filtered off with suction, washed with water and taken up in a little ethyl acetate, and the resulting solution was added dropwise with stirring to 1 litre of petroleum ether. The resulting precipitate was filtered off with suction, taken up in 100 ml of 0.5 N sulphuric acid and 100 ml of acetonitrile, stirred for 30 min and then added to 1 litre of water. After 15 min of stirring, the mixture was once more filtered off with suction and the precipitate was washed with water. The product was taken up in ethyl acetate and, together with silica gel, reconcentrated under reduced pressure. This material was chromatographed on silica gel using a mixture of cyclohexane and ethyl acetate (1:1). The product-containing fractions were concentrated and the residue was dried under reduced pressure. This gave 10.5 g (77% of theory) of the title compound in a purity of 99% (HPLC).


LC-MS (Method 1): Rt=1.02 min; MS (ESpos): m/z=374.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=7.07 (br. s, 2H), 7.31 (dd, 1H), 7.42 (d, 1H), 7.62 (d, 1H), 11.86 (br. s, 1H).


EXAMPLE 43A
2-Amino-5-(3,4-dichlorophenoxy)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 5.53 g (40 mmol) of potassium carbonate, 2.87 g (30 mmol) of guanidinium hydrochloride and 6.70 g (10 mmol; purity 52%) of ethyl 2-(3,4-dichlorophenoxy)-4,4,4-trifluoro-3-oxobutanoate (Example 16A) in 33 ml of dioxane was stirred at 90° C. for 1 h. The reaction mixture was then added to 0.8 litre of water and neutralized with 1 N hydrochloric acid. The precipitated solid was filtered off with suction and washed with 100 ml of water and 200 ml of petroleum ether. The residue was chromatographed on silica gel using a mixture of cyclohexane and ethyl acetate (initially 1:1, then 0:1). The product-containing fractions were concentrated and the residue was dried under reduced pressure. This gave 3.04 g (87% of theory) of the title compound in a purity of 97% (HPLC).


LC-MS (Method 1): Rt=0.99 min; MS (ESpos): m/z=340.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=7.00-7.18 (br. s, 2H), 7.01 (dd, 1H), 7.33 (d, 1H), 7.52 (d, 1H), 11.80 (br. s, 1H).


The intermediates listed in Table 8 were prepared analogously to Example 43A by reacting guanidine hydrochloride with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 8





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







44A
2-amino-5-(3-chloro-4-fluorophenoxy)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.94 min; MS (ESpos): m/z = 324 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.88-7.14 (m, 3H), 7.25 (dd, 1H), 7.32 (t, 1H), 11.79 (br. s, 1H).





45A
2-amino-5-(4-chloro-3-fluorophenoxy)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.96 min; MS (ESpos): m/z = 324 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.87 (dd, 1H), 7.03 (br. s, 2H), 7.18 (dd, 1H), 7.47 (t, 1H), 11.82 (br. s, 1H).





46A
2-amino-5-[4-fluoro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.98 min; MS (ESpos): m/z = 358.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.05 (br. s, 2H), 7.30-7.39 (m, 2H), 7.39-7.48 (m, 1H), 11.81 (br. s, 1H).





47A
2-amino-5-(3-chloro-4-fluorobenzyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.95 min; MS (ESpos): m/z = 322.2 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.74 (s, 2H), 6.80-7.38 (m, 5H), 11.52 (br. s, 1H).





48A
2-amino-5-[4-chloro-3-(trifluoromethyl)benzyl]- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.05 min; MS (ESpos): m/z = 372.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.83 (s, 2H), 7.42 (dd, 1H), 7.63-7.65 (m, 1H), 11.56 (br. s, 1H).





49A
2-amino-5-(3-chlorophenoxy)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.91 min; MS (ESpos): m/z = 306.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.93 (dd, 1H), 6.97-7.11 (m, 3H), 7.31 (t, 1H), 11.79 (br. s, 1H).





50A
2-amino-5-(3,4-difluorophenoxy)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.89 min; MS (ESpos): m/z = 308.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.77-6.84 (m, 1H), 6.88-7.12 (m, 2H), 7.28-7.40 (m, 1H), 11.79 (br. s, 1H).





51A
2-amino-5-(3-chloro-4-methylbenzyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
conditions of prep. HPLC purification: Column: Daicel Chiracel OD-H 5 μm, 250 × 20 mm; flow rate: 20 ml/min; run time: 9 min; detection: 230 nm, mobile phase: isohexane/ethanol 80:20. LC-MS (Method 1): Rt = 1.02 min; MS (ESpos): m/z = 318.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.26 (s, 3H), 3.71 (br. s, 2H), 6.81- 7.34 (m, 5H), 11.49 (br. s, 1H).





52A
2-Amino-5-[(5-chloropyridin-3-yl)oxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.80 min; MS (ESpos): m/z = 349.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.08 (br. s, 2H), 7.72 (t, 2H), 8.31 (d, 1H), 8.36 (d, 1H), 11.90 (br. s, 1H).





53A
2-Amino-5-(4-chloro-3-fluorobenzyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.98 min; MS (ESpos): m/z = 322.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.76 (s, 2H), 6.80-7.10 (m, 3H), 7.16 (d, 1H), 7.45 (t, 1H), 11.50 (br. s, 1H).





54A
2-Amino-5-[3-chloro-4-(trifluoromethyl)benzyl]- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.05 min; MS (ESpos): m/z = 372.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.84 (br. s, 2H), 6.63-8.02 (m, 5H), 11.56 (br. s, 1H).









EXAMPLE 55A
2-Amino-5-[(3,4-dichlorophenyl)sulphanyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 258 mg (1 mmol) of 2-amino-5-bromo-6-(trifluoromethyl)pyrimidin-4(3H)-one [CAS 1583-00-2; preparation analogously to WO 2011/114148, Method XIX], 326 mg (1 mmol) of caesium carbonate and 179 mg (1 mmol) of 3,4-dichlorothiophenol in 5 ml of ethylene glycol was stirred at 110° C. for 6 h. The mixture was then concentrated. The residue was purified by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% of formic acid). The product-containing fractions were concentrated and the residue was dried under reduced pressure. This gave 81 mg (23% of theory) of the title compound in a purity of 100% (HPLC).


LC-MS (Method 1): Rt=1.01 min; MS (ESpos): m/z=356.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=6.15-8.95 (br. s, 2H), 7.09 (dd, 1H), 7.36 (d, 1H), 7.49 (d, 1H), 11.80 (br. s, 1H).


The following intermediates were prepared in an analogous manner:











TABLE 9





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







56A
2-amino-5-[(4-chlorophenyl)sulphanyl]-6- (trifluoromethyl)pyrimidin-4(3H)-one embedded image
  (27% of theory; reaction time: 6 h, 150° C.; solvent: ethylene glycol; 3 eq. of 4-chlorothiophenol, 1 eq. caesium carbonate)

LC-MS (Method 1): Rt = 0.95 min; MS (ESpos): m/z = 322.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.56-8.62 (br. s, 2H), 7.12 (d, 2H), 7.31 (d, 2H), 11.76 (br. s, 1H).





57A
2-amino-5-{[4-chloro-3- (trifluoromethyl)phenyl]sulphanyl}-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (7% of theory; reaction time: 24 h, 150° C.; solvent: ethylene glycol; 3 eq. of 4-chloro-3- (trifluoromethyl)thiophenol, 1 eq. caesium carbonate)

LC-MS (Method 1): Rt = 1.06 min; MS (ESpos): m/z = 390.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.35-8.72 (br. s, 2H), 7.38 (dd, 1H), 7.55-7.62 (m, 2H), 11.85 (br. s, 1H).









EXAMPLE 58A
2-{5-[3-Chloro-4-(trifluoromethyl)benzyl]-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl}acetamide



embedded image


A mixture of 293 mg (2.1 mmol) of potassium carbonate, 219 mg (1.6 mmol) of 3,3-diaminoprop-2-enamide hydrochloride and 200 mg (0.5 mmol) of ethyl 2-[3-chloro-4-(trifluoromethyl)benzyl]-4,4,4-trifluoro-3-oxobutanoate (Example 24A) in 2.3 ml of dioxane was heated under reflux for 18 h. The mixture was then filtered, the residue was washed with dioxane and the filtrate was purified by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% of formic acid). This gave, from two reactions with, in total, 0.66 mmol of ethyl 2-[3-chloro-4-(trifluoromethyl)benzyl]-4,4,4-trifluoro-3-oxobutanoate, 60 mg (20% of theory) of the title compound.


LC-MS (Method 1): Rt=1.01 min; MS (ESpos): m/z=414.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.54 (s, 2H), 4.00 (s, 2H), 7.22-7.34 (m, 2H), 7.54 (s, 1H), 7.65 (br. s, 1H), 7.78 (d, 1H), 13.21 (br. s, 1H).


The intermediates listed in Table 10 were prepared analogously to Example 58A by reacting 3,3-diaminoprop-2-enamide hydrochloride with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 10





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







59A
2-[5-(3-chloro -4-fluorophenoxy)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]acetamide   embedded image
LC-MS (Method 1): Rt = 0.90 min; MS (ESpos): m/z = 366 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.55 (s, 2H), 7.06 (dt, 1H), 7.29 (br. s, 1H), 7.34 (dd, 1H), 7.39 (t, 1H), 7.62 (br. s, 1H), 13.47 (s, 1H).





60A
2-[5-(3,4-difluorophenoxy)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]acetamide   embedded image
LC-MS (Method 1): Rt = 0.86 min; MS (ESpos): m/z = 350 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.55 (s, 2H), 6.83-6.91 (m, 1H), 7.22- 7.34 (m, 2H), 7.40 (q, 1H), 7.62 (br. s, 1H), 13.47 (br. s, 1H).





61A
2-[5-(3-chlorophenoxy)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]acetamide   embedded image
LC-MS (Method 1): Rt = 0.88 min; MS (ESpos): m/z = 348 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.56 (s, 2H), 6.99 (dd, 1H), 7.10-7.19 (m, 2H), 7.29 (br. s, 1H), 7.36 (t, 1H), 7.63 (br. s, 1H), 13.47 (br. s, 1H).





62A
2-[5-(4-chloro-3-fluorophenoxy)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]acetamide   embedded image
LC-MS (Method 1): Rt = 0.92 min; MS (ESpos): m/z = 366 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.54 (s, 2H), 6.91 (dt, 1H), 7.25 (dd, 1H), 7.28 (br. s, 1H), 7.53 (t, 1H), 7.62 (br. s, 1H), 13.26-13.63 (m, 1H).





63A
2-[5-(4-chloro-3-fluorobenzyl)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]acetamide   embedded image
LC-MS (Method 1): Rt = 0.94 min; MS (ESpos): m/z = 364 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.53 (s, 2H), 3.92 (s, 2H), 7.00 (d, 1H), 7.17-7.25 (m, 2H), 7.48 (t, 1H), 7.60-7.64 (m, 1H), 13.05-13.23 (m, 1H).





64A
2-{6-oxo-4-(trifluoromethyl)-5-[3- (trifluoromethyl)benzyl]-1,6-dihydropyrimidin- 2-yl}acetamide   embedded image
  (8% of theory; preparation analogous to Example 78A; 8 eq. 3,3-diaminoprop-2-enamide hydrochloride; 8.5 eq. sodium methoxide; solvent: methanol; reaction time: 10 h, 64° C.)

LC-MS (Method 1): Rt = 0.93 min; MS (ESpos): m/z = 380 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.53 (s, 2H), 4.00 (s, 2H), 7.24 (s, 1H), 7.41-7.48 (m, 1H), 7.48-7.59 (m, 3H), 7.65 (s, 1H), 12.90-13.29 (m, 1H).





65A
2-{5-[3-fluoro-5-(trifluoromethyl)benzyl]-6-oxo- 4-(trifluoromethyl)-1,6-dihydropyrimidin-2- yl}acetamide   embedded image
  (quant. yield; preparation analogous to Example 78A; 8 eq. 3,3-diaminoprop-2-enamide hydrochloride; 8.5 eq. sodium methoxide; solvent: methanol; reaction time: 10 h, 64° C.)

LC-MS (Method 1): Rt = 0.96 min; MS (ESpos): m/z = 398 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.53 (s, 2H), 4.02 (s, 2H), 7.26 (s, 1H), 7.32 (d, 1H), 7.42 (s, 1H), 7.53 (d, 1H), 7.65 (s, 1H), 13.20 (s, 1H).





66A
2-[5-(3-chlorobenzyl)-6-oxo-4-(trifluoromethyl)- 1,6-dihydropyrimidin-2-yl]acetamide   embedded image
  (68% of theory; preparation analogous to Example 78A; 8 eq. 3,3-diaminoprop-2-enamide hydrochloride; 8.5 eq. sodium methoxide; solvent: methanol; reaction time: 10 h, 64° C.)

LC-MS (Method 1): Rt = 0.91 min; MS (ESpos): m/z = 346 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.52 (s, 2H), 3.92 (s, 2H), 7.10 (d, 1H), 7.21-7.27 (m, 3H), 7.31 (q, 1H), 7.64 (s, 1H), 13.14 (s, 1H).





67A
2-[5-(3-chloro-4-fluorobenzyl)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]acetamide   embedded image
LC-MS (Method 1): Rt = 0.91 min; MS (ESpos): m/z = 364.2 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.53 (s, 2H), 3.90 (s, 2H), 7.11-7.43 (m, 4H), 7.64 (br. s, 1H), 13.17 (br. s, 1H).





68A
2-{5-[4-chloro-3-(trifluoromethyl)phenoxy]-6- oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2- yl}acetamide   embedded image
LC-MS (Method 1): Rt = 0.98 min; MS (ESpos): m/z = 416 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.57 (s, 2H), 7.30 (br. s, 1H), 7.36 (dd, 1H), 7.54 (d, 1H), 7.63 (br. s, 1H), 7.69 (d, 1H), 13.54 (br. s, 1H).





69A
2-[5-(3,4-dichlorophenoxy)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]acetamide   embedded image
LC-MS (Method 1): Rt = 0.96 min; MS (ESpos): m/z = 382 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.55 (s, 2H), 7.07 (dd, 1H), 7.30 (br. s, 1H), 7.41 (d, 1H), 7.59 (d, 1H), 7.62 (br. s, 1H), 13.50 (s, 1H).





70A
2-[5-(3-chloro-4-methylbenzyl)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]acetamide   embedded image
conditions of prep. HPLC purification: Column: Daicel Chirapak AS-H 5 μm, 250 × 20 mm; flow rate: 20 ml/min; run time: 7 min; detection: 285 nm, mobile phase: isohexane/(ethanol + 0.1% TFA) 50:50. LC-MS (Method 2): Rt = 2.77 min; MS (ESpos): m/z = 360.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.26 (s, 3H), 3.52 (s, 2H), 3.87 (s, 2H), 7.01 (d, 1H), 7.17-7.27 (m, 3H), 7.64 (br. s, 1H), 13.13 (br. s, 1H).





71A
2-{5-[4-chloro-3-(trifluoromethyl)benzyl]-6-oxo- 4-(trifluoromethyl)-1,6-dihydropyrimidin-2- yl}acetamide   embedded image
LC-MS (Method 1): Rt = 1.00 min; MS (ESpos): m/z = 414 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.52 (s, 2H), 3.98 (s, 2H), 7.25 (s, 1H), 7.43 (d, 1H), 7.62-7.66 (m, 2H), 7.70-7.72 (m, 1H), 13.19 (s, 1H).





72A
2-{5-[4-fluoro-3-(trifluoromethyl)phenoxy]-6- oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2- yl}acetamide   embedded image
LC-MS (Method 1): Rt = 0.93 min; MS (ESpos): m/z = 400 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.56 (s, 2H), 7.29 (br. s, 1H), 7.38- 7.46 (m, 2H), 7.48 (t, 1H), 7.63 (br. s, 1H), 13.50 (br. s, 1H).





73A
2-[5-(3,4-dichlorobenzyl)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl)acetamide   embedded image
  (61% of theory; preparation analogous to Example 78A; 8 eq. 3,3-diaminoprop-2-enamide hydrochloride; 8.5 eq. sodium methoxide; solvent: methanol; reaction time: 10 h, 64° C.)

LC-MS (Method 1): Rt = 0.96 min; MS (ESpos): m/z = 380 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.52 (s, 2H), 3.91 (s, 2H), 7.13 (dd, 1H), 7.25 (br. s, 1H), 7.43 (d, 1H), 7.54 (d, 1H), 7.65 (br. s, 1H), 13.14 (s, 1H).









EXAMPLE 74A
[5-(3,4-Dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetonitrile



embedded image


2 g (5.3 mmol) of 2-[5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetamide (Example 73A) in 30 ml of a 50% strength solution of propanephosphonic anhydride in ethyl acetate were stirred at 45° C. for 2 days. The reaction was then diluted with 300 ml of ethyl acetate and extracted three times with 200 ml of water. The organic phase was dried over sodium sulphate and filtered and the filtrate was concentrated to dryness. This gave 1.95 g (94% of theory) of the title compound.


LC-MS (Method 1): Rt=1.09 min; MS (ESpos): m/z=362.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.93 (s, 2H), 4.20 (s, 2H), 7.14 (dd, 1H), 7.42 (d, 1H), 7.54 (d, 1H), 13.41 (br. s, 1H).


EXAMPLE 75A
(1Z)-2-[5-(3,4-Dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]-N′-hydroxyethanimidamide



embedded image


102.67 mg (1.48 mmol) of hydroxylamine hydrochloride were dissolved in 1.5 ml of DMSO, and 0.21 ml (1.48 mmol) of triethylamine were added at RT. After 10 min, the mixture was filtered, 107 mg (0.30 mmol) of [5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetonitrile (Example 74A) were added to the filtrate and the mixture was stirred at 75° C. for 11 h. The reaction mixture was then diluted with water and extracted three times with ethyl acetate. The combined organic phases were washed three times with saturated sodium chloride solution, dried over magnesium sulphate and concentrated. The residue was purified by preparative HPLC (column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient 10:90→90:10). This gave 127 mg (about 100% of theory; purity 93%) of the title compound.


LC-MS (Method 1): Rt=0.93 min; MS (ESpos): m/z=395.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=13.02 (br. s, 1H), 9.20 (s, 1H), 7.55 (d, J=8.3 Hz, 1H), 7.43 (d, J=1.7 Hz, 1H), 7.14 (dd, J=8.4, 1.8 Hz, 1H), 5.63 (s, 2H), 3.90 (s, 2H), 3.38 (s, 2H).


EXAMPLE 76A
2-{[5-(3,4-Dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetyl}-N-(2,4-dimethoxybenzyl)hydrazinecarboxamide



embedded image


355 mg (0.90 mmol) of 2-[5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetohydrazide (Example 79A) and 208 mg (1.08 mmol) of 2,4-dimethoxybenzyl isocyanate were suspended in 3.5 ml of dichloromethane and the mixture was stirred at 23° C. overnight. The precipitated crystals were filtered off with suction, washed with dichloromethane and dried under high vacuum. 540 mg (96% of theory) of the title compound were obtained.


LC-MS (Method 2): Rt=1.10 min; MS (ESpos): m/z=588.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.61 (s, 2H), 3.70-3.80 (m, 6H), 3.91 (s, 2H), 4.11 (d, 2H), 6.43 (dd, 1H), 6.52 (d, 1H), 6.60-6.69 (m, 1H), 7.06 (d, 1H), 7.10-7.16 (m, 1H), 7.42 (d, 1H), 7.54 (d, 1H), 8.00 (s, 1H), 9.90 (s, 1H), 13.26 (br. s, 1H).


EXAMPLE 77A
5-(3,4-Dichlorobenzyl)-2-{[4-(2,4-dimethoxybenzyl)-5-oxo-4,5-dihydro-1H-1,2,4-triazol-3-yl]methyl}-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


520 mg (0.88 mmol) of 2-{[5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetyl}-N-(2,4-dimethoxybenzyl)hydrazinecarboxamide (Example 76A) were suspended in 125 ml of 2% strength aqueous sodium hydroxide solution and stirred under reflux for 6 h. After cooling, the mixture was acidified slowly with 6 ml of 1 N hydrochloric acid. The precipitated crystals were filtered off with suction, washed with water and dried under high vacuum. This gave 491 mg (97% of theory; purity 100%) of the title compound.


LC-MS (Method 2): Rt=3.43 min; MS (ESpos): m/z=568.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=13.30 (br. s, 1H), 11.63 (s, 1H), 7.52 (d, J=8.3 Hz, 1H), 7.41 (d, J=1.7 Hz, 1H), 7.10 (d, J=8.3 Hz, 1H), 6.87 (d, J=8.6 Hz, 1H), 6.49 (d, J=2.3 Hz, 1H), 6.40 (dd, J=8.4, 2.2 Hz, 1H), 4.65 (s, 2H), 3.86 (br. s, 2H), 3.83 (s, 2H), 3.75 (s, 3H), 3.71 (s, 3H).


EXAMPLE 78A
Methyl [5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetate



embedded image


Under argon and at 23° C., 1.13 g (20.89 mmol) of sodium methoxide were added to a solution of 3 g (19.66 mmol) of methyl 3-amino-3-iminopropanoate hydrochloride in 5 ml of methanol. The mixture was stirred at 23° C. for 15 min, and 0.84 g (2.46 mmol) of ethyl 2-(3,4-dichlorobenzyl)-4,4,4-trifluoro-3-oxobutanoate [CAS 179110-12-4; WO 2012/041817, Intermediate 56], dissolved in 5 ml of methanol, was then added. The mixture was stirred initially at 23° C. for 30 min and then under reflux for 16 h. The mixture was then applied to kieselguhr and purified directly by flash chromatography (40 g of silica gel, mobile phase cyclohexane/ethyl acetate). This gave 302 mg (26% of theory; purity 84%) of the title compound.


LC-MS (Method 1): Rt=1.13 min; MS (ESpos): m/z=395.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.67 (s, 3H), 3.79 (s, 2H), 3.92 (s, 2H), 7.13 (dd, 1H), 7.43 (d, 1H), 7.54 (d, 1H), 13.32 (br. s, 1H).


EXAMPLE 79A
2-[5-(3,4-Dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetohydrazide



embedded image


At 23° C., 557.4 mg (11.14 mmol) of hydrazine hydrate were added to a solution of 880 mg (2.22 mmol) of methyl [5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetate (Example 78A) in 52 ml of THF. The reaction mixture was stirred at 23° C. overnight and then concentrated. The residue was purified by preparative HPLC (column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient: 10:90→90:10). This gave 515 mg (57% of theory; purity 98%) of the title compound.


LC-MS (Method 1): Rt=0.94 min; MS (ESpos): m/z=395.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=9.29 (br. s, 1H), 6.99-7.66 (m, 3H), 4.04-4.89 (m, 1H), 3.91 (s, 2H), 3.49 (s, 2H).


EXAMPLE 80A
5-(3,4-Dichlorophenoxy)-2-(diethoxymethyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 7.05 g (11 mmol) of ethyl 2-(3,4-dichlorophenoxy)-4,4,4-trifluoro-3-oxobutanoate (Example 16A; purity 54%), 10.63 g (44.1 mmol) of 2,2-diethoxyethaneimidamide×sodium chloride (purity 85%) and 7.63 g (55.2 mmol) of potassium carbonate in 89 ml of dioxane was stirred at 85° C. for 1.5 h. After addition of 4 ml of 1 N hydrochloric acid, the reaction mixture was concentrated. The residue was dissolved in ethyl acetate and washed twice with water.


Chromatography on silica gel using the mobile phase cyclohexane/ethyl acetate (3:1) gave 2.7 g (45% of theory, purity 81%) of the title compound.


LC-MS (Method 1): Rt=1.27 min; MS (ESneg): m/z=425 (M−H)



1H-NMR (400 MHz, DMSO-d6): δ=1.20 (t, 6H), 3.60-3.78 (m, 4H), 5.31 (s, 1H), 7.14 (dd, 1H), 7.51 (d, 1H), 7.57 (d, 1H), 13.51 (br. s, 1H).


The following compound was prepared in an analogous manner:


EXAMPLE 81A
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-(diethoxymethyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


Yield: quantitative


LC-MS (Method 1): Rt=1.25 min; MS (ESpos): m/z=461 (M+H)+.


EXAMPLE 82A
5-(3,4-Dichlorophenoxy)-2-[(E)-(hydroxyimino)methyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


4.5 g (10.5 mmol) of 5-(3,4-dichlorophenoxy)-2-(diethoxymethyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 80A) and 0.88 g (12.6 mmol) of hydroxylammonium chloride in 15.5 ml of ethanol and 1.83 ml of water were stirred initially at 60° C. for 12 h and then at 85° C. for 12 h. Water was then added, and the mixture was extracted three times with ethyl acetate. The combined organic phases were washed with saturated sodium chloride solution, dried over magnesium sulphate, filtered and concentrated under reduced pressure. This gave 3.49 g (64% of theory) of the title compound in the form of a brown solid which was reacted further without further purification.


LC-MS (Method 1): Rt=1.08 min; MS (ESpos): m/z=368 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=7.16 (dd, 1H), 7.49 (d, 1H), 7.57 (d, 1H), 7.91 (s, 1H), 12.67 (s, 1H), 13.48 (br. s, 1H).


The following compound was prepared in an analogous manner:


EXAMPLE 83A
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-[(E)-(hydroxyimino)methyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


Yield: 84% of theory


LC-MS (Method 1): Rt=1.12 min; MS (ESpos): m/z=402.1 (M+H)+.


EXAMPLE 84A
5-(3,4-Dichlorophenoxy)-6-oxo-4-(trifluoromethy)-1,6-dihydropyrimidine-2-carbonitrile



embedded image


A mixture of 3.49 g (9.5 mmol) of 5-(3,4-dichlorophenoxy)-2-[(E)-(hydroxyimino)methyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 82A), 7.94 g (77.8 mmol) of acetic anhydride and 31 mg (0.4 mmol) of sodium acetate was heated under reflux for 1 h. After cooling, the solution was added to 800 ml of water, neutralized with potassium carbonate and extracted three times with ethyl acetate. The combined organic extracts were washed with saturated sodium chloride solution, clarified with activated carbon and, after filtration, dried with sodium sulphate and concentrated. Chromatography of the residue on silica gel (mobile phase: ethyl acetate) gave 1.67 g (50% of theory) of the title compound.


LC-MS (Method 1): Rt=1.09 min; MS (ESneg): m/z=347.9 (M−H)



1H-NMR (400 MHz, DMSO-d6): δ=7.10 (dd, 1H), 7.42 (d, 1H), 7.59 (d, 1H).


The following compound was prepared in an analogous manner:


EXAMPLE 85A
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carbonitrile



embedded image


Yield: 56% of theory


LC-MS (Method 1): Rt=1.02 min; MS (ESneg): m/z=381.9 (M−H).


EXAMPLE 86A
5-(3,4-Dichlorophenoxy)-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carboximidamide



embedded image


A mixture of 2.4 g (35 mmol) of hydroxylammonium chloride and 3.53 g (35 mmol) of triethylamine in 94 ml of DMSO was stirred for 10 minutes, and the precipitate was then filtered off with suction. 2.79 g (7 mmol) of 5-(3,4-dichlorophenoxy)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carbonitrile (Example 84A) were then added to the filtrate and the mixture was then stirred at 75° C. for 12 h. 1 litre of water was then added and the precipitate formed was filtered off with suction. The aqueous mother liquor was extracted three times with ethyl acetate, and the combined organic extracts were washed twice with saturated sodium chloride solution, dried with sodium sulphate and concentrated under reduced pressure. This gave 2.17 g (69% of theory) of the title compound.


LC-MS (Method 1): Rt=1.06 min; MS (ESpos): m/z=382.9 (M+H)+.


The following compound was prepared in an analogous manner:


EXAMPLE 87A
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carboximidamide



embedded image


Yield: 94% of theory


LC-MS (Method 1): Rt=1.09 min; MS (ESpos): m/z=417.0 (M+H)+.


The following compounds are known from the literature, commercially available or can be prepared analogously to Example 2A:











TABLE 11





Example




No.
IUPAC name/structure
Analytical data or CAS number







88A
ethyl [4-chloro-3- (trifluoromethoxy)phenoxy]acetate   embedded image
LC-MS (Method 1): Rt = 1.18 min; MS (ESneg): m/z = 297.1 (M − H)





89A
ethyl (4-chlorophenoxy)acetate   embedded image
CAS 14426-42-7





90A
ethyl (3-chloro-4-methylphenoxy)acetate   embedded image
LC-MS (Method 3): Rt = 2.38 min; MS (ESpos): m/z = 229.2 (M + H)+





91A
ethyl (4-chloro-3-methylphenoxy)acetate   embedded image
CAS 30406-61-2









The following synthesis intermediates were prepared analogously to Example 9A:











TABLE 12





Example
IUPAC name/structure



No.
(yield; reaction time)
Analytical data







92A
ethyl 2-[4-chloro-3- (trifluoromethoxy)phenoxy]-4,4,4-trifluoro-3- oxobutanoate   embedded image
LC-MS (Method 1): Rt = 1.07 min; MS (ESneg): m/z = 393.0 (M − H)





93A
ethyl 2-[4-chlorophenoxy]-4,4,4-trifluoro-3- oxobutanoate   embedded image
LC-MS (Method 1): Rt = 0.93 min; MS (ESneg): m/z = 309 (M − H)





94A
ethyl 2-(3-chloro-4-methylphenoxy)-4,4,4- trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 3): Rt = 2.28 min; MS (ESneg): m/z = 323.0 (M − H)





95A
ethyl 2-(4-chloro-3-methylphenoxy)-4,4,4- trifluoro-3-oxobutanoate   embedded image
LC-MS (Method 3): Rt = 2.28 min; MS (ESneg): m/z = 323.0 (M − H)









EXAMPLE 96A
3-(Methylsulphanyl)pyridine-2-carboximidamide



embedded image


Step 1:


5 g (40.9 mmol) of 3-fluoropyridine-2-carbonitrile were dissolved in 40 ml of N,N-dimethylformamide. 3.2 g (45 mmol) of sodium thiomethoxide were then added slowly at room temperature. The mixture was stirred at room temperature for 2 h and then poured onto 500 ml of water. This resulted in the precipitation of a solid which was filtered off and washed with water. Drying under high vacuum gave 5.1 g (79% of theory) of the intermediate 3-(methylsulphanyl)pyridine-2-carbonitrile.


Step 2:


Under an atmosphere of argon, 4.5 g (83 mmol) of ammonium chloride were initially charged in 100 ml of toluene, and the mixture was cooled to 0° C. 36.6 ml (73 mmol) of a 2 M solution of trimethylaluminium in toluene were then added, and the reaction mixture was, with stirring, slowly warmed to room temperature. After the evolution of gas had ceased, 5.0 g (33 mmol) of 3-(methylsulphanyl)pyridine-2-carbonitrile were added and the reaction mixture was stirred at 80° C. for 14 h. After cooling to room temperature, 50 ml of methanol were added a little at a time at 0° C., followed by 40 ml of a methanol/water mixture (4:1). The resulting mixture was then stirred at room temperature for 2 h. The precipitate formed was filtered off with suction and washed with methanol and methyl tert-butyl ether. The mother liquor was concentrated under reduced pressure, 500 ml of dichloromethane/methanol (5:1) were added to the residue and the mixture was filtered again. The filtrate was finally concentrated under reduced pressure. This gave 5.0 g (purity 87%, 78% of theory) of 3-(methylsulphanyl)pyridine-2-carboximidamide.


