WO2021094210A1 - Substituted pyrazine carboxamide derivatives as prostaglandin ep3 receptor antagonists - Google Patents

Substituted pyrazine carboxamide derivatives as prostaglandin ep3 receptor antagonists Download PDF

Info

Publication number
WO2021094210A1
WO2021094210A1 PCT/EP2020/081250 EP2020081250W WO2021094210A1 WO 2021094210 A1 WO2021094210 A1 WO 2021094210A1 EP 2020081250 W EP2020081250 W EP 2020081250W WO 2021094210 A1 WO2021094210 A1 WO 2021094210A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
group
fluoro
alkyl
mmol
Prior art date
Application number
PCT/EP2020/081250
Other languages
French (fr)
Inventor
Lisa CANDISH
Steffen Müller
Niels Lindner
Christoph Gerdes
Elisabeth Pook
Anja BUCHMÜLLER
Fabienne Zdenka GAUGAZ
Stefanie Zimmermann
Dieter Lang
Markus Follmann
Nuria Ortega Hernandez
Frank SÜSSMEIER
Andreas Timmermann
Rudolf Schohe-Loop
Eloisa JIMENEZ NUNEZ
Xiang Gao
Hongping WANG
Xin Guo
Original Assignee
Bayer Aktiengesellschaft
Bayer Pharma Aktiengesellschaft
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bayer Aktiengesellschaft, Bayer Pharma Aktiengesellschaft filed Critical Bayer Aktiengesellschaft
Publication of WO2021094210A1 publication Critical patent/WO2021094210A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid

Definitions

  • the invention relates to substituted pyrazine carboxamide derivatives and to processes for their preparation, and also to their use for preparing medicaments for the treatment and/or prophylaxis of diseases, in particular cardiovascular disorders, preferably thrombotic or thromboembolic disorders, and diabetes, and also urogenital and ophthalmic disorders.
  • Atherothrombosis is the main complication of atherosclerosis and underlies several of the most lethal human diseases, such as myocardial infarction (Ml), ischemic stroke (IS) and peripheral arterial occlusive disease (PAOD).
  • Ml myocardial infarction
  • IS ischemic stroke
  • PAOD peripheral arterial occlusive disease
  • the process is initiated by the deposition of lipids and their subsequent oxidation in the arterial wall which induces the recruitment of inflammatory cells and the formation of atherosclerotic plaques.
  • These plaques are covered by a fibrous cap which maintains the plaque content separated from the blood flow.
  • pro-inflammatory mechanisms drive inflammatory cells to produce matrix metalloproteases which digest the proteins of the fibrous cap.
  • the thinned cap is called “vulnerable”, meaning that the cap may rupture relatively easily in response to stresses.
  • the cap ruptures or its endothelial cover erodes, the plaque content comes into contact with the blood and provokes the formation of an intravascular thrombus by activating platelets and blood coagulation.
  • This process, forming a thrombus on plaques, is called atherothrombosis. If obstructive, the resulting intravascular thrombus interrupts blood flow and causes ischemia of downstream tissues with dramatic clinical consequences representing the leading cause of death and morbidity worldwide.
  • thrombosis which is the pathological formation of intra-vascular plugs.
  • the mechanisms of thrombosis encompass two intertwined pathways, the coagulation cascade and the aggregation of platelets, which once activated by a vessel injury act synergistically to build the intravascular clot obstructing the vessel lumen.
  • Anticoagulant and anti-aggregant drugs, used to prevent atherothrombosis have elicited a considerable reduction of the rate of second myocardial infarctions and a decrease of long term mortality from about 30% prior to the 1980s to less than 10% after the 2000s (Arch. Intern. Med.
  • the COX inhibitor Aspirin and the ADP receptor P2Y12 antagonist Clopidogrel are components of dual antiplatelet therapy.
  • Prasugrel and Ticagrelor are alternative P2Y12 blockers that have demonstrated reductions of cardiovascular ischemic events ( N . Engl. J. Med. 2007, 357, 2001-2015; ibid. 2009, 361, 1045-1057).
  • the coagulation factor Xa inhibitor Rivaroxaban has also shown benefits to patients with stable atherosclerotic vascular disease (N. Engl. J. Med. 2017, 377, 1319-1330).
  • PGE2 prostaglandin E2
  • Increased PGE2 concentrations have been measured in atherosclerotic vascular walls of mice and humans [Circulation 2001 , 104, 921-927; J. Exp. Med. 2007, 204, 311-320; Cardiovasc. Res. 2014, 101, 482- 491]
  • PGE2 binds on four specific receptors EP1, EP2, EP3 and EP4 on cell membranes.
  • PGE2 has been shown to interfere with human and murine platelet function via EP3 and EP4 receptors ( Eur : J. Pharmacol. 1991, 194, 63-70). Stimulation of EP3 potentiates platelet activation and aggregation induced by primary agonists like collagen or ADP, whereas stimulation of EP4 inhibits platelet activation. This PGE2-dependent balance of platelet activation and inhibition can be tipped by modulation of EP3 or EP4 receptors ( Platelets 2010, 21, 329-342; Prostaglandins & Other Upid Mediators 2015, 121, 4-16).
  • blocking the EP3 receptor by specific antagonists should be a beneficial strategy for prevention and treatment of atherothrombosis by local abrogation of platelet activation without altering hemostasis.
  • EP3 antagonists might represent a beneficial strategy in the treatment of patients with type II diabetes.
  • EP3 receptor antagonists might help to improve renal disorders and in particular to resolve bladder hyperactivity, interstitial cystitis or bladder pain syndrome.
  • antithrombotic and anti-inflammatory principles may also be particularly attractive to prevent the mutual enhancement of coagulation and platelet activation.
  • prostaglandin derivatives participate in the regulation of intraocular pressure and inflammation. Therefore, compounds modulating the respective receptors may have a benefit in the prevention and treatment of ocular diseases.
  • prostaglandin derivates play an essential role via their platelet-activating and pro- inflammatory mechanisms in inflammatory disorders like vasculitides, for example Kawasaki disease, Takayasu arteritis and Thrombangiitis obliterans (Buerger’s disease) as well as myocarditis (EMBO Mol Med 2018, e8536).
  • EP3 antagonists might contribute to the treatment and/or prophylaxis of diabetes- related end-organ manifestations like diabetic retinopathy and diabetic nephropathy.
  • prostaglandins are involved in the pathogenesis of neurological disorders like neuropathic pain, Alzheimer’s disease and Parkinson’s disease.
  • PGE2 and EP3 have been reported to play a role in the pathogenesis of respiratory disorders like chronic cough, asthma and COPD (Am J Respir Crit Care 2009, 923-928).
  • W02009/082687 provides azaindolizines with anti-cancer activity.
  • WO 2012/054233 provides imidazo pyrazine compounds for agronomic and nonagronomic uses.
  • WO 2016/057522 provides triazolo pyridine compounds for the treatment of cystic fibrosis.
  • WO 2015/052610 and WO 2016/103097 provide antagonists of prostaglandin EP3 receptor having a pyridinone substituted indazole, indole or quinoline derivative.
  • Amide or pyridinone based EP3 receptor antagonists are also described in ACS Med. Chem. Lett. 2010, 7, 316-320 and in Bioorg. Med. Chem. Lett. 2009, 19, 4292-4295, ibid. 2011, 27, 2806-2811, ibid. 2016, 26, 2670- 2675.
  • certain substituted pyrazine carboxamide derivatives represent highly potent antagonists of prostaglandin EP3 receptor.
  • the invention provides compounds of the formula (I) in which G 1 represents CR 4 or N, G 2 represents CH or N, with the proviso that G 1 is N, if G 2 is N,
  • R 4 represents hydrogen, CrC4-alkyl, CrC4-halogenoalkyl, C3-C6-cycloalkyl or C1-C3- alkoxymethyl
  • R 1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, phenylsulfonyl, C3-C6- cycloalkylsulfonyl, 2,3-dihydro-1H-inden-1-yl or the group -L-R E , where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano, methoxy, methylsulfonyl, carbamoyl, NR a R b (where R a and R b are independently selected from the group consisting of hydrogen, CrC4-alkyl, C2-C6- halogenoalkyl and cyclopropyl) and CrC3-halogenoalkoxy
  • R 5 represents hydrogen, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl or methoxycarbonyl, and where phenylsulfonyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro, chloro and cyano, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and L represents CrC 6 -alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, methoxycarbonyl, carboxyl, carbamoyl, amino, dimethylamino, te/f-butoxycarbonylamino, C3-C6-cycloalkyl (which may be substituted by 1 hydroxy), azetidin-1-yl (which may be substituted by 1
  • R E represents phenyl, pyridinyl, pyrimidinyl, pyrazinyl, thienyl, pyrazolyl, oxazolyl, C3-C6- cycloalkyl, azetidinyl, pyrrolidinyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen, CrC4-alkyl, hydroxy, methoxy, trifluoromethyl, trifluoromethoxy, difluoromethoxy, dimethylaminomethyl, aminosulfonyl and pyrrolidin- 1-y I methyl, where pyridinyl may be substituted by 1 or 2 substituents independently selected from the group consisting of chloro, methyl, trifluoromethyl and methoxy, where pyrimidinyl may be substituted by 1 or 2 methyl substituents, where pyrazolyl may be substituted by 1 to 3 substituents
  • R 2 represents a group of the formula where # is the point of attachment to the pyrazine ring, Q 1 represents CR 8A or N, Q 2 represents CR 8 or N,
  • R 6 represents hydrogen, halogen, CrC4-alkyl, CrC4-halogenoalkyl, CrC4-alkoxy, C1-C4- halogenoalkoxy or C3-C6-cycloalkyl,
  • R 7 represents hydrogen, halogen, CrC4-alkyl, CrC4-halogenoalkyl, CrC4-alkoxy, C1-C4- halogenoalkoxy or C3-C6-cycloalkyl, with the proviso, that at least one of R 6 and R 7 is not hydrogen,
  • R 7A represents hydrogen or halogen
  • R 8 represents hydrogen or halogen
  • R 8A represents hydrogen or halogen
  • R 9 represents hydrogen, halogen or CrC4-alkyl
  • R 3 represents hydrogen, halogen, cyano, CrC4-alkyl or CrC2-halogenoalkyl, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
  • the compounds according to the invention may be divided into three subgroups:
  • substituted means that one or more hydrogen atoms on the designated atom or group are replaced with a selection from the indicated group, provided that the designated atom’s normal valence under the existing circumstances is not exceeded. Combinations of substituents and/or variables are permissible.
  • the term “one or more”, e.g. in the definition of the substituents of the compounds of general formula (I) of the present invention, means “1 , 2, 3, 4 or 5, particularly 1 , 2, 3 or 4, more particularly 1, 2 or 3, even more particularly 1 or 2”.
  • halogen or “halogeno” like in combinations e.g. in halogenoalkyl means a fluorine, chlorine, bromine or iodine atom, particularly a fluorine, chlorine or bromine atom, even more particularly fluorine or chlorine.
  • Ci-C4-alkyl and “Ci-Cyalkyl” means a linear or branched, saturated, monovalent hydrocarbon group having 1, 2, 3, or 4 carbon atoms, and 1, 2, 3, 4, 5, 6 or 7 carbon atoms, e.g.
  • said group has 1, 2, 3 or 4 carbon atoms (“Ci-C4-alkyl”), e.g. a methyl, ethyl, propyl, isopropyl, butyl, sec-butyl isobutyl, or tert- butyl group, more particularly 1, 2 or 3 carbon atoms (“Ci-C3-alkyl”), e.g. a methyl, ethyl, n-propyl or isopropyl group.
  • Ci-C 6 -halogenoalkyl represents a linear or branched, saturated, monovalent hydrocarbon group in which the term “alkyl” is as defined supra, and in which one or more of the hydrogen atoms are replaced, identically or differently, with a halogen atom.
  • said halogen atom is a fluorine atom.
  • Said CrC 6 -halogenoalkyl group is, for example fluoromethyl, difluoromethyl, trifluoromethyl, 2-fluoroethyl, 2,2-difluoroethyl, 2,2, 2-trif luoroethy I , pentafluoroethyl, 3,3,3-trifluoropropan-1-yl, 1,1,1 -trif luoropropan-2-yl , 1,3-diflu- oropropan-2-yl, 3-fluoropropan-1-yl, 1,1,1 -trifluorobutan-2-yl, and 3,3,3-trifluoro-1-methyl-propan- 1-yl.
  • Ci-C4-halogenoalkoxy and “Ci-C3-halogenoalkoxy” represents a linear or branched, saturated, monovalent CrC4-alkoxy or CrC3-alkoxy group (where alkoxy represents a straight- chain or branched, saturated, monovalent alkoxy radical having 1 to 4 or 1 to 3 carbon atoms, by way of example and with preference methoxy, ethoxy, n-propoxy, isopropoxy), in which one or more of the hydrogen atoms is replaced, identically or differently, with a halogen atom. Particularly, said halogen atom is a fluorine atom.
  • Said CrC3-halogenoalkoxy group is, for example, fluoromethoxy, difluoromethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy or pentafluoroethoxy.
  • C3-C6-cycloalkyl means a saturated, monovalent, monocyclic hydrocarbon ring which contains 3, 4, 5 or 6 carbon atoms.
  • Said C3-C6-cycloalkyl group is for example a cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl group.
  • Ci-C 6 -alkanediyl represents a linear or branched, divalent alkyl radical having 1 to 6, 1 to 4, 2 to 5 or 2 to 4 carbon atoms, by way of example and with preference methylene (-CH2-), ethan-1 ,1-diyl [-CH(CH3)-], ethan-1,2-diyl [-(CH 2 )H, propan- 1,1-diyl [-CH(CH 2 CH 3 )-], propan-1, 2-diyl [-CH 2 CH(CH 3 )-], 2- methylpropan-1,1-diyl ⁇ -CH[CH(CH3)2]- ⁇ , 2-methylpropan-1,3-diyl ⁇ -CH 2 [CH(CH 3 )]CH 2 - ⁇ , butan-CH2-), ethan-1 ,1-diyl [-CH(CH3)-], ethan-1,2-diyl [-(CH 2 )H, propan- 1,1-
  • Compounds according to the invention are the compounds of the formula (I) and the salts, solvates and solvates of the salts thereof, and also the compounds encompassed by formula (I) and specified hereinafter as working example(s), and the salts, solvates and solvates of the salts thereof, to the extent that the compounds encompassed by formula (I) and specified hereinafter are not already salts, solvates and solvates of the salts.
  • inventive compounds may, depending on their structure, exist in different stereoisomeric forms, i.e. in the form of configurational isomers or else, if appropriate, of conformational isomers (enantiomers and/or diastereomers, including those in the case of rotamers and atropisomers).
  • the present invention therefore encompasses the enantiomers and diastereomers, and the respective mixtures thereof.
  • the stereoisomerically uniform constituents can be isolated from such mixtures of enantiomers and/or diastereomers in a known manner; chromatography processes are preferably used for this, especially HPLC chromatography on an achiral or chiral phase.
  • the compounds of the present invention may exist as tautomers.
  • the compounds of formula (I) encompass the tautomer of formula (la)
  • the tautomers (l-Aa), (l-Ba) and (l-Ca) of the subgroups of formulas (l-A), (l-B) and (l-C) are shown hereafter:
  • the present invention includes all possible tautomers of the compounds of the present invention as single tautomers, or as any mixture of said tautomers, in any ratio.
  • the term “enantiomerically pure” is understood to mean that the compound in question with respect to the absolute configuration of the chiral centre is present in an enantiomeric excess of more than 95%, preferably more than 97%.
  • the present invention also encompasses all suitable isotopic variants of the compounds according to the invention.
  • An isotopic variant of an inventive compound is understood here as meaning a compound in which at least one atom within the inventive compound 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.
  • isotopes which can be incorporated into a compound according to the invention are those of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as 2 H (deuterium), 3 H (tritium), 13 C, 14 C, 15 N, 17 0, 18 0, 32 P, 33 P, 33 S, 34 S, 35 S, 36 S, 18 F, 36 CI, 82 Br, 123 l, 124 l, 129 l and 131 1.
  • Particular isotopic variants of a compound according to the invention 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 3 H or 14 C isotopes are suitable for this purpose.
  • the incorporation of isotopes, for example of deuterium may lead to particular therapeutic benefits as a consequence of greater metabolic stability of the compound, for example an extension of the half-life in the body or a reduction in the active dose required; such modifications of the inventive compounds 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 the processes known to those skilled in the art, for example by the methods described further below and the procedures described in the working examples, by using corresponding isotopic modifications of the respective reagents and/or starting compounds.
  • Preferred salts in the context of the present invention are physiologically acceptable salts of the compounds according to the invention.
  • the invention also encompasses salts which themselves are unsuitable for pharmaceutical applications but which can be used, for example, for the isolation or purification 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 sulfonic acids, for example salts of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, benzenesulfonic acid, naphthalenedisulfonic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
  • Physiologically acceptable salts of the compounds according to the invention also include salts of 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) and ammonium salts derived from ammonia or organic amines having 1 to 16 carbon atoms, by way of example and with preference ethylamine, diethylamine, tri ethyl amine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, /V-methylmorpholine, arginine, lysine, ethylenediamine, N- methylpiperidine and choline.
  • alkali metal salts e.g. sodium and potassium salts
  • alkaline earth metal salts e.g. calcium and magnesium salts
  • ammonium salts derived
  • the present invention includes all possible salts of the compounds according to the invention as single salts, or as any mixture of said salts, in any ratio.
  • Solvates in the context of the invention are described as those forms of the inventive compounds which form a complex in the solid or liquid state by coordination with solvent molecules.
  • the compounds according to the invention may contain polar solvents, in particular water, methanol or ethanol for example, as structural element of the crystal lattice of the compounds. Hydrates are a specific form of the solvates in which the coordination is with water. It is possible for the amount of polar solvents, in particular water, to exist in a stoichiometric or non-stoichiometric ratio. In the case of stoichiometric solvates, e.g.
  • a hydrate, hemi-, (semi-), mono-, sesqui-, di-, tri-, tetra-, penta- etc. solvates or hydrates, respectively, are possible.
  • the present invention includes all such hydrates or solvates.
  • the compounds according to the invention can exist as N-oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised in a known manner.
  • the present invention includes all such possible N-oxides.
  • the present invention additionally also encompasses prodrugs of the inventive compounds.
  • prodrugs encompasses compounds which for their part may be biologically active or inactive but are converted during their residence time in the body into compounds according to the invention (for example by metabolism or hydrolysis).
  • treatment 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.
  • therapy is understood here to be synonymous with the term “treatment”.
  • prevention means prevention, prophylaxis and “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 advancement 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 end point of the line marked by # in each case does not represent a carbon atom or a CH2 group, but is part of the bond to the atom to which R 2 is attached.
  • G 1 represents CR 4 or N
  • G 2 represents CH or N, with the proviso that G 1 is N, if G 2 is N,
  • R 4 represents hydrogen, CrC 4 -halogenoalkyl or C 3 -C 6 -cycloalkyl
  • R 1 represents CrCyalkyl, C 2 -C 6 -halogenoalkyl, C 3 -C 6 -cycloalkyl, phenylsulfonyl, C 3 -C 6 - cycloalkylsulfonyl, 2,3-dihydro-1H-inden-1-yl or the group -L-R E , where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR 5 , and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, ethyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 or 2 substitu
  • R 5 represents CrC4-alkyl or C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, and where phenylsulfonyl may be substituted by 1 substituent fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
  • L represents Ci-C 6 -alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, carboxyl, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro and CrC4-alkyl,
  • R 2 represents a group of the formula where # is the point of attachment to the pyrazine ring
  • Q 1 represents CR 8A .
  • Q 2 represents CR 8 .
  • R 6 represents halogen or CrC4-alkyl
  • R 7 represents halogen, CrC4-alkyl or CrC4-alkoxy
  • R 7A represents hydrogen
  • R 8 represents hydrogen or halogen
  • R 8A represents hydrogen
  • R 9 represents hydrogen or halogen
  • R 3 represents hydrogen or halogen, and the salts thereof, the solvates thereof and the solvates of the salts thereof. Preference is also given to compounds of the formula (I) in which G 1 represents CR 4 , G 2 represents CH,
  • R 4 represents hydrogen, CrC 4 -halogenoalkyl or C 3 -C 6 -cycloalkyl
  • R 1 represents CrCyalkyl, C 2 -C 6 -halogenoalkyl, C 3 -C 6 -cycloalkyl, phenylsulfonyl, C 3 -C 6 - cycloalkylsulfonyl or the group -L-R E , where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by NR 5 , and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
  • R 5 represents CrC 4 -alkyl, and where phenylsulfonyl may be substituted by 1 substituent fluoro, and
  • L represents Ci-C 6 -alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl or C3-C6-cycloalkyl, where phenyl may be substituted by 1 substituent halogen, and where pyridinyl may be substituted by 1 substituent methoxy,
  • R 2 represents a group of the formula where # is the point of attachment to the pyrazine ring
  • Q 1 represents CR 8A .
  • Q 2 represents CR 8 .
  • R 6 represents halogen or CrC4-alkyl
  • R 7 represents halogen, CrC4-alkyl or CrC4-alkoxy
  • R 7A represents hydrogen
  • R 8 represents hydrogen or fluoro
  • R 8A represents hydrogen
  • R 9 represents hydrogen or fluoro
  • R 3 represents hydrogen or fluoro, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
  • G 2 represents CH
  • R 1 represents CrCyalkyl, C3-C6-cycloalkyl, 2,3-dihydro-1 H-inden-1-yl or the group -L-R E , where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR 5 , and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 substituent fluoro, and
  • R 5 represents CrC4-alkyl or C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
  • L represents Ci-C4-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy and carboxyl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro and CrC4-alkyl,
  • R 2 represents a group of the formula where # is the point of attachment to the pyrazine ring
  • Q 1 represents CR 8A .
  • Q 2 represents CR 8 .
  • R 6 represents halogen or CrC4-alkyl
  • R 7 represents halogen or CrC4-alkyl
  • R 7A represents hydrogen
  • R 8 represents hydrogen
  • R 8A represents hydrogen
  • R 9 represents hydrogen
  • R 3 represents hydrogen, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
  • G 2 represents N
  • R 1 represents the group -L-R E .
  • L represents C 2 -C 4 -alkanediyl, where alkanediyl may be substituted by up to 3 fluoro,
  • R E represents phenyl or pyridinyl, where phenyl may be substituted by 1 substituent of halogen, and where pyridinyl may be substituted by 1 substituent methoxy
  • R 2 represents a group of the formula where # is the point of attachment to the pyrazine ring
  • R 3 represents hydrogen, and the salts thereof, the solvates thereof and the solvates of the salts thereof Preference is also given to compounds of the formula (I) in which G 1 represents CR 4 ,
  • G 2 represents CH
  • R 4 represents hydrogen, trifluoromethyl or cyclopropyl
  • R 1 represents CrCyalkyl, C 2 -C 6 -halogenoalkyl, C 3 -C 6 -cycloalkyl, phenylsulfonyl, cyclopropylsulfonyl or the group -L-R E , where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH 2 group may be replaced by NR 5 , and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 substituent selected from the group consisting of fluoro and methoxy, and
  • R 5 represents methyl, and where phenylsulfonyl may be substituted by 1 substituent fluoro, and
  • L represents Ci-C 6 -alkanediyl, where alkanediyl may be substituted by 1 substituent selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl or cyclopropyl, where phenyl may be substituted by 1 substituent fluoro, and where pyridinyl may be substituted by 1 substituent methoxy
  • R 2 represents a group of the formula where # is the point of attachment to the pyrazine ring
  • Q 1 represents CR 8A ,
  • Q 2 represents CR 8 .
  • R 6 represents fluoro, chloro or methyl
  • R 7 represents chloro, methyl or methoxy
  • R 7A represents hydrogen
  • R 8 represents hydrogen or fluoro
  • R 8A represents hydrogen
  • R 9 represents hydrogen or fluoro
  • R 3 represents hydrogen or fluoro, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
  • G 2 represents CH
  • R 1 represents Ci-Cralkyl, C3-C6-cycloalkyl, 2,3-dihydro-1H-inden-1-yl or the group -L-R E , where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR 5 , and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 substituent fluoro, and
  • R 5 represents methyl or C2-C3-halogenoalkyl substituted by 1 to 3 fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
  • L represents Ci-C4-alkanediyl, where alkanediyl may be substituted by 1 substituent selected from the group consisting of hydroxy, methoxy and carboxyl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro, bromo and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro, methyl and ethyl,
  • R 2 represents a group of the formula where # is the point of attachment to the pyrazine ring
  • Q 1 represents CR 8A .
  • Q 2 represents CR 8 .
  • R 6 represents chloro or methyl
  • R 7 represents fluoro or methyl
  • R 7A represents hydrogen
  • R 8 represents hydrogen
  • R 8A represents hydrogen
  • R 9 represents hydrogen
  • R 3 represents hydrogen, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
  • R 1 represents Ci-Cralkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, 2,3-dihydro-1H-inden-1-yl or the group -L-R E , where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano, methoxy, methylsulfonyl, carbamoyl, NR a R b (where R a and R b are independently selected from the group consisting of hydrogen, CrC4-alkyl, C2-C6- halogenoalkyl and cyclopropyl) and CrC3-halogenoalkoxy, where halogenoalkoxy is substituted by 1 to 3 fluoro substituents, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxycarbonyl and NR
  • R 5 represents hydrogen, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl or methoxycarbonyl, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
  • L represents Ci-C 6 -alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, methoxycarbonyl, carboxyl, carbamoyl, amino, dimethylamino, te/f-butoxycarbonylamino, C3-C6-cycloalkyl (which may be substituted by 1 hydroxy), azetidin-1-yl (which may be substituted by 1 or 2 fluoro), pyrrolidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl, pyrimidinyl, pyrazinyl, thienyl, pyrazolyl, oxazolyl, C3-C6- cycloalkyl, azetidinyl, pyrrolidinyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen, CrC4-alkyl, hydroxy, methoxy, trifluoromethyl, trifluoromethoxy, difluoromethoxy, dimethylaminomethyl, aminosulfonyl and pyrrolidin- 1-y I methyl, where pyridinyl may be substituted by 1 or 2 substituents independently selected from the group consisting of chloro, methyl, trifluoromethyl and methoxy, where pyrimidinyl may be substituted by 1 or 2 methyl substituents, where pyrazolyl may be substituted by 1 to 3 substituents
  • R 1 represents Ci-Cralkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, 2,3-dihydro-1H-inden-1-yl or the group -L-R E , where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR 5 , and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, ethyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
  • R 5 represents CrC4-alkyl or C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
  • L represents Ci-C 6 -alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, carboxyl, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro and CrC4-alkyl.
  • R 1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl or the group -L-R E , where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by NR 5 , and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
  • R 5 represents CrC4-alkyl
  • L represents Ci-C 6 -alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl or C3-C6-cycloalkyl, where phenyl may be substituted by 1 substituent halogen, and where pyridinyl may be substituted by 1 substituent methoxy.
  • R 1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl or the group -L-R E , where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by NR 5 , and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 substituent selected from the group consisting of fluoro and methoxy, and
  • R 5 represents methyl
  • L represents Ci-C 6 -alkanediyl, where alkanediyl may be substituted by 1 substituent selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
  • R E represents phenyl, pyridinyl or cyclopropyl, where phenyl may be substituted by 1 substituent fluoro, and where pyridinyl may be substituted by 1 substituent methoxy.
  • R 2 represents 4-chloro-3- fluorophenyl, 3-chloro-4-methylphenyl, 4-chloro-3-methylphenyl, 3,4-dimethylphenyl, 3-fluoro-4- methylphenyl, 2-napthyl, 2,3-dihydro-1,4-benzodioxine or 5-fluoro-2,3-dihydro-1,4-benzodioxine.
  • R 2 represents 3,4-dimethylphenyl, 2-napthyl or 2,3-dihydro-1 ,4-benzodioxine.
  • R x represents CrCs-alkyl, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, methoxycarbonyl, carboxyl, carbamoyl, amino, dimethylamino, fe/f-butoxycarbonylamino, C3-C6-cycloalkyl (which may be substituted by 1 hydroxy), azetidin- 1-yl (which may be substituted by 1 or 2 fluoro), pyrrolidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro, and
  • R y represents hydrogen, halogen, CrC4-alkyl, hydroxy, methoxy, trifluoromethyl, trifluoromethoxy, difluoromethoxy, dimethylaminomethyl, aminosulfonyl or pyrrolidin-1-ylmethyl.
  • the compounds of the formula (l-X) are a subgroup of the compounds of the formula (I).
  • the invention further provides a process for preparing compounds of the formula (I), or salts thereof, solvates thereof or solvates of the salts thereof, wherein
  • R 1 is as defined above, are reacted with compounds of the formula (III) in which G 1 , G 2 , R 2 and R 3 are as defined above, in the presence of a dehydrating agent to give compounds of the formula (I).
  • the reaction [A] is generally carried out in inert solvents, if appropriate in the presence of a base, preferably in a temperature range from 0°C to 50°C at atmospheric pressure.
  • reaction [A] can also be carried out without a solvent only in one base if the base is a liquid at room temperature.
  • Suitable dehydrating agents are, for example, carbodiimides such as N,N ’-diethyl-, N,N’- dipro- pyl- N,N ’-diisopropyl-, A/,/ ⁇ /-dicydohexylcarbodiimide, A/-(3-dimethylaminoisopropyl)-/ ⁇ /'-ethylcarbo- diimide hydrochloride (EDCI) (optionally in the presence of pentafluorophenol (PFP)), /V-cyclohexyl- carbodiimide-/V-propyloxymethyl-polystyrene (PS-carbodiimide) or carbonyl compounds such as carbonyldiimidazole (CDI), or 1 ,2-oxazolium compounds such as 2-ethyl-5-phenyl-1,2-oxazolium 3- sulphate or 2-terf-butyl-5-methyl-isoxa
  • Bases are, for example, alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate, or organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine; preference is given to condensation with diisopropylethylamine, 4-dimethylaminopyridine, pyridine or /V-methylmorpholine.
  • alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate
  • organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine; preference is given to condensation with diisopropylethylamine, 4-dimethylaminopyridine,
  • Inert solvents are, for example, halogenated hydrocarbons such as dichloromethane or trichloromethane, hydrocarbons such as benzene or toluene, or other solvents such as nitromethane, dioxane, diethyl ether, tetrahydrofuran, ethyl acetate, dimethylformamide, dimethylacetamide, dimethyl sulphoxide, /V-methylpyrrolidone or acetonitrile, or mixtures of the solvents; preference is given to dimethylformamide, ethyl acetate, /V-methylpyrrolidone or dichloromethane.
  • halogenated hydrocarbons such as dichloromethane or trichloromethane
  • hydrocarbons such as benzene or toluene
  • other solvents such as nitromethane, dioxane, diethyl ether, tetrahydrofuran, ethyl
  • the compounds of the formula (II) are known or can be synthesized from the corresponding starting compounds by known processes.
  • G 1 , G 2 , R 2 and R 3 are as defined above, and
  • R 13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with a base.
  • the reaction [B] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • Bases are, for example, alkali metal hydroxides such as sodium hydroxide, lithium hydroxide or potassium hydroxide, or alkali metal carbonates such as caesium carbonate, sodium carbonate or potassium carbonate, or alkoxides such as potassium tert- butoxide or sodium tert- butoxide; preference is given to sodium hydroxide or lithium hydroxide.
  • alkali metal hydroxides such as sodium hydroxide, lithium hydroxide or potassium hydroxide
  • alkali metal carbonates such as caesium carbonate, sodium carbonate or potassium carbonate
  • alkoxides such as potassium tert- butoxide or sodium tert- butoxide
  • Inert solvents are, for example, alcohols such as methanol or ethanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, dimethylacetamide, acetonitrile or pyridine, or mixtures of solvents, or mixtures of solvents with water; preference is given to a mixture of tetrahydrofuran and water or ethanol and water or methanol and water or tetrahydrofuran, methanol and water.
  • alcohols such as methanol or ethanol
  • ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran
  • other solvents such as dimethylformamide, dimethylacetamide, acetonitrile or pyridine, or mixtures of
  • R 13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
  • R 14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with an ammonia equivalent in the presence of an acid.
  • the reaction [C] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • reaction [C] can also be carried out without a solvent only in acid if the acid is a liquid at room temperature.
  • Ammonia equivalents are, for example, ammonium acetate, ammonium formate, ammonium propionate, or ammonium chloride; preference is given to ammonium acetate.
  • Acids are, for example, organic acids such was formic acid, acetic acid, propionic acid, trifluoroacetic acid, trifluoromethanesulfonic acid, or mineral acids such as, for example, hydrogen chloride, or hydrogen bromide; preference is given to acetic acid.
  • G 1 and G 2 are each as defined above,
  • R 13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
  • R 14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with compounds of the formula (VII)
  • R 2 and R 3 are as defined above, and X 1 represents chloro, bromo, iodo Oder triflate, in the presence of a base.
  • the reaction [D] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • Bases are, for example, alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate, or organic bases such as trialkylamines, for example triethylamine, N-methylmorpholine, N-methylpiperidine, 4- dimethylaminopyridine, diisopropylethylamine or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to potassium carbonate.
  • alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate
  • organic bases such as trialkylamines, for example triethylamine, N-methylmorpholine, N-methylpiperidine, 4- dimethylaminopyridine, diisopropylethylamine or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to potassium carbonate.
  • Inert solvents are, for example, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride or 1,2-dichloroethane, alcohols such as methanol or ethanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, N-methyl-pyrrolidone, dimethylacetamide, acetonitrile, acetone or pyridine, or mixtures of solvents, or mixtures of solvents with water; preference is given to acetone.
  • halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride or 1,2-dichloroethane
  • alcohols such as methanol or ethanol
  • ethers such as diethyl ether, methyl tert- but
  • the compounds of the formula (VII) are known or can be synthesized from the corresponding starting compounds by known processes.
  • the compounds of the formula (VI) may be divided into three subgroups ((Vl-A), (Vl-B) and (VI-C)):
  • R 4 is as defined above, and
  • R 13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with compounds of the formula (IX) (IX), in which
  • R 14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, in the presence of a copper(ll) sorce and a ligand.
  • the reaction [E] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • Copper(ll) sorces are, for example, copper(ll)chloride, copper(ll)bromide, copper(ll)iodide or copper(ll)oxide; preference is given to copper(ll)oxide.
  • Ligands are, for example, 1,10-phenanthroline, 3,4,7,8-tetramethyl-1,10-phenanthroline or pathophenanthroline; preference is given to 1,10-phenanthroline.
  • Inert solvents are, for example, ethers such as diethyl ether, methyl tert- butyl ether, 1,2- dimethoxyethane, 1,4-dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, /V-methyl-pyrrolidone, dimethylacetamide or acetonitrile; preference is given to 1,4-dioxane.
  • the compounds of the formula (VIII) and of the formula (IX) are known or can be synthesized from the corresponding starting compounds by known processes.
  • the compounds of the formula (Vl-A) are known or can also be prepared
  • R 4 is as defined above, R 13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
  • R 14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with a base.
  • the reaction [F] is generally carried out in inert solvents, preferably in a temperature range from 0°C up to reflux of the solvents at atmospheric pressure.
  • Bases are, for example, alkali metal te/f-butoxides, such as sodium te/f-butoxide or potassium tert- butoxide or alkali metal hydrides such as sodium hydride or potassium hydride, or alkali metal amides such as lithium diisopropylamide or potassium bis(trimethylsilyl)amide; preference is given to potassium ferf-butoxide.
  • alkali metal te/f-butoxides such as sodium te/f-butoxide or potassium tert- butoxide
  • alkali metal hydrides such as sodium hydride or potassium hydride
  • alkali metal amides such as lithium diisopropylamide or potassium bis(trimethylsilyl)amide
  • Inert solvents are, for example, alcohols such as methanol, ethanol, iso-propanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as toluene, dimethylformamide, N-methyl-pyrrolidone or dimethylacetamide, or mixtures of solvents; preference is given to a mixture of ethanol, toluene and tetrahydrofuran.
  • alcohols such as methanol, ethanol, iso-propanol
  • ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran
  • other solvents such as toluene, dimethylformamide, N-methyl-pyrrolidone or dimethylacetamide, or mixtures of solvents
  • R 4 is as defined above
  • R 13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
  • R 15 represents methyl or ethyl, with a compound of the formula (XII) in which
  • R 14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl.
  • the reaction [G] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • Inert solvents are, for example, alcohols such as methanol, ethanol, iso-propanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, N-methyl-pyrrolidone, dimethylacetamide or acetonitrile; preference is given to ethanol.
  • alcohols such as methanol, ethanol, iso-propanol
  • ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran
  • other solvents such as dimethylformamide, N-methyl-pyrrolidone, dimethylacetamide or acetonitrile; preference is given to ethanol.
  • the compounds of the formula (XII) are known or can be synthesized from the corresponding starting compounds by known processes.
  • the compounds of the formula (XI) are known or can be prepared
  • R 13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
  • R 15 represents methyl or ethyl, with pivaloyl chloride and a base.
  • the reaction [H] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • Bases are, for example, organic bases such as trialkylamines, for example triethylamine, N- methylmorpholine, /V-methylpiperidine, 4-dimethylaminopyridine or diisopropylethylamin, or pyridine; preference is given to triethylamine.
  • organic bases such as trialkylamines, for example triethylamine, N- methylmorpholine, /V-methylpiperidine, 4-dimethylaminopyridine or diisopropylethylamin, or pyridine; preference is given to triethylamine.
  • Inert solvents are, for example, ethers such as diethyl ether, methyl tert- butyl ether, 1,2- dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as toluene, dimethylformamide, N-methyl-pyrrolidone or dimethylacetamide; preference is given to toluene.
  • ethers such as diethyl ether, methyl tert- butyl ether, 1,2- dimethoxyethane, dioxane or tetrahydrofuran
  • solvents such as toluene, dimethylformamide, N-methyl-pyrrolidone or dimethylacetamide; preference is given to toluene.
  • R 13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with compounds of the formula (XVI) in which
  • R 14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, in the presence of a base.
  • the reaction [I] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • Bases are, for example, alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate, or organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to triethylamine.
  • alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate
  • organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to triethylamine.
  • Inert solvents are, for example, aromatic solvents such as toluene, xylene, chlorobenzene or 1,2- dichlorobenzene, alcohols such as methanol or ethanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, dimethylacetamide, /V-methyl-pyrrolidone, acetonitrile, acetone or pyridine, or mixtures of the solvents; preference is given to xylene.
  • aromatic solvents such as toluene, xylene, chlorobenzene or 1,2- dichlorobenzene
  • alcohols such as methanol or ethanol
  • ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydro
  • the compounds of the formula (XV) are known or can be synthesized from the corresponding starting compounds by known processes.
  • R 14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with hydroxylamine hydrochloride in the presence of a base.
  • the reaction [J] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • Bases are, for example, alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate, or organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to sodium carbonate.
  • alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate
  • organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to sodium carbonate.
  • Inert solvents are, for example, alcohols such as methanol or ethanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, dimethylacetamide, /V-methyl-pyrrolidone, acetonitrile, acetone or pyridine, or mixtures of the solvents; preference is given to ethanol.
  • alcohols such as methanol or ethanol
  • ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran
  • other solvents such as dimethylformamide, dimethylacetamide, /V-methyl-pyrrolidone, acetonitrile, acetone or pyridine, or mixtures of the solvents; preference is given to ethanol.
  • the compounds of the formula (XVII) are known or can be synthesized from the corresponding starting compounds by known processes.
  • the compounds of the formula (Vl-C) are known or can be synthesized from the corresponding starting compounds by known processes.
  • R 1 , R 2 and R 4 are as defined above, are known or can be prepared
  • R 1 , R 2 and R 4 are as defined above, and
  • R 3 represents hydrogen with an electrophilic fluorine source and a base, followed by a dehydrating agent.
  • the compounds of the formula (l-A) and the formula (l-A-l) are subgroups of the compounds of the formula (I).
  • the reaction [K] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
  • Suitable electrophilic fluorination reagents are, for example, Selectfluor®, Selectfluor® II, N- fluorobenzenesulfonimide, 2,6-dichloro-1-fluoropyridinium tetrafluoroborate, 2,6-dichloro-1- fluoropyridinium triflate; preference is given to Selectfluor®.
  • Bases are, for example, organic bases such as trialkylamines, for example triethylamine, N- methylmorpholine, /V-methylpiperidine, 4-dimethylaminopyridine or A/./V-diisopropylethylamine, or pyridine; preference is given to pyridine.
  • organic bases such as trialkylamines, for example triethylamine, N- methylmorpholine, /V-methylpiperidine, 4-dimethylaminopyridine or A/./V-diisopropylethylamine, or pyridine; preference is given to pyridine.
  • Inert solvents are, for example, ethers such as dioxane, diethyl ether, tetrahydrofuran, N,N- dimethylformamide, A/,/ ⁇ /-dimethylacetamide, dimethyl sulphoxide, /V-methylpyrrolidone or acetonitrile, or mixtures of the solvents; preference is given to acetonitrile.
  • ethers such as dioxane, diethyl ether, tetrahydrofuran, N,N- dimethylformamide, A/,/ ⁇ /-dimethylacetamide, dimethyl sulphoxide, /V-methylpyrrolidone or acetonitrile, or mixtures of the solvents; preference is given to acetonitrile.
  • Suitable dehydrating agents are, for example, 2,4,6-tripropyl-1,3,5,2,4,6-trioxatriphosphinane- 2,4,6-trioxide (T3P); preference is given to 2, 4, 6-tripropyl-1, 3, 5,2,4, 6-trioxatriphosphinane-2, 4,6- trioxide.
  • the compounds of 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 have an unforeseeable useful pharmacological activity spectrum and good pharmacokinetic behavior, in particular a sufficient exposure of such a compound in the blood above the minimal effective concentration within a given dosing interval after oral administration.
  • Such a profile results in an improved peak-to-trough ratio (quotient of maximum to minimum concentration) within a given dosing interval, which has the advantage that the compound can be administered less frequently and at a significantly lower dose to achieve an effect.
  • They are compounds that inhibit the activation of the EP3 receptor by its ligand Prostaglandin E2 (PGE2).
  • the present invention further provides for the use of the compounds according to the invention for the treatment and/or prophylaxis of disorders, in particular cardiovascular disorders, preferably thrombotic or thromboembolic disorders and/or thrombotic or thromboembolic complications such as acute coronary syndrome or myocardial infarction or ischemic stroke or peripheral arterial occlusive disease , and/or diabetes, and/or ophthalmic disorders and/or urogenital disorders, in particular those associated with excess PGE2.
  • disorders in particular cardiovascular disorders, preferably thrombotic or thromboembolic disorders and/or thrombotic or thromboembolic complications such as acute coronary syndrome or myocardial infarction or ischemic stroke or peripheral arterial occlusive disease , and/or diabetes, and/or ophthalmic disorders and/or urogenital disorders, in particular those associated with excess PGE2.
  • PGE2 concentrations have been measured in atherosclerotic vascular walls of mice and humans. Once released upon plaque rupture, PGE2 binds on four specific receptors EP1 , EP2, EP3 and EP4 on cell membranes. PGE2 has been shown to interfere with human and murine platelet function via EP3 and EP4 receptors. Stimulation of EP3 potentiates platelet activation and aggregation induced by primary agonists like collagen or ADP, whereas stimulation of EP4 inhibits platelet activation. This PGE2-dependent balance of platelet activation and inhibition can be tipped by modulation of EP3 or EP4 receptors.
  • blocking the EP3 receptor by specific antagonists should be a beneficial strategy for prevention and treatment of atherothrombosis by local abrogation of platelet activation without altering hemostasis.
  • the "thrombotic or thromboembolic disorders” include disorders which occur preferably in the arterial vasculature and which can be treated with the compounds according to the invention, in particular disorders leading to peripheral arterial occlusive disorders and in the coronary arteries of the heart, such as acute coronary syndrome (ACS), myocardial infarction with ST segment elevation (STEMI) and without ST segment elevation (non-STEMI), stable angina pectoris, unstable angina pectoris, reocclusions and restenoses after coronary interventions such as angioplasty, stent implantation or aortocoronary bypass, but also thrombotic or thromboembolic disorders in cerebrovascular arteries, such as transitory ischaemic attacks (TIA), ischemic strokes including cardioembolic strokes, such as strokes due to atrial fibrillation, non-cardioembolic strokes, such as lacunar stroke, strokes due to large or small artery diseases, or strokes due to undetermined cause,
  • ACS acute coronary
  • the compounds according to the invention are suitable in particular for the treatment and/or prophylaxis of disorders where, the pro-inflammatory component plays an essential role, including vasculitides like Kawasaki disease, Takayasu arteritis and Thrombangiitis obliterans (Buerger’s disease) as well as inflammatory disorders like myocarditis.
  • the pro-inflammatory component plays an essential role, including vasculitides like Kawasaki disease, Takayasu arteritis and Thrombangiitis obliterans (Buerger’s disease) as well as inflammatory disorders like myocarditis.
  • the compounds according to the invention are suitable for the treatment and/or prophylaxis of disorders of the urogenital tract like overactive bladder, interstitial cystitis and bladder pain syndrome.
  • the compounds according to the invention are suitable for the treatment and/or prophylaxis of diabetes mellitus including its end-organ manifestations like diabetic retinopathy and diabetic nephropathy.
  • the compounds according to the invention are suitable in particular for the treatment and/or prophylaxis of neurological disorders like neuropathic pain, Alzheimer’s disease and Parkinson’s disease. Moreover, the compounds according to the invention are suitable in particular for the treatment and/or prophylaxis of pulmonologic disorders like chronic cough, asthma and COPD.
  • the present invention further provides for the use of the compounds according to the invention for the treatment and/or prophylaxis of disorders, especially the disorders mentioned above.
  • the present invention further provides for the use of the compounds according to the invention for production of a medicament for the treatment and/or prophylaxis of disorders, especially the disorders mentioned above.
  • the present invention further provides a method for the treatment and/or prophylaxis of disorders, especially the disorders mentioned above, using a therapeutically effective amount of a compound according to the invention.
  • the present invention further provides the compounds according to the invention for use in a method for the treatment and/or prophylaxis of disorders, especially the disorders mentioned above, using a therapeutically effective amount of a compound according to the invention.
  • the present invention provides the compounds according to the invention for use in a method for the treatment and/or prophylaxis of thrombotic or thromboembolic, in particular atherothrombotic disorders using a therapeutically effective amount of a compound according to the invention.
  • the present invention further provides medicaments comprising a compound according to the invention and one or more further active compounds.
  • the compounds according to the invention can also be used for preventing coagulation ex vivo, for example for the protection of organs to be transplanted against organ damage caused by formation of clots and for protecting the organ recipient against thromboemboli from the transplanted organ, for preserving blood and plasma products, for cleaning/pretreating catheters and other medical auxiliaries and instruments, for coating synthetic surfaces of medical auxiliaries and instruments used in vivo or ex vivo or for biological samples which may comprise factor Xla or plasma kallikrein.
  • the present invention furthermore provides a method for preventing the coagulation of blood in vitro, in particular in banked blood or biological samples which may comprise factor Xla or plasma kallikrein or both enzymes, which method is characterized in that an anticoagulatory effective amount of the compound according to the invention is added.
  • the compounds of the invention can act systemically and/or locally.
  • 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 for oral administration, it is possible to formulate the compounds according to the invention to dosage forms known in the art that deliver the compounds of the invention rapidly and/or in a modified manner, such as, for example, tablets (uncoated or coated tablets, for example with enteric or controlled release coatings that dissolve with a delay or are insoluble), orally- disintegrating tablets, films/wafers, films/lyophylisates, capsules (for example hard or soft gelatine capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions. It is possible to incorporate the compounds according to the invention in crystalline and/or amorphised and/or dissolved form into said dosage forms.
  • Parenteral administration can be effected with avoidance of an absorption step (for example intravenous, intraarterial, intracardial, intraspinal or intralumbal) or with inclusion of absorption (for example intramuscular, subcutaneous, intracutaneous, percutaneous or intraperitoneal).
  • absorption step for example intravenous, intraarterial, intracardial, intraspinal or intralumbal
  • absorption for example intramuscular, subcutaneous, intracutaneous, percutaneous or intraperitoneal.
  • Administration forms which are suitable for parenteral administration are, inter alia, preparations for injection and infusion in the form of solutions, suspensions, emulsions, lyophylisates or sterile powders.