LC-MS (Method 4): Rt=0.93 min; MS (ESpos): m/z=168 (M+H)+


Analogously to Example 96A/Step 2, the compounds listed in Table 13 were prepared from the corresponding nitriles:











TABLE 13





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







97A
5-methoxypyrimidine-4-carboximidamide   embedded image
  (91% of theory; reaction time: 14 h, 80° C.; 2 eq. of trimethylaluminium, 2 eq. of ammonium chloride; 1 eq. of 5-methoxypyrimidine-4-carbonitrile, CAS 114969-64-1)

LC-MS (Method 4): Rt = 0.48 min; (ESpos): m/z = 153.1 (M + H)+





98A
6-aminopyridazine-3-carboximidamide   embedded image
  (96% of theory; reaction time: 14 h, 80° C.; 2 eq. of trimethylaluminium, 2 eq. of ammonium chloride; 1 eq. of 6-aminopyridazine-3-carbonitrile, CAS 340759-46-8)

LC-MS (Method 3): Rt = 0.85 min; MS (ESIpos): m/z = 138.3 (M + H)+





99A
4,6-dimethoxypyrimidine-2-carboximidamide   embedded image
  (71% of theory; reaction time: 14 h, 80° C.; 2.2 eq. of trimethylaluminium, 2.5 eq. of ammonium chloride; 1 eq. of 4,6-dimethoxypyrimidine-2- carbonitrile, CAS 139539-63-2)

LC-MS (Method 6): Rt = 0.35 min; MS (ESpos): m/z = 183.2 (M + H)+





100A
5-aminopyridine-2-carboximidamide   embedded image
  (15% of theory; reaction time: 14 h, 80° C.; 2 eq. of trimethylaluminium, 2 eq. of ammonium chloride; 1 eq. of 5-aminopyridine-2-carbonitrile, CAS 55338-73-3)

LC-MS (Method 4): Rt = 0.25 min; MS (ESneg): m/z = 135.3 (M − H)





101A
1-methyl-1H-indazole-3-carboximidamide   embedded image
  (89% of theory; reaction time: 14 h, 80° C.; 2 eq. of trimethylaluminium, 2 eq. of ammonium chloride; 1 eq. of 1-methyl-1H-indazole-3-carbonitrile, CAS 31748-44-4)

LC-MS (Method 6): Rt = 0.25 min; (ESpos): m/z = 175.2 (M + H)+





102A
4-chloro-1-methyl-1H-indazole-3- carboximidamide   embedded image
  (72% of theory; reaction time: 14 h, 80° C.; 2 eq. of trimethylaluminium, 2 eq. of ammonium chloride; 1 eq. of 4-chloro-1-methyl-1H-indazole-3- carbonitrile, CAS 1264481-55-1)

LC-MS (Method 6): Rt = 0.30 min; MS (ESpos): m/z = 209.2 (M + H)+





103A
4,5-dichloro-1,2-thiazole-3-carboximidamide   embedded image
  (65% of theory; reaction time: 16 h, 80° C.; 2 eq. of trimethylaluminium, 2 eq. of ammonium chloride; 1 eq. of 4,5-dichloro-1,2-thiazole-3-carbonitrile, CAS 1137210-71-9)

LC-MS (Method 4): Rt = 1.57 min; MS (ESpos): m/z = 196.0 (M + H)+





104A
3-(trifluoromethoxy)pyridine-2-carboximidamide   embedded image
  (19% of theory; reaction time: 18 h, 100° C.; 2.5 eq. of trimethylaluminium, 4 eq. of ammonium chloride; 1 eq. of 3-(trifluoromethoxy)pyridine-2- carbonitrile, CAS 1206983-47-2)


1H-NMR (400 MHz, DMSO-d6): δ = 7.27 (br. s, 1H), 7.90 (dd, 1H), 8.22 (d, 1H), 8.79 (d, 1H), 9.66 (br. s, 2H).






105A
N-(6-Carbamimidoylpyridin-3-yl)acetamide   embedded image
  (11% of theory; reaction time: 18 h, 100° C.; 2.5 eq. of trimethylaluminium, 4 eq. of ammonium chloride; 1 eq. of N-(6-cyanopyridin-3-yl)acetamide, CAS 1223587-77-6)

LC-MS (Method 4): Rt = 0.97 min; MS (ESpos): m/z = 179.1 (M + H)+





106A
4,5-dimethylpyridine-2-carboximidamide   embedded image
  (30% of theory; reaction time: 14 h, 80° C.; 2 eq. of trimethylaluminium, 2 eq. of ammonium chloride; 1 eq. of 4,5-dimethylpyridine-2-carbonitrile, CAS 24559-31-7)

LC-MS (Method 6): Rt = 0.25 min; MS (ESpos): m/z = 150.2 (M + H)+









EXAMPLE 107A
4-Methoxy-1H-pyrazole-3-carboximidamide hydrochloride



embedded image


Step 1:


At 20° C., 88 g (1.1 mol) of sodium acetate were added in one portion to a mixture of 60 g (536 mmol) of hydrazinecarboxamide hydrochloride and 375 g (3.2 mol) of ethyl 2-oxopropanoate in 300 ml of water. The reaction mixture was stirred at 20° C. for 2 h. The precipitate formed was filtered off, washed with water and dried. This gave 79 g (86% of theory) of the intermediate ethyl 2-(carbamoylhydrazone)propanoate as a white solid.


Step 2:


At from −5° C. to 0° C. and under an atmosphere of nitrogen, 81 g (526 mmol) of phosphoryl chloride were added dropwise to 120 ml of N,N-dimethylformamide. After 20 min of stirring at 0° C., 30 g (173 mmol) of ethyl 2-(carbamoylhydrazono)propanoate were added a little at a time at from 0° C. 15 to 50 C over a period of 20 min. The reaction mixture was stirred at 60° C. for 1 h and then at 80° C. for 3 h. After cooling to room temperature, the reaction mixture was hydrolysed carefully by addition of 600 ml of ice-water and then adjusted to pH 10 by addition of sodium hydroxide. The reaction was stirred at 50° C. for 5 min, then cooled to 0° C. using an ice/water bath and adjusted to pH 7 by addition of 10 M hydrochloric acid. The mixture was then extracted three times with 500 ml of ethyl acetate each time, and the combined organic phases were washed with saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 30 g of ethyl 4-formyl-1H-pyrazole-3-carboxylate which was used without further purification in the next step.


Step 3:


At 0° C. and under an atmosphere of nitrogen, 3,4-dihydro-2H-pyran (22.5μ, 268 mmol) were added to a mixture of 30 g (178 mmol) of ethyl 4-formyl-1H-pyrazole-3-carboxylate and p-toluenesulphonic acid (3.4μ, 19.6 mmol) in dichloromethane (300 ml). The reaction mixture was stirred at 12° C. for 15 h. The mixture was then diluted with 300 ml of dichloromethane and adjusted to pH 8 with saturated aqueous sodium bicarbonate solution. After separation of the phases, the aqueous phase was extracted twice with 200 ml of dichloromethane each time. The combined organic phases were washed with saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave a residue which was purified by silica gel chromatography (mobile phase petroleum ether/ethyl acetate 30:1). This gave 23 g (51% of theory) of the intermediate ethyl 4-formyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylate as an oil and a further fraction which was still contaminated (7.5μ, purity 80%) of this intermediate.



1H-NMR (400 MHz, CDCl3): δ=1.45 (t, 3H), 1.59-1.65 (m, 3H), 1.68-2.05 (m, 2H), 2.16-2.18 (m, 1H), 3.71-3.74 (m, 1H), 4.09-4.14 (m, 1H), 4.46-4.51 (m, 2H), 5.47-5.51 (m, 1H), 8.25 (s, 1H), 10.41 (s, 1H).


Step 4:


At 0° C. and under an atmosphere of nitrogen, meta-chloroperbenzoic acid (31.5μ, 155 mmol) was added to 23 g (91 mmol) of ethyl 4-formyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylate in dichloromethane (250 ml). The reaction mixture was stirred initially at 15° C. for 15 h and then at 25° C. for 13 h. The reaction mixture was then diluted with dichloromethane (300 ml) and washed twice with 300 ml each of a saturated aqueous sodium thiosulphate solution and a saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave a residue which was purified by silica gel chromatography (mobile phase petroleum ether/ethyl acetate 15:1). This gave 12 g (purity 60%) of the intermediate ethyl 4-hydroxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylate as an oil which was used without further purification in the next step.


Step 5:


At 12° C. and under an atmosphere of nitrogen, methyl iodide (7.3μ, 51.4 mmol) was added to a mixture of 11.9 g (29.7 mmol) of ethyl 4-hydroxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylate and potassium carbonate (8.2μ, 59.4 mmol) in N,N-dimethylformamide (100 ml). The reaction mixture was stirred at 12° C. for 13 h. The reaction mixture was then cooled to 0° C., and 1 ml of methanol was added. The mixture was stirred at 12° C. for 10 min and then diluted with ethyl acetate (300 ml) and water (400 ml). After separation of the phases, the aqueous phase was extracted twice with 200 ml of ethyl acetate each time. The combined organic phases were washed with saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave a residue which was purified by silica gel chromatography (mobile phase petroleum ether/ethyl acetate 10:1). This gave 6.2 g (82% of theory) of the intermediate ethyl 4-methoxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylate as an oil.



1H-NMR (400 MHz, CDCl3): δ=1.39 (t, 3H), 1.62-1.67 (m, 3H), 1.97-2.07 (m, 3H), 3.68-3.71 (m, 1H), 3.84 (s, 3H), 4.06-4.09 (m, 1H), 4.29-4.33 (m, 2H), 5.37-5.40 (m, 1H), 7.33 (s, 1H).


Step 6:


Under an atmosphere of nitrogen, 4.8 g (90.4 mmol) of ammonium chloride were initially charged in 180 ml of toluene, and the mixture was cooled to 0° C. 45.2 ml (90.5 mmol) of a 2 M solution of trimethylaluminium in toluene were then added over a period of 30 min, and the reaction mixture was, with stirring, slowly warmed to room temperature. After the evolution of gas had ceased, 4.6 g (18.1 mmol) of ethyl 4-methoxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylate, dissolved in 20 ml of toluene, were added dropwise and the reaction mixture was stirred at 80° C. for 20 h. After cooling to room temperature, 100 ml of methanol were added a little at a time at 0° C., and the mixture was stirred at 12° C. for 1 h. The precipitate formed was filtered off with suction and washed twice with 50 ml of methanol each time. The filtrate was then concentrated under reduced pressure. This gave a residue which was purified by silica gel chromatography (mobile phase dichloromethane→dichloromethane/methanol 15:1). This gave 3.8 g (94% of theory) of the intermediate 4-methoxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboximidamide as a white solid.



1H-NMR (400 MHz, CDCl3): δ=1.54-1.56 (m, 2H), 1.66-1.70 (m, 1H), 1.92-1.95 (m, 2H), 2.09-2.12 (m, 1H), 3.62-3.68 (m, 1H), 3.84 (s, 3H), 3.92-3.95 (m, 1H), 5.41-5.43 (m, 1H), 8.09 (s, 1H), 8.72 (br. s, 3H).


Step 7:


A mixture of 2 g (8.92 mmol) of 4-methoxy-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboximidamide in hydrogen chloride/methanol (4 M solution, 50 ml) was stirred at 13° C. for 13 h. The reaction mixture was then concentrated under reduced pressure. This gave 1.29 g (82% of theory) of 4-methoxy-1H-pyrazole-3-carboximidamide hydrochloride as a solid.



1H-NMR (400 MHz, CDCl3): δ=3.84 (s, 3H), 7.89 (s, 1H), 8.57 (br. s, 2H), 9.01 (br. s, 2H), 13.90 (br. s, 1H).


EXAMPLE 108A
5-Ethyl-1,2-oxazole-3-carboximidamide



embedded image


Step 1:


0.42 g (3 mmol) of 5-ethyl-1,2-oxazole-3-carboxylic acid (CAS 52320-59-9) were dissolved in 6 ml of ethanol, and a catalytic amount of conc. sulphuric acid was added. After 3 h of stirring at 80° C., the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was taken up in ethyl acetate and washed with saturated aqueous sodium bicarbonate solution, and the organic phase was dried over sodium sulphate. After removal of the drying agent by filtration, the mixture was concentrated under reduced pressure. This gave 0.42 g (82% of theory) of the intermediate ethyl 5-ethyl-1,2-oxazole-3-carboxylate which was used without further purification in the next step.


Step 2:


Under an atmosphere of argon, 0.791 g (14.8 mmol) of ammonium chloride were initially charged in 6 ml of toluene, and the mixture was cooled to 0° C. 14.8 ml (9.9 mmol) of a 2 M solution of trimethylaluminium in toluene were then added, and the reaction mixture was, with stirring, slowly warmed to room temperature. After the evolution of gas had ceased, 0.42 g (2.47 mmol) of ethyl 5-ethyl-1,2-oxazole-3-carboxylate were added and the reaction mixture was stirred at 80° C. for 14 h. After cooling to room temperature, 50 ml of methanol were added a little at a time at 0° C., followed by 40 ml of a methanol/water mixture (4:1). The resulting mixture was then stirred at room temperature for 2 h. The precipitate formed was filtered off with suction and washed with methanol and methyl tert-butyl ether. The mother liquor was concentrated under reduced pressure, 500 ml of dichloromethane/methanol (5:1) were added to the residue and the mixture was filtered again. The filtrate was finally concentrated under reduced pressure. This gave 48 mg (purity 96%, 14% of theory) of 5-ethyl-1,2-oxazole-3-carboximidamide.


LC-MS (Method 4): Rt=1.27 min; MS (ESpos): m/z=140.0 (M+H)+.


Analogously to Example 108A/Step 2, the compounds listed in Table 14 were prepared from the corresponding carboxylic esters:











TABLE 14





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







109A
6-(dimethylamino)pyridazine-3- carboximidamide   embedded image
  (23% of theory; reaction time: 16 h, 80° C.; 2.5 eq. of trimethylaluminium, 4 eq. of ammonium chloride; 1 eq. of ethyl 6-(dimethylamino)pyridazine-3- carboxylate, CAS 64210-62-4)

LC-MS (Method 3): Rt = 0.37 min; MS (ESpos): m/z = 166.1 (M + H)+





110A
5-propyl-1,2-oxazole-3-carboximidamide   embedded image
  (16% of theory; reaction time: 16 h, 80° C.; 4 eq. of trimethylaluminium, 6 eq. of ammonium chloride; 1 eq. of ethyl 5-propyl-1,2-oxazole-3- carboxylate, CAS 91240-31-2)

LC-MS (Method 4): Rt = 1.57 min; MS (ESpos): m/z = 154.0 (M + H)+





111A
5-cyclopropyl-1,2-oxazole-3-carboximidamide   embedded image
  (62% of theory; reaction time: 16 h, 80° C.; 4 eq. of trimethylaluminium, 6 eq. of ammonium chloride; 1 eq. of ethyl 5-cyclopropyl-1,2-oxazole-3- carboxylate, CAS 21080-81-9)

LC-MS (Method 4): Rt = 1.43 min; MS (ESpos): m/z = 152.0 (M + H)+





112A
1-ethyl-1H-pyrazole-3-carboximidamide   embedded image
  (40% of theory; reaction time: 16 h, 80° C.; 2.5 eq. of trimethylaluminium, 4 eq. of ammonium chloride; 1 eq. of ethyl 1-ethyl-1H-pyrazole-3-carboxylate, CAS 1007503-15-2)

LC-MS (Method 4): Rt = 0.87 min; MS (ESpos): m/z = 138.1 (M + H)+





113A
6-methoxy-1,2-benzoxazole-3-carboximidamide   embedded image
  (4% of theory; reaction time: 16 h, 80° C.; 2.5 eq. of trimethylaluminium, 4 eq. of ammonium chloride; 1 eq. of ethyl 6-methoxy-1,2-benzoxazole-3- carboxylate, CAS 57764-51-9)

LC-MS (Method 4): Rt = 1.37 min; MS (ESpos): m/z = 219.1 (M + H)+









EXAMPLE 114A
5-(Methoxymethyl)-1,2-oxazole-3-carboximidamide



embedded image


Step 1:


1.0 g (5.8 mmol) of ethyl 5-(hydroxymethyl)-1,2-oxazole-3-carboxylate (CAS 123770-62-7) was dissolved in 5 ml of THF. At 0° C., 0.28 g (7.0 mmol) of sodium hydride (60% in paraffin) was added, resulting in evolution of hydrogen in an exothermic reaction. After 1 h of stirring at 23° C., 0.91 g (6.43 mmol) of iodomethane was added, and the mixture was stirred at 23° C. for another 18 h. The mixture was then diluted with ethyl acetate, the solution was washed with water and 1 N aqueous sodium hydroxide solution, the organic phase was dried over sodium sulphate and the drying agent was filtered off. Concentration under reduced pressure and drying of the residue under high vacuum gave 0.38 g (33% of theory, purity 95%) of the intermediate ethyl 5-(methoxymethyl)-1,2-oxazole-3-carboxylate which was used without further purification in the next reaction.


Step 2:


Under an atmosphere of argon, 0.440 g (8.2 mmol) of ammonium chloride were initially charged in 5 ml of toluene, and the mixture was cooled to 0° C. 2.6 ml (5.1 mmol) of a 2 M solution of trimethylaluminium in toluene were then added, and the reaction mixture was, with stirring, slowly warmed to room temperature. After the evolution of gas had ceased, 0.38 g (2.06 mmol) of ethyl 5-(methoxymethyl)-1,2-oxazole-3-carboxylate were added and the reaction mixture was stirred at 80° C. for 48 h. After cooling to room temperature, 50 ml of methanol were added a little at a time at 0° C., followed by 40 ml of a methanol/water mixture (4:1). The resulting mixture was then stirred at room temperature for 2 h. The precipitate formed was filtered off with suction and washed with methanol. The mother liquor was concentrated under reduced pressure, 500 ml of dichloromethane/methanol (5:1) were added to the residue and the mixture was filtered again. The filtrate was finally concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (mobile phase gradient dichloromethane/methanol 1:1→methanol). This gave 120 mg (38% of theory) of 5-(methoxymethyl)-1,2-oxazole-3-carboximidamide.


LC-MS (Method 4): Rt=1.02 min; MS (ESpos): m/z=156.0 (M+H)+.


EXAMPLE 115A
tert-Butyl 4-{5-[4-chloro-3-(trifluoromethyl)phenoxy]-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl}piperidine-1-carboxylate



embedded image


A mixture of 146 mg (1.06 mmol) of potassium carbonate, 144 mg (0.63 mmol) of tert-butyl 4-carbamimidoylpiperidine-1-carboxylate (CAS 885270-23-5) and 100 mg (0.2 mmol) of ethyl 2-[3-chloro-4-(trifluoromethyl)phenoxy]-4,4,4-trifluoro-3-oxobutanoate (Example 9A) in 1.7 ml of dioxane was heated under reflux for 2 h. The mixture was then poured onto water/ethyl acetate and the organic phase was separated off, dried and concentrated. The residue was purified by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% trifluoroacetic acid). 88 mg (77% of theory) of the title compound were obtained.


LC-MS (Method 6): Rt=1.60 min; MS (ESneg): m/z=540.0 (M−H)



1H-NMR (400 MHz, DMSO-d6): δ=1.41 (s, 9H), 1.62-1.65 (m, 2H), 1.87-1.90 (m, 2H), 2.79-2.85 (m, 3H), 4.02-4.05 (m, 2H), 7.38 (dd, 1H), 7.55 (d, 1H), 7.67 (d, 1H), 13.4 (br. s, 1H).


EXAMPLE 116A
5-(3,4-Dichlorobenzyl)-2-(hydroxymethyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 25 g (75 mmol) of ethyl 2-(3,4-dichlorobenzyl)-4,4,4-trifluoro-3-oxobutanoate (CAS 179110-12-4; WO 2012/041817, Intermediate 56), 10 g (90 mmol) of 2-hydroxyethanimidamide hydrochloride and 19.7 ml (113 mmol) of N,N-diisopropylethylamine in 250 ml of DMF was stirred at 100° C. for 3 h. The mixture was then concentrated on a rotary evaporator to half of its original volume and then diluted with ethyl acetate and extracted with water. The organic phase was dried over magnesium sulphate. After filtration and concentration, the residue was purified chromatographically on silica gel (mobile phase cyclohexane/ethyl acetate 3:1-1:1). 9.69 g (36% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=1.03 min; MS (ESpos): m/z=353.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.92 (s, 2H), 4.38 (d, 2H), 5.74 (t, 1H), 7.13 (dd, 1H), 7.35-7.62 (m, 2H), 12.96 (br. s, 1H).


EXAMPLE 117A
5-(3,4-Dichlorobenzyl)-2-{[4-(2,4-dimethoxybenzyl)-1-methyl-5-oxo-4,5-dihydro-1H-1,2,4-triazol-3-yl]methyl}-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 190 mg (0.27 mmol) of 5-(3,4-dichlorobenzyl)-2-{[4-(2,4-dimethoxybenzyl)-5-oxo-4,5-dihydro-1H-1,2,4-triazol-3-yl]methyl}-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 77A) and 33 mg (0.30 mmol) of potassium tert-butoxide in 1 ml of N,N-dimethylformamide was stirred at 23° C. for 15 min. A solution of 19 μl (0.30 mmol) of iodomethane in 0.9 ml of N,N-dimethylformamide was then added. The mixture was stirred at 23° C. for 24 h and the reaction mixture was then concentrated. The residue was purified by preparative HPLC (column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient 90:10-10:90). 106 mg (54% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=1.45 min; MS (ESpos): m/z=704.2 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.38 (s, 3H), 3.66 (s, 3H), 3.67 (s, 3H), 3.73 (s, 3H), 3.89 (s, 2H), 4.19 (s, 2H), 4.67 (s, 2H), 5.18 (s, 2H), 6.18 (dd, 1H), 6.39 (d, 1H), 6.59 (d, 1H), 6.82 (d, 2H), 6.93 (dd, 1H), 7.09 (d, 2H), 7.29 (d, 1H), 7.47 (d, 1H).


EXAMPLE 118A
2-(3-Nitro-1H-pyrazol-1-yl]-5-(3,4-dichlorophenoxy)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


Under argon, 112 mg of 5-nitro-1H-pyrazole (0.99 mmol) and five pellets of molecular sieve (4A) were initially charged in 3 ml of dioxane, the mixture was cooled to −78° C. and 30 μl of glacial acetic acid were added. The mixture was then warmed to 0° C. and 200 mg (0.248 mmol) of 5-(3,4-dichlorophenoxy)-2-(methylsulphonyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 38A) were added. In a microwave apparatus, the reaction mixture was heated at 150° C. for 4 h. The mixture was then filtered and purified directly by preparative HPLC (column: Chromatorex C18, 10 μm, 30×125 mm; mobile phase: acetonitrile/0.1% aq. TFA). Lyophilization of the product fractions gave 92 mg (42% of theory) of the title compound.


LC-MS (Method 3): Rt=2.26 min; MS (ESpos): m/z=436.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=6.87 (m, 1H), 7.10 (d, 1H), 7.18 (d, 1H), 7.51 (m, 1H), 8.60 (s, 1H).


EXAMPLE 119A
N-(3-Carbamimidoyl-1,2-benzoxazol-6-yl)acetamide



embedded image


Step 1:


1.0 g (5.2 mmol) of methyl 6-amino-1,2-benzoxazole-3-carboxylate (CAS 57764-47-3) was dissolved in 15 ml of dichloromethane, and 0.49 ml (5.2 mmol) of acetic anhydride was added at 0° C. After 16 h of stirring at 23° C., the mixture was diluted with dichloromethane and washed with water. The organic phase was dried over sodium sulphate and the drying agent was filtered off. Concentration under reduced pressure and drying of the residue under high vacuum gave 1.07 g (79% of theory, purity 89%) of the intermediate methyl 6-acetamido-1,2-benzoxazole-3-carboxylate which was used without further purification in the next step.


LC-MS (Method 1): Rt=0.68 min; MS (ESpos): m/z=235.1 (M+H)+.


Step 2:


0.5 g (2.1 mmol) of methyl 6-acetamido-1,2-benzoxazole-3-carboxylate was dissolved in 5 ml of methanol, and 20 ml of ammonia solution (35% in water) were added at 23° C. After 16 h of stirring at 23° C., the mixture was concentrated to a volume of about 10 ml and the precipitated solid was filtered off. Drying under high vacuum gave 0.47 g (100% of theory) of the intermediate 6-acetamido-1,2-benzoxazole-3-carboxamide.


LC-MS (Method 1): Rt=0.49 min; MS (ESpos): m/z=220.1 (M+H)+.


Step 3:


0.44 g (2.0 mmol) of 6-acetamido-1,2-benzoxazole-3-carboxamide was dissolved in 20 ml of THF, and 3.8 g (6.0 mmol) of propanephosphonic acid cyclic anhydride (as a 50% by weight solution in ethyl acetate) and 1.0 ml (6.0 mmol) of N,N-diisopropylethylamine were added at 23° C. After 1 h of stirring at 120° C. in a microwave apparatus, the mixture was diluted with ethyl acetate and washed with water. The organic phase was dried over sodium sulphate and the drying agent was filtered off. After concentration under reduced pressure, the solid obtained was crystallized from pentane and diethyl ether. After drying under a high vacuum, 0.3 g (73% of theory, purity 97%) of the intermediate N-(3-cyano-1,2-benzoxazol-6-yl)acetamide was obtained.


LC-MS (Method 1): Rt=0.75 min; MS (ESneg): m/z=200.1 (M−H).


Step 4:


Under an atmosphere of argon, 0.452 g (8.5 mmol) of ammonium chloride were initially charged in 10 ml of toluene, and the mixture was cooled to 0° C. 2.6 ml (5.3 mmol) of a 2 M solution of trimethylaluminium in toluene were then added, and the reaction mixture was, with stirring, slowly warmed to room temperature. After the evolution of gas had ceased, 0.425 g (2.11 mmol) of N-(3-cyano-1,2-benzoxazol-6-yl)acetamide was added and the reaction mixture was stirred at 100° C. for 48 h. After cooling to room temperature, 50 ml of methanol were added a little at a time at 0° C., followed by 40 ml of a methanol/water mixture (4:1). The resulting mixture was then stirred at room temperature for 2 h. The precipitate formed was filtered off with suction and washed with methanol. The mother liquor was concentrated under reduced pressure, 500 ml of dichloromethane/methanol (5:1) were added to the residue and the mixture was filtered again. The filtrate was finally concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (mobile phase gradient dichloromethane/methanol 1:1→methanol). This gave 20 mg (4% of theory; purity 100%) of the target compound N-(3-carbamimidoyl-1,2-benzoxazol-6-yl)acetamide.


LC-MS (Method 4): Rt=1.37 min; MS (ESpos): m/z=219.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=2.15 (s, 3H), 7.55 (dd, 1H), 7.91 (d, 1H), 8.41 (s, 1H), 9.55-10.12 (m, 3H), 10.66 (s, 1H).


Analogously to Example 108A, the compound listed in Table 15 was prepared from the corresponding carboxylic ester:











TABLE 15





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







120A
1-Methyl-1H-pyrazolo[3,4-b]pyridine-3- carboximidamide   embedded image
  (24% of theory; reaction time: 16 h, 80° C.; 2.5 eq. of trimethylaluminium, 4 eq. of ammonium chloride; 1 eq. of ethyl 1-methyl-1H-pyrazolo[3,4- b]pyridine-3-carboxylate,CAS 1367752-05-3)

LC-MS (Method 4): Rt = 1.20 min; MS (ESpos): m/z = 176.1 (M + H)+









EXAMPLE 121A
4-Methoxy-1,2-oxazole-3-carboximidamide hydrochloride



embedded image


Step 1:


5.0 g (38.4 mmol) of ethyl 3-oxobutanoate were dissolved in 30 ml of acetic acid and cooled to 0° C. 6.15 g (38.4 mmol) of bromine were added slowly and the reaction mixture was then stirred at 0° C. for 2 h. Water was then added, and the reaction mixture was extracted twice with ethyl acetate. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 6.0 g (75% of theory) of the intermediate ethyl 4-bromo-3-oxobutanoate as an oil.


Step 2:


2.16 g (31.6 mmol) of sodium nitrite, dissolved in 20 ml of water, were added to a solution, stirred at 0° C., of 6.0 g (28.7 mmol) of ethyl 4-bromo-3-oxobutanoate in 40 ml of acetic acid. The reaction mixture was then stirred at room temperature for 2 h. After addition of water, the mixture was extracted twice with diethyl ether. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 6.0 g (88% of theory) of the intermediate ethyl 4-bromo-2-(hydroximino)-3-oxobutanoate as a colourless liquid.


Step 3:


12.0 g (201 mmol) of urea were added to a stirred solution of 6.0 g (25.2 mmol) of ethyl 4-bromo-2-(hydroximino)-3-oxobutanoate in 100 ml of N,N-dimethylformamide, and the reaction mixture was then heated at 100° C. for 4 h. After cooling to room temperature and addition of water, the mixture was extracted twice with ethyl acetate. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. After drying over sodium sulphate and removal of the solvent, the residue was purified by column chromatography (silica gel, mobile phase petroleum ether/ethyl acetate 80:20). This gave 3.0 g (76% of theory) of the intermediate ethyl 4-hydroxy-1,2-oxazole-3-carboxylate as a white solid.



1H-NMR (400 MHz, CDCl3): δ=1.43 (t, 3H), 4.51 (q, 2H), 6.69 (s, 1H), 8.33 (s, 1H).


Step 4:


At 0° C., 13.5 g (95.5 mmol) of methyl iodide were added to a solution of 5.0 g (31.8 mmol) of ethyl 4-hydroxy-1,2-oxazole-3-carboxylate and 13.1 g (95.5 mmol) of potassium carbonate in 300 ml of acetone. The reaction mixture was then stirred at room temperature for 16 h. After addition of water, the mixture was extracted twice with ethyl acetate. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 5.0 g (92% of theory) of the intermediate ethyl 4-methoxy-1,2-oxazole-3-carboxylate as a white solid.


Step 5:


Under an atmosphere of nitrogen, 14.9 g (280.7 mmol) of ammonium chloride were initially charged in 150 ml of toluene, and the mixture was cooled to 0° C. 93.6 ml (187.1 mmol) of a 2 M solution of trimethylaluminium in toluene were then added over a period of 30 min, and the reaction mixture was, with stirring, slowly warmed to room temperature. After the evolution of gas had ceased, 8.0 g (46.7 mmol) of ethyl 4-methoxy-1,2-oxazole-3-carboxylate, dissolved in 20 ml of toluene, were added dropwise and the reaction mixture was stirred at 80° C. for another 16 h. After cooling to room temperature, 100 ml of methanol were added a little at a time at 0° C., and the mixture was stirred at room temperature for 1 h. The precipitate formed was filtered off with suction and washed twice with 50 ml of methanol each time. The filtrate was then concentrated under reduced pressure and the residue was recrystallized from acetonitrile. This gave 1.1 g (17% of theory) of the title compound 4-methoxy-1,2-oxazole-3-carboximidamide hydrochloride as a white solid.



1H-NMR (400 MHz, DMSO-d6): δ=3.84 (s, 3H), 9.23 (s, 1H), 9.54-9.61 (m, 4H).


EXAMPLE 122A
4-Methoxy-1,2-thiazole-3-carboximidamide hydrochloride



embedded image


Step 1:


73.4 g (1064 mmol) of sodium nitrite, dissolved in 520 ml of water, were added to a solution, stirred at 0° C., of 100 g (769 mmol) of ethyl 3-oxobutanoate in 250 ml of acetic acid. The reaction mixture was stirred at room temperature for 1 h. After addition of water, the mixture was extracted twice with ethyl acetate. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 105 g (86% of theory) of the intermediate ethyl 2-(hydroximino)-3-oxobutanoate as a colourless liquid.


Step 2:


3.2 g of palladium on activated carbon (10%) were added to a solution of 100 g (629 mmol) of ethyl 2-(hydroximino)-3-oxobutanoate in 900 ml of acetic acid and 307 ml of acetic anhydride, and the mixture was hydrogenated (60 psi H2) at room temperature for 3 h. The reaction mixture was then filtered through Celite and the filtercake was washed with acetic acid. The combined organic phase was concentrated and the residue was taken up in ethyl acetate. The organic phase was washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 110 g (93% of theory) of the intermediate ethyl 2-acetamido-3-oxobutanoate as a colourless liquid.


Step 3:


50.0 g (267 mmol) of ethyl 2-acetamido-3-oxobutanoate were dissolved in 400 ml of chloroform and cooled to 0° C. 43.0 g (267 mmol) of bromine were added slowly and the reaction mixture was then stirred at room temperature for 20 h. Water was then added, and the reaction mixture was extracted twice with ethyl acetate. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 70.0 g (98% of theory) of the intermediate ethyl 2-acetamido-4-brom-3-oxobutanoate as a solid.


Step 4:


At room temperature, 38 g (414 mmol) of thioacetic acid and 100 g (376 mmol) of ethyl 2-acetamido-4-bromo-3-oxobutanoate were added to a stirred solution of 23.1 g (414 mmol) of potassium hydroxide in 1000 ml of ethanol. The reaction mixture was stirred at room temperature for 4 h. Water was then added, and the mixture was extracted twice with ethyl acetate. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 90.0 g (92% of theory) of the intermediate ethyl N,S-diacetyl-3-oxohomocysteinate as a colourless liquid.


Step 5:


70.0 g (268 mmol) of ethyl N,S-diacetyl-3-oxohomocysteinate were dissolved in 2200 ml of chloroform and cooled to 0° C. 85.7 g (536 mmol) of bromine were added slowly and the reaction mixture was then stirred at room temperature for 20 h. Water was then added, and the reaction mixture was extracted twice with ethyl acetate. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave a residue which was recrystallized from petroleum ether/ethyl acetate. This gave 25.0 g (54% of theory) of the intermediate ethyl 4-hydroxy-1,2-thiazole-3-carboxylate as a white solid.


Step 6:


At room temperature, 24.5 g (173.2 mmol) of methyl iodide were added to a solution of 10.0 g (57.7 mmol) of ethyl 4-hydroxy-1,2-thiazole-3-carboxylate and 23.9 g (173.2 mmol) of potassium carbonate in 300 ml of acetone. The reaction mixture was stirred at room temperature for 4 h. After addition of water, the mixture was extracted twice with ethyl acetate. The combined organic phases were washed with water and saturated aqueous sodium chloride solution. Drying over sodium sulphate and removal of the solvent gave 10.0 g (93% of theory) of the intermediate ethyl 4-methoxy-1,2-thiazole-3-carboxylate as a colourless liquid.


Step 7:


Under an atmosphere of nitrogen, 17.0 g (320.8 mmol) of ammonium chloride were initially charged in 150 ml of toluene, and the mixture was cooled to 0° C. 107 ml (213.9 mmol) of a 2 M solution of trimethylaluminium in toluene were then added over a period of 30 min, and the reaction mixture was, with stirring, slowly warmed to room temperature. After the evolution of gas had ceased, 10.0 g (53.5 mmol) of ethyl 4-methoxy-1,2-thiazole-3-carboxylate, dissolved in 20 ml of toluene, were added dropwise and the reaction mixture was stirred at 80° C. for another 16 h. After cooling to room temperature, 100 ml of methanol were added a little at a time at 0° C., and the mixture was stirred at room temperature for 1 h. The precipitate formed was filtered off with suction and washed twice with 50 ml of methanol each time. The filtrate was then concentrated under reduced pressure and the residue was recrystallized from acetonitrile. This gave 1.5 g (16% of theory) of the title compound 4-methoxy-1,2-thiazole-3-carboximidamide hydrochloride as a white solid.



1H-NMR (400 MHz, DMSO-d6): δ=3.96 (s, 3H), 8.59 (s, 1H), 9.17-9.52 (m, 4H).


WORKING EXAMPLES
Example 1
5-(3,4-Dichlorobenzyl)-2-(pyridin-2-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 966 mg (7.0 mmol) of potassium carbonate, 826 mg (5.2 mmol) of 2-amidinopyridine hydrochloride and 600 mg (1.7 mmol) of ethyl 2-(3,4-dichlorobenzyl)-4,4,4-trifluoro-3-oxobutanoate (purity 68.3%; CAS 179110-12-4; WO 2012/041817, Intermediate 56) in 4 ml of dioxane was heated under reflux for 8 h. The solution was then filtered, the residue was washed with DMSO and the filtrate was purified by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% of formic acid). This gave 412 mg (59% of theory) of the title compound.


LC-MS (Method 1): Rt=1.31 min; MS (ESpos): m/z=400.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=4.00 (s, 2H), 7.20 (dd, 1H), 7.46-7.58 (m, 2H), 7.69 (dd, 1H), 8.09 (td, 1H), 8.33 (d, 1H), 8.78 (d, 1H), 13.07 (br. s, 1H).


The Exemplary compounds listed in Table 16 were prepared analogously to Example 1 by reacting the appropriate amidinopyridines or their salts with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 16





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data

















2
5-(3-chloro-4-fluorophenoxy)-2-(pyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.19 min; MS (ESpos): m/z = 386.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.14-7.21 (m, 1H), 7.40 (dd, 1H), 7.48 (dd, 1H), 7.69 (dd, 1H), 8.10 (dd, 1H), 8.35 (d, 1H), 8.78 (d, 1H), 13.4 (br. s, 1H).





3
5-(3,4-dichlorophenoxy)-2-(pyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.27 min; MS (ESpos): m/z = 402.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.22 (dd, 1H), 7.54 (d, 1H), 7.60 (d, 1H), 7.69 (dd, 1H), 8.07-8.14 (m, 1H), 8.35 (d, 1H), 8.78 (d, 1H), 13.42 (br. s, 1H).





4
5-(3,4-dichlorophenoxy)-2-(3,5-difluoropyridin- 2-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.25 min; MS (ESpos): m/z = 438.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.21 (dd, 1H), 7.57 (d, 1H), 7.60 (d, 1H), 8.20-8.29 (m, 1H), 8.74 (d, 1H), 13.72 (br. s, 1H).





5
2-(3-chloropyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.27 min; MS (ESpos): m/z = 436.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.20 (dd, 1H), 7.58-7.62 (m, 2H), 7.69 (dd, 1H), 8.20 (d, 1H), 8.71 (d, 1H), 12.76 (br. s, 0.2H), 13.94 (br. s, 0.8H).





6
2-[3-chloro-5-(trifluoromethyl)pyridin-2-yl]-5- (3,4-dichlorophenoxy)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.37 min; MS (ESpos): m/z = 504.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.22 (dd, 1H), 7.57-7.65 (m, 2H), 8.79 (s, 1H), 9.16 (s, 1H), 14.13 (br. s, 1H).





7
5-(3,4-dichlorophenoxy)-2-(5-methylpyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.33 min; MS (ESpos): m/z = 416.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.44 (s, 3H), 7.19 (dd, 1H), 7.54 (d, 1H), 7.59 (d, 1H), 7.91 (dd, 1H), 8.25 (d, 1H), 8.62 (s, 1H), 13.34 (br. s, 1H).





8
2-(5-chloropyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.32 min; MS (ESpos): m/z = 436.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.20 (dd, 1H), 7.54 (d, 1H), 7.60 (d, 1H), 8.23 (dd, 1H), 8.34 (d, 1H), 8.83 (dd, 1H), 13.58 (br. s, 1H).





9
5-(3,4-dichlorophenoxy)-2-(6-methylpyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.32 min; MS (ESpos): m/z = 416.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.61 (s, 3H), 7.20 (dd, 1H), 7.51-7.57 (m, 2H), 7.60 (d, 1H), 7.97 (t, 1H), 8.13 (d, 1H), 13.13 (br. s, 1H).





10
2-(6-chloropyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.30 min; MS (ESpos): m/z = 436.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.20 (dd, 1H), 7.54 (d, 1H), 7.61 (d, 2H), 7.82 (d, 1H), 8.14 (dd, 1H), 8.28 (d, 1H), 13.62 (br. s, 1H).





11
5-(4-chloro-3-fluorophenoxy)-2-(3- chloropyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.21 min; MS (ESpos): m/z = 420.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.05 (d, 1H), 7.44 (d, 1H), 7.55 (t, 2H), 7.69 (dd, 1H), 8.20 (d, 1H), 8.71 (d, 1H), 13.96 (br. s, 1H).





12
5-(3-chlorophenoxy)-2-(3-chloropyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 402.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.10 (dd, 1H), 7.19 (d, 1H), 7.29-7.32 (m, 1H), 7.38 (t, 2H), 7.69 (dd, 1H), 8.20 (d, 1H), 8.71 (d, 1H), 13.92 (br. s, 1H).





13
2-(3-chloropyridin-2-yl)-5-(3,4- difluorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.15 min; MS (ESpos): m/z = 404.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.95-7.05 (m, 1H), 7.35-7.49 (m, 2H), 7.69 (dd, 1H), 8.20 (dd, 1H), 8.71 (dd, 1H), 13.92 (br. s, 1H).





14
5-(3-chloro-4-fluorophenoxy)-2-(3- chloropyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.20 min; MS (ESpos): m/z = 420.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.13-7.22 (m, 1H), 7.40 (t, 1H), 7.53 (dd, 1H), 7.69 (dd, 2H), 8.20 (dd, 1H), 8.71 (dd, 1H), 13.90 (br. s, 1H).





15
5-(3,4-dichlorophenoxy)-2-(3-fluoropyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.21 min; MS (ESpos): m/z = 419.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.21 (dd, 1H), 7.58 (d, 1H), 7.60 (d, 1H), 7.78 (m, 1H), 8.02 (m, 1H), 8.63 (d, 1H), 13.60 (br. s, 1H).





16
2-(3-chloropyridin-2-yl)-5-(3,4-dichlorobenzyl)- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.31 min; MS (ESpos): m/z = 434.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ□□□4.01 (d, 2H), 7.17 (dd, 1H), 7.52 (d, 1H), 7.57 (d, 1H), 7.69 (dd, 1H), 8.19 (dd, 1H), 8.70 (dd, 1H), 13.69 (br. s, 1H).





17
5-(3,4-dichlorobenzyl)-2-(3-fluoropyridin-2-yl)- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.24 min; MS (ESpos): m/z = 418.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ□□□4.01 (d, 2H), 7.20 (dd, 1H), 7.51 (d, 1H), 7.55 (d, 1H), 7.78 (m, 1H), 8.01 (m, 1H), 8.62 (d, 1H), 13.40 (br. s, 1H).





18
5-(3,4-dichlorobenzyl)-2-(3,5-difluoropyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (62% of theory; reaction time: 1 h; solvent: dioxane; 5 eq. of potassium carbonate)

LC-MS (Method 1): Rt = 1.25 min; MS(ESpos): m/z = 436.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ□□□4.05 (d, 2H), 7.20 (dd, 1H), 7.51 (d, 1H), 7.55 (d, 1H), 8.24 (m, 1H), 8.73 (s, 1H), 13.39 (br. s, 1H).





19
5-(3,4-dichlorobenzyl)-2-(3,5-methylpyridin-2-yl)- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (13% of theory; reaction time: 1.5 h; 85° C.; solvent: dioxane; 5 eq. of potassium carbonate)

LC-MS (Method 1): Rt = 1.34 min; MS(ESpos): m/z = 414.0 (M +H)+1H-NMR (400 MHz, DMSO-d6): δ□= 3.99 (s, 2H), 7.20 (dd, 1H), 7.50 (s, 1H), 7.53 (d, 1H), 7.90 (d, 1H), 8.23 (d, 1H), 8.62 (s, 1H), 12.99 (br. s, 1H).





20
2-(6-chloropyridin-2-yl)-5-(3,4-dichlorobenzyl)- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.38 min; MS (ESpos): m/z = 434.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ□= 4.00 (s, 2H), 7.20 (d, 1H), 7.50 (s, 1H), 7.53 (d, 1H), 7.81 (d, 1H), 8.12 (t, 1H), 8.27 (d, 1H), 13.20 (br. s, 1H).





21
2-[3-chloro-5-(trifluoromethyl)pyridin-2-yl]-5- (3,4-dichlorobenzyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.41 min; MS (ESpos): m/z = 502.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ□= 4.03 (s, 2H), 7.19 (d, 1H), 7.53 (s, 1H), 7.57 (d, 1H), 8.79 (s, 1H), 9.15 (s, 1H), 13.86 (br. s, 1H).





22
2-(5-chloropyridin-2-yl)-5-(3,4-dichlorobenzyl)- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.36 min; MS (ESpos): m/z = 434.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ□= 3.99 (s, 2H), 7.20 (d, 1H), 7.50 (s, 1H), 7.54 (d, 1H), 8.21 (dd, 1H), 8.32 (d, 1H), 8.83 (d, 1H), 13.24 (br. s, 1H).





23
5-(3,4-dichlorobenzyl-2-(3-methoxypyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.19 min; MS (ESpos): m/z = 430.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ□= 3.87 (s, 3H), 3.99 (s, 2H), 7.18 (d, 1H), 7.51 (s, 1H), 7.56 (d, 1H), 7.62 (dd, 1H), 7.72 (d, 1H), 8.29 (d, 1H), 13.40 (br. s, 1H).





24
5-(3,4-dichlorophenoxy)-2-(3-methoxypyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.14 min; MS (ESpos): m/z = 432.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ□= 3.89 (s, 3H), 7.17 (d, 1H), 7.55- 7.66 (m, 3H), 7.72 (d, 1H), 8.31 (d, 1H), 13.62 (br. s, 1H).





25
5-(3,4-dichlorobenzyl)-2-(6-methylpyridin-2-yl)- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.38 min; MS (ESpos): m/z = 414.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.61 (s, 3H), 4.01 (s, 2H), 7.20 (dd, 1H), 7.47-7.58 (m, 3H), 7.96 (t, 1H), 8.11 (d, 1H), 12.79 (br. s, 1H).





26
5-(3,4-dichlorobenzyl-2-(3-methoxypyridin-4- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.22 min; MS (ESpos): m/z = 430.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.97 (s, 5H), 7.20 (dd, 1H), 7.47-7.64 (m, 3H), 8.39 (d, 1H), 8.62 (s, 1H), 13.28 (br. s, 1H).





27
5-(3,4-dichlorobenzyl)-2-(pyridin-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.16 min; MS (ESpos): m/z = 400.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.00 (s, 2H), 7.15-7.25 (m, 1H), 7.46- 7.68 (m, 3H), 8.44 (d, 1H), 8.78 (dd, 1H), 9.24 (s, 1H), 13.64 (br. s, 1H).





28
5-(3,4-dichlorobenzyl)-2-(pyridin-4-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.15 min; MS (ESpos): m/z = 400.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.01 (s, 2H), 7.20 (dd, 1H), 7.48-7.59 (m, 2H), 8.02-8.12 (m, 2H), 8.77- 8.86 (m, 2H), 13.71 (br. s, 1H).





29
5-(3,4-dichlorobenzyl)-2-(2-methoxypyridin-4- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 2): Rt = 4.07 min; MS (ESpos): m/z = 430.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.92 (s, 3H), 4.00 (s, 2H), 7.20 (dd, 1H), 7.42-7.70 (m, 4H), 8.37 (d, 1H), 13.63 (br. s, 1H).





30
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(6- hydroxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (98% of theory; reaction time: 1 h, 85° C.; solvent: dioxane; 5eq. of potassium carbonate; 3 eq. of 6-hydroxypyridine-2-carboximidamide)

LC-MS (Method 1): Rt = 1.17 min; MS (ESpos): m/z = 452 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.80-6.94 (m, 1H), 7.47 (dd, 1H), 7.61 (d, 1H), 7.69 (d, 1H), 7.54-7.74 (br. m, 1H), 7.85 (br. s, 1H), 12.34 (br. s, 1H).





31
5-(3,4-dichlorophenoxy)-2-(6-hydroxypyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (50% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-hydroxypyridine-2-carboximidamide)

LC-MS (Method 1): Rt = 1.14 min; MS (ESpos): m/z = 417.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.79-6.95 (m, 1H), 7.18 (dd, 1H), 7.52 (d, 1H), 7.60 (d, 1H), 7.63-7.79 (m, 1H), 7.81-7.91 (m, 1H), 11.20 (br. s, 1H), 12.27 (br. s, 1H).





32
5-(3,4-dichlorophenoxy)-2-(4-hydroxypyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (59% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-hydroxypyridine-2-carboximidamide)

LC-MS (Method 1): Rt = 1.04 min; MS (ESpos): m/z = 418.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.01 (br. m, 1H), 7.16 (dd, 1H), 7.49 (d, 1H), 7.58 (d, 1H), 7.76 (br. s, 1H), 8.43 (br. s, 1H), 11.37 (br. s, 1H), 13.18 (br. s, 1H).





33
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(4- hydroxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (18% of theory; reaction time: 1 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-hydroxypyridine-2-carboximidamide)

LC-MS (Method 1): Rt = 1.07 min; MS (ESpos): m/z = 452.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.01 (br. m, 1H), 7.45 (dd, 1H), 7.59 (d, 1H), 7.68 (d, 1H), 7.78 (br. s, 1H), 8.43(br. d, 1H), 11.45 (br. s, 1H), 13.17 (br. s, 1H).





34
5-[4-chloro-3-(trifluoromethyl)benzyl]-2-(3- fluoropyridin-2-yl)-6- (trifluoromethyl)pyrimidin4(3H)-one   embedded image
  (50% of theory; reactiontime: 1.5 h, 85° C.; solvent: dioxane; 4 eq. of potassium carbonate; 3 eq. of 3-fluoropyridine-2-carboximidamide hydrochloride (CAS 246872-67-3))

LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 452 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.93-4.18 (m, 2H), 7.49-7.53 (m, 1H), 7.65 (d, 1H), 7.72-7.81 (m, 2H), 7.96-8.03 (m, 1H), 8.62 (s, 1H), 13.41 (s, 1H).





35
2-(3-chloropyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)benzyl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (75% of theory; reaction time: 1 h, 101° C.; solvent: dioxane; 4 eq.of potassium carbonate; 3 eq. of 3-chloropyridine-2-carboximidamide hydrochloride (CAS 477902-83-3))

LC-MS (Method 1): Rt = 1.33 min; MS (ESpos): m/z = 468 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.09 (s, 2H), 7.50 (d, 1H), 7.65-7.68 (m, 1H), 7.68-7.71 (m, 1H), 7.77 (d, 1H), 8.20 (dd, 1H), 8.70 (dd, 1H), 13.72 (br. s, 1H).





36
2-(6-chloropyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (68% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 4 eq. of potassium carbonate; 3 eq. of 6-chloropyridine-2-carboximidamide hydrochloride (CAS 1179362-38-9))

LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 470 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.50 (dd, 1H), 7.62 (d, 1H), 7.70 (d, 1H), 7.81 (d, 1H), 8.14 (t, 1H), 8.29 (d, 1H).





37
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(2- methoxypyridin-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (56% of theory; reaction time: 1 h, 101° C.; solvent: dioxane; 4 eq. of potassium carbonate; 3 eq. of 2-methoxypyridin-3-carboximidamide (CAS 1016782-05-0))

LC-MS (Method 3): Rt = 2.90 min; MS (ESpos): m/z = 466 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.97 (s, 3H), 7.22 (dd, 1H), 7.47 (dd, 1H), 7.63 (d, 1H), 7.70 (d, 1H), 8.12 (dd, 1H), 8.41 (dd, 1H), 13.25-13.50 (m, 1H).





38
2-(3-chloropyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (72% of theory; reaction time: 1 h, 101° C.; solvent: dioxane; 4 eq. of potassium carbonate; 3 eq. of 3-chloropyridine-2-carboximidamide hydrochloride (CAS 477902-83-3))

LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 470 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.51 (dd, 1H), 7.69 (m, 3H), 8.19 (dd, 1H), 8.72 (dd, 1H).





39
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(3- fluoropyridin-2-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (80% of theory; reaction time: 1 h, 101° C.; solvent: dioxane; 4 eq. of potassium carbonate; 3 eq. of 3-fluoropyridine-2-carboximidamide hydrochloride (CAS 246872-67-3))

LC-MS (Method 1): Rt = 1.25 min; MS (ESpos): m/z = 454 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.52 (dd, 1H), 7.66 (d, 1H), 7.70 (d, 1H), 7.79 (dt, 1H), 8.02 (m, 1H), 8.63 (d, 1H), 13.39-13.85 (m, 1H).





40
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(3- methoxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (17% of theory; reaction time: 1 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 3-methoxypyridine-2-carboximidamide hydrochloride (CAS 1179362-06-1))

LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 466 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.89 (s, 3H), 7.49 (dd, 1H), 7.59-7.66 (m, 2H), 7.68 (d, 1H), 7.72-7.78 (m, 1H), 8.31 (dd, 1H), 13.67 (br. s, 1H).





41
5-(3,4-dichlorobenzyl-2-(2-methoxypyridin-3- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (56% of theory; reaction time: 1 h, 90° C.; solvent: dioxane; 3 eq. of potassium carbonate; 3 eq. of 2-methoxypyridin-3-carboximidamide (CAS 1016782-05-0))

LC-MS (Method 1): Rt = 1.37 min; MS (ESpos): m/z = 430 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.96 (s, 3H), 3.97 (s, 2H), 7.13-7.25 (m, 2H), 7.51 (d, 1H), 7.56 (d, 1H), 8.11 (dd, 1H), 8.39 (dd, 1H), 12.73- 13.22 (m, 1H).





42
5-(3,4-dichlorobenzyl)-2-(2-methoxypyridin-3- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (56% of theory; reaction time: 1 h, 101° C.; solvent: dioxane; 3 eq. of potassium carbonate; 3 eq. of 2-methoxypyridin-3-carboximidamide (CAS 1016782-05-0))

LC-MS (Method 1): Rt = 1.31 min; MS (ESpos): m/z = 432 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.93 (s, 3H), 7.02-7.11 (m, 1H), 7.13- 7.19 (m, 1H), 7.33-7.43 (m, 1H), 7.53-7.61 (m, 1H), 7.98-8.04 (m, 1H), 8.27-8.37 (m, 1H), 13.23-13.41 (m, 1H).





43
2-(6-aminopyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (75% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 3 eq. of potassium carbonate; 3 eq. of 6-aminopyridine-2-carboximidamide)

LC-MS (Method 1): Rt = 1.20 min; MS (ESpos): m/z = 417.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.56 (br. s, 2H), 6.76 (d, 1H), 7.51 (d, 1H), 7.58 (d, 1H), 7.65-7.71 (m, 2H), 12.42 (br. s, 1H).





44
2-(6-aminopyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (78% of theory; reaction time: 1 h, 85° C.; solvent: dioxane; 3 eq. of potassium carbonate; 3 eq. of 6-aminopyridine-2-carboximidamide)

LC-MS (Method 1): Rt = 1.22 min; MS (ESpos): m/z = 451.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.50 (br. s, 2H), 6.74 (d, 1H), 7.16 (dd, 1H), 7.47-7.52 (m, 2H), 7.58 (d, 1H), 7.64-7.71 (m, 1H), 12.36 (br. s, 1H).





45
2-(4-aminopyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (90% of theory; reaction time: 1 h, 85° C.; solvent: dioxane; 3 eq. of potassium carbonate; 3 eq. of 4-aminopyridine-2-carboximidamide dihydrochloride)

LC-MS (Method 1): Rt = 1.22 min; MS (ESpos): m/z = 451.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.80 (dd, 1H), 7.15 (dd, 1H), 7.29 (d, 1H), 7.61 (d, 1H), 7.69 (d, 1H), 8.01 (d, 1H), 8.02 (s, 2H), 13.0 (br. s, 1H).









EXAMPLE 46
2-(5-Amino-2-oxo-1,3-oxathiol-4-yl)-5-(3,4-dichlorobenzyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


At 23° C., 28.3 μl (0.34 mmol) of chloro(chlorosulphanyl)oxomethane were added to a solution of 100 mg (0.26 mmol) of 2-[5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetamide (Example 73A) in 2 ml of toluene. The reaction mixture was stirred at reflux for 1 h and then concentrated. The residue was taken up in 3-4 ml of DMSO and purified by preparative HPLC (column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient: 10:90→90:10). The product obtained in this manner was triturated with diethyl ether and the precipitated crystals were filtered off with suction and dried under high vacuum. This gave 30 mg (24% of theory; purity 91%) of the title compound.


LC-MS (Method 1): Rt=1.24 min; MS (ESpos): m/z=438.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=12.83 (br. s, 1H), 8.28 (br. s, 2H), 7.52 (d, J=8.3 Hz, 1H), 7.42 (s, 1H), 7.10 (d, J=8.3 Hz, 1H), 3.91 (br. s, 2H).


EXAMPLE 47
2-(4-Amino-1H-pyrazol-1-yl)-5-(3,4-dichlorophenoxy)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


113 mg (0.7 mmol) of 4-amino-1H-pyrazole dihydrochloride (purity 97%) in 3 ml of DMF were stirred with 84 mg (2.1 mmol) of sodium hydride (60% in paraffin) at 23° C. for 30 minutes. 100 mg (0.23 mmol) of 5-(3,4-dichlorophenoxy)-2-(methylsulphonyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 38A) were then added, and the mixture was stirred at 120° C. for 30 minutes. After addition of 1 ml of aqueous buffer solution (pH 7), the mixture was purified directly by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% of formic acid). This gave 50 mg (53% of theory) of the title compound.


LC-MS (Method 1): Rt=1.03 min; MS (ESpos): m/z=406.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=4.15 (br. s, 2H), 6.83 (dd, 2H), 7.01 (d, 1H), 7.19 (m, 1H), 7.49 (d, 1H), 7.71 (m, 1H).


The following compound was prepared in an analogous manner:











TABLE 17





Example
IUPAC name, structure



No.
(yield)
Analytical data







48
2-(5-Amino-1H-1,2,4-triazol-1-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
LC-MS (Method 1): Rt = 0.99 min; MS (ESpos): m/z = 407 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 5.58 (s, 1H), 6.86 (dd, 1H), 7.08 (d, 1H), 7.43 (s, 1H), 7.48 (d, 1H), 7.59 (br. s, 2H). The 1,2,4-triazol-2-yl isomer was obtained as a minor component: δ = 5.58 (s, 1H), 6.83 (dd, 1H), 7.08 (d, 1H), 7.48 (d, 1H), 7.59 (br. s, 2H), 8.60 (s, 1H).









EXAMPLE 49
5-(3,4-Dichlorophenoxy)-2-(1H-pyrazol-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 150 mg (0.435 mmol) of ethyl 2-(3,4-dichlorophenoxy)-4,4,4-trifluoro-3-oxobutanoate (Example 16A), 142 mg (0.869 mmol) of 1H-pyrazole-3-carboximidamide and 0.23 ml (1.3 mmol) of N,N-diisopropylethylamine in 2.5 ml of DMF was stirred at 90° C. for 7 h. The mixture was then purified directly by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% of formic acid). Lyophilization of the product-containing fractions gave, from two reactions with, in total, 0.493 mmol of ethyl 2-(3,4-dichlorophenoxy)-4,4,4-trifluoro-3-oxobutanoate, 12 mg (6% of theory) of the title compound.


LC-MS (Method 2): Rt=3.39 min; MS (ESpos): m/z=390.9 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=6.97 (br. s, 1H), 7.17 (dd, 1H), 7.49-7.64 (m, 2H), 7.97 (br. s, 1H), 13.42 (br. s, 1H), 13.66 (br. s, 1H).


The Exemplary compounds listed in Table 18 were prepared analogously to Example 1 or Example 49 by reacting the appropriate amidines (carboximidamides) or their salts with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 18





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







50
5-(3,4-dichlorobenzyl)-2-(1H-pyrazol-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (16% of theory; 3 eq. of potassium carbonate; 1 eq. of 1H-pyrazole-3-carboximidamide; ethanol, 60° C., 9 h)

LC-MS (Method 2): Rt = 3.50 min; MS (ESpos): m/z = 389.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.95 (br. s, 2H), 6.96 (br. s, 1H), 7.18 (d, 1H), 7.45-7.57 (m, 2H), 7.96 (br. s, 1H), 13.13 (br. s, 1H), 13.66 (br. s, 1H).





51
5-(3,4-dichlorophenoxy)-2-(1H-pyrazol-1-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (18% of theory; 3 eq. of potassium carbonate; 3 eq. of 1H-pyrazole-1-carboximidamide hydrochloride; dioxane, 85° C., 1 h)

LC-MS (Method 1): Rt = 1.19 min; MS (ESpos): m/z = 390.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.42 (m, 1H), 6.84 (dd, 1H), 7.05 (d, 1H), 7.50 (d, 1H), 7.66 (br. s, 1H), 8.40 (br. d, 1H).





52
5-(3,4-dichlorobenzyl)-2-(1-methyl-1H- imidazol-4-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (46% of theory; 8 eq. of potassium carbonate; 3 eq. of 1-methyl-1H-imidazole-4- carboximidamide hydrochloride; dioxane, reflux, 21 h)

LC-MS (Method 1): Rt = 1.13 min; MS (ESpos): m/z = 403.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.92 (s, 2H), 7.16 (dd, 1H), 7.42-7.57 (m, 2H), 7.87 (s, 1H), 8.08 (s, 1H), 12.68 (br. s, 1H).





53
5-(3,4-dichlorobenzyl)-2-(1H-pyrazol-1-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (5% of theory; 3 eq. of potassium carbonate; 5 eq. of 1H-pyrazole-1-carboxamidine hydrochloride; dioxane, 85° C., 1 h)

LC-MS (Method 1): Rt = 1.24 min; MS (ESpos): m/z = 389.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.97 (s, 2H), 6.67 (m, 1H), 7.18 (d, 1H), 7.48 (s, 1H), 7.53 (d, 1H), 7.96 (s, 1H), 8.55 (d, 1H), 9.46 (br. m, 0.5H), 13.75 (br. s, 0.5H) (tautomer mixture; only the main component is shown).