  • Suitable for extraocular (topic) administration are administration forms which operate in accordance with the prior art, which release the active compound rapidly and/or in a modified or controlled manner and which contain the active compound in crystalline and/or amorphized and/or dissolved form such as, for example, eye drops, sprays and lotions (e.g. solutions, suspensions, vesicular/colloidal systems, emulsions, aerosols), powders for eye drops, sprays and lotions (e.g. ground active compound, mixtures, lyophilisates, precipitated active compound), semisolid eye preparations (e.g. hydrogels, in-situ hydrogels, creams and ointments), eye inserts (solid and semisolid preparations, e.g. bioadhesives, films/wafers, tablets, contact lenses).
  • eye drops e.g. solutions, suspensions, vesicular/colloidal systems, emulsions, aerosols
  • powders for eye drops, sprays and lotions
  • Intraocular administration includes, for example, intravitreal, subretinal, subscleral, intrachoroidal, subconjunctival, retrobulbar and subtenon administration.
  • Suitable for intraocular administration are administration forms which operate in accordance with the prior art, which release the active compound rapidly and/or in a modified or controlled manner and which contain the active compound in crystalline and/or amorphized and/or dissolved form such as, for example, preparations for injection and concentrates for preparations for injection (e.g. solutions, suspensions, vesicular/colloidal systems, emulsions), powders for preparations for injection (e.g.
  • gels for preparations for injection semisolid preparations, e.g. hydrogels, in-situ hydrogels
  • implants solid preparations, e.g. biodegradable and nonbiodegradable implants, implantable pumps.
  • Examples which are suitable for other administration routes are pharmaceutical forms for inhalation [inter alia powder inhalers, nebulizers], nasal drops, nasal solutions, nasal sprays; tablets/films/wafers/capsules for lingual, sublingual or buccal administration; suppositories; eye drops, eye ointments, eye baths, ocular inserts, ear drops, ear sprays, ear powders, ear-rinses, ear tampons; vaginal capsules, aqueous suspensions (lotions, mixturae agitandae), lipophilic suspensions, emulsions, ointments, creams, transdermal therapeutic systems (such as, for example, patches), milk, pastes, foams, dusting powders, implants or stents.
  • inhalation inter alia powder inhalers, nebulizers
  • nasal drops nasal solutions, nasal sprays
  • tablets/films/wafers/capsules for lingual, sublingual or buccal
  • the compounds according to the invention can be incorporated into the stated administration forms. This can be effected in a manner known per se by mixing with pharmaceutically suitable excipients.
  • Pharmaceutically suitable excipients include, inter alia,
  • fillers and carriers for example cellulose, microcrystalline cellulose (such as, for example, Avicel ® ), lactose, mannitol, starch, calcium phosphate (such as, for example, Di-Cafos ® )),
  • ointment bases for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols
  • ointment bases for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols
  • bases for suppositories for example polyethylene glycols, cacao butter, hard fat
  • solvents for example water, ethanol, isopropanol, glycerol, propylene glycol, medium chain- length triglycerides fatty oils, liquid polyethylene glycols, paraffins
  • surfactants for example sodium dodecyl sulfate), lecithin, phospholipids, fatty alcohols (such as, for example, Lanette®), sorbitan fatty acid esters (such as, for example, Span®), polyoxyethylene sorbitan fatty acid esters (such as, for example, Tween®), polyoxyethylene fatty acid glycerides (such as, for example, Cremophor®), polyoxethylene fatty acid esters, polyoxyethylene fatty alcohol ethers, glycerol fatty acid esters, poloxamers (such as, for example, Pluronic®),
  • buffers for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine
  • acids and bases for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine
  • isotonicity agents for example glucose, sodium chloride
  • adsorbents for example highly-disperse silicas
  • viscosity-increasing agents for example polyvinylpyrrolidone, methylcellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose-sodium, starch, carbomers, polyacrylic acids (such as, for example, Carbopol®); alginates, gelatine),
  • disintegrants for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate (such as, for example, Explotab®), cross- linked polyvinylpyrrolidone, croscarmellose-sodium (such as, for example, AcDiSol®)
  • disintegrants for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate (such as, for example, Explotab®), cross- linked polyvinylpyrrolidone, croscarmellose-sodium (such as, for example, AcDiSol®)
  • lubricants for example magnesium stearate, stearic acid, talc, highly-disperse silicas (such as, for example, Aerosil®)
  • mould release agents for example magnesium stearate, stearic acid, talc, highly-disperse silicas (such as, for example, Aerosil®)
  • coating materials for example sugar, shellac
  • film formers for films or diffusion membranes which dissolve rapidly or in a modified manner for example polyvinylpyrrolidones (such as, for example, Kollidon®), polyvinyl alcohol, hydroxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, hydroxypropylmethylcellulose phthalate, cellulose acetate, cellulose acetate phthalate, polyacrylates, polymethacrylates such as, for example, Eudragit®)),
  • capsule materials for example gelatine, hydroxypropylmethylcellulose
  • synthetic polymers for example polylactides, polyglycolides, polyacrylates, polymethacrylates (such as, for example, Eudragit®), polyvinylpyrrolidones (such as, for example, Kollidon®), polyvinyl alcohols, polyvinyl acetates, polyethylene oxides, polyethylene glycols and their copolymers and blockcopolymers
  • plasticizers for example polyethylene glycols, propylene glycol, glycerol, triacetine, triacetyl citrate, dibutyl phthalate
  • stabilisers for example antioxidants such as, for example, ascorbic acid, ascorbyl palmitate, sodium ascorbate, butylhydroxyanisole, butylhydroxytoluene, propyl gallate
  • antioxidants for example antioxidants such as, for example, ascorbic acid, ascorbyl palmitate, sodium ascorbate, butylhydroxyanisole, butylhydroxytoluene, propyl gallate
  • preservatives for example parabens, sorbic acid, thiomersal, benzalkonium chloride, chlorhexidine acetate, sodium benzoate
  • colourants for example inorganic pigments such as, for example, iron oxides, titanium dioxide
  • flavourings • flavourings, sweeteners, flavour- and/or odour-masking agents.
  • the present invention furthermore relates to a pharmaceutical composition which comprises at least one compound according to the invention, conventionally together with one or more pharmaceutically suitable excipient(s), and to their use according to the present invention.
  • An embodiment of the invention are pharmaceutical compositions comprising at least one compound of formula (I) according to the invention, preferably together with at least one inert, non toxic, pharmaceutically suitable auxiliary, and the use of these pharmaceutical compositions for the above cited purposes.
  • the present invention covers pharmaceutical combinations, in particular medicaments, comprising at least one compound of general formula (I) of the present invention and at least one or more further active ingredients, in particular for the treatment and/or prophylaxis of cardiovascular disorders, preferably thrombotic or thromboembolic disorders, and diabetes, and also urogenital and ophthalmic disorders.
  • a “fixed combination” in the present invention is used as known to persons skilled in the art and is defined as a combination wherein, for example, a first active ingredient, such as one or more compounds of general formula (I) of the present invention, and a further active ingredient are present together in one unit dosage or in one single entity.
  • a “fixed combination” is a pharmaceutical composition wherein a first active ingredient and a further active ingredient are present in admixture for simultaneous administration, such as in a formulation.
  • Another example of a “fixed combination” is a pharmaceutical combination wherein a first active ingredient and a further active ingredient are present in one unit without being in admixture.
  • a non-fixed combination or “kit-of-parts” in the present invention is used as known to persons skilled in the art and is defined as a combination wherein a first active ingredient and a further active ingredient are present in more than one unit.
  • a non-fixed combination or kit-of-parts is a combination wherein the first active ingredient and the further active ingredient are present separately. It is possible for the components of the non-fixed combination or kit-of-parts to be administered separately, sequentially, simultaneously, concurrently or chronologically staggered.
  • inventive compounds can be employed alone or, if required, in combination with other active ingredients.
  • present invention further provides medicaments comprising at least one of the inventive compounds and one or more further active ingredients, especially for treatment and/or prophylaxis of the aforementioned disorders.
  • suitable active ingredient combinations include:
  • organic nitrates and NO donors for example sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhaled NO;
  • cGMP cyclic guanosine monophosphate
  • PDE phosphodiesterases
  • hypotensive active ingredients by way of example and with preference from the group of the calcium antagonists, angiotensin All antagonists, ACE inhibitors, NEP-inhibitors, vasopeptidase-inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, rho-kinase-inhibitors and the diuretics;
  • antiarrhythmic agents by way of example and with preference from the group of sodium channel blocker, beta-receptor blocker, potassium channel blocker, calcium antagonists, If- channel blocker, digitalis, parasympatholytics (vagoliytics), sympathomimetics and other antiarrhythmics as adenosin, adenosine receptor agonists as well as vernakalant;
  • positive-inotrop agents by way of example cardiac glycoside (Dogoxin), beta-adrenergic and dopaminergic agonists, such as isoprenalin, adrenalin, noradrenalin, dopamin or dobutamin;
  • Dogoxin cardiac glycoside
  • beta-adrenergic and dopaminergic agonists such as isoprenalin, adrenalin, noradrenalin, dopamin or dobutamin
  • vasopressin-receptor-antagonists by way of example and with preference from the group of conivaptan, tolvaptan, lixivaptan, mozavaptan, satavaptan, SR-121463, RWJ 676070 or BAY 86-8050, as well as the compounds described in WO 2010/105770, WO2011/104322 and WO 2016/071212;
  • active ingredients which alter 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, of 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.
  • cholesterol synthesis inhibitors such as, by way of example and preferably, HMG-CoA reductase inhibitors or squalene synthesis inhibitors, of ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid
  • bronchodilatory agents for example and with preference from the group of the beta-adrenergic rezeptor-agonists, such as, by way of example and preferably, albuterol, isoproterenol, metaproterenol, terbutalin, formoterol or salmeterol, or from the group of the anticholinergics, such as, by way of example and preferably, ipratropiumbromid;
  • anti-inflammatory agents for example and with preference from the group of the gluco corticoids, such as, by way of example and preferably, prednison, prednisolon, methylprednisolon, triamcinolon, dexamethason, beclomethason, betamethason, flunisolid, budesonid or fluticason as well as the non-steroidal anti-inflammatory agents (NSAIDs), by way of example and preferably, acetyl salicylic acid (aspirin), ibuprofen and naproxen, 5-amino salicylic acid-derivates, leukotriene-antagonists, TNF-alpha-inhibitors and chemokin-receptor antagonists, such as CCR1, 2 and/or 5 inhibitors;
  • NSAIDs non-steroidal anti-inflammatory agents
  • agents modulating the immune system for example immunoglobulins
  • agents that inhibit the signal transductions cascade for example and with preference from the group of the kinase inhibitors, byway of example and preferably, from the group of the tyrosine kinase- and/or serine/threonine kinase inhibitors;
  • agents that inhibit the degradation and modification of the extracellular matrix, for example and with preference from the group of the inhibitors of the matrix-metalloproteases (MMPs), by way of example and preferably, inhibitors of chymasee, stromelysine, collagenases, gelatinases and aggrecanases (with preference from the group of MMP-1, MMP-3, MMP-8, MMP-9, MMP-10, MMP-11 and MMP-13) as well as of the metallo-elastase (MMP-12) and neutrophil-elastase (HNE), as for example sivelestat or DX-890;
  • MMPs matrix-metalloproteases
  • agents that block the Kunststoff of serotonin to its receptor, for example and with preference antagonists of the 5-HT2b-receptor;
  • organic nitrates and NO-donators for example and with preference sodium nitroprussid, nitro glycerine, isosorbid mononitrate, isosorbid dinitrate, molsidomine or SIN-1, as well as inhaled NO;
  • agents that stimulates the synthesis of cGMP, wiessen sGC Modulatoren, for example and with preference riociguat, cinaciguat, vericiguat or BAY 1101042;
  • prostacyclin-analogs for example and with preference iloprost, beraprost, treprostinil or epoprostenol;
  • agents that inhibit soulble epoxidhydrolase (sEH), for example and with preference N,N'-Di- cyclohexyl urea, 12-(3-Adamantan-1-yl-ureido)-dodecanic acid or 1-Adamantan-1-yl-3- ⁇ 5-[2- (2-ethoxyethoxy)ethoxy]pentyl ⁇ -urea;
  • SEH soulble epoxidhydrolase
  • agents that interact with glucose metabolism for example and with preference insuline, biguanide, thiazolidinedione, sulfonyl urea, acarbose, DPP4 inhibitors, GLP-1 analogs or SGLT-1 inhibitors;
  • natriuretic peptides for example and with preference atrial natriuretic peptide (ANP), natriuretic peptide type B (BNP, Nesiritid) natriuretic peptide type C (CNP) or urodilatin;
  • ABP atrial natriuretic peptide
  • BNP natriuretic peptide type B
  • CNP natriuretic peptide type C
  • urodilatin urodilatin
  • agents that affect the energy metabolism of the heart for example and with preference etomoxir, dichloroacetat, ranolazine or trimetazidine, full or partial adenosine A1 receptor agonists such as GS-9667 (formerly known as CVT-3619), capadenoson, neladenoson and neladenoson bialanate; agents that affect the heart rate, for example and with preference ivabradin; cyclooxygenase inhibitors such as, for example, bromfenac and nepafenac; inhibitors of the kallikrein-kinin system such as, for example, safotibant and ecallantide; inhibitors of the sphingosine 1-phosphate signal paths such as, for example, sonepcizumab; inhibitors of the complement-C5a receptor such as, for example, eculizumab; plasminogen activators (thrombolytics/fibrinolytics)
  • anticoagulatory substances such as, for example, heparin (UFH), low- molecular-weight heparins (LMW), for example tinzaparin, certoparin, parnaparin, nadroparin, ardeparin, enoxaparin, reviparin, dalteparin, danaparoid, semuloparin (AVE 5026), adomiparin (M118) and EP-42675/ORG42675;
  • anticoagulants such as, for example, heparin (UFH), low- molecular-weight heparins (LMW), for example tinzaparin, certoparin, parnaparin, nadroparin, ardeparin, enoxaparin, reviparin, dalteparin, danaparoid, semuloparin (AVE 5026), adomiparin (M118) and EP-42675/ORG42675;
  • DTI direct thrombin inhibitors
  • Pradaxa diabigatran
  • atecegatran AZD- 0837
  • DP-4088 phosphatidylcholine
  • SSR-182289A argatroban
  • argatroban argatroban
  • bivalirudin and tanogitran BIBT-986 and prodrug BIBT-1011
  • hirudin thrombin inhibitors
  • direct factor Xa inhibitors such as, for example, rivaroxaban, apixaban, edoxaban (DU-176b), betrixaban (PRT-54021), R-1663, darexaban (YM-150), otamixaban (FXV-673/R PR- 130673), letaxaban (TAK-442), razaxaban (DPC-906), DX-9065a, LY-517717, tanogitran (BIBT-986, prodrug: BIBT-1011), idraparinux and fondaparinux;
  • direct factor Xa inhibitors such as, for example, rivaroxaban, apixaban, edoxaban (DU-176b), betrixaban (PRT-54021), R-1663, darexaban (YM-150), otamixaban (FXV-673/R PR- 130673), letaxaban (TAK-442), razax
  • inhibitors of coagulation factor XI and Xla such as, for example, FXI ASO-LICA, BAY 121- 3790, MAA868, BMS986177, EP-7041 and AB-022;
  • platelet aggregation inhibitors substances which inhibit the aggregation of platelets
  • thrombocyte aggregation inhibitors such as, for example, acetylsalicylic acid (such as, for example, aspirin), P2Y12 antagonists such as, for example, ticlopidine (Ticlid), clopidogrel (Plavix), prasugrel, ticagrelor, cangrelor and elinogrel, and PAR-1 antagonists such as, for example, vorapaxar, and PAR-4 antagonists;
  • platelet adhesion inhibitors such as GPVI and/or GPIb antagonists such as, for example, Revacept or caplacizumab;
  • fibrinogen receptor antagonists g lycoprotei n- 11 b/l 11 a antagonists
  • fibrinogen receptor antagonists such as, for example, abciximab, eptifibatide, tirofiban, lamifiban, lefradafiban and fradafiban;
  • recombinant human activated protein C such as, for example, Xigris or recombinant thrombomodulin.
  • Antithrombotic agents are preferably understood to mean compounds from the group of the platelet aggregation inhibitors, the anticoagulants or the profibrinolytic substances.
  • the inventive compounds are administered in combination with a platelet aggregation inhibitor, by way of example and with preference aspirin, clopidogrel, prasugrel, ticagrelor, ticlopidin or dipyridamole.
  • a platelet aggregation inhibitor by way of example and with preference aspirin, clopidogrel, prasugrel, ticagrelor, ticlopidin or dipyridamole.
  • the inventive compounds are administered in combination with a thrombin inhibitor, by way of example and with preference ximelagatran, dabigatran, melagatran, bivalirudin or clexane.
  • the inventive compounds are administered in combination with a GPIIb/llla antagonist such as, by way of example and with preference, tirofiban or abciximab.
  • the inventive compounds are administered in combination with a factor Xa inhibitor, by way of example and with preference rivaroxaban (BAY 59- 7939), DU-176b, apixaban, betrixaban, otamixaban, fidexaban, razaxaban, letaxaban, eribaxaban, fondaparinux, idraparinux, PMD-3112, darexaban (YM-150), KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
  • a factor Xa inhibitor by way of example and with preference rivaroxaban (BAY 59- 7939), DU-176b, apixaban, betrixaban, otamixaban, fidexaban, razaxaban, letaxaban, eribaxaban, fondapar
  • the inventive compounds are administered in combination with a factor XI or factor Xla inhibitor, by way of example and with preference FXI ASO- LICA, BAY 121-3790, MAA868, BMS986177, EP-7041 or AB-022.
  • the inventive compounds are administered in combination with heparin or with a low molecular weight (LMW) heparin derivative.
  • LMW low molecular weight
  • the inventive compounds are administered in combination with a vitamin K antagonist, by way of example and with preference coumarin.
  • Hypotensive agents are preferably understood to mean compounds from the group of the calcium antagonists, angiotensin All antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, rho-kinase inhibitors and the diuretics.
  • the inventive compounds are administered in combination with a calcium antagonist, by way of example and with preference nifedipine, amlodipine, verapamil or diltiazem.
  • a calcium antagonist by way of example and with preference nifedipine, amlodipine, verapamil or diltiazem.
  • the inventive compounds are administered in combination with an alpha- 1 -receptor blocker, by way of example and with preference prazosin.
  • the inventive compounds 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.
  • a beta-receptor blocker by way of example and with preference propranolol, atenolol, timolol, pindolol
  • the inventive compounds are administered in combination with an angiotensin All antagonist, by way of example and with preference losartan, candesartan, valsartan, telmisartan or embusartan or a dual angiotensin All antagonist/neprilysin- inhibitor, by way of example and with preference LCZ696 (valsartan/sacubitril).
  • the inventive compounds are administered in combination with an ACE inhibitor, by way of example and with preference enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril ortrandopril.
  • an ACE inhibitor by way of example and with preference enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril ortrandopril.
  • an endothelin antagonist by way of example and with preference bosentan, darusentan, ambrisentan or sitaxsentan.
  • the inventive compounds are administered in combination with a renin inhibitor, by way of example and with preference aliskiren, SPP-600 or SPP-800.
  • the inventive compounds are administered in combination with a mineralocorticoid receptor antagonist, by way of example and with preference spironolactone or eplerenone.
  • the inventive compounds are administered in combination with a loop diuretic, for example furosemide, torasemide, bumetanide and piretanide, with potassium-sparing diuretics, for example amiloride and triamterene, with aldosterone antagonists, for example spironolactone, potassium canrenoate and eplerenone, and also thiazide diuretics, for example hydrochlorothiazide, chlorthalidone, xipamide and indapamide.
  • a loop diuretic for example furosemide, torasemide, bumetanide and piretanide
  • potassium-sparing diuretics for example amiloride and triamterene
  • aldosterone antagonists for example spironolactone
  • potassium canrenoate and eplerenone potassium canrenoate and eplerenone
  • thiazide diuretics for example hydrochlorothiazide, chlorthalidone,
  • Lipid metabolism modifiers are preferably understood to mean compounds from the group of the CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA reduc tase inhibitors or squalene synthesis inhibitors, the 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 the lipoprotein(a) antagonists.
  • the CETP inhibitors such as HMG-CoA reduc tase inhibitors or squalene synthesis inhibitors
  • ACAT inhibitors such as HMG-CoA reduc tase inhibitors or squalene synthesis inhibitors
  • MTP inhibitors MTP inhibitors
  • PPAR-alpha PPAR-gamma and/or PPAR-delta agonists
  • cholesterol absorption inhibitors polymeric
  • the inventive compounds are administered in combination with a CETP inhibitor, by way of example and with preference dalcetrapib.anacetrapib, torcetrapib (CP- 529414), JJT-705 or CETP vaccine (Avant).
  • a CETP inhibitor by way of example and with preference dalcetrapib.anacetrapib, torcetrapib (CP- 529414), JJT-705 or CETP vaccine (Avant).
  • the inventive compounds are administered in combination with a thyroid receptor agonist, by way of example and with preference D-thyroxine, 3,5,3'-triiodothyronine (T3), CGS 23425 or axitirome (CGS 26214).
  • a thyroid receptor agonist by way of example and with preference D-thyroxine, 3,5,3'-triiodothyronine (T3), CGS 23425 or axitirome (CGS 26214).
  • T3 3,5,3'-triiodothyronine
  • CGS 23425 CGS 23425
  • CGS 26214 axitirome
  • the inventive compounds 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.
  • the inventive compounds are administered in combination with a squalene synthesis inhibitor, by way of example and with preference BMS- 188494 or TAK-475.
  • the inventive compounds are administered in combination with an ACAT inhibitor, by way of example and with preference avasimibe, melinamide, pactimibe, eflucimibe or SM P-797.
  • an ACAT inhibitor by way of example and with preference avasimibe, melinamide, pactimibe, eflucimibe or SM P-797.
  • the inventive compounds are administered in combination with an MTP inhibitor, byway of example and with preference implitapide, BMS-201038, R- 103757 or JTT-130.
  • the inventive compounds are administered in combination with a PPAR-gamma agonist, by way of example and with preference pioglitazone or rosiglitazone.
  • the inventive compounds are administered in combination with a PPAR-delta agonist, by way of example and with preference GW 501516 or BAY 68-5042.
  • the inventive compounds are administered in combination with a cholesterol absorption inhibitor, by way of example and with preference ezetimibe, tiqueside or pamaqueside.
  • the inventive compounds are administered in combination with a lipase inhibitor, a preferred example being orlistat.
  • the inventive compounds are administered in combination with a polymeric bile acid adsorbent, by way of example and with preference cholestyramine, colestipol, colesolvam, CholestaGel or colestimide.
  • the inventive compounds are administered in combination with a lipoprotein(a) antagonist, by way of example and with preference, gemcabene calcium (CI-1027) or nicotinic acid.
  • a lipoprotein(a) antagonist by way of example and with preference, gemcabene calcium (CI-1027) or nicotinic acid.
  • the inventive compounds are administered in combination with a lipoprotein(a) antagonist, by way of example and with preference, gemcabene calcium (CI-1027) or nicotinic acid.
  • a lipoprotein(a) antagonist by way of example and with preference, gemcabene calcium (CI-1027) or nicotinic acid.
  • the inventive compounds are administered in combination with sGC modulators, by way of example and with preference, riociguat, cinaciguat or vericiguat.
  • the inventive compounds are administered in combination with an agent affecting the glucose metabolism, by way of example and with preference, insuline, a sulfonyl urea, acarbose, DPP4 inhibitors, GLP-1 analogs or SGLT-1 inhibitors.
  • the compounds according to the invention are administered in combination with a TGFbeta antagonist, by way of example and with preference pirfenidone or fresolimumab.
  • the compounds according to the invention are administered in combination with a CCR2 antagonist, by way of example and with preference CCX- 140.
  • the compounds according to the invention are administered in combination with a TNFalpha antagonist, by way of example and with preference adalimumab.
  • the compounds according to the invention are administered in combination with a galectin-3 inhibitor, by way of example and with preference GCS- 100.
  • the compounds according to the invention are administered in combination with a Nrf-2 inhibitor, by way of example and with preference bardoxolone
  • the compounds according to the invention are administered in combination with a BMP-7 agonist, by way of example and with preference THR- 184.
  • the compounds according to the invention are administered in combination with a NOX1/4 inhibitor, by way of example and with preference GKT- 137831.
  • the compounds according to the invention are administered in combination with a medicament which affects the vitamin D metabolism, by way of example and with preference calcitriol, alfacalcidol, doxercalciferol, maxacalcitol, paricalcitol, cholecalciferol or paracalcitol.
  • the compounds according to the invention are administered in combination with a cytostatic agent, by way of example and with preference cyclophosphamide.
  • the compounds according to the invention are administered in combination with an immunosuppressive agent, by way of example and with preference ciclosporin.
  • the compounds according to the invention are administered in combination with a phosphate binder, by way of example and with preference colestilan, sevelamer hydrochloride and sevelamer carbonate, Lanthanum and lanthanum carbonate.
  • the compounds according to the invention are administered in combination with renal proximal tubule sodium-phosphate co-transporter, by way of example and with preference, niacin or nicotinamide.
  • the compounds according to the invention are administered in combination with a calcimimetic for therapy of hyperparathyroidism.
  • the compounds according to the invention are administered in combination with agents for iron deficit therapy, by way of example and with preference iron products.
  • the compounds according to the invention are administered in combination with agents for the therapy of hyperurikaemia, by way of example and with preference allopurinol or rasburicase.
  • the compounds according to the invention are administered in combination with glycoprotein hormone for the therapy of anaemia, by way of example and with preference erythropoietin.
  • the compounds according to the invention are administered in combination with biologies for immune therapy, by way of example and with preference abatacept, rituximab, eculizumab or belimumab.
  • the compounds according to the invention are administered in combination with vasopressin antagonists (group of the vaptanes) for the treatment of heart failure, by way of example and with preference tolvaptan, conivaptan, lixivaptan, mozavaptan, satavaptan or relcovaptan.
  • vasopressin antagonists group of the vaptanes
  • the compounds according to the invention are administered in combination with Jak inhibitors, by way of example and with preference ruxolitinib, tofacitinib, baricitinib, CYT387, GSK2586184, lestaurtinib, pacritinib (SB1518) or TG101348.
  • Jak inhibitors by way of example and with preference ruxolitinib, tofacitinib, baricitinib, CYT387, GSK2586184, lestaurtinib, pacritinib (SB1518) or TG101348.
  • the compounds according to the invention are administered in combination with prostacyclin analogs for therapy of microthrombi.
  • the compounds according to the invention are administered in combination with an alkali therapy, by way of example and with preference sodium bicarbonate.
  • the compounds according to the invention are administered in combination with an mTOR inhibitor, by way of example and with preference everolimus or rapamycin.
  • the compounds according to the invention are administered in combination with an NHE3 inhibitor, by way of example and with preference AZD1722 or tenapanor.
  • the compounds according to the invention are administered in combination with an eNOS modulator, by way of example and with preference sapropterin.
  • the compounds according to the invention are administered in combination with a CTGF inhibitor, by way of example and with preference FG-3019.
  • the total amount of the active ingredient to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 50 mg/kg body weight per day, and more preferably from about 0.01 mg/kg to about 10 mg/kg body weight per day.
  • Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing.
  • drug holidays in which a patient is not dosed with a drug for a certain period of time, to be beneficial to the overall balance between pharmacological effect and tolerability.
  • a unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day.
  • the average daily dosage for administration by injection including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily.
  • the transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg.
  • the average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like.
  • the desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
  • the compounds of formula (I) according to the invention are administered orally once or twice or three times a day. According to a further embodiment, the compounds of formula (I) according to the invention are administered orally once or twice a day. According to a further embodiment, the compounds of formula (I) according to the invention are administered orally once a day. For the oral administration, a rapid release or a modified release dosage form may be used.
  • NMR peak forms are stated as they appear in the spectra, possible higher order effects have not been considered.
  • the 1 H-NMR data of selected compounds are listed in the form of 1 H-NMR peaklists. For each signal peak the d value in ppm is given, followed by the signal intensity, reported in round brackets. The d value-signal intensity pairs from different peaks are separated by commas. Therefore, a peaklist is described by the general form: di (intensityi), 62 (intens ⁇ ), ... , d, (intensity,), ... , d h (intensity ⁇ .
  • the intensity of a sharp signal correlates with the height (in cm) of the signal in a printed NMR spectrum.
  • a 1 H-NMR peaklist is similar to a classical 1 H-NMR readout, and thus usually contains all the peaks listed in a classical NMR interpretation. Moreover, similar to classical 1 H-NMR printouts, peaklists can show solvent signals, signals derived from stereoisomers of target compounds (also the subject of the invention), and/or peaks of impurities.
  • the peaks of stereoisomers, and/or peaks of impurities are typically displayed with a lower intensity compared to the peaks of the target compounds (e.g., with a purity of >90%).
  • Such stereoisomers and/or impurities may be typical for the particular manufacturing process, and therefore their peaks may help to identify the reproduction of our manufacturing process on the basis of "by-product fingerprints".
  • An expert who calculates the peaks of the target compounds by known methods can isolate the peaks of target compounds as required, optionally using additional intensity filters. Such an operation would be similar to peak-picking in classical 1 H-NMR interpretation.
  • Method 1 Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ⁇ 1.2 min 100% B ® 2.0 min 100% B; column oven: 40°C; flow rate: 1.2 ml/min.
  • Method 2 Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 1.0 ml/min.
  • Method 3 Column: XBridge C18, 3.5 pm, 4.6 c 50 mm; mobile phase A: 5mM ammonium hydrogencarbonate in water, mobile phase B: acetonitrile; gradient: 0.0 min 10% B ® 4.2 min 95% B ® 5.2 min 95% B; column oven: 35°C; flow rate: 1.5 ml/min.
  • Method 4 Column: Ascentis Express C18, 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 100% B ® 1.7 min 100% B; column oven: 40°C; flow rate: 1.0 ml/min.
  • Method 5 Column: Ascentis Express C18, 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.7 min 95% B; column oven: 40°C; flow rate: 1.0 ml/min.
  • Method 6 Column: Ascentis Express C18, 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 100% B ® 1.7 min 100% B; column oven: 40°C; flow rate: 1.0 ml/min.
  • Method 7 Column: Kinetex EVO-C18 (Phenomenex), 2.6 pm, 3.0 c 50 mm.
  • Method 8 Instrument: Waters Acquity SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8 pm, 50 x 1 mm; eluent A: 1 L water + 0.25 ml formic acid, eluent B: 1 L acetonitrile + 0.25 ml formic acid; gradient: 0.0 min 90% A ® 1.2 min 5% A ® 2.0 min 5% A; column oven: 50°C; flow rate: 0.40 ml/min; UV detection: 208-400 nm.
  • Method 9 MS instrument: Thermo Scientific FT-MS; instrument UHPLC+: Thermo Scientific UltiMate 3000; column: Waters HSS T3, 2.1 c 75 mm, C18 1.8 pm; eluent A: 1 L water + 0.01% formic acid, eluent B: 1 L acetonitrile + 0.01% formic acid; gradient: 0.0 min 10% B ® 2.5 min 95% B ® 3.5 min 95% B; oven: 50°C; flow rate: 0.90 ml/min; UV detection: 210 nm/optimum integration path 210-300 nm.
  • Method 10 Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 0.8 ml/min.
  • Method 11 Column: Kinetex EVO-C18, 2.6 pm, 3.0 c 50 mm; mobile phase A: 5mM ammonium hydrogencarbonate in water, mobile phase B: acetonitrile; gradient: 0.0 min 10% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 1.2 ml/min.
  • Method 12 Column: Omega, 3.0 pm, 2.1 c 50 mm; mobile phase A: 0.09% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 1.2 ml/min.
  • Method 13 Column: Ascentis Express C18, 2.7 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.7 min 95% B; column oven: 40°C; flow rate: 1.5 ml/min.
  • Method 14 Column: CORTECS C18+ (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.09% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 100% B -> 1.6 min 100% B; column oven: 40°C; flow rate: 0.8 ml/min.
  • Method 15 Column: Shim-pack XR-ODS (Shimadzu), 2.2 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 95% B ® 1.6 min 95% B; column oven: 40°C; flow rate: 1.5 ml/min.
  • Method 16 Column: Ascentis Express C18, 2.7 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ⁇ 2.1 min 100% B ® 2.8 min 100% B; column oven: 40°C; flow rate: 1.5 ml/min.
  • Method 17 Column: Ascentis Express C18 (Supelco), 2.7 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.7 min 95% B; column oven: 40°C; flow rate: 1.5 ml/min.
  • Method 18 Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 3.7 min 70% B ® 4.2 min 95% B ® 4.7 min 95% B; column oven: 40°C; flow rate: 0.8 ml/min.
  • Method 19 Column: Shim-pack XR-ODS (Shimadzu), 2.2 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.7 min 95% B; column oven: 40°C; flow rate: 1.5 ml/min.
  • Method 20 Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 1.2 ml/min.
  • Method 21 Column: CORTECS C18+ (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.09% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 0.8 ml/min.
  • Method 22 MS instrument: Waters Single Quad MS System; Waters UPLC Acquity; column: Waters BEH C18, 1.7 pm, 50 c 2.1 mm; eluent A: 1 L water + 1.0 ml aq. ammonium hydroxide solution (25% ammonia), eluent B: 1 L acetonitrile; gradient: 0.0 min 92% A ® 0.1 min 92% A ® 1.8 min 5% A ® 3.5 min 5% A; column oven: 50°C; flow rate: 0.45 ml/min; UV detection: 210 nm (208-400 nm).
  • Method 23 Instrument: Waters Acquity SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8 pm, 50 c 1 mm; eluent A: 1 L water + 0.25 ml formic acid, eluent B: 1 L acetonitrile + 0.25 ml formic acid; gradient: 0.0 min 95% A ® 6.0 min 5% A ® 7.5 min 5% A; column oven: 50°C; flow rate: 0.35 ml/min; UV detection: 210 nm.
  • Method 24 MS instrument: Waters Single Quad MS System; Waters UPLC Acquity; column: Waters BEH C18, 1.7 pm, 50 c 2.1 mm; eluent A: 1 L water + 1.0 ml aq. ammonium hydroxide solution (25% ammonia), eluent B: 1 L acetonitrile; gradient: 0.0 min 92% A ® 0.1 min 92% A ® 1.8 min 5% A ® 3.5 min 5% A; column oven: 50°C; flow rate: 0.45 ml/min; UV detection: 210 nm.
  • Method 25 Column: Ascentis Express C18 (Supelco), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 100% B ® 2.0 min 100% B; column oven: 40°C; flow rate: 1.5 ml/min.
  • Method 26 Instrument: Waters ACQUITY SQD UPLC System; Column: Waters Acquity UPLC HSS T3 1.8 pm 50 x 1 mm; eluent A: 1 L water + 0.25 ml 99% formic acid, eluent B: 1 L acetonitrile + 0.25 ml 99% formic acid; gradient: 0.0 min 90% A 1.2 min 5% A 2.0 min 5% A oven: 50°C; flow rate: 0.40 ml/min; UV-Detection: 210 nm.
  • Method 27 Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 3.2 min 50% B ® 4.2 min 95% B® 5.0 min 95% B; column oven: 40°C; flow rate: 1.0 ml/min.
  • Method 28 Instrument MS: Waters SQD; Instrument HPLC: Waters UPLC; column: Zorbax SB- Aq (Agilent), 50 mm x 2.1 mm, 1.8 pm; eluent A: water + 0.025% formic acid, eluent B: acetonitrile (ULC) + 0.025% formic acid; gradient: 0.0 min 98% A ® 0.9 min 25% A ® 1.0 min 5% A ® 1.4 min 5% A ® 1.41 min 98% A -> 1.5 min 98% A; oven: 40°C; flow: 0.600 ml/min; UV-detection: DAD; 210 nm.
  • Method G1 Instrument: Thermo Scientific DSQII, Thermo Scientific Trace GC Ultra; Column: Restek RTX-35MS, 15 m x 200 pm x 0.33 pm; constant flow with helium, flow rate: 1.2 ml/min; oven: 60°C; inlet: 220°C; gradient: 60°C, 30°C/min 300°C (3.33 min stop).
  • Method P1 Instrument: Waters Prep LC/MS System; column: XBridge C18 5 pm, 100 c 30 mm; eluent A: water, eluent B: acetonitrile; flow rate: 80 ml/min plus 5.0 ml of aq. ammonia (2% ammonia in water); at-column injection; gradient: 0.0-2.0 min 0% B, 2.0-10 min 0% B ® 100% B, 10-12 min 100%; column oven: RT; UV detection: 200-400 nm.
  • Method P3 Column: Phenomenex Gemini C18 250 x 50mm x 10 urn; eluent A: water + 0.05% of ammonia; eluent B: acetonitrile; gradient: 0-28 min 10% B ® 35% B.
  • Method P4 Column: Chromatorex C18, 10pm, 205 c 50 mm; eluent A: water + 0.1% of formic acid; eluent B: acetonitrile; gradient: 0.0-5.0 min 10% B, 5.0-17.5 min 10% B to 95% B, 17.5-21.0 min 95% B; flow rate: 150 ml/min, UV-Detection: 210 nm.
  • Microwave Reactions employing microwave irradiation may be run with a Biotage Initator® microwave oven optionally equipped with a robotic unit.
  • the reported reaction times employing microwave heating are intended to be understood as fixed reaction times after reaching the indicated reaction temperature.
  • 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 contain a sufficiently basic or acidic functionality.
  • a salt can be converted to the corresponding free base or acid by various methods known to the person skilled in the art.
  • any compound specified in the form of a salt of the corresponding base or acid is generally a salt of unknown exact stoichiometric composition, as obtained by the respective preparation and/or purification process.
  • names and structural formulae such as “hydrochloride”, “trifluoroacetate”, “sodium salt” or “x HCI”, “x CF 3 COOH”, “x Na + " should not therefore be understood in a stoichiometric sense in the case of such salts, but have merely descriptive character with regard to the salt-forming components present therein.
  • reaction mixture was partitioned between water (50 ml) and dichloromethane (100 ml) and the aqueous phase was extracted with dichloromethane (2 x 100 ml).
  • dichloromethane 2 x 100 ml
  • the combined organic phase was dried, concentrated and purified by silica gel column chromatography (eluent: dichloromethane/methanol) to afford 820 mg (54% of theory, 97% purity) of the title compound.
  • the reaction was heated to 40°C for 72 h, then the reaction was concentrated to remove the THF and water was added.
  • the pH of the aqueous solution was adjusted to 1 to 2 with hydrochloric acid (1.0 N aqueous solution) and the precipitated was collected by filtration and dried to afford 418 mg (87% of theory, 100% purity) of the title compound.
  • the reaction mixture was concentrated to remove the ethanol, then the water (100 ml) was added and the aquoues solution was extracted with MTBE (2 x 100 ml).
  • the aqueous phase was acidified with hydrochloric acid (2.0 N aquoues solution) to adjust the pH value to 1.
  • the product was collected by filtration, washed with water and dried to afford 1.35 g (87% of theory, 86% purity) of the title compound.
  • Ethyl (3S)-3-amino-2,2-difluoro-3-(4-fluorophenyl)propanoate hydrochloride To a solution of ethyl (3S)-3-[[(S)-te/f-butylsulfinyl]amino]-2,2-difluoro-3-(4-fluorophenyl) propanoate (Intermediate 55A, 46.0 g, 131 mmol) in ethanol (300 ml) was added hydrochloric acid (350 ml of a 4.0 M solution in 1 ,4-dioxane) and the mixture was stirred at 30°C for 2 h.
  • hydrochloric acid 350 ml of a 4.0 M solution in 1 ,4-dioxane
  • the aqueous layer was brought to basic pH by addition of sodium hydroxide (1.0 M aqueous solution) and was extracted with ethyl acetate. The combined organic layers were dried over magnesium sulfate and concentrated. The residue was purified by silica gel column chromatography (basified silica gel, eluent: gradient of methanol in dichloromethane). Yield: 203 mg (48% of theory, 95% purity).
  • Triethylamine (5.2 ml, 37 mmol) was added to a solution of thionylchloride (1.4 ml, 19 mmol) in dichloromethane (60.0 ml), and the mixture was cooled to -60°C.
  • a solution of tert.- butyl [( 7S)-2- hydroxy- 1-phenylethyl]carbamate (4.00 g, 16.9 mmol) in dichloromethane (100 ml) was added dropwise, and the reaction was stirred at -60°C for 2 h. After warming to RT, water was added and the layers were separated. The organic layer was washed with brine, dried over magnesium sulfate and evaporated to dryness.
  • 6-(2,3-Dihydro-1,4-benzodioxin-6-yl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2-carboxylic acid To a solution of ethyl 6-(2,3-dihydro-1,4-benzodioxin-6-yl)-8-oxo-7,8-dihydroimidazo[1,2- a]pyrazine-2-carboxylate (Intermediate 80A, 300 mg, 879 pmol) in water (10 ml) and ethanol (10 ml) was added sodium hydroxide (3.5 ml of a 2.0 M aqueous solution, 7.00 mmol).
  • the racemic amide was separated into its enantiomeres by preparative HPLC (column: Daicel OJ- H, 250 x 25 mm; flow rate: 80 ml/min; temperature: 40°C; eluent: 70% supercritical CC> 2 :30% methanol).
  • the racemic amide was separated into its enantiomeres by preparative HPLC (column: Daicel Chiralpak IF, 5 pm, 250 c 20 mm; flow rate: 15 ml/min; temperature: 50°C; eluent: 30% n- heptan:70% ethanol).
  • the racemic amide was separated into its enantiomeres by preparative HPLC (column: Daicel Chiralpak IE, 5 pm, 250 c 20 mm; flow rate: 20 ml/min; temperature: 30°C; eluent: n-heptan ethanol; 0.0 5.0 min, 50% ethanol, 5.1 15.0, 75% ethanol.
  • HPLC columnumn: Daicel Chiralpak IE, 5 pm, 250 c 20 mm; flow rate: 20 ml/min; temperature: 30°C; eluent: n-heptan ethanol; 0.0 5.0 min, 50% ethanol, 5.1 15.0, 75% ethanol.
  • the racemic amide was separated into its enantiomeres by preparative HPLC (column: Daicel Chiralpak IC, 5 pm, 250 c 20 mm; flow rate: 15 ml/min; temperature: 40°C; eluent: 80% n- heptan:20% ethanol).
  • Instrument MS Waters
  • Instrument HPLC Waters (column Phenomenex Luna 5m C18(2) 100A, AXIA Tech. 50 x 21.2 mm, eluent A: water, eluent B: acetonitrile (ULC), gradient; flow: 38.5 ml/min + 1.5 ml/min modifier (10% aq.
  • UV-detection DAD; 210-400 nm
  • Instrument MS Waters
  • Instrument HPLC Waters (column Waters X-Bridge C18, 19 mm x 50 mm, 5 pm
  • eluent A water
  • eluent B acetonitrile (ULC), gradient; flow 38.5 ml/min + 1.5 ml/min modifier (10% aq. ammonia)
  • the product containing fractions were evaporated in vacuo with a centrifugal dryer, dissolved in DMSO, pooled and evaporated again to give the final product.
  • the average value also referred to as the arithmetic mean value, represents the sum of the values obtained divided by the number of times tested
  • the median value represents the middle number of the group of values when ranked in ascending or descending order. If the number of values in the data set is odd, the median is the middle value. If the number of values in the data set is even, the median is the arithmetic mean of the two middle values.
  • Examples were synthesized one or more times. When synthesized more than once, data from biological assays represent average values calculated utilizing data sets obtained from testing of one or more synthetic batch.
  • the identification of antagonists of the EP3 receptor from humans and rats as well as the quantification of the activity of the compounds of the invention was carried out using recombinant cell lines. These cell lines originally derive from a hamster's ovary epithelial cell (Chinese Hamster Ovary, CHO K1, ATCC: American Type Culture Collection, Manassas, VA 20108, USA). The test cell lines constitutively express the human or rat EP3 receptors.
  • the naturally G ai -coupled human and rat EP3 receptors were stably co-transfected with G-ne into cells that are also stably transfected with a mitochondrial form of the calcium-sensitive photoproteins Clytin (human EP3) or Photina (rat EP3), which, after reconstitution with the cofactor coelenterazine, emit light when there are increases in free calcium concentrations ( Nature 1992, 358, 325-327; Gene 1995, 153 (2), 273-274).
  • the resulting EP3 receptor cells react to stimulation of the recombinantly expressed EP3 receptors by the agonist sulprostone with intracellular release of calcium ions, which can be quantified by the resulting photoprotein luminescence.
  • Antagonists inhibit the receptor signaling, leading to reduced calcium release and luminescence.
  • the bioluminescence of the cell lines is detected using a suitable luminometer (Trends Pharmacol. Sci. 1996, 17, 235-237).
  • the cells are plated out in culture medium (OptiMEM, 1% FCS, 2 mM glutamine, 10 mM HEPES, 5 pg/ml coelenterazine) in 384-well microtiter plates and kept in a cell incubator (96% humidity, 5% v/v CO2, 37°C).
  • culture medium OptiMEM, 1% FCS, 2 mM glutamine, 10 mM HEPES, 5 pg/ml coelenterazine
  • test compounds in various concentrations are placed for 10 minutes in the wells of the microtiter plate before the agonist sulprostone at EC 50 concentration is added. The resulting light signal is measured immediately in a luminometer. All concentrations are measured in quadruples. Table A:
  • Blood collection Human blood is collected by antecubital venipuncture using a 20G multifly set (Sarstedt, Num- brecht, Germany) into plastic tubes (Sarstedt 9 NC/10 ml monovettes) containing 3.8% (w/v) sodium citrate (1/10 v/v) from healthy donors who denied having taken any medication for at least 7 days prior to the study. Tubes are gently mixed to prevent coagulation.
  • PRP platelet rich plasma
  • PPP platelet poor plasma
  • Platelet aggregation is analyzed by light transmittance aggregometry according to the method of Born (J. Physiol. 1963, 168, 178-195) using APACT aggregometers (Rolf Greiner BioChemicals, Flacht, Germany).
  • 178 pi of PRP is incubated with test compound in various concentrations dissolved in 2 mI DMSO in the aggregation cuvette at 37°C for 5 min. Then 2 mI sulprostone is added and is incubated at 37°C for 2 min. Aggregation is started by addition of 20 mI of collagen.
  • concentrations of agonists are used: sulprostone 0.02 mM-2.5mM (Sigma-Aldrich), collagen 0.05-0.6 pg/ml (Horm ® , Nycomed).
  • the reaction is followed by monitoring changes in light transmission on PRP aliquots against PPP control. Light transmittance is recorded for 5 minutes and the maximal aggregation value is used for IC 50 calculations.
  • IC50 is calculated with GrapPad Prism (sigmoidal dose-response). All values are expressed as means ⁇ SEM, with n denoting the number of blood donors.
  • GrapPad Prism sigmoidal dose-response
  • reaction buffer 50 mM Tris/HCI, pH 7.5; 10 mM MgCI2; 1 mM EDTA
  • reaction buffer 50 mM Tris/HCI, pH 7.5; 10 mM MgCI2; 1 mM EDTA
  • suitable 96 well clear bottom plate [3H] Prostaglandin E2 ([5,6,8,11,12,14,15-3H(N)] in a final concentration of 1.5 nM;
  • PVT-WGA Beads in a final concentration of 2.5 mg/ml in a final concentration of 2.5 mg/ml.
  • the mixture is incubated for 2 h at room temperature. Scintillation is monitored in a suitable microplate reader equipped with 3H-SPA program. The values are used to plot dose response curves and calculate EC50 values for the test compounds.
  • This study serves to investigate the efficacy of a test substance on thrombus formation, stability and resolution induced by laser injury to microvessels of atherosclerotic mice.
  • mice Male ApoE -/- mice (Charles River) are fed an atherogenic diet (Ssniff S0602-S170, 20.85% fat, 0.15% cholesterol, 19.5% casein) from 6 weeks of age for at least 3 months. Test compound treatment is performed either orally via gavage to conscious animals before anesthesia or intravenously to anesthetized animals prior to injury.
  • Thrombus induction For oral treatment, animals are dosed by gavage in appropriate formulations by gavage prior to anesthesia.
  • a venous catheter will be implanted in the jugular vein for bolus or infusion drug delivery prior to the injury procedure.
  • Mice are anesthetized by intraperitoneal injection of Ketamin (65 mg/kg) and Xylazin (15 mg/kg) The cremaster muscle is exposed after opening the scrotum and removal of a testis with attention to preserve blood supply from the circulation of the mouse.
  • a stainless steel wire loop (Unimed 30.065, diameter 0.4 mm) is used to spread the inverted cremaster pocket in an organ bath constantly perfused with Dulbecco’s phosphate buffered saline temperated at 37°C.
  • the tissue preparation is positioned on top of a transparent macrolon plastic stage for intravital microscopy (Leica AS LMD, water immersion objective Leica 506155 HC APO/ L40x/0.80).
  • Video images are acquired by a digital camera (Basler PCO.EDGE 5.5) for identification of arterioles suitable for vascular wall injury (50 - 100 pm diameter, bifurcation-free segment).
  • a microdissection UV-Laser (wavelength 355 nm, Cell Surgeon, LLS Rowiak, Hannover, Germany) coupled into the optical path of the intravital microscope is used for 2-stage injury of the vessel wall: (1) Linear detachment of the endothelium along a length of approximately 150 pm. (2) Punctiform deep injury with local damage of the media. The resulting thrombus formation is visualized and recorded over a duration of 5 minutes. The lesion procedure will be repeated on an upstream segment of the identical vessel for at least two times.
  • Video sequences of thrombus development are quantified by morphometric analysis of images at 10-second intervals using a customized software for semi-automatic vessel and thrombus detection (Zeta, Fraunhofer-lnstitut fur Angewandte Informationstechnik FIT, St. Augustin, Germany). Thrombus area and degree of stenosis are calculated to generate a thrombus-over-time-curve. The main derived result is the time above 75% occlusion of the vessel lumen. The mean of at least three repeated injuries per animal will be calculated. In case of intravenous testing, the treatment effect will be compared to baseline control injuries in the identical animals (t-test for repeated measurements). In case of oral treatment, thrombus development will be compared to vehicle- treated animals (ANOVA and t-test for independent measurements). A minimum of 5 animals will be tested to obtain sufficient statistical power.

Abstract

The invention relates to substituted pyrazine carboxamide derivatives of formula (I) and to processes for their preparation, and also to their use for preparing medicaments for the treatment and/ or prophylaxis of diseases, in particular cardiovascular disorders, preferably thrombotic or thromboembolic disorders, and diabetes, and also urogenital and ophthalmic disorders.

Description

SUBSTITUTED PYRAZINE CARBOXAMIDE DERIVATIVES AS PROSTAGLANDIN EP3 RECEPTOR ANTAGONISTS
The invention relates to substituted pyrazine carboxamide derivatives and to processes for their preparation, and also to their use for preparing medicaments for the treatment and/or prophylaxis of diseases, in particular cardiovascular disorders, preferably thrombotic or thromboembolic disorders, and diabetes, and also urogenital and ophthalmic disorders.
Atherothrombosis is the main complication of atherosclerosis and underlies several of the most lethal human diseases, such as myocardial infarction (Ml), ischemic stroke (IS) and peripheral arterial occlusive disease (PAOD). The process is initiated by the deposition of lipids and their subsequent oxidation in the arterial wall which induces the recruitment of inflammatory cells and the formation of atherosclerotic plaques. These plaques are covered by a fibrous cap which maintains the plaque content separated from the blood flow. Inside the plaque, pro-inflammatory mechanisms drive inflammatory cells to produce matrix metalloproteases which digest the proteins of the fibrous cap. The thinned cap is called “vulnerable”, meaning that the cap may rupture relatively easily in response to stresses. When the cap ruptures or its endothelial cover erodes, the plaque content comes into contact with the blood and provokes the formation of an intravascular thrombus by activating platelets and blood coagulation. This process, forming a thrombus on plaques, is called atherothrombosis. If obstructive, the resulting intravascular thrombus interrupts blood flow and causes ischemia of downstream tissues with dramatic clinical consequences representing the leading cause of death and morbidity worldwide.
Given the mechanism of atherothrombotic vessel obstruction, current prevention targets the mechanisms of thrombosis, which is the pathological formation of intra-vascular plugs. The mechanisms of thrombosis encompass two intertwined pathways, the coagulation cascade and the aggregation of platelets, which once activated by a vessel injury act synergistically to build the intravascular clot obstructing the vessel lumen. Anticoagulant and anti-aggregant drugs, used to prevent atherothrombosis, have elicited a considerable reduction of the rate of second myocardial infarctions and a decrease of long term mortality from about 30% prior to the 1980s to less than 10% after the 2000s (Arch. Intern. Med. 2002, 162, 2405-2410; Lancet 2011, 377, 2193-2204). Specifically, the COX inhibitor Aspirin and the ADP receptor P2Y12 antagonist Clopidogrel are components of dual antiplatelet therapy. Prasugrel and Ticagrelor are alternative P2Y12 blockers that have demonstrated reductions of cardiovascular ischemic events ( N . Engl. J. Med. 2007, 357, 2001-2015; ibid. 2009, 361, 1045-1057). The coagulation factor Xa inhibitor Rivaroxaban has also shown benefits to patients with stable atherosclerotic vascular disease (N. Engl. J. Med. 2017, 377, 1319-1330). However, activation of platelets and blood coagulation is also crucial for hemostasis to prevent excessive blood loss after vascular injury. Consequently, currently marketed antiplatelet drugs and anticoagulants achieve their therapeutic effect at the cost of increased bleeding rates. Therefore, a safe antithrombotic drug must preserve the competence of hemostasis. An approach to widen the therapeutic window between antithrombotic-effective dose and the hemo stasis-compromising dose is to target mechanisms which are restricted to the site of atherosclerotic vessel walls. It has been widely shown that plaques host and maintain inflammation. A pro- thrombotic mediator produced by inflammatory mechanisms within plaques, but not by healthy vascular walls, would be an ideal target to impact atherothrombosis only at the site of the plaque without an impact on systemic hemostasis. Among other factors, local synthesis of prostanoids from arachidonic acid (AA) in the arterial vessel wall may play a profound role in atherosclerosis. Besides TXA2, the AA pathway generates several other mediators, e.g. prostaglandin E2 (PGE2). Increased PGE2 concentrations have been measured in atherosclerotic vascular walls of mice and humans [Circulation 2001 , 104, 921-927; J. Exp. Med. 2007, 204, 311-320; Cardiovasc. Res. 2014, 101, 482- 491] Once released upon plaque rupture, PGE2 binds on four specific receptors EP1, EP2, EP3 and EP4 on cell membranes. PGE2 has been shown to interfere with human and murine platelet function via EP3 and EP4 receptors ( Eur : J. Pharmacol. 1991, 194, 63-70). Stimulation of EP3 potentiates platelet activation and aggregation induced by primary agonists like collagen or ADP, whereas stimulation of EP4 inhibits platelet activation. This PGE2-dependent balance of platelet activation and inhibition can be tipped by modulation of EP3 or EP4 receptors ( Platelets 2010, 21, 329-342; Prostaglandins & Other Upid Mediators 2015, 121, 4-16).
Therefore, blocking the EP3 receptor by specific antagonists should be a beneficial strategy for prevention and treatment of atherothrombosis by local abrogation of platelet activation without altering hemostasis.
In patients suffering from PAOD, chronically inflamed vessel walls produce PGE2 to activate EP3 receptors not only on platelets but also on vascular smooth muscle cells thus preventing microvascular relaxation and contributing to malperfusion of peripheral tissues. Therefore, an antagonist to the EP3 receptor might be expected to provide therapeutic benefit specifically in PAOD.
Furthermore, PGE2 originating from inflammatory processes has been shown to compromise glucose-stimulated insulin secretion from pancreatic beta cells via the EP3 receptor pathway (Diabetes 2013, 62, 1904-1912). Therefore, EP3 antagonists might represent a beneficial strategy in the treatment of patients with type II diabetes.
In the kidney and urogenital tract, PGE2 participates in the regulation of renal microcirculation, diuresis and bladder excitability. EP3 receptor antagonists might help to improve renal disorders and in particular to resolve bladder hyperactivity, interstitial cystitis or bladder pain syndrome.
Furthermore, for many disorders the combination of antithrombotic and anti-inflammatory principles may also be particularly attractive to prevent the mutual enhancement of coagulation and platelet activation.
In the field of ophthalmology, prostaglandin derivatives participate in the regulation of intraocular pressure and inflammation. Therefore, compounds modulating the respective receptors may have a benefit in the prevention and treatment of ocular diseases.
Moreover, prostaglandin derivates play an essential role via their platelet-activating and pro- inflammatory mechanisms in inflammatory disorders like vasculitides, for example Kawasaki disease, Takayasu arteritis and Thrombangiitis obliterans (Buerger’s disease) as well as myocarditis (EMBO Mol Med 2018, e8536).
Furthermore, EP3 antagonists might contribute to the treatment and/or prophylaxis of diabetes- related end-organ manifestations like diabetic retinopathy and diabetic nephropathy.
Moreover, prostaglandins are involved in the pathogenesis of neurological disorders like neuropathic pain, Alzheimer’s disease and Parkinson’s disease.
In addition, PGE2 and EP3 have been reported to play a role in the pathogenesis of respiratory disorders like chronic cough, asthma and COPD (Am J Respir Crit Care 2009, 923-928).
W02009/082687 provides azaindolizines with anti-cancer activity. WO 2012/054233 provides imidazo pyrazine compounds for agronomic and nonagronomic uses. WO 2016/057522 provides triazolo pyridine compounds for the treatment of cystic fibrosis.
WO 2015/052610 and WO 2016/103097 provide antagonists of prostaglandin EP3 receptor having a pyridinone substituted indazole, indole or quinoline derivative. Amide or pyridinone based EP3 receptor antagonists are also described in ACS Med. Chem. Lett. 2010, 7, 316-320 and in Bioorg. Med. Chem. Lett. 2009, 19, 4292-4295, ibid. 2011, 27, 2806-2811, ibid. 2016, 26, 2670- 2675.
It is therefore an object of the present invention to provide novel compounds for the treatment of cardiovascular disorders, in particular of thrombotic or thromboembolic disorders, in humans and animals, which compounds have a wide therapeutic window and, in addition, a good pharmacokinetic behavior. Surprisingly, it has now been found that certain substituted pyrazine carboxamide derivatives represent highly potent antagonists of prostaglandin EP3 receptor.
The invention provides compounds of the formula (I)
Figure imgf000004_0001
in which G1 represents CR4 or N, G2 represents CH or N, with the proviso that G1 is N, if G2 is N,
R4 represents hydrogen, CrC4-alkyl, CrC4-halogenoalkyl, C3-C6-cycloalkyl or C1-C3- alkoxymethyl, R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, phenylsulfonyl, C3-C6- cycloalkylsulfonyl, 2,3-dihydro-1H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano, methoxy, methylsulfonyl, carbamoyl, NRaRb (where Ra and Rb are independently selected from the group consisting of hydrogen, CrC4-alkyl, C2-C6- halogenoalkyl and cyclopropyl) and CrC3-halogenoalkoxy, where halogenoalkoxy is substituted by 1 to 3 fluoro substituents, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxycarbonyl and NRcRd (where Rc and Rd are independently selected from the group consisting of hydrogen, CrC4-alkyl, C2-C6-halogenoalkyl and cyclopropyl), and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, ethyl, n-propyl, isopropyl, hydroxy, hydroxymethyl, trifluoromethyl and phenyl, or may be substituted by 1 or 2 fluoro, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro, chloro, cyano and methoxy, and
R5 represents hydrogen, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl or methoxycarbonyl, and where phenylsulfonyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro, chloro and cyano, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and L represents CrC6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, methoxycarbonyl, carboxyl, carbamoyl, amino, dimethylamino, te/f-butoxycarbonylamino, C3-C6-cycloalkyl (which may be substituted by 1 hydroxy), azetidin-1-yl (which may be substituted by 1 or 2 fluoro), pyrrolidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, pyrimidinyl, pyrazinyl, thienyl, pyrazolyl, oxazolyl, C3-C6- cycloalkyl, azetidinyl, pyrrolidinyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen, CrC4-alkyl, hydroxy, methoxy, trifluoromethyl, trifluoromethoxy, difluoromethoxy, dimethylaminomethyl, aminosulfonyl and pyrrolidin- 1-y I methyl, where pyridinyl may be substituted by 1 or 2 substituents independently selected from the group consisting of chloro, methyl, trifluoromethyl and methoxy, where pyrimidinyl may be substituted by 1 or 2 methyl substituents, where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro, CrC4-alkyl, trifluoromethyl, difluoromethyl, cyclopropyl and phenyl, where azetidinyl may be substituted by 1 substituent selected from the group consisting of fluoro, hydroxy, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl and methoxycarbonyl, and where pyrrolidinyl may be substituted by 1 substituent selected from the group consisting of fluoro, hydroxy, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl and methoxycarbonyl,
R2 represents a group of the formula
Figure imgf000006_0001
where # is the point of attachment to the pyrazine ring, Q1 represents CR8A or N, Q2 represents CR8 or N,
R6 represents hydrogen, halogen, CrC4-alkyl, CrC4-halogenoalkyl, CrC4-alkoxy, C1-C4- halogenoalkoxy or C3-C6-cycloalkyl,
R7 represents hydrogen, halogen, CrC4-alkyl, CrC4-halogenoalkyl, CrC4-alkoxy, C1-C4- halogenoalkoxy or C3-C6-cycloalkyl, with the proviso, that at least one of R6 and R7 is not hydrogen,
R7A represents hydrogen or halogen,
R8 represents hydrogen or halogen,
R8A represents hydrogen or halogen,
R9 represents hydrogen, halogen or CrC4-alkyl,
R3 represents hydrogen, halogen, cyano, CrC4-alkyl or CrC2-halogenoalkyl, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
The compounds according to the invention may be divided into three subgroups:
Figure imgf000007_0001
The term “substituted” means that one or more hydrogen atoms on the designated atom or group are replaced with a selection from the indicated group, provided that the designated atom’s normal valence under the existing circumstances is not exceeded. Combinations of substituents and/or variables are permissible.
As used herein, the term “one or more”, e.g. in the definition of the substituents of the compounds of general formula (I) of the present invention, means “1 , 2, 3, 4 or 5, particularly 1 , 2, 3 or 4, more particularly 1, 2 or 3, even more particularly 1 or 2”.
In the context of the present invention, unless specified otherwise, the substituents are defined as follows:
The term “halogen” or “halogeno” like in combinations e.g. in halogenoalkyl means a fluorine, chlorine, bromine or iodine atom, particularly a fluorine, chlorine or bromine atom, even more particularly fluorine or chlorine.
The term “Ci-C4-alkyl” and “Ci-Cyalkyl” means a linear or branched, saturated, monovalent hydrocarbon group having 1, 2, 3, or 4 carbon atoms, and 1, 2, 3, 4, 5, 6 or 7 carbon atoms, e.g. a methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert- butyl, pentyl, isopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-di ethylpropyl, neo-pentyl, 1,1-dimethylpropyl, hexyl, 1-methylpentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 1-ethylbutyl, 2-ethylbutyl,
1.1-dimethylbutyl, 2,2-dimethylbutyl, 3,3-di ethylbutyl, 2,3-di ethylbutyl, 1,2-di ethylbutyl or 1,3-dimethylbutyl group, or an isomer thereof. Particularly, said group has 1, 2, 3 or 4 carbon atoms (“Ci-C4-alkyl”), e.g. a methyl, ethyl, propyl, isopropyl, butyl, sec-butyl isobutyl, or tert- butyl group, more particularly 1, 2 or 3 carbon atoms (“Ci-C3-alkyl”), e.g. a methyl, ethyl, n-propyl or isopropyl group.
The term “Ci-C6-halogenoalkyl”, “C2-C6-halogenoalkyl”, “Ci-C4-halogenoalkyl”, “C2-C4- halogenoalkyl”, “Ci-C3-halogenoalkyl” and “Ci-C2-halogenoalkyl” represents a linear or branched, saturated, monovalent hydrocarbon group in which the term “alkyl” is as defined supra, and in which one or more of the hydrogen atoms are replaced, identically or differently, with a halogen atom. Particularly, said halogen atom is a fluorine atom. Said CrC6-halogenoalkyl group is, for example fluoromethyl, difluoromethyl, trifluoromethyl, 2-fluoroethyl, 2,2-difluoroethyl, 2,2, 2-trif luoroethy I , pentafluoroethyl, 3,3,3-trifluoropropan-1-yl, 1,1,1 -trif luoropropan-2-yl , 1,3-diflu- oropropan-2-yl, 3-fluoropropan-1-yl, 1,1,1 -trifluorobutan-2-yl, and 3,3,3-trifluoro-1-methyl-propan- 1-yl.
The term “Ci-C4-halogenoalkoxy” and “Ci-C3-halogenoalkoxy” represents a linear or branched, saturated, monovalent CrC4-alkoxy or CrC3-alkoxy group (where alkoxy represents a straight- chain or branched, saturated, monovalent alkoxy radical having 1 to 4 or 1 to 3 carbon atoms, by way of example and with preference methoxy, ethoxy, n-propoxy, isopropoxy), in which one or more of the hydrogen atoms is replaced, identically or differently, with a halogen atom. Particularly, said halogen atom is a fluorine atom. Said CrC3-halogenoalkoxy group is, for example, fluoromethoxy, difluoromethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy or pentafluoroethoxy.
The term “C3-C6-cycloalkyl” means a saturated, monovalent, monocyclic hydrocarbon ring which contains 3, 4, 5 or 6 carbon atoms. Said C3-C6-cycloalkyl group is for example a cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl group.
The term “Ci-C6-alkanediyl”, “Ci-C4-alkanediyl”, “C2-C5-alkanediyl” and “C2-C4-alkanediyl” represents a linear or branched, divalent alkyl radical having 1 to 6, 1 to 4, 2 to 5 or 2 to 4 carbon atoms, by way of example and with preference methylene (-CH2-), ethan-1 ,1-diyl [-CH(CH3)-], ethan-1,2-diyl [-(CH2)H, propan- 1,1-diyl [-CH(CH2CH3)-], propan-1, 2-diyl [-CH2CH(CH3)-], 2- methylpropan-1,1-diyl {-CH[CH(CH3)2]-}, 2-methylpropan-1,3-diyl {-CH2[CH(CH3)]CH2-}, butan-
1.1-diyl {-CH[(CH2)2CH3]-}, butan-1, 2-diyl [-CH2CH(CH2CH3)-], 3-methylbutan-1,1-diyl [- (CH2)2CH(CH3)2-] or 3-methylbutan-1, 2-diyl {-CH2CH[CH(CH3)2]-}.
Compounds according to the invention are the compounds of the formula (I) and the salts, solvates and solvates of the salts thereof, and also the compounds encompassed by formula (I) and specified hereinafter as working example(s), and the salts, solvates and solvates of the salts thereof, to the extent that the compounds encompassed by formula (I) and specified hereinafter are not already salts, solvates and solvates of the salts.
The inventive compounds may, depending on their structure, exist in different stereoisomeric forms, i.e. in the form of configurational isomers or else, if appropriate, of conformational isomers (enantiomers and/or diastereomers, including those in the case of rotamers and atropisomers). The present invention therefore encompasses the enantiomers and diastereomers, and the respective mixtures thereof. The stereoisomerically uniform constituents can be isolated from such mixtures of enantiomers and/or diastereomers in a known manner; chromatography processes are preferably used for this, especially HPLC chromatography on an achiral or chiral phase.
Further, it is possible for the compounds of the present invention to exist as tautomers. For example the compounds of formula (I) encompass the tautomer of formula (la)
Figure imgf000009_0001
The tautomers (l-Aa), (l-Ba) and (l-Ca) of the subgroups of formulas (l-A), (l-B) and (l-C) are shown hereafter:
Figure imgf000009_0002
Within this description the compounds according to the invention are drawn in the 4-oxo form.
The present invention includes all possible tautomers of the compounds of the present invention as single tautomers, or as any mixture of said tautomers, in any ratio. In the context of the present invention, the term “enantiomerically pure" is understood to mean that the compound in question with respect to the absolute configuration of the chiral centre is present in an enantiomeric excess of more than 95%, preferably more than 97%. The enantiomeric excess (ee value) is calculated in this case by evaluation of the corresponding HPLC chromatogram on a chiral phase with the aid of the formula below: ee = [EA (area%) - EB (area%)] x 100% / [EA(area%) + EB (area%)]
(EA: enantiomer in excess, EB: enantiomer in deficiency)
The present invention also encompasses all suitable isotopic variants of the compounds according to the invention. An isotopic variant of an inventive compound is understood here as meaning a compound in which at least one atom within the inventive compound 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, sulfur, fluorine, chlorine, bromine and iodine, such as 2H (deuterium), 3H (tritium), 13C, 14C, 15N, 170, 180, 32P, 33P, 33S, 34S, 35S, 36S, 18F, 36CI, 82Br, 123l, 124l, 129l and 1311. 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, may lead to particular therapeutic benefits as a consequence of greater metabolic stability of the compound, for example an extension of the half-life in the body or a reduction in the active dose required; such modifications of the inventive compounds 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 the processes known to those skilled in the art, for example by the methods described further below and the procedures described in the working examples, by using corresponding isotopic modifications of the respective reagents and/or starting compounds.
Preferred salts in the context of the present invention are physiologically acceptable salts of the compounds according to the invention. However, the invention also encompasses salts which themselves are unsuitable for pharmaceutical applications but which can be used, for example, for the isolation or purification 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 sulfonic acids, for example salts of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, benzenesulfonic acid, naphthalenedisulfonic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
Physiologically acceptable salts of the compounds according to the invention also include salts of 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) and ammonium salts derived from ammonia or organic amines having 1 to 16 carbon atoms, by way of example and with preference ethylamine, diethylamine, tri ethyl amine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, /V-methylmorpholine, arginine, lysine, ethylenediamine, N- methylpiperidine and choline.
The present invention includes all possible salts of the compounds according to the invention as single salts, or as any mixture of said salts, in any ratio.
Solvates in the context of the invention are described as those forms of the inventive compounds which form a complex in the solid or liquid state by coordination with solvent molecules. The compounds according to the invention may contain polar solvents, in particular water, methanol or ethanol for example, as structural element of the crystal lattice of the compounds. Hydrates are a specific form of the solvates in which the coordination is with water. It is possible for the amount of polar solvents, in particular water, to exist in a stoichiometric or non-stoichiometric ratio. In the case of stoichiometric solvates, e.g. a hydrate, hemi-, (semi-), mono-, sesqui-, di-, tri-, tetra-, penta- etc. solvates or hydrates, respectively, are possible. The present invention includes all such hydrates or solvates.
Further, the compounds according to the invention can exist as N-oxides, which are defined in that at least one nitrogen of the compounds of the present invention is oxidised in a known manner. The present invention includes all such possible N-oxides.
The present invention additionally also encompasses prodrugs of the inventive compounds. The term “prodrugs” encompasses compounds which for their part may be biologically active or inactive but are converted during their residence time in the body into compounds according to the invention (for example by metabolism or hydrolysis).
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" and "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 advancement 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. In the formulae of the group which may represent R2, the end point of the line marked by # in each case does not represent a carbon atom or a CH2 group, but is part of the bond to the atom to which R2is attached.
Preference is given to compounds of the formula (I) in which
G1 represents CR4 or N, G2 represents CH or N, with the proviso that G1 is N, if G2 is N,
R4 represents hydrogen, CrC4-halogenoalkyl or C3-C6-cycloalkyl,
R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, phenylsulfonyl, C3-C6- cycloalkylsulfonyl, 2,3-dihydro-1H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, ethyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
R5 represents CrC4-alkyl or C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, and where phenylsulfonyl may be substituted by 1 substituent fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, carboxyl, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro and CrC4-alkyl,
R2 represents a group of the formula
Figure imgf000013_0001
where # is the point of attachment to the pyrazine ring,
Q1 represents CR8A,
Q2 represents CR8,
R6 represents halogen or CrC4-alkyl,
R7 represents halogen, CrC4-alkyl or CrC4-alkoxy,
R7A represents hydrogen,
R8 represents hydrogen or halogen,
R8A represents hydrogen,
R9 represents hydrogen or halogen,
R3 represents hydrogen or halogen, and the salts thereof, the solvates thereof and the solvates of the salts thereof. Preference is also given to compounds of the formula (I) in which G1 represents CR4, G2 represents CH,
R4 represents hydrogen, CrC4-halogenoalkyl or C3-C6-cycloalkyl,
R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, phenylsulfonyl, C3-C6- cycloalkylsulfonyl or the group -L-RE, where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by NR5, and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
R5 represents CrC4-alkyl, and where phenylsulfonyl may be substituted by 1 substituent fluoro, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl or C3-C6-cycloalkyl, where phenyl may be substituted by 1 substituent halogen, and where pyridinyl may be substituted by 1 substituent methoxy,
R2 represents a group of the formula where # is the point of attachment to the pyrazine ring,
Q1 represents CR8A,
Q2 represents CR8,
R6 represents halogen or CrC4-alkyl,
R7 represents halogen, CrC4-alkyl or CrC4-alkoxy,
R7A represents hydrogen,
R8 represents hydrogen or fluoro,
R8A represents hydrogen,
R9 represents hydrogen or fluoro,
R3 represents hydrogen or fluoro, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
Preference is also given to compounds of the formula (I) in which G1 represents N,
G2 represents CH,
R1 represents CrCyalkyl, C3-C6-cycloalkyl, 2,3-dihydro-1 H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 substituent fluoro, and
R5 represents CrC4-alkyl or C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
L represents Ci-C4-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy and carboxyl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro and CrC4-alkyl,
R2 represents a group of the formula
Figure imgf000016_0001
where # is the point of attachment to the pyrazine ring,
Q1 represents CR8A,
Q2 represents CR8,
R6 represents halogen or CrC4-alkyl,
R7 represents halogen or CrC4-alkyl,
R7A represents hydrogen,
R8 represents hydrogen,
R8A represents hydrogen,
R9 represents hydrogen,
R3 represents hydrogen, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
Preference is also given to compounds of the formula (I) in which G1 represents N,
G2 represents N,
R1 represents the group -L-RE,
L represents C2-C4-alkanediyl, where alkanediyl may be substituted by up to 3 fluoro,
RE represents phenyl or pyridinyl, where phenyl may be substituted by 1 substituent of halogen, and where pyridinyl may be substituted by 1 substituent methoxy, R2 represents a group of the formula
Figure imgf000017_0001
where # is the point of attachment to the pyrazine ring,
R3 represents hydrogen, and the salts thereof, the solvates thereof and the solvates of the salts thereof Preference is also given to compounds of the formula (I) in which G1 represents CR4,
G2 represents CH,
R4 represents hydrogen, trifluoromethyl or cyclopropyl,
R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, phenylsulfonyl, cyclopropylsulfonyl or the group -L-RE, where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by NR5, and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 substituent selected from the group consisting of fluoro and methoxy, and
R5 represents methyl, and where phenylsulfonyl may be substituted by 1 substituent fluoro, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 substituent selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl or cyclopropyl, where phenyl may be substituted by 1 substituent fluoro, and where pyridinyl may be substituted by 1 substituent methoxy, R2 represents a group of the formula
Figure imgf000018_0001
where # is the point of attachment to the pyrazine ring, Q1 represents CR8A,
Q2 represents CR8,
R6 represents fluoro, chloro or methyl,
R7 represents chloro, methyl or methoxy,
R7A represents hydrogen,
R8 represents hydrogen or fluoro,
R8A represents hydrogen,
R9 represents hydrogen or fluoro,
R3 represents hydrogen or fluoro, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
Preference is also given to compounds of the formula (I) in which G1 represents N,
G2 represents CH, R1 represents Ci-Cralkyl, C3-C6-cycloalkyl, 2,3-dihydro-1H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 substituent fluoro, and
R5 represents methyl or C2-C3-halogenoalkyl substituted by 1 to 3 fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
L represents Ci-C4-alkanediyl, where alkanediyl may be substituted by 1 substituent selected from the group consisting of hydroxy, methoxy and carboxyl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro, bromo and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro, methyl and ethyl,
R2 represents a group of the formula where # is the point of attachment to the pyrazine ring,
Q1 represents CR8A,
Q2 represents CR8,
R6 represents chloro or methyl, R7 represents fluoro or methyl, R7A represents hydrogen,
R8 represents hydrogen,
R8A represents hydrogen,
R9 represents hydrogen,
R3 represents hydrogen, and the salts thereof, the solvates thereof and the solvates of the salts thereof.
Preference is also given to compounds of the formula (I) in which
R1 represents Ci-Cralkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, 2,3-dihydro-1H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano, methoxy, methylsulfonyl, carbamoyl, NRaRb (where Ra and Rb are independently selected from the group consisting of hydrogen, CrC4-alkyl, C2-C6- halogenoalkyl and cyclopropyl) and CrC3-halogenoalkoxy, where halogenoalkoxy is substituted by 1 to 3 fluoro substituents, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxycarbonyl and NRcRd (where Rc and Rd are independently selected from the group consisting of hydrogen, CrC4-alkyl, C2-C6-halogenoalkyl and cyclopropyl), and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, ethyl, n-propyl, isopropyl, hydroxy, hydroxymethyl, trifluoromethyl and phenyl, or may be substituted by 1 or 2 fluoro, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro, chloro, cyano and methoxy, and
R5 represents hydrogen, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl or methoxycarbonyl, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, methoxycarbonyl, carboxyl, carbamoyl, amino, dimethylamino, te/f-butoxycarbonylamino, C3-C6-cycloalkyl (which may be substituted by 1 hydroxy), azetidin-1-yl (which may be substituted by 1 or 2 fluoro), pyrrolidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, pyrimidinyl, pyrazinyl, thienyl, pyrazolyl, oxazolyl, C3-C6- cycloalkyl, azetidinyl, pyrrolidinyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen, CrC4-alkyl, hydroxy, methoxy, trifluoromethyl, trifluoromethoxy, difluoromethoxy, dimethylaminomethyl, aminosulfonyl and pyrrolidin- 1-y I methyl, where pyridinyl may be substituted by 1 or 2 substituents independently selected from the group consisting of chloro, methyl, trifluoromethyl and methoxy, where pyrimidinyl may be substituted by 1 or 2 methyl substituents, where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro, Ci-C4-alkyl, trifluoromethyl, difluoromethyl, cyclopropyl and phenyl, where azetidinyl may be substituted by 1 substituent selected from the group consisting of fluoro, hydroxy, Ci-C4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl and methoxycarbonyl, and where pyrrolidinyl may be substituted by 1 substituent selected from the group consisting of fluoro, hydroxy, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl and methoxycarbonyl.
Preference is also given to compounds of the formula (I) in which R1 represents Ci-Cralkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, 2,3-dihydro-1H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, ethyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
R5 represents CrC4-alkyl or C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, carboxyl, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro and CrC4-alkyl.
Preference is also given to compounds of the formula (I) in which
R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl or the group -L-RE, where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by NR5, and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
R5 represents CrC4-alkyl, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl or C3-C6-cycloalkyl, where phenyl may be substituted by 1 substituent halogen, and where pyridinyl may be substituted by 1 substituent methoxy.
Preference is also given to compounds of the formula (I) in which R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl or the group -L-RE, where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by NR5, and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 substituent selected from the group consisting of fluoro and methoxy, and
R5 represents methyl, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 substituent selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl or cyclopropyl, where phenyl may be substituted by 1 substituent fluoro, and where pyridinyl may be substituted by 1 substituent methoxy.
Preference is also given to compounds of the formula (I) in which R2 represents 4-chloro-3- fluorophenyl, 3-chloro-4-methylphenyl, 4-chloro-3-methylphenyl, 3,4-dimethylphenyl, 3-fluoro-4- methylphenyl, 2-napthyl, 2,3-dihydro-1,4-benzodioxine or 5-fluoro-2,3-dihydro-1,4-benzodioxine. Preference is also given to compounds of the formula (I) in which R2 represents 3,4-dimethylphenyl, 2-napthyl or 2,3-dihydro-1 ,4-benzodioxine.
Preference is also given to compounds of the formula (I) in which R3 represents hydrogen or fluoro. Preference is also given to compounds of the formula (I) in which R3 represents hydrogen.
Also preferred are compounds having the formula (l-X)
Figure imgf000024_0001
in which G1, G2, R2 and R3 are as defined above, Rx represents CrCs-alkyl, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, methoxycarbonyl, carboxyl, carbamoyl, amino, dimethylamino, fe/f-butoxycarbonylamino, C3-C6-cycloalkyl (which may be substituted by 1 hydroxy), azetidin- 1-yl (which may be substituted by 1 or 2 fluoro), pyrrolidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro, and
Ry represents hydrogen, halogen, CrC4-alkyl, hydroxy, methoxy, trifluoromethyl, trifluoromethoxy, difluoromethoxy, dimethylaminomethyl, aminosulfonyl or pyrrolidin-1-ylmethyl. The compounds of the the formula (l-X) are a subgroup of the compounds of the formula (I).
The invention further provides a process for preparing compounds of the formula (I), or salts thereof, solvates thereof or solvates of the salts thereof, wherein
[A] the compounds of the formula (II)
R1/N H2
(II), in which
R1 is as defined above, are reacted with compounds of the formula (III)
Figure imgf000025_0001
in which G1, G2, R2 and R3 are as defined above, in the presence of a dehydrating agent to give compounds of the formula (I).
The reaction [A] is generally carried out in inert solvents, if appropriate in the presence of a base, preferably in a temperature range from 0°C to 50°C at atmospheric pressure.
Alternatively, the reaction [A] can also be carried out without a solvent only in one base if the base is a liquid at room temperature.
Suitable dehydrating agents here are, for example, carbodiimides such as N,N ’-diethyl-, N,N’- dipro- pyl- N,N ’-diisopropyl-, A/,/\/-dicydohexylcarbodiimide, A/-(3-dimethylaminoisopropyl)-/\/'-ethylcarbo- diimide hydrochloride (EDCI) (optionally in the presence of pentafluorophenol (PFP)), /V-cyclohexyl- carbodiimide-/V-propyloxymethyl-polystyrene (PS-carbodiimide) or carbonyl compounds such as carbonyldiimidazole (CDI), or 1 ,2-oxazolium compounds such as 2-ethyl-5-phenyl-1,2-oxazolium 3- sulphate or 2-terf-butyl-5-methyl-isoxazolium perchlorate, or acylamino compounds such as 2-eth- oxy-1-ethoxycarbonyl-1,2-dihydroquinoline, or propanephosphonic anhydride, or isobutyl chloro- formate, or bis-(2-oxo-3-oxazolidinyl)phosphoryl chloride or benzotriazolyloxytri(dimethylamino)- phosphonium hexafluorophosphate, or 0-(benzotriazol-1-yl)-/V,/V,/V',/V'-tetramethyluronium hexa- fluorophosphate (HBTU), 2-(2-oxo-1-(2H)-pyridyl)-1,1,3,3-tetramethyluronium tetrafluoroborate (TPTU), (benzotriazol-l-yloxy)bisdimethylaminomethylium fluoroborate (TBTU) or 0-(7-azabenzo- triazol-1-yl)-/V,/V,/V',/V'-tetramethyluronium hexafluoro-phosphate (HATU), or 1-hydroxybenzotriazole (HOBt), or benzotriazol-1-yloxytris(dimethyl-amino)phosphonium hexafluorophosphate (BOP), or ethyl cyano(hydroxyimino)acetate (Oxyma), or (1-cyano-2-ethoxy-2-oxoethylidenaminooxy)- dimethylamino-morpholino-carbenium hexafluorophosphate (COMU), or N-[(dimethylamino)(3/-/- [1,2,3]triazolo[4,5-b]pyridin-3-yloxy)methylidene]-N-methylmethanaminium hexafluorophosphate, or 2,4,6-tripropyl-1,3,5,2,4,6-trioxatriphosphinane-2,4,6-trioxide (T3P), or mixtures of these with bases, the condensation with HATU, EDCI, CDI or T3P being preferred.
Bases are, for example, alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate, or organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine; preference is given to condensation with diisopropylethylamine, 4-dimethylaminopyridine, pyridine or /V-methylmorpholine.
Inert solvents are, for example, halogenated hydrocarbons such as dichloromethane or trichloromethane, hydrocarbons such as benzene or toluene, or other solvents such as nitromethane, dioxane, diethyl ether, tetrahydrofuran, ethyl acetate, dimethylformamide, dimethylacetamide, dimethyl sulphoxide, /V-methylpyrrolidone or acetonitrile, or mixtures of the solvents; preference is given to dimethylformamide, ethyl acetate, /V-methylpyrrolidone or dichloromethane.
The compounds of the formula (II) are known or can be synthesized from the corresponding starting compounds by known processes.
The compounds of the formula (III) are known or can be prepared
[B] by reacting compounds of the formula (IV)
Figure imgf000026_0001
in which
G1, G2, R2 and R3 are as defined above, and
R13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with a base. The reaction [B] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
Bases are, for example, alkali metal hydroxides such as sodium hydroxide, lithium hydroxide or potassium hydroxide, or alkali metal carbonates such as caesium carbonate, sodium carbonate or potassium carbonate, or alkoxides such as potassium tert- butoxide or sodium tert- butoxide; preference is given to sodium hydroxide or lithium hydroxide.
Inert solvents are, for example, alcohols such as methanol or ethanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, dimethylacetamide, acetonitrile or pyridine, or mixtures of solvents, or mixtures of solvents with water; preference is given to a mixture of tetrahydrofuran and water or ethanol and water or methanol and water or tetrahydrofuran, methanol and water.
The compounds of the formula (IV) are known or can be prepared
[C] by reacting compounds of the formula (V)
Figure imgf000027_0001
in which G1, G2, R2 and R3 are each as defined above,
R13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
R14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with an ammonia equivalent in the presence of an acid. The reaction [C] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
Alternatively, the reaction [C] can also be carried out without a solvent only in acid if the acid is a liquid at room temperature. Ammonia equivalents are, for example, ammonium acetate, ammonium formate, ammonium propionate, or ammonium chloride; preference is given to ammonium acetate.
Acids are, for example, organic acids such was formic acid, acetic acid, propionic acid, trifluoroacetic acid, trifluoromethanesulfonic acid, or mineral acids such as, for example, hydrogen chloride, or hydrogen bromide; preference is given to acetic acid.
The compounds of the formula (V) are known or can be prepared
[D] by reacting compounds of the formula (VI)
Figure imgf000028_0001
in which
G1 and G2 are each as defined above,
R13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
R14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with compounds of the formula (VII)
(VII),
Figure imgf000028_0002
in which
R2 and R3 are as defined above, and X1 represents chloro, bromo, iodo Oder triflate, in the presence of a base. The reaction [D] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
Bases are, for example, alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate, or organic bases such as trialkylamines, for example triethylamine, N-methylmorpholine, N-methylpiperidine, 4- dimethylaminopyridine, diisopropylethylamine or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to potassium carbonate. Inert solvents are, for example, halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachloride or 1,2-dichloroethane, alcohols such as methanol or ethanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, N-methyl-pyrrolidone, dimethylacetamide, acetonitrile, acetone or pyridine, or mixtures of solvents, or mixtures of solvents with water; preference is given to acetone.
The compounds of the formula (VII) are known or can be synthesized from the corresponding starting compounds by known processes.
The compounds of the formula (VI) may be divided into three subgroups ((Vl-A), (Vl-B) and (VI-C)):
Figure imgf000029_0001
(Vl-A) (VI- B) (VI-C)
The compounds of the formula (Vl-A) are known or can be prepared
[E] by reacting compounds of the formula (VIII)
Figure imgf000029_0002
in which
R4 is as defined above, and
R13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with compounds of the formula (IX)
Figure imgf000029_0003
(IX), in which
R14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, in the presence of a copper(ll) sorce and a ligand.
The reaction [E] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure. Copper(ll) sorces are, for example, copper(ll)chloride, copper(ll)bromide, copper(ll)iodide or copper(ll)oxide; preference is given to copper(ll)oxide.
Ligands are, for example, 1,10-phenanthroline, 3,4,7,8-tetramethyl-1,10-phenanthroline or pathophenanthroline; preference is given to 1,10-phenanthroline. Inert solvents are, for example, ethers such as diethyl ether, methyl tert- butyl ether, 1,2- dimethoxyethane, 1,4-dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, /V-methyl-pyrrolidone, dimethylacetamide or acetonitrile; preference is given to 1,4-dioxane.
The compounds of the formula (VIII) and of the formula (IX) are known or can be synthesized from the corresponding starting compounds by known processes. The compounds of the formula (Vl-A) are known or can also be prepared
[F] by reacting compounds of the formula (X)
Figure imgf000030_0001
(X), in which
R4 is as defined above, R13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
R14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with a base.
The reaction [F] is generally carried out in inert solvents, preferably in a temperature range from 0°C up to reflux of the solvents at atmospheric pressure.
Bases are, for example, alkali metal te/f-butoxides, such as sodium te/f-butoxide or potassium tert- butoxide or alkali metal hydrides such as sodium hydride or potassium hydride, or alkali metal amides such as lithium diisopropylamide or potassium bis(trimethylsilyl)amide; preference is given to potassium ferf-butoxide. Inert solvents are, for example, alcohols such as methanol, ethanol, iso-propanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as toluene, dimethylformamide, N-methyl-pyrrolidone or dimethylacetamide, or mixtures of solvents; preference is given to a mixture of ethanol, toluene and tetrahydrofuran.
The compounds of the formula (X) are known or can be prepared [G] by reacting compounds of the formula (XI)
Figure imgf000031_0001
in which
R4 is as defined above,
R13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
R15 represents methyl or ethyl, with a compound of the formula (XII)
Figure imgf000031_0002
in which
R14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl.
The reaction [G] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
Inert solvents are, for example, alcohols such as methanol, ethanol, iso-propanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, N-methyl-pyrrolidone, dimethylacetamide or acetonitrile; preference is given to ethanol.
The compounds of the formula (XII) are known or can be synthesized from the corresponding starting compounds by known processes. The compounds of the formula (XI) are known or can be prepared
[H] by reacting compounds of the formula (XIII)
Figure imgf000031_0003
(XIII), in which R4 is as defined above, with compounds of the formula (XIV)
Figure imgf000032_0001
(XIV), in which
R13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, and
R15 represents methyl or ethyl, with pivaloyl chloride and a base.
The reaction [H] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
Bases are, for example, organic bases such as trialkylamines, for example triethylamine, N- methylmorpholine, /V-methylpiperidine, 4-dimethylaminopyridine or diisopropylethylamin, or pyridine; preference is given to triethylamine.
Inert solvents are, for example, ethers such as diethyl ether, methyl tert- butyl ether, 1,2- dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as toluene, dimethylformamide, N-methyl-pyrrolidone or dimethylacetamide; preference is given to toluene.
The compounds of the formula (XIII) and of the formula (XIV) are known or can be synthesized from the corresponding starting compounds by known processes.
The compounds of the formula (Vl-B) are known or can be prepared [I] by reacting compounds of the formula (XV)
Figure imgf000032_0002
in which
R13 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with compounds of the formula (XVI) in which
R14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, in the presence of a base. The reaction [I] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
Bases are, for example, alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate, or organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to triethylamine.
Inert solvents are, for example, aromatic solvents such as toluene, xylene, chlorobenzene or 1,2- dichlorobenzene, alcohols such as methanol or ethanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, dimethylacetamide, /V-methyl-pyrrolidone, acetonitrile, acetone or pyridine, or mixtures of the solvents; preference is given to xylene.
The compounds of the formula (XV) are known or can be synthesized from the corresponding starting compounds by known processes.
The compounds of the formula (XVI) are known or can be prepared [J] by reacting compounds of the formula (XVII)
Figure imgf000033_0001
(XVII), in which
R14 represents methyl, ethyl, propyl, isopropyl, tert- butyl or benzyl, with hydroxylamine hydrochloride in the presence of a base. The reaction [J] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure.
Bases are, for example, alkali metal carbonates such as sodium carbonate or potassium carbonate, or sodium bicarbonate or potassium bicarbonate, or organic bases such as trialkylamines, for example triethylamine, /V-methylmorpholine, /V-methylpiperidine, 4-dimethyl- aminopyridine, diisopropylethylamin or pyridine, or other bases such as sodium hydride or lithium diisopropylamide; preference is given to sodium carbonate. Inert solvents are, for example, alcohols such as methanol or ethanol, ethers such as diethyl ether, methyl tert- butyl ether, 1,2-dimethoxyethane, dioxane or tetrahydrofuran, or other solvents such as dimethylformamide, dimethylacetamide, /V-methyl-pyrrolidone, acetonitrile, acetone or pyridine, or mixtures of the solvents; preference is given to ethanol.
The compounds of the formula (XVII) are known or can be synthesized from the corresponding starting compounds by known processes.
The compounds of the formula (Vl-C) are known or can be synthesized from the corresponding starting compounds by known processes.
The compounds of the formula (l-A-l)
Figure imgf000034_0001
(l-A-l), in which
R1, R2 and R4 are as defined above, are known or can be prepared
[K] by reacting compounds of formular (l-A)
Figure imgf000034_0002
in which
R1, R2 and R4 are as defined above, and
R3 represents hydrogen with an electrophilic fluorine source and a base, followed by a dehydrating agent. The compounds of the the formula (l-A) and the formula (l-A-l) are subgroups of the compounds of the formula (I).
The reaction [K] is generally carried out in inert solvents, preferably in a temperature range from room temperature up to reflux of the solvents at atmospheric pressure. Suitable electrophilic fluorination reagents are, for example, Selectfluor®, Selectfluor® II, N- fluorobenzenesulfonimide, 2,6-dichloro-1-fluoropyridinium tetrafluoroborate, 2,6-dichloro-1- fluoropyridinium triflate; preference is given to Selectfluor®.
Bases are, for example, organic bases such as trialkylamines, for example triethylamine, N- methylmorpholine, /V-methylpiperidine, 4-dimethylaminopyridine or A/./V-diisopropylethylamine, or pyridine; preference is given to pyridine.
Inert solvents are, for example, ethers such as dioxane, diethyl ether, tetrahydrofuran, N,N- dimethylformamide, A/,/\/-dimethylacetamide, dimethyl sulphoxide, /V-methylpyrrolidone or acetonitrile, or mixtures of the solvents; preference is given to acetonitrile.
Suitable dehydrating agents are, for example, 2,4,6-tripropyl-1,3,5,2,4,6-trioxatriphosphinane- 2,4,6-trioxide (T3P); preference is given to 2, 4, 6-tripropyl-1, 3, 5,2,4, 6-trioxatriphosphinane-2, 4,6- trioxide.
The preparation of the starting compounds and of the compounds of the formula (I) can be illustrated by the synthesis schemes which follow.
Scheme 1
Figure imgf000035_0001
Scheme 2
Figure imgf000036_0002
Scheme 4
Figure imgf000036_0001
The compounds of 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 have an unforeseeable useful pharmacological activity spectrum and good pharmacokinetic behavior, in particular a sufficient exposure of such a compound in the blood above the minimal effective concentration within a given dosing interval after oral administration. Such a profile results in an improved peak-to-trough ratio (quotient of maximum to minimum concentration) within a given dosing interval, which has the advantage that the compound can be administered less frequently and at a significantly lower dose to achieve an effect. They are compounds that inhibit the activation of the EP3 receptor by its ligand Prostaglandin E2 (PGE2).
They are therefore suitable for use as medicaments for the treatment and/or prophylaxis of diseases in humans and animals.
The present invention further provides for the use of the compounds according to the invention for the treatment and/or prophylaxis of disorders, in particular cardiovascular disorders, preferably thrombotic or thromboembolic disorders and/or thrombotic or thromboembolic complications such as acute coronary syndrome or myocardial infarction or ischemic stroke or peripheral arterial occlusive disease , and/or diabetes, and/or ophthalmic disorders and/or urogenital disorders, in particular those associated with excess PGE2.
Increased PGE2 concentrations have been measured in atherosclerotic vascular walls of mice and humans. Once released upon plaque rupture, PGE2 binds on four specific receptors EP1 , EP2, EP3 and EP4 on cell membranes. PGE2 has been shown to interfere with human and murine platelet function via EP3 and EP4 receptors. Stimulation of EP3 potentiates platelet activation and aggregation induced by primary agonists like collagen or ADP, whereas stimulation of EP4 inhibits platelet activation. This PGE2-dependent balance of platelet activation and inhibition can be tipped by modulation of EP3 or EP4 receptors.
In contrast to established platelet antagonists, for example COX inhibitors like acetyl salicylic acid (Aspirin®) and P2Y12 receptor antagonists like clopidogrel, prasugrel or ticagrelor, modulation of platelet activity via the EP3 receptor does not interfere with physiologic hemostasis and therefore is not expected to induce or increase the risk of bleeding.
Therefore, blocking the EP3 receptor by specific antagonists should be a beneficial strategy for prevention and treatment of atherothrombosis by local abrogation of platelet activation without altering hemostasis.