54
5-(3,4-dichlorophenoxy)-2-(1,5-dimethyl-1H- pyrazol-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (69% of theory; 3 eq. of potassium carbonate; 3 eq. of 1,5-dimethyl-1H-pyrazole-3- carboximidamide hydrochloride; dioxane, 85° C., 1 h)

LC-MS (Method 1): Rt = 1.23 min; MS (ESpos): m/z = 419.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.33 (s, 3H), 3.86 (s, 3H), 6.77 (s, 1H), 7.15 (dd, 1H), 7.50 (d, 1H), 7.57 (d, 1H), 13.33 (br. s, 1H).





55
5-(3,4-dichlorobenzyl)-2-(1,5-dimethyl-1H- pyrazol-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (61% of theory; 4 eq. of potassium carbonate; 3 eq. of 1,5-dimethyl-1H-pyrazole-3- carboximidamide hydrochloride; dioxane, reflux, 10 h)

LC-MS (Method 1): Rt = 1.26 min; MS (ESpos): m/z = 417.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.32 (s, 3H), 3.85 (s, 3H), 3.93 (s, 2H), 6.77 (s, 1H), 7.17 (dd, J = 8.3, 1.8 Hz, 1H), 7.43-7.59 (m, 2H), 13.02 (br. s, 1H).





56
5-(3,4-dichlorophenoxy)-2-(1-methyl-1H- pyrazol-5-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (53% of theory; 5 eq. of potassium carbonate; 3 eq. of 1-methyl-1H-pyrazole-5- carboximidamide; dioxane, 85° C., 1 h)

LC-MS (Method 3): Rt = 2.56 min; MS (ESpos): m/z = 405 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.19 (s, 3H), 7.19 (dd, 1H), 7.21 (d, 1H), 7.53 (d, 1H), 7.60 (d, 1H), 7.62 (s, 1H), 13.60-13.78 (m, 1H).





57
5-(3,4-dichlorobenzyl)-2-(3,5-dimethyl-1H- pyrazol-1-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (30% of theory; 5 eq. of potassium carbonate; 3 eq. of 3,5-dimethyl-1H-pyrazole-1- carboximidamide nitrate; DMF, 80° C., 10 h)

LC-MS (Method 1): Rt = 1.40 min; MS (ESpos): m/z = 417.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.24 (s, 3H), 2.58 (s, 3H), 3.93 (s, 2H), 6.28 (s, 1H), 7.19 (dd, 1H), 7.43-7.59 (m, 2H), 13.15 (br. s, 1H).





58
Ethyl 1-[5-(3,4-dichlorobenzyl)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2-yl]-5- hydroxy-1H-pyrazole-3-carboxylate   embedded image
  (7% of theory; 3.5 eq. of potassium carbonate; 3 eq. of ethyl 1-carbamimidoyl-5-hydroxy-1H- pyrazole-3-carboxylate nitrate ethanol solvate; dioxane, 101° C., 16 h)

LC-MS (Method 1): Rt = 1.08 min; MS (ESpos): m/z = 449 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.29 (t, 3H), 3.96 (s, 2H), 4.28 (q, 2H), 5.65 (s, 1H), 7.15 (dd, 1H), 7.47 (d, 1H), 7.53 (d, 1H).





59
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(1- methyl-1H-imidazol-4-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (20% of theory; 2 eq. of 1-methyl-1H-imidazole- 4-carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 4 h)

LC-MS (Method 2): Rt = 3.44 min; MS (ESpos): m/z = 439.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.77 (s, 3H), 7.44 (dd, 1H), 7.57 (d, 1H), 7.66 (d, 1H), 7.92 (s, 1H), 8.10 (s, 1H).





60
2-(5-amino-1H-pyrazol-4-yl)-5-(3,4- dichlorobenzyl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (2% of theory; 2 eq. of 5-amino-1H-pyrazole-4- carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 8 h)

LC-MS (Method 2): Rt = 3.04 min; MS (ESpos): m/z = 404.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.97 (s, 2H), 7.13 (dd, 1H), 7.40 (d, 1H), 7.49 (d, 1H), 8.22 (br. s, 2H), 8.58 (s, 1H), 8.63 (br. s, 2H).





61
2-(5-amino-1-methyl-1H-pyrazol-4-yl)-5-(3,4- dichlorobenzyl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (5% of theory; 2 eq. of 5-amino-1-methyl-1H- pyrazole-4-carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 8 h)

LC-MS (Method 2): Rt = 3.39 min; MS (ESpos): m/z = 418.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.58 (s, 3H), 3.88 (s, 2H), 6.83 (s, 2H), 7.16 (dd, 1H), 7.40-7.59 (m, 2H), 8.09 (s, 1H), 12.77 (s, 1H).





62
2-(5-amino-1H-pyrazol-4-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one trifluoroacetate   embedded image
  (3% of theory; 2 eq. of 5-amino-1H-pyrazole-4- carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 4 h)

LC-MS (Method 2): Rt = 2.91 min; MS (ESpos): m/z = 406.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.00 (m, 1H), 7.28 (d, 1H), 7.52 (d, 1H), 8.26 (br. s, 2H), 8.81-8.54 (m, 3H).





63
5-(3,4-dichlorobenzyl)-2-(1H-pyrazol-4-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (10% of theory; 2 eq. of 1H-pyrazole-4- carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 7 h)

conditions of prep. HPLC purification: Column: Shield RP18 5 μm, 100 × 19 mm; flow rate: 25 ml/min; run time: 9 min; detection: 210 nm, mobile phase: acetonitrile/water 65:35 with 0.05% TFA. LC-MS (Method 2): Rt = 3.25 min; MS (ESpos): m/z = 388.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.93 (s, 2H), 7.17 (dd, 1H), 7.40-7.60 (m, 2H), 8.21 (br. s, 1H), 8.57 (br. s, 1H), 13.20 (s, 1H), 13.50 (br. s, 1H).





64
2-(4-amino-1H-imidazol-5-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (7% of theory; 2 eq. of 5-amino-1H-imidazole-4- carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 4 h)

LC-MS (Method 2): Rt = 3.14 min; MS (ESpos): m/z = 406.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.03 (dd, 1H), 7.31 (d, 1H), 7.51 (d, 1H), 8.15 (br. s, 2H), 8.33 (br. s, 3H).





65
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(1H- pyrazol-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (15% of theory; 2 eq. of 1H-pyrazole-3- carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 7 h)

LC-MS (Method 2): Rt = 3.54 min; MS (ESpos): m/z = 425.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 8.49-6.53 (m, 5H), 14.08-13.09 (m, (2H).





66
5-(3,4-dichlorophenoxy)-2-(1H-pyrazol-4-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (15% of theory; 2 eq. of 1H-pyrazole-4- carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 7 h)

LC-MS (Method 2): Rt = 3.17 min; MS (ESpos): m/z = 391.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.15 (dd, 1H), 7.50 (d, 1H), 7.57 (d, 1H), 8.22 (br. s, 1H), 8.56 (br. s, 1H), 13.30-13.67 (m, 2H).





67
2-(5-amino-1-methyl-1H-pyrazol-4-yl)-5-[4- chloro-3-(trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (17% of theory; 2 eq. of 5-amino-1-methyl-1H- pyrazole-4-carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 4 h)

LC-MS (Method 2): Rt = 3.39 min; MS (ESpos): m/z = 454.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.59 (s, 3H), 6.83 (s, 2H), 7.43 (dd, 1H), 7.56 (d, 1H), 7.66 (d, 1H), 8.08 (s, 1H), 13.02 (s, 1H).





68
2-(5-amino-1-methyl-1H-pyrazol-4-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-on   embedded image
  (6% of theory; 2 eq. of 5-amino-1-methyl-1H- pyrazole-4-carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 4 h)

LC-MS (Method 2): Rt = 3.24 min; MS (ESpos): m/z = 420.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.59 (s, 3H), 6.81 (s, 2H), 7.12 (m, 1H), 7.47 (d, 1H), 7.56 (d, 1H), 8.08 (s, 1H), 12.99 (br. s, 1H).





69
2-(4-amino-1H-imidazol-5-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (15% of theory; 2 eq. of 5-amino-1H-imidazole- 4-carboximidamide; 3 eq. of DIPEA; DMF, 90° C., 4 h)

LC-MS (Method 2): Rt = 3.27 min; MS (ESpos): m/z = 440.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.31 (dd, 1H), 7.44 (d, 1H), 7.60 (d, 1H), 8.17 (br. s, 2H), 8.34 (br. s, 3H).





70
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(1H- pyrazol-4-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (17% of theory; 2 eq. of 1H-pyrazole-4- carboximidamide hydrochloride; 3 eq. of DIPEA; DMF, 90° C., 7 h)

LC-MS (Method 2): Rt = 3.30 min; MS (ESpos): m/z = 425.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.46 (dd, 1H), 7.59 (d, 1H), 7.67 (d, 1H), 8.22 (br. s, 1H), 8.57 (br. s, 1H), 13.28-13.78 (m, 2H).





71
5-[4-chloro-3-(trifluoromethyl)benzyl]-2-(1H- pyrazol-1-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (11% of theory; 3 eq. of 1H-pyrazole-1- carboximidamide hydrochloride; 6 eq. of potassium carbonate; dioxane, 101° C., 1 h)

LC-MS (Method 1): Rt = 1.28 min; MS (ESpos): m/z = 423 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.00-4.08 (m, 2H), 6.59-6.71 (m, 1H), 7.41-7.53 (m, 1H), 7.60-7.66 (m, 1H), 7.70-7.76 (m, 1H), 7.92- 7.97 (m, 1H), 8.54 (d, 1H), 9.39-9.57 (m, 1H).





72
5-[4-chloro-3-(trifluoromethyl)benzyl]-2-(1H- pyrazol-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (59% of theory; 3 eq. of 1H-pyrazole-3- carboximidamide hydrochloride; 4 eq. of potassium carbonate; dioxane, 101° C., 1 h)

LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 423 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.03 (s, 2H), 6.96 (br. s, 1H), 7.49 (d, 1H), 7.63 (d, 1H), 7.74 (d, 1H), 7.96 (br. s, 1H), 12.96-13.31 (m, 1H), 13.56-13.77 (m, 1H).





73
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(1H- pyrazol-1-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (22% of theory; 3 eq. of 1H-pyrazole-1- carboximidamide hydrochloride; 4 eq. of potassium carbonate; dioxane, 101° C., 1 h)

LC-MS (Method 3): Rt = 2.74 min; MS (ESpos): m/z = 425 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.69 (dd, 1H), 7.47 (dd, 1H), 7.60 (d, 1H), 7.69 (d, 1H), 7.98 (d, 1H), 8.57 (d, 1H).





74
2-(4-chloro-1H-pyrazol-3-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (20% of theory; 1.5 eq. of 4-chloro-1H-pyrazole- 3-carboximidamide; 2 eq. of DIPEA; DMF, 90° C., 8 h)

LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 424.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.18 (dd, 1H), 7.51-7.62 (m, 2H), 8.26 (s, 1H), 13.43 (br. s, 1H), 13.98 (br. s, 1H).





75
2-(4-chloro-1H-pyrazol-3-yl)-5-(3,4- dichlorobenzyl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (11% of theory; 1.5 eq. of 4-chloro-1H-pyrazole- 3-carboximidamide; 2 eq. of DIPEA; DMF, 90° C., 8 h)

LC-MS (Method 1): Rt = 1.23 min; MS (ESpos): m/z = 423.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.96 (s, 2H), 7.18 (dd, 1H), 7.47-7.57 (m, 2H), 8.25 (s, 1H), 13.15 (br. s, 1H), 13.97 (br. s, 1H).





76
2-(4-chloro-1H-pyrazol-3-yl)-5-[4-chloro-3- (trifluoromethyl)benzyl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (9% of theory; 1.5 eq. of 4-chloro-1H-pyrazole- 3-carboximidamide; 2 eq. of DIPEA; DMF, 90° C., 4 h)

LC-MS (Method 1): Rt = 1.27 min; MS (ESpos): m/z = 457.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.04 (s, 2H), 7.49 (d, 1H), 7.63 (d, 1H), 7.75 (d, 1H), 8.25 (s, 1H), 13.18 (br. s, 1H), 13.98 (br. s, 1H).





77
2-(4-chloro-1H-pyrazol-3-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (23% of theory; 1.5 eq. of 4-chloro-1H-pyrazole- 3-carboximidamide; 2 eq. of DIPEA; DMF, 90° C., 3 h)

LC-MS (Method 1): Rt = 1.20 min; MS (ESpos): m/z = 459.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.50 (dd, 1H), 7.61-7.72 (m, 2H), 8.27 (s, 1H), 13.48 (br. s, 1H), 13.99 (br. s, 1H).









EXAMPLE 78
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-(1,2,4-oxadiazol-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A drop of boron trifluoride diethyl ether complex was added to 250 mg (0.6 mmol) of 5-[4-chloro-3-(trifluoromethyl)phenoxy]-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carboximidamide (Example 87A) and 93 mg (0.88 mmol) of trimethyl orthoformate in 7.6 ml of dioxane, and the mixture was stirred at 100° C. for 4 h. The mixture was then purified directly by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 45 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→95% A (35.00-40.00 min)->10% A (40.50-45.00 min)]. The product-containing fractions were combined and concentrated by evaporation. Yield: 22 mg (8% of theory).


LC-MS (Method 1): Rt=1.12 min; MS (ESpos): m/z=426.9 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=7.15 (dd, 1H), 7.28 (d, 1H), 7.61 (d, 1H), 9.66 (s, 1H).


The following compound was prepared in an analogous manner:











TABLE 19





Example
IUPAC name/structure



No.
(yield)
Analytical data







79
5-(3,4-Dichlorophenoxy)-2-(1,2,4-oxadiazol- 3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
LC-MS (Method 1): Rt = 1.09 min; MS (ESpos): m/z = 392.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.20 (dd, 1H), 7.55 (d, 1H), 7.61 (d, 1H), 9.95 (s, 1H).









EXAMPLE 80
5-(3,4-Dichlorophenoxy)-2-[5-(propan-2-yl)-1,2,4-oxadiazol-3-yl]-6-(trifluoromethyl)pyrimidin-4(3H)-one x N,N-diisopropylethylamine



embedded image


100 mg (0.25 mmol) of 5-(3,4-dichlorophenoxy)-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carboximidamide (Example 86A), 35 mg (0.33 mmol) of 2-methylpropionyl chloride and 53 mg (0.4 mmol) of N,N-diisopropylethylamine in 2 ml of dioxane were stirred at 90° C. for 16 h. The mixture was then purified directly by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 45 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→95% A (35.00-40.00 min)→10% A (40.50-45.00 min)]. The product-containing fractions were combined and concentrated by evaporation and the residue was triturated with water, filtered off with suction and dried again. Yield: 22 mg (16% of theory).


LC-MS (Method 1): Rt=1.23 min; MS (ESpos): m/z=435 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=1.21-1.29 (m, 15H), 1.37 (d, 6H), 3.09-3.18 (m, 2H), 3.55-3.68 (m, 2H), 6.86 (br. d, 1H), 7.08 (br. s, 1H), 7.51 (d, 1H), 8.16 (br. s, 1H).


The following compounds were prepared in an analogous manner:











TABLE 20





Example
IUPAC name/structure



No.
(starting material, reaction time, yield)
Analytical data







81
5-(3,4-dichlorophenoxy)-2-[5-(methoxymethyl)- 1,2,4-oxadiazol-3-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one x N,N- diisopropylethylamine   embedded image
  (from methoxyacetyl chloride; 8 h; 38% of theory)

LC-MS (Method 1): Rt = 1.13 min; MS (ESpos): m/z = 437 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.18-1.30 (m, 12H), 3.08-3.19 (m, 2H), 3.42 (s, 3H), 3.56-3.68 (m, 2H), 4.82 (s, 2H), 5.76 (s, 2H), 6.84-6.92 (m, 1H), 7.07-7.15 (m, 1H), 7.52 (d, 1H), 8.10-8.26 (m, 1H).





82
2-(5-cyclopropyl-1,2,4-oxadiazol-3-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one x N,N-diisopropylethylamine   embedded image
  (from cyclopropanecarbonyl chloride; 12 h; 9% of theory)

LC-MS (Method 1): Rt = 1.19 min; MS (ESpos): m/z = 433 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.16-1.36 (m, 20H), 2.44 (br. m, 1H), 3.14 (m, 2H), 3.62 (m, 2H), 7.0 (br. s, 1H), 7.29 (br. s, 1H), 7.55 (d, 1H), 8.17 (br. s, 1H).









EXAMPLE 83
5-(3,4-Dichlorophenoxy)-2-(5-hydroxypyridin-2-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


At 0° C., 40 mg (0.5 mmol) of acetyl chloride were added to 100 mg (0.25 mmol) of 5-[3,4-dichlorophenoxy)-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1, 6-dihydropyrimidine-2-carboximidamide (Example 86A) in 2 ml of pyridine, and the mixture was stirred at 23° C. for 12 h. The mixture was then purified directly by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 45 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→95% A (35.00-40.00 min)→10% A (40.50-45.00 min)]. The product-containing fractions were combined and concentrated by evaporation. Yield: 25 mg (24% of theory).


LC-MS (Method 1): Rt=1.12 min; MS (ESpos): m/z=407 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=2.74 (s, 3H), 7.19 (dd, 1H), 7.53 (d, 1H), 7.60 (d, 1H).


EXAMPLE 84
5-(3,4-Dichlorophenoxy)-6-(trifluoromethyl)-2-[5-(trifluoromethyl)-1,2,4-oxadiazol-3-yl]pyrimidin-4(3H)-one



embedded image


At 0° C., 107 mg (0.5 mmol) of trifluoroacetic anhydride were added to 100 mg (0.25 mmol) of 5-(3,4-dichlorophenoxy)-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carboximidamide (Example 86A) in 2 ml of pyridine, and the mixture was then heated under reflux for 30 minutes. After cooling, water was added, the mixture was extracted three times with ethyl acetate and the combined organic phases were dried over sodium sulphate. After concentration, the residue was purified by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 45 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→95% A (35.00-40.00 min)→10% A (40.50-45.00 min)]. The product-containing fractions were combined and concentrated by evaporation. Yield: 21 mg (16% of theory).


LC-MS (Method 1): Rt=1.23 min; MS (ESneg): m/z=458.9 (M−H)



1H-NMR (400 MHz, DMSO-d6): δ=7.14 (dd, 1H), 7.47 (d, 1H), 7.59 (d, 1H).


EXAMPLE 85
5-(3,4-Dichlorophenoxy)-2-(5-imino-4,5-dihydro-1,2,4-oxadiazol-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


At 23° C., 58 mg (0.42 mmol) of potassium carbonate were added to 150 mg (0.38 mmol) of 5-(3,4-dichlorophenoxy)-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1, 6-dihydropyrimidine-2-carboximidamide (Example 86A) in 2 ml of ethanol and 2 ml of water. 0.14 ml (0.42 mmol) of a 3 M solution of cyanogen bromide in methylene chloride were added dropwise and the mixture was stirred at room temperature for 15 minutes. After concentration, the residue was purified by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 45 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→95% A (35.00-40.00 min)-10% A (40.50-45.00 min)]. The product-containing fractions were combined and concentrated by evaporation. Yield: 3 mg (2% of theory).


LC-MS (Method 1): Rt=1.03 min; MS (ESpos): m/z=408.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=7.18 (br. d, 1H), 7.53 (br. s, 1H), 7.59 (d, 1H), 8.26 (br. s, 2H).


EXAMPLE 86
5-(3,4-Dichlorophenoxy)-2-(5-oxo-4,5-dihydro-1,2,4-thiadiazol-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


150 mg (0.38 mmol) of 5-(3,4-dichlorophenoxy)-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carboximidamide (Example 86A) were dissolved in 4.6 ml of THF, 81.5 mg (0.46 mmol) of 1,1′-thiocarbonyldiimidazole were added and the mixture was stirred at 23° C. for 2 h. Subsequently, 54 mg (0.38 mmol) of boron trifluoride diethyl ether complex were added and the mixture was stirred at 23° C. for a further 68 h. After addition of 2.5 ml of water, the mixture was purified directly by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 45 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→95% A (35.00-40.00 min)→10% A (40.50-45.00 min)]. The product-containing fractions were combined and concentrated by evaporation. Yield: 6 mg (4% of theory).


LC-MS (Method 2): Rt=3.03 min; MS (ESneg): m/z=422.8 (M−H)



1H-NMR (400 MHz, DMSO-d6): δ=7.15 (dd, 1H), 7.49 (d, 1H), 7.60 (d, 1H), 13.90 (br. s, 1H).


EXAMPLE 87
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-(5,5-dimethyl-4,5-dihydro-1,2,4-oxadiazol-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


The title compound (26 mg) was obtained as a by-product in the preparation of 5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(1,2,4-oxadiazol-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 78) (apparently owing to acetone impurities in the reaction).


LC-MS (Method 1): Rt=1.15 min; MS (ESpos): m/z=457.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=1.68 (s, 6H), 7.47 (dd, 1H), 7.62 (d, 1H), 7.70 (d, 1H), 8.51 (s, 1H), 10.47 (s, 1H).


EXAMPLE 88
1-[5-(3,4-Dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]-5-hydroxy-H-pyrazole-3-carboxylic acid



embedded image


17 mg (0.034 mmol) of ethyl 1-[5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]-5-hydroxy-1H-pyrazole-3-carboxylate (Example 58) in 0.13 ml of THF were stirred with 0.14 ml of 1 N aqueous lithium hydroxide solution at 23° C. for 6 h. The reaction was then neutralized with 1 N hydrochloric acid. The precipitated solid was filtered off with suction, washed with water and dried under high vacuum. This gave 11 mg (73% of theory) of the title compound.


LC-MS (Method 1): Rt=1.08 min; MS (ESpos): m/z=449.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.98 (s, 2H), 5.78 (br. m, 1H), 7.17 (d, 1H), 7.47 (d, 1H), 7.54 (d, 1H), 13.07 (br. s, 1H).


EXAMPLE 89
5-(3,4-Dichlorobenzyl)-2-[(5-oxo-4,5-dihydro-1,3,4-oxadiazol-2-yl)methyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


At RT, 32 mg (0.20 mmol) of 1,1′-carbonyldiimidazole were added to a solution of 65 mg (0.16 mmol) of 2-[5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetohydrazide (Example 79A) in 1.3 ml of dry THF. The mixture was stirred under reflux for 1 h. 2 drops of water were then added, and the reaction mixture was purified directly by preparative HPLC (column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient: 10:90→90:10). This gave 61 mg (88% of theory; purity 100%) of the title compound.


LC-MS (Method 1): Rt=1.02 min; MS (ESpos): m/z=421.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=13.43 (br. s, 1H), 12.32 (s, 1H), 7.54 (d, J=8.3 Hz, 1H), 7.43 (d, J=1.8 Hz, 1H), 7.14 (dd, J=8.3, 1.8 Hz, 1H), 4.06 (s, 2H), 3.92 (s, 2H).


EXAMPLE 90
5-(3,4-Dichlorobenzyl)-2-[(5-oxo-4,5-dihydro-1H-1,2,4-triazol-3-yl)methyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


At 23° C., 161.7 μl (3.03 mmol) of conc. sulphuric acid were added to a solution of 25 mg (0.04 mmol) of 5-(3,4-dichlorobenzyl)-2-{[4-(2,4-dimethoxybenzyl)-5-oxo-4,5-dihydro-1H-1,2,4-triazol-3-yl]methyl}-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 77A) in 431.1 μl of acetic acid. The mixture was stirred at 50° C. for 30 min. A few drops of water were then added with ice cooling, and the reaction mixture was purified directly by preparative HPLC (column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient: 10:90→90:10). This gave 11 mg (60% of theory; purity 100%) of the title compound.


LC-MS (Method 1): Rt=0.91 min; MS (ESpos): m/z=420.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=13.34 (br. s, 1H), 11.23-11.38 (m, 2H), 7.55 (d, J=8.3 Hz, 1H), 7.43 (d, J=1.8 Hz, 1H), 7.14 (dd, J=8.3, 1.9 Hz, 1H), 3.80-3.96 (m, 4H).


EXAMPLE 91
5-(3,4-Dichlorobenzyl)-2-[(3-methyl-1,2,4-oxadiazol-5-yl)methyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


Under argon and at 23° C., 222.7 mg (5.57 mmol) of sodium hydride (60% in mineral oil) were added to a solution of 374.9 mg (5.06 mmol) of (1Z)—N′-hydroxyethanimidamide in 10 ml of DMF. The mixture was stirred at 50° C. for 10 min and then at 23° C. for 40 min. 200 mg (0.51 mmol) of methyl [5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetate (Example 78A) were then added, and the reaction mixture was treated in a microwave at 120° C. for 3 h. Water, formic acid, methanol and DMSO were then added, and the mixture was purified directly by preparative HPLC (column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient: 10:90→90:10). This gave 79 mg (37% of theory; purity 100%) of the title compound.


LC-MS (Method 2): Rt=3.45 min; MS (ESpos): m/z=419.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=13.50 (br. s, 1H), 7.55 (d, J=8.3 Hz, 1H), 7.44 (d, J=2.0 Hz, 1H), 7.15 (dd, J=8.3, 2.0 Hz, 1H), 4.41 (s, 2H), 3.93 (s, 2H), 2.34 (s, 3H).


The Exemplary compounds listed in Table 21 were prepared analogously to Example 1 by reacting the appropriate amidines (imidamides) or their salts with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 21





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







92
5-(3,4-dichlorobenzyl)-2-(1H-1,2,4-triazol-5- ylmethyl)-6-(trifluoromethyl)pyrimidin-4(3H)- one   embedded image
  (28% of theory; 3eq. of 2-(4H-1,2,4-triazol-3- yl)ethanimidamide hydrochloride; 6 eq. of potassium carbonate; dioxane, 101° C., 16 h)

LC-MS (Method 1): Rt = 0.97 min; MS (ESpos): m/z = 404 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.92 (s, 2H), 4.04 (s, 1.5H), 4.17 (s, 0.5H), 7.13 (dd, 1H), 7.44 (d, 1H), 7.55 (d, 1H), 7.90 (br. s, 0.3H), 8.49 (br. s, 0.7H). 13.37 (br. s, 1H), 13.93 (br. s, 1H).





93
5-(3,4-dichlorobenzyl)-2-(pyridin-4-ylmethyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (23% of theory; 8 eq. of 2-(pyridin-4- yl)ethanimidamide hydrochloride; 8.5 eq. sodium methoxide; methanol, 65° C., 10 h)

LC-MS (Method 3): Rt = 2.19 min; MS (ESpos): m/z = 414 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.90 (s, 2H), 4.00 (s, 2H), 7.13 (dd, 1H), 7.34 (d, 2H), 7.44 (d, 1H), 7.51 (d, 1H), 8.52 (d, 2H), 13.17-13.63 (m, 1H).





94
5-(3,4-dichlorobenzyl)-2-(pyridin-2-ylmethyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (18% of theory; 8 eq. of 2-(pyridin-2- yl)ethanimidamide hydrochloride; 8.5 eq. sodium methoxide; methanol, 65° C., 16 h)

LC-MS (Method 1): Rt = 1.14 min; MS (ESpos): m/z = 414 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.92 (br. s, 2H), 4.16 (s, 2H), 7.13 (dd, 1H), 7.26-7.34 (m, 1H), 7.41 (d, 1H), 7.44-7.46 (m, 1H), 7.55 (d, 1H), 7.78 (dt, 1H), 8.49 (d, 1H), 13.28- 13.45 (m, 1H).





95
5-(3,4-dichlorophenoxy)-2-(1,2-oxazol-5- ylmethyl)-6-(trifluoromethyl)pyrimidin-4(3H)- one   embedded image
  (36% of theory; 4 eq. of 2-(1,2-oxazol-5- yl)ethanimidamide; 5 eq. of potassium carbonate; dioxane, 85° C., 1.5 h)

LC-MS (Method 1): Rt = 1.11 min; MS (ESpos): m/z = 406 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.12 (s, 2H), 6.65 (d, 1H), 7.11 (dd, 1H), 7.45 (d, 1H), 7.58 (d, 1H), 8.91 (d, 1H), 13.74 (br. s, 1H).





96
5-(3,4-dichlorobenzylylmethyl)-6- (trifluoromethyl)pyrimidin-4(3H)- one   embedded image
  (58% of theory; 4 eq. of 2-(1,2-oxazol-5- yl)ethanimidamide; 5 eq. of potassium carbonate; dioxane, 85° C., 1.5 h)

LC-MS (Method 1): Rt = 1.13 min; MS (ESpos): m/z = 404 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.91 (s, 2H), 4.10 (s, 2H), 6.62 (d, 1H), 7.14 (dd, 1H), 7.44 (d, 1H), 7.54 (d, 1H), 8.89 (d, 1H), 13.44 (br. s, 1H).





97
5-(3,4-dichlorobenzyl)-2-(3-thienylmethyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (58% of theory; 3 eq. of 2-(thiophen-3- yl)ethanimidamide acetate; 4 eq. of potassium carbonate; dioxane, 101° C., 16 h)

LC-MS (Method 1): Rt = 1.27 min; MS (ESpos): m/z = 419 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.88 (s, 2H), 3.94 (s, 2H), 7.08 (d, 1H), 7.12 (dd, 1H), 7.33-7.37 (m, 1H), 7.43 (d, 1H), 7.49-7.53 (m, 2H), 13.15-13.40 (m, 1H).





98
5-(3,4-dichlorobenzyl)-2- (pyridin-3-ylmethyl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (34% of theory; 8 eq. of 2-(pyridin-3- yl)ethanimidamide hydrochloride; 8.5 eq of sodium methoxide; methanol, 65° C., 16 h)

LC-MS (Method 1): Rt = 0.99 min; MS (ESpos): m/z = 414 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.89 (s, 2H), 3.99 (s, 2H), 7.13 (dd, 1H), 7.33-7.39 (m, 1H), 7.44 (d, 1H), 7.52 (d, 1H), 7.74 (d, 1H), 8.48 (dd, 1H), 8.56 (d, 1H), 13.17-13.57 (m, 1H).









EXAMPLE 99
5-(3,4-Dichlorobenzyl)-2-[(3-methyl-1H-1,2,4-triazol-5-yl)methyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


100 mg (0.28 mmol) of [5-(3,4-dichlorobenzyl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]acetonitrile (Example 74A) in 3 ml of methanol were stirred with 49 mg (0.9 mmol) of sodium methoxide for 30 minutes. 20.5 mg of acetohydrazide were then added, and the mixture was heated under reflux for 8 h. After a further 2.5 days at room temperature, 6.6 mg (0.3 mmol) of sodium hydride were added and the mixture was heated under reflux for another 8 h. 1 ml of water was then added, and the reaction was purified directly by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 35 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→80% A (25.00 min)→95% A (25.50-30.00 min)→10% A (30.50-35.00 min)]. The product-containing fractions were combined and concentrated by evaporation. The residue was re-purified by preparative thin-layer chromatography (silica gel, mobile phase cyclohexane/ethyl acetate 1:3). This gave 6 mg (5% of theory) of the title compound.


LC-MS (Method 1): Rt=0.98 min; MS (ESpos): m/z=418 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.77-4.04 (peak cluster), 4.30 (br. m), 7.04 (br. s), 7.14 (m), 7.21-7.35 (br. m), 7.43 (m), 7.54 (m), 13.45 (br. s) (tautomer mixture; integration not possible).