In patients suffering from PAOD, chronically inflamed vessel walls produce PGE2 to activate EP3 receptors not only on platelets but also on vascular smooth muscle cells thus preventing microvascular relaxation and contributing to malperfusion of peripheral tissues. Therefore, an antagonist to the EP3 receptor might be expected to provide therapeutic benefit specifically in PAOD.
For the purpose of the present invention, the "thrombotic or thromboembolic disorders" include disorders which occur preferably in the arterial vasculature and which can be treated with the compounds according to the invention, in particular disorders leading to peripheral arterial occlusive disorders and in the coronary arteries of the heart, such as acute coronary syndrome (ACS), myocardial infarction with ST segment elevation (STEMI) and without ST segment elevation (non-STEMI), stable angina pectoris, unstable angina pectoris, reocclusions and restenoses after coronary interventions such as angioplasty, stent implantation or aortocoronary bypass, but also thrombotic or thromboembolic disorders in cerebrovascular arteries, such as transitory ischaemic attacks (TIA), ischemic strokes including cardioembolic strokes, such as strokes due to atrial fibrillation, non-cardioembolic strokes, such as lacunar stroke, strokes due to large or small artery diseases, or strokes due to undetermined cause, cryptogenic strokes, embolic strokes, embolic strokes of undetermined source, or events of thrombotic and/or thromboembolic origin leading to stroke or TIA.
Moreover, the compounds according to the invention are suitable in particular for the treatment and/or prophylaxis of disorders where, the pro-inflammatory component plays an essential role, including vasculitides like Kawasaki disease, Takayasu arteritis and Thrombangiitis obliterans (Buerger’s disease) as well as inflammatory disorders like myocarditis.
Furthermore, the compounds according to the invention are suitable for the treatment and/or prophylaxis of disorders of the urogenital tract like overactive bladder, interstitial cystitis and bladder pain syndrome.
Moreover, the compounds according to the invention are suitable for the treatment and/or prophylaxis of diabetes mellitus including its end-organ manifestations like diabetic retinopathy and diabetic nephropathy.
Furthermore, the compounds according to the invention are suitable in particular for the treatment and/or prophylaxis of neurological disorders like neuropathic pain, Alzheimer’s disease and Parkinson’s disease. Moreover, the compounds according to the invention are suitable in particular for the treatment and/or prophylaxis of pulmonologic disorders like chronic cough, asthma and COPD.
The present invention further provides for the use of the compounds according to the invention for the treatment and/or prophylaxis of disorders, especially the disorders mentioned above.
The present invention further provides for the use of the compounds according to the invention for production of a medicament for the treatment and/or prophylaxis of disorders, especially the disorders mentioned above.
The present invention further provides a method for the treatment and/or prophylaxis of disorders, especially the disorders mentioned above, using a therapeutically effective amount of a compound according to the invention. The present invention further provides the compounds according to the invention for use in a method for the treatment and/or prophylaxis of disorders, especially the disorders mentioned above, using a therapeutically effective amount of a compound according to the invention.
Particularly the present invention provides the compounds according to the invention for use in a method for the treatment and/or prophylaxis of thrombotic or thromboembolic, in particular atherothrombotic disorders using a therapeutically effective amount of a compound according to the invention.
The present invention further provides medicaments comprising a compound according to the invention and one or more further active compounds.
In addition, the compounds according to the invention can also be used for preventing coagulation ex vivo, for example for the protection of organs to be transplanted against organ damage caused by formation of clots and for protecting the organ recipient against thromboemboli from the transplanted organ, for preserving blood and plasma products, for cleaning/pretreating catheters and other medical auxiliaries and instruments, for coating synthetic surfaces of medical auxiliaries and instruments used in vivo or ex vivo or for biological samples which may comprise factor Xla or plasma kallikrein.
The present invention furthermore provides a method for preventing the coagulation of blood in vitro, in particular in banked blood or biological samples which may comprise factor Xla or plasma kallikrein or both enzymes, which method is characterized in that an anticoagulatory effective amount of the compound according to the invention is added.
The compounds of the invention can 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.
For these administration routes, it is possible for the compounds according to the invention to be administered in suitable administration forms.
For oral administration, it is possible to formulate the compounds according to the invention to dosage forms known in the art that deliver the compounds of the invention rapidly and/or in a modified manner, such as, for example, tablets (uncoated or coated tablets, for example with enteric or controlled release coatings that dissolve with a delay or are insoluble), orally- disintegrating tablets, films/wafers, films/lyophylisates, capsules (for example hard or soft gelatine capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions. It is possible to incorporate the compounds according to the invention in crystalline and/or amorphised and/or dissolved form into said dosage forms.
Parenteral administration can be effected with avoidance of an absorption step (for example intravenous, intraarterial, intracardial, intraspinal or intralumbal) or with inclusion of absorption (for example intramuscular, subcutaneous, intracutaneous, percutaneous or intraperitoneal). Administration forms which are suitable for parenteral administration are, inter alia, preparations for injection and infusion in the form of solutions, suspensions, emulsions, lyophylisates or sterile powders.
Suitable for extraocular (topic) administration are administration forms which operate in accordance with the prior art, which release the active compound rapidly and/or in a modified or controlled manner and which contain the active compound in crystalline and/or amorphized and/or dissolved form such as, for example, eye drops, sprays and lotions (e.g. solutions, suspensions, vesicular/colloidal systems, emulsions, aerosols), powders for eye drops, sprays and lotions (e.g. ground active compound, mixtures, lyophilisates, precipitated active compound), semisolid eye preparations (e.g. hydrogels, in-situ hydrogels, creams and ointments), eye inserts (solid and semisolid preparations, e.g. bioadhesives, films/wafers, tablets, contact lenses).
Intraocular administration includes, for example, intravitreal, subretinal, subscleral, intrachoroidal, subconjunctival, retrobulbar and subtenon administration. Suitable for intraocular administration are administration forms which operate in accordance with the prior art, which release the active compound rapidly and/or in a modified or controlled manner and which contain the active compound in crystalline and/or amorphized and/or dissolved form such as, for example, preparations for injection and concentrates for preparations for injection (e.g. solutions, suspensions, vesicular/colloidal systems, emulsions), powders for preparations for injection (e.g. ground active compound, mixtures, lyophilisates, precipitated active compound), gels for preparations for injection (semisolid preparations, e.g. hydrogels, in-situ hydrogels) and implants (solid preparations, e.g. biodegradable and nonbiodegradable implants, implantable pumps).
Preference is given to oral administration or, in the case of ophthalmologic disorders, extraocular and intraocular administration.
Examples which are suitable for other administration routes are pharmaceutical forms for inhalation [inter alia powder inhalers, nebulizers], nasal drops, nasal solutions, nasal sprays; tablets/films/wafers/capsules for lingual, sublingual or buccal administration; suppositories; eye drops, eye ointments, eye baths, ocular inserts, ear drops, ear sprays, ear powders, ear-rinses, ear tampons; vaginal capsules, aqueous suspensions (lotions, mixturae agitandae), lipophilic suspensions, emulsions, ointments, creams, transdermal therapeutic systems (such as, for example, patches), milk, pastes, foams, dusting powders, implants or stents.
The compounds according to the invention can be incorporated into the stated administration forms. This can be effected in a manner known per se by mixing with pharmaceutically suitable excipients. Pharmaceutically suitable excipients include, inter alia,
• fillers and carriers (for example cellulose, microcrystalline cellulose (such as, for example, Avicel®), lactose, mannitol, starch, calcium phosphate (such as, for example, Di-Cafos®)),
• ointment bases (for example petroleum jelly, paraffins, triglycerides, waxes, wool wax, wool wax alcohols, lanolin, hydrophilic ointment, polyethylene glycols),
• bases for suppositories (for example polyethylene glycols, cacao butter, hard fat),
• solvents (for example water, ethanol, isopropanol, glycerol, propylene glycol, medium chain- length triglycerides fatty oils, liquid polyethylene glycols, paraffins),
• surfactants, emulsifiers, dispersants or wetters (for example sodium dodecyl sulfate), lecithin, phospholipids, fatty alcohols (such as, for example, Lanette®), sorbitan fatty acid esters (such as, for example, Span®), polyoxyethylene sorbitan fatty acid esters (such as, for example, Tween®), polyoxyethylene fatty acid glycerides (such as, for example, Cremophor®), polyoxethylene fatty acid esters, polyoxyethylene fatty alcohol ethers, glycerol fatty acid esters, poloxamers (such as, for example, Pluronic®),
• buffers, acids and bases (for example phosphates, carbonates, citric acid, acetic acid, hydrochloric acid, sodium hydroxide solution, ammonium carbonate, trometamol, triethanolamine),
• isotonicity agents (for example glucose, sodium chloride),
• adsorbents (for example highly-disperse silicas),
• viscosity-increasing agents, gel formers, thickeners and/or binders (for example polyvinylpyrrolidone, methylcellulose, hydroxypropylmethylcellulose, hydroxypropylcellulose, carboxymethylcellulose-sodium, starch, carbomers, polyacrylic acids (such as, for example, Carbopol®); alginates, gelatine),
• disintegrants (for example modified starch, carboxymethylcellulose-sodium, sodium starch glycolate (such as, for example, Explotab®), cross- linked polyvinylpyrrolidone, croscarmellose-sodium (such as, for example, AcDiSol®)),
• flow regulators, lubricants, glidants and mould release agents (for example magnesium stearate, stearic acid, talc, highly-disperse silicas (such as, for example, Aerosil®)),
• coating materials (for example sugar, shellac) and film formers for films or diffusion membranes which dissolve rapidly or in a modified manner (for example polyvinylpyrrolidones (such as, for example, Kollidon®), polyvinyl alcohol, hydroxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, hydroxypropylmethylcellulose phthalate, cellulose acetate, cellulose acetate phthalate, polyacrylates, polymethacrylates such as, for example, Eudragit®)),
• capsule materials (for example gelatine, hydroxypropylmethylcellulose), • synthetic polymers (for example polylactides, polyglycolides, polyacrylates, polymethacrylates (such as, for example, Eudragit®), polyvinylpyrrolidones (such as, for example, Kollidon®), polyvinyl alcohols, polyvinyl acetates, polyethylene oxides, polyethylene glycols and their copolymers and blockcopolymers),
• plasticizers (for example polyethylene glycols, propylene glycol, glycerol, triacetine, triacetyl citrate, dibutyl phthalate),
• penetration enhancers,
• stabilisers (for example antioxidants such as, for example, ascorbic acid, ascorbyl palmitate, sodium ascorbate, butylhydroxyanisole, butylhydroxytoluene, propyl gallate),
• preservatives (for example parabens, sorbic acid, thiomersal, benzalkonium chloride, chlorhexidine acetate, sodium benzoate),
• colourants (for example inorganic pigments such as, for example, iron oxides, titanium dioxide),
• flavourings, sweeteners, flavour- and/or odour-masking agents.
The present invention furthermore relates to a pharmaceutical composition which comprises at least one compound according to the invention, conventionally together with one or more pharmaceutically suitable excipient(s), and to their use according to the present invention.
An embodiment of the invention are pharmaceutical compositions comprising at least one compound of formula (I) according to the invention, preferably together with at least one inert, non toxic, pharmaceutically suitable auxiliary, and the use of these pharmaceutical compositions for the above cited purposes.
In accordance with another aspect, the present invention covers pharmaceutical combinations, in particular medicaments, comprising at least one compound of general formula (I) of the present invention and at least one or more further active ingredients, in particular for the treatment and/or prophylaxis of cardiovascular disorders, preferably thrombotic or thromboembolic disorders, and diabetes, and also urogenital and ophthalmic disorders.
The term “combination” in the present invention is used as known to persons skilled in the art, it being possible for said combination to be a fixed combination, a non-fixed combination or a kit-of- parts.
A “fixed combination” in the present invention is used as known to persons skilled in the art and is defined as a combination wherein, for example, a first active ingredient, such as one or more compounds of general formula (I) of the present invention, and a further active ingredient are present together in one unit dosage or in one single entity. One example of a “fixed combination” is a pharmaceutical composition wherein a first active ingredient and a further active ingredient are present in admixture for simultaneous administration, such as in a formulation. Another example of a “fixed combination” is a pharmaceutical combination wherein a first active ingredient and a further active ingredient are present in one unit without being in admixture.
A non-fixed combination or “kit-of-parts” in the present invention is used as known to persons skilled in the art and is defined as a combination wherein a first active ingredient and a further active ingredient are present in more than one unit. One example of a non-fixed combination or kit-of-parts is a combination wherein the first active ingredient and the further active ingredient are present separately. It is possible for the components of the non-fixed combination or kit-of-parts to be administered separately, sequentially, simultaneously, concurrently or chronologically staggered.
The inventive compounds can be employed alone or, if required, in combination with other active ingredients. The present invention further provides medicaments comprising at least one of the inventive compounds and one or more further active ingredients, especially for treatment and/or prophylaxis of the aforementioned disorders. Preferred examples of suitable active ingredient combinations include:
• organic nitrates and NO donors, for example sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SIN-1, and inhaled NO;
• compounds which inhibit the breakdown of cyclic guanosine monophosphate (cGMP), for example inhibitors of phosphodiesterases (PDE) 1, 2 and/or 5, especially PDE 5 inhibitors such as sildenafil, vardenafil, tadalafil, udenafil, desantafil, avanafil, mirodenafil, lodenafil or PF-00489791;
• hypotensive active ingredients, by way of example and with preference from the group of the calcium antagonists, angiotensin All antagonists, ACE inhibitors, NEP-inhibitors, vasopeptidase-inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, rho-kinase-inhibitors and the diuretics;
• antiarrhythmic agents, by way of example and with preference from the group of sodium channel blocker, beta-receptor blocker, potassium channel blocker, calcium antagonists, If- channel blocker, digitalis, parasympatholytics (vagoliytics), sympathomimetics and other antiarrhythmics as adenosin, adenosine receptor agonists as well as vernakalant;
• positive-inotrop agents, by way of example cardiac glycoside (Dogoxin), beta-adrenergic and dopaminergic agonists, such as isoprenalin, adrenalin, noradrenalin, dopamin or dobutamin;
• vasopressin-receptor-antagonists, by way of example and with preference from the group of conivaptan, tolvaptan, lixivaptan, mozavaptan, satavaptan, SR-121463, RWJ 676070 or BAY 86-8050, as well as the compounds described in WO 2010/105770, WO2011/104322 and WO 2016/071212;
• active ingredients which alter 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, of 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.
• bronchodilatory agents, for example and with preference from the group of the beta-adrenergic rezeptor-agonists, such as, by way of example and preferably, albuterol, isoproterenol, metaproterenol, terbutalin, formoterol or salmeterol, or from the group of the anticholinergics, such as, by way of example and preferably, ipratropiumbromid;
• anti-inflammatory agents, for example and with preference from the group of the gluco corticoids, such as, by way of example and preferably, prednison, prednisolon, methylprednisolon, triamcinolon, dexamethason, beclomethason, betamethason, flunisolid, budesonid or fluticason as well as the non-steroidal anti-inflammatory agents (NSAIDs), by way of example and preferably, acetyl salicylic acid (aspirin), ibuprofen and naproxen, 5-amino salicylic acid-derivates, leukotriene-antagonists, TNF-alpha-inhibitors and chemokin-receptor antagonists, such as CCR1, 2 and/or 5 inhibitors;
• agents modulating the immune system, for example immunoglobulins;
• agents that inhibit the signal transductions cascade, for example and with preference from the group of the kinase inhibitors, byway of example and preferably, from the group of the tyrosine kinase- and/or serine/threonine kinase inhibitors;
• agents, that inhibit the degradation and modification of the extracellular matrix, for example and with preference from the group of the inhibitors of the matrix-metalloproteases (MMPs), by way of example and preferably, inhibitors of chymasee, stromelysine, collagenases, gelatinases and aggrecanases (with preference from the group of MMP-1, MMP-3, MMP-8, MMP-9, MMP-10, MMP-11 and MMP-13) as well as of the metallo-elastase (MMP-12) and neutrophil-elastase (HNE), as for example sivelestat or DX-890;
• agents, that block the bindung of serotonin to its receptor, for example and with preference antagonists of the 5-HT2b-receptor;
• organic nitrates and NO-donators, for example and with preference sodium nitroprussid, nitro glycerine, isosorbid mononitrate, isosorbid dinitrate, molsidomine or SIN-1, as well as inhaled NO;
• NO-independent, but heme-dependent stimulators of the soluble guanylate cyclase, for example and with preference the compounds described in WO 00/06568, WO 00/06569, WO 02/42301, WO 03/095451, WO 2011/147809, WO 2012/004258, WO 2012/028647 and WO 2012/059549;
• NO-independent and heme-independent activators of the soluble guanylate cyclase, for example and with preference the compounds described in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 and WO 02/070510 beschriebenen Verbindungen;
• agents, that stimulates the synthesis of cGMP, wie beispielsweise sGC Modulatoren, for example and with preference riociguat, cinaciguat, vericiguat or BAY 1101042;
• prostacyclin-analogs, for example and with preference iloprost, beraprost, treprostinil or epoprostenol;
• agents, that inhibit soulble epoxidhydrolase (sEH), for example and with preference N,N'-Di- cyclohexyl urea, 12-(3-Adamantan-1-yl-ureido)-dodecanic acid or 1-Adamantan-1-yl-3-{5-[2- (2-ethoxyethoxy)ethoxy]pentyl}-urea;
• agents that interact with glucose metabolism, for example and with preference insuline, biguanide, thiazolidinedione, sulfonyl urea, acarbose, DPP4 inhibitors, GLP-1 analogs or SGLT-1 inhibitors;
• natriuretic peptides, for example and with preference atrial natriuretic peptide (ANP), natriuretic peptide type B (BNP, Nesiritid) natriuretic peptide type C (CNP) or urodilatin;
• activators of the cardiac myosin, for example and with preference omecamtiv mecarbil (CK- 1827452);
• calcium-sensitizers, for example and with preference levosimendan;
• agents that affect the energy metabolism of the heart, for example and with preference etomoxir, dichloroacetat, ranolazine or trimetazidine, full or partial adenosine A1 receptor agonists such as GS-9667 (formerly known as CVT-3619), capadenoson, neladenoson and neladenoson bialanate; agents that affect the heart rate, for example and with preference ivabradin; cyclooxygenase inhibitors such as, for example, bromfenac and nepafenac; inhibitors of the kallikrein-kinin system such as, for example, safotibant and ecallantide; inhibitors of the sphingosine 1-phosphate signal paths such as, for example, sonepcizumab; inhibitors of the complement-C5a receptor such as, for example, eculizumab; plasminogen activators (thrombolytics/fibrinolytics) and compounds which promote thrombolysis/fibrinolysis such as inhibitors of the plasminogen activator inhibitor (PAI inhibitors) or inhibitors of the thrombin-activated fibrinolysis inhibitor (TAFI inhibitors) such as, for example, tissue plasminogen activator (t-PA, for example Actilyse®), streptokinase, reteplase and urokinase or plasminogen-modulating substances causing increased formation of plasmin;
• anticoagulatory substances (anticoagulants) such as, for example, heparin (UFH), low- molecular-weight heparins (LMW), for example tinzaparin, certoparin, parnaparin, nadroparin, ardeparin, enoxaparin, reviparin, dalteparin, danaparoid, semuloparin (AVE 5026), adomiparin (M118) and EP-42675/ORG42675;
• direct thrombin inhibitors (DTI) such as, for example, Pradaxa (dabigatran), atecegatran (AZD- 0837), DP-4088, SSR-182289A, argatroban, bivalirudin and tanogitran (BIBT-986 and prodrug BIBT-1011) and hirudin;
• direct factor Xa inhibitors such as, for example, rivaroxaban, apixaban, edoxaban (DU-176b), betrixaban (PRT-54021), R-1663, darexaban (YM-150), otamixaban (FXV-673/R PR- 130673), letaxaban (TAK-442), razaxaban (DPC-906), DX-9065a, LY-517717, tanogitran (BIBT-986, prodrug: BIBT-1011), idraparinux and fondaparinux;
• inhibitors of coagulation factor XI and Xla such as, for example, FXI ASO-LICA, BAY 121- 3790, MAA868, BMS986177, EP-7041 and AB-022;
• substances which inhibit the aggregation of platelets (platelet aggregation inhibitors, thrombocyte aggregation inhibitors), such as, for example, acetylsalicylic acid (such as, for example, aspirin), P2Y12 antagonists such as, for example, ticlopidine (Ticlid), clopidogrel (Plavix), prasugrel, ticagrelor, cangrelor and elinogrel, and PAR-1 antagonists such as, for example, vorapaxar, and PAR-4 antagonists;
• platelet adhesion inhibitors such as GPVI and/or GPIb antagonists such as, for example, Revacept or caplacizumab;
• fibrinogen receptor antagonists (g lycoprotei n- 11 b/l 11 a antagonists) such as, for example, abciximab, eptifibatide, tirofiban, lamifiban, lefradafiban and fradafiban;
• recombinant human activated protein C such as, for example, Xigris or recombinant thrombomodulin.
Antithrombotic agents are preferably understood to mean compounds from the group of the platelet aggregation inhibitors, the anticoagulants or the profibrinolytic substances.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a platelet aggregation inhibitor, by way of example and with preference aspirin, clopidogrel, prasugrel, ticagrelor, ticlopidin or dipyridamole.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a thrombin inhibitor, by way of example and with preference ximelagatran, dabigatran, melagatran, bivalirudin or clexane.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a GPIIb/llla antagonist such as, by way of example and with preference, tirofiban or abciximab.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a factor Xa inhibitor, by way of example and with preference rivaroxaban (BAY 59- 7939), DU-176b, apixaban, betrixaban, otamixaban, fidexaban, razaxaban, letaxaban, eribaxaban, fondaparinux, idraparinux, PMD-3112, darexaban (YM-150), KFA-1982, EMD-503982, MCM-17, MLN-1021, DX 9065a, DPC 906, JTV 803, SSR-126512 or SSR-128428.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a factor XI or factor Xla inhibitor, by way of example and with preference FXI ASO- LICA, BAY 121-3790, MAA868, BMS986177, EP-7041 or AB-022.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with heparin or with a low molecular weight (LMW) heparin derivative.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a vitamin K antagonist, by way of example and with preference coumarin.
Hypotensive agents are preferably understood to mean compounds from the group of the calcium antagonists, angiotensin All 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 inventive compounds 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 inventive compounds 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 inventive compounds 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 inventive compounds are administered in combination with an angiotensin All antagonist, by way of example and with preference losartan, candesartan, valsartan, telmisartan or embusartan or a dual angiotensin All antagonist/neprilysin- inhibitor, by way of example and with preference LCZ696 (valsartan/sacubitril).
In a preferred embodiment of the invention, the inventive compounds are administered in combination with an ACE inhibitor, by way of example and with preference enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril ortrandopril. In a preferred embodiment of the invention, the inventive compounds 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 inventive compounds 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 inventive compounds 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 inventive compounds are administered in combination with a loop diuretic, for example furosemide, torasemide, bumetanide and piretanide, with potassium-sparing diuretics, for example amiloride and triamterene, with aldosterone antagonists, for example spironolactone, potassium canrenoate and eplerenone, and also thiazide diuretics, for example hydrochlorothiazide, chlorthalidone, xipamide and indapamide.
Lipid metabolism modifiers are preferably understood to mean compounds from the group of the CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA reduc tase inhibitors or squalene synthesis inhibitors, the 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 the lipoprotein(a) antagonists.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a CETP inhibitor, by way of example and with preference dalcetrapib.anacetrapib, torcetrapib (CP- 529414), JJT-705 or CETP vaccine (Avant).
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a thyroid receptor agonist, by way of example and with preference D-thyroxine, 3,5,3'-triiodothyronine (T3), CGS 23425 or axitirome (CGS 26214). In a preferred embodiment of the invention, the inventive compounds 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 inventive compounds 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 inventive compounds are administered in combination with an ACAT inhibitor, by way of example and with preference avasimibe, melinamide, pactimibe, eflucimibe or SM P-797.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with an MTP inhibitor, byway of example and with preference implitapide, BMS-201038, R- 103757 or JTT-130.
In a preferred embodiment of the invention, the inventive compounds 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 inventive compounds 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 inventive compounds 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 inventive compounds are administered in combination with a lipase inhibitor, a preferred example being orlistat. In a preferred embodiment of the invention, the inventive compounds 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 inventive compounds 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 inventive compounds are administered in combination with a lipoprotein(a) antagonist, by way of example and with preference, gemcabene calcium (CI-1027) or nicotinic acid.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with a lipoprotein(a) antagonist, by way of example and with preference, gemcabene calcium (CI-1027) or nicotinic acid.
In a preferred embodiment of the invention, the inventive compounds are administered in combination with sGC modulators, by way of example and with preference, riociguat, cinaciguat or vericiguat. In a preferred embodiment of the invention, the inventive compounds are administered in combination with an agent affecting the glucose metabolism, by way of example and with preference, insuline, a sulfonyl urea, acarbose, DPP4 inhibitors, GLP-1 analogs or SGLT-1 inhibitors.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a TGFbeta antagonist, by way of example and with preference pirfenidone or fresolimumab.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a CCR2 antagonist, by way of example and with preference CCX- 140. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a TNFalpha antagonist, by way of example and with preference adalimumab.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a galectin-3 inhibitor, by way of example and with preference GCS- 100.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a Nrf-2 inhibitor, by way of example and with preference bardoxolone
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a BMP-7 agonist, by way of example and with preference THR- 184.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a NOX1/4 inhibitor, by way of example and with preference GKT- 137831. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a medicament which affects the vitamin D metabolism, by way of example and with preference calcitriol, alfacalcidol, doxercalciferol, maxacalcitol, paricalcitol, cholecalciferol or paracalcitol.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a cytostatic agent, by way of example and with preference cyclophosphamide.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an immunosuppressive agent, by way of example and with preference ciclosporin. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a phosphate binder, by way of example and with preference colestilan, sevelamer hydrochloride and sevelamer carbonate, Lanthanum and lanthanum carbonate. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with renal proximal tubule sodium-phosphate co-transporter, by way of example and with preference, niacin or nicotinamide.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a calcimimetic for therapy of hyperparathyroidism. In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with agents for iron deficit therapy, by way of example and with preference iron products.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with agents for the therapy of hyperurikaemia, by way of example and with preference allopurinol or rasburicase.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with glycoprotein hormone for the therapy of anaemia, by way of example and with preference erythropoietin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with biologies for immune therapy, by way of example and with preference abatacept, rituximab, eculizumab or belimumab.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with vasopressin antagonists (group of the vaptanes) for the treatment of heart failure, by way of example and with preference tolvaptan, conivaptan, lixivaptan, mozavaptan, satavaptan or relcovaptan.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with Jak inhibitors, by way of example and with preference ruxolitinib, tofacitinib, baricitinib, CYT387, GSK2586184, lestaurtinib, pacritinib (SB1518) or TG101348.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with prostacyclin analogs for therapy of microthrombi.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an alkali therapy, by way of example and with preference sodium bicarbonate.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an mTOR inhibitor, by way of example and with preference everolimus or rapamycin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an NHE3 inhibitor, by way of example and with preference AZD1722 or tenapanor.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with an eNOS modulator, by way of example and with preference sapropterin.
In a preferred embodiment of the invention, the compounds according to the invention are administered in combination with a CTGF inhibitor, by way of example and with preference FG-3019.
The total amount of the active ingredient to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 50 mg/kg body weight per day, and more preferably from about 0.01 mg/kg to about 10 mg/kg body weight per day. Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing. In addition, it is possible for “drug holidays”, in which a patient is not dosed with a drug for a certain period of time, to be beneficial to the overall balance between pharmacological effect and tolerability. It is possible for a unit dosage to contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day. The average daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily. The transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg. The average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.
Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
Nevertheless, it may optionally be necessary to deviate from the stated amounts, namely depending on body weight, route of administration, individual response to the active substance, type of preparation and time point or interval when application takes place. Thus, in some cases it may be sufficient to use less than the aforementioned minimum amount, whereas in other cases the stated upper limit must be exceeded. When applying larger amounts, it may be advisable to distribute these in several individual doses throughout the day.
According to a further embodiment, the compounds of formula (I) according to the invention are administered orally once or twice or three times a day. According to a further embodiment, the compounds of formula (I) according to the invention are administered orally once or twice a day. According to a further embodiment, the compounds of formula (I) according to the invention are administered orally once a day. For the oral administration, a rapid release or a modified release dosage form may be used.
Unless stated otherwise, the percentages in the tests and examples which follow are percentages by weight; parts are parts by weight. Solvent ratios, dilution ratios and concentration data for the liquid/liquid solutions are based in each case on volume “w/v” means “weight/volume”. For example, “10% w/v” means: 100 ml of solution or suspension comprise 10 g of substance.
EXPERIMENTAL SECTION
Abbreviations and acronyms: aq. aqueous (solution)
Boc fert.-butoxycarbonyl br. broad (signal in NMR)
CDI 1,T-carbonyldiimidazole
COMU (1-cyano-2-ethoxy-2-oxoethylidenaminooxy)dimethylamino-morpholino- carbenium hexafluorophosphate d day(s); doublet (in NMR)
DBU 1 ,8-diazabicyclo[5.4.0]undec-7-en
DCI direct chemical ionization (in MS)
DCM dichloromethane dd doublet of doublets (in NMR)
DIPEA A/,/\/-diisopropylethylamine
DMAP 4-/\/,/\/-dimethylaminopyridine
DMF A/,/\/-dimethylformamide
DMSO dimethylsulfoxide dppp 1 ,3-bis(diphenylphosphino)propane
EDC /V-ethyl-/V-(3-dirnethylarninopropyl)carbodiirnide hydrochloride
EDCI 1-(3-dimethyla inopropyl)-3-ethylcarbodii ide equiv. equivalent(s)
ESI electrospray ionization (in MS) h hour(s)
HATU 0-(7-azabenzotriazol-1-yl)-/V,/\/,/\/',/\/-tetramethyluronium-hexafluorophosphate
HOAt 1-hydroxy-7-azabenzotriazole
HOBt 1 -hydroxy- 1/-/-benzotriazole hydrate
HPLC high-pressure / high-performance liquid chromatography
HV high vacuum
LC-MS liquid chromatography-coupled mass spectrometry m multiplet (in NMR) min minute(s)
MS mass spectrometry
MTBE methyl tert.- butyl ether
NMP /V-methylpyrrolidone
NMM /V-methylmorpholine
NMR nuclear magnetic resonance spectrometry
PTSA p-toluenesulfonic acid q quartet or quadruplet (in NMR) quant. quantitative (yield) quin quintet (in NMR)
RP reverse phase (in HPLC)
RT or rt room temperature
Rt retention time (in HPLC, LC-MS) s singlet (in NMR) sep septet (in NMR) sext sextet (in NMR)
SFC supercritical fluid chromatography t triplet (in NMR)
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC thin-layer chromatography
T3P® propylphosphonic anhydride (2,4,6-tripropyl-1 ,3,5,2,4,6-trioxatriphosphinane
2, 4, 6- trioxide)
Other abbreviations not specified herein have their meanings customary to the skilled person.
The various aspects of the invention described in this application are illustrated by the following examples which are not meant to limit the invention in any way. All publications mentioned herein are incorporated by reference in their entirety.
The example testing experiments described herein serve to illustrate the present invention and the invention is not limited to the examples given.
EXPERIMENTAL SECTION - GENERAL PART All reagents, for which the synthesis is not described in the experimental part, are either commercially available, or are known compounds or may be formed from known compounds by known methods by a person skilled in the art.
NMR peak forms are stated as they appear in the spectra, possible higher order effects have not been considered. The 1H-NMR data of selected compounds are listed in the form of 1H-NMR peaklists. For each signal peak the d value in ppm is given, followed by the signal intensity, reported in round brackets. The d value-signal intensity pairs from different peaks are separated by commas. Therefore, a peaklist is described by the general form: di (intensityi), 62 (intens^), ... , d, (intensity,), ... , dh (intensity^. The intensity of a sharp signal correlates with the height (in cm) of the signal in a printed NMR spectrum. When compared with other signals, this data can be correlated to the real ratios of the signal intensities. In the case of broad signals, more than one peak, or the center of the signal along with their relative intensity, compared to the most intense signal displayed in the spectrum, are shown. A 1H-NMR peaklist is similar to a classical 1H-NMR readout, and thus usually contains all the peaks listed in a classical NMR interpretation. Moreover, similar to classical 1H-NMR printouts, peaklists can show solvent signals, signals derived from stereoisomers of target compounds (also the subject of the invention), and/or peaks of impurities. The peaks of stereoisomers, and/or peaks of impurities are typically displayed with a lower intensity compared to the peaks of the target compounds (e.g., with a purity of >90%). Such stereoisomers and/or impurities may be typical for the particular manufacturing process, and therefore their peaks may help to identify the reproduction of our manufacturing process on the basis of "by-product fingerprints". An expert who calculates the peaks of the target compounds by known methods (MestReC, ACD simulation, or by use of empirically evaluated expectation values), can isolate the peaks of target compounds as required, optionally using additional intensity filters. Such an operation would be similar to peak-picking in classical 1H-NMR interpretation. A detailed description of the reporting of NMR data in the form of peaklists can be found in the publication "Citation of NMR Peaklist Data within Patent Applications" (cf. Research Disclosure Database Number 605005, 2014, 01 Aug 2014, or http://www.researchdisclosure.com/searching- disclosures). In the peak picking routine, as described in the Research Disclosure Database Number 605005, the parameter "MinimumHeight" can be adjusted between 1% and 4%. Depending on the chemical structure and/or depending on the concentration of the measured compound it may be reasonable to set the parameter "MinimumHeight" <1%.
In NMR spectra of mixtures of stereoisomers, numbers mentioned with 7” indicate that the stereoisomers show separate signals for the respective hydrogen atom, i.e. “.... / . (2s, 1 H)” means that one hydrogen atom is represented by 2 singlets, each singlet from one or more different stereoisomer(s). lUPAC names of the following intermediates and example compounds were generated using the ACD/Name software (batch version 14.00; Advanced Chemistry Development, Inc.) or the naming tool implemented in the BIOVIA Draw software (version 4.2 SP1; Dassault Systemes SE).
Analytical LC-MS methods
Method 1: Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ^ 1.2 min 100% B ® 2.0 min 100% B; column oven: 40°C; flow rate: 1.2 ml/min. Method 2: Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 1.0 ml/min.
Method 3: Column: XBridge C18, 3.5 pm, 4.6 c 50 mm; mobile phase A: 5mM ammonium hydrogencarbonate in water, mobile phase B: acetonitrile; gradient: 0.0 min 10% B ® 4.2 min 95% B ® 5.2 min 95% B; column oven: 35°C; flow rate: 1.5 ml/min.
Method 4: Column: Ascentis Express C18, 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 100% B ® 1.7 min 100% B; column oven: 40°C; flow rate: 1.0 ml/min. Method 5: Column: Ascentis Express C18, 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.7 min 95% B; column oven: 40°C; flow rate: 1.0 ml/min.
Method 6: Column: Ascentis Express C18, 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 100% B ® 1.7 min 100% B; column oven: 40°C; flow rate: 1.0 ml/min.
Method 7: Column: Kinetex EVO-C18 (Phenomenex), 2.6 pm, 3.0 c 50 mm. mobile mhase A: 5 mM NH4HCO3 in water, mobile phase B: acetonitrile; gradient: 0.0 min 10% B ® 2.1 min 95% B ® 3.0 min 95% B; column oven: 40°C; flow rate: 1.0 ml/min.
Method 8: Instrument: Waters Acquity SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8 pm, 50 x 1 mm; eluent A: 1 L water + 0.25 ml formic acid, eluent B: 1 L acetonitrile + 0.25 ml formic acid; gradient: 0.0 min 90% A ® 1.2 min 5% A ® 2.0 min 5% A; column oven: 50°C; flow rate: 0.40 ml/min; UV detection: 208-400 nm.
Method 9: MS instrument: Thermo Scientific FT-MS; instrument UHPLC+: Thermo Scientific UltiMate 3000; column: Waters HSS T3, 2.1 c 75 mm, C18 1.8 pm; eluent A: 1 L water + 0.01% formic acid, eluent B: 1 L acetonitrile + 0.01% formic acid; gradient: 0.0 min 10% B ® 2.5 min 95% B ® 3.5 min 95% B; oven: 50°C; flow rate: 0.90 ml/min; UV detection: 210 nm/optimum integration path 210-300 nm.
Method 10: Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 0.8 ml/min.
Method 11: Column: Kinetex EVO-C18, 2.6 pm, 3.0 c 50 mm; mobile phase A: 5mM ammonium hydrogencarbonate in water, mobile phase B: acetonitrile; gradient: 0.0 min 10% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 1.2 ml/min. Method 12: Column: Omega, 3.0 pm, 2.1 c 50 mm; mobile phase A: 0.09% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 1.2 ml/min.
Method 13: Column: Ascentis Express C18, 2.7 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.7 min 95% B; column oven: 40°C; flow rate: 1.5 ml/min.
Method 14: Column: CORTECS C18+ (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.09% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 100% B -> 1.6 min 100% B; column oven: 40°C; flow rate: 0.8 ml/min. Method 15: Column: Shim-pack XR-ODS (Shimadzu), 2.2 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 95% B ® 1.6 min 95% B; column oven: 40°C; flow rate: 1.5 ml/min.
Method 16: Column: Ascentis Express C18, 2.7 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ^ 2.1 min 100% B ® 2.8 min 100% B; column oven: 40°C; flow rate: 1.5 ml/min.
Method 17: Column: Ascentis Express C18 (Supelco), 2.7 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.7 min 95% B; column oven: 40°C; flow rate: 1.5 ml/min.
Method 18: Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 3.7 min 70% B ® 4.2 min 95% B ® 4.7 min 95% B; column oven: 40°C; flow rate: 0.8 ml/min.
Method 19: Column: Shim-pack XR-ODS (Shimadzu), 2.2 pm, 3.0 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.7 min 95% B; column oven: 40°C; flow rate: 1.5 ml/min. Method 20: Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 1.2 ml/min.
Method 21: Column: CORTECS C18+ (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.09% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 2.0 min 95% B ® 2.6 min 95% B; column oven: 40°C; flow rate: 0.8 ml/min.
Method 22: MS instrument: Waters Single Quad MS System; Waters UPLC Acquity; column: Waters BEH C18, 1.7 pm, 50 c 2.1 mm; eluent A: 1 L water + 1.0 ml aq. ammonium hydroxide solution (25% ammonia), eluent B: 1 L acetonitrile; gradient: 0.0 min 92% A ® 0.1 min 92% A ® 1.8 min 5% A ® 3.5 min 5% A; column oven: 50°C; flow rate: 0.45 ml/min; UV detection: 210 nm (208-400 nm).
Method 23: Instrument: Waters Acquity SQD UPLC System; column: Waters Acquity UPLC HSS T3 1.8 pm, 50 c 1 mm; eluent A: 1 L water + 0.25 ml formic acid, eluent B: 1 L acetonitrile + 0.25 ml formic acid; gradient: 0.0 min 95% A ® 6.0 min 5% A ® 7.5 min 5% A; column oven: 50°C; flow rate: 0.35 ml/min; UV detection: 210 nm.
Method 24: MS instrument: Waters Single Quad MS System; Waters UPLC Acquity; column: Waters BEH C18, 1.7 pm, 50 c 2.1 mm; eluent A: 1 L water + 1.0 ml aq. ammonium hydroxide solution (25% ammonia), eluent B: 1 L acetonitrile; gradient: 0.0 min 92% A ® 0.1 min 92% A ® 1.8 min 5% A ® 3.5 min 5% A; column oven: 50°C; flow rate: 0.45 ml/min; UV detection: 210 nm.
Method 25: Column: Ascentis Express C18 (Supelco), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.05% TFA in water, mobile phase B: 0.05% TFA in acetonitrile; gradient: 0.0 min 5% B ® 1.1 min 100% B ® 2.0 min 100% B; column oven: 40°C; flow rate: 1.5 ml/min.
Method 26: Instrument: Waters ACQUITY SQD UPLC System; Column: Waters Acquity UPLC HSS T3 1.8 pm 50 x 1 mm; eluent A: 1 L water + 0.25 ml 99% formic acid, eluent B: 1 L acetonitrile + 0.25 ml 99% formic acid; gradient: 0.0 min 90% A 1.2 min 5% A 2.0 min 5% A oven: 50°C; flow rate: 0.40 ml/min; UV-Detection: 210 nm.
Method 27: Column: CORTECS C18 (Waters), 2.7 pm, 2.1 c 50 mm; mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; gradient: 0.0 min 5% B ® 3.2 min 50% B ® 4.2 min 95% B® 5.0 min 95% B; column oven: 40°C; flow rate: 1.0 ml/min.
Method 28: Instrument MS: Waters SQD; Instrument HPLC: Waters UPLC; column: Zorbax SB- Aq (Agilent), 50 mm x 2.1 mm, 1.8 pm; eluent A: water + 0.025% formic acid, eluent B: acetonitrile (ULC) + 0.025% formic acid; gradient: 0.0 min 98% A ® 0.9 min 25% A ® 1.0 min 5% A ® 1.4 min 5% A ® 1.41 min 98% A -> 1.5 min 98% A; oven: 40°C; flow: 0.600 ml/min; UV-detection: DAD; 210 nm.
Analytical GCMS methods
Method G1: Instrument: Thermo Scientific DSQII, Thermo Scientific Trace GC Ultra; Column: Restek RTX-35MS, 15 m x 200 pm x 0.33 pm; constant flow with helium, flow rate: 1.2 ml/min; oven: 60°C; inlet: 220°C; gradient: 60°C, 30°C/min 300°C (3.33 min stop). Preparative HPLC methods
Method P1: Instrument: Waters Prep LC/MS System; column: XBridge C18 5 pm, 100 c 30 mm; eluent A: water, eluent B: acetonitrile; flow rate: 80 ml/min plus 5.0 ml of aq. ammonia (2% ammonia in water); at-column injection; gradient: 0.0-2.0 min 0% B, 2.0-10 min 0% B ® 100% B, 10-12 min 100%; column oven: RT; UV detection: 200-400 nm.
Method P2: Column: Reprosil C18, 10 mM, 125 x 30 mm;, eluent: A = water + 0.01% formic acid, eluent B = acetonitrile; gradient: 0.0-5.00 min = 10% B, 6.50 min = 20% B, 17.0-19.75 min = 100% B, 19.75-23.00 min 90% B. Flow rate: 75 ml/min, UV-Detection: 210 nm.
Method P3: Column: Phenomenex Gemini C18 250 x 50mm x 10 urn; eluent A: water + 0.05% of ammonia; eluent B: acetonitrile; gradient: 0-28 min 10% B ® 35% B.
Method P4: Column: Chromatorex C18, 10pm, 205 c 50 mm; eluent A: water + 0.1% of formic acid; eluent B: acetonitrile; gradient: 0.0-5.0 min 10% B, 5.0-17.5 min 10% B to 95% B, 17.5-21.0 min 95% B; flow rate: 150 ml/min, UV-Detection: 210 nm.
Microwave: Reactions employing microwave irradiation may be run with a Biotage Initator® microwave oven optionally equipped with a robotic unit. The reported reaction times employing microwave heating are intended to be understood as fixed reaction times after reaching the indicated reaction temperature.
When compounds according to the invention are purified by preparative HPLC by the above- described methods in which the eluents contain additives, 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 contain a sufficiently basic or acidic functionality. Such a salt can be converted to the corresponding free base or acid by various methods known to the person skilled in the art.
In the case of the synthesis intermediates and working examples of the invention described hereinafter, any compound specified in the form of a salt of the corresponding base or acid is generally a salt of unknown exact stoichiometric composition, as obtained by the respective preparation and/or purification process. Unless specified in more detail, additions to names and structural formulae, such as “hydrochloride”, “trifluoroacetate”, “sodium salt” or "x HCI", "x CF3COOH", "x Na+" should not therefore be understood in a stoichiometric sense in the case of such salts, but have merely descriptive character with regard to the salt-forming components present therein.
This applies correspondingly if synthesis intermediates or working examples or salts thereof were obtained in the form of solvates, for example hydrates, of unknown stoichiometric composition (if they are of a defined type) by the preparation and/or purification processes described. Starting compounds and intermediates
Intermediate 1A
Diethyl 1 H-pyrrole-2,4-dicarboxylate
Figure imgf000061_0001
To a solution of ethyl prop-2-ynoate (31 ml, 310 mmol) in 1,4-dioxane (400 ml) was added ethyl isocyanoacetate (40 ml, 370 mmol), copper(l)oxide (2.19 g, 15.3 mmol) and 1,10-phenanthroline (5.51 g, 30.6 mmol) under a nitrogen atmosphere. The solution was stirred at 100°C for 2 h. After cooling to RT, the reaction mixture was filtered over Celite® and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (330 g silica gel, eluent: petroleum ether/ethyl acetate, 10:1) to give 45.0 g (65% of theory, 97% purity) of the title compound.
LC-MS (Method 1): Rt = 0.88 min, MS (ESIpos): m/z = 212 [M+H]+
1H-NMR (300 MHz, DM SO-de) : d [ppm] = 12.53 (brs, 1H), 7.56 (s, 1H), 7.07 (s, 1H), 4.29-4.16 (m, 4H), 1.32-1.24 (m, 6H). Intermediate 2A
Diethyl 1-[2-(naphthalen-2-yl)-2-oxoethyl]-1 H-pyrrole-2,4-dicarboxylate
Figure imgf000061_0002
To a solution of diethyl 1H-pyrrole-2,4-dicarboxylate (Intermediate 1A, 4.00 g, 18.9 mmol) in acetone (50 ml) was added 2-bromo-1-(naphthalen-2-yl)ethan-1-one (6.29 g, 75% purity, 18.9 mmol) and potassium carbonate (3.93 g, 28.4 mmol). The mixture was stirred for 2 h at RT, then reaction was filtered and the filtrate was concentrated. The crude residue was suspended in ethyl acetate/petroleum ether (150 ml, 1:10) and stirred for 30 min, then the solid was collected by filtration to give to afford 2.60 g (33% of theory, 90% purity) of the title compound.
LC-MS (Method 2): Rt = 1.27 min; MS (ESIpos): m/z = 380 [M+H]+ 1H-NMR (300 MHz, DMSO-d6): d [ppm] 8.83 (s, 1H), 8.19 (d, 1H), 8.12-8.01 (m, 3H), 7.85 (s, 1H), 7.76-7.65 ( , 2H), 7.24 (s, 1H), 6.12 (s, 2H), 4.24 (q, 2H), 4.11 (q, 2H), 1.27 (t, 3H), 1.16 (t, 3H).
Intermediate 3A
Ethyl 3-(naphthalen-2-yl)-1 -oxo-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate
Figure imgf000062_0001
To a solution of diethyl 1-[2-(naphthalen-2-yl)-2-oxoethyl]-1H-pyrrole-2,4-dicarboxylate (Intermedaite 2A, 1.50 g, 3.95 mmol) in acetic acid (15 ml) was added ammonium acetate (7.62 g, 98.8 mmol). After refluxing for 48 h the reaction mixture poured onto ice-water and neutralized with sodium hydroxide. The precipitate was collected by filtration, washed with water and dried at 100°C to afford 0.91 g (63% of theory, 92% purity) of the title compound.
1H-NMR (300 MHz, DMSO-de): d [ppm] 11.19 (s, 1H), 8.30 (s, 1H), 8.05-7.94 (m, 5H), 7.78 (d, 1H), 7.61-7.58 (m, 2 H), 7.24 (s, 1H), 4.27 (q, 2H), 1.32 (t, 3H).
Intermediate 4A
3-(Naphthalen-2-yl)-1 -oxo-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid
Figure imgf000062_0002
To a solution of ethyl 3-(naphthalen-2-yl)-1-oxo-1,2-dihydropyrrolo[1,2-a]pyrazine-7-carboxylate (Intermediate 3A, 990 mg, 2.98 mmol) in water (10 ml) and ethanol (10 ml) was added sodium hydroxide (12 ml of a 2.0 N aqueous solution, 24.0 mmol). After stirring at RT for 2 h the mixture was diluted with water and the pH was adjusted to 1 to 2 with hydrochloric acid. The solid was collected by filtration and dried to afford 663 mg (71 % of theory, 96% purity) of the title compound.
LC-MS (Method 3): Rt = 1.50 min; MS (ESIpos): m/z = 305 [M+H]+
1H-NMR (400 MHz, DMSO-de): d [ppm] 12.57 (s, 1H), 11.16 (s, 1H), 8.30 (s, 1H), 8.20-7.94 (m, 5H), 7.78 (d, 1H), 7.67-7.57 (m, 2 H), 7.21 (s, 1H). Intermediate 5A
Diethyl 1-[2-(3,4-dimethylphenyl)-2-oxoethyl]-1 H-pyrrole-2,4-dicarboxylate
Figure imgf000063_0001
To a solution of diethyl 1H-pyrrole-2,4-dicarboxylate (Intermediate 1A, 1.40 g, 6.63 mmol) in acetone (20 ml) was addded 2-bromo-1-(3,4-dimethylphenyl)ethan-1-one (1.81 g, 7.95 mmol) and potassium carbonate (1.83 g, 13.3 mmol). The mixture was stirred overnight at RT, then filtered. The filtrate was concentrated, then diluted with water and extracted with ethyl acetate. The organic layers were combined, washed with brine, dried over anhydrous sodium sulfate and concentrated to afford 1.87 g (72% of theory, 92% purity) of the title compound. LC-MS (Method 4): Rt = 1.23 min; MS (ESIpos): m/z = 358 [M+H]+
1H-NMR (300 MHz, DMSO-d6): d [ppm] 7.82-7.76 (m, 3H), 7.37 (d, 1H), 7.20 (s, 1H), 5.93 (s, 2H), 4.23 (q, 2H), 4.10 (q, 2H), 2.33-2.30 (m, 6H), 1.28 (t, 3H), 1.17 (t, 3H).
Intermediate 6A
Ethyl 3-(3,4-dimethylphenyl)-1 -oxo-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylate
Figure imgf000063_0002
To a solution of diethyl 1-[2-(3,4-dimethylphenyl)-2-oxoethyl]-1H-pyrrole-2,4-dicarboxylate (Intermediate 5A, 1.50 g, 90% purity, 3.78 mmol) in acetic acid (15 ml) was added ammonium acetate (7.28 g, 94.4 mmol). After refluxing for 48 h the reaction mixture was poured onto ice-water and neutralized with sodium hydroxide. The precipitate was collected by filtration, washed with water and dried at 100°C under reduced pressure to afford 900 mg (69% of theory, 90% purity) of the title compound.
1H-NMR (300 MHz, DMSO-d6): d [ppm] 11.21 (br s, 1H), 7.98 (s, 1H), 7.71 (s, 1H), 7.47 (s, 1H), 7.38 (d, 1H), 7.24-7.17 (m, 2H), 4.26 (q, 2H), 2.30-2.20 (m, 6H), 1.28 (t, 3H). Intermediate 7A
3-(3,4-Dimethylphenyl)-1-oxo-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid
Figure imgf000064_0002
To a solution of ethyl 3-(3,4-dimethylphenyl)-1-oxo-1,2-dihydropyrrolo[1,2-a]pyrazine-7- carboxylate (Intermediate 6A, 1.20 g, 90% purity, 3.48 mmol) in water (10 ml) and ethanol (10 ml) was added sodium hydroxide (14 ml of a 2.0 N aqueous solution, 28.0 mmol). After stirring at RT for 2 h the mixture was diluted with water and the pH of the solution was adjusted to 1 to 2 with hydrochloric acid. The solid was collected by filtration and dried to afford 638 mg (63% of theory 98% purity) of the title compound. LC-MS (Method 5): Rt = 1.22 min; MS (ESIpos): m/z = 283 [M+H]+
1H-NMR (400 MHz, DMSO-d6): d [ppm] 12.50 (s, 1H), 10.93 (s, 1H), 7.92 (s, 1H), 7.71 (s, 1H), 7.46 (s, 1H), 7.38 (d, 1H), 7.23 (d, 1H), 7.15 (s, 1H), 2.32 (s, 3H), 2.27 (s, 3H).
Intermediate 8A
Diethyl 1-[2-(2,3-dihydro-1 ,4-benzodioxin-6-yl)-2-oxoethyl]-1 H-pyrrole-2,4-dicarboxylate
Figure imgf000064_0001
To a solution of diethyl 1H-pyrrole-2,4-dicarboxylate (Intermediate 1A, 1.50 g, 7.10 mmol) in acetone (15 ml) was addded 2-bromo-1-(2,3-dihydro-1,4-benzodioxin-6-yl)ethan-1-one (2.19 g, 8.52 mmol) and potassium carbonate (1.96 g, 14.2 mmol). The mixture was stirred overnight at RT, then filtered. The filtrate was concentrated, then the residue was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated to afford 1.50 g (50% yield, 91% purity) of the title compound.
LC-MS (Method 6): Rt = 1.16 min; MS (ESIpos): m/z = 388 [M+H]+ 1H-NMR (300 MHz, DMSO-d6): d [ppm] 7.77 (s, 1H), 7.61-7.52 (m, 2H), 7.18 (s, 1H), 7.04 (d, 1H), 5.87 (s, 2H), 4.38-4.29 (m, 4H), 4.24 (q, 2H), 4.10 (q, 2H), 1.27 (t, 3H), 1.19 (t, 3H).
Intermediate 9A
Ethyl 3-(2,3-dihydro-1,4-benzodioxin-6-yl)-1 -oxo-1 , 2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate
Figure imgf000065_0001
To a solution of diethyl 1-[2-(2,3-dihydro-1,4-benzodioxin-6-yl)-2-oxoethyl]-1H-pyrrole-2,4- dicarboxylate (Intermediate 8A, 1.50 g, 3.87 mmol) in acetic acid (15 ml) was added ammonium acetate (7.46 g, 96.8 mmol]). After refluxing for 48 h the reaction mixture was poured into ice-water and neutralized with sodium hydroxide. The solid was collected by filtration, washed with water and dried at 100°C under reduced pressure to afford 940 mg (62% of theory, 87% purity) of the title compound.
LC-MS (Method 7): Rt = 1.65 min; MS (ESIpos): m/z = 341 [M+H]+
Intermediate 10A 3-(2,3-Dihydro-1 ,4-benzodioxin-6-yl)-1-oxo-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid
Figure imgf000065_0002
To a solution of ethyl 3-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-oxo-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 9A, 1.00 g, 80% purity, 2.35 mmol) in water (10 ml) and ethanol (10 ml) was added sodium hydroxide (19 ml of a 1.0 N aqueous solution, 19.0 mmol). After stirring at RT for 2 h the mixture was diluted with water and the pH of the solution was adjusted to 1 to 2 with hydrochloric acid. The solid was collected by filtration and dried to afford 422 mg (56% of theory, 97% purity) of the title compound.
LC-MS (Method 7): Rt = 0.88 min; MS (ESIpos): m/z = 313 [M+H]+
1H-NMR (300 MHz, DMSO-d6): d [ppm] 12.49 (s, 1H), 10.91 (s, 1H), 7.90 (s, 1H), 7.68 (s, 1H), 7.20 (s, 1H), 7.12-7.16 (m, 2H), 6.95 (d, 1H), 4.29-4.23 (m, 4H). Intermediate 11A
2-Bromo-1-(5-fluoro-2,3-dihydro-1,4-benzodioxin-6-yl)ethan-1-one
Figure imgf000066_0001
To a solution of 1-(5-fluoro-2,3-dihydro-1,4-benzodioxin-6-yl)ethan-1-one (1.27 g, 6.47 mmol) in acetic acid (10 ml) at 40°C was added bromine (330 mI, 6.5 mmol) dropwise. The reaction was cooled to RT, then concentrated to afford 1.70 g (57% of theory, 60% purity) of the title compound, which was used without further purification
GC-MS (Method 7): Rt = 6.86 min; MS (Elpos): m/z = 276 [M]+
Intermediate 1A Diethyl 1-[2-(5-fluoro-2,3-dihydro-1 ,4-benzodioxin-6-yl)-2-oxoethyl]-1 H-pyrrole-2,4- dicarboxylate
Figure imgf000066_0002
To a solution of diethyl 1H-pyrrole-2,4-dicarboxylate (Intermediate 1A, 768 mg, 3.64 mmol), 2- bromo-1-(5-fluoro-2,3-dihydro-1,4-benzodioxin-6-yl)ethan-1-one (Intermediate 11 A, 1.00 g, 3.64 mmol) and potassium carbonate (553 mg, 4.00 mmol) in acetone (14 ml) was added water (5 drops). The reacton was stirred overnight at RT, then a further portion of potassium carbonate was added to the reaction (350 mg, 2.55 mmol) and the reaction was stirred overnight at RT. The reaction mixture was partitioned between water (50 ml) and dichloromethane (100 ml) and the aqueous phase was extracted with dichloromethane (2 x 100 ml). The combined organic phase was dried, concentrated and purified by silica gel column chromatography (eluent: dichloromethane/methanol) to afford 820 mg (54% of theory, 97% purity) of the title compound.
LC-MS (Method 8): Rt = 1.06 min; MS (ESIpos): m/z = 406 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: 1.159 (7.43), 1.177 (15.98), 1.195 (7.66), 1.256 (7.58), 1.273 (16.00), 1.291 (7.64), 4.086 (2.34), 4.104 (7.52), 4.122 (7.43), 4.139 (2.28), 4.192 (2.31), 4.210 (7.28), 4.227 (7.20), 4.245 (2.22), 4.386 (3.36), 4.399 (6.28), 4.411 (6.16), 4.424 (3.17), 5.738 (6.12), 5.745 (6.06), 5.753 (1.55), 6.892 (2.53), 6.895 (2.49), 6.914 (2.79), 6.917 (2.74), 7.180 (4.51), 7.185 (4.55), 7.379 (2.23), 7.399 (2.60), 7.401 (2.43), 7.421 (2.03), 7.775 (5.32), 7.779 (5.19).
Intermediate 2A Ethyl 3-(5-fluoro-2,3-dihydro-1,4-benzodioxin-6-yl)-1 -oxo-1 , 2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate
Figure imgf000067_0001
To a solution of diethyl 1-[2-(5-fluoro-2,3-dihydro-1,4-benzodioxin-6-yl)-2-oxoethyl]-1H-pyrrole- 2,4-dicarboxylate (Intermediate 12A, 820 mg, 2.02 mmol) in acetic acid (8.2 ml) was added ammonium acetate (3.90 g, 50.6 mmol). The mixture was heated at 125°C overnight, then the reaction was cooled to RT and concentrated. The crude residue was partitioned between water (2 ml) and dichloromethane (5 ml) and the pH of the solution was adjusted to 6 with sodium hydroxide (concentrated aqueous solution). The aqueous phase was extracted with dichloromethane (3 ml) and the combined organic phase was dried and concentrated to afford 566 mg (72% yield, 92% purity) of the title compound, which was used without further purification.
LC-MS (Method 9): Rt = 1.57 min; MS (ESIpos): m/z = 359 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.008 (1.28), 0.008 (0.86), 1.280 (4.22), 1.298 (8.77), 1.316 (4.16), 2.524 (1.14), 4.232 (1.33), 4.250 (3.91), 4.268 (3.83), 4.286 (1.23), 4.350 (16.00), 6.807 (1.41), 6.811 (1.37), 6.829 (1.71), 6.833 (1.66), 6.980 (1.43), 6.999 (1.83), 7.021 (1.11), 7.188 (2.53), 7.190 (2.49), 7.500 (3.87), 8.031 (3.42), 8.035 (3.26), 10.958 (1.37).
Intermediate 14A
3-(5-Fluoro-2,3-dihydro-1 ,4-benzodioxin-6-yl)-1-oxo-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylic acid
Figure imgf000067_0002
To a solution of ethyl 3-(5-fluoro-2,3-dihydro-1,4-benzodioxin-6-yl)-1-oxo-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 13A, 565 g, 92% purity, 1.45 mmol) in THF (10 ml) was added lithium hydroxide (3.0 ml of a 1.9 M aqueous solution, 5.80 mmol). The reaction was heated to 40°C for 72 h, then the reaction was concentrated to remove the THF and water was added. The pH of the aqueous solution was adjusted to 1 to 2 with hydrochloric acid (1.0 N aqueous solution) and the precipitated was collected by filtration and dried to afford 418 mg (87% of theory, 100% purity) of the title compound.
LC-MS (Method 9): Rt = 1.10 min; MS (ESIpos): m/z = 331 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: 1.031 (0.41), 4.350 (16.00), 6.805 (1.39), 6.808 (1.37), 6.826 (1.74), 6.829 (1.71), 6.982 (1.34), 7.002 (1.92), 7.023 (1.07), 7.156 (1.98), 7.499 (3.20), 7.963 (3.12), 7.967 (3.04), 10.922 (2.16), 12.483 (1.38). Intermediate 15A
4-Chloro-2-fluoro-/\/,3-dimethoxy-/\/-methylbenzamide
Figure imgf000068_0001
To a solution of 4-chloro-2-fluoro-3-methoxybenzoic acid (5.35 g, 26.2 mmol) in acetonitrile was added EDCI (7.52 g, 39.2 mmol), HOBT (881 mg, 5.75 mmol), /V-methoxymethanamine hydrochloride (3.83 g, 39.2 mmol) and triethylamine (11 ml, 78 mmol). The mixture was stirred overnight at RT. The reaction was concentrated and water (20 ml) was added and the mixture was extracted with ethyl acetate (3 x 100 ml). The combined organic phase was dried, concentrated and purified by silica gel column chromatography (eluent: cyclohexane/ethyl acetate) to afford 5.95 g (88% of theory, 96% purity) of the title compound. LC-MS (Method 8): Rt = 0.79 min; MS (ESIpos): m/z = 248 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: 3.313 (3.37), 3.519 (2.61), 3.912 (16.00), 3.915 (15.09), 5.754 (0.44), 7.204 (1.96), 7.220 (2.19), 7.225 (2.59), 7.241 (2.45), 7.392 (2.54), 7.396 (2.42), 7.413 (2.04), 7.417 (1.96).
Intermediate 3A 1-(4-Chloro-2-fluoro-3-methoxyphenyl)ethan-1-one To a solution of 4-chloro-2-fluoro-/\/,3-dimethoxy-/\/-methylbenzamide (Intermediate, 15A, 1.00 g, 96% purity, 3.88 mmol) in THF under an Ar atmosphere at 0°C was added bromido(methyl)magnesium (3.9 ml of a 3.0 M in diethyl ether, 12 mmol) dropwise.The reaction was stirred for 2 h at RT, then quenched by the addition of ammonium chloride (20 ml of an aqueous solution) and diluted with ethyl acetate (50 ml). The aqueous phase was extracted with ethyl acetate and the combined organic phase was washed with brine, dried and concentrated to afford 800 mg (quantitative, 98% purity) of the title compound, which was used without purification.
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: 1.357 (0.42), 2.580 (11.17), 2.591 (10.97), 3.923 (16.00), 7.436 (1.61), 7.439 (1.47), 7.457 (2.38), 7.461 (2.28), 7.536 (2.04), 7.555 (2.36), 7.577 (1.40).
Intermediate 4A
2-Bromo-1-(4-chloro-2-fluoro-3-methoxyphenyl)ethan-1-one
Figure imgf000069_0001
To a solution of 1-(4-chloro-2-fluoro-3-methoxyphenyl)ethan-1-one (Intermediate 16A, 500 mg, 2.47 mmol) in dichloromethane (6.7 ml) was added trimethylphenylammonium tribromide (974 mg, 2.59 mmol) at RT. The reaction was stirred for 16 h, then concentrated and the crude residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate) to afford 520 mg (64% of theory, 85% purity) of the title compound.
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: 1.356 (0.50), 2.580 (2.88), 2.590 (2.88), 3.925 (16.00), 3.928 (14.88), 3.941 (0.75), 4.487 (1.48), 4.848 (8.12), 4.853 (8.15), 7.458 (0.54), 7.462 (0.56),
7.487 (1.66), 7.491 (1.63), 7.509 (2.24), 7.513 (2.28), 7.537 (0.53), 7.555 (0.60), 7.558 (0.49), 7.628 (2.14), 7.647 (2.29), 7.650 (1.88), 7.668 (1.65).
Intermediate 5A
Diethyl 1-[2-(4-chloro-2-fluoro-3-methoxyphenyl)-2-oxoethyl]-1H-pyrrole-2,4-dicarboxylate To a solution of diethyl 1H-pyrrole-2,4-dicarboxylate (Intermedatie 1A, 332 mg, 1.57 mmol), 2- bromo-1-(4-chloro-2-fluoro-3-methoxyphenyl)ethan-1-one (Intermediate 17A, 520 mg, 85% purity, 1.57 mmol) and potassium carbonate (239 mg, 1.73 mmol) in acetone (6.1 ml) was added water (1 drop). The reacton was stirred overnight at RT. The reaction mixture concentrated and the crude residue was partitioned between water and dichloromethane and the aqueous phase was extracted with dichloromethane. The combined organic phase was dried, concentrated and purified by silica gel column chromatography (eluent: cyclohexane/ethyl acetate) to afford 264 mg (35% of theory, 86% purity) of the title compound. LC-MS (Method 8): Rt = 1.17 min; MS (ESIpos): m/z = 412 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: 1.170 (2.23), 1.187 (4.68), 1.205 (2.29), 1.258 (2.39),
1.276 (4.77), 1.285 (0.44), 1.293 (2.30), 1.398 (16.00), 2.061 (0.89), 3.823 (0.78), 3.894 (0.79),
3.969 (7.19), 4.100 (0.72), 4.118 (2.22), 4.136 (2.18), 4.153 (0.70), 4.196 (0.74), 4.214 (2.23),
4.231 (2.21), 4.249 (0.71), 5.803 (2.14), 5.808 (2.16), 7.193 (1.53), 7.198 (1.59), 7.538 (0.77), 7.561 (1.15), 7.630 (0.84), 7.647 (0.89), 7.669 (0.56), 7.782 (1.62), 7.787 (1.64).
Intermediate 6A
Ethyl 3-(4-chloro-2-fluoro-3-methoxyphenyl)-1 -oxo-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate
Figure imgf000070_0001
To a solution of diethyl 1-[2-(4-chloro-2-fluoro-3-methoxyphenyl)-2-oxoethyl]-1H-pyrrole-2,4- dicarboxylate (Intermediate 18A, 440 mg, 86% purity, 919 pmol) in acetic acid (9.4 ml) was added ammonium acetate (1.42 g, 18.4 mmol). The mixture was heated at 125°C overnight, then the reaction mixture was poured into ice-water. The pH of the solution was adjusted to neutral with sodium hydroxide (concentrated aqueous solution) and the resulting precipitate was collected by filtration and washed with water, dichloromethane and acetonitrile to afford 273 mg (81% of theory, 100% purity) of the title compound.
LC-MS (Method 9): Rt = 1.77 min; MS (ESIpos): m/z = 365 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.149 (0.43), 0.146 (0.43), 1.285 (5.04), 1.302 (9.35), 1.320 (4.93), 2.328 (1.11), 2.367 (0.65), 2.670 (1.09), 2.710 (0.50), 3.941 (16.00), 4.241 (1.86),
4.258 (4.70), 4.275 (4.65), 4.293 (1.72), 7.215 (3.12), 7.310 (1.45), 7.329 (2.72), 7.348 (1.97), 7.437 (3.04), 7.458 (2.24), 7.621 (4.91), 8.057 (4.62), 11.100 (3.38).
Intermediate 20A
3-(4-Chloro-2-fluoro-3-methoxyphenyl)-1-oxo-1,2-dihydropyrrolo[1,2-a]pyrazine-7-carboxylic acid
Figure imgf000071_0001
To a solution of ethyl 3-(4-chloro-2-fluoro-3-methoxyphenyl)-1-oxo-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 19A, 272 mg, 746 pmol) in THF (5.1 ml) was added lithium hydroxide (1.7 ml of a 1.9 M aqueous solution, 3.23 mmol). The reaction was heated to 40°C for 72 h, then the reaction was concentrated to remove the THF and water was added. The pH of the aqueous solution was adjusted to 1 to 2 with hydrochloric acid (1.0 N aqueous solution) and the precipitated was collected by filtration and dried to afford 221 mg (85% of theory, 97% purity) of the title compound.
LC-MS (Method 9): Rt = 1.33 min; MS (ESIpos): m/z = 337 [M+H]+ Ή-NMR (400 MHz, DMSO-d6) d [ppm]: 1.169 (0.46), 1.302 (0.42), 2.328 (0.45), 2.670 (0.41), 3.907 (0.42), 3.941 (16.00), 7.184 (2.73), 7.312 (1.46), 7.333 (2.41), 7.352 (1.99), 7.434 (2.53), 7.455 (1.75), 7.623 (4.47), 7.986 (4.07), 7.990 (4.00), 11.060 (2.37), 12.540 (0.60).
Intermediate 7A
Diethyl 3-cyclopropyl- 1 H-pyrrole-2,4-dicarboxylate
Figure imgf000071_0002
H3C To a solution of ethyl 3-cyclopropyl prop-2-ynoate (45.0 g, 90% purity, 293 mmol) in DMF (400 ml) were added ethyl isocyanoacetate (36.5 g, 322 mmol) and copper(l)oxide (1.90 g, 14.7 mmol). The reaction mixture was stirred overnight at 90°C, then poured into ice-water (1.0 L). The aqueous layer was extracted with ethyl acetate and the combined organic phase was dried over anhydrous sodium sulfate, filtered and concentrated. The crude residue was purified by silica gel column chromatography (120 g silica gel, eluent: petroleum ether/ethyl acetate, 4:1) to afford 25.0 g (34% of theory, 99% purity) of the title product.
LC-MS (Method 10): Rt = 1.62 min, MS (ESIpos): m/z = 252 [M+H]+
1H-NMR (300 MHz, DMSO-de): d [ppm] = 12.04 (br s, 1H), 7.40 (d, 1H), 4.24 (q, 2H), 4.16 (q, 2H), 2.37-2.28 (m, 1H), 1.30 (t, 3H), 1.27 (t, 3H), 0.92-0.88 (m, 4H).
Intermediate 8A
Diethyl 3-cyclopropyl-1-[2-(3,4-dimethylphenyl)-2-oxoethyl]-1 H-pyrrole-2,4-dicarboxylate
Figure imgf000072_0001
To a solution of 2-bromo-1-(3,4-dimethylphenyl)ethan-1-one (5.76 g, 80% purity, 20.3 mmol) in acetone (150 ml) was added diethyl 3-cyclopropyl- 1 H-pyrrole-2,4-dicarboxylate (Intermediate 21A, 3.00 g, 85% purity, 10.1 mmol) and potassium carbonate (5.00 g, 30.4 mmol). The mixture was stirred 2 h at RT, then filtered. The filtrate was concentrated and the crude residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate, 5:2) to afford 5.05 g (28% of theory, 46% purity) of the title compound. LC-MS (Method 11): Rt = 1.95 min, MS (ESIpos): m/z = 398 [M+H]+
1H-NMR (400 MHz, DMSO-cfe): d [ppm] 7.80-7.74 (m, 2H), 7.64 (s, 1H), 7.45-7.26 (m, 1H), 5.82 (s, 2H), 4.19 (q, 2H), 4.08 (q, 2H), 2.33 (s, 6H), 2.30-2.27 (m, 1H), 1.27 (t, 3H), 1.12 (t, 3H), 0.91- 0.84 (m, 2H), 0.69-0.63 (m, 2H).
Intermediate 23A Ethyl 8-cyclopropyl-3-(3,4-dimethylphenyl)-1 -oxo-1 , 2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate To a solution of diethyl 3-cyclopropyl-1-[2-(3,4-dimethylphenyl)-2-oxoethyl]-1H-pyrrole-2,4- dicarboxylate (Intermediate 22A, 3.54 g, 46% purity, 4.05 mmol) in acetic acid (50 ml) was added ammonium acetate (20.6 g, 122 mmol). The mixture was heated overnight at 110°C, then cooled to RT. The reaction mixture was poured into ice-water and the resulting precipitate was collected by filtration, washed with water and dried to afford 1.40 g (93% of theory, 95% purity) of the title compound.
LC-MS (Method 12): Rt = 1.54 min, MS (ESIpos): m/z = 351 [M+H]+
1H-NMR (400 MHz, DMSO-cfe): d [ppm] 10.59 (s, 1H), 7.89 (s, 1H), 7.61 (d, 1H), 7.44 (d, 1H), 7.37- 7.35 (m, 1H), 7.22 (d, 1H), 4.23 (q, 2H), 2.91-2.83 (m, 1H), 2.26 (s, 3H), 2.25 (s, 3H), 1.30 (t, 3H),
1.24-1.20 (m, 2H), 0.89-0.84 (m, 2H).
Intermediate 24A
8-Cyclopropyl-3-(3,4-dimethylphenyl)-1-oxo-1,2-dihydropyrrolo[1,2-a]pyrazine-7-carboxylic acid
Figure imgf000073_0001
To a solution of ethyl 8-cyclopropyl-3-(3,4-dimethylphenyl)-1-oxo-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 23A, 1.40 g, 95% purity, 3.80 mmol) in ethanol (50 ml) and sodium hydroxide (22 ml of a 3.5 M aqueous solution, 77 mmol). After stirring overnight at 40°C the reaction mixture was concentrated under reduced pressure to remove the ethanol. The crude residue was diluted with water and the pH of the mixture was adjusted to 3 to 4 with hydrochloric acid. The resulting precipitate was collected by filtration, washed with water and dried to afford 1.07 g (83% of theory, 95% purity) of the title compound.
LC-MS (Method 13): Rt = 1.34 min, MS (ESIpos): m/z = 323 [M+H]+ 1H-NMR (400 MHz, DMSO-cfe): d [ppm] 12.26 (s, 1H), 10.54 (s, 1H), 7.84 (s, 1H), 7.59 (d, 1H), 7.45 (s, 1H), 7.36 (d, 1H), 7.26-7.20 (m, 1H), 2.99-2.92 (m, 1H), 2.26 (s, 3H), 2.25 (s, 3H), 1.30- 1.26 (m, 2H), 0.88- 0.82 (m, 2H).
Intermediate 25A Diethyl 1-[2-(4-chloro-3-methylphenyl)-2-oxoethyl]-3-cyclopropyl-1 H-pyrrole-2,4-dicarboxylate
Figure imgf000074_0001
To a solution of diethyl 3-cyclopropyl- 1 H-pyrrole-2,4-dicarboxylate (Intermediate 21 A, 2.00 g, 90% purity, 7.16 mmol) in acetone (25 ml) was added potassium carbonate (2.97 g, 21.5 mmol) and 2- bromo-1-(4-chloro-3-methylphenyl)ethan-1-one (2.29 g, 85% purity, 7.88 mmol) at RT. The reaction was stirred overnight, then the mixture was queched with water (20 ml) and extracted with ethyl acetate (2 x 25 ml). The organic phase was washed with brine, dried with anhydrous sodium sulfate and concentrated and the crude product was suspended in ethyl acetate/hexane (20 ml, 1:5) and stirred for 30 min. The solid was collected by filtration and dried to afford 2.80 g (82% of theory, 96% purity) of the title compound LC-MS (Method 14): Rt = 1.41 min, MS (ESIpos): m/z = 418 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 8.03 (d, 1H), 7.87-7.85 (m, 1H), 7.65 (d, 2H), 5.84 (s, 2H), 4.19 (q, 2H), 4.09 (q, 2H), 2.44 (s, 3H), 2.10-2.05 (m, 1H), 1.27 (t, 3H), 1.13 (t, 3H), 0.91-0.86 (m, 2H), 0.66-0.63 (m, 2H).
Intermediate 26A Ethyl 3-(4-chloro-3-methylphenyl)-8-cyclopropyl-1-oxo-1,2-dihydropyrrolo[1,2-a]pyrazine-7- carboxylate
Figure imgf000074_0002
To a solution of diethyl 1-[2-(4-chloro-3-methylphenyl)-2-oxoethyl]-3-cyclopropyl-1H-pyrrole-2,4- dicarboxylate (Intermediate 25A, 2.80 g, 96% purity, 6.43 mmol) in acetic acid (30 ml) was added ammonium acetate (19.8 g, 257 mmol) and the resulting mixture was heated overnight at 110°C. After cooling to RT the reaction mixture was poured into ice-water. The solid was collected by filtration, washed with water and dried to afford 1.80 g (72% of theory, 96% purity) of the title compound.
LC-MS (Method 15): Rt = 1.34 min, MS (ESIpos): m/z = 371 [M+H]+
1H-NMR (400 MHz, DMSO-cfe): d [ppm] d 10.69 (s, 1H), 7.89 (s, 1H), 7.68-7.66 (m, 2H), 7.52-7.46 (m, 2H), 4.23 (q, 2H), 2.89-2.84 (m, 1H), 2.38 (s, 3H), 1.30 (t, 3H), 1.24-1.20 (m, 2H), 0.89-0.85 (m, 2H).
Intermediate 27A
3-(4-Chloro-3-methylphenyl)-8-cyclopropyl-1-oxo-1 ,2-dihydropyrrolo[1,2-a]pyrazine-7- carboxylic acid
Figure imgf000075_0001
To a solution of ethyl 3-(4-chloro-3-methylphenyl)-8-cyclopropyl-1-oxo-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 26A, 1.70 g, 96% purity, 4.40 mmol) in ethanol (30 ml) was added sodium hydroxide (10 ml of a 4.4 M aqueous solution, 44.0 mmol). After stirring overnight at 40°C the reaction mixture was concentrated to remove the ethanol and the mixture was diluted with water (10 ml) and extracted with MTBE (2 x 20 ml). The pH of the aqueous phase was adjusted to 1 by addition of hydrochloric acid (2.0 N) and the product was collected by filtration, washed with water and dried to afford 1.10 g (72% of theory, 99% purity) of the title compound.
LC-MS (Method 11): Rt = 0.90 min, MS (ESIpos): m/z = 343 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 12.31 (s, 1H), 10.64 (s, 1H), 7.85 (s, 1H), 7.67 (t, 2H), 7.52-7.46 (m, 2H), 3.00-2.90 (m, 1H), 2.38 (s, 3H), 1.33-1.28 (m, 2H), 0.88-0.82 (m, 2H). Intermediate 28A
Diethyl 1-[2-(3-chloro-4-methylphenyl)-2-oxoethyl]-3-cyclopropyl-1 H-pyrrole-2,4-dicarboxylate To a solution of 2-bromo-1-(3-chloro-4-methylphenyl)ethan-1-one (2.20 g, 95% purity, 8.44 mmol) in acetone (30 ml) was added potassium carbonate (3.18 g, 23.0 mmol) and diethyl 3-cyclopropyl- 1 H-pyrrole-2,4-dicarboxylate (Intermediate 21 A, 1.95 g, 99% purity, 7.68 mmol) at RT. After stirring 2 h at room temperature the solids was filtered off and the filtrate was evaporated. The residue was suspended in ethyl acetate/n-hexane (80 ml, 1:20) and stirred for 30 min, then the precipitate was collected by filtration and dried to afford give 2.20 g (67% of theory, 98% purity) of the title compound.
LC-MS (Method 21): Rt = 2.09 min, MS (ESIpos): m/z = 418 [M+H]+ 1H-NMR (300 MHz, DMSO-cfe): d [ppm] 8.04 (d, 1H), 7.90 (dd, 1H), 7.64-7.59 (m, 2H), 5.85 (s, 2H), 4.19 (q, 2H), 4.10 (q, 2H), 2.43 (s, 3H), 2.10-2.05 (m, 1H), 1.27 (t, 3H), 1.13 (t, 3H), 0.92-0.86 (m, 2H), 0.68-0.64 (m, 2H).
Intermediate 29A
Ethyl 3-(3-chloro-4-methylphenyl)-8-cyclopropyl-1-oxo-1,2-dihydropyrrolo[1,2-a]pyrazine-7- carboxylate
Figure imgf000076_0001
To a solution of diethyl 1-[2-(3-chloro-4-methylphenyl)-2-oxoethyl]-3-cyclopropyl-1H-pyrrole-2,4- dicarboxylate (Intermediate 28A, 2.20 g, 99% purity, 5.21 mmol) in acetic acid (120 ml) was added ammonium acetate (12.0 g, 156 mmol) and the resulting mixture was heated overnight at 110°C. After cooling to RT the reaction mixture was poured into ice-water. The solid was collected by filtration, washed with water and dried to afford 1.74 g (85% of theory, 95% purity) of the title compound.
LC-MS (Method 13): Rt = 1.99 min, MS (ESIpos): m/z = 371 [M+H]+ 1H-NMR (300 MHz, DMSO-cfe): d [ppm] 10.74 (s, 1H), 7.89 (s, 1H), 7.72 (s, 2H), 7.54-7.44 (m, 2H), 4.25-4.22 ( , 2H), 2.87 (d, 1H), 2.37 (s, 3H), 1.32-1.22 ( , 5H), 0.89-0.85 ( , 2H).
Intermediate 30A
3-(3-Chloro-4-methylphenyl)-8-cyclopropyl-1-oxo-1 ,2-dihydropyrrolo[1,2-a]pyrazine-7- carboxylic acid
Figure imgf000077_0001
To a solution of ethyl 3-(3-chloro-4-methylphenyl)-8-cyclopropyl-1-oxo-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 29A, 1.64 g, 95% purity, 4.20 mmol) in ethanol (30 ml) was added sodium hydroxide (15 ml of a 2.8 M aqueous solution, 42.0 mmol). After stirring overnight at 40°C the reaction mixture was concentrated to remove the ethanol and the mixture was diluted with water (10 ml) and extracted with MTBE (2 x 20 ml). The pH of the aqueous phase was adjusted to 4 by addition of hydrochloric acid (4.0 N) and the product was collected by filtration, washed with water and dried to afford 1.38 g (94% of theory, 98% purity) of the title compound.
LC-MS (Method 11): Rt = 0.89 min, MS (ESIpos): m/z = 343 [M+H]+ 1H-NMR (300 MHz, DMSO-cfe): d [ppm] 12.32 (s, 1H), 10.67 (s, 1H), 7.83 (s, 1H), 7.80-7.70 (m, 2H), 7.52 (dd, 1H), 7.43 (d, 1H), 3.00-2.90 (m, 1H), 2.36 (s, 3H), 1.31-1.23 (m, 2H), 0.89-0.84 (m,
2H).
Intermediate 31A
Diethyl 3-cyclopropyl- 1-[2-(3-fluoro-4-methylphenyl)-2-oxoethyl]-1H-pyrrole-2,4-dicarboxylate
Figure imgf000077_0002
To a solution of 2-bromo-1-(3-fluoro-4-methylphenyl)ethan-1-one (3.59 g, 80% purity, 12.4 mmol) in acetone (50 ml) was added potassium carbonate (5.62 g, 18.6 mmol) and diethyl 3-cyclopropyl- 1 H-pyrrole-2,4-dicarboxylate (Intermediate 21 A, 1.56 g, 6.21 mmol) at RT. The reaction was stirred for 2 h, then filtered and the filtrate concentrated. The crude residue was suspended in ethyl acetate/petroleum ether (200 ml, 1:20) and stirred for 30 min. The solid was collected by filtration and dried to afford 2.28 g (72% of theory, 79% purity) of the title compound.
LC-MS (Method 16): Rt = 1.78 min, MS (ESIpos): m/z = 402 [M+H]+ Intermediate 32A
Ethyl 8-cyclopropyl-3-(3-fluoro-4-methylphenyl)-1 -oxo-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate
Figure imgf000078_0001
To a solution of diethyl 3-cyclopropyl-1-[2-(3-fluoro-4-methylphenyl)-2-oxoethyl]-1H-pyrrole-2,4- dicarboxylate (Intermediate 31A, 670 mg, 79% purity, 1.32 mmol) in acetic acid (10 ml) was added ammonium acetate (3.05 g, 39.6 mmol) and the resulting mixture was heated overnight at 110°C. After cooling to RT the reaction mixture was poured into ice-water. The solid was collected by filtration, washed with water and dried to afford 220 mg (40% of theory, 86% purity) of the title compound. 1H-NMR (300 MHz, DMSO-cfe): d [ppm] 10.67 (s, 1H), 7.86 (s, 1H), 7.68 (d, 1H), 7.46-7.33 (m, 3H), 4.24-4.17 (m, 2H), 2.88-2.79 (m, 1H), 2.19 (s, 3H), 1.30-1.17 (m, 5H), 0.88-0.82 (m, 2H).
Intermediate 33A
8-Cyclopropyl-3-(3-fluoro-4-methylphenyl)-1 -oxo-1, 2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylic acid
Figure imgf000078_0002
To a solution of ethyl 8-cyclopropyl-3-(3-fluoro-4-methylphenyl)-1-oxo-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 32A, 220 mg, 86% purity, 534 pmol) in ethanol (10 ml) was added sodium hydroxide (13 ml of a 0.8 M aqueous solution, 10.4 mmol). After stirring for 2 h at RT the reaction mixture was concentrated to remove the ethanol and the mixture was diluted with water (100 ml) and the pH of the aqueous phase was adjusted to 3 to 4 by addition of hydrochloric acid (2.0 N) and the product was collected by filtration, washed with water and dried to afford 173 g (93% of theory, 94% purity) of the title compound.
LC-MS (Method 11): Rt = 0.84 min, MS (ESIpos): m/z = 327 [M+H]+ 1H-NMR (300 MHz, DMSO-cfe): d [ppm] d 12.29 (s, 1H), 10.61 (s, 1H), 7.89 (s, 1H), 7.52-7.46 (m, 2H), 7.39-7.37 (m, 1H), 7.27-7.21 (m, 1H), 2.99-2.90 (m, 1H), 2.28 (d, 3H), 1.30-1.20 (m, 2H), 0.88-0.81 (m, 2H).
Intermediate 34A
Diethyl 1-[2-(4-chloro-3-fluorophenyl)-2-oxoethyl]-3-cyclopropyl-1 H-pyrrole-2,4-dicarboxylate
Figure imgf000079_0001
To a solution of 2-bromo-1-(4-chloro-3-fluorophenyl)ethan-1-one (330 mg, 1.31 mmol) in acetone (11 ml) was added potassium carbonate (412 mg, 2.98 mmol) and diethyl 3-cyclopropyl-1H- pyrrole-2,4-dicarboxylate (Intermediate 21 A, 300 mg, 1.19 mmol) at RT. The reaction was stirred for 72 h, then 2-bromo-1-(4-chloro-3-fluorophenyl)ethan-1-one (165 mg, 0.655 mmol) and (206 mg, 1.49 mmol) was added and the reaction was stirred overnight at RT. The reaction was filtered, and the filtrate concentrated. The crude residue was purified by silica gel column chromatography (eluent: cyclohexane/ethyl acetate) to afford 447 mg (50% of theory, 56% purity) of the title compound.
LC-MS (Method 9): Rt = 2.35 min; MS (ESIpos): m/z = 422 [M+H]+ Intermediate 35A
Ethyl 3-(4-chloro-3-fluorophenyl)-8-cyclopropyl-1-oxo-1,2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate
Figure imgf000079_0002
To a solution of diethyl 1-[2-(4-chloro-3-fluorophenyl)-2-oxoethyl]-3-cyclopropyl-1H-pyrrole-2,4- dicarboxylate (Intermediate 34A, 447 mg, 56% purity, 598 pmol) in acetic acid (5.0 ml) was added ammonium acetate (461 mg, 5.98 mmol) and the resulting mixture was heated for 72 h at 110°C. After cooling to RT the reaction mixture was poured into ice-water (50 ml). The solid was collected by filtration, washed with water and dried to afford 106 mg g (47% of theory, 100% purity) of the title compound
LC-MS (Method 9): Rt = 2.14 min; MS (ESIpos): m/z = 375 [M+H]+
Intermediate 36A
3-(4-Chloro-3-fluorophenyl)-8-cyclopropyl-1-oxo-1,2-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid
Figure imgf000080_0001
To a solution of ethyl 3-(4-chloro-3-fluorophenyl)-8-cyclopropyl-1-oxo-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 35A, 106 mg, 283 pmol) in THF/methanol (2.0 ml, 5:1) was added lithium hydroxide (1.4 ml of a 1.0 M aqueous solution, 1.4 mmol). After stirring for 6 days at 60°C the reaction mixture was concentrated to remove the organic solvent. The pH of the aqueous phase was adjusted to 3 to 4 by addition of hydrochloric acid (1.0 N) and the product was collected by filtration, washed with water and dried to afford 115 mg (82% of theory, 70% purity) of the title compound.
LC-MS (Method 9): Rt = 1.61 min; MS (ESIneg): m/z = 345 [M-H] Intermediate 37A
Ethyl (2Z)-2-[(dimethylamino)methylidene]-4,4,4-trifluoro-3-oxobutanoate
Figure imgf000080_0002
To a solution of triethylamine (311 g, 3.07 mol) in toluene (1.0 L) were added trifluoroacetic acid (100 g, 877 mmol) and ethyl (2Z)-3-(dimethylamino)prop-2-enoate (127 g, 886 mmol) at RT. After stirring at RT for 30 min, pivaloyl chloride (242 g, 2.02 mol) was added at 0°C and the reaction was stirred at RT for 6 h. The reaction mixture was then concentrated and the residue was diluted with water (3.0 L) and extracted with ethyl acetate (2 x 1.0 L). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate, 4:1) to afford 150 g (96% of theory, 80% purity) of the title compound.
LC-MS (Method 17): Rt = 1.18 min; MS (ESIpos): m/z = 240 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] = 7.84 (s, 1H), 4.08 (q, 2H), 3.38 (s, 3H), 2.83 (s, 3H), 1.20 (t, 3H).
Intermediate 9A Ethyl 2-{[(2-ethoxy-2-oxoethyl)amino]methylidene}-4,4,4-trifluoro-3-oxobutanoate
Figure imgf000081_0001
To a solution of ethyl (2Z)-2-[(dimethylamino)methylidene]-4,4,4-trifluoro-3-oxobutanoate (Intermediate 37A, 150 g, 90% purity, 564 mmol) in ethanol (800 ml) was added ethyl glycinate hydrochloride (64.0 g, 621 mmol) at RT. After stirring at 85°C for 2 h, the reaction mixture was concentrated and the residue was diluted with water (1.0 L) and extracted with ethyl acetate (3 x 800 ml). The combined organic phases were dried over anhydrous sodium sulfate and concentrated to afford 180 g (91% of theory, 85% purity) of the title compound which was used without purification.
LC-MS (Method 17): Rt = 1.32 min; MS (ESIpos): m/z = 298 [M+H]+ Intermediate 39A
Diethyl 3-(trifluoromethyl)-1 H-pyrrole-2,4-dicarboxylate
Figure imgf000081_0002
To a solution of ethyl 2-{[(2-ethoxy-2-oxoethyl)amino]methylidene}-4,4,4-trifluoro-3-oxobutanoate (Intermediate 38A, 46.5 g, 85% purity, 133 mmol) in ethanol (390 ml) and toluene (200 ml) was added ethyl 4,4,4-trifluoroacetoacetate (20 ml, 130 mmol) under nitrogen at RT. A solution of potassium te/f-butoxide (21.1 g, 293 mmol) in THF (320 ml) was added dropwise at RT over 30 min. After stirring overnight at 63°C the reaction was concentrated and the residue was dissolved in ethyl acetate (400 ml) and washed with water (300 ml). The organic phase was dried with anhydrous sodium sulfate and concentrated. The crude residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acatate, 5:1) to afford 15.0 g (40% of theory, 98% purity) of the title compound.
LC-MS (Method 18): Rt = 2.68 min; MS (ESIpos): m/z = 302 [M+Na]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] = 13.00 (br s, 1H), 7.60 (s, 1H), 4.29 (q, 2H), 4.19 (q, 2H), 1.28 (t, 3H), 1.24 (t, 3H). Intermediate 40A
Diethyl 1-[2-(3-fluoro-4-methylphenyl)-2-oxoethyl]-3-(trifluoromethyl)-1 H-pyrrole-2,4- dicarboxylate
Figure imgf000082_0001
To a solution of 2-bromo-1-(3-fluoro-4-methylphenyl)ethan-1-one (1.89 g, 78% purity, 6.38 mmol) in acetone (100 ml) was added diethyl 3-(trifluoromethyl)-1H-pyrrole-2,4-dicarboxylate (Intermediate 39A, 1.50 g, 99% purity, 5.32 mmol) and potassium carbonate (2.20 g, 16.0 mmol). The mixture was stirred overnight at RT, then filtered and the filtrate was concentrated under reduced pressure to provide the crude product. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate, 5:2) to give 1.60 g (63% of theory, 90% purity) of the title compound.
LC-MS (Method 19): Rt = 1.95 min; MS (ESIpos): m/z = 430 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] = 7.82-7.76 (m, 3H), 7.58-7.53 (m, 1H), 5.91 (s, 2H), 4.28- 4.11 (m, 4H), 2.36 (d, 3H), 1.27 (t, 3H), 1.11 (t, 3H).
Intermediate 41A Ethyl 3-(3-fluoro-4-methylphenyl)-1-oxo-8-(trifluoromethyl)-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate To a solution of diethyl 1-[2-(3-fluoro-4-methylphenyl)-2-oxoethyl]-3-(trifluoromethyl)-1H-pyrrole- 2,4-dicarboxylate (Intermediate 40A, 2.10 g, 90% purity, 4.40 mmol) in acetic acid (150 ml) was added ammonium acetate (10.2 g, 132 mmol). The resulting mixture was heated overnight at 110°C. After cooling to RT, the reaction mixture was poured into ice-water. The solid was collected by filtration, washed with water and dried to afford 1.60 g (90% of theory, 95% purity) of the title compound.
LC-MS (Method 20): Rt = 1.61 min; MS (ESIpos): m/z = 383 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] = 11.25 (s, 1H), 8.05 (s, 1H), 7.87 (s, 1H), 7.52-7.39 (m, 3H), 4.29 (q, 2H), 2.29 (d, 3H), 1.30 (t, 3H).
Intermediate 42A
3-(3-Fluoro-4-methylphenyl)-1-oxo-8-(trifluoromethyl)-1 ,2-dihydropyrrolo[1,2-a]pyrazine-7- carboxylic acid
Figure imgf000083_0001
To a solution of ethyl 3-(3-fluoro-4-methylphenyl)-1-oxo-8-(trifluoromethyl)-1 ,2-dihydropyrrolo[1 ,2- a]pyrazine-7-carboxylate (Intermediate 41 A, 1.60 g, 90% purity, 3.77 mmol) in ethanol (40 ml) was added sodium hydroxide (11 ml of a 3.4 M aqueous solution, 37.67 mmol). After stirring at RT for 2 h, the reaction mixture was concentrated to remove the ethanol, then the water (100 ml) was added and the aquoues solution was extracted with MTBE (2 x 100 ml). The aqueous phase was acidified with hydrochloric acid (2.0 N aquoues solution) to adjust the pH value to 1. The product was collected by filtration, washed with water and dried to afford 1.35 g (87% of theory, 86% purity) of the title compound.
LC-MS (Method 11): Rt = 0.78 min; MS (ESIpos): m/z = 355 [M+H]+ 1H-NMR (300 MHz, DMSO-cfe): d [ppm] 13.01 (s, 1H), 11.37 (s, 1H), 7.99 (s, 1H), 7.89 (s, 1H), 7.54-7.37 (m, 3H), 2.28 (d, 3H).
Intermediate 43A
Diethyl 1-[2-(3,4-di ethylphenyl)-2-oxoethyl]-3-(trifluoro ethyl)-1 H-pyrrole-2,4-dicarboxylate
Figure imgf000084_0001
To a solution of diethyl 3-(trifluoromethyl)-1H-pyrrole-2,4-dicarboxylate (Intermediate 39A, 801 mg, 95% purity, 2.72 mmol) in acetone (15 ml) was added 2-bromo-1-(3,4-dimethylphenyl)ethan-1-one (1.16 g, 80% purity, 4.09 mmol) and potassium carbonate (1.13 g, 8.17 mmol). The mixture was stirred 2 h at RT. The reaction was filtered and the filtrate was concentrated under reduced pressure. The crude residue was suspended in ethyl acetate/petroleum ether (100 ml, 1:20) and stirred for 30 min. The solid was collected by filtrated to afford 1.10 g (90% of theory, 95% purity) of the title compound.
LC-MS (Method 19): Rt = 1.97 min; MS (ESIpos): m/z = 426 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 7.81-7.75 (m, 3H), 7.38 (d, 1H), 5.90 (s, 2H), 4.25 (q, 2H), 4.16 (q, 2H), 2.33 (s, 6H), 1.27 (t, 3H), 1.10 (t, 3H).
Intermediate 44A
Ethyl 3-(3,4-dimethylphenyl)-1-oxo-8-(trifluoromethyl)-1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate
Figure imgf000084_0002
To a solution of diethyl 1-[2-(3,4-dimethylphenyl)-2-oxoethyl]-3-(trifluoromethyl)-1H-pyrrole-2,4- dicarboxylate (Intermediate 43A, 1.10 g, 95% purity, 2.46 mmol) in acetic acid (30 ml) was added ammonium acetate (5.67 g, 73.7 mmol). The resulting mixture was heated overnight at 110°C. After cooling to RT, the reaction mixture was poured into ice-water. The solid was collected by filtration, washed with water and dried to afford 800 mg (82% of theory, 95% purity) of the title compound.
LC-MS (Method 11): Rt = 1.62 min; MS (ESIpos): m/z = 442 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 9.58 (s, 1H), 8.05 (s, 1H), 7.78 (s, 1H), 7.47 (d, 1H), 7.41- 7.38 (m, 1H), 7.26 (d, 1H), 4.29 (q, 2H), 2.28 (s, 3H), 2.27 (s, 3H), 1.30 (t, 3H).
Intermediate 45A
3-(3,4-Dimethylphenyl)-1-oxo-8-(trifluoromethyl)-1,2-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxylic acid
Figure imgf000085_0001
To a solution of ethyl 3-(3,4-dimethylphenyl)-1-oxo-8-(trifluoromethyl)-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxylate (Intermediate 44A, 1.00 g, 99% purity, 2.62 mmol) in ethanol (25 ml) was added sodium hydroxide (7.5 ml of a 3.5 M aqueous solution, 26.2 mmol). After stirring at RT for 2 h the reaction mixture was concentrated to remove ethanol. Water (50 ml) was added and the aqueous mixture was extracted with MTBE (2 x 50 ml). The aqueous phase was acidified with hydrochloric acid (2.0 N) to adjust the pH value to 1 and the product was collected by filtration, washed with water and dried to afford 0.43 g (41% of theory, 87% purity) of the title compound.
LC-MS (Method 11): Rt = 0.82 min; MS (ESIpos): m/z = 351 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 10.81 (s, 1H), 7.68 (s, 1H), 7.49 (s, 1H), 7.42-7.40 (m, 1H), 7.41 (d, 1H), 7.29 (s, 1H), 7.21 (d, 1H), 2.262.25 (s, 3H). Intermediate 46A
Diethyl 1-[2-(3-chloro-4-methylphenyl)-2-oxoethyl]-3-(trifluoromethyl)-1H-pyrrole-2,4- dicarboxylate
Figure imgf000085_0002
To a solution of 2-bromo-1-(3-chloro-4-methylphenyl)ethan-1-one (2.00 g, 95% purity, 7.68 mmol) in acetone (30 ml) was added diethyl 3-(trifluoromethyl)-1H-pyrrole-2,4-dicarboxylate (Intermediate 39A, 2.05 g, 95% purity, 6.98 mmol) and potassium carbonate (2.89 g, 20.9 mmol) at RT. After stirring 2 h the solids was filtered and the filtrate was evaporated under reduced pressure. The residue was suspended in ethyl acetate/n-hexane (60 ml, 1:20) and stirred for 30 min. The solid was collected by filtration and dried to afford 2.70 g (78% of theory, 99% purity) of the title compound.
LC-MS (Method 21): Rt = 2.05 min; MS (ESIpos): m/z = 468 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 8.05 (d, 1H), 7.91 (dd, 1H), 7.80 (s, 1H), 7.62 (d, 1H), 5.93 (s, 2H), 4.28-4.13 (m, 4H), 2.45 (s, 3H), 1.27 (t, 3H), 1.11 (t, 3H).
Intermediate 47A
Ethyl 3-(3-chloro-4-methylphenyl)-1-oxo-8-(trifluoromethyl)-1,2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate
Figure imgf000086_0001
To a solution of diethyl 1-[2-(3-chloro-4-methylphenyl)-2-oxoethyl]-3-(trifluoromethyl)-1H-pyrrole- 2,4-dicarboxylate (Intermediate 46A, 2.80 g, 99% purity, 6.22 mmol) in acetic acid (150 ml) was added ammonium acetate (14.4 g, 187 mmol). The resulting mixture was stirred overnight at 110°C. After cooling to RT, the reaction mixture was poured into ice-water and the solid was collected by filtration, washed with water and dried to afford 2.50 g (99% of theory, 99% purity) of the title compound.
LC-MS (Method 21): Rt = 1.88 min; MS (ESIpos): m/z = 399 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 11.44 (s, 1H), 8.02 (s, 1H), 7.86 (s, 1H), 7.74 (d, 1H), 7.53 (dd, 1H), 7.46 (d, 1H), 4.28 (q, 2H), 2.37 (s, 3H), 1.30 (t, 3H).
Intermediate 48A 3-(3-Chloro-4-methylphenyl)-1-oxo-8-(trifluoromethyl)-1,2-dihydropyrrolo[1,2-a]pyrazine-7- carboxylic acid To a solution of ethyl 3-(3-chloro-4-methylphenyl)-1-oxo-8-(trifluoromethyl)-1 ,2-dihydropyrrolo[1 ,2- a]pyrazine-7-carboxylate (Intermediate 47A, 2.40 g, 99% purity, 5.96 mmol) in ethanol (60 ml) was added sodium hydroxide (20 ml of a 3.0 M aqueous solution, 60.0 mmol). After stirring at RT for 2 h the reaction mixture was concentrated under reduced pressure. The aqueous phase was acidified with hydrochloric acid (4.0 N) to adjust the pH value to 4 and the product was collected by filtration, washed with water and dried to afford 2.18 g (96% of theory, 97% purity) of the title compound.
LC-MS (Method 21): Rt = 1.55 min; MS (ESIpos): m/z = 371 [M+H]+ 1H-NMR (300 MHz, DMSO-cfe): d [ppm] 13.01 (s, 1H), 11.40 (s, 1H), 8.11 (s, 1H), 7.99 (s, 1H), 7.75 (dd, 1H), 7.55 (d, 1H), 7.47 (d, 1H), 2.37 (s, 3H).
Intermediate 49A
Diethyl 1-[2-(4-chloro-3-methylphenyl)-2-oxoethyl]-3-(trifluoromethyl)-1H-pyrrole-2,4- dicarboxylate
Figure imgf000087_0001
To a solution of 2-bromo-1-(4-chloro-3-methylphenyl)ethan-1-one (1.85 g, 90% purity, 6.74 mmol) in acetone (25 ml) was added diethyl 3-(trifluoromethyl)-1H-pyrrole-2,4-dicarboxylate (Intermediate 39A, 1.80 g, 95% purity, 6.12 mmol) and potassium carbonate (2.54 g, 18.37 mmol). The mixture was stirred for 2 h at RT, then filtered and the filtrate was concentrated. The crude residue was suspended in ethyl acetate/petroleum ether (80 ml, 1 :20) and stirred for 30 min, then the solid was collected by filtration to give 2.50 g (89% of theory, 97% purity) of the title compound.
LC-MS (Method 19): Rt = 2.08 min; MS (ESIpos): m/z = 446 [M+H]+ 1H-NMR (300 MHz, DMSO-cfe): d [ppm] 8.04 (d, 1H), 7.87 (dd,1H), 7.81 (s, 1H), 7.68 (d, 1H), 5.93 (s, 2H), 4.24 (q, 2H), 4.17 (q, 2H), 2.45 (s, 3H), 1.27 (t, 3H), 1.11 (t, 3H).
Intermediate 50A
Ethyl 3-(4-chloro-3-methylphenyl)-1-oxo-8-(trifluoromethyl)-1,2-dihydropyrrolo[1 ,2-a]pyrazine-7- carboxylate
Figure imgf000088_0001
To a solution of diethyl 1-[2-(4-chloro-3-methylphenyl)-2-oxoethyl]-3-(trifluoromethyl)-1H-pyrrole- 2,4-dicarboxylate (Intermediate 49A, 2.50 g, 97% purity, 5.44 mmol) in acetic acid (120 ml) was added ammonium acetate (12.6 g, 163 mmol). The resulting mixture was stirred overnight at 110°C. After cooling to RT the reaction mixture was poured into ice-water. The solid was collected by filtration, washed with water and dried to afford 2.10 g (96% of theory, 99% purity) of the title compound.
LC-MS (Method 21): Rt = 1.87 min; MS (ESIpos): m/z = 399 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 11.31 (s, 1H), 8.05 (s, 1H), 7.83 (s, 1H), 7.68 (d, 1H), 7.56- 7.48 (m, 2H), 4.28 (q, 2H), 2.39 (s, 3H), 1.30 (t, 3H).
Intermediate 51A
3-(4-Chloro-3-methylphenyl)-1-oxo-8-(trifluoromethyl)-1,2-dihydropyrrolo[1,2-a]pyrazine-7- carboxylic acid
Figure imgf000088_0002
To a solution of ethyl 3-(4-chloro-3-methylphenyl)-1-oxo-8-(trifluoromethyl)-1 ,2-dihydropyrrolo[1 ,2- a]pyrazine-7-carboxylate (Intermediate 50A, 2.10 g, 99% purity, 5.21 mmol) in ethanol (45 ml) was added sodium hydroxide (15 ml of a 3.5 M aqueous solution, 52.0 mmol). After stirring at RT for 2 h, the reaction mixture was concentrated to remove ethanol, then water (150 ml) was added. The aqueous phase was acidified with hydrochloric acid (2.0 N) to adjust the pH value to 1 and the product was collected by filtration, washed with water and dried to afford 1.89 g (94% of theory, 96% purity) of the title compound.
LC-MS (Method 18): Rt = 2.71 min; MS (ESIpos): m/z = 371 [M+H]+
1H-NMR (300 MHz, DMSO-cfe): d [ppm] 13.03 (s, 1H), 11.36(s, 1H), 8.00 (s, 1H), 7.82 (s, 1H), 7.68 (d, 1 H), 7.54-7.49 (m, 2H), 2.38 (s, 3H).
Intermediate 52A
1-[2-(2,3-Dihydro-1,4-benzodioxin-6-yl)-1-fluoro-2-oxo-ethyl]-N4-[(1R)-3,3,3-trifluoro-1-methyl- propyl]pyrrole-2, 4-dicarboxamide
Figure imgf000089_0001
To a solution of 3-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-oxo-/\/-[(2R)-4,4,4-trifluorobutan-2-yl]-1,2- dihydropyrrolo[1,2-a]pyrazine-7-carboxamide (Example 1-36, 52.0 mg, 91% purity, 112 pmol) in dry acetonitrile (1.1 ml) was added A/,/\/-diisopropylethylamine (49 pi, 280 pmol) and Selectfluor® (104 mg, 95% purity, 279 pmol). The reaction was stirred at RT for 30 min, then further Selectfluor® (52 mg, 95% purity, 140 pmol) and A/,/\/-diisopropylethylamine (25 pi, 140 pmol) were added and the reaction was stirred for 15 min. The reaction was quenched with sodium thiosulfate (10% w/w solution) and the mixture was partitioned between ethyl acetate and water. The aqueous phase was extracted two times with ethyl acetate and the combined organic phase was washed with brine, dried and concentrated to afford 72.7 mg (quantitative, 70% purity) of the title compound, which was used without further purification. LC-MS (Mehtod 9): Rt = 1.36 min; MS (ESIpos): m/z = 458 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.149 (0.42), -0.008 (3.46), 0.008 (2.99), 0.834 (0.69), 0.852 (1.18), 0.870 (0.59), 1.092 (0.99), 1.108 (1.04), 1.157 (2.30), 1.175 (4.58), 1.194 (5.74),
1.205 (6.26), 1.211 (7.15), 1.221 (6.51), 1.243 (11.23), 1.259 (11.46), 1.273 (7.15), 1.988 (6.94),
2.327 (0.62), 2.366 (0.62), 2.431 (0.45), 2.459 (0.91), 2.523 (2.56), 2.670 (0.74), 2.710 (0.69), 3.128 (0.86), 3.139 (0.88), 3.146 (0.88), 3.157 (0.86), 3.359 (0.62), 3.374 (0.42), 3.600 (0.51),
3.609 (0.56), 3.616 (0.72), 3.626 (0.72), 3.633 (0.54), 3.642 (0.51), 4.003 (0.58), 4.021 (1.68),
4.038 (1.71), 4.056 (0.54), 4.227 (0.45), 4.257 (8.24), 4.266 (16.00), 4.288 (1.79), 4.304 (1.14), 4.320 (0.86), 6.068 (0.77), 6.083 (0.77), 6.202 (0.77), 6.217 (0.72), 6.825 (1.18), 6.847 (1.90),
6.896 (2.27), 6.909 (0.50), 6.919 (2.83), 6.982 (0.59), 7.084 (1.92), 7.097 (3.17), 7.152 (1.92),
7.159 (1.95), 7.271 (1.34), 7.283 (1.84), 7.317 (0.56), 7.338 (0.82), 7.430 (1.10), 7.739 (0.74),
7.749 (0.85), 7.762 (3.09), 8.136 (1.55), 8.156 (1.90), 8.189 (0.48), 8.465 (0.62), 8.823 (2.61). Intermediate 53A
3-(2,3-Dihydro-1,4-benzodioxin-6-yl)-4-fluoro-3-methoxy-1-oxo-/\/-[(1R)-3,3,3-trifluoro-1-methyl- propyl]-2,4-dihydropyrrolo[1,2-a]pyrazine-7-carboxamide
Figure imgf000090_0001
To a solution of 1-[2-(2,3-dihydro-1,4-benzodioxin-6-yl)-1-fluoro-2-oxoethyl]-/\/4-[(2R)-4,4,4- trifluorobutan-2-yl]-1H-pyrrole-2, 4-dicarboxamide (Intermediate 52A, 71.0 mg, 70% purity, 109 pmol)in ethanol (5.4 ml) was added Amberlyst® 15 (74.2 mg) and magnesium sulfate (144 mg, 1.20 mmol) and the reaction was stirred for 2 h at RT. The mixture was filtered over diatomaceous earth, washed with ethanol and concentrated. The crude residue was purified by silica gel column chromatography (eluent: cyclohexane/ethyl acetate, gradient 0-100%) to afford 21.2 mg (33% of theory, 82% purity) of the title compound.
LC-MS (Method 9): Rt = 1.59 min; MS (ESIneg): m/z = 438 [M-H]-
Intermediate 54A
(S)-/\/-[(4-Fluorophenyl)methylene]-2-methyl-propane-2-sulfinamide
Figure imgf000090_0002
To a solution of 4-fluorobenzaldehyde (30 g, 242 mmol) and copper(ll) sulfate (116 g, 725 mmol) in dichloromethane (300 ml), were added 2-methylpropane-2-sulfinamide (29.3 g, 242 mmol) and PTSA (1.50 g, 8.70 mmol) and the mixture was stirred at 30°C for 60 h under nitrogen atmosphere. The reaction mixture was filtered and the filter cake was washed with dichloromethane. The filtrate was washed with sodium bicarbonate solution (120 ml of a saturated aqueous solution) and brine (50 ml), dried over anhydrous sodium sulfate, filtered and concentrated to afford (44.5 g, 195.7 mmol, 81% of theory) the title compound.
1H-NMR (400 MHz, CHC -d): d [ppm] = 8.55 (s, 1H), 7.87 (m, 2H), 7.17 (m, 2H), 1.69 (s, 1H), 1.27 (s, 9H).
Intermediate 55A
Ethyl (3S)-3-[[(S)-te/f-butylsulfinyl]amino]-2,2-difluoro-3-(4-fluorophenyl)propanoate
Figure imgf000091_0001
A suspension of zinc (153 g, 2.30 mol) in THF (600 ml) was stirred in 30°C at nitrogen atmosphere. Chloro(trimethyl)silane (6.80 g, 63.0 mmol) was added and the mixture was stirred for 15 min. Then ethyl 2-bromo-2,2-difluoro-acetate (237 g, 1.17 mol, 150 ml) in THF (700 ml) was added dropwise and the temperature of the reaction was kept below 50°C. The mixture was stirred and cooled to RT and (S)-/\/-[(4-fluorophenyl)methylene]-2-methyl-propane-2-sulfinamide (Intermediate 54A, 53.0 g, 233 mmol) in THF (200 ml) was added. The mixture was stirred at 30°C for 20 h under nitrogen atmosphere. The mixture was filtered and the filtrate was concentrated, quenched with ammonium chloride (1.6 L of a saturated aqueous solution), washed with brine (1.0 L) and dried with anhydrous sodium sulfate. The mixture was purified by silica gel column chromatography (eluent: gradient of ethyl acetate in cyclohexane). The compound was purified for second time by silica gel column chromatography (eluent: gradient of ethyl acetate in cyclohexane), to afford 47.7 g (58% of theory) of the title compound.
1H-NMR (400 MHz, CHC -d) d [ppm] = 7.37 (m, 2H), 7.06-7.10 (m, 2H), 5.01-4.94 (dd, 1H), 4.4 (m, 1H), 4.26 (m, 2H), 1.29 (dd, 3H), 1.24 (s, 9H).
Intermediate 56A
Ethyl (3S)-3-amino-2,2-difluoro-3-(4-fluorophenyl)propanoate hydrochloride To a solution of ethyl (3S)-3-[[(S)-te/f-butylsulfinyl]amino]-2,2-difluoro-3-(4-fluorophenyl) propanoate (Intermediate 55A, 46.0 g, 131 mmol) in ethanol (300 ml) was added hydrochloric acid (350 ml of a 4.0 M solution in 1 ,4-dioxane) and the mixture was stirred at 30°C for 2 h. The mixture was concentrated then MTBE (500 ml) was added and the suspension was stirred for 1 hour and filtered to give the title compound (29.3 g, 102.3 mmol, 78% of theory, 99% purity, 91.9% ee) as solid. From this, a part was recrystallized (14.2 g, 50.1 mmol) from a mixture of ethyl acetate/methanol (1.5:1, 125 ml) to give ethyl (3S)-3-amino-2,2-difluoro-3-(4- fluorophenyl)propanoate (3.28 g, 11.6 mmol, 23% of theory, 96.7% ee) as solid. 1H-NMR (400 MHz, DMSO-de): d [ppm] = 9.42-9.38 (br s, 2H), 7.61 (br s, 2H), 7.39-7.33 (m, 2H), 5.36-5.29 (m, 1H), 4.24-4.19 (dd, 2H), 1.12 (dd, 3H).
Intermediate 57A
(3S)-3-Amino-2,2-difluoro-3-(4-fluorophenyl)propan-1-ol
Figure imgf000092_0001