EXAMPLE 100
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-[(6-methoxypyridin-2-yl)amino]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 100 mg (0.26 mmol) of ethyl 2-[4-chloro-3-(trifluoromethyl)phenoxy]-4,4,4-trifluoro-3-oxobutanoate (Example 9A), 80 mg (0.39 mmol) of 1-(6-methoxypyridin-2-yl)guanidine (Example 33A) and 92 μl of N,N-diisopropylethylamine in 1.5 ml of DMF was stirred at 110° C. for 3 h. After cooling, the reaction mixture was purified directly by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% of formic acid). The product-containing fractions were combined and concentrated by evaporation. This gave 40 mg (32% of theory) of the title compound.


LC-MS (Method 1): Rt=1.27 min; MS (ESpos): m/z=481.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.91 (s, 3H), 6.59 (d, 1H), 6.88 (d, 1H), 7.42 (dd, 1H), 7.53 (d, 1H), 7.67 (d, 1H), 7.79 (t, 1H), 11.24 (s, 1H), 13.62 (s, 1H).


The Exemplary compounds below were prepared analogously to Example 100 from the guanidines mentioned and the appropriate phenoxy-substituted trifluoromethyl keto esters:











TABLE 22





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







101
5-(3,4-dichlorophenoxy)-2-[(6-methoxypyridin- 2-yl)amino]-6-(trifluoromethyl)pyrimidin-4(3H)- one   embedded image
  (21% of theory; 1.5 eq. of 1-(6-methoxypyridin- 2-yl)guanidine; 2 eq. of DIPEA; DMF, 110° C., 3 h)

LC-MS (Method 1): Rt = 1.25 min; MS (ESpos): m/z = 447.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.91 (s, 3H), 6.59 (d, 1H), 6.88 (d, 1H), 7.12 (dd, 1H), 7.43 (d, 1H), 7.57 (d, 1H), 7.79 (t, 1H), 11.22 (s, 1H), 13.59 (s, 1H).





102
5-(3,4-dichlorophenoxy)-2-[(3-methoxypyridin- 2-yl)amino]-6-(trifluoromethyl)pyrimidin-4(3H)- one   embedded image
  (27% of theory; 2 eq. of 1-(3-methoxypyridin-2- yl)guanidine (preparation described in Bioorg. Med. Chem. Lett. 2002, 12 (2), 181-184); 3 eq. of DIPEA; DMF, 110° C., 20 h)

LC-MS (Method 1): Rt = 1.23 min; MS (ESpos): m/z = 447.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.91 (s, 3H), 7.06-7.25 (m, 2H), 7.43 (d, 1H), 7.56 (d, 2H), 7.97 (dd, 1H), 10.08 (s, 1H), 14.26 (br. s, 1H).





103
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-[(3- methoxypyridin-2-yl)amino]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (33% of theory; 2 eq. of 1-(3-methoxypyridin-2- yl)guanidine (preparation described in Bioorg. Med. Chem. Lett. 2002, 12 (2), 181-184); 3 eq. of DIPEA; dioxane, reflux, 10 h)

LC-MS (Method 1): Rt = 1.26 min; MS (ESpos): m/z = 481.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.91 (s, 3H), 7.20 (dd, 1H), 7.42 (dd, 1H), 7.49-7.59 (m, 2H), 7.66 (d, 1H), 7.92-8.01 (m, 1H), 10.13 (s, 1H), 14.29 (br. s, 1H).









EXAMPLE 104
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-(1H-pyrazol-4-ylamino)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


5 ml of toluene were added to 100 mg (0.22 mmol) of 5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(methylsulphonyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 40A) and 57 mg (0.67 mmol) of 4-amino-1H-pyrazole, and the mixture was then re-concentrated under reduced pressure. A drop of DMSO was added to the residue, and the mixture was then stirred at 150° C. for 1 h. The mixture was then purified directly by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 45 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: water; gradient: 10% A (5.00 min)→95% A (35.00-40.00 min)→10% A (40.50-45.00 min)]. This gave 30 mg (31% of theory) of the title compound.


LC-MS (Method 1): Rt=1.03 min; MS (ESpos): m/z=440.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=7.37 (m, 1H), 7.48 (d, 1H), 7.64 (m, 2H), 7.92 (br. s, 1H), 9.10 (br. s, 1H), 11.98 (br. s, 1H), 12.66 (br. s, 1H).


The Exemplary compounds below were prepared analogously to Example 104 from the appropriate 2-methylsulphonyl-substituted pyrimidinones and the respective amine components:











TABLE 23





Example
IUPAC name/structure



No.
(yield)
Analytical data







105
5-[4-chloro-3- (trifluoromethypbenzyl]-2-(1H- pyrazol-4-ylamino)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (51% of theory)

LC-MS (Method 1): Rt = 1.05 min; MS (ESpos): m/z = 438 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.88 (s, 2H), 7.46 (d, 1H), 7.61 (d, 1H), 7.68 (s, 1H), 8.88-9.30 (m, 1H), 11.48-11.83 (m, 1H), 12.37-12.80 (m, 1H).





106
5-(3,4-dichlorophenoxy)-2- [(1-methyl-1H-1,2,4- triazol- 3-yl)amino]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (20% of theory; without addition of DMSO)

LC-MS (Method 1): Rt = 0.99 min; MS (ESpos): m/z = 421 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.70 (s, 3H), 6.86 (dd, 1H), 7.12 (d, 1H), 7.52 (d, 1H), 8.97 (s, 1H), 9.12- 9.24 (m, 1H).









EXAMPLE 107
2-(4-Amino-1H-pyrazol-1-yl)-5-[4-chloro-3-(trifluoromethyl)benzyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


46 mg (0.53 mmol) of 4-amino-1H-pyrazole were dissolved in 2.4 ml of DMF and stirred with 21 mg (0.5 mmol) of sodium hydride (60% in paraffin) at 23° C. for 30 minutes. 100 mg (0.18 mmol) of 5-[4-chloro-3-(trifluoromethyl)benzyl]-2-(methylsulphonyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 39A, purity 77%) were then added, and the mixture was stirred at 120° C. for 30 minutes. The mixture was then purified directly by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 35 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: water; gradient: 10% A (5.00 min)→80% A (25.00 min)→95% A (25.50-30.00 min)→10% A (30.50-35.00 min)]. The product-containing fractions were combined and concentrated. The residue was re-purified by preparative thin-layer chromatography (silica gel, mobile phase dichloromethane/methanol 20:1). This gave 41 mg (53% of theory) of the title compound.


LC-MS (Method 1): Rt=1.13 min; MS (ESpos): m/z=438 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=4.04 (s, 2H), 7.47 (d, 1H), 7.63 (d, 1H), 7.73 (d, 1H), 7.77-7.83 (m, 1H), 8.15-8.25 (m, 1H).


The following exemplary compounds were prepared in an analogous manner:











TABLE 24





Example
IUPAC name/structure



No.
(yield)
Analytical data







108
2-(4-amino-1H-pyrazol- 1-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (37% of theory)

LC-MS (Method 1): Rt = 1.07 min; MS (ESpos): m/z = 440 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.30 (br. m), 7.15 (br. m), 7.28 (br. m), 7.52 (br. m), 7.76 (br. m).





109
5-(3,4-dichlorophenoxy)-2-[(3-methyl-1,2- oxazol-5-yl)aminol-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (54% of theory)

LC-MS (Method 1): Rt = 1.16 min; MS (ESpos): m/z = 421 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.12 (s, 3H), 6.10 (s, 1H), 6.81 (dd, 1H), 6.97 (d, 1H), 7.48 (d, 1H), 10.09-10.25 (m, 1H).





110
5-(3,4-dichlorophenoxy)-2-[(1-methyl-1H- pyrazol-4-yl)amino]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (25% of theory)

LC-MS (Method 1): Rt = 1.07 min; MS (ESpos): m/z = 420.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.81 (s, 3H), 7.05 (dd, 1H), 7.37 (d, 1H), 7.51-7.56 (m, 2H), 7.85 (s, 1H), 9.30 (br. s, 1H).









EXAMPLE 111
5-(3,4-Dichlorobenzyl)-2-(2-oxoimidazolidin-1-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


80 mg (0.24 mmol) of 2-amino-5-(3,4-dichlorobenzyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 41A) were initially charged in 2 ml of DMF, 10.4 mg (0.26 mmol) of sodium hydride (60% in paraffin) were added and the mixture was stirred at 23° C. for 1 h. 71 mg (0.47 mmol) of 2-bromoethyl isocyanate were then added, and the mixture was stirred at 23° C. for 2 h. Water was then added, the mixture was stirred for 10 minutes and the precipitate formed was filtered off with suction. After concentration by evaporation, the mother liquor was purified by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 35 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→80% A (25.00 min)→95% A (25.50-30.00 min)→10% A (30.50-35.00 min)]. The product-containing fractions were combined and concentrated by evaporation. This gave 16 mg (17% of theory) of the title compound.


LC-MS (Method 3): Rt=2.57 min; MS (ESpos): m/z=407 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.42-3.51 (m, 2H), 3.86 (s, 2H), 3.92-3.99 (m, 2H), 7.13 (dd, 1H), 7.42 (d, 1H), 7.52 (d, 1H), 8.29 (s, 1H), 12.22 (s, 1H).


EXAMPLE 112
5-(3,4-Dichlorobenzyl)-2-(2-oxo-1,3-oxazolidin-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


100 mg (0.3 mmol) of 2-amino-5-(3,4-dichlorobenzyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 41A) were dissolved in 2 ml of dichloromethane and 0.04 ml of pyridine, and 58 mg (0.3 mmol) of 2-bromoethyl chloroformate were added dropwise at 0° C. The mixture was stirred initially at 0° C. for 10 min and then at 23° C. for 20 h. A further 0.63 mmol of 2-bromoethyl chloroformate were then added, and the mixture was stirred at 23° C. for 70 h. 153 mg (1.2 mmol) of N,N-diisopropylethylamine were then added, and the mixture was stirred at 23° C. for another 22 h. Another 58 mg (0.3 mmol) of 2-bromoethyl chloroformate and 38 mg of N,N-diisopropylethylamine were then added, and the mixture was stirred at 23° C. for a further 12 h. Finally, a further 174 mg (0.9 mmol) of 2-bromoethyl chloroformate and 114 mg (0.9 mmol) of N,N-diisopropylethylamine were then added, and the mixture was stirred at 23° C. for a further 4 h. The mixture was then concentrated by evaporation and the residue was purified by preparative HPLC [column: Chromatorex C18 10 μm, 250×30 mm; flow rate: 50 ml/min; run time: 35 min; detection: 210 nm; injection after 3 min of run time; mobile phase A: acetonitrile, mobile phase B: 0.1% aq. formic acid; gradient: 10% A (5.00 min)→80% A (25.00 min)→95% A (25.50-30.00 min)→10% A (30.50-35.00 min)]. The product-containing fractions were combined and concentrated by evaporation. This gave 50 mg (41% of theory) of the title compound.


LC-MS (Method 1): Rt=1.19 min; MS (ESpos): m/z=408 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.89 (s, 2H), 4.09 (t, 2H), 4.53 (t, 2H), 7.14 (dd, 1H), 7.41 (d, 1H), 7.54 (d, 1H).


EXAMPLE 113
2-(5-Aminopyridin-2-yl)-5-[4-chloro-3-(trifluoromethyl)phenoxy]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 283 mg (2.0 mmol) of potassium carbonate, 212 mg (1.2 mmol) of 5-aminopyridine-2-carboximidamide hydrochloride and 225 mg (0.41 mmol) of ethyl 2-[4-chloro-3-(trifluoromethyl)phenoxy]-4,4,4-trifluoro-3-oxobutanoate (Example 9A, purity 69%) in 3.3 ml of dioxane was stirred at 85° C. for 1 h. 1 ml of 1 N hydrochloric acid was then added, and the mixture was purified directly by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% of formic acid). This gave 142 mg (77% of theory) of the title compound.


LC-MS (Method 1): Rt=0.99 min; MS (ESpos): m/z=413.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=5.89 (s, 2H), 7.10-7.26 (m, 3H), 7.37 (m, 1H), 7.47 (m, 1H), 7.60 (m, 1H), 8.62 (s, 1H), 8.69 (dd, 1H), 8.89 (dd, 1H).


The Exemplary compounds listed in Table 25 were prepared analogously to Example 1 by reacting the appropriate amidines (carboximidamides) or their salts with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 25





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







114
5-[4-chloro-3-(trifluoromethyl)phenoxyl-2-[3- (methylsulphanyl)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (62% of theory; reaction time: 1 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 2 eq. of 3-(methylsulphanyl)pyridine-2- carboximidamide)

LC-MS (Method 1): Rt = 1.27 min; MS (ESpos): m/z = 482.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.49 (s, 3H), 7.50 (dd, 1H), 7.61-7.69 (m, 3H), 7.99 (dd, 1H), 8.51 (dd, 1H), 13.3 (br. s, 1H).





115
5-(3,4-dichlorophenoxy)-2-[3- (methylsulphanyl)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (64% of theory; reaction time: 1 h,85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 2 eq. of 3-(methylsulphanyl)pyridine-2- carboximidamide)

LC-MS (Method 6): Rt = 1.54 min; MS (ESpos): m/z = 448.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.49 (s, 3H), 7.20 (dd, 1H), 7.55-7.64 (m, 3H), 7.99 (dd, 1H), 8.51 (dd, 1H), 13.3 (br. s, 1H).





116
5-[4-chloro-3-(trifluoromethyl)phenoxyl-2-(3- methoxypyrazin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (25% of theory; reaction time: 4 h, 50° C.; solvent: dioxane; 3.5 eq. of potassium carbonate; 2 eq. of 3-methoxypyrazin-2- carboximidamide, CAS 1247573-36-9

LC-MS (Method 1): Rt = 1.15 min; MS (ESpos): m/z = 467.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.92 (s, 3H), 7.19 (dd, 1H), 7.33 (d, 1H), 7.63 (d, 1H), 8.24 (d, 1H), 8.31 (d, 1H).





117
5-(3,4-dichlorophenoxy)-2-(3-methoxypyrazin- 2-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (18% of theory; reaction time: 3 h, 75° C.; solvent: dioxane; 3 eq. of potassium carbonate; 1.1 eq. of 3-methoxypyrazin-2-carboximidamide, CAS 1247573-36-9

LC-MS (Method 1): Rt = 1.14 min; MS (ESpos): m/z = 433.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.07 (s, 3H), 7.20 (dd, 1H), 7.58-7.60 (m, 2H), 8.42 (d, 1H), 8.50 (d, 1H), 13.7 (br. s, 1H).





118
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2- (pyridazin-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (29% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of pyridazine-3-carboximidamide hydrochloride, CAS 405219-28-5)

LC-MS (Method 6): Rt = 1.40 min; MS (ESpos): m/z = 436.9 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.51 (dd, 1H), 7.64 (d, 1H), 7.71 (d, 1H), 8.01 (dd, 1H), 8.49 (dd, 1H), 9.48 (dd, 1H), 14.1 (br. s, 1H).





119
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(5- methoxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (63% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 5-methoxypyridine-2-carboximidamide, CAS 1179359-60-4

LC-MS (Method 1): Rt = 1.25 min; MS (ESpos): m/z = 466.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.96 (s, 3H), 7.49 (dd, 1H), 7.62 (d, 1H), 7.65-7.69 (m, 2H), 8.33 (d, 1H), 8.44 (d, 1H), 13.2 (br. s, 1H).





120
5-[4-chloro-3-(trifluoromethyl)phenoxy]-5′- methoxy-6-(trifluoromethyl)-2,4′-bipyrimidin- 4(3H)-one   embedded image
  (44% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 5-methoxypyrimidine-4- carboximidamide

LC-MS (Method 6): Rt = 1.39 min; MS (ESpos): m/z = 467.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.02 (s, 3H), 7.52 (dd, 1H), 7.67-7.70 (m, 2H), 8.96 (s, 1H), 8.99 (s, 1H), 13.9 (br. s, 1H).





121
2-(6-aminopyridazin-3-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (6% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 6-aminopyridazine-3-carboximidamide hydrochloride)

LC-MS (Method 6): Rt = 1.31 min; MS (ESpos): m/z = 418.0 (M+H)+





122
5-[4-chloro-3-(trifluoromethyl) phenoxy]-4′,6′- dimethoxy-6-(trifluoromethyl)- 2,2′-bipyrimidin-4(3H)-one   embedded image
  (10% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 4,6-dimethoxypyrimidine-2- carboximidamide hydrochloride)

LC-MS (Method 6): Rt = 1.52 min; MS (ESpos): m/z = 497.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.05 (s, 6H), 6.48 (s, 1H), 7.54 (dd, 1H), 7.66-7.71 (m, 2H), 13.6 (br. s, 1H).





123
2-(5-aminopyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (14% of theory; reaction time: 3 h, 75° C.; solvent: dioxane; 3 eq. of potassium carbonate; 1.1 eq. of 5-aminopyridine-2-carboximidamide hydrochloride)

LC-MS (Method 1): Rt = 1.14 min; MS (ESpos): m/z = 417.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.34 (s, 2H), 7.06-7.16 (m, 2H), 7.48 (d, 1H), 7.57 (d, 1H), 8.04-8.06 (m, 2H), 12.7 (br. s, 1H).





124
2-(3-chloropyridin-4-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (13% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-chloropyridine-4-carboximidamide CAS 1256825-46-3)

LC-MS (Method 6): Rt = 1.42 min; MS (ESpos): m/z = 436.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.17 (dd, 1H), 7.52 (d, 1H), 7.63 (d, 1H), 7.72 (d, 1H), 8.75 (d, 1H), 8.87 (s, 1H), 14.2 (br. s, 1H).





125
5-(3,4-dichlorophenoxy)-2-(pyridazin-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (6% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq.of potassium carbonate; 3 eq. of 3-pyridazinecarboximidamide hydrochloride, CAS 405219-28-5)

LC-MS (Method 1): Rt = 1.09 min; MS (ESpos): m/z = 403.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.20 (dd, 1H), 7.53 (d, 1H), 7.61 (d, 1H), 7.99 (dd, 1H), 8.48 (dd, 1H), 9.48 (dd, 1H), 14.1 (br. s, 1H).





126
5-(3,4-dichlorophenoxy)-2-(pyridazin- 4-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (6% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-pyridazinecarboximidamide hydrochloride, CAS 1426089-20-4)

LC-MS (Method 6): Rt = 1.30 min; MS (ESpos): m/z = 403.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.18 (dd, 1H), 7.50 (d, 1H), 7.62 (d, 1H), 8.27 (dd, 1H), 9.52 (dd, 1H), 9.79 (dd, 1H).





127
5-(3,4-dichlorophenoxy)-2-(pyrazin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (16% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of pyrazine-2-carboximidamide hydrochloride, CAS 138588-41-7)

LC-MS (Method 6): Rt = 1.41 min; MS (ESpos): m/z = 403.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.84 (dd, 1H), 7.03 (d, 1H), 7.50 (d, 1H), 8.65 (d, 1H), 8.70 (dd, 1H), 9.32 (d, 1H).





128
2-(4-aminopyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (38% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-amino-2-pyridinecarboximidamide, CAS 1342267-80-4)

LC-MS (Method 6): Rt = 1.15 min; MS (ESpos): m/z = 417.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.81 (dd, 1H), 6.89 (dd, 2H), 7.12 (d, 1H), 7.52 (d, 1H), 7.68 (d, 1H), 8.00- 8.03 (m, 3H), 13.07 (br. s, 1H).





129
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2- (pyridazin-4-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (43% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-pyridazinecarboximidamide hydrochloride, CAS 1426089-20-4)

LC-MS (Method 6): Rt = 1.35 min; MS (ESpos): m/z = 437.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.47 (dd, 1H), 7.62 (d, 1H), 7.72 (d, 1H), 8.27 (dd, 1H), 9.52 (dd, 1H), 9.79 (dd, 1H).





130
5-[4-chloro-3-(trifluoromethyl)phenoxyl-2-(4,5- dimethylpyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (20% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4,5-dimethylpyridine-2- carboximidamide)

LC-MS (Method 6): Rt = 1.63 min; MS (ESpos): m/z = 464.2 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.35 (s, 3H), 2.40 (s, 3H), 7.48 (dd, 1H), 7.62 (d, 1H), 7.68 (d, 1H), 8.13 (s, 1H), 8.51 (s, 1H), 13.3 (br. s, 1H).





131
2-(3-chloropyridin-4-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (40% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-chloropyridine-4-carboximidamide, CAS 1256825-46-3)

LC-MS (Method 6): Rt = 1.46 min; MS (ESpos): m/z = 469.8 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.47 (dd, 1H), 7.63 (d, 1H), 7.72-7.73 (m, 2H), 8.75 (d, 1H), 8.87 (s, 1H), 14.2 (br. s, 1H).





132
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2- (pyrazin-2-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (37% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of pyrazine-2-carboximidamide hydrochloride, CAS 138588-41-7)

LC-MS (Method 6): Rt = 1.44 min; MS (ESpos): m/z = 436.8 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.50 (dd, 1H), 7.63 (d, 1H), 7.70 (d, 1H), 8.86 (dd, 1H), 8.92 (d, 1H), 9.45 (d, 1H), 13.8 (br. s, 1H).





133
5-(3,4-dichlorophenoxy)-2-[5- (dimethylamino)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (45% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 5-(dimethylamino)pyridine-2- carboximidamide, CAS 1265277-51-7)

LC-MS (Method 1): Rt = 1.44 min; MS (ESpos): m/z = 445.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.10 (s, 6H), 7.15 (dd, 1H), 7.24 (dd, 1H), 7.49 (d, 1H), 7.85 (d, 1H), 8.14 (d, 1H), 8.17 (d, 1H), 12.8 (br. s, 1H).





134
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-[5- (dimethylamino)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (32% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 5-(dimethylamino)pyridine-2- carboximidamide, CAS 1265277-51-7)

LC-MS (Method 1): Rt = 1.34 min; MS (ESpos): m/z = 479.2 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.10 (s, 6H), 7.25 (dd, 1H), 7.46 (dd, 1H), 7.59 (d, 1H), 7.66 (d, 1H), 8.14- 8.19 (m, 2H), 12.8 (br. s, 1H).





135
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-[6- (dimethylamino)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (35% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-(dimethylamino)pyridine-2- carboximidamide, CAS 1341182-11-3)

LC-MS (Method 1): Rt = 1.40 min; MS (ESpos): m/z = 479.2 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.15 (s, 6H), 6.90 (d, 1H), 7.49-7.76 (m, 5H), 12.8 (br. s, 1H).





136
5-(3,4-dichlorophenoxy)-2-(1-methyl-1H- indazol-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (39% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 1-methyl-1H-indazole-3- carboximidamide)

LC-MS (Method 6): Rt = 1.61 min; MS (ESpos): m/z = 455.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.23 (s, 3H), 7.20 (dd, 1H), 7.40-7.44 (m, 1H), 7.54-7.60 (m, 3H), 7.84 (d, 1H), 8.37 (d, 1H), 13.6 (br. s, 1H).





137
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(1- methyl-1H-indazol-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (57% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 1-methyl-1H-indazole-3- carboximidamide)

LC-MS (Method 6): Rt = 1.62 min; MS (ESpos): m/z = 489.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.23 (s, 3H), 7.40-7.44 (m, 1H), 7.50- 7.52 (dd, 1H), 7.55-7.59 (m, 1H), 7.63 (d, 1H), 7.69 (d, 1H), 7.84 (d, 1H), 8.37 (d, 1H), 13.6 (br. s, 1H).





138
5-[4-chloro-3-(trifluoromethoxy)phenoxy]-2-(3- methoxypyrazin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (42% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-methoxypyridine-2-carboximidamide hydrochloride, CAS 1179362-06-1)

LC-MS (Method 1): Rt = 1.16 min; MS (ESpos): m/z = 482.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.89 (s, 3H), 7.24-7.27 (dd, 1H), 7.48-7.49 (m, 1H), 7.61-7.65 (m, 2H), 7.73 (dd, 1H), 8.31 (d, 1H), 13.7 (br. s, 1H).





139
5-(3,4-Dichlorophenoxy)-2-(1H-indazol-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (33% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 1H-indazole-3-carboximidamide, CAS 1518586-60-1)

LC-MS (Method 6): Rt = 1.54 min; MS (ESpos): m/z = 441.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.20 (dd, 1H), 7.38 (dd, 1H), 7.49- 7.60 (m, 3H), 7.69-7.71 (d, 1H), 8.38 (d, 1H), 13.6 (br. s, 1H), 14.1 (br. s, 1H).





140
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(1H- indazol-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (13% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 1H-indazole-3-carboximidamide, CAS 1518586-60-1)

LC-MS (Method 6): Rt = 1.56 min; MS (ESpos): m/z = 475.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.38 (dd, 1H), 7.49-7.53 (m, 2H), 7.64-7.71 (m, 3H), 8.38 (d, 1H), 13.6 (br. s, 1H), 14.1 (br. s, 1H).





141
2-(4-chloro-1-methyl-1H-indazol-3-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (35% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 4-chloro-1-methyl-1H-indazole-3- carboximidamide)

LC-MS (Method 6): Rt = 1.58 min; MS (ESpos): m/z = 489.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.21 (s, 3H), 7.18 (dd, 1H), 7.40 (d, 1H), 7.51-7.61 (m, 3H), 7.83 (d, 1H), 13.9 (br. s, 1H).





142
2-(4-chloro-1-methyl-1H-indazol-3-yl)-5-[4- chloro-3-(trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (30% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 4-chloro-1-methyl-1H-indazole-3- carboximidamide)

LC-MS (Method 6): Rt = 1.60 min; MS (ESpos): m/z = 523.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.21 (s, 3H), 7.40 (d, 1H), 7.49-7.55 (m, 2H), 7.64 (d, 1H), 7.70 (d, 1H), 7.83 (d, 1H), 14.0 (br. s, 1H).





143
5-(3,4-dichlorophenoxy)-5′-methoxy-6- (trifluoromethyl)-2,4′-bipyrimidin-4(3H)-one   embedded image
  (12% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 5-methoxypyrimidine-4- carboximidamide

LC-MS (Method 6): Rt = 1.34 min; MS (ESpos): m/z = 432.8 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.02 (s, 3H), 7.21 (dd, 1H), 7.58-7.60 (m, 2H), 8.96 (s, 1H), 8.99 (s, 1H), 13.9 (br. s, 1H).





144
5-(3,4-dichlorophenoxy)-2-(6-methoxypyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (18% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-methoxypyridine-2-carboximidamide hydrochloride, CAS 1179361-69-3)

LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 432.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.94 (s, 3H), 6.89-6.90 (m, 1H), 7.10 (d, 1H), 7.20 (d, 1H), 7.50 (d, 1H), 7.80-7.85 (m, 1H), 8.01 (dd, 1H), 9.67 (br. s, 1H).





145
5-(3,4-dichlorophenoxy)-2-(4- isopropoxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (25% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-isopropoxypyridine-2- carboximidamide, CAS 1179533-57-3)

LC-MS (Method 1): Rt = 1.32 min; MS (ESpos): m/z = 460.0 (M+H)+1H-NMR (500 MHz, DMSO-d6): δ = 1.35 (d, 6H), 4.89 (sept, 1H), 7.18 (dd, 1H), 7.26 (dd, 1H), 7.51 (d, 1H), 7.59 (d, 1H), 7.76 (d, 1H), 8.57 (d, 1H), 13.2 (br. s, 1H).





146
2-(6-tent-butoxypyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (74% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-tert-butoxypyridine-2- carboximidamide, CAS 1339092-12-4)

LC-MS (Method 1): Rt = 1.43 min; MS (ESneg): m/z = 472.1 (M−H)1H-NMR (500 MHz, DMSO-d6): δ = 1.61 (s, 9H), 7.00 (d, 1H), 7.21 (dd, 1H), 7.55-7.60 (m, 2H), 7.88-7.94 (m, 2H), 13.3 (br. s, 1H).





147
5-(3,4-dichlorophenoxy)-2-(4-methoxypyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (23% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-methoxypyridine-2-carboximidamide hydrochloride, CAS 1179361-66-0)

LC-MS (Method 1): Rt = 1.21 min; MS (ESpos): m/z = 432.0 (M+H)+1H-NMR (500 MHz, DMSO-d6): δ = 3.96 (s, 3H), 7.18 (dd, 1H), 7.28 (dd, 1H), 7.52 (d, 1H), 7.59 (d, 1H), 7.80 (d, 1H), 8.59 (d, 1H), 13.3 (br. s, 1H).





148
5-(3,4-dichlorophenoxy)-2-(5-methoxypyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (23% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 5-methoxypyridine-2-carboximidamide hydrochloride, CAS 1179359-60-4)

LC-MS (Method 1): Rt = 1.22 min; MS (ESpos): m/z = 432.0 (M+H)+1H-NMR (500 MHz, DMSO-d6): δ = 3.96 (s, 3H), 7.18 (dd, 1H), 7.51 (d, 1H), 7.58 (d, 1H), 7.66 (dd, 1H), 8.32 (d, 1H), 8.44 (d, 1H), 13.2 (br. s, 1H).





149
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(6- methoxypyridin- 2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (76% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-methoxypyridine-2-carboximidamide hydrochloride, CAS 1179361-69-3)

LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 466.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.08 (s, 3H), 7.08-7.11 (m, 1H), 7.52 (dd, 1H), 7.63 (d, 1H), 7.69 (d, 1H), 7.96-7.97 (m, 2H), 13.3 (br. s, 1H).





150
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(4- isopropoxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (64% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-isopropoxypyridine-2- carboximidamide, CAS 1179533-57-3)

LC-MS (Method 1): Rt = 1.33 min; MS (ESpos): m/z = 494.0 (M+H)+1H-NMR (500 MHz, DMSO-d6): δ = 1.35 (d, 6H), 4.89 (sept, 1H), 7.26 (dd, 1H), 7.48 (dd, 1H), 7.62 (d, 1H), 7.69 (d, 1H), 7.77 (d, 1H), 8.57 (d, 1H), 13.4 (br. s, 1H).





151
2-(6-tert-butoxypyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (78% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-tert-butoxypyridine-2- carboximidamide, CAS 1339092-12-4)

LC-MS (Method 1): Rt = 1.43 min; MS (ESneg): m/z = 506.1 (M−H)1H-NMR (400 MHz, DMSO-d6): δ = 1.61 (s, 9H), 7.00 (dd, 1H), 7.52 (dd, 1H), 7.64 (d, 1H), 7.69 (d, 1H), 7.88- 7.95 (m, 2H), 13.4 (br. s, 1H).





152
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(4- methoxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (79% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-methoxypyridine-2-carboximidamide hydrochloride, CAS 1179361-66-0)

LC-MS (Method 1): Rt = 1.23 min; MS (ESpos): m/z = 466.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.96 (s, 3H), 7.27 (dd, 1H), 7.48 (dd, 1H), 7.61 (d, 1H), 7.69 (d, 1H), 7.80 (d, 1H), 8.59 (d, 1H), 13.4 (br. s, 1H).





153
2-(6-aminopyridazin-3-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (7% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 6-aminopyridazine-3-carboximidamide hydrochloride)

LC-MS (Method 6): Rt = 1.35 min; MS (ESpos): m/z = 452.0 (M+H)+





154
2′-amino-5-(3,4-dichlorophenoxy)-6- (trifluoromethyl)-2,5′-bipyrimidin-4(3H)-one   embedded image
  (6% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq.of potassium carbonate; 3 eq. of 2-aminopyrimidine-5-carboximidamide, CAS 497099-72-6)

LC-MS (Method 6): Rt = 1.28 min; MS (ESpos): m/z = 418.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.14 (dd, 1H), 7.48 (d, 1H), 7.53 (br. s, 2H), 7.58 (d, 1H), 8.90 (s, 2H), 13.5 (br. s, 1H).





155
2′-amino-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6-(trifluoromethyl)- 2,5′-bipyrimidin-4(3H)-one   embedded image
  (6% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 2-aminopyrimidine-5-carboximidamide, CAS 497099-72-6)

LC-MS (Method 6): Rt = 1.32 min; MS (ESpos): m/z = 452.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.45 (dd, 1H), 7.54 (br. s, 2H), 7.58 (d, 1H), 7.68 (d, 1H), 8.91 (s, 2H), 13.5 (br. s, 1H).





156
5-(3,4-dichlorophenoxy)-4′,6′-dimethoxy-6- (trifluoromethyl)-2,2′-bipyrimidin-4(3H)-one   embedded image
  (10% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 4,6-dimethoxypyrimidine-2- carboximidamide hydrochloride)

LC-MS (Method 6): Rt = 1.50 min; MS (ESpos): m/z = 463.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.05 (s, 6H), 6.48 (s, 1H), 7.22 (dd, 1H), 7.57-7.61 (m, 2H), 13.6 (br. s, 1H).





157
5-(3,4-dichlorophenoxy)-2-(5,6-dimethylpyridin- 2-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (49% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 5,6-dimethylpyridin-2-carboximidamide, CAS 760907-02-6)

LC-MS (Method 6): Rt = 1.64 min; MS (ESpos): m/z = 430.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.32 (s, 3H), 2.38 (s, 3H), 7.52 (d, 1H), 7.63 (d, 1H), 7.87 (d, 1H), 7.95- 8.02 (m, 2H), 9.52 (br. s, 1H).





158
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(5,6- dimethylpyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (52% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 5,6-dimethylpyridin-2-carboximidamide, CAS 760907-02-6)

LC-MS (Method 6): Rt = 1.65 min; MS (ESpos): m/z = 464.2 (M+H)+





159
2-(4-tert-butoxypyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one embedded image
  (73% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-tert-butoxypyridine-2- carboximidamide hydrochloride, CAS 1179360-94-1)

LC-MS (Method 1): Rt = 1.38 min; MS (ESpos): m/z = 474.1 (M+H)+1H-NMR (500 MHz, DMSO-d6): δ = 1.52 (s, 9H), 7.19 (dd, 1H), 7.31 (d, 1H), 7.51 (d, 1H), 7.59 (d, 1H), 7.83 (s, 1H), 8.55 (d, 1H), 13.3 (br. s, 1H).





160
2-(4-tert-butoxypyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (76% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-tert-butoxypyridine-2- carboximidamide hydrochloride, CAS 1179360-94-1)

LC-MS (Method 1): Rt = 1.39 min; MS (ESpos): m/z = 508.1 (M+H)+1H-NMR (500 MHz, DMSO-d6): δ = 1.52 (s, 9H), 7.31 (dd, 1H), 7.49 (dd, 1H), 7.61 (d, 1H), 7.68 (d, 1H), 7.84 (d, 1H), 8.56 (d, 1H).





161
5-(3,4-dichlorophenoxy)-2-(1-oxidopyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (69% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 1-oxidopyridine-2-carboximidamide hydrochloride, CAS 845505-67-1)

LC-MS (Method 1): Rt = 1.10 min; MS (ESpos): m/z = 418.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.18 (dd, 1H), 7.50 (d, 1H), 7.61 (d, 1H), 7.79-7.82 (m, 2H), 8.56-8.58 (m, 1H), 8.65-8.67 (m, 1H), 15.8 (br. s, 1H).





162
2-(6-ethylpyridin-2-yl)-5-(3,4-dichlorophenoxy)- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (70% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 6-ethylpyridine-2-carboximidamide hydrochloride, CAS 112451-63-5)

LC-MS (Method 6): Rt = 1.68 min; MS (ESpos): m/z = 430.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.32 (t, 3H), 2.88 (q, 2H), 7.20 (dd, 1H), 7.54-7.60 (m, 3H), 7.99 (m, 1H), 8.15 (d, 1H), 13.1 (br. s, 1H).





163
5-(3,4-dichlorophenoxy)-2-(6- isopropoxypyridin-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (85% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 10 eq. of potassium carbonate; 3 eq. of 6-isopropoxypyridine-3- carboximidamide, CAS 1016838-19-9)

LC-MS (Method 6): Rt = 1.67 min; MS (ESpos): m/z = 460.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.34 (d, 6H), 5.36 (sept, 1H), 6.93 (d, 1H), 7.17 (dd, 1H), 7.50 (d, 1H), 7.59 (d, 1H), 8.33 (dd, 1H), 8.88 (d, 1H), 13.7 (br. s, 1H).





164
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2- (pyridin-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (22% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of pyridine-3-carboximidamide hydrochloride, CAS 7356-60-7)

LC-MS (Method 6): Rt = 1.40 min; MS (ESpos): m/z = 436.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.48 (dd, 1H), 7.61-7.64 (m, 2H), 7.70 (d, 1H), 8.44 (dd, 1H), 8.80 (dd, 1H), 9.23 (d, 1H), 13.95 (br. s, 1H).





165
5-[4-chloro-3-(trifluoromethyl)benzyl]-2- (pyridin-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one embedded image
  (14% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of pyridine-3-carboximidamide hydrochloride, CAS 7356-60-7)

LC-MS (Method 6): Rt = 1.43 min; MS (ESpos): m/z = 434.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.08 (s, 2H), 7.50 (dd, 1H), 7.61-7.65 (m, 2H), 7.78 (d, 1H), 8.45 (dd, 1H), 8.79 (dd, 1H), 9.24 (d, 1H), 13.66 (br. s, 1H).





166
5-(3,4-dichlorophenoxy)-2-[6- (methylamino)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (60% of theory; reaction time: 2 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-(methylamino)pyridine-2- carboximidamide, CAS 1343877-15-5)

LC-MS (Method 1): Rt = 1.26 min; MS (ESpos): m/z = 431.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.94 (s, 3H), 6.72 (d, 1H), 7.00 (br. s, 1H), 7.19 (dd, 1H), 7.49 (d, 1H), 7.54 (d, 1H), 7.57-7.62 (m, 2H), 12.6 (br. s, 1H).





167
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-[6- (methylamino)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (50% of theory; reaction time: 4 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-(methylamino)pyridine-2- carboximidamide, CAS 1343877-15-5)

LC-MS (Method 1): Rt = 1.28 min; MS (ESpos): m/z = 465.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.94 (s, 3H), 6.72 (d, 1H), 6.93 (br. s, 1H), 7.48-7.51 (m, 2H), 7.59-7.63 (m, 2H), 7.69 (d, 1H), 12.6 (br. s, 1H).





168
5-(3,4-dichlorophenoxy)-2-[4- (methylamino)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (56% of theory; reaction time: 4 h, 85° C.; solvent:dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-(methylamino)pyridine-2- carboximidamide, CAS 1342157-44-1)

LC-MS (Method 1): Rt = 0.93 min; MS (ESpos): m/z = 431.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.96 (s, 3H), 6.88 (d, 1H), 7.00 (dd, 1H), 7.25 (s, 1H), 7.56 (d, 1H), 7.63 br. s, 1H), 8.13 (br. s, 1H), 8.74 (br. s, 1H).





169
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-[4- (methylamino)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (53% of theory; reaction time: 4 h, 85° C.; solvent:dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-(methylamino)pyridine-2- carboximidamide, CAS 1343877-15-5)

LC-MS (Method 1): Rt = 0.96 min; MS (ESpos): m/z = 465.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.97 (s, 3H), 6.88 (d, 1H), 7.26 (d, 1H), 7.39 (s, 1H), 7.65 (d, 2H), 8.13 (br. s, 1H), 8.76 (br. s, 1H).





170
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(1,5- dimethyl-1H- pyrazol-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (88% of theory; reaction time: 2 h, 101° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 1,5-dimethyl-1H-pyrazole-3- carboximidamide, CAS 1517554-16-3)

LC-MS (Method 6): Rt = 1.50 min; MS (ESpos): m/z = 453.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.33 (s, 3H), 3.86 (s, 3H), 6.78 (s, 1H), 7.45 (dd, 1H), 7.58 (d, 1H), 7.66 (d, 1H), 13.3 (br. s, 1H).





171
5-(3,4-dichlorobenzyl-2-(tetrahydro-2H-pyran- 4-ylmethyl)-6-(trifluoromethyl)pyrimidin-4(3H)- one   embedded image
  (42% of theory; reaction time: 16 h, 65° C.; solvent: methanol; 8.5 eq. of sodium methoxide; 8 eq. of 2-(tetrahydro-2H-pyran-4- yl)ethaneimidamide, CAS 1247212-70-9)

LC-MS (Method 1): Rt = 1.19 min; MS (ESpos): m/z = 421.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.18-1.32 (m, 2H), 1.50-1.59 (m, 2H), 1.95-2.08 (m, 1H), 3.21-3.31 (m, 2H), 3.77-3.86 (m, 2H), 3.89 (s, 2H), 7.13 (dd, 1H), 7.44 (d, 1H), 7.53 (d, 1H), 13.11 (br. s, 1H).





172
5-(3,4-dichlorobenzyl-2-(thiophen-2-ylmethyl)- 6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (33% of theory; reaction time: 16 h, 101° C.; solvent: dioxane; 4 eq.of potassium carbonate; 3 eq. of 2-(thiophen-2-yl)ethaneimidamide acetate, CAS 28424-54-6)

LC-MS (Method 1): Rt = 1.26 min; MS (ESpos): m/z = 419.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.90 (s, 2H), 4.15 (s, 2H), 6.97-7.00 (m, 1H), 7.01-7.04 (m, 1H), 7.12 (dd, 1H), 7.40-7.44 (m, 2H), 7.52 (d, 1H), 13.4 (br. s, 1H).





173
5-(3,4-dichlorobenzyl)-2-(1H-1,2,4-triazol-1- ylmethyl)-6-(trifluoromethyl)pyrimidin-4(3H)- one   embedded image
  (26% of theory; reaction time: 16 h, 101° C.; solvent: dioxane; 4 eq.of potassium carbonate; 3 eq. of 2-(1H-1,2,4-triazol-1-yl)ethaneimidamide, CAS 1400872-25-4)

LC-MS (Method 1): Rt = 1.02 min; MS (ESpos): m/z = 404.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.91 (s, 2H), 5.41 (s, 2H), 7.12 (dd, 1H), 7.42 (d, 1H), 7.53 (d, 1H), 8.01 (s, 1H), 8.61 (s, 1H), 13.55 (br. s, 1H).





174
5-(3,4-dichlorobenzyl)-2-[(2-methyl-1,3-thiazol- 4-yl)methyl]-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (91% of theory; reaction time: 16 h, 101° C.; solvent: dioxane; 4 eq. of potassium carbonate; 3 eq.of 2-(2-methyl-1,3-thiazol-4- yl)ethaneimidamide, CAS 1343275-83-1)

LC-MS (Method 1): Rt = 1.20 min; MS (ESpos): m/z = 434.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.61 (s, 3H), 3.91 (s, 2H), 4.04 (s, 2H), 7.14 (dd, 1H), 7.34 (s, 1H), 7.44 (d, 1H), 7.53 (d, 1H), 13.31 (br. s, 1H).





175
5-(3,4-dichlorobenzyl)-2-(pyrrolidin-1-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (6% of theory; reaction time: 18 h, 101° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of pyrrolidine-1-carboximidamide hydrochloride, CAS 49755-46-6)

LC-MS (Method 1): Rt = 1.30 min; MS (ESpos): m/z = 392.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.83-1.95 (m, 4H), 3.40-3.51 (m, 4H), 3.77 (s, 2H), 7.11 (dd, 1H), 7.37 (d, 1H), 7.51 (d, 1H), 11.63 (br. s, 1H).





176
ethyl 1-[5-(3,4-dichlorobenzyl)-6-oxo-4- (trifluoromethyl)-1,6-dihydropyrimidin-2- yl]piperidine-4-carboxylate   embedded image
  (9% of theory; reaction time: 18 h, 101° C.; solvent: dioxane; 1.5 eq. of potassium carbonate; 1.5 eq. of ethyl 1-carbamimidoylpiperidine-4- carboxylate acetate, CAS 1208081-80-4)

LC-MS (Method 1): Rt = 1.35 min; MS (ESpos): m/z = 478.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.19 (t, 3H), 1.47-1.60 (m, 2H), 1.84- 1.93 (m, 2H), 3.02-3.13 (m, 2H), 3.77 (s, 2H), 4.08 (q, 2H), 4.18-4.33 (m, 2H), 7.11 (d, 1H), 7.39 (s, 1H), 7.51 (d, 1H), 11.80 (br. s, 1H).





177
5-(3,4-dichlorophenoxy)-2-(6-methoxyquinolin- 2-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (17% of theory; reaction time: 1.5 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 6-methoxyquinoline-2-carboximidamide hydrochloride, CAS 1267494-78-9)

LC-MS (Method 1): Rt = 1.41 min; MS (ESpos): m/z = 482.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.96 (s, 3H), 7.23 (dd, 1H), 7.51-7.63 (m, 4H), 8.12 (d, 1H), 8.37 (d, 1H), 8.52 (d, 1H), 13.39 (br. s, 1H).





178
5-(3,4-dichlorophenoxy)-2-(quinolin-2- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (31% of theory; reaction time: 1 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of quinoline-2-carboximidamide acetate, CAS 251294-66-3)

LC-MS (Method 1): Rt = 1.37 min; MS (ESpos): m/z = 452.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.23 (d, 1H), 7.58 (d, 1H), 7.62 (d, 1H), 7.78 (t, 1H), 7.93 (t, 1H), 8.14 (d, 1H), 8.23 (d, 1H), 8.42 (d, 1H), 8.66 (d, 1H), 13.54 (br. s, 1H).





179
5-(3,4-dichlorophenoxy)-2-(4-methoxy-1,2- oxazol-3-yl)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (13% of theory; reaction time: 4 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-methoxy-1,2-oxazole-3- carboximidamide hydrochloride (Example 121A))

LC-MS (Method 1): Rt = 1.12 min; MS (ESpos): m/z = 422.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.85 (s, 3H), 7.21 (dd, 1H), 7.57-7.60 (m, 2H), 9.10 (s, 1H), 13.94 (br. s, 1H).





180
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(4- methoxy-1,2-oxazol-3-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (61% of theory; reaction time: 4 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-methoxy-1,2-oxazole-3- carboximidamide hydrochloride (Example 121A))

LC-MS (Method 1): Rt = 1.17 min; MS (ESpos): m/z = 456.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.85 (s, 3H), 7.52 (dd, 1H), 7.65-7.69 (m, 2H), 9.11 (s, 1H), 13.99 (br. s, 1H).





181
2-(3-bromopyridin-2-yl)-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (65% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-bromopyridin-2-carboximidamide hydrochloride, CAS 1179360-60-1)

LC-MS (Method 1): Rt = 1.38 min; MS (ESpos): m/z = 481.9 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.18 (dd, 1H), 7.57-7.61 (m, 3H), 8.34 (dd, 1H), 8.74 (dd, 1H), 13.9 (br. s, 1H).





182
2-(3-bromopyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (59% of theory; reaction time: 3 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-bromopyridin-2-carboximidamide hydrochloride, CAS 1179360-60-1)

LC-MS (Method 1): Rt = 1.24 min; MS (ESpos): m/z = 515.9 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.50 (dd, 1H), 7.60 (dd, 1H), 7.66- 7.70 (m, 2H), 8.34 (dd, 1H), 8.74 (dd, 1H), 14.0 (br. s, 1H).





183
5-(3,4-dichlorophenoxy)-4′-methoxy-6′-methyl- 6-(trifluoromethyl)-2,2′-bipyrimidin-4(3H)-one   embedded image
  (5% of theory; reaction time: 14 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-methoxy-6-methylpyrimidine-2- carboximidamide hydrochloride, CAS 192203-63-7)

LC-MS (Method 1): Rt = 1.17 min; MS (ESpos): m/z = 447.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.51 (s, 3H), 4.08 (s, 3H), 7.03 (s, 1H), 7.21 (dd, 1H), 7.56-7.61 (m, 2H), 13.5 (br. s, 1H).





184
5-[4-chloro-3-(trifluoromethyl)phenoxy]-4′- methoxy-6′-methyl-6-(trifluoromethyl)-2,2′- bipyrimidin-4(3H)-one   embedded image
  (7% of theory; reaction time: 14 h, 85° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 4-methoxy-6-methylpyrimidine-2- carboximidamide hydrochloride, CAS 192203-63-7)

LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 481.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.51 (s, 3H), 4.08 (s, 3H), 7.04 (s, 1H), 7.53-7.68 (m, 3H), 13.6 (br. s, 1H).





185
5-(3,4-dichlorophenoxy)-6-(trifluoromethyl)-2- [3-(trifluoromethyl)pyridin-2-yl]pyrimidin- 4(3H)-one   embedded image
  (44% of theory; reaction time: 14 h, 100° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-(trifluoromethyl)pyridine-2- carboximidamide, CAS 1179533-41-5)

LC-MS (Method 1): Rt = 1.23 min; MS (ESpos): m/z = 470.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.17 (dd, 1H), 7.57-7.62 (m, 2H), 7.91 (dd, 1H), 8.49 (d, 1H), 9.02 (d, 1H), 14.1 (br. s, 1H).





186
5-[4-chloro-3-(trifluoromethyl)phenoxy)-6- (trifluoromethyl)-2-[3-(trifluoromethyl)pyridin- 2-yl]pyrimidin-4(3H)-one   embedded image
  (28% of theory; reaction time: 14 h, 100° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-(trifluoromethyl)pyridine-2- carboximidamide, CAS 1179533-41-5)

LC-MS (Method 1): Rt = 1.25 min; MS (ESpos): m/z = 504.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.49 (dd, 1H), 7.65 (d, 1H), 7.71 (d, 1H), 7.91 (dd, 1H), 8.49 (d, 1H), 9.02 (d, 1H), 14.2 (br. s, 1H).





187
5-(3,4-dichlorophenoxy)-2-(3-phenylpyridin-2- yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (77% of theory; reaction time: 14 h, 100° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-phenylpyridine-2-carboximidamide hydrochloride, CAS 1179361-80-8)

LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 478.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.29-7.31 (m, 2H), 7.36-7.44 (m, 4H), 7.51 (d, 1H), 7.68-7.75 (m, 2H), 8.03 (dd, 1H), 8.76 (dd, 1H), 13.9 (br. s, 1H).





188
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(3- phenylpyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (63% of theory; reaction time: 14 h, 100° C.; solvent: dioxane; 5 eq. of potassium carbonate; 3 eq. of 3-phenylpyridine-2-carboximidamide hydrochloride, CAS 1179361-80-8)

LC-MS (Method 1): Rt = 1.31 min; MS (ESpos): m/z = 512.2 (M+H)+





189
2-[3-(tert-butylsulphanyl)pyridin-2-yl)]-5-(3,4- dichlorophenoxy)-6-(trifluoromethyl)pyrimidin- 4(3H)-one   embedded image
  (81% of theory; reaction time: 1 h, 100° C.; solvent: dioxane; 5 eq. of potassium carbonate; 4 eq. of 3-(tert-butylsulphanyl)pyridine-2- carboximidamide, CAS 1521672-29-6)

LC-MS (Method 6): Rt = 1.60 min; MS (ESneg): m/z = 488.0 (M−H)1H-NMR (400 MHz, DMSO-d6): δ = 1.22 (s, 9H), 7.14 (dd, 1H), 7.49 (d, 1H), 7.61-7.67 (m, 2H), 8.18 (dd, 1H), 8.73 (dd, 1H), 13.9 (br. s, 1H).





190
2-[3-(tert-butylsulphanyl)pyridin-2-yl)]-5-[4- chloro-3-(trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (98% of theory; reaction time: 1 h, 100° C.; solvent: dioxane; 5 eq. of potassium carbonate; 4 eq. of 3-(tert-butylsulphanyl)pyridine-2- carboximidamide, CAS 1521672-29-6)

LC-MS (Method 6): Rt = 1.62 min; MS (ESneg): m/z = 522.0 (M−H)1H-NMR (400 MHz, DMSO-d6): δ = 1.23 (s, 9H), 7.48 (dd, 1H), 7.56 (d, 1H), 7.65-7.68 (m, 1H), 7.73 (d, 1H), 8.19 (dd, 1H), 8.74 (dd, 1H), 14.0 (br. s, 1H).





191
5-[4-chloro-3-(trifluoromethyl)phenoxy)-2-[4- (methylsulphanyl)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (78% of theory; reaction time: 14 h, 100° C.; solvent: dioxane; 5 eq. of potassium carbonate; 5 eq.of 4-(methylthio)pyridine-2-carboximidamide, CAS 1342120-26-6)

LC-MS (Method 1): Rt = 1.34 min; MS (ESpos): m/z = 482.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.62 (s, 3H), 7.47-7.55 (m, 2H), 7.62 (d, 1H), 7.69 (d, 1H), 8.10 (d, 1H), 8.57 (dd, 1H), 13.9 (br. s, 1H).





192
5-(3,4-dichlorophenoxy)-2-[4- (methylsulphanyl)pyridin-2-yl]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (83% of theory; reaction time: 14 h, 100° C.; solvent: dioxane; 5 eq. of potassium carbonate; 5 eq. of 4-(methylthio)pyridine-2-carboximidamide, CAS 1342120-26-6)

LC-MS (Method 1): Rt = 1.32 min; MS (ESpos): m/z = 448.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.62 (s, 3H), 7.18 (dd, 1H), 7.52-7.54 (m, 2H), 7.59 (d, 1H), 8.08 (d, 1H), 8.57 (d, 1H), 13.4 (br. s, 1H).





193
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(5- methylpyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (93% of theory; reaction time: 18 h, 80° C.; solvent: dioxane; 6 eq. of potassium carbonate; 5 eq. of 5-methylpyridine-2-carboximidamide, CAS 875401-87-9)

LC-MS (Method 1): Rt = 1.27 min; MS (ESpos): m/z = 450.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.44 (s, 3H), 7.50 (dd, 1H), 7.63 (d, 1H), 7.69 (d, 1H), 7.89-7.95 (m, 1H), 8.26 (d, 1H), 8.62 (s, 1H), 13.38 (br. s, 1H).





194
5-(4-chloro-3-methylphenoxy)-2-(3- methoxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (22% of theory; reaction time: 18 h, 80° C.; solvent: dioxane; 3 eq. of potassium carbonate; 2 eq. of 3-methoxypyridine-2-carboximidamide hydrochloride, CAS 1179362-06-1)

LC-MS (Method 1): Rt = 1.09 min; MS (ESpos): m/z = 412.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.31 (s, 3H), 3.89 (s, 3H), 6.93 (dd, 1H), 7.13 (d, 1H), 7.35 (d, 1H), 7.62 (dd, 1H), 7.73 (dd, 1H), 8.30 (dd, 1H), 13.58 (br. s, 1H).





195
2-(5-chloropyridin-2-yl)-5-[4-chloro-3- (trifluoromethyl)phenoxy]-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (39% of theory; reaction time: initially 2 h, 110° C., then 18 h, RT; solvent: dioxane; 6 eq. of potassium carbonate; 5 eq. of 5-chloropyridine- 2-carboximidamide hydrochloride, CAS 1179360-48-5)

LC-MS (Method 1): Rt = 1.28 min; MS (ESpos): m/z = 470.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.50 (dd, 1H), 7.63 (d, 1H), 7.69 (d, 1H), 8.23 (dd, 1H), 8.35 (dd, 1H), (dd, 1H), 13.49-13.73 (br. s, 1H).





196
5-(3-chloro-4-methylphenoxy)-2-(3- methoxypyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (44% of theory; reaction time: 18 h, 80° C.; solvent: dioxane; 3 eq. of potassium carbonate; 2 eq. of 3-methoxypyridine-2-carboximidamide hydrochloride, CAS 1179362-06-1)

LC-MS (Method 1): Rt = 1.10 min; MS (ESpos): m/z = 412.0 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.28 (s, 3H), 3.89 (s, 3H), 6.97 (dd, 1H), 7.21 (d, 1H), 7.30 (d, 1H), 7.62 (dd, 1H), 7.73 (dd, 1H), 8.30 (dd, 1H), 13.57 (br. s, 1H).





197
5-(4-chloro-3-methylphenoxy)-2-(3- chloropyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (29% of theory; reaction time: 18 h, 80° C.; solvent: dioxane; 3 eq. of potassium carbonate; 2 eq. of 3-chloropyridine-2-carboximidamide hydrochloride. CAS 477902-83-3)

LC-MS (Method 1): Rt = 1.23 min; MS (ESpos): m/z = 416.1 (M+H)+1H-NMR (500 MHz, CD3OD): δ = 2.34 (s, 3H), 6.83 (dd, 1H), 6.97 (d, 1H), 7.29 (d, 1H), 7.59 (dd, 1H), 8.09 (dd, 1H), 8.66 (dd, 1H).





198
5-(4-chloro-3-methylphenoxy)-2-(3- chloropyridin-2-yl)-6- (trifluoromethyl)pyrimidin-4(3H)-one   embedded image
  (69% of theory; reaction time: 18 h, 80° C.; solvent: dioxane; 3 eq. of potassium carbonate; 2 eq. of 3-chloropyridine-2-carboximidamide hydrochloride, CAS 477902-83-3)

LC-MS (Method 1): Rt = 1.23 min; MS (ESpos): m/z = 416.1 (M+H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.29 (s, 3H), 7.00 (dd, 1H), 7.25 (d, 1H), 7.31 (d, 1H), 7.69 (dd, 1H), 8.20 (dd, 1H), 8.71 (dd, 1H), 13.88 (br. s, 1H).









The Exemplary compounds listed in Table 26 were prepared analogously to Example 1 or Example 49 by reacting the appropriate amidines (carboximidamides) or their salts with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 26





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







199


embedded image


LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 493.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.14 (s, 3H), 7.48 (dd, 1H), 7.61 (d, 1H), 7.68 (d, 1H), 8.27 (m, 1H), 8.32 (d, 1H), 8.95 (d, 1H), 10.55 (s, 1H), 13.21-13.29 (br. s, 1H).





200


embedded image


LC-MS (Method 1): Rt = 1.16 min; MS (ESpos): m/z = 446.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.24 (s, 6H), 7.17 (m, 1H), 7.25 (d, 1H), 7.51 (d, 1H), 7.58 (d, 1H), 8.08 (d, 1H), 13.52 (br. s, 1H).





201


embedded image


LC-MS (Method 1): Rt = 1. 18 min; MS (ESpos): m/z = 480.0 (M + H)+1H-NMR (500 MHz, CD2Cl2): δ = 3.32 (br. s, 6H), 6.92-7.03 (m, 1H), 7.10-7.21 (m, 1H), 7.39 (br. s, 1H), 7.52 (d, 1H), 8.23 (br. s, 1H), 11.61 (br. s, 1H).





202


embedded image


LC-MS (Method 1): Rt = 1.15 min; MS (ESpos): m/z = 459.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.14 (s, 3H), 7.18 (dd, 1H), 7.52 (d, 1H), 7.58 (d, 1H), 8.26 (m, 1H), 8.29-8.33 (m, 1H), 8.95 (d, 1H), 10.55 (s, 1H), 13.21 (br. s, 1H).





203


embedded image


LC-MS (Method 1): Rt = 1.27 min; MS (ESpos): m/z = 486.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.19 (dd, 1H), 7.56 (d, 1H), 7.60 (d, 1H), 7.85 (dd, 1H), 8.14-8.17 (m, 1H), 8.80 (dd, 1H), 13.72 (br. s, 1H).





204


embedded image


LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 520.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.51 (dd, 1H), 7.64 (d, 1H), 7.70 (d, 1H), 7.85 (dd, 1H), 8.14-8.19 (m, 1H), 8.80 (dd, 1H), 13.77 (br. s, 1H).





205


embedded image


LC-MS (Method 1): Rt = 1.21 min; MS (ESpos): m/z = 456.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.23 (s, 3H), 7.20 (dd, 1H), 7.49-7.55 (m, 2H), 7.59 (d, 1H), 8.70 (m, 1H), 8.74 (dd, 1H), 13.83 (br. s, 1H).





206


embedded image


LC-MS (Method 1): Rt = 1.24 min; MS (ESpos): m/z = 490.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.23 (s, 3H), 7.49-7.54 (m, 2H), 7.61- 7.64 (m, 1H), 7.69 (d, 1H), 8.70 (dd, 1H), 8.74 (dd, 1H), 13.86 (br. s, 1H).





207


embedded image


LC-MS (Method 1): Rt = 1.33 min; MS (ESneg): m/z = 504.0 (M − H)1H-NMR (400 MHz, DMSO-d6): δ = 3.92 (s, 3H), 7.22 (dd, 1H), 7.47-7.54 (m, 2H), 7.65 (m, 1H), 7.71 (d, 1H), 8.16 (d, 1H).





208


embedded image


LC-MS (Method 1): Rt = 1.32 min: MS (ESpos): m/z = 472.2 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.92 (s, 3H), 7.19-7.24 (m, 2H), 7.48- 7.57 (m, 2H), 7.61 (d, 1H), 8.15 (d, 1H).





209


embedded image


LC-MS (Method 1): Rt = 1.16 min: MS (ESpos): m/z = 533.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.14 (s, 3H), 7.47-7.55 (m, 2H), 7.65 (d, 1H), 7.71 (d, 1H), 8.20 (d, 1H), 8.39-8.43 (m, 1H), 10.55 (br. s, 1H).





210


embedded image


LC-MS (Method 1): Rt = 1.16 min; MS (ESpos): m/z = 402.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.10-7.17 (m, 2H), 7.35-7.42 (m, 2H), 7.66-7.72 (m, 1H), 8.17-8.23 (m, 1H), 8.68-8.73 (m, 1H), 13.93 (br. s, 1H).





211


embedded image


LC-MS (Method 1): Rt = 1.21 min; MS (ESpos): m/z = 401.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 4.00 (s, 2H), 7.20-7.27 (m, 2H), 7.31- 7.39 (m, 2H), 7.63-7.71 (m, 1H), 8.14-8.22 (m, 1H), 8.66-8.72 (m, 1H), 13.66 (br. s, 1H).





212


embedded image


LC-MS (Method 6): Rt = 1.61 min; MS (ESpos): m/z = 431.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.35 (s, 3H), 2.40 (s, 3H), 7.18 (dd, 1H), 7.52 (d, 1H), 7.58 (d, 1H), 8.12 (s, 1H), 8.51 (s, 1H), 13.24 (br. s, 1H).