To a suspension of lithium borohydride (1.43 g, 65.6 mmol) in THF (100 ml) at 0°C, was added ethyl (3S)-3-amino-2,2-difluoro-3-(4-fluorophenyl)propanoate (Intermediate 56A, 6.20 g, 21.8 mmol). The mixture was warmed to RT and stirred for 2 h. The reaction mixture was poured to a solution of ammonium hydroxide (20 ml) and water (50ml). The mixture was stirred for 1 hour, then extracted with ethyl acetate (3 c 150 ml), washed with brine (100 ml), dried over sodium sulfate and concentrated. The residue was purified by preparative HPLC (Method P3) to afford 3.5 g (75% yield, 96% purity) of the title compound.
1H-NMR (400 MHz, DMSO-de): d [ppm] = 7.46-7.42 (dd, 2H), 7.18-7.14 (m, 2H), 5.46 (br s, 1H), 4.28-4.22 (m, 1 H), 3.77 (m, 1 H), 3.54 (m, 1 H), 2.21 (br s, 2H). Intermediate 58A tert- Butyl 3-(4-fluorophenyl)-3-hydroxyazetidine-1-carboxylate
Figure imgf000093_0001
To a solution of tert- butyl 3-oxoazetidine-1-carboxylate (10.0 g, 58.4 mmol) in THF (200 ml) was added (4-fluorophenyl)magnesium bromide (70 ml of a 1.0 M solution in THF, 70 mmol) with stirring at 0°C under an atmosphere of nitrogen. After stirring for 3 h at RT, the reaction mixture was quenched with ammonium chloride (saturated aqueous solution). After extracting with dichloromethane, the combined organic phases were dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate, 3:1) to afford 10.2 g (73% of theory, 92% purity) of the title compound.
LC/MS [Method 13]: Rt = 1.07 min; MS (ESIpos): m/z = 212 [M+H-C4H8]+.
1H-NMR (400 MHz, CDC ): d [ppm] = 7.50-7.46 (m, 2H), 7.10-7.05 (m, 2H), 4.22 (d, 2H), 4.16 (d, 2H), 1.46 (s, 9H).
Intermediate 59A tert- Butyl 3-chloro-3-(4-fluorophenyl)azetidine-1-carboxylate
Figure imgf000093_0002
To a solution of tert- butyl 3-(4-fluorophenyl)-3-hydroxyazetidine-1-carboxylate (Intermediate 58A, 10.6 g, 39.8 mmol) in dichloromethane (160 ml) were added triethylamine (8.3 ml, 59.7 mmol) and methanesulfonyl chloride (3.7 ml, 47.7 mmol) dropwise at 0°C under a nitrogen atmosphere. The reaction mixture was stirred at RT for 24 h, then poured into water (200 ml) and extracted with dichloromethane. The combined organic layers were dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate, 4:1) to afford 7.80 g (69% of theory) of the title compound.
1H-NMR (300 MHz, DMSO-de): d [ppm] = 7.59-7.54 (m, 2H), 7.30-7.24 (m, 2H), 4.64 (d, 2H), 4.40 (d, 2H), 1.40 (s, 9H).
Intermediate 60A tert- Butyl 3-azido-3-(4-fluorophenyl)azetidine-1-carboxylate To a solution of tert.- butyl 3-chloro-3-(4-fluorophenyl)azetidine-1-carboxylate (Intermediate 59A, 7.20 g, 25.2 mmol) in A/,/\/-dimethylformamide (100 ml) was added sodium azide (8.19 g, 126 mmol) in portions. The resulting mixture was heated to 100°C overnight. After cooling to RT, the reaction mixture was poured into water (100 ml) and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate, 4:1) to give 6.30 g (85% of theory) of the title compound.
1H-NMR (300 MHz, CDC ): d [ppm] = 7.41-7.36 (m, 2H), 7.18-7.12 (m, 2H), 4.36-4.27 (m, 4H), 1.49 (s, 9H).
Intermediate 61A
3-(4-Fluorophenyl)-1-methylazetidin-3-amine
Figure imgf000094_0001
To a solution of tert- butyl 3-azido-3-(4-fluorophenyl)azetidine-1-carboxylate (Intermediate 60A, 8.40 g, 25.9 mmol, 90% purity) in THF (200 ml) was added lithium aluminium hydride (3.93 g, 104 mmol) in portions at 0°C. The resulting mixture was stirred for 30 min at RT and then heated to 55°C for 40 min. After cooling to room temperature, the reaction mixture was quenched with sodium sulfate decahydrate (30.0 g). The solids were filtered off and washed with methanol. The combined filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: dichloromethane/methanol, 19:1 containing 0.05% triethylamine) to give 2.14 g (40% of theory, 88% purity) of the title compound.
1H-NMR (300 MHz, DMSO-d6): d [ppm] = 7.65-7.57 (m, 2H), 7.15-7.09 (m, 2H), 3.52-3.49 (m, 2H), 3.14-3.11 (m, 2H), 2.29 (s, 3H).
Intermediate 62A (7S)-1-Amino-1-(4-fluorophenyl)-2-methylpropan-2-ol Methylmagnesium bromide (13 ml of a 1.0 M solution in THF, 13 mmol) was added slowly to a solution of methyl (2S)-amino(4-fluorophenyl)ethanoate hydrochloride (485 mg, 2.21 mmol) in THF (9.7 ml). After complete addition, the reaction mixture was slowly warmed to RT and stirred at this temperature overnight. Hydrochloric acid solution (1.0 M aqueous solution) was then added, and the mixture was washed with MTBE. The layers were separated and the organic layer was discarded. The aqueous layer was brought to basic pH by addition of sodium hydroxide (1.0 M aqueous solution) and was extracted with ethyl acetate. The combined organic layers were dried over magnesium sulfate and concentrated. The residue was purified by silica gel column chromatography (basified silica gel, eluent: gradient of methanol in dichloromethane). Yield: 203 mg (48% of theory, 95% purity).
LC/MS (Method 22): Rt = 1.08 min; MS (ESIpos): m/z = 184 [M+H]+.
1H-NMR (500 MHz, DMSO-de): d [ppm] = 7.44-7.30 (m, 2H), 7.13-7.03 (m, 2H), 4.36 (br. s, 1H), 3.69 (s, 1H), 2.07-1.75 (m, 2H), 1.00 (s, 3H), 0.94 (s, 3H). Intermediate 63A tert- Butyl (4S)-4-phenyl-1 ,2,3-oxathiazolidine-3-carboxylate 2,2-dioxide
Figure imgf000095_0001
Triethylamine (5.2 ml, 37 mmol) was added to a solution of thionylchloride (1.4 ml, 19 mmol) in dichloromethane (60.0 ml), and the mixture was cooled to -60°C. A solution of tert.- butyl [( 7S)-2- hydroxy- 1-phenylethyl]carbamate (4.00 g, 16.9 mmol) in dichloromethane (100 ml) was added dropwise, and the reaction was stirred at -60°C for 2 h. After warming to RT, water was added and the layers were separated. The organic layer was washed with brine, dried over magnesium sulfate and evaporated to dryness. The residue was dissolved in acetonitrile (40 ml), and sodium periodate (3.97 g, 18.5 mmol) and ruthenium(lll) chloride trihydrate (441 mg, 1.69 mmol) were added at 0°C. The mixture was stirred overnight at 0°C and for 1 d at RT. Water and ethyl acetate were then added, and the insoluble material was removed by filtration. The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over magnesium sulfate and evaporated to dryness to afford 4.20 g (73% of theory, 88% purity) of the title compound. The product was used in the next step without further purification.
LC/MS (Method 9): Rt = 1.91 min; MS (ESIneg): m/z = 344 [M-H+HC02H] . Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.008 (0.49), 0.008 (0.42), 1.195 (3.59), 1.227 (0.54),
1.329 (16.00), 1.481 (1.04), 4.597 (1.14), 4.606 (1.16), 4.621 (1.28), 4.630 (1.30), 5.024 (1.28),
5.040 (1.46), 5.048 (1.22), 5.064 (1.18), 5.521 (0.92), 5.529 (1.00), 5.537 (0.95), 5.545 (0.84),
7.318 (0.58), 7.322 (0.75), 7.330 (1.08), 7.372 (2.24), 7.392 (4.18), 7.408 (1.84), 7.439 (2.16),
7.444 (0.84), 7.456 (2.05), 7.460 (1.26), 7.475 (0.74), 7.478 (0.60). Intermediate 64A tert- Butyl [(7S)-2-(morpholin-4-yl)-1-phenylethyl]carbamate
Figure imgf000096_0001
A solution of te/t-butyl (4S)-4-phenyl-1,2,3-oxathiazolidine-3-carboxylate 2,2-dioxide (Intermedi ate 63A, 986 mg, 58% purity, 1.91 mmol) in THF (16.0 ml) was cooled to 0°C, and morpholine (670 pi, 7.6 mmol) was added. The reaction mixture was stirred at RT overnight before an aqueous solution of ammonium carbonate (1.0 M, 5.0 ml) was added. The mixture was again stirred at RT overnight before the pH was adjusted to 5, and stirring was continued at RT for 3 d. The reaction mixture was then evaporated to dryness to obtain the title compound. Yield: 660 mg (61 % of theory, 54% purity). The product was used in the next step without further purification. LC/MS (Method 9): Rt = 1.01 min; MS (ESIpos): m/z = 307 [M+H]+.
Intermediate 65A
(1S)-2-(Morpholin-4-yl)-1-phenylethanamine hydrochloride
Figure imgf000096_0002
A solution of tert- butyl [(7S)-2-(morpholin-4-yl)-1-phenylethyl]carbamate (Intermediate 64A, 660 g, 54% purity, 1.16 mmol) in dichloromethane (4.0 ml) was treated with anisole (630 pi, 5.8 mmol) and hydrogen chloride (2.9 ml, 4.0 M solution in dioxane, 12 mmol) at RT. After stirring at RT overnight, the reaction mixture was evaporated to dryness to afford the title compound. Yield: 453 mg (98% of theory, 70% purity). The product was used in the next step without further purification.
Intermediate 66A tert- Butyl [(1S)-2-(3-fluoroazetidin-1-yl)-1-(4-fluorophenyl)ethyl]carbamate
Figure imgf000097_0001
To a solution of tert-butyl (4S)-4-(4-fluorophenyl)-2,2-dioxo-1,2lambda6,3-oxathiazolidine-3- carboxylate (500 mg, 94% purity, 1.48 mmol) in acetonitrile (10 ml) was added 3-fluoroazetidine hydrochloride (215 mg, 1.93 mmol) and triethylamine (310 mI, 2.2 mmol). The reaction was stirred for 5 h at RT, then the solvent was removed and the residue was suspended in ethyl acetate and washed with sodium hydrogen carbonate (2 x 50 ml of a saturated aqueous solution) and brine. The organic phase was dried, concentrated and purified by preparative HPLC to afford 98.3 mg (21% yield, 100% purity) of the title compound.
LC-MS (Method 23): Rt = 1.79 min; MS (ESIpos): m/z = 313 [M+H]+
Ή-NMR (600 MHz, DMSO-d6) d [ppm]: 1.143 (16.00), 1.383 (1.07), 2.425 (0.42), 3.955 (0.50), 4.028 (0.63), 4.510 (0.42), 5.368 (0.59), 5.464 (0.58), 5.563 (0.45), 7.170 (2.04), 7.185 (4.20), 7.200 (2.27), 7.395 (1.73). Intermediate 67A
(1S)-2-(3-Fluoroazetidin-1-yl)-1-(4-fluorophenyl)ethan-1-amine hydrochloride
Figure imgf000097_0002
To a solution of tert- butyl [(1S)-2-(3-fluoroazetidin-1-yl)-1-(4-fluorophenyl)ethyl]carbamate (Intermediate 66A, 98.3 mg, 94% purity, 296 pmol) in methanol (2.0 ml) was added hydrochloric acid (62 mI of a 4.0 M solution in 1,4-dioxane, 1.2 mmol) and the reaction was stirred overnight at RT. After this time, a further portion of hydrochloric acid (62 mI of a 4.0 M solution in 1,4-dioxane, 1.2 mmol) was added and the reaction was again stirred overnight at RT. The solvent was then removed and the residue was dissolved and acetonitrile and water and lyophilized to afford 98 g (quantitative, 40% purity) of the title compound. The material was used without further purification.
LC-MS (Method 24): Rt = 1.18 min; MS (ESIpos): m/z = 212 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: 1.180 (2.90), 2.328 (8.23), 2.367 (4.88), 2.671 (8.53), 2.710 (5.18), 3.168 (12.50), 3.958 (5.64), 7.349 (16.00), 7.578 (13.41), 8.742 (3.20).
Intermediate 68A tert- Butyl 3-hydroxy-3-(4-methoxyphenyl)azetidine-1-carboxylate
Figure imgf000098_0001
To a solution of tert- butyl 3-oxoazetidine-1-carboxylate (20.00 g, 117 mmol) in THF (200 ml) was added (4-methoxyphenyl)magnesium bromide (140 ml, 140 mmol, 1 M solution in THF). The reaction mixture was stirred at 0°C for 3 h under an atmosphere of nitrogen. Then, a saturated aqueous solution of ammonium chloride (200 ml) was added, and the mixture was extracted with dichloromethane (2 x 400 ml). The combined organic layers were washed with water (2 x 400 ml) and brine (2 x 400 ml) and dried over anhydrous sodium sulfate. Filtration, concentration and chromatography on silica gel (eluent: petroleum ether/ethyl acetate 3:1) gave the title compound. Yield: 27.10 g (79% of theory, 95% purity).
LC/MS [Method 8]: Rt = 1.14 min; MS (ESIpos): m/z = 559 [2M+H]+.
1H-NMR (300 MHz, CDC ): d [ppm] = 1.50 (s, 9H), 3.84 (s, 3H), 4.17 (d, 2H), 4.26 (d, 2H), 6.94 (d, 2H), 7.42 (d, 2H). Intermediate 69A tert- Butyl 3-azido-3-(4-methoxyphenyl)azetidine-1-carboxylate
Figure imgf000098_0002
To a solution of tert- butyl 3-hydroxy-3-(4-methoxyphenyl)azetidine-1-carboxylate (Intermediate 68A, 10.0 g, 35.8 mmol) in THF (200 ml) were added triphenylphosphine (11.7 g, 44.8 mmol), diphenyl phosphoroazidate (10.0 ml, 46.5 mmol) and diisopropyl azodicarboxylate (9.16 ml, 46.5 mmol) under nitrogen atmosphere. After stirring at RT for 16 h, the reaction mixture was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with water, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by flash-chromatography on silica gel (eluent: petroleum ether/ethyl acetate 15:1 ® 4:1) to give the title compound. Yield: 7.80 g (71% of theory).
5 1H-NMR (400 MHz, DMSO-d6): d [ppm] = 7.39 (d, 2H), 7.01 (d, 2H), 4.33 (d, 2H), 4.16 (d, 2H), 3.79 (s, 3H), 1.39 (s, 9H).
Intermediate 70A tert- Butyl 3-amino-3-(4-methoxyphenyl)azetidine-1-carboxylate
Figure imgf000099_0001
io tert- Butyl 3-azido-3-(4-methoxyphenyl)azetidine-1-carboxylate (Intermediate 69A, 4.8 g, 15.8 mmol) was dissolved in methanol (50.0 ml), and 10% palladium on active carbon (800 mg) was added. The mixture was stirred under a hydrogen atmosphere (2-3 atm) at RT overnight. Then, the catalyst was filtered off, and the solvent was removed under reduced pressure. The residue was purified by flash- chromatography on silica gel (eluent: petroleum ether/0-100% ethyl acetate) to afford the title 15 compound. Yield: 3.16 g (67% of theory, 93% purity).
LC/MS [Method 1]: Rt = 1.43 min; MS (ESIpos): m/z = 557 [2M+H]+.
1H-NMR (300 MHz, DMSO-d6): d [ppm] = 7.40 (d, 2H), 6.91 (d, 2H), 3.99 (d, 2H), 3.88 (d, 2H), 3.74 (s, 3H), 2.45 (s, 2H), 1.39 (s, 9H).
Intermediate 71A
20 Ethyl amino(hydroxyimino)acetate
Figure imgf000099_0002
Water (172 ml) was added dropwise over a period of 2 h to a vigorously stirred mixture of ethyl cyanoformate (28.3 g, 285 mmol), hydroxylamine hydrochloride (30.0 g, 428 mmol) and sodium carbonate (23.4 g, 22.0 mmol) in ethanol (286 ml) at RT. The resulting mixture was stirred until the 25 starting material has been consumed and then the solvent was removed under reduced pressure. The residue was extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was evaporated under reduced pressure to give 25.0 g (65% of theory, 99% purity) of the title compound.
LC-MS (Method 7): Rt = 0.38 min; MS (ESIpos): m/z = 133 [M+H]+.
1H-NMR (300 MHz, CDC ): d [ppm] 1.35 (t, 3H), 4.32 (q, 2H), 5.20 (brs, 2H), 9.50 (brs, 1H). Intermediate 72A
Diethyl 1 H-imidazole-2, 4-dicarboxylate
Figure imgf000100_0001
To a solution of ethyl amino(hydroxyimino)acetate (Intermediate 71A, 10.0 g, 7.6 mmol) in xylene (200 ml) were added triethylamine (12.7 ml, 93.1 mmol) and ethyl propiolate (9.2 ml, 93.1 mmol) at 0°C. After stirring at RT for 12 h, the reaction mixture was concentrated under reduced pressure, and the residue was dissolved in xylene (300 ml) and heated under reflux for 16 h. After cooled to RT, the resulting mixture was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (eluent: petroleum ether-ethyl acetate 20:1, 3:1) to give 3.00 g (18% of theory, 99% purity) of the title compound. LC-MS (Method 25): Rt = 0.71 min; MS (ESIpos): m/z = 213 [M+H]+.
1H-NMR (300 MHz, DMSO-d6): d [ppm] 1.21-1.36 (m, 6H), 4.20-4.38 (m, 4H), 8.02 (s, 1H), 13.90 (s, 1H).
Intermediate 73A
Diethyl 1-[2-(3,4-dimethylphenyl)-2-oxoethyl]-1 H-imidazole-2,4-dicarboxylate
Figure imgf000100_0002
To a solution of diethyl 1H-imidazole-2,4-dicarboxylate (Intermediate 72A, 6.00 g, 28.3 mmol) in acetone (80 ml) was added 2-bromo-1-(3,4-dimethylphenyl)ethan-1-one (8.03 g, 88% purity, 31.1 mmol) and potassium carbonate (4.29 g, 31.1 mmol). The mixture was stirred 3 h at RT, then the mixture was filtered and the filtrate was concentrated. The crude residue was washed with ethyl acetate/petroleum ether (80 ml, 1:10) and stirred for 30 min, then the solid was collected by filtration to afford 8.00 g (72% of theory, 91% purity) of the title compound.
LC-MS (Method 25): Rt = 1.09 min; MS (ESIpos): m/z = 359 [M+H]+
1H-NMR (400 MHz, DMSO-d6): d [ppm] 8.14 (s, 1H), 7.84-7.75 (m, 2H), 7.36 (d, 1H), 6.00 (s, 2H), 4.29-4.16 (m, 4H), 2.31 (s, 6H), 1.28 (t, 3H), 1.18 (t, 3H). Intermediate 74A
Ethyl 6-(3,4-dimethylphenyl)-8-oxo-7,8-dihydroimidazo[1 ,2-a]pyrazine-2-carboxylate
Figure imgf000101_0001
To a solution of diethyl 1-[2-(3,4-dimethylphenyl)-2-oxoethyl]-1H-imidazole-2,4-dicarboxylate (Intermediate 73A, 10.5 g, 29.3 mmol) in acetic acid (200 ml) was added ammonium acetate (67.7 g, 879 mmol). The reaction mixture was stirred for 18 h at 120°C, then the solution was poured into ice-water. The resulting precipitate was filtered and the filter cake was washed with water and dried to afford 7.77 g (77% of theory, 90% purity) of the title compound.
LC-MS (Method 25): Rt = 0.94 min; MS (ESIpos): m/z = 312 [M+H]+
Intermediate 75A 6-(3,4-Dimethylphenyl)-8-oxo-7,8-dihydroimidazo[1 ,2-a]pyrazine-2-carboxylic acid
Figure imgf000101_0002
A suspension of ethyl 6-(3,4-dimethylphenyl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2- carboxylate (Intermediate 74A, 2.00 g, 6.42 mmol) and lithium hydroxide (1.54 g, 64.2 mmol) in ethanol (26 ml) and water (13 ml) was stirred at RT overnight. The reaction mixture was diluted with water and the pH of the solution was adjusted to 3 by addition of hydrochloric acid (1.0 N aqueous solution). The precipitate was collected by filtration and dried to afford the title compound. Yield: 1.20 g (64% of theory, 97% purity).
LC-MS (Method 9): Rt = 1.16 min; MS (ESIpos): m/z = 284 [M+H]+.
1H-NMR (400 MHz, DMSO-de): d [ppm] = 12.88 (br s, 1H), 11.55 (br s, 1H), 8.29 (s, 1H), 7.82 (s, 1 H), 7.48 (s, 1 H), 7.40 (d, 1 H), 7.26 (d, 1 H), 2.29 (s, 3H), 2.27 (s, 3H).
Intermediate 76A
Diethyl 1-[2-(4-chloro-3-fluorophenyl)-2-oxoethyl]-1 H-imidazole-2,4-dicarboxylate
Figure imgf000102_0001
A mixture of diethyl 1H-imidazole-2,4-dicarboxylate (Intermediate 72A, 500 mg, 2.36 mmol) and 2-bromo-1-(4-chloro-3-fluorophenyl)ethan-1-one (711 mg, 2.83 mmol) in acetone (21 ml) was treated with potassium carbonate (814 mg, 5.89 mmol) and the mixture was stirred at RT for three days. The insoluble material was filtered off and washed with acetone. The combined filtrates were concentrated under reduced pressure and the residue was dried to afford the crude title compound which was used in the next step without further purification. Yield: 1.35 g (99% of theory, 80% purity).
LC-MS (Method 9): Rt = 1.83 min; MS (ESIpos): m/z = 383 [M+H]+.
Intermediate 77A
Ethyl 6-(4-chloro-3-fluorophenyl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2-carboxylate
Figure imgf000102_0002
A mixture of diethyl 1-[2-(4-chloro-3-fluorophenyl)-2-oxoethyl]-1H-imidazole-2,4-dicarboxylate (Intermediate 76A, 1.35 g, 3.53 mmol) and ammonium acetate (6.80 g, 88.2 mmol) in acetic acid (35 ml) was heated to 110°C for three days. After cooling to RT, the reaction mixture was poured into water and the precipitate was collected by filtration and dried to afford the title compound which was used in the next step without further purififaction. Yield: 530 mg (36% of theory, 81% purity).
LC-MS (Method 26): Rt = 0.78 min; MS (ESIpos): m/z = 336 [M+H]+.
1H-NMR (600 MHz, DMSO-cfe): d [ppm] = 11.78 (brs, 1H), 8.37 (s, 1H), 7.94 (s, 1H), 7.79 (dd, 1H), 7.76-7.73 (m, 1 H), 7.56 (dd, 1 H), 4.32 (q, 2H), 1.33 (t, 3H).
Intermediate 78A
6-(4-Chloro-3-fluorophenyl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2-carboxylic acid
Figure imgf000103_0001
A mixture of ethyl 6-(4-chloro-3-fluorophenyl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2- carboxylate (Intermediate 77A, 530 mg, 1.58 mmol) and lithium hydroxide (189 mg, 7.89 mmol) in water (3.0 ml) and ethanol (6.0 ml) was stirred at RT overnight. The ethanol was distilled off and the aqueous layer was diluted with water and the pH of the solution was adjusted to 3 by addition of hydrochloric acid (10 ml of a 1.0 N aqueous solution). The precipitate was collected by filtration and dried to afford the title compound. Yield: 415 mg (85% of theory). LC-MS (Method 9): Rt = 1.11 min; MS (ESIpos): m/z = 308 [M+H]+.
1H-NMR (600 MHz, DMSO-de): d [ppm] = 13.60 (br s, 1H), 11.73 (br s, 1H), 8.31 (s, 1H), 7.98 (s, 1 H), 7.81-7.73 (m, 2H), 7.57 (br d, 1 H).
Intermediate 79A
Diethyl 1-[2-(2,3-dihydro-1 ,4-benzodioxin-6-yl)-2-oxoethyl]-1 H-imidazole-2,4-dicarboxylate
Figure imgf000103_0002
To a solution of diethyl 1H-imidazole-2,4-dicarboxylate (Intermediate 72A, 450 mg, 2.12 mmol) in acetone (15 ml) was 2-bromo-1-(2,3-dihydro-1,4-benzodioxin-6-yl)ethan-1-one (545 mg, 2.12 mmol) and potassium carbonate (322 mg, 2.33 mmol). The mixture was stirred overnight at RT, then filter. The filtrate was concentrated, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated to give 820 mg (83% of theory, 84% purity) of the title compound, which was used without further purification. LC-MS (Method 25): Rt = 0.97 min; MS (ESIpos): m/z = 389 [M+H]+.
Intermediate 80A
Ethyl 6-(2,3-dihydro-1,4-benzodioxin-6-yl)-8-oxo-7,8-dihydroimidazo[1 ,2-a]pyrazine-2- carboxylate
Figure imgf000104_0001
To a solution of diethyl 1-[2-(2,3-dihydro-1,4-benzodioxin-6-yl)-2-oxoethyl]-1H-imidazole-2,4- dicarboxylate (Intermediate 79A, 850 g, 2.19 mmol) in acetic acid (20 ml) was added ammonium acetate (4.22 g, 54.7 mmol). After refluxing for 48 h, the reaction mixture was poured onto ice water and neutralized with sodium hydroxide. The precipitate was collected by filitration, washed thoroughly with water and dried at 100°C under reduced pressure to give 350 mg (39% of theory, 84% purity) of the title compound.
LC-MS (Method 25): Rt = 0.85 min; MS (ESIpos): m/z = 342 [M+H]+.
Intermediate 81A
6-(2,3-Dihydro-1,4-benzodioxin-6-yl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2-carboxylic acid
Figure imgf000104_0002
To a solution of ethyl 6-(2,3-dihydro-1,4-benzodioxin-6-yl)-8-oxo-7,8-dihydroimidazo[1,2- a]pyrazine-2-carboxylate (Intermediate 80A, 300 mg, 879 pmol) in water (10 ml) and ethanol (10 ml) was added sodium hydroxide (3.5 ml of a 2.0 M aqueous solution, 7.00 mmol). After stirring at RT for 2 h, the mixture was diluted with water, then adjusted to pH 2 with hydrochloric acid (1.0 M aqueous solution). The solid was collected by filtration and dried to give 197.7 mg (69% of theory, 96% purity) of the title compound. LC-MS (Method 5): Rt = 0.88min; MS (ESIpos): m/z = 314 [M+H]+.
Intermediate 82A
Diethyl 1-[2-(naphthalen-2-yl)-2-oxoethyl]-1 H-imidazole-2,4-dicarboxylate
Figure imgf000105_0001
To a solution of diethyl 1H-imidazole-2,4-dicarboxylate (Intermediate 72A, 400 g, 1.88 mmol) in acetone (20 ml) was added 2-bromo-1-(2-naphthyl)ethenone (472 mg, 1.88 mmol) and potassium carbonate (286 mg, 2.07 mmol). The mixture was stirred overnight at RT, then filtered. The filtrate was concentrated, then diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated to give 600 mg (78% of theory, 94% purity) of the title compound.
LC-MS (Method 25): Rt = 1.09 min; MS (ESIpos): m/z = 381 [M+H]+.
Intermediate 83A
Ethyl 6-(naphthalen-2-yl)-8-oxo-7,8-dihydroimidazo[1 ,2-a]pyrazine-2-carboxylate
Figure imgf000105_0002
To a solution of diethyl 1-[2-(naphthalen-2-yl)-2-oxoethyl]-1H-imidazole-2,4-dicarboxylate (Intermediate 82A, 600 mg, 1.58 mmol) in acetic acid (50 ml) was added ammonium acetate (3.04 g, 39.4 mmol). After refluxing for 48 h, the reaction mixture was poured onto ice water and neutralized with sodium hydroxide. The precipitate was collected by filtration, washed thoroughly with water and dried at 100°C under reduced pressure to give 460 mg (71% of theory, 82% purity) of the title compound.
LC-MS (Method 25): Rt = 0.96 min; MS (ESIpos): m/z = 334 [M+H]+.
Intermediate 84A
6-(Naphthalen-2-yl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2-carboxylic acid To a solution of ethyl 6-(naphthalen-2-yl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2-carboxylate (Intermediate 83A, 400 g, 1.20 mmol) in water (15 ml) and ethanol (15 ml) was added sodium hydroxide (4.8 ml of a 2.0 M aqueous solution, 9.6 mmol). After stirring at RT for 2 h, the mixture was diluted with water, then adjusted to pH 2 with hydrochloric acid (1.0 M aqueous solution). The solid was collected by filtration and dried to give 286 mg (75% of theory, 96% purity) of the title compound.
LC-MS (Method 27): Rt = 2.13 min; MS (ESIpos): m/z = 306 [M+H]+.
Intermediate 85A 4-(4-Fluorophenyl)-1-(2,2,2-trifluoroethyl)piperidin-4-ol
Figure imgf000106_0001
Under an argon atmosphere was bromido(4-fluorophenyl)magnesium (2.8 ml of a 1.0 M solution in THF, 2.8 mmol) diluted with further THF (3.0 ml) and cool to 0°C. Then, a solution of 1 -(2,2,2- trifluoroethyl)piperidin-4-one (426 mg, 2.35 mmol) in THF (8 ml) was added dropwise and the reaction was warmed to RT and stirred for 4.5 h. The mixture was cooled to 0°C and the reaction quenched by addition of sodium hydrogencarbonate (40 ml of a saturated aqueous solution). The mixture was extracted four times with ethyl acetate and the combined organic phases weredried with magnesium sulfate, filtered and concentrated to afford 585 mg (75% of theory, 84% purity) of the title compound, which was used without further purification. LC-MS (Method 9): Rt = 1.49 min; MS (ESIpos): m/z = 278 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.149 (0.44), -0.008 (3.65), 0.008 (3.26), 0.146 (0.41), 1.356 (2.88), 1.378 (0.82), 1.406 (1.03), 1.426 (0.84), 1.538 (6.36), 1.568 (7.76), 1.668 (0.58),
1.688 (0.41), 1.701 (0.49), 1.800 (0.55), 1.813 (0.54), 1.885 (2.96), 1.897 (3.21), 1.917 (5.30),
1.927 (5.33), 1.948 (2.94), 1.960 (2.69), 2.270 (0.71), 2.283 (0.41), 2.292 (0.46), 2.306 (0.96), 2.318 (0.63), 2.328 (0.65), 2.345 (0.51), 2.366 (0.43), 2.473 (1.09), 2.585 (1.68), 2.600 (1.75), 2.634 (1.22), 2.665 (1.17), 2.709 (3.65), 2.733 (6.77), 2.774 (5.58), 2.801 (7.18), 2.828 (2.78), 2.875 (0.41), 2.891 (0.60), 2.914 (0.54), 2.926 (0.96), 2.948 (0.74), 2.958 (1.25), 2.978 (0.76), 3.022 (0.66), 3.037 (0.57), 3.052 (1.20), 3.078 (2.32), 3.104 (2.23), 3.130 (0.81), 3.144 (4.09), 3.170 (11.87), 3.195 (11.53), 3.221 (4.13), 3.233 (0.66), 3.246 (0.54), 3.339 (1.14), 3.353 (1.75), 5 3.364 (1.49), 3.378 (1.28), 3.389 (1.28), 3.402 (0.52), 3.414 (0.41), 3.856 (0.81), 4.356 (2.78),
4.910 (14.58), 5.152 (0.52), 5.163 (0.51), 6.725 (0.41), 6.736 (0.51), 6.748 (0.47), 6.949 (0.46), 6.971 (0.79), 7.083 (0.85), 7.090 (7.62), 7.113 (16.00), 7.135 (8.57), 7.315 (0.60), 7.329 (0.62), 7.336 (0.55), 7.351 (0.41), 7.486 (1.01), 7.494 (8.82), 7.499 (3.68), 7.508 (9.77), 7.515 (8.85), 7.524 (3.49), 7.529 (7.87), 7.537 (0.87). io Intermediate 86A
A/-[4-(4-Fluorophenyl)-1-(2,2,2-trifluoroethyl)piperidin-4-yl]acetamide
Figure imgf000107_0001
To a solution of 4-(4-fluorophenyl)-1-(2,2,2-trifluoroethyl)piperidin-4-ol (364 mg, 1.31 mmol) in acetonitrile (5.5 ml) was added concentrated sulfuric acid (0.42 ml) dropwise at 0°C. Then, the is reaction was diluted with ice-water (40 ml) and extracted twice with MTBE (2 x 20 ml) and the organic phase was discarded. The pH of the aqueous phase was adjusted to 12 with sodium hydroxide (1.0 M aqueous solution) and the mixture was extracted with ethyl acetate (3 x 30 ml). The organic phase was dried with magnesium sulfate, filtered and concentrated to give 318 mg (73% of theory, 96% purity) of the title compound, which was used without further purification.
20 LC-MS (Method 26): Rt = 0.71 min; MS (ESIpos): m/z = 319 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.008 (0.42), 0.008 (0.53), 1.795 (0.56), 1.805 (0.71), 1.818 (0.92), 1.827 (1.26), 1.836 (1.28), 1.858 (16.00), 2.280 (1.54), 2.311 (1.33), 2.601 (0.91),
2.629 (1.82), 2.657 (1.17), 2.696 (0.57), 2.710 (0.41), 2.740 (1.67), 2.769 (1.09), 3.122 (0.57),
3.133 (1.17), 3.158 (2.71), 3.184 (2.84), 3.209 (1.19), 3.397 (0.44), 3.423 (0.42), 7.073 (1.73),
25 7.095 (3.79), 7.112 (0.70), 7.118 (2.08), 7.349 (2.12), 7.354 (1.00), 7.362 (2.33), 7.371 (2.10),
7.379 (0.83), 7.385 (1.84), 7.872 (2.38).
Intermediate 87A
4-(4-Fluorophenyl)-1-(2,2,2-trifluoroethyl)piperidin-4-amine A/-[4-(4-Fluorophenyl)-1-(2,2,2-trifluoroethyl)piperidin-4-yl]acetamide (316 mg, 992 pmol) was heated to 100°C for 3 days in a hydrochloric acid (4.0 ml of a 6.0 M aqueous solution, 24 mmol). Following this, ice-water (80 ml) was added to the reaction and the pH of the mixture was adjusted to 12 with sodium hydroxide (1.0 M aqueous solution). The mixture was extracted with ethyl acetate and the combined organic phases were dried with magnesium sulfate, filtered and concentrated to give 118 mg (28% of theory, 66% purity) of the title compound, which was used without further purification.
LC-MS (Method 26): Rt = 1.52 min; MS (ESIpos): m/z = 277 [M+H]+ Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.008 (0.87), 0.008 (0.83), 1.526 (1.85), 1.557 (2.14), 1.858 (0.55), 1.902 (1.31), 1.913 (1.39), 1.935 (1.85), 1.942 (1.97), 1.964 (1.31), 1.974 (1.32),
2.008 (0.47), 2.328 (0.46), 2.473 (0.94), 2.629 (1.19), 2.637 (2.09), 2.648 (1.28), 2.657 (1.62),
2.666 (2.80), 2.675 (1.52), 2.696 (0.83), 2.710 (0.61), 2.823 (1.61), 2.828 (1.77), 2.851 (3.24),
2.857 (2.89), 2.863 (2.07), 2.879 (1.78), 2.944 (0.41), 3.078 (0.46), 3.106 (0.53), 3.120 (2.13), 3.145 (4.67), 3.157 (0.64), 3.171 (4.58), 3.182 (1.07), 3.197 (1.73), 3.208 (0.90), 3.243 (0.62),
3.268 (1.41), 3.294 (2.23), 3.311 (16.00), 3.398 (0.59), 3.423 (0.53), 6.100 (0.51), 7.082 (2.57), 7.087 (0.96), 7.099 (1.20), 7.105 (5.44), 7.121 (1.09), 7.127 (3.03), 7.133 (0.97), 7.155 (1.38),
7.177 (0.78), 7.449 (0.76), 7.463 (0.80), 7.471 (0.75), 7.485 (0.69), 7.529 (0.45), 7.537 (3.11),
7.543 (1.38), 7.551 (3.35), 7.559 (3.22), 7.568 (1.21), 7.573 (2.87). Intermediate 88A
Dimethyl 1 H-1 ,2,4-triazole-3,5-dicarboxylate
Figure imgf000108_0001
1H-1,2,4-Triazole-3,5-dicarboxylic acid (Intermediate 88A, 5.00 g, 31.8 mmol) was added to an anhydrous solution of methanolic hydrochloric acid (10 ml) at RT. After stirring for 24 h, the methanol was evaporated under reduced pressure to give 6.20 g of the title compound, which was used for the next step directly without further purification. H-NMR (300 MHz, DMSO-d6): d [ppm] 8.66 (s, 1H), 3.84 (s, 3H), 3.90 (s, 3H).
Intermediate 89A
Dimethyl 1-[2-(naphthalen-2-yl)-2-oxoethyl]-1 H-1,2,4-triazole-3,5-dicarboxylate
Figure imgf000109_0001
Dimethyl 1H-1,2,4-triazole-3,5-dicarboxylate (Intermediate 88A, 800 mg, 4.32 mmol), 2-bromo-1- (2-naphthyl)ethanone (1.18 g, 4.75 mmol) and potassium carbonate (1.25 g, 9.07 mmol) were stirred overnight at RT in acetone (20 ml). After filtering off the solids, the filtrate was evaporated and partitioned between dichloromethane and water. The organic phase was washed with water and brine, then dried over sodium sulphate and evaporated to give 850 mg (50% of theory, 90% purity) of the title compound.
1 H-NMR (400 MHz, DMSO-d6): d [ppm] 8.18 (d, 1H), 8.12 (d, 1H), 8.06 (d, 1H), 8.02 (dd, 1H), 7.76-7.67 (m, 2H), 6.49 (s, 1H), 3.92 (s, 3H), 3.84 (s, 3H).
Intermediate 90A
Methyl 6-(naphthalen-2-yl)-8-oxo-7,8-dihydro[1 ,2,4]triazolo[1 ,5-a]pyrazine-2-carboxylate
Figure imgf000109_0002
A solution of dimethyl 1-[2-(2-naphthyl)-2-oxoethyl]-1H-1,2,4-triazole-3,5-dicarboxylate (Intermediate 89A, 850 mg, 2.18 mmol, 90% purity) in 20 ml acetic acid, then ammonium acetate (3.36 g, 43.6 mmol) was added and stirred overnight at 110°C, the solution was poured into ice- water, filtered. The filter cake was washed with water and dried in air to give 650 mg (73% of theory, 76% purity) of the title compound.
LC-MS (Method 5): Rt = 0.93 min; MS (ESIpos): m/z = 321 [M+H]+. Intermediate 91A
6-(Naphthalen-2-yl)-8-oxo-7,8-dihydro[1 ,2,4]triazolo[1 ,5-a]pyrazine-2-carboxylic acid
Figure imgf000110_0001
To a solution of methyl 6-(2-naphthyl)-8-oxo-7,8-dihydro[1,2,4]triazolo[1,5-a]pyrazine-2- carboxylate (Intermediate 90A, 670 mg, 1.58 mmol, 76% purity) and methanol (10 ml) was added sodium hydroxide (5 ml of a 3.0 N aqueous solution) and the reaction was stirred for 2 h at RT. The pH of aqueous phase was adjusted to 6 with hydrochloric acid (3.0 M aqueous solution). The resulting precipitate was filtered and the solid was washed with water and dried to give 450 mg (88% of theory, 95% purity) of the title compound. 1H-NMR (400 MHz, DMSO-de): d [ppm] 13.82 (br s, 1H), 12.40 (s, 1H), 8.53 (s, 1H), 8.43 (s, 1H), 8.07-8.00 (m, 3H), 7.90-7.88 (m, 1H), 7.63-7.61 (m, 2H).
Example compounds
The compounds found in the following Table 1 are prepared using standard amide coupling conditions, and when appropriate, enantiomeric separation, using one of the standard conditions listed.
General procedure 1-A
To a solution of carboxylic acid (1.0 equiv.) and the required amine (1.5 equiv.) in pyridine at 50°C was added 2,4,6-tripropyl-2,4,6-trioxo-1,3,5,2,4,6-trioxatriphosphorinane (T3P®) (³50 wt.% in ethyl acetate, 3.0 equiv.) dropwise over 45 - 90 min. The reaction mixture was stirred for 1 hour, then cooled to RT and purified by either preparative HPLC or silica gel column chromatography.
General procedure 1-B
To a solution of carboxylic acid (1.0 equiv.), HATU (1.0 - 2.0 equiv.) and DIPEA (2.0 - 2.7 equiv.) in DMF at either 0°C or RT was added the required amine (1.05 - 1.5 equiv.) and the reaction was stirred at RT. Upon consumption of the carboxylic acid, the reaction mixture was purified by either preparative HPLC or silica gel column chromatography.
General procedure 1-C
To a solution of carboxylic acid (1.0 equiv.), EDCI (1.5 equiv.) and DMAP (2.3 equiv.) in dichloromethane was added the required amine (1.5 equiv.) and the reaction mixture was stirred overnight at 50°C. The reaction mixture was then cooled to RT and purified by either preparative HPLC or silica gel column chromatography.
General procedure 1-D
The racemic amide was separated into its enantiomeres by preparative HPLC (column: Daicel OJ- H, 250 x 25 mm; flow rate: 80 ml/min; temperature: 40°C; eluent: 70% supercritical CC>2:30% methanol).
General procedure 1-E
The racemic amide was separated into its enantiomeres by preparative HPLC (column: Daicel Chiralpak IF, 5 pm, 250 c 20 mm; flow rate: 15 ml/min; temperature: 50°C; eluent: 30% n- heptan:70% ethanol).
General procedure 1-F
The racemic amide was separated into its enantiomeres by preparative HPLC (column: Daicel Chiralpak IE, 5 pm, 250 c 20 mm; flow rate: 20 ml/min; temperature: 30°C; eluent: n-heptan ethanol; 0.0 5.0 min, 50% ethanol, 5.1 15.0, 75% ethanol. General procedure 1-G
The racemic amide was separated into its enantiomeres by preparative HPLC (column: Daicel Chiralpak IC, 5 pm, 250 c 20 mm; flow rate: 15 ml/min; temperature: 40°C; eluent: 80% n- heptan:20% ethanol).
Table 1
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0002
Example 1-99
3-(2,3-Dihydro-1,4-benzodioxin-6-yl)-4-fluoro-1-oxo-/\/-[(2R)-4,4,4-trifluorobutan-2-yl]-1,2- dihydropyrrolo[1 ,2-a]pyrazine-7-carboxamide
Figure imgf000164_0001
To a solution of 3-(2,3-dihydro-1,4-benzodioxin-6-yl)-3-ethoxy-4-fluoro-1-oxo-/\/-[(2R)-4,4,4- trifluorobutan-2-yl]-1 ,2,3,4-tetrahydropyrrolo[1 ,2-a]pyrazine-7-carboxamide (Intermediate 53A, 21.0 g, 82% purity, 35.5 pmol) in 1,4-dioxane was added Amberlyst® 15 (24.2 mg) and magnesium sulfate (47.0 mg, 390 pmol) and the reaction was stirred at 80°C for 1 h. The mixture was filtered over diatomaceous earth, washed with 1,4-dioxane and concentrated. The crude residue was purified by silica gel column chromatography (eluent: cyclohexane/ethyl acetate, gradient 0-100%) to afford 3.2 mg (20% of theory, 98% purity) of the title compound.
LC-MS (Method 8): Rt = 0.82 min; MS (ESIpos): m/z = 440 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.008 (3.25), 0.008 (2.93), 1.234 (6.73), 1.251 (6.66), 2.328 (0.56), 2.465 (0.54), 2.559 (1.28), 2.579 (0.86), 2.587 (0.66), 2.670 (0.55), 4.296 (16.00),
4.340 (0.92), 4.356 (0.71), 6.974 (3.39), 6.995 (5.09), 7.069 (2.03), 7.073 (1.68), 7.090 (1.44), 7.094 (1.18), 7.116 (3.23), 7.423 (2.09), 7.427 (2.42), 7.433 (2.23), 7.437 (2.25), 7.922 (4.11), 7.926 (4.09), 8.289 (1.91), 8.309 (1.86), 10.817 (1.66). Example 1-100
3-(2,3-Dihydro-1,4-benzodioxin-6-yl)-4-fluoro-/\/-[(1R)-1-(4-fluorophenyl)ethyl]-1-oxo-1,2- dihydropyrrolo[1 ,2-a]pyrazine-7-carboxamide
Figure imgf000165_0001
To a solution of 3-(2,3-dihydro-1,4-benzodioxin-6-yl)-/\/-[(1R)-1-(4-fluorophenyl)ethyl]-1-oxo-1,2- dihydropyrrolo[1,2-a]pyrazine-7-carboxamide (Example 1-30, 147 g, 97% purity, 329 pmol) and A/,/\/-diisopropylethylamine (110 pi, 660 pmol) in dry acetonitrile (3.3 ml) at 0°C was added Selectfluor® (245 mg, 95% purity, 658 pmol). The mixture was sonicated for 5 min, then stirred at RT for 30 min. A further portion of Selectfluor® (245 mg, 95% purity, 658 pmol) and N,N- diisopropylethylamine (110 pi, 660 pmol) were added to the reaction and the reaction was stirred for a further 30 min. The reaction was cooled to 0°C, quenched with brine and the aqueous solution was extracted with ethyl acetate. The combined organic phase was washed with brine, dried and concentrated. The crude residue was suspended in pyridine (1.7 ml) and treated with T3P® (590 pi, ³50 wt.% in ethyl acetate, 990 pmol) and the reaction was heated to 90°C for 80 min. The reaction was cooled to RT, and purified by preparative HPLC to afford 34.0 mg (22% yield, 97% purity) of the title compound.
LC-MS (Method 8): Rt = 0.90 min; MS (ESIpos): m/z = 452 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.008 (2.61), 0.008 (2.49), 1.311 (0.43), 1.432 (0.40),
1.452 (7.17), 1.469 (7.08), 2.523 (0.99), 4.295 (16.00), 5.135 (1.01), 5.154 (1.44), 5.172 (0.98), 6.974 (2.82), 6.995 (4.30), 7.069 (1.93), 7.090 (1.41), 7.117 (3.32), 7.125 (2.86), 7.148 (4.90),
7.170 (2.73), 7.399 (2.70), 7.413 (3.01), 7.421 (2.66), 7.434 (2.33), 7.491 (1.84), 7.495 (2.08),
7.500 (1.94), 7.504 (1.90), 8.008 (3.70), 8.012 (3.67), 8.622 (1.99), 8.642 (1.93), 10.804 (2.05).
Example 1-101
3-(3,4-Dimethylphenyl)-4-fluoro-1-oxo-/\/-[(2R)-4,4,4-trifluorobutan-2-yl]-1,2-dihydropyrrolo[1,2- a]pyrazine-7-carboxamide To a solution of 3-(3,4-dimethylphenyl)-1-oxo-/\/-[(2R)-4,4,4-trifluorobutan-2-yl]-1,2- dihydropyrrolo[1,2-a]pyrazine-7-carboxamide (Example 1-23, 20.0 g, 90% purity, 46.0 pmol) and A/,/\/-diisopropylethylamine (16 pi, 92 pmol) in dry acetonitrile (460 mI) at 0°C was added Selectfluor® (34.3 mg, 95% purity, 92.0 pmol). The mixture was sonicated for 5 min, then stirred at RT for 30 min. A further portion of Selectfluor® (68.6 mg, 95% purity, 184 pmol) and N,N- diisopropylethylamine (32 pi, 184 pmol) were added to the reaction and the reaction was stirred for a further 60 min. The reaction was cooled to 0°C, quenched with brine and the aqueous solution was extracted with ethyl acetate. The combined organic phase was washed with brine, dried and concentrated. The crude residue was suspended in pyridine (140 mI) and treated with T3P® (41 pi, ³50 wt.% in ethyl acetate, 140 pmol) and the reaction was heated to 90°C for 80 min. The reaction was cooled to RT, and purified by preparative HPLC to afford 4.90 mg (26% yield, 99% purity) of the title compound.
LC-MS (Method 8): Rt = 0.95 min; MS (ESIpos): m/z = 410 [M+H]+ Ή-NMR (500 MHz, DMSO-d6) d [ppm]: -0.006 (1.18), 0.006 (0.72), 1.239 (6.30), 1.253 (6.13), 2.261 (0.75), 2.278 (14.56), 2.283 (16.00), 2.465 (0.55), 2.475 (0.89), 2.518 (1.37), 2.559 (0.71), 2.565 (0.72), 2.582 (0.65), 2.589 (0.53), 4.332 (0.65), 4.344 (0.78), 4.356 (0.62), 7.263 (1.77), 7.279 (3.01), 7.320 (1.94), 7.336 (1.14), 7.395 (2.91), 7.442 (1.87), 7.446 (2.08), 7.450 (1.96), 7.453 (1.87), 7.926 (3.66), 7.929 (3.57), 8.301 (1.77), 8.318 (1.72), 10.833 (1.24). Example 1-102
3-(2,3-Dihydro-1,4-benzodioxin-6-yl)-4-fluoro-/\/-[(1R)-1-(6-methoxypyridin-3-yl)ethyl]-1-oxo- 1 ,2-dihydropyrrolo[1 ,2-a]pyrazine-7-carboxamide
Figure imgf000166_0001
To a solution of 3-(2,3-dihydro-1,4-benzodioxin-6-yl)-N-[(1R)-1-(6-methoxypyridin-3-yl)ethyl]-1- oxo-1, 2-dihydropyrrolo[1,2-a]pyrazine-7-carboxamide (Example 1-26, 48.0 g, 98% purity, 105 pmol) in DMF (960 pi) at RT was added /V-fluorobenzenesulfonimide (33.2 mg, 105 pmol). The reaction was stirred for 2.5 h at RT, then pyridine (480 mI) and treated with T3P® (250 mI, ³50 wt.% in ethyl acetate, 420 pmol) were added and the reaction was heated to 90°C for 2 h. The reaction was cooled to RT, and purified by preparative HPLC to afford 30.0 mg (57% yield, 93% purity) of the title compound.
LC-MS (Method 9): Rt = 1.55 min; MS (ESIpos): m/z = 465 [M+H]+
Ή-NMR (400 MHz, DMSO-d6) d [ppm]: -0.008 (2.35), 1.445 (0.52), 1.465 (4.69), 1.483 (4.47), 2.524 (1.37), 3.822 (16.00), 4.281 (2.06), 4.294 (10.06), 5.111 (0.75), 5.130 (1.04), 5.148 (0.69),
6.784 (2.07), 6.806 (2.07), 6.972 (1.88), 6.993 (2.82), 7.066 (1.31), 7.088 (0.94), 7.115 (2.09),
7.475 (1.36), 7.479 (1.52), 7.485 (1.44), 7.489 (1.38), 7.713 (1.35), 7.719 (1.43), 7.734 (1.26),
7.740 (1.31), 7.992 (2.52), 7.996 (2.50), 8.159 (2.10), 8.165 (1.99), 8.605 (1.33), 8.624 (1.27),
10.804 (1.43).
The compounds found in the following Table 2 are prepared using standard amide coupling conditions using one of the standard conditions listed.
General procedure 2-A To a solution of carboxylic acid (1.0 equiv.) and the required amine (1.5-2.0 equiv.) in pyridine (0.06 - 0.3 M) at RT was added 2,4,6-tripropyl-2,4,6-trioxo-1,3,5,2,4,6-trioxatriphosphorinane (T3P®) (³50 wt.% in ethyl acetate, 3.0 equiv.) dropwise. The reaction mixture was heated to 50°C for 1 h, then cooled to RT and purified by either preparative HPLC or silica gel column chromatography. General procedure 2-B
A solution of carboxylic acid (1.0 equiv.), HATU (1.2 equiv.) and DIPEA (2.5 - 3.0 equiv.) in NMP (0.07 - 0.2 M) was heated for 1 h at 50°C, then the corresponding amine (1.1 - 2.0 equiv) was added and the reaction was stirred overnight at 50°C. The reaction mixture was cooled to RT and purified by either preparative HPLC or silica gel column chromatography. General procedure 2-C
A solution of carboxylic acid (1.0 equiv.), CDI (1.1 equiv.) and DMAP (0.5 equiv.) in NMP (0.01 M) was stirred for 1 h at 50°C, then the corresponding amine (1.1 equiv.) was added and the reaction mixture was stirred overnight at 50°C. The reaction mixture was then cooled to RT and purified by either preparative HPLC or silica gel column chromatography. Table 2
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
General procedure 3 (parallel synthesis of example compounds )
6-(3,4-Dimethylphenyl)-8-oxo-7,8-dihydroimidazo[1,2-a]pyrazine-2-carboxylic acid (Intermediate 75A, 28.3 g, 0.1 mmol) was dissolved in DMF (0.8 ml) and HATU (53.2 mg, 0.14 mmol) was added and the mixture was stirred for 30 min. The corresponding amine (more than 0.2 mmol) was placed in one well of a 96 deep well plate and the carboxylic acid containing reaction mixture was added, followed by NMM (50 pi). The plate was sealed and shaken over night at RT, then the reaction mixture was filtered and the filtrate was purified by preparative LC-MS with one of the then following methods:
Instrument MS: Waters, Instrument HPLC: Waters (column Phenomenex Luna 5m C18(2) 100A, AXIA Tech. 50 x 21.2 mm, eluent A: water, eluent B: acetonitrile (ULC), gradient; flow: 38.5 ml/min + 1.5 ml/min modifier (10% aq. formic acid); UV-detection: DAD; 210-400 nm), or Instrument MS: Waters, Instrument HPLC: Waters (column Waters X-Bridge C18, 19 mm x 50 mm, 5 pm, eluent A: water, eluent B: acetonitrile (ULC), gradient; flow 38.5 ml/min + 1.5 ml/min modifier (10% aq. ammonia); UV-detection: DAD; 210 - 400 nm). The product containing fractions were evaporated in vacuo with a centrifugal dryer, dissolved in DMSO, pooled and evaporated again to give the final product.
The example compounds specified in the following Table 3 were prepared according to general procedure 3.
Table 3
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
The compounds found in the following Table 4 were prepared using standard amide coupling conditions. General procedure 4
To a 6-(naphthalen-2-yl)-8-oxo-7,8-dihydro[1,2,4]triazolo[1 ,5-a]pyrazine-2-carboxylic acid (Intermediate 91 A, 1.0 equiv.) and the required amine (1.5 equiv.) in pyridine at 50°C was added 2,4,6-tripropyl-2,4,6-trioxo-1,3,5,2,4,6-trioxatriphosphorinane (T3P®) (³50wt.% in ethyl acetate, 3.0 equiv.) dropwise over 45 - 90 min. The reaction mixture was stirred for 1 hour, then cooled to RT and purified by either preparative HPLC or silica gel column chromatography.
Table 4
Figure imgf000190_0001
Figure imgf000191_0001
EXPERIMENTAL SECTION - BIOLOGICAL ASSAYS
Biological investigations
The example testing experiments described herein serve to illustrate the present invention and the invention is not limited to the examples given.
The following assays can be used to illustrate the commercial utility of the compounds according to the present invention.
Examples were tested in selected biological assays one or more times. When tested more than once, data are reported as either average values or as median values, wherein
• the average value, also referred to as the arithmetic mean value, represents the sum of the values obtained divided by the number of times tested, and
• the median value represents the middle number of the group of values when ranked in ascending or descending order. If the number of values in the data set is odd, the median is the middle value. If the number of values in the data set is even, the median is the arithmetic mean of the two middle values.
Examples were synthesized one or more times. When synthesized more than once, data from biological assays represent average values calculated utilizing data sets obtained from testing of one or more synthetic batch.
The in vitro activity of the compounds of the present invention can be demonstrated in the following assays.
Biological assays
B-1. Cellular in vitro assay for determining EP3 receptor activity
The identification of antagonists of the EP3 receptor from humans and rats as well as the quantification of the activity of the compounds of the invention was carried out using recombinant cell lines. These cell lines originally derive from a hamster's ovary epithelial cell (Chinese Hamster Ovary, CHO K1, ATCC: American Type Culture Collection, Manassas, VA 20108, USA). The test cell lines constitutively express the human or rat EP3 receptors. The naturally Gai-coupled human and rat EP3 receptors were stably co-transfected with G-ne into cells that are also stably transfected with a mitochondrial form of the calcium-sensitive photoproteins Clytin (human EP3) or Photina (rat EP3), which, after reconstitution with the cofactor coelenterazine, emit light when there are increases in free calcium concentrations ( Nature 1992, 358, 325-327; Gene 1995, 153 (2), 273-274). The resulting EP3 receptor cells react to stimulation of the recombinantly expressed EP3 receptors by the agonist sulprostone with intracellular release of calcium ions, which can be quantified by the resulting photoprotein luminescence. Antagonists inhibit the receptor signaling, leading to reduced calcium release and luminescence. The bioluminescence of the cell lines is detected using a suitable luminometer (Trends Pharmacol. Sci. 1996, 17, 235-237).
Test procedure:
Human and rat EP3 cell lines:
On the day before the assay, the cells are plated out in culture medium (OptiMEM, 1% FCS, 2 mM glutamine, 10 mM HEPES, 5 pg/ml coelenterazine) in 384-well microtiter plates and kept in a cell incubator (96% humidity, 5% v/v CO2, 37°C). On the day of the assay, test compounds in various concentrations are placed for 10 minutes in the wells of the microtiter plate before the agonist sulprostone at EC50 concentration is added. The resulting light signal is measured immediately in a luminometer. All concentrations are measured in quadruples. Table A:
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
B-2. Inhibition of human platelet aggregation in vitro
Blood collection Human blood is collected by antecubital venipuncture using a 20G multifly set (Sarstedt, Num- brecht, Germany) into plastic tubes (Sarstedt 9 NC/10 ml monovettes) containing 3.8% (w/v) sodium citrate (1/10 v/v) from healthy donors who denied having taken any medication for at least 7 days prior to the study. Tubes are gently mixed to prevent coagulation.
Preparation of platelet rich plasma (PRP) and platelet poor plasma (PPP) Blood is centrifuged at 140 g for 20 min at room temperature and the supernatant PRP is immediately collected into Falcon tubes. PRP is further centrifuged (11.000 rpm, 1 min) to produce platelet poor plasma (PPP).
Measurement of platelet aggregation
Platelet aggregation is analyzed by light transmittance aggregometry according to the method of Born (J. Physiol. 1963, 168, 178-195) using APACT aggregometers (Rolf Greiner BioChemicals, Flacht, Germany).
178 pi of PRP is incubated with test compound in various concentrations dissolved in 2 mI DMSO in the aggregation cuvette at 37°C for 5 min. Then 2 mI sulprostone is added and is incubated at 37°C for 2 min. Aggregation is started by addition of 20 mI of collagen. The following concentrations of agonists are used: sulprostone 0.02 mM-2.5mM (Sigma-Aldrich), collagen 0.05-0.6 pg/ml (Horm®, Nycomed).
The reaction is followed by monitoring changes in light transmission on PRP aliquots against PPP control. Light transmittance is recorded for 5 minutes and the maximal aggregation value is used for IC50 calculations.
Statistical analysis
IC50 is calculated with GrapPad Prism (sigmoidal dose-response). All values are expressed as means ± SEM, with n denoting the number of blood donors. B-3. EP3 Binding Assay to identify inhibitors of PGE2 binding using scintillation proximity assay technique
To compound of interest (in DMSO, 2 pi, typical final concentration up to 20 mM) the following reagents are added in a total volume of 152 mI in reaction buffer (50 mM Tris/HCI, pH 7.5; 10 mM MgCI2; 1 mM EDTA) in a suitable 96 well clear bottom plate: [3H] Prostaglandin E2 ([5,6,8,11,12,14,15-3H(N)] in a final concentration of 1.5 nM;
EP3 Prostanoid Receptor Membrane Preparation (2 mg/ml) and
PVT-WGA Beads in a final concentration of 2.5 mg/ml.
The mixture is incubated for 2 h at room temperature. Scintillation is monitored in a suitable microplate reader equipped with 3H-SPA program. The values are used to plot dose response curves and calculate EC50 values for the test compounds.
B-4. Determination of the effect on laser-induced thrombus formation in mice (in vivo)
This study serves to investigate the efficacy of a test substance on thrombus formation, stability and resolution induced by laser injury to microvessels of atherosclerotic mice.
Animals Male ApoE -/- mice (Charles River) are fed an atherogenic diet (Ssniff S0602-S170, 20.85% fat, 0.15% cholesterol, 19.5% casein) from 6 weeks of age for at least 3 months. Test compound treatment is performed either orally via gavage to conscious animals before anesthesia or intravenously to anesthetized animals prior to injury.
Thrombus induction For oral treatment, animals are dosed by gavage in appropriate formulations by gavage prior to anesthesia. For intravenous treatment, a venous catheter will be implanted in the jugular vein for bolus or infusion drug delivery prior to the injury procedure. Mice are anesthetized by intraperitoneal injection of Ketamin (65 mg/kg) and Xylazin (15 mg/kg) The cremaster muscle is exposed after opening the scrotum and removal of a testis with attention to preserve blood supply from the circulation of the mouse. A stainless steel wire loop (Unimed 30.065, diameter 0.4 mm) is used to spread the inverted cremaster pocket in an organ bath constantly perfused with Dulbecco’s phosphate buffered saline temperated at 37°C. The tissue preparation is positioned on top of a transparent macrolon plastic stage for intravital microscopy (Leica AS LMD, water immersion objective Leica 506155 HC APO/ L40x/0.80). Video images are acquired by a digital camera (Basler PCO.EDGE 5.5) for identification of arterioles suitable for vascular wall injury (50 - 100 pm diameter, bifurcation-free segment).
A microdissection UV-Laser (wavelength 355 nm, Cell Surgeon, LLS Rowiak, Hannover, Germany) coupled into the optical path of the intravital microscope is used for 2-stage injury of the vessel wall: (1) Linear detachment of the endothelium along a length of approximately 150 pm. (2) Punctiform deep injury with local damage of the media. The resulting thrombus formation is visualized and recorded over a duration of 5 minutes. The lesion procedure will be repeated on an upstream segment of the identical vessel for at least two times. Statistical analysis
Video sequences of thrombus development are quantified by morphometric analysis of images at 10-second intervals using a customized software for semi-automatic vessel and thrombus detection (Zeta, Fraunhofer-lnstitut fur Angewandte Informationstechnik FIT, St. Augustin, Germany). Thrombus area and degree of stenosis are calculated to generate a thrombus-over-time-curve. The main derived result is the time above 75% occlusion of the vessel lumen. The mean of at least three repeated injuries per animal will be calculated. In case of intravenous testing, the treatment effect will be compared to baseline control injuries in the identical animals (t-test for repeated measurements). In case of oral treatment, thrombus development will be compared to vehicle- treated animals (ANOVA and t-test for independent measurements). A minimum of 5 animals will be tested to obtain sufficient statistical power.