The Exemplary compounds listed in Table 27 were prepared analogously to Example 1 or Example 49 by reacting the appropriate amidines (carboximidamides) or their salts with the appropriate benzyl- or phenoxy-substituted trifluoromethyl keto esters:











TABLE 7





Example
IUPAC name/structure



No.
(yield, reaction conditions)
Analytical data







213


embedded image


LC-MS (Method 6): Rt = 1.33 min; MS (ESpos): m/z = 421.0 (M + H)+1H-NMR (500 MHz, DMSO-d6): δ = 3.79 (s, 3H), 7.16 (dd, 1H), 7.51 (d, 1H), 7.57 (d, 1H), 7.77 (s, 1H), 12.86 (br. s, 1H), 13.32 (br. s, 1H).





214


embedded image


LC-MS (Method 1): Rt = 1.09 min; MS (ESpos): m/z = 455.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.80 (s, 3H), 7.46 (dd, 1H), 7.59 (d, 1H), 7.66 (d, 1H), 7.77 (s, 1H), 12.88 (br. s, 1H), 13.32 (br. s, 1H).





215


embedded image


LC-MS (Method 1): Rt = 1.28 min; MS (ESneg): m/z = 432.0 (M − H)1H-NMR (400 MHz, DMSO-d6): δ = 0.95 (t, 3H), 1.66-1.78 (m, 2H), 2.85 (t, 2H), 6.84 (s, 1H), 7.18 (dd, 1H), 7.52 (d, 1H), 7.59 (d, 1H), 14.05 (br. s, 1H).





216


embedded image


LC-MS (Method 1): Rt = 1.29 min; MS (ESneg): m/z = 466.1 (M − H)1H-NMR (400 MHz, DMSO-d6): δ = 0.95 (t, 3H), 1.72 (m, 2H), 2.83 (t, 2H), 6.75 (s, 1H), 7.31-7.40 (m, 1H), 7.45-7.52 (m, 1H), 7.66 (d, 1H), 14.06 (br. s, 1H).





217


embedded image


LC-MS (Method 1): Rt = 1.24 min; MS (ESneg): m/z = 452.1 (M − H)1H-NMR (400 MHz, DMSO-d6): δ = 1.28 (t, 3H), 2.89 (q, 2H), 6.84 (s, 1H), 7.49 (dd, 1H), 7.63 (d, 1H), 7.69 (d, 1H), 14.09 (br. s, 1H).





218


embedded image


LC-MS (Method 1): Rt = 1.22 min; MS (ESpos): m/z = 420.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.28 (t, 3H), 2.88 (q, 2H), 6.84 (s, 1H), 7.19 (dd, 1H), 7.53 (d, 1H), 7.59 (d, 1H), 14.06 (br. s, 1H).





219


embedded image


LC-MS (Method 1): Rt = 1.27 min; MS (ESpos): m/z = 432.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.01 (m, 2H), 1.11-1.18 (m, 2H), 2.23-2.31 (m, 1H), 6.77 (s, 1H), 7.17 (d, 1H), 7.52 (s, 1H), 7.59 (d, 1H), 14.07 (br. s, 1H).





220


embedded image


LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 466.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.01 (m, 2H), 1.15 (m, 2H), 2.23-2.31 (m, 1H), 6.78 (s, 1H), 7.49 (dd, 1H), 7.62 (d, 1H), 7.69 (d, 1H), 14.03 (br. s, 1H).





221


embedded image


LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 470.0 (M + H)+1H-NMR (500 MHz, CD3OD): δ = 3.47 (s, 3H), 4.68 (s, 2H), 7.02 (s, 1H), 7.27 (m, 1H), 7.47 (m, 1H), 7.57 (d, 1H).





222


embedded image


LC-MS (Method 1): Rt = 1.14 min; MS (ESneg): m/z = 434.0 (M − H)1H-NMR (400 MHz, DMSO-d6): δ = 3.35 (s, 3H), 4.67 (s, 2H), 7.09 (s, 1H), 7.19 (dd, 1H), 7.53 (d, 1H), 7.60 (d, 1H), 14.26 (br. s, 1H).





223


embedded image


LC-MS (Method 1): Rt = 1.08 min; MS (ESpos): m/z = 371.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.29 (s, 3H), 6.93 (dd, 1H), 6.96 (br. s, 1H), 7.11 (d, 1H), 7.32 (d, 1H), 7.81-8.05 (m, 1H), 13.34 (br. s, 1H), 13.64 (br. s, 1H).





224


embedded image


LC-MS (Method 1): Rt = 1.07 min; MS (ESpos): m/z = 371.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.27 (s, 3H), 6.98 (m, 2H), 7.20 (d, 1H), 7.28 (d, 1H), 7.89-8.02 (m, 1H), 13.34 (br. s, 1H), 13.63 (br. s, 1H).





225


embedded image


LC-MS (Method 1): Rt = 1.15 min; MS (ESpos): m/z = 405.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.98 (s, 3H), 6.97 (d, 1H), 7.15 (dd, 1H), 7.50 (d, 1H), 7.57 (d, 1H), 7.91 (d, 1H), 13.39 (br. s, 1H).





226


embedded image


LC-MS (Method 1): Rt = 1.15 min; MS (ESpos): m/z = 439.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.98 (s, 3H), 6.98 (d, 1H), 7.46 (dd, 1H), 7.59 (d, 1H), 7.67 (d, 1H), 7.92 (d, 1H), 13.44 (br. s, 1H).





227


embedded image


LC-MS (Method 1): Rt = 1.34 min; MS (ESpos): m/z = 476.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.21 (dd, 1H), 7.56 (d, 1H), 7.60 (d, 1H), 14.08 (br. s, 1H).





228


embedded image


LC-MS (Method 1): Rt = 1.35 min; MS (ESpos): m/z = 510.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.52 (dd, 1H), 7.65 (d, 1H), 7.70 (d, 1H), 4.04 (br. s, 1H).





229


embedded image


LC-MS (Method 1): Rt = 1.16 min; MS (ESpos): m/z = 392.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.17 (d, 1H), 7.20 (dd, 1H), 7.54 (d, 1H), 7.60 (d, 1H), 9.22 (d, 1H), 14.15 (br. s, 1H).





230


embedded image


LC-MS (Method 1): Rt = 1.17 min; MS (ESpos): m/z = 426.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.17 (d, 1H), 7.50 (dd, 1H), 7.63 (d, 1H), 7.70 (d, 1H), 9.23 (d, 1H), 14.12 (br. s, 1H).





231


embedded image


LC-MS (Method 1): Rt = 1.26 min; MS (ESneg): m/z = 406.0 (M − H)1H-NMR (400 MHz, DMSO-d6): δ = 2.53 (d, 3H), 6.83 (d, 1H), 7.19 (dd, 1H), 7.54 (d, 1H), 7.59 (d, 1H), 14.05 (br. s, 1H).





232


embedded image


LC-MS (Method 1): Rt = 1.28 min; MS (ESneg): m/z = 439.0 (M − H)1H-NMR (400 MHz, DMSO-d6): δ = 2.53 (d, 3H), 6.84 (d, 1H), 7.50 (dd, 1H), 7.63 (d, 1H), 7.69 (d, 1H), 14.14 (br. s, 1H).





233


embedded image


LC-MS (Method 1): Rt = 1.16 min; MS (ESpos): m/z = 407.9 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.19 (dd, 1H), 7.54 (d, 1H), 7.59 (d, 1H), 8.03 (d, 1H), 9.27 (d, 1H), 13.85 (br. s, 1H).





234


embedded image


LC-MS (Method 1): Rt = 1.19 min; MS (ESpos): m/z = 442.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.50 (dd, 1H), 7.63 (d, 1H), 7.69 (d, 1H), 8.03 (d, 1H), 9.27 (d, 1H), 13.86 (br. s, 1H).





235


embedded image


LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 469.2 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.79 (s, 3H), 3.93 (s, 3H), 7.43 (dd, 1H), 7.56 (d, 1H), 7.66 (d, 1H), 8.32 (s, 1H), 12.15 (br. s, 1H).





236


embedded image


LC-MS (Method 1): Rt = 1.18 min; MS (ESpos): m/z = 435.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.78 (s, 3H), 3.93 (s, 3H), 7.12 (dd, 1H), 7.47 (d, 1H), 7.56 (d, 1H), 8.32 (s, 1H), 12.09 (br. s, 1H).





237


embedded image


LC-MS (Method 1): Rt = 1.13 min; MS (ESpos): m/z = 438.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.86 (s, 3H), 6.96-7.05 (m, 1H), 7.29 (br. s, 1H), 7.54 (d, 1H), 8.33 (s, 1H), 13.50 (br. s, 1H).





238


embedded image


LC-MS (Method 6): Rt = 1.44 min; MS (ESpos): m/z = 472.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.92 (s, 3H), 7.50 (dd, 1H), 7.64 (d, 1H), 7.68 (d, 1H), 8.46 (s, 1H), 13.45 (br. s, 1H).





239


embedded image


LC-MS (Method 6): Rt = 1.49 min; MS (ESpos): m/z = 458.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.49 (dd, 1H), 7.62 (d, 1H), 7.69 (d, 1H), 8.27 (s, 1H), 10.73 (br. s, 1H).





240


embedded image


LC-MS (Method 1): Rt = 1.21 min; MS (ESpos): m/z = 453.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.44 (t, 3H), 4.27 (q, 2H), 6.97 (d, 1H), 7.46 (dd, 1H), 7.60 (d, 1H), 7.67 (d, 1H), 7.97 (d, 1H), 13.39 (br. s, 1H).





241


embedded image


LC-MS (Method 1): Rt = 0.94 min; MS (ESpos): m/z = 405.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.16 (s, 3H), 6.73 (br. s, 1H), 6.82 (dd, 1H), 6.99 (d, 1H), 7.49 (d, 1H), 12.15 (br. s, 1H).





242


embedded image


LC-MS (Method 1): Rt = 0.98 min; MS (ESpos): m/z = 439.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.11-2.22 (m, 3H), 6.59-6.82 (m, 1H), 7.08 (dd, 1H), 7.18 (d, 1H), 7.59 (d, 1H), 11.95-12.27 (m, 1H).









The following compound was prepared in a manner analogous to Example 80:











TABLE 28





Example
IUPAC name/structure



No.
(starting material, reaction conditions, yield)
Analytical data







243


embedded image


LC-MS (Method 1): Rt = 1.28 min; MS (ESpos): m/z = 487.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.20 (dd, 1H), 7.53-7.55 (m, 1H), 7.61 (d, 1H), 7.68-7.73 (m, 2H), 7.76- 7.82 (m, 1H), 8.21-8.26 (m, 1H).









The following compound was prepared analogously to Example 88:











TABLE 29





Example
IUPAC name/structure



No.
(yield, starting material, reaction conditions)
Analytical data







244


embedded image


LC-MS (Method 1): Rt = 1.15 min; MS (ESpos): m/z = 450.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.45-1.59 (m, 2H), 1.82-1.92 (m, 2H), 3.03-3.14 (m, 2H), 3.77 (s, 2H), 4.18-4.33 (m, 2H), 7.11 (d, 1H), 7.39 (s, 1H), 7.51 (d, 1H), 11.78 (br. s, 1H), 12.31 (br. s, 1H).









The exemplary compounds below were prepared analogously to Example 104 from the appropriate 2-methylsulphonyl-substituted pyrimidinones and the respective amine components:











TABLE 30





Example
IUPAC name/structure



No.
(yield, starting material)
Analytical data







245


embedded image


LC-MS (Method 1): Rt = 1.12 min; MS (ESpos): m/z = 410.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.81-1.91 (m, 1H), 2.14-2.26 (m, 1H), 3.62 (dd, 1H), 3.67-3.75 (m, 1H), 3.76-3.88 (m, 2H), 4.34-4.43 (m, 1H), 7.03 (dd, 1H), 7.60-7.30 (br. s, 1H), 7.34 (d, 1H), 7.53 (d, 1H), 11.50 (br. s, 1H).





246


embedded image


LC-MS (Method 1): Rt = 1.19 min; MS (ESpos): m/z = 386.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.54-3.64 (m, 2H), 4.06-4.14 (m, 2H), 6.83 (dd, 1H), 7.03 (d, 1H), 7.51 (d, 1H), 8.52 (br. s, 1H), 8.84 (br. s, 1H), 9.90 (br. s, 1H).









The following exemplary compounds were obtained in analogy to Example 107:











TABLE 31





Example
IUPAC name/structure



No.
(yield, starting material)
Analytical data







247


embedded image


LC-MS (Method 6): Rt = 1.36 min; MS (ESpos): m/z = 426.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.08 (dd, 1H), 7.37 (d, 1H), 7.58 (d, 1H), 9.33 (s, 1H).





248


embedded image


LC-MS (Method 1): Rt = 1.26 min; MS (ESpos): m/z = 437.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 2.48 (s, 3H), 7.15 (dd, 1H), 7.49 (d, 1H), 7.58 (d, 1H), 8.01 (s, 1H), 9.33 (s, 1H).





249


embedded image


LC-MS (Method 1): Rt = 1.17 min; MS (ESpos): m/z = 421.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.82 (s, 3H), 7.16 (dd, 1H), 7.50 (d, 1H), 7.58 (d, 1H), 7.86 (s, 1H), 8.16 (s, 1H).





250


embedded image


LC-MS (Method 1): Rt = 1.28 min; MS (ESpos): m/z = 449.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.29 (d, 6H), 4.46 (sept, 1H), 7.16 (dd, 1H), 7.50 (d, 1H), 7.58 (d, 1H), 7.84 (s, 1H), 8.14 (s, 1H).





251


embedded image


LC-MS (Method 1): Rt = 1.29 min; MS (ESpos): m/z = 448.2 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.36 (s, 9H), 7.09-7.13 (dd, 1H), 7.41 (d, 1H), 7.58 (d, 1H), 9.16 (s, 1H).





252


embedded image


LC-MS (Method 6): Rt = 1.51 min; MS (ESpos): m/z = 458.8 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.10-7.15 (m, 2H), 7.44 (d, 1H), 7.59 (d, 1H), 8.73-8.74 (m, 1H).





253


embedded image


LC-MS (Method 6): Rt = 1.51 min; MS (ESpos): m/z = 458.8 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 7.15 (dd, 1H), 7.45 (d, 1H), 7.59 (d, 1H), 8.39 (s, 1H), 9.11 (s, 1H).





254


embedded image


LC-MS (Method 6): Rt = 1.32 min; MS (ESpos): m/z = 437.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 3.90 (s, 3H), 7.15 (dd, 1H), 7.47 (d, 1H), 7.58 (d, 1H), 7.83 (br. s, 2H).









EXAMPLE 255
5-(3,4-Dichlorophenoxy)-2-(1H-1,2,4-triazol-1-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


Under argon, 34 mg of 1,2,4-triazole (0.496 mmol) and 5 pellets of molecular sieve (4A) were initially charged in 3 ml of dioxane, the mixture was cooled to −78° C. and 30 μl of glacial acetic acid were added. The mixture was subsequently warmed to 0° C. and 100 mg (0.248 mmol) of 5-(3,4-dichlorophenoxy)-2-(methylsulphonyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 38A) were then added. In a microwave apparatus, the mixture was heated at 150° C. for 4 h. The reaction mixture was then filtered and purified directly by preparative HPLC (column: Chromatorex C18 10 μm, 30×125 mm; mobile phase: acetonitrile/0.1% aq. TFA). The product fractions were concentrated and lyophilized. This gave 16 mg (16% of theory) of the title compound.


LC-MS (Method 1): Rt=1.19 min; MS (ESpos): m/z=392.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=7.13 (m, 1H), 7.45 (d, 1H), 7.59 (m, 1H), 8.36 (s, 1H), 9.31 (s, 1H).


EXAMPLE 256
2-(3-Amino-1H-pyrazol-1-yl)-5-(3,4-dichlorophenoxy)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


50 mg of 5-(3,4-dichlorophenoxy)-2-(3-nitro-1H-pyrazol-1-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one were dissolved in 1 ml of THF, 5 mg of palladium on activated carbon (10%) were added and the mixture was hydrogenated at RT for 12 h. Another 10 mg of Pd/C (10%) were then added, and the mixture was hydrogenated for a further 24 h. The mixture was then filtered through a syringe filter, the filtrate was concentrated and the residue was purified by preparative HPLC (column: Chromatorex C18 10 μm, 30×125 mm; mobile phase: acetonitrile/0.1% aq. TFA). The product fractions were concentrated and lyophilized. This gave 25.2 mg (42% of theory) of the title compound.


LC-MS (Method 4): Rt=2.06 min; MS (ESpos): m/z=406.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=6.01 (d, 1H), 7.14 (m, 1H), 7.47 (d, 1H), 7.56 (m, 1H), 8.20 (s, 1H).


The following compounds were prepared in a manner analogous to Example 47:











TABLE 32





Example
IUPAC name/structure



No.
(yield, starting material, reaction conditions)
Analytical data







257


embedded image


LC-MS (Method 1): Rt = 1.06 min; MS (ESpos): m/z = 457.0 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 5.81 (br. s, 2H), 6.86-6.92 (m, 1H), 7.05-7.13 (m, 1H), 7.52 (d, 1H), 7.97- 8.00 (m, 1H), 8.05 (d, 1H), 8.11-8.15 (m, 1H).





258


embedded image


LC-MS (Method 1): Rt = 1.21 min; MS (ESpos): m/z = 442.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 6.97-7.07 (m, 1H), 7.27 (br. s, 1H), 7.55 (d, 1H), 7.61 (dd, 1H), 8.64 (s, 1H), 8.69 (d, 1H), 8.94 (d, 1H).









EXAMPLE 259
5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-(piperidin-4-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 85 mg (0.2 mmol) of tert-butyl 4-{5-[4-chloro-3-(trifluoromethyl)phenoxy]-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl}piperidine-1-carboxylate and 0.24 ml (3.13 mmol) of trifluoroacetic acid in 1 ml of dichloromethane was stirred at room temperature for 2 h. The mixture was then concentrated and the residue was purified by preparative HPLC (mobile phase: acetonitrile/water gradient with 0.1% trifluoroacetic acid). 69 mg (99% of theory) of the title compound were obtained.


LC-MS (Method 6): Rt=0.99 min; MS (ESpos): m/z=442.2 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=1.88-1.98 (m, 2H), 2.06-2.10 (m, 2H), 2.90-3.01 (m, 3H), 3.30-3.41 (m, 2H), 7.39 (dd, 1H), 7.54 (d, 1H), 7.68 (d, 1H), 8.28 (br. s, 1H), 8.64 (br. s, 1H), 13.5 (br. s, 1H).


EXAMPLE 260
Ethyl 3-[5-(3,4-dichlorophenoxy)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidin-2-yl]-1,2,4-oxadiazole-5-carboxylate x N,N-diisopropylethylamine



embedded image


A mixture of 150 mg (0.38 mmol) of 5-(3,4-dichlorophenoxy)-N′-hydroxy-6-oxo-4-(trifluoromethyl)-1,6-dihydropyrimidine-2-carboximidamide (Example 86A), 41 μl (0.36 mmol) of ethyl chloroxoacetate and 106 μl of N,N-diisopropylethylamine in 3 ml of dioxane was stirred at 90° C. for 4 h. The reaction mixture was then concentrated and the residue was purified by preparative HPLC. 66 mg (26% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=1.18 min; MS (ESpos): m/z=465.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=1.39 (t, 3H), 4.46 (q, 2H), 6.86 (dd, 1H), 7.09 (d, 1H), 7.51 (d, 1H), 8.20 (br. s, 1H).


EXAMPLE 261
5-(3,4-Dichlorobenzyl)-2-(morpholin-4-ylmethyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 100 mg (0.28 mmol) of 5-(3,4-dichlorobenzyl)-2-(hydroxymethyl)-6-(trifluoromethyl)pyrimidin-4(3H)-one and 30 μl (0.42 mmol) of thionyl chloride in 1 ml of dichloromethane was stirred at 23° C. for 24 h. The mixture was then concentrated on a rotary evaporator and the residue was dried under high vacuum. This residue was then dissolved in 1.3 ml of dioxane, and 46 mg (0.54 mmol) of morpholine and 80 mg (0.81 mmol) of potassium bicarbonate were added. The reaction mixture was stirred at room temperature overnight and then concentrated. The residue was purified by double preparative HPLC (1st column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient 90:10→10:90; 2nd column: Sunfire C18, 5 μm, 250×20 mm; mobile phase: acetonitrile/water with 0.1% TFA, 50:50). 22 mg (18% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=1.06 min; MS (ESpos): m/z=422.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=2.72-4.69 (m, 12H), 7.16 (dd, 1H), 7.44 (d, 1H), 7.55 (d, 1H), 13.3 (br. s, 1H).


The following compound was prepared analogously to Example 261:











TABLE 33





Example
IUPAC name/structure



No.
(yield, starting material, reaction conditions)
Analytical data







262


embedded image


LC-MS (Method 1): Rt = 0.87 min; MS (ESpos): m/z = 420.1 (M + H)+1H-NMR (400 MHz, DMSO-d6): δ = 1.08 (d, 3H), 1.36-1.43 (m, 1H), 1.68-1.72 (m, 2H), 1.91-1.96 (m, 1H), 2.44 (q, 1H), 2.66-2.71 (m, 1H), 3.07-3.12 (m, 1H), 3.43 (d, 1H), 3.85-3.90 (m, 3H), 7.13 (dd, 1H), 7.43 (d, 1H), 7.53 (d, 1H).









EXAMPLE 263
5-(3,4-Dichlorobenzyl)-2-[(1-methyl-5-oxo-4,5-dihydro-1H-1,2,4-triazol-3-yl)methyl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


A mixture of 96 mg (0.14 mmol) of 5-(3,4-dichlorobenzyl)-2-{[4-(2,4-dimethoxybenzyl)-1-methyl-5-oxo-4,5-dihydro-H-1,2,4-triazol-3-yl]methyl}-6-(trifluoromethyl)pyrimidin-4(3H)-one and 490 μl (9 mmol) of conc. sulphuric acid in 1.3 ml of acetic acid was stirred at 50° C. for 30 min. The mixture was then concentrated on a rotary evaporator. The residue was purified by preparative HPLC (column: Reprosil C18, 10 μm, 125×30 mm; mobile phase: acetonitrile with 0.1% formic acid/water with 0.1% formic acid; gradient 90:10-10:90). 28 mg (48% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=0.96 min; MS (ESpos): m/z=434.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.24 (s, 3H), 3.88 (s, 2H), 3.92 (s, 2H), 7.15 (dd, 1H), 7.43 (d, 1H), 7.55 (d, 1H), 11.45 (s, 1H), 13.3 (br. s, 1H).


EXAMPLE 264
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-(4-methoxy-1-methyl-1H-pyrazol-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


27 mg (0.059 mmol) of 5-[4-chloro-3-(trifluoromethyl)phenoxy)-2-(4-methoxy-1H-pyrazol-3-yl)-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 214) were dissolved in 1.5 ml of THF, and 3 mg (0.065 mmol) of sodium hydride (60% in paraffin) were added at 0° C., resulting in the evolution of hydrogen in an exothermic reaction. After 1 h of stirring at 23° C., 9 mg (0.0065 mmol) of iodomethane, dissolved in 0.5 ml of THF, were added, and the mixture was stirred at 23° C. for 18 h. The mixture was then diluted was ethyl acetate and washed with 1 N hydrochloric acid and water, and the organic phase was dried over magnesium sulphate. After removal of the drying agent by filtration, the mixture was concentrated under reduced pressure. The resulting oil was dissolved in acetonitrile and washed with n-hexane. Concentration under reduced pressure and drying under high vacuum gave 14 mg (51% of theory) of the target compound.


LC-MS (Method 1): Rt=1.13 min; MS (ESpos): m/z=469.0 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.78 (s, 3H), 3.89 (s, 3H), 7.39-7.47 (m, 1H), 7.53-7.59 (m, 1H), 7.66 (d, 1H), 7.76 (s, 1H), 12.78 (br. s, 1H).


EXAMPLE 265
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-[3-(methylsulphinyl)pyridin-2-yl]-6-trifluoromethyl)pyrimidin-4(3H)-one



embedded image


100 mg (0.208 mmol) of 5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-[3-(methylsulphanyl)pyridin-2-yl]-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 114) were dissolved in 1.5 ml of dichloromethane, and 77 mg (0.31 mmol) of 3-chloroperbenzoic acid were added at 0° C. After 4 h of stirring at 23° C., the mixture was diluted with dichloromethane and washed with saturated aqueous sodium sulphite solution, and the organic phase was dried over sodium sulphate. After the drying agent had been removed by filtration, the mixture was concentrated under reduced pressure and the residue was purified by preparative HPLC (mobile phase: acetonitrile/water gradient). 15 mg (13% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=1.08 min; MS (ESpos): m/z=498.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=2.92 (s, 3H), 7.51 (dd, 1H), 7.63 (d, 1H), 7.71 (d, 1H), 7.98 (dd, 1H), 8.66 (dd, 1H), 8.90 (dd, 1H), 13.68 (br. s, 1H).


EXAMPLE 266
5-[4-Chloro-3-(trifluoromethyl)phenoxy]-2-[3-(methylsulphonyl)pyridin-2-yl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


100 mg (0.208 mmol) of 5-[4-chloro-3-(trifluoromethyl)phenoxy]-2-[3-(methylsulphanyl)pyridin-2-yl]-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 114) were dissolved in 1.5 ml of dichloromethane, and 77 mg (0.31 mmol) of 3-chloroperbenzoic acid were added at 0° C. After 4 h of stirring at 23° C., the mixture was diluted with dichloromethane and washed with saturated aqueous sodium sulphite solution, and the organic phase was dried over sodium sulphate. After the drying agent had been removed by filtration, the mixture was concentrated under reduced pressure and the residue was purified by preparative HPLC (mobile phase: acetonitrile/water gradient). 14 mg (13% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=1.13 min; MS (ESpos): m/z=514.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.52 (s, 3H), 7.38-7.47 (m, 1H), 7.55-7.63 (m, 1H), 7.73 (d, 1H), 7.95 (dd, 1H), 8.54 (dd, 1H), 9.01-9.05 (m, 1H), 14.29 (br. s, 1H).


EXAMPLE 267
5-(3,4-Dichlorophenoxy)-2-[3-(methylsulphinyl)pyridin-2-yl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


47 mg (0.106 mmol) of 5-(3,4-dichlorophenoxy)-2-[3-(methylsulphanyl)pyridin-2-yl]-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 115) were dissolved in 3 ml of dichloromethane, and 39 mg (0.16 mmol) of 3-chloroperbenzoic acid were added at 0° C. After 4 h of stirring at 23° C., the mixture was diluted with dichloromethane and washed with saturated aqueous sodium sulphite solution, and the organic phase was dried over sodium sulphate. After the drying agent had been removed by filtration, the mixture was concentrated under reduced pressure and the residue was purified by preparative HPLC (mobile phase: acetonitrile/water gradient). 9 mg (18% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=1.03 min; MS (ESpos): m/z=464.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=2.92 (s, 3H), 7.21 (dd, 1H), 7.53 (d, 1H), 7.61 (d, 1H), 7.98 (dd, 1H), 8.66 (dd, 1H), 8.90 (dd, 1H), 13.64 (br. s, 1H).


EXAMPLE 268
5-(3,4-Dichlorophenoxy)-2-[3-(methylsulphonyl)pyridin-2-yl]-6-(trifluoromethyl)pyrimidin-4(3H)-one



embedded image


47 mg (0.106 mmol) of 5-(3,4-dichlorophenoxy)-2-[3-(methylsulphanyl)pyridin-2-yl]-6-(trifluoromethyl)pyrimidin-4(3H)-one (Example 115) were dissolved in 3 ml of dichloromethane, and 39 mg (0.16 mmol) of 3-chloroperbenzoic acid were added at 0° C. After 4 h of stirring at 23° C., the mixture was diluted with dichloromethane and washed with saturated aqueous sodium sulphite solution, and the organic phase was dried over sodium sulphate. After the drying agent had been removed by filtration, the mixture was concentrated under reduced pressure and the residue was purified by preparative HPLC (mobile phase: acetonitrile/water gradient). 16 mg (32% of theory) of the title compound were obtained.


LC-MS (Method 1): Rt=1.08 min; MS (ESpos): m/z=480.1 (M+H)+



1H-NMR (400 MHz, DMSO-d6): δ=3.52 (s, 3H), 7.12 (dd, 1H), 7.51 (d, 1H), 7.63 (d, 1H), 7.96 (dd, 1H), 8.54 (dd, 1H), 9.04 (dd, 1H), 14.23 (br. s, 1H).


B. ASSESSMENT OF PHARMACOLOGICAL EFFICACY

The pharmacological activity of the compounds according to the invention can be demonstrated by in vitro and in vivo studies, as known to the person skilled in the art. The application examples which follow describe the biological action of the compounds according to the invention, without restricting the invention to these examples.


ABBREVIATIONS AND ACRONYMS


















BSA
bovine serum albumin



DMEM
Dulbecco's modified Eagle's medium



DMSO
dimethyl sulphoxide



FCS
foetal calf serum



HEPES
4-(2-hydroxyethyl)piperazine-1-ethanesulphonic acid



LPS
lipopolysaccharide(s)



MEM
minimum essential medium



PBMC
peripheral blood mononuclear cells



PBS
phosphate-buffered saline solution



PEG
polyethylene glycol



RNA
ribonucleic acid(s)



Tris
tris(hydroxymethyl)aminomethane



v/v
ratio by volume (of a solution)



w/v
weight to volume ratio (of a solution)



WBC
white blood cells










B-1. Functional Ca2+ Release Test


The antagonistic action of test substances on CCR2 was determined in a functional Ca2+ release test. Binding of CCL2/MCP-1 to CCR2 leads to a change in the conformation of the receptor resulting in Gi/Gq protein activation and intracellular signal cascade. This involves, inter alia, an intracellular Ca2+ release. The test cell used was a Chem-1 cell line transfected with human CCR2 (ChemiSCREEN™ CCR2B Calcium-Optimized FLIPR Cell Line, Merck Millipore).


The test substances were dissolved in dimethyl sulphoxide (DMSO) at a concentration of 10 mM and serially diluted with DMSO in steps of 1:3.16 for a 10-point dose/activity analysis. According to the desired test concentrations, the substances were pre-diluted in Tyrode with 2 mM CaCl2 and 0.05% BSA.


The cells, cultivated in DMEM high glucose [supplemented with 10% FCS, 1 mM pyruvate, 15 mM HEPES, 500 μg/ml geniticin and non-essential amino acids (NEAA)], were sown at 5000 cells/25 μl in 384 well, μCLEAR/black cell culture plates from Greiner (#781092) and incubated at 37° C. for 24 h. The sowing medium consisted of DMEM high glucose [supplemented with 5% FCS, 1 mM pyruvate, 15 mM HEPES, 50 U/ml penicillin, 50 μg/ml streptomycin and non-essential amino acids (NEAA)]. The medium was then removed and the cells were charged for 60 min at 37° C. with Fluo-4 dye [25 μl Tyrode with 3 μM Fluo-4 AM (1 mM DMSO stock solution), 0.4 mg/ml Brilliant Black, 2.5 mM probenicid, 0.03% Pluronic F-127]. The cells were pre-incubated for 10 min with 10 μl of the test substances diluted in buffer, and 20 μl of agonist solution (MCP-1 in Tyrode with 0.05% BSA) were then added. MCP-1 was employed at the concentration which corresponds to the EC50 which had been determined in a preliminary test (usually about 5 nM). Ca2+ release was monitored over a period of 120 s in 1 s increments in a proprietary fluorescence imaging reader. The molar concentration of the test substance which caused 50% inhibition of the MCP-1 effect (IC50) was determined using a 4-parameter logistic function (Hill function).