Claims

Claims
1. Compound of the formula (I)
Figure imgf000198_0001
in which G1 represents CR4 or N,
G2 represents CH or N, with the proviso that G1 is N, if G2 is N,
R4 represents hydrogen, CrC4-alkyl, CrC4-halogenoalkyl, C3-C6-cycloalkyl or Ci- C3-alkoxymethyl, R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, phenylsulfonyl, C3-C6- cycloalkylsulfonyl, 2,3-dihydro-1H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano, methoxy, methylsulfonyl, carbamoyl, NRaRb (where Ra and Rb are independently selected from the group consisting of hydrogen, C1-C4- alkyl, C2-C6-halogenoalkyl and cyclopropyl) and CrC3-halogenoalkoxy, where halogenoalkoxy is substituted by 1 to 3 fluoro substituents, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxycarbonyl and NRcRd (where Rc and Rd are independently selected from the group consisting of hydrogen, CrC4-alkyl, C2-C6-halogenoalkyl and cyclopropyl), and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, ethyl, n-propyl, isopropyl, hydroxy, hydroxymethyl, trifluoromethyl and phenyl, or may be substituted by 1 or 2 fluoro, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro, chloro, cyano and methoxy, and
R5 represents hydrogen, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl or methoxycarbonyl, and where phenylsulfonyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro, chloro and cyano, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, methoxycarbonyl, carboxyl, carb amoyl, amino, dimethylamino, te/f-butoxycarbonylamino, C3-C6-cycloalkyl (which may be substituted by 1 hydroxy), azetidin-1-yl (which may be substituted by 1 or
2 fluoro), pyrrolidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin- 4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, pyrimidinyl, pyrazinyl, thienyl, pyrazolyl, oxazolyl, C3- C6-cycloalkyl, azetidinyl, pyrrolidinyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen, CrC4-alkyl, hydroxy, methoxy, trifluoromethyl, trifluoromethoxy, difluoromethoxy, dimethylaminomethyl, aminosulfonyl and pyrrolidin-1-ylmethyl, where pyridinyl may be substituted by 1 or 2 substituents independently selected from the group consisting of chloro, methyl, trifluoromethyl and methoxy, where pyrimidinyl may be substituted by 1 or 2 methyl substituents, where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro, CrC4-alkyl, trifluoromethyl, difluoromethyl, cyclopropyl and phenyl, where azetidinyl may be substituted by 1 substituent selected from the group consisting of fluoro, hydroxy, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to
3 fluoro, cyclopropyl, methylcarbonyl and methoxycarbonyl, and where pyrrolidinyl may be substituted by 1 substituent selected from the group consisting of fluoro, hydroxy, CrC4-alkyl, C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, cyclopropyl, methylcarbonyl and methoxycarbonyl,
R2 represents a group of the formula
Figure imgf000200_0001
where # is the point of attachment to the pyrazine ring,
Q1 represents CR8A or N,
Q2 represents CR8 or N,
R6 represents hydrogen, halogen, CrC4-alkyl, CrC4-halogenoalkyl, CrC4-alkoxy, CrC4-halogenoalkoxy or C3-C6-cycloalkyl,
R7 represents hydrogen, halogen, CrC4-alkyl, CrC4-halogenoalkyl, CrC4-alkoxy, CrC4-halogenoalkoxy or C3-C6-cycloalkyl, with the proviso, that at least one of R6 and R7 is not hydrogen,
R7A represents hydrogen or halogen,
R8 represents hydrogen or halogen,
R8A represents hydrogen or halogen,
R9 represents hydrogen, halogen or CrC4-alkyl,
R3 represents hydrogen, halogen, cyano, CrC4-alkyl or CrC2-halogenoalkyl, or one of the salts thereof, solvates thereof or solvates of the salts thereof. 2. Compound according to Claim 1, characterized in that
G1 represents CR4 or N,
G2 represents CH or N, with the proviso that G1 is N, if G2 is N,
R4 represents hydrogen, CrC4-halogenoalkyl or C3-C6-cycloalkyl,
R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, phenylsulfonyl, C3-C6- cycloalkylsulfonyl, 2,3-dihydro-1H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, ethyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
R5 represents CrC4-alkyl or C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, and where phenylsulfonyl may be substituted by 1 substituent fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy, carboxyl, dimethylamino, azetidin- 1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro and CrC4-alkyl,
R2 represents a group of the formula where # is the point of attachment to the pyrazine ring,
Q1 represents CR8A,
Q2 represents CR8,
R6 represents halogen or CrC4-alkyl,
R7 represents halogen, CrC4-alkyl or CrC4-alkoxy,
R7A represents hydrogen,
R8 represents hydrogen or halogen,
R8A represents hydrogen,
R9 represents hydrogen or halogen,
R3 represents hydrogen or halogen, or one of the salts thereof, solvates thereof or solvates of the salts thereof.
3. Compound according to Claim 1 or 2, characterized in that G1 represents CR4,
G2 represents CH,
R4 represents hydrogen, CrC4-halogenoalkyl or C3-C6-cycloalkyl,
R1 represents CrCyalkyl, C2-C6-halogenoalkyl, C3-C6-cycloalkyl, phenylsulfonyl, C3-C6- cycloalkylsulfonyl or the group -L-RE, where alkyl may be substituted by 1 substituent hydroxy, and where halogenoalkyl is substituted by 1 to 6 fluoro substituents and may be further substituted by 1 substituent hydroxy, and where in the cycloalkyl ring one CH2 group may be replaced by NR5, and where the cycloalkyl may be substituted by 1 substituent phenyl, where the phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of fluoro and methoxy, and
R5 represents CrC4-alkyl, and where phenylsulfonyl may be substituted by 1 substituent fluoro, and
L represents Ci-C6-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, dimethylamino, azetidin-1-yl (which may be substituted by 1 or 2 fluoro) and morpholin-4-yl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl or C3-C6-cycloalkyl, where phenyl may be substituted by 1 substituent halogen, and where pyridinyl may be substituted by 1 substituent methoxy,
R2 represents a group of the formula
Figure imgf000203_0001
where # is the point of attachment to the pyrazine ring, Q1 represents CR8A,
Q2 represents CR8,
R6 represents halogen or CrC4-alkyl,
R7 represents halogen, CrC4-alkyl or CrC4-alkoxy, R7A represents hydrogen,
R8 represents hydrogen or fluoro,
R8A represents hydrogen,
R9 represents hydrogen or fluoro,
R3 represents hydrogen or fluoro, or one of the salts thereof, solvates thereof or solvates of the salts thereof.
4. Compound according to Claim 1 or 2, characterized in that
G1 represents N,
G2 represents CH,
R1 represents CrCyalkyl, C3-C6-cycloalkyl, 2,3-dihydro-1H-inden-1-yl or the group -L-RE, where alkyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, cyano and methoxy, and where in the cycloalkyl ring one CH2 group may be replaced by O, SO2 or NR5, and where the cycloalkyl may be substituted by 1 substituent selected from the group consisting of methyl, hydroxymethyl and phenyl, where the phenyl may be substituted by 1 substituent fluoro, and
R5 represents CrC4-alkyl or C2-C6-halogenoalkyl substituted by 1 to 3 fluoro, and where 2,3-dihydro-1H-inden-1-yl may be substituted by 1 hydroxy substituent, and
L represents Ci-C4-alkanediyl, where alkanediyl may be substituted by 1 or 2 substituents independently selected from the group consisting of hydroxy, methoxy and carboxyl, and additionally by up to 3 fluoro,
RE represents phenyl, pyridinyl, thienyl, pyrazolyl, oxazolyl, C3-C6-cycloalkyl or oxanyl, where phenyl may be substituted by 1 or 2 substituents independently selected from the group consisting of halogen and trifluoromethyl, where pyridinyl may be substituted by 1 substituent methoxy, and where pyrazolyl may be substituted by 1 to 3 substituents independently selected from the group consisting of chloro and CrC4-alkyl,
R2 represents a group of the formula where # is the point of attachment to the pyrazine ring,
Q1 represents CR8A,
Q2 represents CR8,
R6 represents halogen or CrC4-alkyl,
R7 represents halogen or CrC4-alkyl,
R7A represents hydrogen,
R8 represents hydrogen,
R8A represents hydrogen,
R9 represents hydrogen,
R3 represents hydrogen, or one of the salts thereof, solvates thereof or solvates of the salts thereof.
5. Compound according to Claim 1 or 2, characterized in that G1 represents N,
G2 represents N,
R1 represents the group -L-RE,
L represents C2-C4-alkanediyl, where alkanediyl may be substituted by up to 3 fluoro,
RE represents phenyl or pyridinyl, where phenyl may be substituted by 1 substituent of halogen, and where pyridinyl may be substituted by 1 substituent methoxy,
R2 represents a group of the formula
Figure imgf000205_0001
where # is the point of attachment to the pyrazine ring,
R3 represents hydrogen, or one of the salts thereof, solvates thereof or solvates of the salts thereof.
6. Process for preparing a compound of the formula (I) or one of the salts thereof, solvates thereof or solvates of the salts thereof according to one of Claims 1 to 5, characterized in that
[A] a compound of the formula (II)
NH2
R1/ (II), in which
R1 is as defined in claims 1 to 5, is reacted with a compound of the formula (III)
Figure imgf000206_0001
in which
G1, G2, R2 and R3 are as defined in claims 1 to 5, in the presence of a dehydrating agent to give compounds of the formula (I).
7. Compound according to any of Claims 1 to 5 for the treatment and/or prophylaxis of diseases.
8. Compound according to any of Claims 1 to 5 for the treatment and/or prophylaxis of thrombotic or thromboembolic disorders, diabetes, urogenital and ophthalmic disorders.
9. Use of a compound according to any of Claims 1 to 5 for producing a medicament for the treatment and/or prophylaxis of diseases.
10. Use of a compound according to any of Claims 1 to 5 for producing a medicament for the treatment and/or prophylaxis of thrombotic or thromboembolic disorders, diabetes, urogenital and ophthalmic disorders.
11. Medicament comprising a compound according to any of Claims 1 to 5 in combination with an inert, nontoxic, pharmaceutically suitable excipient.
12. Medicament according to Claim 11 for the treatment and/or prophylaxis of thrombotic or thromboembolic disorders, diabetes, urogenital and ophthalmic disorders.
13. Method for the treatment and/or prophylaxis of thrombotic or thromboembolic disorders, diabetes, urogenital and ophthalmic disorders in humans and animals by administration of a therapeutically effective amount of at least one compound according to any of Claims 1 to 5, of a medicament according to Claim 11 or 12 or of a medicament obtained according to
Claim 9 or 10.
INTERNATIONAL SEARCH REPORT
Box No. IV Text of the abstract (Continuation of item 5 of the first sheet)
Figure imgf000208_0001
Substituted pyrazine carboxamide derivatives
The invention relates to substituted pyrazine carboxamide derivatives of formula (I) and to processes for their preparation, and also to their use for preparing medicaments for the treatment and/ or prophylaxis of diseases, in particular cardiovascular disorders, preferably thrombotic or thromboembolic disorders, and diabetes, and also urogenital and ophthalmic disorders.
Figure imgf000208_0002
Form PCT/ISA/210 (continuation of first sheet (3)) (July2009)
PCT/EP2020/081250 2019-11-12 2020-11-06 Substituted pyrazine carboxamide derivatives as prostaglandin ep3 receptor antagonists WO2021094210A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019117537 2019-11-12
CNPCT/CN2019/117537 2019-11-12

Publications (1)

Publication Number Publication Date
WO2021094210A1 true WO2021094210A1 (en) 2021-05-20

Family

ID=73172719

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2020/081250 WO2021094210A1 (en) 2019-11-12 2020-11-06 Substituted pyrazine carboxamide derivatives as prostaglandin ep3 receptor antagonists

Country Status (1)

Country Link
WO (1) WO2021094210A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114409681A (en) * 2022-03-18 2022-04-29 信义核新(北京)生物科技有限公司 Quinolone analog for treating DNA damage repair defect tumor and application thereof

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000006569A1 (en) 1998-07-29 2000-02-10 Bayer Aktiengesellschaft Substituted pyrazole derivatives condensed with six-membered heterocyclic rings
WO2000006568A1 (en) 1998-07-29 2000-02-10 Bayer Aktiengesellschaft Substituted pyrazole derivatives
WO2001019778A1 (en) 1999-09-13 2001-03-22 Bayer Aktiengesellschaft Novel dicarboxylic acid derivatives with pharmaceutical properties
WO2001019780A2 (en) 1999-09-13 2001-03-22 Bayer Aktiengesellschaft Novel derivatives of dicarboxylic acid having pharmaceutical properties
WO2001019355A2 (en) 1999-09-13 2001-03-22 Bayer Aktiengesellschaft Novel dicarboxylic acid derivatives with pharmaceutical properties
WO2001019776A2 (en) 1999-09-13 2001-03-22 Bayer Aktiengesellschaft Novel derivatives of dicarboxylic acid having pharmaceutical properties
WO2002042301A1 (en) 2000-11-22 2002-05-30 Bayer Aktiengesellschaft Novel pyridine-substituted pyrazolopyridine derivatives
WO2002070462A1 (en) 2001-03-07 2002-09-12 Bayer Aktiengesellschaft Substituted amino dicarboxylic acid derivatives
WO2002070510A2 (en) 2001-03-07 2002-09-12 Bayer Aktiengesellschaft Amino dicarboxylic acid derivatives with pharmaceutical properties
WO2003095451A1 (en) 2002-05-08 2003-11-20 Bayer Healthcare Ag Carbamate-substituted pyrazolopyridines
WO2009082687A1 (en) 2007-12-21 2009-07-02 Genentech, Inc. Azaindolizines and methods of use
WO2010105770A1 (en) 2009-03-18 2010-09-23 Bayer Schering Pharma Aktiengesellschaft Substituted 2-acetamido-5-aryl-1,2,4-triazolones and use thereof
WO2011104322A1 (en) 2010-02-27 2011-09-01 Bayer Pharma Aktiengesellschaft Bisaryl-bonded aryltriazolones and use thereof
WO2011147809A1 (en) 2010-05-26 2011-12-01 Bayer Pharma Aktiengesellschaft Substituted 5-fluoro-1h-pyrazolopyridines and use thereof
WO2012004258A1 (en) 2010-07-09 2012-01-12 Bayer Pharma Aktiengesellschaft Ring-fused pyrimidines and triazines and use thereof for the treatment and/or prophylaxis of cardiovascular diseases
WO2012028647A1 (en) 2010-09-03 2012-03-08 Bayer Pharma Aktiengesellschaft Bicyclic aza heterocycles, and use thereof
WO2012054233A1 (en) 2010-10-18 2012-04-26 E. I. Du Pont De Nemours And Company Nematocidal sulfonamides
WO2012059549A1 (en) 2010-11-04 2012-05-10 Bayer Pharma Aktiengesellschaft Substituted 6-fluoro-1h-pyrazolo[4,3-b]pyridines and use thereof
US20150099782A1 (en) * 2013-10-09 2015-04-09 Pfizer Inc. Antagonists of prostaglandin ep3 receptor
WO2016057522A1 (en) 2014-10-06 2016-04-14 Flatley Discovery Lab Triazolopyridine compounds and methods for the treatment of cystic fibrosis
WO2016071212A1 (en) 2014-11-03 2016-05-12 Bayer Pharma Aktiengesellschaft Hydroxyalkyl-substituted phenyltriazole derivatives and uses thereof
US20160176851A1 (en) * 2014-12-22 2016-06-23 Pfizer Inc. Antagonists of prostaglandin ep3 receptor
WO2019219517A1 (en) * 2018-05-17 2019-11-21 Bayer Aktiengesellschaft Substituted dihydropyrazolo pyrazine carboxamide derivatives

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000006569A1 (en) 1998-07-29 2000-02-10 Bayer Aktiengesellschaft Substituted pyrazole derivatives condensed with six-membered heterocyclic rings
WO2000006568A1 (en) 1998-07-29 2000-02-10 Bayer Aktiengesellschaft Substituted pyrazole derivatives
WO2001019778A1 (en) 1999-09-13 2001-03-22 Bayer Aktiengesellschaft Novel dicarboxylic acid derivatives with pharmaceutical properties
WO2001019780A2 (en) 1999-09-13 2001-03-22 Bayer Aktiengesellschaft Novel derivatives of dicarboxylic acid having pharmaceutical properties
WO2001019355A2 (en) 1999-09-13 2001-03-22 Bayer Aktiengesellschaft Novel dicarboxylic acid derivatives with pharmaceutical properties
WO2001019776A2 (en) 1999-09-13 2001-03-22 Bayer Aktiengesellschaft Novel derivatives of dicarboxylic acid having pharmaceutical properties
WO2002042301A1 (en) 2000-11-22 2002-05-30 Bayer Aktiengesellschaft Novel pyridine-substituted pyrazolopyridine derivatives
WO2002070462A1 (en) 2001-03-07 2002-09-12 Bayer Aktiengesellschaft Substituted amino dicarboxylic acid derivatives
WO2002070510A2 (en) 2001-03-07 2002-09-12 Bayer Aktiengesellschaft Amino dicarboxylic acid derivatives with pharmaceutical properties
WO2003095451A1 (en) 2002-05-08 2003-11-20 Bayer Healthcare Ag Carbamate-substituted pyrazolopyridines
WO2009082687A1 (en) 2007-12-21 2009-07-02 Genentech, Inc. Azaindolizines and methods of use
WO2010105770A1 (en) 2009-03-18 2010-09-23 Bayer Schering Pharma Aktiengesellschaft Substituted 2-acetamido-5-aryl-1,2,4-triazolones and use thereof
WO2011104322A1 (en) 2010-02-27 2011-09-01 Bayer Pharma Aktiengesellschaft Bisaryl-bonded aryltriazolones and use thereof
WO2011147809A1 (en) 2010-05-26 2011-12-01 Bayer Pharma Aktiengesellschaft Substituted 5-fluoro-1h-pyrazolopyridines and use thereof
WO2012004258A1 (en) 2010-07-09 2012-01-12 Bayer Pharma Aktiengesellschaft Ring-fused pyrimidines and triazines and use thereof for the treatment and/or prophylaxis of cardiovascular diseases
WO2012028647A1 (en) 2010-09-03 2012-03-08 Bayer Pharma Aktiengesellschaft Bicyclic aza heterocycles, and use thereof
WO2012054233A1 (en) 2010-10-18 2012-04-26 E. I. Du Pont De Nemours And Company Nematocidal sulfonamides
WO2012059549A1 (en) 2010-11-04 2012-05-10 Bayer Pharma Aktiengesellschaft Substituted 6-fluoro-1h-pyrazolo[4,3-b]pyridines and use thereof
US20150099782A1 (en) * 2013-10-09 2015-04-09 Pfizer Inc. Antagonists of prostaglandin ep3 receptor
WO2015052610A1 (en) 2013-10-09 2015-04-16 Pfizer Inc. Antagonists of prostaglandin ep3 receptor
WO2016057522A1 (en) 2014-10-06 2016-04-14 Flatley Discovery Lab Triazolopyridine compounds and methods for the treatment of cystic fibrosis
WO2016071212A1 (en) 2014-11-03 2016-05-12 Bayer Pharma Aktiengesellschaft Hydroxyalkyl-substituted phenyltriazole derivatives and uses thereof
US20160176851A1 (en) * 2014-12-22 2016-06-23 Pfizer Inc. Antagonists of prostaglandin ep3 receptor
WO2016103097A1 (en) 2014-12-22 2016-06-30 Pfizer Inc. Antagonists of prostaglandin ep3 receptor
WO2019219517A1 (en) * 2018-05-17 2019-11-21 Bayer Aktiengesellschaft Substituted dihydropyrazolo pyrazine carboxamide derivatives

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
ACS MED. CHEM. LETT., vol. 1, 2010, pages 316 - 320
AM J RESPIR CRIT CARE, 2009, pages 923 - 928
ARCH. INTERN. MED., vol. 162, 2002, pages 2405 - 2410
BIOORG. MED. CHEM. LETT., vol. 19, 2009, pages 4292 - 4295
BORN, J. PHYSIOL., vol. 168, 1963, pages 178 - 195
CARDIOVASC. RES., vol. 101, 2014, pages 482 - 491
CIRCULATION, vol. 104, 2001, pages 921 - 927
CITATION OF NMR PEAKLIST DATA WITHIN PATENT APPLICATIONS, 1 August 2014 (2014-08-01), Retrieved from the Internet <URL:http://www.researchdisclosure.com/searching-disclosures>
DIABETES, vol. 62, 2013, pages 1904 - 1912
EMBO MOL MED, 2018, pages e8536
EUR. J. PHARMACOL., vol. 194, 1991, pages 63 - 70
GENE, vol. 153, no. 2, 1995, pages 273 - 274
J. EXP. MED., vol. 204, 2007, pages 311 - 320
LANCET, vol. 377, 2011, pages 2193 - 2204
N. ENGL. J. MED., vol. 357, 2007, pages 2001 - 2015
N. ENGL. J. MED., vol. 377, 2017, pages 1319 - 1330
NATURE, vol. 358, 1992, pages 325 - 327
PLATELETS, vol. 21, 2010, pages 329 - 342
PROSTAGLANDINS & OTHER LIPID MEDIATORS, vol. 121, 2015, pages 4 - 16
TRENDS PHARMACOL. SCI., vol. 17, 1996, pages 235 - 237

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114409681A (en) * 2022-03-18 2022-04-29 信义核新(北京)生物科技有限公司 Quinolone analog for treating DNA damage repair defect tumor and application thereof

Similar Documents

Publication Publication Date Title
CA3100221A1 (en) Substituted dihydropyrazolo pyrazine carboxamide derivatives
KR20180026761A (en) Substituted oxopyridine derivatives
US9695131B2 (en) Substituted uracils as chymase inhibitors
KR102438965B1 (en) Substituted 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridin-3(2H)-one and 2,5,6,7-tetrahydro-3H- pyrrolo[2,1-c][1,2,4]triazol-3-one, and uses thereof
AU2018354785A1 (en) Substituted imidazopyridine amides and use thereof
US20230265072A1 (en) Substituted pyrazolo piperidine carboxylic acids
EP4259617A1 (en) Substituted pyrazolo piperidine carboxylic acids
WO2021094210A1 (en) Substituted pyrazine carboxamide derivatives as prostaglandin ep3 receptor antagonists
US20230346777A1 (en) Use of sgc activators for the treatment of ophthalmologic diseases
WO2022122913A1 (en) Substituted pyrazolo piperidine carboxylic acids
EP4259618A1 (en) Substituted pyrazolyl piperidine carboxylic acids
WO2021094208A1 (en) Substituted imidazo pyrimidine ep3 antagonists
WO2021094209A1 (en) Substituted pyrrolo triazine carboxamide derivatives as prostaglandin ep3 receptor antagonists

Legal Events

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

Ref document number: 20803529

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20803529

Country of ref document: EP

Kind code of ref document: A1