The IC50 values determined in this manner from this assay for individual working examples are given in Table 1 below (in some cases as means of a plurality of independent individual determinations):












TABLE 1







Example No.
IC50 [nM]



















1
3.5



2
4.5



3
1.2



4
2.9



5
1.0



6
4.8



7
2.2



8
3.9



9
13



10
15



11
4.3



12
75



13
56



14
11



15
2.0



16
2.1



17
1.4



18
14



19
9.7



20
12



21
28



22
7.0



23
0.4



24
0.4



25
33



26
3.2



27
35



28
49



29
92



30
140



31
150



32
51



33
46



34
2.7



35
1.5



36
6.2



37
0.9



38
0.3



39
0.9



40
0.8



41
4.8



42
1.5



43
6.6



44
2.2



45
4.7



46
59



47
1.0



48
4.2



49
0.6



50
0.2



51
0.5



52
1.3



53
1.8



54
2.8



55
4.4



56
38



57
83



58
66



59
0.6



60
91



61
36



62
110



63
5.5



64
320



65
0.3



66
4.4



67
8.3



68
27



69
130



70
0.8



71
3.0



72
0.7



73
0.4



74
1.1



75
1.3



76
1.5



77
2.0



78
3.7



79
4.2



80
32



81
19



82
19



83
20



84
9.2



85
31



86
7.7



87
14



88
18



89
11



90
58



91
22



92
57



93
83



94
90



95
27



96
30



97
100



98
120



99
72



100
65



101
430



102
150



103
50



104
9.4



105
41



106
180



107
2.4



108
0.8



109
59



110
41



111
59



112
27



113
0.4



114
0.1



115
0.2



116
0.3



117
0.3



118
0.4



119
0.4



120
0.5



121
0.5



122
0.6



123
0.6



124
2.9



125
9.5



126
1120



127
2.1



128
3.2



129
222



131
2.8



132
3.8



133
0.8



134
2.1



135
14



136
39



137
16



138
1.4



139
0.7



140
0.8



141
5.1



142
3.3



143
1.1



144
17



145
322



146
404



147
121



148
42



149
0.7



150
23



151
96



152
8.2



153
0.7



154
37



155
23



156
0.7



157
14



158
8.7



159
62



160
62



161
2.3



162
1.9



163
129



164
16



165
14



166
8.8



167
2.7



168
25



169
36



170
0.7



171
400



172
62



173
260



174
150



175
360



176
45



177
51



178
33



179
1.8



180
1.0



181
0.7



182
0.3



183
2.9



184
0.8



185
27



186
4.8



187
16



188
1.5



189
53



190
62



191
5.0



192
11



193
0.7



194
0.3



195
1.1



196
1.7



197
1.3



198
9.5



199
0.6



200
1.2



201
0.7



202
0.9



203
15



204
9.2



205
26



206
12



207
81



208
77



209
22



210
3.0



211
1.5



212
2.6



213
0.6



214
0.6



215
7.9



216
4.6



217
1.8



218
3.9



219
7.2



220
3.5



221
1.3



222
6.5



223
0.1



224
0.3



225
0.3



226
0.5



227
6.4



228
5.3



229
0.5



230
0.4



231
0.1



232
0.3



233
0.2



234
0.3



235
20



236
692



237
1.0



238
0.9



239
2.6



240
0.5



241
0.8



242
1.8



243
16



244
18



245
160



246
240



247
123



248
2.2



249
0.9



250
4.1



251
76



252
113



253
240



254
15



255
38



256
0.8



257
2.8



258
9.9



259
624



260
13



261
360



262
160



263
180



264
0.4



265
12



266
1.8



267
103



268
5.8










B-2a. Functional β-Arrestin Recruiting Test with Human MCP-1


The antagonistic action of test substances on CCR2 was determined in a β-arrestin test. The PathHunter β-arrestin GPCR test system (DiscoveRx Corporation, Ltd.) is a cell-based functional method for detecting binding of β-arrestin to an activated receptor. The molecular basis is a 3-galactosidase complementation measured by the enzymatic conversion of a chemiluminescent substrate. The test cell used was a U2OS β-arrestin cell line transfected with murine CCR2 (93-0543C3, DiscoveRx Corporation, Ltd.).


The test substances were dissolved in dimethyl sulphoxide (DMSO) at a concentration of 10 mM and serially diluted with DMSO in steps of 1:3.16 for a 10-point dose/activity analysis. According to the desired test concentrations, the substances were pre-diluted in Tyrode with 2 mM CaCl2 and 0.05% BSA.


The cells, cultivated in MEM Eagle (supplemented with 10% FCS, 50 U/ml of penicillin, 50 μg/ml of streptomycin, 250 μg/ml of hygromycin and 500 μg/ml of geniticin), were sown at 2000 cells/25 μl in 384 well, μCLEAR/black cell culture plates from Greiner (#781092) and incubated at 37° C. for 24 h. The sowing medium consisted of Opti-MEM (supplemented with 1% FCS, 50 U/ml of penicillin and 50 μg/ml of streptomycin). The cells were pre-incubated for 10 min with 10 μl of the test substances diluted in buffer, and 10 μl of agonist solution [human MCP-1 (PeproTech, #300-04) in Tyrode with 0.05% BSA) were then added. The human MCP-1 was employed at the concentration which corresponds to the EC50 which had been determined in a preliminary test (usually about 3 nM). After 90 min of incubation at 37° C., the solution was removed, and recruitment of β-arrestin to CCR2 was detected with the aid of the PathHunter detection reagent (93-001, DiscoveRx Corporation, Ltd.) according to the instructions of the manufacturer. Luminescence was measured after an incubation time of 60 min using a proprietary luminescence imaging measuring instrument. The molar concentration of the test substance which caused 50% inhibition of the MCP-1 effect (IC50) was determined using a 4-parameter logistic function (Hill function).


The IC50 values determined in this manner from this assay for individual working examples are given in Table 2a below (in some cases as means of a plurality of independent individual determinations):












TABLE 2a







Example No.
IC50 [nM]



















1
110



2
88



3
31



4
87



5
13



6
150



7
120



8
180



9
180



10
140



11
28



12
100



13
120



14
25



15
30



16
21



17
43



18
140



19
300



20
380



21
230



22
400



23
25



24
8.2



25
790



26
190



27
480



28
1000



29
2600



30
530



31
1400



32
1100



33
650



34
110



35
35



36
140



37
40



38
5.1



39
20



40
12



41
410



42
44



43
220



44
74



45
86



46
230



47
3.6



48
30000



49
2.5



50
4.7



51
20



52
85



53
67



54
51



55
110



56
900



57
1300



58
1600



59
12



60
180



61
220



62
140



63
100



64
6500



65
1.8



66
110



67
150



68
310



69
1100



70
56



71
57



72
19



73
12



74
2.9



75
29



76
26



77
6.3



78
16



79
53



80
160



81
110



82
150



83
79



84
86



85
96



86
100



87
54



88
6100



89
370



90
620



91
270



92
220



93
370



94
420



95
77



96
160



97
490



98
460



99
630



100
920



101
11000



102
260



103
370



104
160



105
380



106
1400



107
28



108
2.7



109
480



110
730



111
360



112
380



113
8.3



171
1000



172
140



173
980



174
1100



175
1400



176
460



177
1500



178
535



243
300



244
1400



245
1400



246
1400



260
120



261
630



262
2500



263
680










B-2b. Functional β-Arrestin Recruiting Test with Murine MCP-1


The test was carried out in a manner identical to that described above under B-2a, but using murine MCP-1 (PeproTech, #250-10) as agonist.


The IC50 values from this assay for individual working examples are given in Table 2b below (in some cases as means of a plurality of independent individual determinations):












TABLE 2b







Example No.
IC50 [nM]



















2
1040



3
205



4
380



5
50



7
435



11
194



14
260



15
144



16
307



17
191



23
135



24
54



35
160



37
122



38
8.9



39
50



40
44



42
812



44
222



45
553



47
95



49
12



51
112



52
439



53
440



54
458



59
106



65
4.8



70
154



71
1080



73
37



74
59



75
102



76
111



77
18



78
50



79
195



83
308



95
449



107
53



108
27



113
16



114
6.1



115
7.6



116
37



117
36



118
73



119
185



120
44



121
149



122
164



123
28



124
320



125
475



126
13900



127
372



128
360



129
2710



131
158



132
259



133
21700



134
51



135
1570



136
1220



137
334



138
412



139
229



140
75



141
274



142
246



143
86



144
759



145
2810



146
3500



147
373



148
454



149
526



150
1340



151
3680



152
838



153
87



154
1930



155
1080



156
244



157
1510



158
983



159
2470



160
1960



161
432



162
1070



163
7680



164
798



165
3370



166
277



167
153



168
1210



169
624



170
84



179
73



180
105



181
7.9



182
8.4



183
145



184
221



185
405



186
513



187
1600



188
828



189
1030



190
2110



191
693



192
758



193
79



194
59



195
157



196
585



197
83



198
105



199
110



200
359



201
300



202
212



203
423



204
282



205
2490



206
1990



207
4490



208
9580



209
368



210
349



211
299



212
1700



213
49



214
8.5



215
1150



216
1320



217
874



218
584



219
1040



220
775



221
447



222
551



223
21



224
47



225
150



226
87



227
164



228
227



229
44



230
44



231
249



232
170



233
43



234
27



235
796



236
720



237
80



238
39



239
89



240
107



241
76



242
153



247
5530



248
1230



249
254



250
942



251
6760



252
6940



253
4230



254
613



255
1340



256
100



257
276



258
949



259
3530



264
53



265
257



266
196



267
679



268
484










B-3. Test of Selectivity for Human CC Receptors


The antagonistic effect of test substances on human CC receptors was determined in functional Ca2+ release tests using Ca2+-sensitive fluorescent dyes. The test cells used were Chem-1 or Chem-5 cell lines transfected with the respective receptor (ChemiSCREEN™ CCR Calcium-Optimized FLIPR Cell Lines, Merck Millipore; CCR1: HTS005C; CCR3: HTS008C; CCR4: HTS009C; CCR5 rhesus monkey: HTS010C; CCR6: HTS011C; CCR7: HTS012C; CCR8: HTS013C; CCR9: HTS036C; CCR10: HTS014C).


The substance test was carried out in a FLIPR tetra instrument (Molecular Devices). The agonist in question was added in a concentration corresponding to the EC80. Ca2+ release was measured over a period of 180 sec.


B-4. Test of Selectivity for Murine CC Receptors


The antagonistic effect of test substances on murine CC receptors was determined in the PathHunter β-arrestin GPCR test system (DiscoveRx Corporation, Ltd.). The test cells used were U2OS or CHO-K1β-arrestin cell lines transfected with the respective murine receptor (DiscoveRx Corporation, Ltd.; mCCR1: 93-0561C3; mCCR3: 93-0522C2; mCCR4: 93-0515C2; mCCR5: 93-0470C2; mCCR6: 93-0694C2; mCCR7: 93-0528C2; mCCR8: 93-0556C2; mCCR9: 93-0734C2).


The substance test was carried out with an EnVision microplate reader (Perkin Elmer) which detects the chemiluminescent conversion of the β-galactosidase substrate. The agonist in question was added in a concentration corresponding to the EC80.


B-5. Activity Test for CCR2 (Rat) and CCR5 (Rat)


The antagonistic effect of test substances on CCR2 (rat) and CCR5 (rat) was determined in functional Ca2+ release tests using the Ca2+-sensitive photoprotein aequorin [Vakili et al., J. Immunol. 167, 3406 (2001); Fichna et al., J. Pharmacol. Exp. Ther. 317, 1150 (2006); Silvano et al., Mol. Pharmacol. 78, 925 (2010)]. The test cells used were CHO-K1 cell lines transfected with the respective receptor and aequorin (Euroscreen SA; rCCR2: FAST-0616A; rCCR5: FAST-0617A).


Luminescent detection of Ca2+ release was carried out using a Functional Drug Screening System 6000 (FDSS 6000) luminometer (Hamamatsu). The agonist in question was added in a concentration corresponding to the EC80.


B-6. THP-1 Migration Assay


The migration of THP-1 cells is analysed using a CytoSelect 96-well cell migration assay (5 μm membrane pores), Fluormetric (BioCat GmbH) or a comparable assay, and the effect of test substances on the migration behaviour is investigated. Alternatively, macrophages are isolated from whole blood (canine, porcine or human) and used for carrying out a migration assay.


B-7. THP-1 Gene Expression Assay


THP-1 cells are incubated for 7-24 h with 9-cis-retinoic acid to initiate cell differentiation. During the incubation, test substance is added to the medium, and the RNA is then isolated (TRIzol®, Invitrogen). After work-up of the RNA and reverse transcription (ImProm-II™ Reverse Transcription System, Promega A3800), an MCP-1 gene expression analysis is carried out using TaqMan.


B-8. Human Whole Blood Assay (PBMC Assay)/MCP-1-Induced Gene Expression


The blood was removed into heparin monovettes (Sarstedt) and the blood was then collected and 2.5 ml each are pipetted into the wells of a 12-well plate. 2.5 μl of solvent or test substance solution are pipetted into each well, the contests of the individual wells are mixed for about 5 min on a plate shaker and the plates are then incubated in an incubator at 37° C. for 20 min. The hMCP-1 (100 ng/ml) was then added, followed by about 4 min of mixing on a plate shaker and subsequent incubation in an incubator at 37° C. for 4 h. The blood is then transferred into PAXgene® blood RNA tubes (PreAnalytix) and, after work-up of the RNA and reverse transcription (ImProm-II™ Reverse Transcription System, Promega A3800), a gene expression analysis is carried out using TaqMan.


B-9. Acute Myocardial Infarction (aMI) in the Rat


Male Wistar rats (280-300 g; Harlan Nederland) are anaesthetized with 160 mg/kg of ketamine and 8 mg/kg of xylazine, intubated, connected to a ventilation pump (ugo basile 7025 rodent; 0.4-0.5 litre/min, 60×/min) and ventilated with 60% compressed air/40% O2. The body temperature is maintained at 37-38° C. by a heating mat. If appropriate, 0.03 mg/kg s.c. of Temgesic® may be administered as analgesic. The area to be operated on is disinfected (for example with Cutasept®), the thorax of the animal is opened between the 3rd and the 4th rib and fixated using a rib spreader. The heart of the animal is exposed under the auricula atrii and a 5-0 Prolene thread is passed underneath about 2 mm from the end of the auricula atrii. Both ends of the thread are pushed into a PE50 plunger and the ends of the thread are coiled around a needle holder. Owing to the resulting tension, the coronary artery of the left ventricle (LAD) is clamped. A bulldog clamp is placed on top of the PE50 plunger and used to occlude the LAD (occlusion time 30 minutes). After this time, the bulldog clamp is loosened and the PE50 plunger is removed; the thread remains in place. The thorax is closed again, and the muscle layers and the epidermis are sutured using coated Vicryl L 5-0 (V990H). Antisedan® i.m. is then injected to reverse anaesthesia.


After 1-4 days of treatment with the test substance, the animals are again anaesthetized (2% isoflurane/compressed air/O2) and a pressure catheter (Millar SPR-320 2F) is inserted via the carotid artery into the left ventricle after measurement of the systemic blood pressure. The heart rate, left ventricular pressure (LVP), left-ventricular end-diastolic pressure (LVEDP), contractility (dp/dt) and relaxation rate (tau) are measured there and analysed with the aid of the Powerlab system (AD Instruments) and LabChart software. A blood sample is then taken to determine the plasma levels of the substance and plasma biomarkers, and the animals are sacrificed. Area at risk (the non-perfused area) and infarct size are determined by perfusion with Evans Blue (0.2%) and subsequent TTC staining.


B-10. Cronic Myocardial Infarction (cMI) in the Rat


Male Wistar rats (280-300 g; Harlan Nederland) are anaesthetized with 5% isoflurane in an anaesthesia cage, intubated, connected to a ventilation pump (ugo basile 7025 rodent; 0.4-0.5 litre/min, 60×/min) and ventilated with 5% enflurane/compressed air/O2. The body temperature is maintained at 37-38° C. by a heating mat. If appropriate, 0.03 mg/kg s.c. of Temgesic® may be administered as analgesic. The chest is opened laterally between the third and fourth ribs, and the heart is exposed. The coronary artery of the left ventricle (LAD) is permanently ligated with an occlusion thread (Prolene Ethicon 5-0, EH7401H) passed underneath shortly below its origin (below the left atrium). The thorax is closed again, and the muscle layers and the epidermis are sutured using coated Vicryl L 5-0 (V990H). The surgical suture is wetted with spray dressing (for example Nebacetin® N spray dressing, active ingredient neomycin sulphate), and anaesthesia is then terminated. Alternatively, the occlusion thread may initially be passed around the LAD without occluding it. After closure of the thorax and a healing phase (up to 1 week later), the LAD is then occluded by pulling the occlusion thread, which had been led outside of the body.


The animals are randomized by troponine determination and divided into individual treatment groups and a control group with no substance treatment. A further control included is a sham group in which only the surgical procedure, but not the LAD occlusion, was performed. Treatment with the test substance takes place over 8 weeks by gavage or by adding the test substance to the feed or drinking water.


After treatment for 8 weeks, the animals are again anaesthetized (2% isoflurane/compressed air/O2) and a pressure catheter (Millar SPR-320 2F) is inserted via the carotid artery into the left ventricle. The heart rate, left ventricular pressure (LVP), left-ventricular end-diastolic pressure (LVEDP), contractility (dp/dt) and relaxation rate (tau) are measured there and analysed with the aid of the Powerlab system (AD Instruments) and LabChart software. A blood sample is then taken to determine the plasma levels of the substance and plasma biomarkers, and the animals are sacrificed. The heart (heart chambers, left ventricle plus septum, right ventricle), liver, lung and kidney are removed and weighed.


B-11. Acute Lung Injury (ALI) in the Rat


Male Sprague Dawley rats (200-250 g; Charles River) are anaesthetized with 5% isoflurane in an anaesthesia cage. In the tolerance stage, the animals are intubated, with the aid of a guide wire, with a peripheral venous catheter (Brauniile, 16G), and the harmful substance (3 mg/kg of LPS in 100 μl of physiological saline) is administered via the tube. Control animals receive 100 μl of saline. 24 hours after administration of the harmful substance, a pulmonary lavage is carried out. Prior to the lavage, the animals are weighed again to determine the lung index (weight of the lung/body weight). For the lavage, the animals are anaesthetized with isoflurane. The trachea is prepared, and a Braunfile (16G) is inserted and fixed. Via the Brauniile, the lung is rinsed three times with 1.5 ml of physiological saline. The lavage is stored on ice, and the lavages of individual animals are combined and measured on a CellDyn 3700 to determine the number of inflammatory cells (leukocytes, neutrophiles, monocytes).


B-12. Acute Lung Injury (ALI) in the Mouse


Male mice (Balb/cAnN, about 20 g; Charles River) are anaesthetized with compressed air/oxygen/5% isoflurane. Using a pipette, 100 μl of a solution of the harmful substance to be administered (3 mg/kg of LPS or 10 ng of LPS/MCP-1; see Maus et al., Am. J. Resp. Crit. Care Med. 2001, 164 (3), 406-411) are administered deep into the mouth above the larynx. The animal inhales all of the liquid. 24 to 48 hours after the administration of the harmful substance, pulmonary lavage is carried out. To this end, the mice are anaesthetized again as described above. The thorax is opened and the trachea is exposed. An indwelling cannula (20 G) is introduced into the trachea and fixed with a thread. Via the cannula, 0.5 ml of physiological saline is administered to the lung. This is used to rinse the lung three times. The lavage obtained in this manner is transferred into a vessel. In this manner, the lung is rinsed with a total of 1.5 ml of saline. The lavage is stored on ice, and the inflammatory cells (leukocytes, neutrophiles and monocytes) are quantified on a CellDyn 3700.


B-13. Analysis of db/db Mice


Leptin receptor-deficient db/db mice (Jackson Laboratory) serve as murine model of type 2 diabetes. These animals have, firstly, contractile defects of the heart and, secondly, also renal dysfunction [Belke et al., in: Animal Models in Diabetes Research, Methods in Molecular Biology, Vol. 933 (2012); Sayyed et al., Kidney Int. 2011, 80, 68-78; Li et al., Acta Pharmacol. Sin. 2010, 31, 560-569]. Male db/db mice with or without unilateral nephrectomy are treated with test substances, and the effect on heart and kidney function is examined.


B-14. Analysis in the Renal Ischaemia Reperfusion Model (Mouse and Rat)


Experimental data confirm a reduction of the reperfusion damage after renal ischaemia/reperfusion in CCR2-knock out animals [Furuichi et al., J. Am. Soc. Nephrol. 2003, 14, 2503-2515]. In this model, mice or rats are treated with test substances and the effect on kidney function is examined.


B-15. Analysis in the UUO Model (Mouse and Rat)


Experimental data confirm reduced fibrosis in the unilateral ureteral obstruction (UUO) model in CCR2-knock out animals [Kitagawa et al., Am. J. Pathol. 2004, 165 (1), 237-246]. In this model, mice or rats are treated with test substances and the effect on kidney function is examined.


B-16. Streptozotocin-Induced Diabetes (Mouse and Rat)


Experimental data confirm reduced kidney damage in the streptozotocin-(STZ)-induced type 1 diabetes model in CCR2-knock out animals or animals that were treated with a CCR2 antagonist [Awad et al., Am. J. Physiol. Renal Physiol. 2011, 301 (6), F1358-F1366; Novikova et al., J. Diabetes Res. 2013, online, Article-ID 965832; WO 2012/041817-A1, pages 87-88]. In this model, mice or rats are treated with test substances and the effect on kidney function is examined.


B-17. Alport Mouse Model


The effect of test substances can also be demonstrated in the Alport mouse model of kidney damage [Clauss et al., J. Pathol. 2009, 218 (1), 40-47].


B-18. MCP-1-Induced Monocyte Recruitment in the Rat


Male Sprague Dawley rats (200-250 g; Charles River) are anaesthetized with 5% isoflurane in an anaesthesia cage. In the tolerance stage, MCP-1 (10 μg in 200 μl of NaCl solution) is administered via the tail vein, thus inducing the recruitment of monocytes from bone marrow. 60 minutes after the administration of MCP-1, the rats are re-anaesthetized and sacrificed painlessly, and the blood count (neutrophiles, monocytes) is determined (Advia 2120i, Siemens). The effect of test substances on the MCP-1-induced increase of monocytes measured in the blood is examined.


C. WORKING EXAMPLES FOR PHARMACEUTICAL COMPOSITIONS

The compounds according to the invention can be converted to pharmaceutical formulations as follows:


Tablet:


Composition:


100 mg of the compound according to the invention, 50 mg of lactose (monohydrate), 50 mg of corn starch (native), 10 mg of polyvinylpyrrolidone (PVP 25) (BASF, Ludwigshafen, Germany) and 2 mg of magnesium stearate.


Tablet weight 212 mg, diameter 8 mm, radius of curvature 12 mm.


Production:


The mixture of compound according to the invention, lactose and starch is granulated with a 5% solution (w/w) of the PVP in water. The granules are dried and mixed with the magnesium stearate for 5 minutes. This mixture is pressed with a conventional tableting press (for tablet dimensions see above). The guide value used for the pressing is a pressing force of 15 kN.


Suspension which can be Administered Orally:


Composition:


1000 mg of the compound according to the invention, 1000 mg of ethanol (96%), 400 mg of Rhodigel® (xanthan gum from FMC, Pennsylvania, USA) and 99 g of water.


A single dose of 100 mg of the compound according to the invention corresponds to 10 ml of oral suspension.


Production:


The Rhodigel is suspended in ethanol and the compound according to the invention is added to the suspension. The water is added while stirring. The mixture is stirred for approx. 6 h until swelling of the Rhodigel has ended.


Solution for Oral Administration:


Composition:


500 mg of the compound according to the invention, 2.5 g of polysorbate and 97 g of polyethylene glycol 400. A single dose of 100 mg of the compound according to the invention corresponds to 20 g of oral solution.


Production:


The compound according to the invention is suspended in the mixture of polyethylene glycol and polysorbate while stirring. The stirring operation is continued until dissolution of the compound according to the invention is complete.


i.v. Solution:


The compound according to the invention is dissolved in a concentration below the saturation solubility in a physiologically acceptable solvent (e.g. isotonic saline, glucose solution 5% and/or PEG 400 solution 30%). The solution is subjected to sterile filtration and dispensed into sterile and pyrogen-free injection vessels.

Claims
  • 1. A compound of the formula (I)
  • 2. The compound of the formula (I) according to claim 1 in which A represents C—H,E represents CH2 or O,R1 represents fluorine, chlorine, methyl or trifluoromethyl,R2 represents hydrogen, fluorine, chlorine, methyl or trifluoromethyl,L represents a bond, CH2 or NH, andHet represents pyridyl, pyrimidinyl or pyrazinyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, bromine, trifluoromethyl, methyl, phenyl, hydroxy, trifluoromethoxy, methoxy, methylsulphanyl, methylsulphinyl, methylsulphonyl, amino, methylamino, dimethylamino and acetylamino, represents 5-membered heteroaryl which contains one ring nitrogen atom and may additionally contain one or two further ring heteroatoms from the group consisting of N, O and S and which (i) may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, (C1-C3)-alkyl, cyclopropyl, phenyl, hydroxy, methoxy, methylsulphanyl, methylsulphinyl, methylsulphonyl, amino, methylamino, dimethylamino and acetylamino and which (ii) may be fused with a phenyl or pyridyl ring which for its part may be substituted by fluorine, chlorine, methyl, trifluoromethyl, methoxy, trifluoromethoxy, amino or acetylamino orrepresents 5-membered saturated or partially unsaturated heterocyclyl which contains one ring nitrogen atom and may additionally contain one or two further ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of methyl, hydroxy, oxo and amino, and their salts, solvates and solvates of the salts.
  • 3. The compound of the formula (I) according to claim 1 in which A represents C—H,E represents CH2 or O,R1 represents fluorine, chlorine, methyl or trifluoromethyl,R2 represents fluorine or chlorine,L represents a bond andHet represents pyridyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, bromine, trifluoromethyl, methyl, hydroxy, methoxy, methylsulphanyl, methylsulphinyl, methylsulphonyl and amino, represents pyrazolyl, imidazolyl, 1,2-oxazolyl, 1,2-thiazolyl, 1,2,4-triazolyl or 1,2,4-oxadiazolyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, methoxy, methylsulphanyl, methylsulphinyl, methylsulphonyl and amino, orrepresents 2-oxoimidazolidin-1-yl or 2-oxo-1,3-oxazolidin-3-yl, and their salts, solvates and solvates of the salts.
  • 4. The compound of the formula (I) according to claim 1 in which A represents C—H, C—F or N,E represents CH2, O or S,R1 and R2 independently of one another represent hydrogen, fluorine, chlorine, methyl or trifluoromethyl, where at least one of the two radicals R1 and R2 represents fluorine, chlorine, methyl or trifluoromethyl,L represents a bond, CH2 or NH, andHet represents pyridyl or pyrimidinyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, (C1-C4)-alkyl, hydroxy, trifluoromethoxy, (C1-C4)-alkoxy and amino, represents 5-membered heteroaryl which contains one, two or three identical or different ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, (C1-C4)-alkyl, cyclopropyl, hydroxy, (C1-C4)-alkoxy, (C1-C4)-alkoxymethyl, amino, hydroxycarbonyl and (C1-C4)-alkoxycarbonyl orrepresents 5- or 6-membered saturated or partially unsaturated heterocyclyl which contains one, two or three identical or different ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of (C1-C4)-alkyl, hydroxy, oxo, amino and imino, and their salts, solvates and solvates of the salts.
  • 5. The compound of the formula (I) according to claim 1 in which A represents C—H,E represents CH2 or O,R1 represents fluorine, chlorine, methyl or trifluoromethyl,R2 represents hydrogen, fluorine, chlorine, methyl or trifluoromethyl,L represents a bond, CH2 or NH, andHet represents pyridyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, trifluoromethoxy, methoxy and amino, represents 5-membered heteroaryl which contains one ring nitrogen atom and may additionally contain one or two further ring heteroatoms selected from the group consisting of N and O and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, cyclopropyl, hydroxy, methoxy and amino orrepresents 5-membered saturated or partially unsaturated heterocyclyl which contains one ring nitrogen atom and may additionally contain one or two further ring heteroatoms selected from the group consisting of N, O and S and which may be mono- or disubstituted by identical or different radicals selected from the group consisting of methyl, hydroxy, oxo and amino, and their salts, solvates and solvates of the salts.
  • 6. The compound of the formula (I) according to claim 1 in which A represents C—H,E represents CH2 or O,R1 represents fluorine, chlorine or trifluoromethyl,R2 represents fluorine or chlorine,L represents a bond andHet represents pyridyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, methoxy and amino, represents pyrazolyl, imidazolyl, 1,2,4-triazolyl or 1,2,4-oxadiazolyl which may be mono- or disubstituted by identical or different radicals selected from the group consisting of fluorine, chlorine, trifluoromethyl, methyl, hydroxy, methoxy and amino, orrepresents 2-oxoimidazolidin-1-yl or 2-oxo-1,3-oxazolidin-3-yl, and their salts, solvates and solvates of the salts.
  • 7. A process for preparing a compound of the formula (I) as defined in claim 1, characterized in that [A] a compound of the formula (II)
  • 8. The compound as defined in claim 1 for treatment and/or prevention of diseases.
  • 9. A method for the treatment and/or prevention of acute coronary syndrome, myocardial infarction, acute and chronic heart failure, acute and chronic kidney failure and acute lung damage using the compound as defined in claim 1.
  • 10. A method for the treatment and/or prevention of acute coronary syndrome, myocardial infarction, acute and chronic heart failure, acute and chronic kidney failure and acute lung damage using the compound as defined in claim 1.
  • 11. A medicament comprising the compound as defined in claim 1 in combination with one or more inert, non-toxic, pharmaceutically suitable excipients.
  • 12. A medicament comprising the compound as defined in claim 1 in combination with one or more further active compounds selected from the group of the antihyperglycaemic agents (antidiabetics), the hypotensive agents, the platelet aggregation inhibitors, the anticoagulants and the HMG-CoA reductase inhibitors (statins).
  • 13. A method for the treatment and/or prevention of acute coronary syndrome, myocardial infarction, acute and chronic heart failure, acute and chronic kidney failure and acute lung damage using the medicament of claim 11.
  • 14. A method for the treatment and/or prevention of acute coronary syndrome, myocardial infarction, acute and chronic heart failure, acute and chronic kidney failure and acute lung damage in humans and animals by administration of an effective amount of the compound as defined in claim 1.
  • 15. A method for the treatment and/or prevention of acute coronary syndrome, myocardial infarction, acute and chronic heart failure, acute and chronic kidney failure and acute lung damage in humans and animals by administration of an effective amount of the medicament as defined in claim 11.
Priority Claims (1)
Number Date Country Kind
13184483.9 Sep 2013 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2014/069537 9/12/2014 WO 00